Sunteți pe pagina 1din 602

Monitoring in

Neurocritical Care
Monitoring in
Neurocritical Care

Peter D. le Roux, MD, FACS


Associate Professor of Neurosurgery
Perelman School of Medicine at the University of
Pennsylvania;
Department of Neurosurgery
Pennsylvania Hospital
Philadelphia, Pennsylvania

Joshua M. Levine, MD
Assistant Professor
Departments of Neurology, Neurosurgery, and
Anesthesiology and Critical Care
Perelman School of Medicine at the University of
Pennsylvania;
Co-Director, Neurointensive Care Unit
Hospital of the University of Pennsylvania
Philadelphia, Pennsylvania

W. Andrew Kofke, MD, MBA, FCCM


Professor
Departments of Anesthesiology and Critical Care and
Neurosurgery
Perelman School of Medicine at the University of
Pennsylvania;
Director, Neuroanesthesia
Department of Anesthesiology and Critical Care
University of Pennsylvania Health System;
Co-Director, Neurointensive Care Unit
Hospital of the University of Pennsylvania
Philadelphia, Pennsylvania
1600 John F. Kennedy Blvd.
Ste 1800
Philadelphia, PA 19103-2899

MONITORING IN NEUROCRITICAL CARE ISBN: 978-1-4377-0167-8


Copyright © 2013 by Saunders, an imprint of Elsevier Inc.
Chapter 22: “Renal, Electrolyte, and Acid Base Assessment”: Guy M. Dugan retains copyright to his original
contribution.
Chapter 23: “Gastrointestinal and Hepatic Disorders” by Christiana E. Hall and Aashish R. Patel:
Christiana E. Hall retains copyright to her original portion of the contribution only.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic or
mechanical, including photocopying, recording, or any information storage and retrieval system, without
permission in writing from the Publisher. Details on how to seek permission, further information about the
Publisher’s permissions policies and our arrangements with organizations such as the Copyright Clearance
Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.

This book and the individual contributions contained in it are protected under copyright by the Publisher
(other than as may be noted herein).

Notices

Knowledge and best practice in this field are constantly changing. As new research and experience broaden
our understanding, changes in research methods, professional practices, or medical treatment may become
necessary.
Practitioners and researchers must always rely on their own experience and knowledge in evaluating
and using any information, methods, compounds, or experiments described herein. In using such
information or methods, they should be mindful of their own safety and the safety of others, including
parties for whom they have a professional responsibility.
With respect to any drug or pharmaceutical products identified, readers are advised to check the most
current information provided (i) on procedures featured or (ii) by the manufacturer of each product to be
administered, to verify the recommended dose or formula, the method and duration of administration,
and contraindications. It is the responsibility of practitioners, relying on their own experience and
knowledge of their patients, to make diagnoses, to determine dosages and the best treatment for each
individual patient, and to take all appropriate safety precautions.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume
any liability for any injury and/or damage to persons or property as a matter of products liability,
negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas
contained in the material herein.

Library of Congress Cataloging-in-Publication Data


  Monitoring in neurocritical care / [edited by] Peter D. Le Roux, Joshua M. Levine, W. Andrew Kofke.
    p. ; cm.
   Includes bibliographical references and index.
   ISBN 978-1-4377-0167-8 (hardcover : alk. paper)
   I. Le Roux, Peter D.  II. Levine, Joshua M.  III. Kofke, W. Andrew.
   [DNLM:1.  Monitoring, Intraoperative–methods.  2.  Neurosurgical Procedures.  3.  Central Nervous
System Diseases–surgery.  4.  Intensive Care–methods.  5.  Intensive Care Units–organization &
administration. WL 368]
   616.02’8–dc23
2012036807

Content Strategist: Charlotta Kryhl


Senior Content Development Specialist: Janice Gaillard
Content Development Specialist: Angela Rufino
Publishing Services Manager: Anne Altepeter
Senior Project Manager: Cheryl A. Abbott
Project Manager: Louise King
Design Direction: Louis Forgione Working together to grow
libraries in developing countries
Printed in China www.elsevier.com | www.bookaid.org | www.sabre.org

Last digit is the print number:  9  8  7  6  5  4  3  2  1 


To the ICU nurses who take care of all our patients
Preface
Neurocritical care has evolved rapidly in the past 10 years. understanding of what information the technology provides.
It is a specialty that focuses on the critical care manage- These advances have been chronicled in numerous contribu-
ment of patients with catastrophic neurologic diseases, tions to the scientific literature, the sheer volume of which
including primary neurologic pathologies such as traumatic makes it difficult for healthcare providers and device engi-
brain injury (TBI), ischemic stroke, intracerebral hemorrhage neers to keep up to date with the knowledge necessary to
(ICH), subarachnoid hemorrhage (SAH), brain tumors, provide the best patient care. In addition, although several
infection (e.g., HIV, TB, meningitis), spinal cord injury, and textbooks on critical care or head injury briefly discuss moni-
acute ascending neuropathies. In addition, neurologic dys- toring in a chapter or two, there is a paucity of information
function occurs in many diverse systemic disorders, including that summarizes all aspects of neuromonitoring and no text-
hypoxia (e.g., post cardiac arrest, near drowning), liver dys- book that is dedicated to monitoring in neurocritical care.
function, electrolyte abnormalities, high altitude sickness, This book, Monitoring in Neurocritical Care, represents a
eclampsia, lead intoxication, and malignant hypertension. comprehensive review of neuromonitoring. We have designed
There are many reasons why brain injury or damage occurs this textbook to provide the reader with a practical but in-
in patients with neurologic disorders. In particular, multiple depth reference that describes the scientific basis and rationale
experimental and clinical studies have demonstrated a close for use of a particular monitor, the information it provides,
relationship between variables such as hypoxia, increased and how this information can be used to manage the neuro-
intracranial pressure, arterial hypotension, hyperglycemia, critical care patient in an integrated fashion. We have been
fever, and seizures with neurologic outcome, and accumulat- fortunate to have chapters written by authors from around the
ing evidence suggests that brain damage evolves over time. world. The contributors are clinicians, engineers, information
Minimizing the burden of this delayed, or “secondary,” brain technology experts, and researchers who have extensive expe-
injury, has become the focus of modern neurocritical care. rience in the field, and each has provided an excellent and
However, despite much research, trials in neuroprotection timely review. We hope that the reader will gain a comprehen-
have largely failed, in part because of their association with sive understanding about neuromonitoring and neurocritical
prognostic heterogeneity, multiple mechanisms of cellular care and an insight into existent controversies and potential
damage, and a paucity of early mechanistic endpoints. This future management. Its contents will be relevant to neurolo-
has led to a realization that strategies of care, or “bundles,” gists, neurosurgeons, neuroanesthesiologists, neurointensiv-
rather than single agents, and approaches that are tailored to ists, and neuroscience nurses, and will serve as a useful resource
individual patient physiology and pathophysiology are neces- to intensivists working in medical and surgical ICUs. For those
sary to improve outcome. who are interested in clinical or laboratory research on brain
In daily practice neurointensivists focus on the recognition injury in its broad sense, this book will provide many ideas
of subtle changes in the neurologic condition, interactions and references and will be a stepping-stone to further progress
between the brain and systemic derangements, and brain in understanding a complex problem. The text also will serve
physiology. The challenge for intensivists today is to identify as a reference and guide for many engineers, bioengineers, and
individuals who are at risk of developing disease or secondary computer experts who work on medical device and bioinfor-
injury, determine disease severity, and distinguish responders matics development.
from nonresponders to therapy (i.e., individualized and tar- We have divided the book into seven sections. Section I,
geted medicine). Monitoring is one tool that may answer these Background, provides information about cerebral metabo-
challenges, and it has become central to the management of lism, the principles of neurocritical care, informatics, quality
secondary brain injury and to individualized care. In recent assessment, the role of ICU design and nursing, specific con-
years (interestingly in concert with the evolution of neuro- siderations in children, the effects of anesthetic agents on
critical care as a distinct specialty), technology developments monitors, and a discussion on the relationship between bio-
have resulted in several new monitoring techniques that ethics and monitoring. Section II, Clinical and Laboratory
provide the neurointensivist with information about brain Assessment, reviews clinical evaluation, sedation, pain, delir-
and cellular function. Techniques to better monitor function ium, outcomes such as neuropsychological and brain death,
of the heart, lung, liver, kidney, and blood also have evolved. extracerebral organ systems, laboratory analysis, and the role
In addition, when the various techniques are combined (“mul- of biomarkers. Section III, Electrophysiology, is devoted to
timodal monitoring”), a more accurate overall picture of brain evoked potentials and electroencephalography. Section IV,
function is produced. This approach, along with new com- Radiology, discusses the use and integration of various tech-
puter systems that integrate data at the bedside, and the niques, including computed tomography, xenon-CT, MRI,
emerging field of bioinformatics may change the way patients PET, and SPECT in neurocritical care. Section V, Cerebral
with brain injury are managed in the future. Blood Flow, is a review of techniques such as neurosonology,
In the last decade, there have been many advances in laser Doppler flowmetry, thermal diffusion flowmetry, jugular
neurocritical care monitoring technology and a better bulb oximetry, and near infrared spectroscopy. Section VI,
vii
viii Preface

Intracranial Monitoring, provides an in-depth review of We would like to express our appreciation and acknowledge
invasive techniques, including intracranial pressure, brain the efforts of all contributors to this volume. We also thank
oxygen, cerebral microdialysis, and brain temperature. The the editorial, design, and production staff at Elsevier Science,
final section, Computers, Engineering, and the Future, pro- in particular Janice Gaillard, Charlotta Kryhl, Julie Goolsby,
vides a description of device development, engineering, sim- Kate Crowley, Angela Rufino, Louis Forgione, Cheryl Abbott,
ulation, telemedicine, robotics, information processing, data Louise King, and Anne Altepeter, who have been very helpful
acquisition and storage, medical informatics and multimo- in producing this volume, and Yolanda Caban who provided
dality monitoring, noninvasive brain monitoring, and a dis- excellent administrative assistance. Finally, we thank Barbara
cussion of potential future developments. It is important for Williams who provided outstanding editorial assistance and
the reader to realize that the “ideal brain monitor” does not made this book possible.
yet exist and no single monitor will by itself affect outcome. Peter D. le Roux, MD, FACS
Instead, it is the information provided by a monitor and how Joshua M. Levine, MD
we as healthcare providers interpret and apply the informa- W. Andrew Kofke, MD, MBA, FCCM
tion that has the potential to improve outcome and lead to
new insights into disease processes.
Contributors
Pippa G. Al-Rawi, BSc Rosette C. Biester, PhD Maurizio Cereda, MD
Research Associate Polytrauma Neuropsychologist Assistant Professor
Neurosurgery Unit Department of Behavioral Health Department of Anesthesiology and Critical Care
Department of Clinical Neurosciences Philadelphia Veterans Affairs Medical Center; Perelman School of Medicine at the University
University of Cambridge/Addenbrooke’s Auxiliary Health Care Provider of Pennsylvania
Hospital Department of Physical Medicine and Philadelphia, Pennsylvania
Cambridge, United Kingdom Rehabilitation
University of Pennsylvania Health System Randall M. Chesnut, MD, FCCM, FACS
Pamela J. Amelung, MD Philadelphia, Pennsylvania Integra Endowed Professor of Neurotrauma
Clinical Associate Professor Department of Neurological Surgery
Department of Medicine Peter M. Black, MD, PhD Department of Orthopaedic Surgery
University of Maryland School of Medicine; Center for Advanced Brain and Spine Surgery Adjunct Professor, School of Global Health
Physician Liaison Natick, Massachusetts; Harborview Medical Center, University of
Philips VISICU Professor of Neurosurgery Washington
Baltimore, Maryland Department of Surgery Seattle, Washington
Harvard Medical School
Michal Arkuszewski, MD, PhD Boston, Massachusetts Jan Claassen, MD
Department of Neurology, Central University Attending Neurointensivist
Hospital Thomas P. Bleck, MD, FCCM Department of Neurology
Medical University of Silesia Professor Division of Neurocritical Care
Katowice, Poland; Departments of Neurological Sciences, Columbia University College of Physicians and
Department of Radiology, Neuroradiology Neurosurgery, Anesthesiology, and Medicine Surgeons
Division Rush Medical College; New York, New York
Perelman School of Medicine at the University Associate Chief Medical Officer for Critical Care
of Pennsylvania Rush University Medical Center Wendy A. Cohen, MD
Philadelphia, Pennsylvania Chicago, Illinois Professor
Departments of Radiology and Neurological
Syed T. Arshad, MD Jens Bracht, Dipl.-Phys. Surgery
Neuro-Intensivist Technical Director R&D, PD University of Washington School of Medicine
Department of Neurosurgery GMS mbH Seattle, Washington
Sacramento Medical Center/Kaiser Permanente Kiel, Germany
North Valley E. Sander Connolly, Jr., MD
Sacramento, California M. Ross Bullock, MD, PhD Bennett M. Stein Professor and Vice Chairman
Professor Department of Neurological Surgery
Ramani Balu, MD, PhD Department of Neurological Surgery Columbia University College of Physicians and
Fellow Director, Clinical Neurotrauma Surgeons;
Department of Neurology Department of Neurological Surgery Director, Cerebrovascular Research Laboratory
Division of Neurocritical Care University of Miami Miller School of Medicine Surgical Director, Neuro-Intensive Care Unit
Perelman School of Medicine at the University Miami, Florida Department of Neurological Surgery
of Pennsylvania Columbia University Medical Center/New
Philadelphia, Pennsylvania Andrew P. Carlson, MD York-Presbyterian
Department of Neurological Surgery New York, New York
Sarice L. Bassin, MD University of New Mexico School of Medicine
Assistant Professor Albuquerque, New Mexico Marek Czosnyka, PhD
Department of Neurology Reader in Brain Physics
Northwestern University Feinberg School of Emmanuel Carrera, MD Department of Clinical Neurosciences,
Medicine; Department of Neurology Neurosurgery Unit
Program Director, Neurocritical Care Fellowship Centre Hospitalier Universitaire Vaudois University of Cambridge/Addenbrooke’s
Division of Neurocritical Care (CHUV) Hospital
Northwestern Memorial Hospital Lausanne University Hospital Cambridge, United Kingdom
Chicago, Illinois Lausanne, Switzerland
John A. Detre, MD
David M. Benglis, Jr., MD Professor
Atlanta Brain and Spine Care Departments of Neurology and Radiology
Atlanta, Georgia Perelman School of Medicine at the University
of Pennsylvania
Philadelphia, Pennsylvania

ix
x Contributors

Martin E. Doerfler, MD Thomas Geeraerts, MD, PhD K.T. Henrik Huttunen, MD


Associate Professor Anesthesiology and Critical Care Department Clinical Assistant Professor of Anesthesiology
Departments of Medicine and Science Education University Hospital of Toulouse Department of Anesthesiology, Pharmacology,
Hofstra North Shore-LIJ School of Medicine at University Paul Sabatier and Therapeutics
Hofstra University Toulouse, France University of British Columbia Faculty of
Hempstead, New York; Medicine;
Vice President, Evidence Based Clinical Practice Vicente H. Gracias, MD Attending Anesthesiologist, Vancouver Acute
and Clinical Integration Professor Department of Anesthesia
North Shore-LIJ Health System Department of Surgery Division of Neuroanesthesia
Lake Success, New York Chief, Trauma/Surgical Critical Care Vancouver General Hospital
UMDNJ-Robert Wood Johnson Medical School; Vancouver, British Columbia, Canada
Guy M. Dugan, MD, FCCP Director, Level I Trauma Center
Director Robert Wood Johnson University Hospital Peter J. Kirkpatrick, MSc, FRCS(SN),
Department of Critical Care and Neurocritical New Brunswick, New Jersey FMedSci
Care Fellow of Medical Academy of Sciences
Alexian Brothers Medical Center David M. Greer, MD, MA, FCCM, FAHA Society of British Neurosurgeons (SBNS)
Elk Grove Village, Illinois Dr. Harry M. Zimmerman and Dr. Nicholas and Consultant Neurosurgeon
Viola Spinelli Professor and Vice Chairman Honorary University Lecturer
Richard P. Dutton, MD, MBA Director, Neurosciences Intensive Care Unit Department of Neurosurgery
Clinical Associate Neurology Residency Program Director Addenbrooke’s Hospital
Department of Anesthesia and Critical Care Director of Medical Studies Cambridge University Hospitals
University of Chicago; Department of Neurology Cambridge, United Kingdom
Executive Director Yale School of Medicine
American Quality Institute (AQI) New Haven, Connecticut Michel Kliot, MD
American Society of Anesthesiologists Professor of Clinical Neurosurgery
Park Ridge, Illinois Christiana E. Hall, MD, MS Department of Neurological Surgery
Associate Professor of Neurology and University of California, San Francisco School of
E. Wesley Ely, MD, MPH, FACP, FCCM Neurotherapeutics and Neurological Surgery Medicine;
Professor of Medicine and Critical Care Division of Neurocritical Care Director, Center for Management and Surgery of
Department of Medicine University of Texas Southwestern Peripheral Nerve Disorders
Center for Health Services Research Dallas, Texas San Francisco, California
Vanderbilt University School of Medicine;
Associate Director of Aging Research, VA J. Claude Hemphill III, MD, MAS W. Andrew Kofke, MD, MBA, FCCM
GRECC Professor of Clinical Neurology Professor
Veterans Affairs Tennessee Valley Healthcare Department of Neurology Departments of Anesthesiology and Critical
System University of California, San Francisco School of Care and Neurosurgery
Nashville, Tennessee Medicine; Perelman School of Medicine at the University
Director, Neurocritical Care of Pennsylvania;
Ronald G. Emerson, MD Department of Neurology Director, Neuroanesthesia
Adjunct Professor of Clinical Neurology San Francisco General Hospital Department of Anesthesiology and Critical Care
Department of Neurology San Francisco, California University of Pennsylvania Health System;
Columbia University Medical Center; Co-Director, Neurointensive Care Unit
Attending Neurologist Jiri Horak, MD Hospital of the University of Pennsylvania
Department of Neurology Assistant Professor of Clinical Anesthesiology Philadelphia, Pennsylvania
Hospital for Special Surgery and Critical Care
New York, New York Department of Anesthesiology and Critical Care Jaroslaw Krejza, MD, PhD
Perelman School of Medicine at the University Research Associate Professor
Per Enblad, MD, PhD of Pennsylvania Department of Radiology
Professor of Neurosurgery Philadelphia, Pennsylvania Perelman School of Medicine at the University
Department of Neuroscience of Pennsylvania
Section of Neurosurgery Peter Horn, MD Philadelphia, Pennsylvania;
Uppsala University Associate Professor of Neurosurgery Al-Imam Muhammad Ibn Saud Islamic
Uppsala University Hospital Department of Neurosurgery University
Uppsala, Sweden Charité Universitätsmedizin Berlin Riyadh, Saudi Arabia
Berlin, Germany
Anthony A. Figaji, MD, FCS, PhD Monisha A. Kumar, MD
Professor David A. Horowitz, MD Assistant Professor
Division of Neurosurgery Assistant Professor of Clinical Medicine Departments of Neurology, Neurosurgery, and
University of Cape Town; Perelman School of Medicine at the University Anesthesiology and Critical Care
Head of Pediatric Neurosurgery of Pennsylvania; Perelman School of Medicine at the University
Red Cross Children’s Hospital Associate Chief Medical Officer of Pennsylvania;
Cape Town, Western Cape, South Africa University of Pennsylvania Health System Director, Neurocritical Care Fellowship Program
Philadelphia, Pennsylvania Hospital of the University of Pennsylvania
Damien Galanaud, MD, PhD Philadelphia, Pennsylvania
CNRS UPR 640 LENA
Université Pierre et Marie Curie (Paris VI);
Neuroradiology
Pitié Salêtrière Hospital
Paris, France
Contributors xi

Arthur M. Lam, MD, FRCPC David K. Menon, MD, PhD, FRCP, FRCA, DaiWai M. Olson, PhD, RN
Medical Director of Neuroanesthesia and FFICM, FMedSci Assistant Professor
Neurocritical Care Professor and Head Department of Medicine
Swedish Neuroscience Institute Division of Anaesthesia Division of Neurology
Swedish Medical Center; University of Cambridge/Addenbrooke’s Duke University School of Medicine
Clinical Professor of Anesthesiology and Pain Hospital; Durham, North Carolina
Medicine Honorary Consultant
University of Washington Perioperative Care Pratik P. Pandharipande, MD, MSCI
Member, Physician Anesthesia Services Addenbrooke’s Hospital; Associate Professor
Seattle, Washington Co-Chair, Acute Brain Injury Program Department of Anesthesiology
Wolfson Brain Imaging Centre Division of Critical Care and Perioperative
Peter D. le Roux, MD, FACS University of Cambridge Medicine
Associate Professor of Neurosurgery Cambridge, United Kingdom Vanderbilt University School of Medicine
Perelman School of Medicine at the University Nashville, Tennessee
of Pennsylvania; Asako Miyakoshi, MD
Department of Neurosurgery Assistant Professor Jose L. Pascual, MD, PhD, FRCS(C), FACS
Pennsylvania Hospital Department of Radiology Assistant Professor
Philadelphia, Pennsylvania Division of Neuroradiology Department of Surgery
University of Washington School of Medicine Division of Traumatology, Surgical Critical Care,
Joshua M. Levine, MD Seattle, Washington and Emergency Surgery
Assistant Professor Perelman School of Medicine at the University
Departments of Neurology, Neurosurgery, and Richard S. Moberg, MSE of Pennsylvania;
Anesthesiology and Critical Care President Attending Surgeon
Perelman School of Medicine at the University Moberg Research, Inc. Department of Surgery
of Pennsylvania; Ambler, Pennsylvania Hospital of the University of Pennsylvania
Co-Director, Neurointensive Care Unit Philadelphia, Philadelphia
Hospital of the University of Pennsylvania Pierre D. Mourad, PhD
Philadelphia, Pennsylvania Associate Professor Aashish R. Patel, DO
Department of Neurological Surgery Fellow in Neurocritical Care
Geoffrey T. Manley, MD, PhD Principal Physicist Department of Neurological Surgery
Professor and Vice Chairman Applied Physics Laboratory University of Texas Southwestern
Co-Director, Brain and Spinal Injury Center University of Washington Dallas, Texas
Department of Neurological Surgery Seattle, Washington
University of California, San Francisco School of Frederik A. Pennings, MD, PhD
Medicine; Barnett R. Nathan, MD Assistant Professor
Chief of Neurosurgery Associate Professor Department of Neurosurgery
San Francisco General Hospital Departments of Neurology and Internal University of Massachusetts Medical School
San Francisco, California Medicine Worcester, Massachusetts
University of Virginia
Basil F. Matta, MB, MA, BCh, FRCA Charlottesville, Virginia Ian Piper, PhD
Associate Lecturer Clinical Scientist
Department of Medicine Patrick J. Neligan, MD Clinical Physics
Division of Anaesthesia Senior Clinical Lecturer of Anaesthesia and University of Glasgow;
University of Cambridge/Addenbrooke’s Intensive Care Brain-IT Group Coordinator, Intensive Care
Hospital; Galway University Hospitals Monitoring
Divisional Director and Associate Medical National University of Ireland, Galway Department of Clinical Physics
Director Galway, Ireland Southern General Hospital Trust
Emergency and Perioperative Care Glasgow, United Kingdom
Cambridge University Foundation Trust Anoma Nellore, MD
Hospitals Fellow Amit Prakash, MBBS, MD, FRCA, EDIC
Cambridge, United Kingdom Department of Medicine Consultant, Department of Anaesthesia and
Division of Infectious Disease Intensive Care
Jonathan McEwen, MD Massachusetts General Hospital Addenbrooke’s Hospital
Clinical Assistant Professor of Anesthesiology Harvard Medical School Cambridge University Hospital Foundation
Department of Anesthesiology, Pharmacology, Boston, Massachusetts Trust
and Therapeutics Cambridge, United Kingdom
University of British Columbia Faculty of Mauro Oddo, MD
Medicine; Staff Physician J. Javier Provencio, MD, FCCM
Attending Anesthesiologist, Vancouver Acute Department of Intensive Care Medicine Associate Professor
Department of Anesthesia, Division of CHUV-University Hospital Departments of Neurology, Neurological
Neuroanesthesia Faculty of Biology and Medicine University of Surgery, and Neurosciences
Vancouver General Hospital Lausanne Cleveland Clinic Lerner College of Medicine of
Vancouver, British Columbia, Canada Lausanne University Hospital Case Western Reserve University;
Lausanne, Switzerland Director, Neurocritical Care Fellowship Program
Cleveland Clinic
Cleveland, Ohio
xii Contributors

Louis Puybasset, MD, PhD J. Michael Schmidt, PhD, MSc Martin Smith, MBBS, FRCA, FFICM
Département d’Anesthésie-Réanimation Assistant Professor of Clinical Neurophysiology Consultant and Honorary Professor in
Université Pierre et Marie Curie (Paris VI); in Neurology Neurocritical Care
Neurosurgical Intensive Care Unit Department of Neurology The National Hospital for Neurology and
Pitié Salêtrière Hospital Columbia University College of Physicians and Neurosurgery
Paris, France Surgeons; University College London Hospitals
Director London, United Kingdom
Rohan Ramakrishna, MD Neuro-ICU Neuromonitoring and Informatics
Resident Columbia University Medical Center Marco D. Sorani, PhD
Department of Neurological Surgery New York, New York Adjunct Assistant Professor
University of Washington Medical Center Department of Neurological Surgery
Seattle, Washington Sarah E. Schmitt, MD University of California, San Francisco
Assistant Professor of Clinical Neurology San Francisco, California
Mahbub Rashid, PhD Department of Neurology
Professor Perelman School of Medicine at the University Alejandro M. Spiotta, MD
University of Kansas School of Architecture, of Pennsylvania; Resident
Design and Planning Director, Electroencephalography Laboratory Department of Neurosurgery
Lawrence, Kansas Department of Neurology Cleveland Clinic
Hospital of the University of Pennsylvania Cleveland, Ohio
Gerald P. Roston, PhD, PE Philadelphia, Pennsylvania
Technology Consultant and Managing Partner John J. Stern, MD
Pair of Docs Consulting, LLC Patricia D. Scripko, MD Clinical Professor
Saline, Michigan Resident Department of Medicine
Department of Neurology Perelman School of Medicine at the University
Stuart Russell, PhD Massachusetts General Hospital of Pennsylvania;
Professor of Computer Science Brigham and Women’s Hospital Chief, Division of Infectious Diseases
Michael H. Smith and Lotfi A. Zadeh Chair in Boston, Massachusetts Department of Medicine
Engineering Pennsylvania Hospital
Computer Science Division †John M. Sewell, BSEE Philadelphia, Pennsylvania
University of California, Berkeley Chief Engineer
Berkeley, California; Active Signal Technologies, Inc. Nino Stocchetti, MD
Adjunct Professor Linthicum, Maryland Professor of Anesthesia and Intensive Care
Department of Neurological Surgery Terapia Intensiva Neuroscienze
University of California, San Francisco Robert G. Siman, PhD Fondazione IRCCS Cà Granda
San Francisco, California Research Professor University of Milan
Department of Neurosurgery Milan, Italy
Owen B. Samuels, MD Perelman School of Medicine at the University
Associate Professor of Neurosurgery and of Pennsylvania Jose I. Suarez, MD
Neurology Philadelphia, Pennsylvania Professor
Emory University School of Medicine; Departments of Neurology and Neurosurgery
Director, Division of Neuroscience Critical Care Carrie A. Sims, MD, FACS Baylor College of Medicine;
Emory Healthcare Assistant Professor Director
Atlanta, Georgia Department of Surgery Vascular Neurology and Neurocritical Care
Division of Traumatology, Surgical Critical Care, Baylor College of Medicine
Matthew R. Sanborn, MD and Emergency Surgery Houston, Texas
Resident Perelman School of Medicine at the University
Department of Neurosurgery of Pennsylvania Farzana Tariq, MD
Perelman School of Medicine at the University Philadelphia, Pennsylvania Cerebrovascular and Skull Base Fellow
of Pennsylvania Department of Neurological Surgery
Philadelphia, Pennsylvania Richard O. Sinnott, PhD University of Washington
Director, eResearch Seattle, Washington
Bernhard Schmidt, PhD University of Melbourne
Department of Neurology Melbourne, Australia Kyla P. Terhune, MD
Chemnitz Medical Centre Assistant Professor of Surgery and
Chemnitz, Germany Alan Siu Anesthesiology
Resident Division of General Surgery
Eric Albert Schmidt, MD, PhD Department of Neurological Surgery Vanderbilt University School of Medicine
Department of Neurosurgery The George Washington University Medical Nashville, Tennessee
Hôpital Purpan Center
Toulouse, France Washington, District of Columbia Brett Trimble, BSME
Director, Advanced Technology
Integra LifeSciences Corporation
†Deceased San Diego, California
Contributors xiii

David K. Vawdrey, PhD Brandon von Tobel, MD, MBE Elisa R. Zanier, MD
Assistant Professor of Clinical Biomedical Vice President of Finance and Operations Department of Neuroscience
Informatics ImaCor, Inc. Instituto Mario Negri
Department of Biomedical Informatics New York, New York Milan, Italy
Columbia University College of Physicians and
Surgeons Howard Yonas, MD Craig Zimring, PhD
New York, New York Professor and Chairman Professor of Architecture and Psychology
Department of Neurosurgery Colleges of Architecture and Psychology
Paul M. Vespa, MD University of New Mexico School of Medicine Georgia Institute of Technology
Professor of Neurosurgery and Neurology Albuquerque, New Mexico Atlanta, Georgia
Department of Neurosurgery
David Geffen School of Medicine at UCLA; Brad E. Zacharia, MD
Director Resident
Neurocritical Care Department of Neurological Surgery
Ronald Reagan UCLA Medical Center Columbia University Medical Center/New
Los Angeles, California York-Presbyterian
New York, New York
Abbreviations
NOTE: Abbreviations may have more than one meaning, depending on their context.

3-H  hypertension, hemodilution, and hypervolemia ASTM  American Society for Testing and Materials
AACN  American Association of Critical Care Nurses AT  antithrombin
AAN  American Academy of Neurology ATC  automatic tube compensation
AARP  American Association of Retired Persons ATN  acute tubular necrosis
ABA  American Bar Association ATP  adenosine triphosphate
ABM  acute bacterial meningitis ATS  American Thoracic Society
ABP  arterial blood pressure AU  arbitrary units
ACA  anterior cerebral artery AV  audiovisual
ACE  angiotensin converting enzyme AVDO2  arteriovenous difference in oxygen
ACEI/ARBs  angiotensin-converting enzyme inhibitor/ AVM  arteriovenous malformation
angiotensin-receptor blocker BA  basilar artery
ACGME  Accreditation Council for Graduate Medical Education BAEP  brainstem auditory evoked potential
AChE  acetylcholinesterase BAM  brain acoustic monitor
ACCP  American College of Chest Physicians BANN  Bayesian Artificial Neural Network
ACoA  anterior communicating artery BBB  blood-brain barrier
ACS  abdominal compartment syndrome BFV  blood flow velocity
ACT  activated clotting time BG  blood glucose
ACTH  adrenocorticotrophic hormone BHI  breath holding index
ACV  assist control ventilation BIS  bispectral index
AD  axial diffusivity BMI  body mass index
ADC  apparent diffusion coefficient BMR  basal metabolic rate
ADH  antidiuretic hormone BOLD  blood oxygen level dependent
ADL  activities of daily living BOOST  Brain Oxygen and Outcome Study in Traumatic Brain Injury
ADNI  Alzheimer’s Disease Neuroimaging Database BP  blood pressure
ADP  adenosine diphosphate BPI  bactericidal permeability-increasing protein
ADQI  Acute Dialysis Quality Initiative BrainIT  brain monitoring with information technology
ADR  alpha/delta ratio BSI  bloodstream infection
ADT  admission/discharge/transfer BSM  bedside monitors
AED  antiepileptic drug BT  brain temperature
aEEG  amplitude-integrated electroencephalography BTO  balloon test occlusion
AEP  auditory evoked potentials BUN  blood urea nitrogen
AF  atrial fibrillation CA  cerebral autoregulation (Chapters 30, 46)
AG  anion gap CA  cardiac arrest (Chapter 25)
AHA/ASA  American Heart Association/American Stroke Association CA-BSI  catheter-associated bloodstream infection
AI  artificial intelligence CAD  coronary artery disease
AIS  acute ischemic stroke CAM-ICU  Confusion Assessment Method for the ICU
AKI  acute kidney injury CAP  College of American Pathologists
AKIN  Acute Kidney Injury Network CAR  cerebral arterial resistance
ALF  acute liver failure CAS  carotid angioplasty and stenting
ALFSG  Acute Liver Failure Study Group CASL  continuous arterial spin labeling
ALI  acute lung injury CBF  cerebral blood flow
AMA  American Medical Association CBFV  cerebral blood flow velocity
AMID  active implantable medical device CBV  cerebral blood volume
ANH  artificial nutrition and hydration CCAT  Computerized Cognitive Assessment Tool
ANN  artificial neural network CCO  continuous cardiac output
APACHE  Acute Physiology and Chronic Health Evaluation CCT  central conduction time
aPL  antiphospholipid antibodies Ccw  compliance of the chest wall
APN  advanced practice nurse CDC  Centers for Disease Control and Prevention
APP  abdominal perfusion pressure CDSA  color density spectral array
aPTT  activated partial thromboplastin time CEA  carotid endarterectomy
ARAS  ascending reticular activating system cEEG  continuous electroencephalography
ARC  absolute reticulocyte count CES  cholesterl emboli syndrome
ARDS  acute respiratory distress syndrome CEUs  continuing education units
ARi (or ARI)  autoregulation index CFM  cerebral function monitoring
ASA  American Society of Anesthesiology C-FMZ  C-flumazenil
aSAH  aneurysmal subarachnoid hemorrhage Cho  choline
ASIA  American Spinal Injury Association CHr  reticulocyte hemoglobin content
ASL  arterial spin labeling CI  coagulation index

xv
xvi Abbreviations

CINMA  critical illness neuromuscular abnormalities DHHS  Department of Health and Human Services
CIPM  critical illness polyneuromyopathy DI  diabetes insipidus
CIPNM  critical illness polyneuropathy and myopathy DIC  disseminated intravascular coagulation
CIRCI  critical illness related corticosteroid insufficiency DIND  delayed ischemic neurologic deficit
CK  creatine kinase DIT  drug-induced thrombocytopenia
Cl  compliance of the lung DITP  drug-induced immune thrombocytopenia
CLIA 88  Clinical Laboratory Improvements Amendments of 1988 dIVC  inferior vena cava diameter
CLAB  central line-associated bacteraemia DLCO  diffusing capacity of the lung for carbon monoxide
CLABSI  central line-associated bloodstream infection DMN  default mode network
CMAP  compound muscle action potentials DO2  oxygen delivery
CMO  comfort measures only DoD  Department of Defense
CMRO2  cerebral metabolic rate of oxygen DoE  Department of Energy
CMRGluc  cerebral metabolic rate of glucose DRG  diagnostic related group
CMS  Centers for Medicare and Medicaid Services DRS  Disability Rating Scale
CMV  cytomegalovirus DS  Down syndrome
CNS  central nervous system DSA  digital subtraction angiography
CO  cardiac output DSM  Diagnostic and Statistical Manual of Mental Disorders
CO2  carbon dioxide DSP  digital signal processing
COGIF  Consensus on Grading Intracranial Flow DTI  diffusion tensor imaging
COI  cerebral oxygenation index DUS  duplex ultrasonography
COM  communication DV  data validation
COMBI  Center for Outcome Measurement in Brain Injury DVT  deep vein thrombosis
COMPACCS  Committee on Manpower for Pulmonary and Critical DWI  diffusion-weighted imaging
Care Societies EAA  excitatory amino acids
COPD  chronic obstructive pulmonary disease EBM  evidence-based medicine
COx  cerebral oximetry index EBNP  evidence-based nursing practice
CPAP  continuous positive airway pressure EBP  evidence-based practice
CPOE  computerized order entry EC-IC  extracranial to intracranial
CPP  cerebral perfusion pressure ECA  external carotid artery
CPR  cardiopulmonary resuscitation ECCO  Essentials of Critical Care Orientation
CPSE  complex partial status epilepticus ECF  extracellular fluid
CPT  current procedural terminology ECoG  electrocorticogram
Cr  creatine ED  emergency department
CRH  corticotropin-releasing hormone EDC  extended differential count
CRM  crew/crisis resource management EDH  epidural hematoma
CRMP  collapsin response mediator protein EDM  esophageal Doppler monitor
CRP  C-reactive protein EDTA  ethylene diamine tetra acetate
CRRT  continuous renal replacement therapy EEG  electroencephalography; electroencephalogram
CRS  Coma Recovery Scale EF  ejection fraction
CRS-R  Coma Recovery Scale–Revised E-GOS  Extended Glasgow Outcome Scale
Crs  compliance of respiratory system EHR  electronic health record
CSA  cross-sectional area EIT  electrical impedance tomography
CSD  cortical spreading depression EKG  electrocardiogram
CSE  convulsive status epilepticus ELISA  enzyme-linked immunological sample assay
CSF  cerebrospinal fluid EMG  electromyogram
CSW  cerebral salt wasting EMI  electromagnetic interference
CT  computed tomography EMR  electronic medical record
CTA  computed tomography angiography EMS  emergency medical services
CTP  computed tomography perfusion (Chapters 13, 26) EN  enteral nutrition
CTP  Child-Turcotte-Pugh (Chapter 23) eNAA  extracellular N-acetyl aspartate
CTT  central conduction time EOG  electrooculogram
CTV  cerebral venous thrombosis EP  evoked potential
CVC  central venous catheter EPIC  extended prevalence of infection in intensive care
CVP  central venous pressure EPL  estimated percent lysis
CVR  cerebrovascular resistance EPO  erythropoietin
CVT  cerebral venous thrombosis EPOR  erythropoietin receptor
CVVH  continuous veno-venous hemofiltration ESA  erythropoiesis-stimulating agents
CXR  chest x-ray ESICM  European Society of Intensive Care Medicine
D  diameter of conduit ESO  European Stroke Organization
DAI  diffuse axonal injury ESRD  end-stage renal disease
dARi  dynamic autoregulation index ET  endotracheal tube
DBN  dynamic Bayesian network etCO2  end-tidal carbon dioxide
DBP  diastolic blood pressure ETF  Emerging Technology Fund
DBS  deep brain stimulator EU  European Union
DC  decompressive craniectomy EVD  external ventricular drain
DCI  delayed cerebral ischemia EVLWI  extravascular lung water index
DCS  diffuse correlation spectroscopy FA  fractional anisotropy
DHCA  deep hypothermic circulatory arrest FC  Foley catheter
Abbreviations xvii

FDA  Food and Drug Administration HSE  Herpes simplex encephalitis


FD&C  Food, Drug, and Cosmetic Act HSV  Herpes simplex virus
FDG  fluorodeoxyglucose HUS  hemolytic uremic syndrome
Fe  iron HV  hyperventilation
FENa  fractional excretion of sodium IAP  intra-abdominal pressure
FET  field effect transistor IBW  ideal body weight
FFF  family, friends, and fools ICA  internal carotid artery
FFP  fresh frozen plasma ICAMs  intracellular adhesion molecules
FFT  fast-Fourier transformation ICE  integrated clinical environment
FIM  Functional Independence Measure ICG  indocyanine green
FiO2  inspiratory oxygen fraction ICH  intracerebral hemorrhage
FLAIR  fluid-attenuated inversion recovery ICP  intracranial pressure
fMRI  functional magnetic resonance imaging ICDSC  Intensive Care Delirium Screening Checklist
FNHTR  febrile non-hemolytic transfusion reactions ICU  intensive care unit
FNN  Foundations of Neuroscience Nursing IDE  investigational device exemption
FOIA  Freedom of Information Act IDSA  Infectious Diseases Society of America
FOUR  Full Outline of UnResponsiveness IEEE  Institute of Electrical and Electronic Engineers
FRBC  fragmented RBC iEEG  intermittent electroencephalography
FTc  flow time correction IFNα  interferon-α
F/V  Flotrac-Vigileo IH  intracranial hypertension
FV  flow velocity IHD  ischemic heart disease
FVl  flow velocity, left IIT  intensive insulin therapy
FVL  Factor V Leiden IJV  internal jugular vein
FVr  flow velocity, right IL  interleukin
GAAP  generally accepted accounting practices IMZ  iomazenil
GABA  gamma-aminobutyric acid IND  investigational new drug
GAP  growth associated protein INR  international normalized ratio
GB  gigabyte IOM  Institute of Medicine
GCS  Glasgow Coma Scale IOP  intraocular pressure
GDP  gross domestic product IP  intellectual property
GFAP  glial fibrillary acidic protein IPC  intermittent pneumatic compression
GFR  glomerular filtration rate IPF  immature platelet fraction
GH  growth hormone IPS  intensive care unit physician staffing
GHBP  growth-hormone binding protein IRF  immature reticulocyte fraction
GHRP  GH-releasing peptide ISF  International Sepsis Forum
GI  gastrointestinal ISHEN  International Society for Hepatic Encephalopathy and
GMDI  Glioma Molecular Diagnostic Initiative Nitrogen Metabolism
GMP  good manufacturing practices ISO  International Organization for Standardization
GN  glomerulonephritis ISS  Injury Severity Score
GNP  gross national product IT  information technology
GOS  Glasgow Outcome Scale ITAA  Information Technology Association of America
G-PEDs  generalized periodic epileptiform discharges ITBVI  intrathoracic blood volume index
1
H-MRS  proton magnetic resonance spectroscopy IV  intravenous
HAC  hospital-acquired condition IVC  inferior vena cava
HAI  hospital-acquired infection IVIg  intravenous immunoglobulin
HAP  hospital-acquired pneumonia KIM-1  kidney injury molecular 1
HbO2  oxyhemoglobin LC  liquid chromatography
HCAP  heathcare-associated pneumonia LCD  liquid crystal display
Hct  hematocrit LDF  laser Doppler flowmetry
HE  hepatic encephalopathy LDH  lactate dehydrogenase
HELLP  hemolytic anemia, elevated liver enzymes, low platelets LDUH  low-dose unfractionated heparin
syndrome LGIB  lower gastrointestinal bleeding
Hgb  hemoglobin LGR  lactate : glucose ratio
HHb  deoxyhemoglobin LIP  lower inflection point
HHCFA  Health Care Financing Organization LMWH  low molecular weight heparin
HHNK  hyperglycemic hyperosmolar nonketotic lp(a)  lipoprotein (a)
HIE  hypoxic-ischemic encephalopathy LOC  loss of consciousness
HIPAA  Health Insurance Portability and Accountability Act LOH  Loop of Henle
HIT  heparin-induced thrombocytopenia LOI  lactate oxygen index
HiTT  high dose thrombin time LOS  length of stay
HIV  human immunodeficiency virus LP  lumbar puncture
HLA  human leukocyte antigens LPR  lactate : pyruvate ratio
HMG CoA  3-hydroxy-3-methylglutaryl coenzyme A LUS  lung ultrasound
HMS  Haemostasis Management System LV  left ventricle
HPA  hypothalamic-pituitary axis LVEDA  left ventricular end-diastolic area
HPS  human patient stimulator LVEDV  left ventricular end-diastolic volume
HR  heart rate MA  maximum amplitude
HRS  hepatorenal syndrome MAC  mid arm circumference (Chapter 14)
xviii Abbreviations

MAC  minimum alveolar concentration (Chapter 9) NiCO  noninvasive cardiac output


MAP  mean arterial pressure NICU  neurointensive care unit
MAP2  microtubule-associate protein type 2 NIH  National Institutes of Health
MB  megabyte NIHSS  National Institutes of Health Stroke Scale
MBP  myelin basic protein NINDS  National Institute of Neurological Disease and Stroke
MBs  microbubbles NIRS  near infrared spectroscopy
MCA  middle cerebral artery NMDA  N-methyl-D-aspartic
MCHC  mean corpuscular hemoglobin concentration NNIS  National Nosocomial Infections Surveillance
MCI  mild cognitive impairment NO  nitric oxide
MCMC  Monte Carlo Markov Chain NOS  nitric oxide synthase
MCS  minimally conscious state NOx  nitric oxide metabolite
MCV  mean corpuscular volume NPH  normal pressure hydrocephalus
MCVr  reticulocyte volume NPV  negative predictive value
MD  mean diffusion (Chapter 28) NRBC  nucleated red blood cell
MD  microdialysis (Chapters 36, 48) NRCPR  National Registry of Cardiopulmonary Resuscitation
MDCT  multidetector computed tomography NRI  Nutritional Risk Index
MDPnP  Medical Device Plug and Play NSAID  nonsteroidal anti-inflammatory drug
MDS  myelodysplastic syndrome NSE  neuron-specific enolase
MEE  measured energy expenditure NSF  National Science Foundation (Chapter 38)
MEMS  micro electro-mechanical system NSF  nephrogenic systemic fibrosis (Chapter 22)
MEP  motor evoked potential NTIS  nonthyroidal illness syndrome
MES  microembolic signal NTP  Network Time Protocol
MeSH  medical subject heading OAST  Optimizing Analysis of Stroke Trials
MFI  microcirculatory flow index OEF  oxygen extraction fraction
mGy  milli-Gray OGR  oxygen-glucose ratio
MI  Maastricht Index (Chapter 14) ONS  optic nerve sheath
MI  myocardial infarction (Chapter 15) OPS  orthogonal polarizing spectral
MIB  Medical Information Bus OR  operating room
MICU  medical intensive care unit P4P  pay-for-performance
MIPS  maximum intensity projections PA  pulmonary artery
MMM  multi modality monitor PAC  pulmonary artery catheter
MMP  matrix metalloproteinases PaCO2  arterial carbon dioxide tension
MMPF  multimodal pressure-flow PACS  picture archiving and communication system
MMSE  Mini-Mental Status Examination PAD  pulmonary artery diastolic
MOANS  Mayo’s Older Americans Normative Studies PAGE  polyacrylamide gel electrophoresis
MOCA  Montreal Cognitive Assessment PAI  plasminogen activator inhibitor
MODS  Multiple Organ Dysfunction Syndrome Palv  alveolar pressure
MPAI  Mayo Portland Adaptability Inventory PaO2  arterial oxygen tension
MPM  Mortality Probability Model PAOP  pulmonary artery occlusion pressure
MPV  mean platelet volume PAP  pulmonary pressures
MR  magnetic resonance PAR  pressure autoregulation
MRA  magnetic resonance angiography PAS  pulmonary artery systolic
MRI  magnetic resonance imaging PASL  pulsed arterial spin labeling
MRP  magnetic resonance perfusion PAV  percent alpha variability
MRS  magnetic resonance spectroscopy PAV+  proportional assist ventilation
mRS  modified Rankin Scale Paw  airway pressures
MRSA  methicillin resistant Staphylococcus aureus PbtO2  brain tissue oxygen tension
MS  mass spectrometry PCA  posterior cerebral artery
mSv  milli-Sieverts pCAM-ICU  Pediatric Confusion Assessment Method for Intensive
MTT  mean transit time Care Unit
Mx  autoregulation PCI  percutaneous coronary interventions
NAA  N-acetyl aspartate PCM  pulse contour method
NADH  nicotinamide adenine dinucleotide PCR  polymerase chain reaction
NAG  N-acetyl-glucosaminidase PCT  proximal convoluted tubule
NBH  normobaric hyperoxia PCWP  pulmonary capillary wedge pressure
nCPPl  noninvasive cerebral perfusion pressure, left PDSA  Plan-Do-Study-Act
nCCPr  noninvasive cerebral perfusion pressure, right PE  pulmonary embolism
NCCU  neurocritical care unit PED  periodic epileptiform discharges
NCS  nonconvulsive seizures PEEP  positive end-expiratory pressure
NCSE  nonconvulsive status epilepticus PEEPi  intrinsic positive end-expiratory pressure
NEMS  nano electro-mechanical system Pes  esophageal pressure
NFH  neurofilament heavy chain PET  positron emission tomography
NFL  neurofilament light chain PF4  platelet factor 4
NFM  neurofilament middle chain PI  pulsatility index
NGAL  neutrophil gelatinase-associated lipocalin PICARD  Program to Improve Care in Acute Renal Disease
NHSN  National Healthcare Safety Network PICC  peripherally inserted central catheter
NIBP  noninvasive blood pressure PID  peri-infarct depolarizations
NICE  National Institute for Clinical Excellence PICU  pediatric intensive care unit
Abbreviations xix

PILOT  Pediatric Intensity Level of Therapy rTEG  rapid thromboelastography


PIM  Pediatric Index of Mortality rtPA  recombinant tissue plasminogen activator
Pip  peak inspiratory pressure RTS  Revised Trauma Score
Pl  transpulmonary pressure RTV silicone  room temperature vulcanization silicone
PLED  periodic lateralized epileptiform discharge RV  right ventricle
Pleth-HR  heart rate from oxygen saturation monitor SAH  subarachnoid hemorrhage
PMA  premarket approval SaO2  arterial oxygen saturation
PN  parenteral nutrition SAPS  Simplified Acute Physiology Score
PO2  partial pressure of oxygen SAS  Riker Sedation-Agitation Scale
POC  point of care SATs  spontaneous awakening trial
PoCoA  posterior communicating artery SBDP  spectrin breakdown degradation product
POCT  point of care testing SBI  secondary brain insults
Pplat  plateau pressure SBIR  Small Business Innovative Research
PPV  pulse pressure variation SBP  systolic blood pressure
PRAM  pressure recording analytical method SBP 120  spectrin breakdown products
PRBC  packed red blood cell SBTs  spontaneous breathing trials
PRESS  posterior reversible leukoencephalopathy syndrome SC  serum creatinine
PRx  cerebrovascular pressure reactivity index SCAT  Standardized Concussion Assessment Tool
PSV  pressure support ventilation SCCM  Society of Critical Care Medicine
PT  prothrombin time SCI  spinal cord injury
PT/INR  prothrombin time/international normative ratio SCM  sternocleidomastoid muscle
PTS  Pediatric Trauma Score SCUF  slow continuous ultrafiltration
PTSD  post-traumatic stress disorder ScVO2  oxygen saturation in superior vena cava (Chapters 19, 40)
PTT  partial thromboplastin time ScvO2  mixed venous oxygen saturation (Chapter 23)
PVD  patient-ventilator dyssynchrony SDE  subdural empyema
PVI  pressure-volume index SDF  sidestream dark field
PvO2  oxygen partial pressure in venous blood SDH  subdural hematoma
PVS  persistent vegetative state SE  status epilepticus
PWI  perfusion-weighted imaging SF  short form
QFD  Quality Functional Deployment SIADH  syndrome of inappropriate antidiuretic hormone
QI  quality initiatives sICH  spontaneous intracerebral hemorrhage
qMRA  quantitative magnetic resonance angiography SIMV  Synchronized Intermittent Mechanical Ventilation
QODD  quality of dying and death SIP  Sickness Impact Profile
QOL  quality of life SIRS  systemic inflammatory response syndrome
QUASAR  quantitative STAR labeling of arterial regions SjvO2  jugular venous oxygen saturation
RA  right atrium SLE  systemic lupus erythematosus
RAAS  renin-angiotensin-II-aldosterone system SLT  scanning laser tomography
RAI  relative adrenal insufficiency SMBG  self-monitoring blood glucose
RAIDs  redundant arrays of independent disks SMR  standardized mortality ratio
RALS  remote automated laboratory system SNAP  superlattice nano wire pattern transfer
RAP  cerebrospinal compensatory reserve SOFA  Sequential Organ Failure Assessment
RASS  Richmond Agitation-Sedation Scale SPECT  single photon emission computed tomography
RBC  red blood cell SpO2  oxygen saturation
RBCT  red blood cell transfusion SR  suppression ratio
RC  reticulocyte count SSC  Surviving Sepsis Campaign
rCBF  regional cerebral blood flow SSEP  somatosensory evoked potential
RCM  radiocontrast media STC  shock trauma center
RCT  randomized clinical trial StcO2  transcranial oxygen saturation
RDW  red cell distribution width StO2  tissue oxygen saturation
REE  resting energy expenditure STTR  Small Business Technology Transfer
REG  rheoencephalography SV  stroke volume
RF  radio frequency SVC  superior vena cava
rhuEPO  recombinant human erythropoietin SVM  support vector machine
Ri  inspiratory resistance SVO2  venous oxygen saturation
RN  registered nurse SVV  stroke volume variability
ROI  regions of interest SWI  susceptibility-weighted imaging
ROSC  return of spontaneous circulation T  body temperature (Chapters 45, 48)
ROTEM  rotational thromboelastometry T  translational (Chapter 6)
RPT  robotic telepresence T3  triiodothyronine
RQ  respiratory quotient T4  thyroxine
RR  respiratory rate TBI  traumatic brain injury
RRT  renal replacement therapy Tc99m-ECD  technetium-99m-ethylcysteinate dimer
RS  Ramsay Sedation Scale Tc99m-HMPAO  technetium-99m-hexamethylpropyleneamine oxide
RSE  refractory status epilepticus TCCS  transcranial color-coded duplex sonography
rSO2  regional cerebral oxygen saturation TCD  transcranial Doppler
rSO2C  regional cerebral tissue oxygenation TcE  transcranial electrical (stimulation)
rSO2R  regional renal tissue oxygenation TcEMEP  transcranial electrical motor evoked potential
rSO2S  regional splanchnic tissue oxygenation TcMMEP  transcranial magnetic motor evoked potential
xx Abbreviations

TDF  thermal diffusion flowmetry UO  urine output


TDM  time division multiplex US  ultrasound
TEE  transesophageal echocardiography UTI  urinary tract infection
TEG  thromboelastography UUN  urine urea nitrogen
TF  tissue factor VA  vertebral artery
TH  therapeutic hypothermia VALI  ventilator-associated lung injury
THb  total hemoglobin VAP  ventilator-associated pneumonia
THR  transient hyperemic response VASO  vascular space occupancy
THx  total hemoglobin reactivity VC  venture capital (Chapter 38)
TI  thermal index VC  vital capacity (Chapter 20)
TIBC  total iron binding capacity VCAM-1  vascular cell adhesion molecule-1
TIBI  thrombolysis in brain ischemia VE  expired minute volume
TIL  Therapeutic Intensity Level vEEG  video electroencephalography
TLC  total lymphocyte count VEGF  vascular endothelial cell growth factor
TNF  tumor necrosis factor VEP  visual evoked potentials
TOI  tissue oxygen index V’I  inspiratory flow
TOR  brain tissue oxygen response VILI  ventilator-induced lung injury
tPA  tissue plasminogen activator VKA  vitamin K antagonist
TPL  transplant VMRr  vasomotor reactivity
TR  tricuspid regurgitation VO  virtual organization
TRALI  transfusion-related acute lung injury VO2  oxygen consumption
TRC  tanned red cell VPR  volume-pressure response
TRH  thyrotropin-releasing hormone VRE  vancomycin-resistant enterococcus
TRICC  transfusion requirements in critical care VS  vegetative state
TRISS  Trauma Injury Severity Score VSP  vasospasm
TS  test standard vT  tidal volume
TSF  triceps skinfold thickness VTE  venous thromboembolism
TSH  thyroid stimulating hormone VTI  velocity time integral
T-tau  total tau vWD  von Willebrand’s disease
TTE  transthoracic echocardiography vWF  von Willebrand multimers
TTP  thrombotic thrombocytopenic purpura WBC  white blood cell
TV  tidal volume WDM  wavelength division multiplex
UCH  ubiquitin C-terminal hydrolase WFNS  World Federation of Neurological Societies
UFH  unfractionated heparin WHO  World Health Organization
UGIB  upper gastrointestinal bleeding WNV  West Nile virus
UIP  upper inflection point WNVE  West Nile virus encephalitis
UMLS  Unified Medical Language System Xe-CT  xenon-enhanced computed tomography
UNa  urinary sodium concentration
I
Chapter
1  

Principles of Cerebral
Metabolism and Blood Flow
Brad E. Zacharia and E. Sander Connolly, Jr.

consequently lower in the microcirculation. This effect is


Introduction known as the Fahraeus-Lindquist effect.1,4
The brain’s survival and function depend on its ability to The most powerful factor in Poiseuille’s law that can influ-
maintain a constant supply of oxygen and energy-rich sub- ence CBF is vessel radius. For example, the maximum
strate. To accomplish this feat the complex architecture of the constriction that can be obtained by hyperventilation is
brain that accounts for approximately 2% of total body mass approximately 20% from baseline. This, however, leads to a
demands approximately 20% of the total cardiac output, and decrease in CBF of approximately 60%.5 From a practical
consumes roughly one quarter of resting total body oxygen standpoint, all of this diameter regulation takes place in the
consumption.1,2 microcirculation.
With knowledge about how much blood flows to the brain
and how much oxygen the brain extracts from this blood
Cerebral Hemodynamics (arteriovenous difference in oxygen, AVDO2), one can calcu-
An understanding of the complex cerebral circulation first late cerebral metabolic rate of oxygen (CMRO2) consumption.
requires an understanding of basic physical principles about CMRO2 = CBF × AVDO2
flow. Ohm’s law predicts that flow (Q) is proportional to the
pressure gradient between inflow and outflow (ΔP) divided by
the resistance to flow (R). Physiology of Cerebral Blood Flow
Q = ∆P / R and Cerebral Blood Volume
Analogously in the brain, cerebral perfusion pressure (CPP), Normal CBF is approximately 50 mL/100 g brain tissue/min.6
the difference between arterial inflow and venous outflow Flow is normally greater in gray matter than white matter.
pressure, represents the driving pressure for cerebral blood Under normal conditions there are critical thresholds for CBF
flow (CBF). CPP is the difference between mean arterial pres- in the brain to maintain tissue health and cellular integrity.
sure (MAP) and the pressure in the thin-walled veins.1 Venous When CBF is reduced to 25 mL/100 g/min there is electroen-
pressure changes with changes in intracranial pressure (ICP) cephalographic slowing, and at 20 mL/100 g/min loss of con-
and is typically 2 to 5 mm Hg higher than the central venous sciousness occurs. When CBF is less than 18 mL/100 g/min,
pressure. CPP, therefore, can be described as: cellular homeostasis becomes jeopardized and neurons convert
CPP = MAP − ICP to anaerobic metabolism.2,7,8 At a CBF of 10 mL/100 g/min,
membrane integrity is compromised and irreversible brain
Poiseuille’s law demonstrates that in addition to CPP (ΔP), damage is inevitable (Table 1.1). Tissue infarction, however, is
blood viscosity (η) and vessel radius (r) are key determinants related not only to CBF, but to time as well.7
of CBF (Q). Vessel length (L) is generally not measured in Many different factors are postulated to maintain adequate
physiologic systems. CBF, but it is believed that local metabolic factors are of
primary importance. Under normal circumstances, in areas of
Q = (πr 4 ∆P)/ 8 η L)
increased cerebral activity, vasoactive substances are released,
Viscosity, the internal friction of blood flow, is frequently which alters vascular tone and local perfusion. The compen-
overlooked because direct measurement is difficult. Impor- satory increase in perfusion then creates a local washout
tantly, however, viscosity is felt to vary directly with changes effect, which leads to a reduction in perfusion. Key local
in hematocrit and any other process that alters blood’s cellular metabolites include, but are not limited to, carbon dioxide
composition. Blood viscosity also varies inversely with vessel (CO2), potassium, adenosine, nitric oxide, histamine, and
diameter. This is a consequence of the increased velocity gra- prostaglandins.2
dient of laminar flow as vessel size decreases, a parameter Cerebral blood volume (CBV) is determined by CBF and
known as the shear rate.3 Thus for a given blood velocity, shear capacitance vessel diameter. CBV thus increases with vasodila-
rates are greater in smaller vessels and apparent viscosity is tion and decreases with vasoconstriction. The relationship
2 © Copyright 2013 Elsevier Inc. All rights reserved.
Section I—Background 3

Table 1.1  Cerebral Blood Flow Thresholds


Cerebral Blood Flow (mL/100 g/min) Threshold Consequences
40-60 — Normal
20-30 Neurologic function Start of neurologic symptoms
Altered mental status
16-20 Electrical failure Isoelectric electroencephalogram
Loss of evoked potentials
10-12 Ionic pump failure Na+ and K+ pump failure
Cytotoxic edema
<10 Metabolic failure Complete metabolic failure with gross
disturbance of cellular energy homeostasis

From Doberstein C, Martin NA. Cerebral blood flow in clinical neurosurgery. In Youmans R, editor. Neurological surgery: a comprehensive reference guide to the
diagnosis and management of neurosurgical problems. Philadelphia: WB Saunders; 1996. p. 521, Table 21-4.

between CBF and CBV, however, is complex and under both Vasodilatory Vasoconstrictory
cascade cascade
normal and pathologic situations these variables may be
inversely related. Furthermore, blood volume is not equally Autoregulation
distributed throughout the brain, with greater volume per Passive Zone of normal breakthrough
collapse autoregulation zone
unit weight in gray matter than white matter. The central
volume principle relates the volume that intravascular blood Cerebral blood flow
occupies within the brain (CBV in mL) and the volume of VASCULAR CALIBER
75 60
(mL/100 g/min)
blood, which moves through the brain per unit time (CBF in

ICP (mmHg)
mL/min).9 50 40
CBF = CBV /τ
25 20
This equation implies that a change in vascular diameter will
directly affect CBV, but not necessarily CBF if mean transit
time (τ) is simultaneously altered. Normally, increases in CBV 25 50 75 100 125 150 175
are handled by the vasculature in two ways: (1) restricted Cerebral perfusion pressure (mm Hg)
inflow via constriction of the major feeding arteries and (2)
increased venous outflow. On the other hand, dog studies have Fig. 1.1  Relationship between extremes of cerebral perfusion pressure
shown that within the autoregulatory range, increased CBV (CPP) and intracranial pressure (ICP) in states of normal (purple line) and
reduced intracranial compliance (blue line). In the vasodilatory cascade
and mean transit time maintain CBF constancy.10 In healthy
zone, CPP insufficiency and intact pressure autoregulation lead to reflex
volunteers with intact autoregulation, transcranial Doppler cerebral vasodilation and increased ICP; the treatment is to raise CPP. In
studies suggest that a sudden steep decrease of arterial blood the autoregulation breakthrough zone, pressure and volume overload,
pressure results in an increase of intracranial blood volume,11 which overwhelms the brains capacity to autoregulate, leads to increased
that is to say, vasodilatation may protect the brain from a cerebral blood volume and ICP; the treatment is to lower CPP. (From Rose
decrease in CBF during a decrease in CPP. In pathologic states JA, Mayer SA. Optimizing blood pressure in neurological emergencies. Neurocrit
the relationship between CBV and CBF is more complex and Care 2004;1:289.)
may depend on the region affected, CO2 reactivity, autoregula-
tion, and blood-brain barrier permeability.12,13
the blood is increased in an attempt to maintain oxygen deliv-
ery (DO2) for metabolism and to prevent ischemia. When OEF
Cerebral Autoregulation is increased (oligemia), the brain may not be able to compen-
The normal cerebral vasculature is able to regulate its own sate for added reductions in DO2 and, if not corrected, infarc-
blood flow through metabolic or pressure regulation. In meta- tion is likely. Clinical symptoms develop once the reduction
bolic autoregulation, CBF changes proportionally with in CPP exceeds the brain’s ability to extract enough oxygen to
changes in CMRO2 demand. In pressure autoregulation14 arte- satisfy its metabolic demands. Breakthrough of the upper
rial diameter increases or decreases to actively control CBF limits of autoregulation also is accompanied by segmental
and maintain constant flow over a range of perfusion pres- dilation of arterial vessels and breakdown of the blood-brain
sures; that is to say, CBF remains constant despite altered CPP. barrier (BBB) and vascular endothelium, which ultimately
When CPP exceeds the limits of the autoregulatory plateau, contributes to cerebral edema.
cerebral resistance vessels respond passively to further changes The mechanisms responsible for CBF autoregulation are
in pressure. Thus once the limits of autoregulation are not yet clearly understood. Several mechanisms are proposed:
exceeded, CBF changes passively with increases or reductions (1) The myogenic hypothesis states that smooth muscle in
in perfusion pressure (Fig. 1.1). In an attempt to compensate resistance arteries responds directly to alterations in CPP;15
for reduced CBF, the oxygen extraction fraction (OEF) from (2) The metabolic hypothesis states that reduced CBF
4 Section I—Background

stimulates the release of vasoactive substances from the brain Intracranial Pressure and
that then stimulate the dilatation of cerebral vessels; (3) Auto-
regulation also may be modified by the sympathetic nervous
Cerebral Hemodynamics
system, which shifts both the upper and lower limits of auto- The Monro-Kellie hypothesis states that the brain, its vascular
regulation to higher perfusion pressures.1 network, and cerebrospinal fluid (CSF) are contained within
a rigid bony skull and membranous dura mater with a fixed
volume and so has a limited potential for expansion. ICP
Carbon Dioxide Reactivity reflects the volume of the three compartments. Increases in
The cerebral vasculature is exquisitely sensitive to changes in any one component are compensated for by removal of an
CO2, a phenomenon known as CO2 reactivity (Fig. 1.2). With equivalent amount of another; if not ICP will increase. The
hyperventilation (and resultant hypocapnea) cerebral vaso- adult intracranial space is composed of approximately 87%
constriction ensues and CBF decreases, whereas with hyper- brain, 9% CSF, and 4% blood.29 These compartments allow
capnea the reverse occurs.16 The effect can be profound with fluid and chemical shifts under normal as well as pathologic
an increase or decrease in CBF of approximately 4% for a conditions (Fig. 1.3). Normally, no net change occurs in brain
corresponding change in PaCO2 of 1 mm Hg.17 Autoregula- water or sodium content because the influx into the brain
tion is fundamentally different from CO2 reactivity. In both from the blood vessels is matched closely by extracellular bulk
metabolic and pressure autoregulation, vessel diameter flow into the CSF.30
changes are compensatory responses to maintain a constant An increase in ICP can affect neurologic function through
AVDO2. In CO2 reactivity the diameter changes are primary its effects on cerebral blood flow.31-33 However, there also are
and not related to metabolic needs such that CBF and AVDO2 circumstances in which increased flow (hyperemia) can cause
follow passively.2 increased ICP. The pressures in arteries of the subarachnoid
space are estimated to be about the same as the systemic arte-
rial pressures.1 Normally cerebral venous pressure is approxi-
Cerebral Oxygen Tension mately the same as ICP because the pressure is transmitted
There are many factors that influence brain oxygen tension through the subarachnoid space to compliant veins. However,
(PbtO2). Under physiologic conditions, PbtO2 has a close rela- when the ICP is pathologically elevated, it can become dissoci-
tionship with arterial oxygen tension (PaO2). In acute brain ated from CVP. CBV is a consequence of arterial inflow,
injury PbtO2 appears to depend on the interaction between venous drainage, and cerebrovascular tone. Cerebral vasodila-
plasma oxygen tension and CBF: PbtO2 = CBF × AVTO2.18 tion increases cerebral blood volume, whereas vasoconstric-
Because oxygen is consumed in the tissue, PbtO2 can be con- tion decreases blood volume. Manipulation of these factors
sidered as a continuum that can vary from approximately can be taken advantage of to treat elevated ICP, but to be used
90 mm Hg close to the arteriolar side of capillaries to approxi-
mately 35 mm Hg in the more distal regions.19 Reductions of
partial pressure of arterial PaO2 can alter CBF to compensate
for reduced oxygen delivery. This may depend also in part on Elastance
hemoglobin concentration among other factors but once PaO2 Low High
is less than 65 mm Hg the ability to perform complex tasks is
C
impaired in humans. Loss of consciousness occurs when PaO2
is equal to or less than 30 mm Hg.20 Observational clinical
studies and microdialysis studies suggest that cerebral infarc-

High
tion may occur in a time-dependent fashion when PbtO2 is less
than 10 to 15 mm Hg.21-28

Pressure
intracranial pressure

125 PaCO2
A B
Increasing

100
Cerebral blood flow

Low
(mL/100 g/min)

Mean arterial
75 blood pressure

50 PaO2
Increasing volume
25
Fig. 1.3  Elastance curve showing the region of low intracranial pressure
and low elastance (A), low intracranial pressure and high elastance (B),
25 50 75 100 125 150 175 and high intracranial pressure and high elastance (C). Region A describes
a state of normal intracranial pressure and a “safe” amount of volume
Pressure (mm Hg)
buffering capacity. Region B includes patients with normal intracranial
Fig. 1.2  Comparison of the response of cerebral blood flow to mean pressure with a dangerously low amount of volume reserve. Region C is
arterial blood pressure, arterial oxygen tension (PaO2), and arterial carbon clearly abnormal and readily diagnosed by the level of intracranial
dioxide tension (PaCO2). ICP, Intracranial pressure. (From Lee KR, Hoff JT. pressure. (From Lee KR, Hoff JT. Intracranial pressure. In: Youmans JR, editor.
Intracranial pressure. In: Youmans JR, editor. Neurological surgery: a comprehensive Neurological surgery: a comprehensive reference guide to the diagnosis and
reference guide to the diagnosis and management of neurosurgical problems. management of neurosurgical problems. Philadelphia: WB Saunders; 1996. p. 503,
Philadelphia: WB Saunders; 1996. p. 510, Fig. 20-14.) Fig. 20-8.)
Section I—Background 5

correctly requires knowledge of autoregulation, and the effect oxidation, and the citric acid cycle. The electrons are donated
may depend in part on the autoregulatory state.34 to molecular oxygen, resulting in a large amount of free
CBV can vary between 13 and 51 mL in the microcircula- energy, which is used to generate ATP.45
tion.2 The pressure volume index (PVI) can be used to
understand what these volume differences mean to ICP (see
also Chapter 34). The PVI is defined as the fluid volume Coupling of Cerebral Metabolism
change (in mL) in the intracranial space that would produce
a 10-fold increase in ICP [PVI = V/loge(Po/Pm)].35 Normal
and Blood Flow
PVI is 20 to 25 mL and reflects intracranial elastance.36 Because There is a relationship between cerebral metabolism and CBF;
maximal constriction can reduce blood volume by almost that is to say, blood flow is determined by regional energy
40 mL, this equation helps explain how going from normo- demands. Strong evidence of tight coupling between local
capnea to hypocapnea can reduce ICP.2,37 This may not, metabolism and flow has been obtained with in vivo radioac-
however, always be desired because the effect of vasoconstric- tive deoxyglucose methods.46,47 Many local metabolites have
tion on blood vessel diameter is raised to the fourth power been implicated in this coupling. Among them, adenosine
(Poiseuille’s equation) and so can diminish CBF.7 appears to provide a consistent link between flow and metabo-
lism.48 This substance is a potent cerebral vasodilator and a
product of dephosphorylation of adenosine monophosphate
Cerebral Metabolism that accumulates during increased neuronal activity. This
The brain only comprises 2% to 3% of the total body weight, “metabolic theory” of coupling has been challenged,49 and it
yet uses up to 20% of energy generated. Approximately 50% is proposed that there is an intrinsic neural system capable of
of the energy produced by the brain is for synaptic activity, influencing CBF independent of changes in metabolism. Evi-
25% is used for restoring ionic gradients across the cell mem- dence for this hypothesis comes from examples of dispropor-
brane, and the remaining energy is spent on biosynthesis.2 The tionate increases in flow compared to metabolism during
majority of energy is consumed by neurons. Although glial manipulation of cerebellar and brainstem centers.49,50 It seems
cells account for almost half of the brain volume, they have a reasonable that many neurotransmitters can overcome meta-
much lower metabolic rate and account for less than 10% of bolic regulation and exert a primary effect on blood flow, or
total cerebral energy consumption.38 The brain is metaboli- can directly influence metabolism and regulate blood flow in
cally unique in its use of fuel relative to other organs because kind.49-51
it lacks the ability to store fuel and thus requires a constant
supply of energy metabolites.39 Brain energy metabolism in
humans with acute brain injury is complex.40-43 Although Cerebral Hemodynamic
glucose is the major substrate, a large body of evidence sug- and Metabolic Response
gests that endogenous lactate, produced by aerobic glycolysis,
also can be an important substrate for neurons.44 Further-
to Neurologic Injury
more, there is a glucose lactate shuttle between astrocytes and Although a detailed discussion of the hemodynamic and met-
neurons. abolic responses of the brain in various pathologic conditions
is beyond the scope of this introduction, a basic understand-
ing will provide a contextual foundation upon which to
Cerebral Energy Production understand the indications for, and potential benefits of, mon-
High-energy phosphates, such as adenosine triphosphate itoring in neurocritical care.
(ATP), are the main energy substrates in the brain. Under
normal physiologic conditions, a constant supply of these sub-
strates is made possible through aerobic metabolism of Primary and Secondary Injury
glucose. In the cell, the complete oxidation of 1 mole of The pathophysiology of brain injury can be thought of in
glucose is associated with the formation of 38 moles of ATP, terms of primary and secondary events. Primary brain injury
according to the following equation: is the physical brain injury sustained at the moment of impact
in traumatic brain injury (TBI) or at the time of an insult, for
Glucose + 6 O2 + 38 ADP + 38 Pi → 6 CO2 + 44 H2O + 38 ATP
example, aneurysm rupture or reduced flow with ischemic
The above equation is a summary of many individual reac- stroke, whereas secondary injury are events that occur at any
tions. The first of these reactions, glycolysis, occurs in the later stage. Primary injuries generally are believed to be imme-
cytoplasm. Glucose is initially phosphorylated to glucose-6- diate and irreversible, but some mechanisms of cell death
phosphate and then through a series of reactions is converted from primary injury may occur over hours, suggesting the
into pyruvate. Once pyruvate has been synthesized it can possibility of reversibility.52,53 Secondary insults consist of a
either be reversibly converted to lactate or in the presence of wide range of ischemic, metabolic, and immunologic insults
oxygen, enter the citric acid cycle. The citric acid or Krebs cycle often initiated by the primary event that can occur at a lower
represents a cyclic series of reactions that occur in the mito- threshold in a susceptible brain and lead to further brain
chondria and achieve the complete oxidation of pyruvate to damage. The etiology of theses secondary events is diverse and
carbon dioxide and water. The Krebs cycle is the common can include systemic and intracranial phenomena such as
pathway for the oxidation of fuel molecules and also serves as hypotension, hypoxemia, intracranial hypertension, edema, or
a source of building blocks for biosynthesis. The electron seizures.53-56 In addition, secondary events may be iatrogenic
transport chain is the final step in the production of ATP from and cumulative. Secondary injuries of some form occur in
the NADH and FADH2 formed during glycolysis, fatty acid nearly all patients treated in a neurointensive care unit. It is
6 Section I—Background

the goal of monitoring in neurocritical care to detect poten- autoregulation can be impaired even in mild TBI, and in the
tially harmful pathophysiologic events before they cause irre- setting of normal ICP and MAP values.67 It is unknown why
versible secondary brain damage, and so allow effective autoregulatory mechanisms are so vulnerable to injury, but
treatment and improved patient outcomes.53,57-59 disturbed autoregulation may have a variable time course
that various monitoring techniques may help detect.68-70
Although pressure autoregulation commonly is disturbed
Ischemia following TBI, CO2 vasoreactivity may be preserved. Dis-
Ischemia is defined as a decrease in blood flow below the level turbed CO2 reactivity, however, is usually seen in patients
necessary to sustain normal cell structure and function. Isch- with poor neurologic status.1,68,71 Defective autoregulation is
emia can be global, as in cardiac arrest or severe oligemia as often associated with poor outcome, and in some studies
may be seen with intracranial hypertension, or focal as in focally altered autoregulation may have a greater impact on
occlusion of an intracranial vessel by embolism or thrombus. poor outcome than globally altered autoregulation.71,72
Focal ischemia does not necessarily lead to irreversible isch- Xenon blood flow studies demonstrate that cerebral isch-
emic damage in the entire area of reduced perfusion because emia occurs early after TBI and often is more severe in patients
of collateral blood flow. The central pathophysiology of cere- with intracranial hematomas and those with diffuse edema.73
bral ischemia is energy failure; that is to say, when metabolic It has been proposed that focal ischemia may be due to
demands are not satisfied, there is a rapid decrease in high- increased ICP that compresses small parenchymal arteries or
energy intermediates, and anaerobic glycolysis is stimulated. results from increased diffusion distance for oxygen from the
The loss of an adequate energy supply impairs membrane- microvasculature secondary to tissue swelling and cytotoxic
bound ionic pumps, causing a loss of ionic homeostasis and edema.32 In these situations the brain may require higher
a dramatic rise in the intracellular concentration of calcium, tissue oxygen tensions to maintain sufficient tissue oxygen-
an event of major pathophysiologic importance through its ation.19 An early reduction in CBF often is followed by a
activation of several deleterious enzyme systems and intracel- period of hyperemia; that is to say, blood flow is in excess of
lular signaling pathways.1,60 cellular needs.69,74,75 Hyperemia then can be a significant con-
Conceptually there are two distinct areas of hemodynamic tributor to increased ICP elevations following TBI in some
and metabolic functioning in focal ischemia: (1) a central patients, and can contribute to or aggravate diffuse cerebral
densely ischemic core destined to die, and (2) a collateral edema (i.e., there can be two CBF patterns associated with
area composed of viable cells at risk of ischemic damage, ICP: oligemic or hyperemic).1
known as the penumbra.60,61 Blood flow within the penum-
bra lies in a range that is between the threshold for energy
failure and complete infarction. The ultimate survival of Aneurysmal Subarachnoid Hemorrhage
penumbral cells depends on restoration of adequate blood Immediately following aneurysm rupture, global reductions
flow, which leads to the concept of a flow-dependent thera- in CBF and even circulatory arrest can be observed, often
peutic time window.1 proportional to the clinical severity of the SAH.76,77 Patients in
Ischemia commonly impairs the normal regulatory poor neurologic condition frequently develop intracranial
responses of the cerebral circulation and can lead to a state of hypertension and vasospasm. The relationship between clini-
vasomotor paralysis, which is manifest by a completely passive cal grade and CBF generally remains even in the absence of
blood flow response to changes in perfusion pressure. A major increased ICP.1 It is possible that the initial flow reduction is
factor thought to impair autoregulatory mechanisms in isch- related to direct metabolic depression from blood or its
emia is reduced tissue pH secondary to lactate accumulation. by-products.78,79 Associated with a decrease in CBF, many
Positron emission tomography studies measure cerebral patients have a reduction in CMRO2 and CBV that can reduce
hemodynamics and have provided valuable information about cerebral perfusion.37,78,80-83 There often is a progressive decrease
the events that take place during ischemia. The first alteration in CBF during the first 14 days after hemorrhage, after which
is a decrease in the ratio of CBF to CBV secondary to mainte- it usually increases.84 Cerebral infarction is one of the princi-
nance of blood flow through autoregulatory vasodilation. pal causes of death and disability in patients who survive the
When this mechanism is exhausted, oxygen extraction increases initial hemorrhage and have their aneurysm secured.84,85 Arte-
in hypoperfused areas. If this increased oxygen extraction rial vasospasm is considered the most common cause of isch-
cannot maintain an adequate oxygen supply, then CMRO2 emia; particularly delayed cerebral ischemia following SAH
declines and the threshold for an infarct will be reached.1,62 In and its severity is associated with the development of ischemic
part the goal of monitoring is to help define the penumbra or deficits. However, there are many other causes for delayed
detect pathologic changes while they still are reversible. infarction including intracranial hypertension, cerebral
edema, microthrombosis, and systemic hypotension among
others. Both pressure and CO2 autoregulation are impaired
Traumatic Brain Injury after SAH, particularly in the acute period. Disturbances in
TBI commonly is associated with disturbed cerebral hemo- autoregulation are worse with poor clinical grade and the
dynamics, which is particularly detrimental given the vulner- occurrence of vasospasm.
ability of injured brain to secondary insults.1,63,64 Secondary
cerebral ischemia is very common after severe TBI, and can
be associated with an unfavorable outcome.65,66 However, not
Conclusion
all secondary injury after TBI is ischemic in nature. Follow- A thorough understanding of the basic principles that govern
ing trauma, autoregulatory mechanisms can fail and lead cerebral blood flow and metabolism is essential for clinicians
to a detrimental uncoupling of CBF and CMRO2. Cerebral who care for patients with critical neurologic disease. It is only
Section I—Background 7

when equipped with this knowledge that we can fully take 24. Valadka AB, Hlatky R, Furuya Y, et al. Brain tissue PO2: correlation with
cerebral blood flow. Acta Neurochir Suppl 2002;81:299–301.
advantage of neuromonitoring to attenuate secondary pro-
25. Maas AI, Fleckenstein W, de Jong DA, et al. Monitoring cerebral oxygenation:
cesses leading to worse brain damage. It further stands to experimental studies and preliminary clinical results of continuous
reason that information learned from multimodality neuro- monitoring of cerebrospinal fluid and brain tissue oxygen tension. Acta
monitoring may alter and enhance our understanding of the Neurochir Suppl (Wien) 1993;59:50–7.
fundamental mechanisms of cerebral hemodynamic and met- 26. Hlatky R, Valadka AB, Goodman JC, et al. Patterns of energy substrates
during ischemia measured in the brain by microdialysis. J Neurotrauma
abolic regulation. 2004;21(7):894–906.
27. Hlatky R, Valadka AB, Goodman JC, et al. Evolution of brain tissue injury
References after evacuation of acute traumatic subdural hematomas. Neurosurgery
2007;61(1 Suppl):249–54; discussion 254–5.
1. Dobserstein C, Martin NA. Cerebral blood flow in clinical neurosurgery. In: 28. Doppenberg EM, Zauner A, Watson JC, et al. Determination of the ischemic
Winn RH, editor. Youmans neurological surgery. 5th ed. Philadelphia: WB threshold for brain oxygen tension. Acta Neurochir Suppl 1998;71:166–9.
Saunders; 2004. pp. 519–69. 29. Plum F, Posner JB. The diagnosis of stupor and coma. Philadelphia: FA
2. Verweij BH, Amelink GJ, Muizelaar JP. Current concepts of cerebral oxygen Davis; 1980.
transport and energy metabolism after severe traumatic brain injury. Prog 30. Cserr HF, Knopf PM. Cervical lymphatics, the blood-brain barrier and the
Brain Res 2007;161:111–24. immunoreactivity of the brain: a new view. Immunol Today 1992;13:507–12.
3. Chien S. Shear dependence of effective cell volume as a determinant of 31. Lee KR, Hoff JT. Intracranial pressure. In: Youmans JR, editor. Neurological
blood viscosity. Science 1970;168:977–9. surgery: a comprehensive reference guide to the diagnosis and management
4. Albrecht KH, Gaehtgens P, Pries A, et al. The Fahraeus effect in narrow of neurosurgical problems. 4th ed. Philadelphia: WB Saunders; 1996.
capillaries (i.d. 3.3 to 11.0 micron). Microvasc Res 1979;18:33–47. 32. Lindenberg R. Compression of the brain arteries as pathogenetic factor for
5. Kontos HA, Raper AJ, Patterson JL. Analysis of vasoactivity of local pH, tissue necroses and their areas of predilection. J Neuropathol Exper Neurol
PaCO2 and bicarbonate on pial vessels. Stroke 1977;8:358–60. 1955;14:223–43.
6. Sokoloff L. Metabolism of the central nervous system in vivo. In: Field J, 33. Miller J. The pathophysiology of raised intracranial pressure. In: Greenfield
Magoun HW, Hall VE, editors. Handbook of physiology. Washington, DC: JG, Adams JH, Corsellis JAN, et al, editors. Greenfield’s neuropathology.
American Physiological Society; 1960. pp. 1843–64. London: Edward Arnold; 1984. pp. 53–84.
7. Jones TH, Morawetz RB, Crowell RM, et al. Thresholds of focal cerebral 34. Huseby JS, Pavlin EG, Butler J. Effect of positive end-expiratory pressure on
ischemia in awake monkeys. J Neurosurg 1981;54:773–82. intracranial pressure in dogs. J Appl Physiol 1978;44(1):25–7.
8. Schroder ML, Muizelaar JP, Kuta AJ, et al. Thresholds for cerebral ischemia 35. Marmarou A, Shulman K, Rosende RM. A nonlinear analysis of the
after severe head injury: relationship with late CT findings and outcome. cerebrospinal fluid system and intracranial pressure dynamics. J Neurosurg
J Neurotrauma 1996;13:17–23. 1978;48:332–44.
9. Celsis P, Chan M, Marc-Vergnes JP, et al. Measurement of cerebral 36. Shapiro K, Marmarou A, Shulman K. Characterization of clinical CSF
circulation time in man. Eur J Nucl Med 1985;10:426–31. dynamics and neural axis compliance using the pressure-volume index. I.
10. Ferrari M, Wilson DA, Hanley DF, et al. Effects of graded hypotension on The normal pressure-volume index. Ann Neurol 1980;7:508–14.
cerebral blood flow, blood volume, and mean transit time in dogs. Am J 37. Bouma GJ, Muizelaar JP, Bandoh K, et al. Blood pressure and intracranial
Physiol 1992;262(6 Pt 2):H1908–14. pressure-volume dynamics in severe head injury: relationship with cerebral
11. Rosengarten B, Rüskes D, Mendes I, et al. A sudden arterial blood pressure blood flow. J Neurosurg 1992;77:15–9.
decrease is compensated by an increase in intracranial blood volume. 38. Siesjo BK. Cerebral circulation and metabolism. J Neurosurg
J Neurol 2002;249(5):538–41. 1984;60:883–908.
12. Singer OC, de Rochemont Rdu M, Foerch C, et al. Relation between relative 39. Guyton A. Textbook of medical physiology. Philadelphia: WB Saunders;
cerebral blood flow, relative cerebral blood volume, and mean transit time in 1986.
patients with acute ischemic stroke determined by perfusion-weighted MRI. 40. Vespa P, Bergsneider M, Hattori N, et al. Metabolic crisis without brain
J Cereb Blood Flow Metab 2003;23(5):605–11. ischemia is common after traumatic brain injury: a combined microdialysis
13. Nordström CH. Physiological and biochemical principles underlying and positron emission tomography study. J Cereb Blood Flow Metab
volume-targeted therapy—the “Lund concept.” Neurocrit Care 2005;25(6):763–74.
2005;2(1):83–95. 41. Pellerin L. How astrocytes feed hungry neurons. Mol Neurobiol
14. Muizelaar JP, Lutz HA, 3rd, Becker DP. Effect of mannitol on ICP and CBF 2005;32(1):59–72.
and correlation with pressure autoregulation in severely head-injured 42. Schurr A, Miller JJ, Payne RS, et al. An increase in lactate output by brain
patients. J Neurosurg 1984;61:700–6. tissue serves to meet the energy needs of glutamate-activated neurons.
15. Mitagvaria NP. Regulation of local cerebral blood flow. Adv Exp Med Biol J Neurosci 1999;19(1):34–9.
1984;180:861–79. 43. Pellerin L, Magistretti PJ. How to balance the brain energy budget while
16. Muizelaar JP, Marmarou A, Ward JD, et al. Adverse effects of prolonged spending glucose differently. J Physiol 2003;546(Pt 2):325.
hyperventilation in patients with severe head injury: a randomized clinical 44. Gallagher CN, Carpenter KL, Grice P, et al. The human brain utilizes lactate
trial. J Neurosurg 1991;75:731–9. via the tricarboxylic acid cycle: a 13C-labelled microdialysis and high-
17. Rangel-Castilla L, Gasco J, Nauta HJ, et al. Cerebral pressure autoregulation resolution nuclear magnetic resonance study. Brain 2009;132(Pt 10):2839–
in traumatic brain injury. Neurosurg Focus 2008;25:E7. 49. Epub 2009 Aug 20.
18. Rosenthal G, Hemphill JC III, Sorani M, et al. Brain tissue oxygen tension is 45. Pasqualin A. Cerebral metabolism. In: Youmans JR, editor. Neurological
more indicative of oxygen diffusion than oxygen delivery and metabolism in surgery: a comprehensive guide to the diagnosis and management of
patients with traumatic brain injury. Crit Care Med 2008;36:1917–24 neurosurgical problems. 4th ed. Philadelphia: WB Saunders; 1996.
19. Zauner A, Daugherty WP, Bullock MR, et al. Brain oxygenation and energy 46. Sokoloff L. Relationships among local functional activity, energy metabolism,
metabolism: part I-biological function and pathophysiology. Neurosurgery and blood flow in the central nervous system. Fed Proc 1981;40:2311–6.
2002;51:289–301; discussion 2. 47. Sokoloff L. Localization of functional activity in the central nervous system
20. Siesjo B. Brain energy metabolism. Chichester, UK: Wiley; 1978. by measurement of glucose utilization with radioactive deoxyglucose.
21. Maloney-Wilensky E, Le Roux P. The physiology behind direct brain oxygen J Cereb Blood Flow Metab 1981;1:7–36.
monitors and practical aspects of their use. Childs Nerv Syst 48. Wahl M, Kuschinsky W. The dilatatory action of adenosine on pial arteries
2010;26(4):419–30. of cats and its inhibition by theophylline. Pflugers Arch 1976;362:55–9.
22. Scheufler K-M, Lehnert A, Rohrborn H-J, et al. Individual values of brain 49. Edvinsson L, MacKenzie ET, McCulloch J. Cerebral blood flow and
tissue oxygen pressure, microvascular oxygen saturation, cytochrome redox metabolism. New York: Raven Press; 1993.
level and energy metabolites in detecting critically reduced cerebral energy 50. Bonvento G, Lacombe P, Seylaz J. Effects of electrical stimulation of the
state during acute changes in global cerebral perfusion. J Neurosurg dorsal raphe nucleus on local cerebral blood flow in the rat. J Cereb Blood
Anesthesiol 2004;16:210–19 Flow Metab 1989;9:251–5.
23. Valadka AB, Goodman JC, Gopinath SP, et al. Comparison of brain tissue
oxygen tension to microdialysis-based measures of cerebral ischemia in A complete list of references for this chapter can be found online at
fatally head-injured humans. J Neurotrauma 1998;15(7):509–19. www.expertconsult.com.
Section I—Background 7.e1

References 27. Hlatky R, Valadka AB, Goodman JC, et al. Evolution of brain tissue injury
after evacuation of acute traumatic subdural hematomas. Neurosurgery
1. Dobserstein C, Martin NA. Cerebral blood flow in clinical neurosurgery. In: 2007;61(1 Suppl):249–54; discussion 254–5.
Winn RH, editor. Youmans neurological surgery. 5th ed. Philadelphia: WB 28. Doppenberg EM, Zauner A, Watson JC, et al. Determination of the ischemic
Saunders; 2004. pp. 519–69. threshold for brain oxygen tension. Acta Neurochir Suppl 1998;71:166–9.
2. Verweij BH, Amelink GJ, Muizelaar JP. Current concepts of cerebral oxygen 29. Plum F, Posner JB. The diagnosis of stupor and coma. Philadelphia: FA
transport and energy metabolism after severe traumatic brain injury. Prog Davis; 1980.
Brain Res 2007;161:111–24. 30. Cserr HF, Knopf PM. Cervical lymphatics, the blood-brain barrier and the
3. Chien S. Shear dependence of effective cell volume as a determinant of immunoreactivity of the brain: a new view. Immunol Today 1992;13:507–12.
blood viscosity. Science 1970;168:977–9. 31. Lee KR, Hoff JT. Intracranial pressure. In: Youmans JR, editor. Neurological
4. Albrecht KH, Gaehtgens P, Pries A, et al. The Fahraeus effect in narrow surgery: a comprehensive reference guide to the diagnosis and management
capillaries (i.d. 3.3 to 11.0 micron). Microvasc Res 1979;18:33–47. of neurosurgical problems. 4th ed. Philadelphia: WB Saunders; 1996.
5. Kontos HA, Raper AJ, Patterson JL. Analysis of vasoactivity of local pH, 32. Lindenberg R. Compression of the brain arteries as pathogenetic factor for
PaCO2 and bicarbonate on pial vessels. Stroke 1977;8:358–60. tissue necroses and their areas of predilection. J Neuropathol Exper Neurol
6. Sokoloff L. Metabolism of the central nervous system in vivo. In: Field J, 1955;14:223–43.
Magoun HW, Hall VE, editors. Handbook of physiology. Washington, DC: 33. Miller J. The pathophysiology of raised intracranial pressure. In: Greenfield
American Physiological Society; 1960. pp. 1843–64. JG, Adams JH, Corsellis JAN, et al, editors. Greenfield’s neuropathology.
7. Jones TH, Morawetz RB, Crowell RM, et al. Thresholds of focal cerebral London: Edward Arnold; 1984. pp. 53–84.
ischemia in awake monkeys. J Neurosurg 1981;54:773–82. 34. Huseby JS, Pavlin EG, Butler J. Effect of positive end-expiratory pressure on
8. Schroder ML, Muizelaar JP, Kuta AJ, et al. Thresholds for cerebral ischemia intracranial pressure in dogs. J Appl Physiol 1978;44(1):25–7.
after severe head injury: relationship with late CT findings and outcome. 35. Marmarou A, Shulman K, Rosende RM. A nonlinear analysis of the
J Neurotrauma 1996;13:17–23. cerebrospinal fluid system and intracranial pressure dynamics. J Neurosurg
9. Celsis P, Chan M, Marc-Vergnes JP, et al. Measurement of cerebral 1978;48:332–44.
circulation time in man. Eur J Nucl Med 1985;10:426–31. 36. Shapiro K, Marmarou A, Shulman K. Characterization of clinical CSF
10. Ferrari M, Wilson DA, Hanley DF, et al. Effects of graded hypotension on dynamics and neural axis compliance using the pressure-volume index. I.
cerebral blood flow, blood volume, and mean transit time in dogs. Am J The normal pressure-volume index. Ann Neurol 1980;7:508–14.
Physiol 1992;262(6 Pt 2):H1908–14. 37. Bouma GJ, Muizelaar JP, Bandoh K, et al. Blood pressure and intracranial
11. Rosengarten B, Rüskes D, Mendes I, et al. A sudden arterial blood pressure pressure-volume dynamics in severe head injury: relationship with cerebral
decrease is compensated by an increase in intracranial blood volume. blood flow. J Neurosurg 1992;77:15–9.
J Neurol 2002;249(5):538–41. 38. Siesjo BK. Cerebral circulation and metabolism. J Neurosurg
12. Singer OC, de Rochemont Rdu M, Foerch C, et al. Relation between relative 1984;60:883–908.
cerebral blood flow, relative cerebral blood volume, and mean transit time in 39. Guyton A. Textbook of medical physiology. Philadelphia: WB Saunders; 1986.
patients with acute ischemic stroke determined by perfusion-weighted MRI. 40. Vespa P, Bergsneider M, Hattori N, et al. Metabolic crisis without brain
J Cereb Blood Flow Metab 2003;23(5):605–11. ischemia is common after traumatic brain injury: a combined microdialysis
13. Nordström CH. Physiological and biochemical principles underlying and positron emission tomography study. J Cereb Blood Flow Metab
volume-targeted therapy—the “Lund concept.” Neurocrit Care 2005;2(1): 2005;25(6):763–74.
83–95. 41. Pellerin L. How astrocytes feed hungry neurons. Mol Neurobiol
14. Muizelaar JP, Lutz HA, 3rd, Becker DP. Effect of mannitol on ICP and CBF 2005;32(1):59–72.
and correlation with pressure autoregulation in severely head-injured 42. Schurr A, Miller JJ, Payne RS, et al. An increase in lactate output by brain
patients. J Neurosurg 1984;61:700–6. tissue serves to meet the energy needs of glutamate-activated neurons.
15. Mitagvaria NP. Regulation of local cerebral blood flow. Adv Exp Med Biol J Neurosci 1999;19(1):34–9.
1984;180:861–79. 43. Pellerin L, Magistretti PJ. How to balance the brain energy budget while
16. Muizelaar JP, Marmarou A, Ward JD, et al. Adverse effects of prolonged spending glucose differently. J Physiol 2003;546(Pt 2):325.
hyperventilation in patients with severe head injury: a randomized clinical 44. Gallagher CN, Carpenter KL, Grice P, et al. The human brain utilizes lactate
trial. J Neurosurg 1991;75:731–9. via the tricarboxylic acid cycle: a 13C-labelled microdialysis and high-
17. Rangel-Castilla L, Gasco J, Nauta HJ, et al. Cerebral pressure autoregulation resolution nuclear magnetic resonance study. Brain 2009;132(Pt 10):2839–
in traumatic brain injury. Neurosurg Focus 2008;25:E7. 49. Epub 2009 Aug 20.
18. Rosenthal G, Hemphill JC III, Sorani M, et al. Brain tissue oxygen 45. Pasqualin A. Cerebral metabolism. In: Youmans JR, editor. Neurological
tension is more indicative of oxygen diffusion than oxygen delivery and surgery: a comprehensive guide to the diagnosis and management of
metabolism in patients with traumatic brain injury. Crit Care Med 2008;36: neurosurgical problems. 4th ed. Philadelphia: WB Saunders; 1996.
1917–24. 46. Sokoloff L. Relationships among local functional activity, energy metabolism,
19. Zauner A, Daugherty WP, Bullock MR, et al. Brain oxygenation and energy and blood flow in the central nervous system. Fed Proc 1981;40:2311–6.
metabolism: part I-biological function and pathophysiology. Neurosurgery 47. Sokoloff L. Localization of functional activity in the central nervous system
2002;51:289–301; discussion 2. by measurement of glucose utilization with radioactive deoxyglucose.
20. Siesjo B. Brain energy metabolism. Chichester, UK: Wiley; 1978. J Cereb Blood Flow Metab 1981;1:7–36.
21. Maloney-Wilensky E, Le Roux P. The physiology behind direct brain oxygen 48. Wahl M, Kuschinsky W. The dilatatory action of adenosine on pial arteries
monitors and practical aspects of their use. Childs Nerv Syst of cats and its inhibition by theophylline. Pflugers Arch 1976;362:55–9.
2010;26(4):419–30. 49. Edvinsson L, MacKenzie ET, McCulloch J. Cerebral blood flow and
22. Scheufler K-M, Lehnert A, Rohrborn H-J, et al. Individual values of brain metabolism. New York: Raven Press; 1993.
tissue oxygen pressure, microvascular oxygen saturation, cytochrome redox 50. Bonvento G, Lacombe P, Seylaz J. Effects of electrical stimulation of the
level and energy metabolites in detecting critically reduced cerebral energy dorsal raphe nucleus on local cerebral blood flow in the rat. J Cereb Blood
state during acute changes in global cerebral perfusion. J Neurosurg Flow Metab 1989;9:251–5.
Anesthesiol 2004;16:210–19 51. Tuor UI, Edvinsson L, McCulloch J. Catecholamines and the relationship
23. Valadka AB, Goodman JC, Gopinath SP, et al. Comparison of brain tissue between cerebral blood flow and glucose use. Am J Physiol
oxygen tension to microdialysis-based measures of cerebral ischemia in 1986;251:H824–33.
fatally head-injured humans. J Neurotrauma 1998;15(7):509–19. 52. Reilly PL. Brain injury: the pathophysiology of the first hours. “Talk and die
24. Valadka AB, Hlatky R, Furuya Y, et al. Brain tissue PO2: correlation with revisited.” J Clin Neurosci 2001;8:398–403.
cerebral blood flow. Acta Neurochir Suppl 2002;81:299–301. 53. Tisdall MM, Smith M. Multimodal monitoring in traumatic brain injury:
25. Maas AI, Fleckenstein W, de Jong DA, et al. Monitoring cerebral oxygenation: current status and future directions. Br J Anaesth 2007;99:61–7.
experimental studies and preliminary clinical results of continuous 54. Chesnut RM. Secondary brain insults after head injury: clinical perspectives.
monitoring of cerebrospinal fluid and brain tissue oxygen tension. Acta New Horiz 1995;3:366–75.
Neurochir Suppl (Wien) 1993;59:50–7. 55. Choi DW. Ionic dependence of glutamate neurotoxicity. J Neurosci
26. Hlatky R, Valadka AB, Goodman JC, et al. Patterns of energy substrates 1987;7:369–79.
during ischemia measured in the brain by microdialysis. J Neurotrauma 56. Unterberg AW, Stover J, Kress B, et al. Edema and brain trauma.
2004;21(7):894–906. Neuroscience 2004;129:1021–9.
7.e2 Section I—Background

57. Fakhry SM, Trask AL, Waller MA, et al. Management of brain-injured 72. Schmidt EA, Piechnik SK, Smielewski P, et al. Symmetry of cerebral
patients by an evidence-based medicine protocol improves outcomes and hemodynamic indices derived from bilateral transcranial Doppler.
decreases hospital charges. J Trauma 2004;56:492–9; discussion 9–500. J Neuroimaging 2003;13:248–54.
58. Jones PA, Andrews PJ, Midgley S, et al. Measuring the burden of secondary 73. Bouma GJ, Muizelaar JP, Stringer WA, et al. Ultra-early evaluation of
insults in head-injured patients during intensive care. J Neurosurg regional cerebral blood flow in severely head-injured patients using
Anesthesiol 1994;6:4–14. xenon-enhanced computerized tomography. J Neurosurg 1992;77:360–8.
59. Patel HC, Menon DK, Tebbs S, et al. Specialist neurocritical care and 74. Gobiet W, Grote W, Bock WJ. The relation between intracranial pressure,
outcome from head injury. Intensive Care Med 2002;28:547–53. mean arterial pressure and cerebral blood flow in patients with severe head
60. Siesjo BK. Pathophysiology and treatment of focal cerebral ischemia. Part I: injury. Acta Neurochir (Wien) 1975;32:13–24.
Pathophysiology. J Neurosurg 1992;77:169–84. 75. Thomas DJ, du Boulay GH, Marshall J, et al. Cerebral blood-flow in
61. Hakim AM. The cerebral ischemic penumbra. Can J Neurol Sci 1987;14:557–9. polycythaemia. Lancet 1977;2:161–3.
62. Baron JC, Rougemont D, Soussaline F, et al. Local interrelationships of 76. Eng CC, Lam AM, Byrd S, et al. The diagnosis and management of a
cerebral oxygen consumption and glucose utilization in normal subjects and perianesthetic cerebral aneurysmal rupture aided with transcranial Doppler
in ischemic stroke patients: a positron tomography study. J Cereb Blood ultrasonography. Anesthesiology 1993;78:191–4.
Flow Metab 1984;4:140–9. 77. Kobayashi KI, Koike T. Cerebral blood flow and metabolism in patients with
63. Hlatky R, Valadka AB, Robertson CS. Intracranial pressure response to ruptured aneurysms. Acta Neurol Scand Suppl 1979;60:292–3.
induced hypertension: role of dynamic pressure autoregulation. 78. Fein JM. Cerebral energy metabolism after subarachnoid hemorrhage. Stroke
Neurosurgery 2005;57:919–23. 1975;6:1–8.
64. Miller JD, Sweet RC, Narayan R, et al. Early insults to the injured brain. 79. Gelmers HJ, Beks JW, Journee HL. Regional cerebral blood flow in
JAMA 1978;240:439–42. patients with subarachnoid haemorrhage. Acta Neurochir (Wien) 1979;47:
65. Bouma GJ, Muizelaar JP, Choi SC, et al. Cerebral circulation and metabolism 245–51.
after severe traumatic brain injury: the elusive role of ischemia. J Neurosurg 80. Fein J, Boulos R. Local cerebral blood flow in experimental middle cerebral
1991;75:685–93. artery vasospasm. J Neurosurg 1973;39:337–47.
66. Graham DI, Adams JH. Ischaemic brain damage in fatal head injuries. 81. Ishii R. Regional cerebral blood flow in patients with ruptured intracranial
Lancet 1971;1:265–6. aneurysms. J Neurosurg 1979;50:587–94.
67. Junger EC, Newell DW, Grant GA, et al. Cerebral autoregulation following 82. Nornes H, Knutzen HB, Wikeby P. Cerebral arterial blood flow and
minor head injury. J Neurosurg 1997;86:425–32. aneurysm surgery. Part 2: Induced hypotension and autoregulatory capacity.
68. Enevoldsen EM, Cold G, Jensen FT, et al. Dynamic changes in regional CBF, J Neurosurg 1977;47:819–27.
intraventricular pressure, CSF pH and lactate levels during the acute phase 83. Meyer CH, Lowe D, Meyer M, et al. Progressive change in cerebral blood
of head injury. J Neurosurg 1976;44:191–214. flow during the first three weeks after subarachnoid hemorrhage.
69. Fieschi C, Battistini N, Beduschi A, et al. Regional cerebral blood flow and Neurosurgery 1983;12:58–76.
intraventricular pressure in acute head injuries. J Neurol Neurosurg 84. Sundt TM, Jr, Whisnant JP. Subarachnoid hemorrhage from intracranial
Psychiatry 1974;37:1378–88. aneurysms. Surgical management and natural history of disease. N Engl J
70. Bouma GJ, Muizelaar JP. Cerebral blood flow, cerebral blood volume, and Med 1978;299:116–22.
cerebrovascular reactivity after severe head injury. J Neurotrauma 1992;9 85. Adams HP, Jr, Kassell NF, Torner JC, et al. Early management of aneurysmal
Suppl 1:S333–48. subarachnoid hemorrhage. A report of the Cooperative Aneurysm Study.
71. Obrist WD, Langfitt TW, Jaggi JL, et al. Cerebral blood flow and metabolism J Neurosurg 1981;54:141–5.
in comatose patients with acute head injury. Relationship to intracranial
hypertension. J Neurosurg 1984;61:241–53.
I
Chapter
2  

Why Monitor and Principles


of Neurocritical Care
Syed T. Arshad and Jose I. Suarez

World War II further expanded the field of intensive care with


Introduction the designation of “shock wards.”
The collective burden of critical neurologic illnesses is over- The advent of polio vaccines and few important develop-
whelming. Stroke occurs in about 800,000 individuals each ments in neurosurgical techniques led to a somewhat a nihil-
year in the United States, with case fatality rates from acute istic approach to critically ill neurologic and neurosurgical
ischemic stroke (AIS) being close to 15% to 20%. Although patients in the 1950s. By 1960, health care witnessed a rapid
primary intracerebral hemorrhage (ICH) and aneurysmal spread of resuscitation and surgical techniques, including the
subarachnoid hemorrhage (SAH) are less common than isch- creation of recovery rooms. Toward the late 1960s almost 95%
emic stroke, their case fatality rates are nearly two to three of all acute care hospitals in the United States had some sort
times higher than AIS.1 Traumatic brain injury (TBI), accord- of critical care unit. Advances in neuroanesthesia and the
ing to the World Health Organization, will surpass many dis- organization of critical care protocols allowed for a more com-
eases as the major cause of death and disability worldwide by prehensive care model for the critically ill. In the 1960s, tech-
the year 2020.2 The number of patients with hypoxic-ischemic niques to measure cerebral blood flow (CBF) and intracranial
encephalopathy following resuscitation is between 100,000 pressure (ICP) were introduced and evolved. During the
and 200,000 per year in the United States.3 The mortality in 1970s, neurologists David Jackson and Alan Ropper, and an
this particular cohort of critically ill neurologic patients has anesthesiologist, Sean Kennedy, spearheaded the concept of a
been decreasing steadily, in part because of advances in scien- “neuro-ICU” in North America. Later pioneers included Dr.
tific understanding of the clinical disorder but perhaps more Daniel Hanley and Dr. Thomas Bleck. In 2003, the Neurocriti-
so because of the impact of neurologically oriented critical cal Care Society was formed with Dr. Bleck as its first presi-
care. In addition, patients with AIS have better outcomes dent.5,6 The scope of neurocritical care has further evolved
when admitted to a dedicated stroke/neurocritical care unit with the advent of more sophisticated monitoring techniques
rather than a general medical intensive care unit (ICU). In and informatics and computer systems and guidelines for
addition, studies have shown that admission of ICH patients program requirements for neurocritical care training and a
to specialized neurocritical care units is associated with core curriculum and competencies for training.7,8
improved clinical outcome. The institution of such dedicated
neurocritical care units has improved resource utilization,
introduced more efficient patient care protocols, established Scope of Practice of the
neuroprotective measures, reduced the impact of comorbid
illness, and allowed for prevention and management of post-
Modern Neurointensivist
stroke complications.4 In day-to-day clinical practice, neurointensivists focus on
subtle changes in the neurologic exam and physiologic or
pathophysiologic interactions between the brain and other
Principles of Neurocritical Care organ systems. The main argument in favor of staffing
neuro-ICUs with full-time neurointensivists stems from the
A Brief History of Neurocritical Care notion that these individuals are specially trained to recog-
Historians date the beginning of neurocritical care back to the nize the specific interactions between the intracranial physi-
16th century, at the time resuscitation practices and attempts ology and systemic derangements of the neurocritically ill
at artificial ventilation first appeared. Modern neurocritical patient.9 The members of a neurocritical care team are more
care probably started with the polio epidemics and evolved likely to be aware of secondary physiologic insults to the
thereafter with the introduction of the iron lung in the early brain that can include fever, hyperglycemia, anemia, hypo-
to mid-20th century. At that time neurologists were the natremia, and delirium. A neurocritical care team typically
primary treating physicians for these patients and probably will care for patients with AIS, ICH, SAH, intracranial neo-
laid the groundwork for the first large-scale use of mechanical plasms, TBI and spinal cord injury, status epi­lepticus, neuro-
ventilation. In 1923, a neurosurgeon, Dr. W.E. Dandy, created muscular respiratory failure, postoperative neurosurgical
a three-bed unit for postoperative neurosurgical patients. care, hypoxic-ischemic brain injury after cardiac arrest,
8 © Copyright 2013 Elsevier Inc. All rights reserved.
Section I—Background 9

postprocedure neurovascular care, and acutely ill medical relationship is expressed by the Fick’s equation: CMRO2 = CBF
patients with neurologic injury requiring critical care, for × AVDO2, in which AVDO2 represents arteriovenous differ-
example, hepatic encephalopathy. Neurointensivists have ence of oxygen. Under normal conditions the brain maintains
intimate knowledge of acute circulatory, respiratory, and a constant AVDO2 by responding to changes in metabolism,
metabolic disturbances and general skills required for cerebral perfusion pressure (CPP), and blood viscosity with
advanced cardiac life support, cardioversion, and intubation changes in vessel caliber, a phenomenon referred to as
and ventilator management, and for the insertion of invasive autoregulation.21-23 Ischemia occurs if oxygen delivery is below
hemodynamic monitoring. In addition, given their knowl- the metabolic demand of the tissue (despite the increased
edge with nervous system function, neurointensivists have AVDO2). Hyperemia, or “luxury perfusion,” occurs if oxygen
significant experience in end-of-life questions, prognosis in delivery is greater than the metabolic demand. These various
severe brain injury, and defining brain death. Consequently changes may not always manifest clinical features, and moni-
neurointensivists lead the way at the interface for organ pro- toring techniques can help detect and define these parameters
curement and donation. before irreversible injury occurs.
CPP, defined as mean arterial pressure (MAP) minus ICP,
is well maintained in healthy subjects. In pathologic states
Impact of Specialized there can be initial compensatory changes to maintain CPP;
Neurocritical Care Team for example, small increases in intracranial volume such as an
Several observational studies suggest that the evolution intracranial mass lesion can be accommodated for by translo-
of neurocritical care units (NCCUs) has led to improved out- cation of cerebrospinal fluid (CSF) into distensible spinal sub-
comes and optimal utilization of resources for patients with arachnoid space with little effect on ICP. This is expressed as
severe acute brain injury (Table 2.1).9-17 Mortality after TBI the Monro-Kellie doctrine. Exhaustion of this compensatory
has improved because of prehospital advanced life support mechanism can result in large increases in ICP, and so decrease
and specialized neurointensivist-led teams.10 In patients with CPP or CBF. Uncorrected, this results in a vicious cycle of
AIS and who are critically ill, admission to neurointensivist- further increases in ICP and decreases in CBF. Various moni-
led NCCU is asso­ciated with improved condition at discharge tors can help define and detect this delicate balance.
and shortened hospital length of stay.4,11 ICH traditionally has A large body of evidence suggests that knowledge of normal
carried the highest mortality rate and been the most disabling CBF physiology and alterations caused by specific diseases is
form of stroke. Recent data suggest that those patients admit- paramount in the treatment of critically ill patients with acute
ted to the NCCU have improved outcomes, shorter lengths of brain injury, although not all cellular dysfunction results from
stay, and lower total cost of care compared with a national altered blood flow. CBF is equal to CPP divided by the cere-
benchmark.12,13 In addition, studies show that mortality brovascular resistance (CVR) (CBF = CPP/CVR). Measure-
among patients who undergo surgery for ruptured intracra- ment of CBF in the critically ill can be technically difficult, so
nial aneurysms is significantly reduced in high-volume centers CPP (i.e., ICP and MAP) is monitored as a surrogate for the
that provide access to specialized multidisciplinary care.14 In adequacy of CBF. Exactly what optimal CPP is remains
addition, aggressive treatment through specialized care is debated,24 but adequate values for CPP generally are consid-
associated with improved outcome and proven to be cost ered to be between 55 and 100 mm Hg. This is a wide range,
effective in poor-grade subarachnoid hemorrhage patients.15,16 and these values, however, may depend on the specific patient
Hypoxic-ischemic brain injury after cardiac arrest is one of and pathology and so emphasize a need for monitoring. In
the most devastating conditions; historically only 15% of addition, a normal CPP represents a normal CBF only if CVR
patients successfully resuscitated in the field have survived to is normal. If CVR is high, a normal CPP still may be accom-
hospital discharge. Studies have shown that induced therapeu- panied by ischemia. Alternatively, if CVR is low, a normal CPP
tic hypothermia after cardiac arrest is associated with improved may be associated with hyperemia and increased ICP. Thus it
survival and better functional outcomes.17,18 In many institu- is important to maintain CPP in acute neurologic disorders,
tions neurointensivists have become the ideal practitioners to preferably in a range that is targeted to the patient especially
care for these critically ill patients. Studies also have shown in situations in which autoregulation is impaired. Although
that availability of neurointensivists is associated with subtle changes in serial neurologic examinations may suggest
improved documentation in the medical records and an altered CBF, the sensitivity of these clinical findings can be
increased rate of organ and tissue donation after brain death enhanced by careful attention to other bedside indicators,
declarations.19,20 such as CPP.

Physiologic Parameters in Neurocritical


Care Patients Neurocritical Care Delivery
The adult brain (1200-1400 g) comprises 2% to 3% of total
and Patient Outcome
body weight and yet receives 15% to 20% of cardiac output. The Senate and House of Representatives of the United States
However, it has a high cerebral metabolic rate of oxygen designated the decade beginning January 1, 1990, as the
(CMRO2) and uses glucose predominantly as a substrate for “Decade of the Brain.” The goal of this proclamation was to
its energy needs. Normal CBF in humans averages 50 mL/100  stimulate multidisciplinary efforts of scientists from diverse
g/min brain tissue per minute. Irreversible neuronal damage areas to better understand “the structure of the brain and how
occurs when CBF is less than 10 mL/100 g/min. The brain it affects our development, health and behavior.” An immedi-
has no significant storage capacity; hence cerebral metabo- ate consequence of this has been a significant increase in the
lism, CBF, and oxygen extraction are tightly coupled. This number of new tools available to monitor the central nervous
Table 2.1  Summary of Studies That Investigate the Impact of Specialized Neurocritical Care on Outcomes of Critically Ill 10
Neurologic and Neurosurgical Patients.
Author Objective Design Outcome Measure Conclusion
Diringer To determine whether mortality rate after Outcomes, Hospital mortality, hospital For patients with acute ICH, admission to a neuro- vs.
et al, 2001 intracerebral hemorrhage (ICH) is lower cross-sectional length of stay (LOS), general ICU is associated with reduced mortality rate. The
in patients admitted to a neurologic or with concurrent readmission rates, presence of a full-time intensivist was associated with a
neurosurgical (neuro) intensive care unit control, long-term mortality lower mortality rate
(ICU) compared with those admitted to retrospective
general ICUs
Patel et al, To document the effect of neurocritical Cohort with Hospital mortality Specialist neurocritical care with protocol-driven therapy is
2002 care, delivered by specialist staff and historical control, associated with a significant improvement in outcome for
based on protocol-driven therapy aimed retrospective all patients with severe head injury. Such management
at intracranial pressure (ICP) and may also benefit patients requiring no surgical therapy,
cerebral perfusion pressure (CPP) some of whom may need complex therapeutic
Section I—Background

targets, on outcome in acute head injury interventions


Wilby et al, To prospectively assess outcome and cost Outcomes, Hospital mortality, hospital Poor-grade aneurysmal subarachnoid hemorrhage is
2003 for poor-grade subarachnoid cross-sectional length of stay, cost associated with a high mortality, but a significant subset
hemorrhage patients presenting to a analysis of patients can achieve favorable outcomes.
regional neurosurgical center
Berman et al, To examine the impact of hospital Outcomes, Hospital mortality, hospital Hospital procedural volume and the propensity of a
2003 characteristics on outcome after the cross-sectional length of stay hospital to use endovascular therapy are both
treatment of ruptured and unruptured independently associated with better outcome
cerebral aneurysms
Suarez et al, Length of stay and mortality in Cohort with Hospital mortality, hospital Introduction of a neurocritical care team, including a
2004 neurocritically ill patients: impact of a historical control, length of stay, readmission full-time neurointensivist who coordinated care, was
specialized neurocritical care team retrospective rates, long-term mortality associated with significantly reduced in-hospital mortality
and length of stay without changes in readmission rates
or long-term mortality.
Helms et al, Measuring the effect of uncoupling and Cohort with End-of-life care and organ Neurointensivist-led policy change resulted in an increase in
2004 removal of the treating physician from historical control, donation consent rates for organ donation.
organ and tissue donation requests on retrospective
consent rates for donation in the
neurocritical care unit
Varelas et al, To evaluate the impact of a newly Cohort with Hospital mortality, hospital The institution of a neurointensivist-led team model was
2004 appointed neurointensivist on historical control, length of stay, readmission associated with an independent positive impact on
neurosciences intensive care unit (NICU) retrospective rates, long-term mortality patient outcomes, including a lower ICU mortality, LOS,
patient outcomes and quality of care and discharge to a skilled nursing facility and a higher
variables discharge home.
Varelas et al, Examined the hypothesis that a newly Cohort with Length of stay, readmission A major change was implemented in the NICU regarding
2005 appointed neurointensivist may alter historical control, rates documentation after a neurointensivist was appointed.
documentation practices in a university retrospective
hospital setting
Varelas et al, Impact of a neurointensivist on outcomes Cohort with Hospital mortality, hospital Neurointensivist-led team model had an independent,
2006 in patients with head trauma treated in historical control, length of stay, readmission positive impact on patient outcomes, including a lower
an NICU retrospective rates, long-term mortality NICU-associated mortality rate and hospital LOS,
improved disposition, and better chart documentation
Bershad Impact of a specialized neurointensive care Cohort with Hospital mortality, hospital Institution of a dedicated neurocritical care team was
et al, 2008 team on outcomes of critically ill acute historical control, length of stay, readmission associated with a reduction in resource utilization and
ischemic stroke patients retrospective rates, long-term mortality improved patient outcomes at hospital discharge.
Varelas et al, The impact of a neurointensivist on Cohort with Hospital mortality, hospital The direct patient care offered and the organizational
2008 patients with stroke admitted to an historical control, length of stay, readmission changes implemented by a neurointensivist shortened
NICU retrospective rates, long-term mortality the NCCU stay and hospital LOS and improved the
disposition of patients with strokes admitted to an NCCU.
Section I—Background 11

system.25 A multidisciplinary team comprised of neurologists, future of neurocritical care will see a further development of
neurointensivists, neurosurgeons, and neuroradiologists is multimodality monitoring in large part because it now is real-
frequently seen in the NCCU of the academic medical center ized that no one monitor can provide all the necessary infor-
of today. The efficiency and core proficiency of any NCCU mation about what is happening in the brain. In addition, the
further depends on a central core of nurses with neurocritical evolution of computing power and informatics means that the
care training. information from various monitors can in the future be used
There is ongoing debate about whether general ICUs for neurophysiologic decision support. Advanced monitoring
require a dedicated, full-time team. Several studies have shown techniques that can provide real-time information about the
that admission to closed units is associated with a decrease in relative health or distress of the brain and specifically cellular
morbidity and mortality rates, average ventilation time, health, such as brain tissue oxygen tension, signal-processed
reduced length of stay, complications, resource utilization, and continuous EEG, and neurochemical analysis using microdi-
total hospital charges. For patients with ICH, TBI, and severe alysis likely will be used to develop strategies to maintain an
AIS, the introduction of a multidisciplinary NCCU team has optimal physiologic environment for the comatose injured
been associated with decreased mortality, shortened the hos- brain. This may be particularly important because most
pital length of stay, and lowered total cost of care, and has led studies of single agents or neuroprotection, although success-
to better disposition at discharge, that is to say, increased rate ful in the laboratory, have not proven beneficial in clinical
of home discharges and a concomitant decrease in nursing practice. This suggests that strategies that are based on under-
home discharges.16,26 Differences in care, including the use of standing of physiology and pathophysiology (i.e., that are
more invasive intracranial monitoring, tracheotomy, and less monitoring based) may be useful given the complex and het-
use of intravenous (IV) sedation, may account for some of the erogeneous consequences of any injury to the brain.
observed better outcome in NCCUs.27 It remains to be seen
whether true “closed” or “open” NCCUs provide better out-
comes. However, it does appear that patients with neurologic
Primary and Secondary Brain Injury
disorders that require ICU admission are better in dedicated It is now well established that any acute insult to the brain,
NCCUs than general ICUs.12 Although few studies have whether it be trauma, ischemia, hemorrhage, edema, or
directly addressed the value of using information from a infection among others, is associated with primary and sec-
monitor in the NCCU, a recent meta-analysis of the TBI lit- ondary brain injury. The same is true for spinal cord injury.
erature that included more than 100,000 patients suggests In large part the prevention and management of secondary
there is a value to using an ICP monitor and aggressive treat- injury through early detection with a variety of monitors has
ment in severe TBI.28 Similarly clinical studies suggest that use become the focus of modern NCCUs. Recognition of primary
of a brain oxygen monitor and ICP monitor and management and secondary brain injury allows the NCCU team to orga-
based on that may help improve outcome,29 whereas microdi- nize the proper resources to influence clinical outcome in the
alysis studies suggest that changes in the brain can be observed critically ill patient. Secondary injury can be complex and
before altered ICP occurs.30 represent an interaction between systemic and intracranial
factors. Therefore knowledge of cellular mechanisms of
injury through monitoring becomes important to allow early
Why Monitor Patients in the NCCU: intervention. There are many biochemical and pathophysio-
logic processes that are activated. For example, when CPP
Historical Perspective and as a consequence, CBF are reduced, failure of aerobic
Between the 1960s and 1980s monitoring in the ICU was metabolism and thus the electron transport system may
restricted to clinical examination, heart and respiratory rate, result. This in turn leads to disruption of the mitochondrial
blood pressure, oxygen saturation, body temperature, and membrane potential and depletion of adenosine triphos-
central venous pressure. NCCUs at that time focused almost phate (ATP). During the process oxygen radicals are released
exclusively on the care of postoperative neurosurgical patients, and the ensuing anaerobic metabolism lowers the pH and
and neuromonitoring was restricted primarily to serial neu- can induce cellular edema; this prevents repolarization
rologic examination and, in some units, ICP monitoring. This further and exacerbates ATP depletion. There is also release
era can be thought of as the age of clinical neuromonitoring. of excitatory amino acids, and CSF concentrations of gluta-
Between 1980 and 2000, use of ICP monitoring became more mate, glycine, and aspartate can increase significantly.21 A
widespread and the age of physiologic neuromonitoring was change in the immune response triggers the production of
ushered in. The idea was to detect and treat increases in ICP excitatory cytokines (interleukin [IL]-6 and tumor necrosis
before they led to obvious and often irreversible clinical dete- factor) that contribute to dysfunction of the blood-brain
rioration. The current decade has focused on monitoring and barrier. These various intracranial processes generally are
management based on brain-derived physiologic information detected by indirect measures (e.g., ICP, glucose metabolism
to detect secondary injury in its earliest phase. Monitoring through microdialysis, EEG, brain oxygen), but newer tech-
techniques in the ICU can be divided into “whole-brain moni- niques are evolving to allow more direct assessments. In
toring,” which includes ICP monitoring devices, jugular bulb addition it appears that processes considered to be “symp-
catheters, electroencephalography (EEG), evoked potentials, toms,” for example, seizures, are actually secondary insults.
and “regional brain monitoring,” which includes transcranial Many secondary injuries also result from systemic factors
Doppler (TCD) ultrasonography, xenon-133 clearance, laser such as hypoxia, hypotension, hyperglycemia, hyperthermia,
Doppler flowmetry, thermal diffusion flowmetry, microdialy- shivering anemia, coagulation disorders, and organ
sis catheters, and brain-tissue oxygen probes. These various dysfunction.31-34 In addition secondary injury may result
monitors can be supplemented with radiologic studies. The from treatment; for example, hyperventilation may reduce
12 Section I—Background

ICP but simultaneously decrease CBF. The ability to monitor each technique is presented in other chapters in this book.
for systemic dysfunction and the effects of treatment hence Broadly these monitors provide either single measurements at
become important in caring for a patient in the NCCU. a specific point in time (radiographic techniques) or continu-
ous information.
An important advance in the management of critically ill
Rationale for Systemic Monitoring patients has been the emergence of functional imaging and
Patient monitoring is an integral part of the management of sophisticated magnetic resonance imaging (MRI) techniques.
severely brain-injured patients in the NCCU and can be clas- Consequently radiology has undergone a paradigm shift from
sified broadly into systemic monitoring and brain-specific simply providing an anatomic image to providing informa-
techniques. Brain-specific techniques may be further classified tion about tissue metabolism. These techniques remain limited
into radiographic techniques or continuous methods. Second- by fixed time windows despite the development of faster scan-
ary brain injury can result from systemic insults. Furthermore, ners and higher-resolution detectors.
efforts to treat the intracranial pathophysiology, although suc- Positron emission tomography (PET) may be considered
cessful in reversing abnormalities in the brain, can aggravate the current gold-standard method to image brain metabolism.
function in other organ systems and so not improve outcome.35 Although largely a research tool, it has been shown to be
Observational data suggest that efforts to correct systemic specific for changes in CBF, cerebral blood volume (CBV),
insults can help improve outcome in the NCCU.36 Systemic and oxygen and glucose requirements (rCMRO2/rCMRGlu)
monitoring includes invasive arterial blood pressure record- including regional and relative changes. PET also can be used
ing, core body temperature, heart rate, systemic arterial oxygen to identify some traumatic brain injuries not associated with
saturation (SaO2), and central venous pressure. Invasive blood overt anatomic abnormalities. For example, diffuse axonal
pressure monitoring may be indicated when excessive blood injury (DAI) following TBI is associated with diffuse cortical
pressure elevations may lead to ICH or hematoma expansion hypometabolism and a decrease in CMRO2 in the visual
such as in the post-thrombolysis patient or with primary ICH; cortex.39 Newer MRI techniques are largely replacing some of
in those with loss of cerebral autoregulation, for example, PET’s role. In addition PET is expensive and available in only
poor-grade SAH; or in patients with autonomic instability a few major medical centers.
such as severe Guillain-Barré syndrome. In addition, invasive Single-photon emission computed tomography (SPECT) is
blood pressure monitoring can help guide therapy in those available in most medical centers and shares similar basic
patients with neurologic injury with septic shock or those principles to PET scanning but has less spatial resolution and
with critical vascular stenosis and fluctuations of blood provides qualitative rather than quantitative information.
pressure. SPECT can be used to measure CBV and mean transit time
Monitoring central venous pressure (CVP) and in special (MTT). CBF is then calculated from the equation: CBF =
circumstances the use of pulmonary artery catheterization CBV/MTT. Abnormalities detected using SPECT within 24
that can provide an estimate of intravascular volume status in hours after TBI even with “negative” initial head computed
conditions such as pulmonary edema, cardiac failure, cerebral tomography (CT) scans are associated with patient outcome.40
vasospasm, severe preeclampsia, and sepsis are useful. Heart CT perfusion is a newer imaging modality that provides
rate, telemetry, respiratory rate, and oxygen saturation help similar qualitative information as SPECT and is able to
alert the neurocritical care team to early signs of sepsis, pul- measure rCBF, rCBV and rMTT. In addition CT perfusion can
monary embolism, myocardial infarction, ventricular tachy- be used to detect of the state of cerebral autoregulation. Even
cardia, and pneumonia. Temperature monitoring in the though a single examination in time, this information can
NCCU is essential because hyperthermia is common and have important clinical value because in patients with normal
known to aggravate outcome in a variety of conditions admit- autoregulation, alterations of mean arterial pressure are
ted to the NCCU, whereas normothermia, perhaps through unlikely to affect CPP, whereas in those with defective auto-
suppression of the immune response, can increase the likeli- regulation, elevation of the MAP is more likely to result in
hood of favorable outcome.37,38 Regular laboratory analysis elevated ICP rather than CPP.41
of blood samples and chest x-rays supplement continuous ICP monitoring is the central monitoring technique in
monitoring. much of NCCU care. Increased ICP (>20 mm Hg) is an
important secondary insult and associated with mortality
after TBI. Patients with an admission Glasgow Coma Scale
Rationale for Neurologic Monitoring (GCS) score between 3 and 8 and an abnormal CT scan are
most likely to develop intracranial hypertension, and it is rec-
The “ideal” neurologic monitor does not exist. Instead what ommended that these patients receive an ICP monitor. Knowl-
has evolved in recent years in NCCUs is the concept of mul- edge about ICP is necessary to calculate CPP that is a
timodality monitoring, that is, the use of more than one com- “surrogate” of CBF where direct CBF monitoring is not used.
plementary method to monitor a single organ, when no one The use of an ICP monitor is described in detail in the Brain
single method can provide complete information. This is par- Trauma Foundation’s Guidelines for severe TBI. Despite
ticularly true for the brain, although more relevant than the nearly 50 years of use, there is only class II evidence literature
monitor is what is done with the information. Current neu- that supports a role for ICP monitors,42-45 although meta-
romonitoring techniques involve a range of tools that have analysis of the literature suggests use of an ICP is associated
evolved from the study of cerebral physiology and from with better outcome after TBI.28
advances in the understanding of the pathophysiology of Histopathologic evidence for ischemia is a common finding
acute brain injury. The following text presents a brief overview in autopsy studies in TBI and SAH, and it is likely that inad-
of several available techniques. A more detailed discussion on equate CBF contributes to the occurrence of post-traumatic
Section I—Background 13

secondary brain insults and increases the probability of a poor manifest, occur in more than 20% of patients in the NCCU
outcome. In addition CBF threshold values for infarction and with a variety of pathologies including TBI, SAH, or ICH,
the penumbra (which remains potentially salvageable) have among others. In addition EEG can be used to detect ischemia
been defined. The tissue with a CBF between these two thresh- that may be particularly useful in patients with SAH who
olds will proceed to infarction if CBF is not restored within a develop vasospasm. Other applications of cEEG include
limited time window. Therefore CBF monitoring offers a prognostication of outcome, for example, the use of alpha
rational approach to detect and prevent secondary insults.46-48 variability in cEEG recordings may help predict outcome
Bedside CBF monitors can be characterized as quantitative or after TBI.62-64
qualitative and use either direct or indirect methods. Jugular
oximetry and TCD provide nonquantitative or “adequacy of
CBF” data and are the methods used most often in the NCCU.
Conclusion
Jugular venous oxygen saturation (SjvO2) provides informa- Neuromonitoring has evolved and become an integral part
tion about the adequacy of global CBF in relation to metabolic of the care of patients with neurocritical care diseases. The
demands. Reduction in SjvO2 to less than 50% after TBI is primary justification for monitoring is that detection of early
associated with poor outcome, and there is some evidence to neurologic worsening can prevent irreversible brain damage.
suggest that therapies based on this information may help Neuromonitoring may help to individualize patient care
improve outcomes. However, SjvO2 is limited by its lack of decisions, guide patient management, and monitor the ther-
sensitivity to regional changes.49,50 TCD was introduced to apeutic response of interventions and so avoid any potential
clinical practice in 1981 and is based on the Doppler principle. adverse effects. In addition, monitoring may allow physicians
It is noninvasive and can be used repeatedly but its interpreta- and nurses to understand the pathophysiology of complex
tion is operator dependent. TCD measures CBF velocity and disorders, to design and implement management protocols
not CBF. However, reasonable correlations have been reported based on real-time data, and to improve neurologic outcome
between TCD and xenon CT or PET CBF measurements. and quality of life in survivors of severe brain injury. Tradi-
Invasive techniques to measure CBF continually include tional monitoring in the NCCU has been largely reactive, but
such techniques as laser Doppler flowmetry (LDF) and with the introduction of more advanced neuromonitoring
thermal diffusion flowmetry (TDF). Both are available as the information provided by newer monitoring techniques
intraparenchymal probes placed through a small craniotomy now allows trends to be defined and for the proactive treat-
or held in place by a skull bolt and offer the advantage of ment of patients. The application of clinical informatics
assessing tissue perfusion in the microcirculation rather than and the use of multimodal monitoring are evolving and
the major vessels. LDF measures erythrocyte flux and relative have become standard in some NCCUs; this is centered
changes in CBF. In TDF the catheter contains a distal thermis- on patient-specific physiologic targets. Further evolution of
tor and a second, more proximal located temperature probe. monitoring and its integration is likely in the coming
The thermistor is heated to few degrees above tissue tempera- years such that decision support will become commonplace
ture, and the temperature probe samples temperature con- in the NCCU.
stantly. The temperature difference is thus a reflection of heat
transfer and may be translated to a measure of CBF. Initial
data suggest that TDF provides a sensitive real-time assess- References
ment of intraparenchymal CBF that agrees with the xenon CT 1. Rosamond W, Flegal K, Furie K, et al. Heart disease and stroke statistics—
CBF measurements.51 Monitors that provide a measure of the 2008 Update: a report from the American Heart Association Statistics
adequacy of CBF and insights into metabolism include direct Committee and Stroke Statistics Subcommittee. Circulation 2008;117:
brain oxygen tension measurement and cerebral microdialy- e25–146.
2. Hyder AA, Wunderlich CA, Puvanachandra P, et al. The impact of
sis. Brain oxygen monitoring has demonstrated that many traumatic brain injuries: a global perspective. NeuroRehabilitation 2007;22:
of the secondary processes that complicate primary brain 341–53.
injury—hypotension, hypoxia, intracranial hypertension—all 3. Wijdicks EFM, Hijdra A, Young GB, et al. Practice parameter: prediction of
can decrease brain oxygen tension.52,53 The result of oxygen outcome in comatose survivors after cardiopulmonary resuscitation (an
evidence-based review): report of the Quality Standards Subcommittee of
deprivation is increased anaerobic metabolism, which in turn the American Academy of Neurology. Neurology 2006;67:203–10.
can lead to cell death and progressive inflammation.54 Cere- 4. Bershad EM, Feen ES, Hernandez OH, et al. Impact of a specialized
bral microdialysis can be used to detect such metabolic neurointensive care team on outcomes of critically ill acute ischemic stroke
changes before irreversible injury and may complement cere- patients. Neurocrit Care 2008;9:287–92.
bral oximetry or ICP measurements. Severe hypoxia or isch- 5. Suárez JI. Critical care neurology and neurosurgery. Totowa, NJ: Humana
Press; 2004.
emia is typically associated with marked increases in the 6. Ropper AH, Gress DR, Diringer MN, et al. Neurological and neurosurgical
lactate/pyruvate ratio that correlates with the PET-measured intensive care. 4th ed. Philadelphia: Lippincott Williams & Wilkins; 2003.
oxygen extraction fraction. An increase in the lactate- 7. Mayer SA, Coplin WM, Chang C, et al. Neurocritical Care Society; American
to-pyruvate ratio greater than what is considered normal Academy of Neurology Section on Critical Care and Emergency Neurology;
Society of Neurosurgical Anesthesia and Critical Care. Program
(20 : 25) is associated with a poor outcome in severe TBI. Both requirements for fellowship training in neurological intensive care: United
brain oxygen monitors and cerebral microdialysis can be used Council for Neurologic Subspecialties guidelines. Neurocrit Care
in patients with epilepsy, stroke, SAH, and TBI, where their 2006;5(2):166–71.
use often may detect pathology that is not detected by more 8. Mayer SA, Coplin WM, Chang C, et al. Neurocritical Care Society; American
conventional means.55-61 Academy of Neurology Section on Critical Care and Emergency Neurology;
Society of Neurosurgical Anesthesia and Critical care. Core curriculum and
Seizures are a source of secondary insult to the injured competencies for advanced training in neurological intensive care: United
brain. Recent data from continuous electroencephalography Council for Neurologic Subspecialties guidelines. Neurocrit Care 2006;5(2):
(cEEG) studies demonstrate that seizures, mostly not clinically 159–65.
14 Section I—Background

9. Suarez JI. Outcome in neurocritical care: advances in monitoring and 31. Chesnut RM, Marshall LF, Klauber MR, et al. The role of secondary brain
treatment and effect of a specialized neurocritical care team. Crit Care Med injury in determining outcome from severe head injury. J Trauma 1993;34:
2006;34:S232–8. 216–22.
10. Varelas PN, Eastwood D, Yun HJ, et al. Impact of a neurointensivist on 32. Oddo M, Schmidt JM, Carrera C, et al. Impact of tight glycemic control on
outcomes in patients with head trauma treated in a neurosciences intensive cerebral glucose metabolism after severe brain injury: a microdialysis study.
care unit. J Neurosurg 2006;104:713–19. Crit Care Med 2008;36:3233–8.
11. Varelas PN, Schultz L, Conti M, et al. The impact of a neuro-intensivist on 33. Oddo M, Milby A, Chen I, et al. Hemoglobin concentration and cerebral
patients with stroke admitted to a neurosciences intensive care unit. metabolism in patients with aneurysmal subarachnoid hemorrhage: a
Neurocrit Care 2008;9:293–9. microdialysis study. Stroke 2009;40(4):1275–81.
12. Diringer MN, Edwards DF. Admission to a neurologic/neurosurgical 34. Oddo M, Frangos S, Maloney-Wilensky E, et al. Effect of shivering on brain
intensive care unit is associated with reduced mortality rate after tissue oxygenation during induced normothermia in patients with severe
intracerebral hemorrhage. Crit Care Med 2001;29:635–40. brain injury. Neurocrit Care 2010;12(1):10–6.
13. Mirski MA, Chang CW, Cowan R. Impact of a neuroscience intensive care 35. Robertson CS, Valadka AB, Hannay HJ, et al. Prevention of secondary
unit on neurosurgical patient outcomes and cost of care: evidence-based ischemic insults after severe head injury. Crit Care Med 1999;27:2086–95.
support for an intensivist-directed specialty ICU model of care. J Neurosurg 36. Elf K, Nilsson P, Enblad P. Outcome after traumatic brain injury improved
Anesthesiol 2001;13:83–92. by an organized secondary insult program and standardized neurointensive
14. Berman MF, Solomon RA, Mayer SA, et al. Impact of hospital-related factors care. Crit Care Med 2002;30:2129–34.
on outcome after treatment of cerebral aneurysms. Stroke 2003;34:2200–7. 37. Markgraf C, Clifton G, Moody M. Treatment window for hypothermia in
15. Wilby MJ, Sharp M, Whitfield PC, et al. Cost-effective outcome for treating brain injury. J Neurosurg 2001;95:979–83.
poor-grade subarachnoid hemorrhage. Stroke 2003;34:2508–11. 38. Clifton GL, Choi SC, Miller ER, et al. Intercenter variance in clinical trials of
16. Suarez JI, Zaidat OO, Suri MF, et al. Length of stay and mortality in head trauma—experience of the National Acute Brain Injury Study:
neurocritically ill patients: impact of a specialized neurocritical care team. hypothermia. J Neurosurg 2001;95:751–5.
Crit Care Med 2004;32:2311–17. 39. Newberg AB, Alavi A. Neuroimaging in patients with head injury. Semin
17. Bernard SA, Gray TW, Buist MD, et al. Treatment of comatose survivors of Nucl Med 2003;33:136–47.
out-of-hospital cardiac arrest with induced hypothermia. N Engl J Med 40. Jacobs A, Put E, Ingels M, et al. Prospective evaluation of technetium-99m-
2002;346:557–63. HMPAO SPECT in mild and moderate traumatic brain injury. J Nucl Med
18. Hypothermia After Cardiac Arrest Study Group. Mild therapeutic 1994;35:942–47.
hypothermia to improve the neurologic outcome after cardiac arrest. N Engl 41. Wintermark M, Chiolero R, Van Melle G, et al. Cerebral vascular
J Med 2002;346:549–56. autoregulation assessed by perfusion-CT in severe head trauma patients.
19. Varelas PN, Spanaki MV, Hacein-Bey L. Documentation in medical records J Neuroradiol 2006;33:27–37.
improves after a neurointensivist’s appointment. Neurocrit Care 42. The Brain Trauma Foundation. The American Association of Neurological
2005;3:234–6. Surgeons. The Joint Section on Neurotrauma and Critical Care. Indications
20. Helms AK, Torbey MT, Hacein-Bey L, et al. Standardized protocols increase for intracranial pressure monitoring. J Neurotrauma 2000;17:479–91.
organ and tissue donation rates in the neurocritical care unit. Neurology 43. Becker DP, Miller JD, Ward JD, et al. The outcome from severe head injury
2004;63:1955–7. with early diagnosis and intensive management. J Neurosurg 1977;47:
21. Rosenthal G, Hemphill 3rd JC, Sorani M, et al. Brain tissue oxygen tension is 491–502.
more indicative of oxygen diffusion than oxygen delivery and metabolism in 44. Ghajar JB, Hairiri RJ, Paterson RH, et al. Improved outcome from traumatic
patients with traumatic brain injury. Crit Care Med 2008;36:1917–24. coma using only ventricular CSF drainage for ICP control. Adv Neurosurg
22. Obrist WD, Langfitt TW, Jaggi JL, et al. Cerebral blood flow and metabolism 1993;21:173–7.
in comatose patients with acute head injury: relationship to intracranial 45. Narayan RK, Kishore PR, Becker DP, et al. Intracranial pressure: to monitor
hypertension. J Neurosurg 1984;61:241–53. or not to monitor? A review of our experience with severe head injury.
23. Lassen N. Cerebral blood flow and oxygen consumption in man. Physiol Rev J Neurosurg 1982;56:650–9.
1959;39:183–238. 46. Bouma GJ, Muizelaar JP, Choi SC, et al. Cerebral circulation and metabolism
24. Rosner M, Rosner S, Johnson A. Cerebral perfusion pressure: management after severe traumatic brain injury: the elusive role of ischemia. J Neurosurg
protocol and clinical results. J Neurosurg 1995;83:949–62. 1991;75:685–93.
25. Sahuquillo J. Does multimodality monitoring make a difference in 47. Robertson CS, Contant CF, Gokaslan ZL, et al. Cerebral blood flow,
neurocritical care? Eur J Anaesthesiol Suppl 2008;42:83–6. arteriovenous oxygen difference, and outcome in head injured patients.
26. Varelas PN, Conti MM, Spanaki MV, et al. The impact of a neurointensivist- J Neurol Neurosurg Psychiatry 1992;55:594–603.
led team on a semiclosed neurosciences intensive care unit. Crit Care Med 48. Symon L, Held K, Dorsch N. A study of regional autoregulation in the
2004;32:2191–8. cerebral circulation to increased perfusion pressure in normocapnia and
27. Kurtz P, Fitts V, Sumer Z, et al. How does care differ for neurological patients hypercapnia. Stroke 1973;4:139–47.
admitted to a neurocritical care unit versus a general ICU? Neurocrit Care 49. Murr R, Schurer L. Correlation of jugular venous oxygen saturation to
2011;Apr 26. Epub ahead of print. spontaneous fluctuations of cerebral perfusion pressure in patients with
28. Stein SC, Georgoff P, Meghan S, et al. Relationship of aggressive monitoring severe head injury. Neurol Res 1995;17:329–33.
and treatment to improved outcomes in severe traumatic brain injury. 50. Robertson CS, Gopinath SP, Goodman JC, et al. SjvO2 monitoring in
J Neurosurg 2010;112(5):1105–12. head-injured patients. J Neurotrauma 1995;12:891–6.
29. Spiotta AM, Stiefel MF, Gracias VH, et al. Brain tissue oxygen-directed
management and outcome in patients with severe traumatic brain injury. A complete list of references for this chapter can be found online at
J Neurosurg 2010;113:571–80. www.expertconsult.com.
30. Adamides AA, Rosenfeldt FL, Winter CD, et al. Brain tissue lactate elevations
predict episodes of intracranial hypertension in patients with traumatic
brain injury. J Am Coll Surg 2009;209:531–9.
Section I—Background 14.e1

References 24. Rosner M, Rosner S, Johnson A. Cerebral perfusion pressure: management


protocol and clinical results. J Neurosurg 1995;83:949–62.
1. Rosamond W, Flegal K, Furie K, et al. Heart disease and stroke statistics— 25. Sahuquillo J. Does multimodality monitoring make a difference in
2008 Update: a report from the American Heart Association Statistics neurocritical care? Eur J Anaesthesiol Suppl 2008;42:83–6.
Committee and Stroke Statistics Subcommittee. Circulation 2008;117: 26. Varelas PN, Conti MM, Spanaki MV, et al. The impact of a neurointensivist-
e25–146. led team on a semiclosed neurosciences intensive care unit. Crit Care Med
2. Hyder AA, Wunderlich CA, Puvanachandra P, et al. The impact of traumatic 2004;32:2191–8.
brain injuries: a global perspective. NeuroRehabilitation 2007;22:341–53. 27. Kurtz P, Fitts V, Sumer Z, et al. How does care differ for neurological patients
3. Wijdicks EFM, Hijdra A, Young GB, et al. Practice parameter: prediction of admitted to a neurocritical care unit versus a general ICU? Neurocrit Care
outcome in comatose survivors after cardiopulmonary resuscitation (an 2011;Apr 26. [Epub ahead of print]
evidence-based review): report of the Quality Standards Subcommittee of 28. Stein SC, Georgoff P, Meghan S, et al. Relationship of aggressive monitoring
the American Academy of Neurology. Neurology 2006;67:203–10. and treatment to improved outcomes in severe traumatic brain injury.
4. Bershad EM, Feen ES, Hernandez OH, et al. Impact of a specialized J Neurosurg 2010;112(5):1105–12.
neurointensive care team on outcomes of critically ill acute ischemic stroke 29. Spiotta AM, Stiefel MF, Gracias VH, et al. Brain tissue oxygen-directed
patients. Neurocrit Care 2008;9:287–92. management and outcome in patients with severe traumatic brain injury.
5. Suárez JI. Critical care neurology and neurosurgery. Totowa, NJ: Humana J Neurosurg 2010;113:571–80.
Press; 2004. 30. Adamides AA, Rosenfeldt FL, Winter CD, et al. Brain tissue lactate elevations
6. Ropper AH, Gress DR, Diringer MN, et al. Neurological and neurosurgical predict episodes of intracranial hypertension in patients with traumatic
intensive care. 4th ed. Philadelphia: Lippincott Williams & Wilkins; 2003. brain injury. J Am Coll Surg 2009;209:531–9.
7. Mayer SA, Coplin WM, Chang C, et al. Neurocritical Care Society; American 31. Chesnut RM, Marshall LF, Klauber MR, et al. The role of secondary brain
Academy of Neuorology Section on Critical Care and Emergency Neurology; injury in determining outcome from severe head injury. J Trauma
Society of Neurosurgical Anesthesia and Critical Care. Program 1993;34:216–22.
requirements for fellowship training in neurological intensive care: United 32. Oddo M, Schmidt JM, Carrera C, et al. Impact of tight glycemic control on
Council for Neurologic Subspecialties guidelines. Neurocrit Care cerebral glucose metabolism after severe brain injury: a microdialysis study.
2006;5(2):166–71. Crit Care Med 2008;36:3233–8.
8. Mayer SA, Coplin WM, Chang C, et al. Neurocritical Care Society; American 33. Oddo M, Milby A, Chen I, et al. Hemoglobin concentration and cerebral
Academy of Neurology Section on Critical Care and Emergency Neurology; metabolism in patients with aneurysmal subarachnoid hemorrhage: a
Society of Neurosurgical Anesthesia and Critical care. Core curriculum and microdialysis study. Stroke 2009;40(4):1275–81.
competencies for advanced training in neurological intensive care: United 34. Oddo M, Frangos S, Maloney-Wilensky E, et al. Effect of shivering on brain
Council for Neurologic Subspecialties guidelines. Neurocrit Care 2006;5(2): tissue oxygenation during induced normothermia in patients with severe
159–65. brain injury. Neurocrit Care 2010;12(1):10–6.
9. Suarez JI. Outcome in neurocritical care: advances in monitoring and 35. Robertson CS, Valadka AB, Hannay HJ, et al. Prevention of secondary
treatment and effect of a specialized neurocritical care team. Crit Care Med ischemic insults after severe head injury. Crit Care Med 1999;27:2086–95.
2006;34:S232–8. 36. Elf K, Nilsson P, Enblad P. Outcome after traumatic brain injury improved
10. Varelas PN, Eastwood D, Yun HJ, et al. Impact of a neurointensivist on by an organized secondary insult program and standardized neurointensive
outcomes in patients with head trauma treated in a neurosciences intensive care. Crit Care Med 2002;30:2129–34.
care unit. J Neurosurg 2006;104:713–19. 37. Markgraf C, Clifton G, Moody M. Treatment window for hypothermia in
11. Varelas PN, Schultz L, Conti M, et al. The impact of a neuro-intensivist on brain injury. J Neurosurg 2001;95:979–83.
patients with stroke admitted to a neurosciences intensive care unit. 38. Clifton GL, Choi SC, Miller ER, et al. Intercenter variance in clinical trials of
Neurocrit Care 2008;9:293–9. head trauma—experience of the National Acute Brain Injury Study:
12. Diringer MN, Edwards DF. Admission to a neurologic/neurosurgical hypothermia. J Neurosurg 2001;95:751–5.
intensive care unit is associated with reduced mortality rate after 39. Newberg AB, Alavi A. Neuroimaging in patients with head injury. Semin
intracerebral hemorrhage. Crit Care Med 2001;29:635–40. Nucl Med 2003;33:136–47.
13. Mirski MA, Chang CW, Cowan R. Impact of a neuroscience intensive care 40. Jacobs A, Put E, Ingels M, et al. Prospective evaluation of technetium-99m-
unit on neurosurgical patient outcomes and cost of care: evidence-based HMPAO SPECT in mild and moderate traumatic brain injury. J Nucl Med
support for an intensivist-directed specialty ICU model of care. J Neurosurg 1994;35:942–47.
Anesthesiol 2001;13:83–92. 41. Wintermark M, Chiolero R, Van Melle G, et al. Cerebral vascular
14. Berman MF, Solomon RA, Mayer SA, et al. Impact of hospital-related autoregulation assessed by perfusion-CT in severe head trauma patients.
factors on outcome after treatment of cerebral aneurysms. Stroke 2003;34: J Neuroradiol 2006;33:27–37.
2200–7. 42. The Brain Trauma Foundation. The American Association of Neurological
15. Wilby MJ, Sharp M, Whitfield PC, et al. Cost-effective outcome for treating Surgeons. The Joint Section on Neurotrauma and Critical Care. Indications
poor-grade subarachnoid hemorrhage. Stroke 2003;34:2508–11. for intracranial pressure monitoring. J Neurotrauma 2000;17:479–91.
16. Suarez JI, Zaidat OO, Suri MF, et al. Length of stay and mortality in 43. Becker DP, Miller JD, Ward JD, et al. The outcome from severe head injury
neurocritically ill patients: impact of a specialized neurocritical care team. with early diagnosis and intensive management. J Neurosurg
Crit Care Med 2004;32:2311–17. 1977;47:491–502.
17. Bernard SA, Gray TW, Buist MD, et al. Treatment of comatose survivors of 44. Ghajar JB, Hairiri RJ, Paterson RH, et al. Improved outcome from traumatic
out-of-hospital cardiac arrest with induced hypothermia. N Engl J Med coma using only ventricular CSF drainage for ICP control. Adv Neurosurg
2002;346:557–63. 1993;21:173–7.
18. Hypothermia After Cardiac Arrest Study Group. Mild therapeutic 45. Narayan RK, Kishore PR, Becker DP, et al. Intracranial pressure: to monitor
hypothermia to improve the neurologic outcome after cardiac arrest. N Engl or not to monitor? A review of our experience with severe head injury.
J Med 2002;346:549–56. J Neurosurg 1982;56:650–9.
19. Varelas PN, Spanaki MV, Hacein-Bey L. Documentation in medical records 46. Bouma GJ, Muizelaar JP, Choi SC, et al. Cerebral circulation and metabolism
improves after a neurointensivist’s appointment. Neurocrit Care after severe traumatic brain injury: the elusive role of ischemia. J Neurosurg
2005;3:234–6. 1991;75:685–93.
20. Helms AK, Torbey MT, Hacein-Bey L, et al. Standardized protocols increase 47. Robertson CS, Contant CF, Gokaslan ZL, et al. Cerebral blood flow,
organ and tissue donation rates in the neurocritical care unit. Neurology arteriovenous oxygen difference, and outcome in head injured patients.
2004;63:1955–7. J Neurol Neurosurg Psychiatry 1992;55:594–603.
21. Rosenthal G, Hemphill 3rd JC, Sorani M, et al. Brain tissue oxygen tension is 48. Symon L, Held K, Dorsch N. A study of regional autoregulation in the
more indicative of oxygen diffusion than oxygen delivery and metabolism in cerebral circulation to increased perfusion pressure in normocapnia and
patients with traumatic brain injury. Crit Care Med 2008;36:1917–24. hypercapnia. Stroke 1973;4:139–47.
22. Obrist WD, Langfitt TW, Jaggi JL, et al. Cerebral blood flow and metabolism 49. Murr R, Schurer L. Correlation of jugular venous oxygen saturation to
in comatose patients with acute head injury: relationship to intracranial spontaneous fluctuations of cerebral perfusion pressure in patients with
hypertension. J Neurosurg 1984;61:241–53. severe head injury. Neurol Res 1995;17:329–33.
23. Lassen, N. Cerebral blood flow and oxygen consumption in man. Physiol 50. Robertson CS, Gopinath SP, Goodman JC, et al. SjvO2 monitoring in
Rev 1959;39:183–238. head-injured patients. J Neurotrauma 1995;12:891–6.
14.e2 Section I—Background

51. Vajkoczy P, Roth H, Horn P, et al. Continuous monitoring of regional 59. Ståhl N, Mellergård P, Hallström A, et al. Intracerebral microdialysis and
cerebral blood flow: experimental and clinical validation of a novel thermal bedside biochemical analysis in patients with fatal traumatic brain lesions.
diffusion microprobe. J Neurosurg 2000;93:265–74. Acta Anaesthesiol Scand 2001;45:977–85.
52. Gopinath SP, Valadka AB, Uzura M, et al. Comparison of jugular venous 60. Reinstrup P, Ståhl N, Mellergård P, et al. Intracerebral microdialysis in
oxygen saturation and brain tissue Po2 as monitors of cerebral ischemia after clinical practice: baseline values for chemical markers during wakefulness,
head injury. Crit Care Med 1999;27:2337–45. anesthesia, and neurosurgery. Neurosurgery 2000;47:701–9.
53. Kiening KL, Härtl R, Unterberg AW, et al. Brain tissue pO2-monitoring in 61. Vespa PM, McArthur D, O’Phelan K, et al. Persistently low extracellular
comatose patients: implications for therapy. Neurol Res 1997;19:233–40. glucose correlates with poor outcome 6 months after human traumatic brain
54. Siesjö BK. Pathophysiology and treatment of focal cerebral ischemia. Part II: injury despite a lack of increased lactate: a microdialysis study. J Cereb Blood
mechanisms of damage and treatment. J Neurosurg 1992;77:337–54. Flow Metab 2003;23:865–77.
55. Clausen T, Alves OL, Reinert M, et al. Association between elevated brain 62. Scheuer ML. Continuous EEG monitoring in the intensive care unit.
tissue glycerol levels and poor outcome following severe traumatic brain Epilepsia 2002;43(Suppl 3):114–27.
injury. J Neurosurg 2005;103:233–8. 63. Vespa PM, Boscardin WJ, Hovda DA, et al. Early and persistent impaired
56. Meixensberger J, Kunze E, Barcsay E, et al. Clinical cerebral microdialysis: percent alpha variability on continuous electroencephalography monitoring
brain metabolism and brain tissue oxygenation after acute brain injury. as predictive of poor outcome after traumatic brain injury. J Neurosurg
Neurol Res 2001;23:801–6. 2002;97:84–92.
57. Hutchinson PJ, Gupta AK, Fryer TF, et al. Correlation between cerebral 64. Vespa PM, Nuwer MR, Nenov V, et al. Increased incidence and impact of
blood flow, substrate delivery, and metabolism in head injury: a combined nonconvulsive and convulsive seizures after traumatic brain injury as
microdialysis and triple oxygen positron emission tomography study. J Cereb detected by continuous electroencephalographic monitoring. J Neurosurg
Blood Flow Metab 2002;22:735–45. 1999;91:750–60.
58. Vespa P, Martin NA, Nenov V, et al. Delayed increase in extracellular glycerol
with post-traumatic electrographic epileptic activity: support for the theory
that seizures induce secondary injury. Acta Neurochir 2002;81(Suppl.):355–7.
Chapter
3  
I

Designing the Neurocritical


Care Unit for Better Patient Care
Mahbub Rashid, Craig Zimring, and Owen B. Samuels

the layout include: (1) direct monitoring of patients by clinical


Introduction staff, (2) an outside window for each patient room, and (3)
There is growing evidence that the physical design of clinical reduction of cross-infection, traffic volume, and noise level,
spaces affects the quality, efficiency, and experience of care in among other factors.
intensive care units (ICUs) and other settings.1-4 These effects
are complex. In some cases the design directly leads to out-
comes that might have clinical significance, for example, Monitoring
whether improved air filtering reduces airborne pathogens. In In NCCUs, patients need constant visual monitoring because
other cases the environment contributes to noise or lighting their conditions may change quickly and unpredictably, and
that can increase or decrease stress for patient, families, or these changes need to be recognized early. NCCU patients also
staff. In yet other cases, the environment contributes to staff require frequent neurologic assessments or bedside proce-
or patient behavior that may affect care. For instance, if staff dures that may involve multiple members of a care team. A
see and encounter each other informally over the course of physical layout that supports effective face-to-face interaction
their day, they may better coordinate care,5 or if they see hand in a patient’s room and within the unit is thus required. In a
hygiene sinks or rubs, they may increase their compliance with teaching hospital the rooms or corridors also need to support
handwashing.3 the large number of participants who make patient rounds.
This chapter examines the effects of the ICU design and
particularly the design of the neurocritical care unit (NCCU)
on patient care. Addressed are the important areas within the Windows
ICU, key design considerations, main design guidelines, and U.S. law requires that ICU patients must have direct access to
emerging evidence on how ICU design influences patient care. natural light. This same requirement also applies in several
The chapter ends with a case study in which the authors were other countries (e.g., India, Saudi Arabia, and the United
involved, from the perspective of the medical director. Arab Emirates). The number and arrangement of patient
rooms consequently depend on the amount of peri­meter wall
available in the unit. Therefore a review of best practice
Evidence-Based Intensive Care examples shows that designers often select compact shapes
with high area-to-perimeter ratios to accommodate the
Unit Design maximum number of patient rooms for any given area. They
Research in ICU environmental design is an emerging field of also put most support areas, including nurse work areas, in
study, and therefore high-quality research articles are still the core that do not require any outside window to reduce the
limited in number. In addition, many confounding variables walking distance between clinical support areas and patient
can influence the outcome of ICU design research. Therefore rooms.6
along with the findings of ICU design research, the authors
also present expert opinions and findings of research studies
in other health care settings that may be relevant to ICU Limiting Infection, Traffic, and Noise
design. This section reviews the primary functional and pro- Cross-infection, traffic volume, and noise level often may help
cedural issues associated with ICU design and addresses the shape unit configurations, and each of these factors is of
key areas of the ICU. greater significance in larger hospital units. Studies suggest
that patients in larger units have greater risk of hospital-
acquired infection.7,8 Larger units also have more traffic and
Unit Layout noise sources. These noise sources commonly include noises
Unit layout defines the size and location of functional areas of other patients (e.g., snoring, crying), monitor alarms, tele-
and their relationships within a unit. Major determinants of phone rings and conversations, conversations among staff,
© Copyright 2013 Elsevier Inc. All rights reserved. 15
16 Section I—Background

staff entering or leaving, staff wandering, sudden voices, foot- that matches their level of acuity.17-19 This rate may be higher
steps, falling objects, noises of respirators, doors closing, and for NCCU patients. Delays, communication discontinuities,
visitors talking, among others.9 Breaking a larger unit into loss of information, and changes in computers and systems
smaller units, pods, or clusters may reduce infection and noise. during patient transfer can contribute to increased medical
However, pods can break down the visual and social cohesive- errors and loss of staff time and productivity.18,20 Patients can
ness of a unit, and multiple pods may make movement of get hurt, and most nurses’ back injuries occur during patient
supplies difficult because they create more service stops. Thus transfer (see the following text). Thus design interventions
the appropriate configuration for a large ICU remains a matter must include factors that help reduce the time and effort
of striking the right balance among various contradictory involved in patient transfer.
factors.
Patient Room Design
Unit Size A patient’s room is the basic working unit of an ICU, and it
According to the Society of Critical Care Medicine’s Guide- affects patient care including safety, privacy, and comfort. For
lines for Intensive Care Unit Design (henceforth, the Guide- example, nurses can save multiple trips to nursing stations or
lines), 8 to 12 beds per unit are considered best from a storage rooms if a patient’s room includes a space for chart-
functional perspective.10-13 In one survey, a group of ICU ing or storage space for supplies. A longer distance between
experts also suggested that the ideal number of patient beds the bed and the toilet can increase physical stress for a nurse
in an ICU should be 9 or 10, and no less than 6.14 An NCCU who needs to help the patient to the toilet. If there is a well-
with fewer than 6 beds may be inefficient to operate and defined area within the room for families, they can be present
manage. In contrast, in a very large unit without proper unit for patient comfort and help caregivers by providing infor-
design and a sufficiently large nursing staff, patient monitor- mation and assistance with care. Important design consider-
ing and care may become difficult. ations of patient rooms in the NCCU, similar to other ICUs,
The total gross area of a unit, another indicator for unit include to: (1) create well-defined functional zones to elimi-
size, is somewhat related to the number of beds in the unit— nate conflicts, (2) provide enough space that is appropriate
the more beds the greater the ICU gross area per bed.6 The for patient care, (3) provide necessary life support systems,
amount of circulation spaces, another determinant of the (4) balance visibility and privacy, (5) provide toilet facilities,
gross area of a unit, is an important indicator of the square (6) reduce hospital-acquired infections and psychosis among
footage efficiency of an ICU. Circulation spaces within an patients, and (7) reduce patient and staff injury.
ICU consist of internal hallways, corridors, or aisles used by
all ICU users—patients, ICU staff, and visitors. Like any
other facility, an ICU may be inefficient with too much or too Patient Room Layout
little circulation space. Circulation spaces are sociologically The patient room layout defines the size and location of func-
important because they determine the interconnectedness of tions and their relationships within the room. It affects how
people and functions within a facility. A narrow corridor functions are performed and how patients and caregivers
within an ICU can impede transfer of knowledge as much as interface in the room. Jastremski and Harvey suggest that an
it can impede the flow of goods and people. Properly designed ideal room should have three zones: a patient zone, a family
circulation spaces in ICUs may hold a great potential to zone, and a caregiver zone.21 Hamilton and Shepley22 defined
enhance the transfer of tacit knowledge through face-to-face four different zones within a patient’s room: (1) patient, (2)
interactions.15 hygiene, (3) staff, and (4) family zones. The patient zone
In the United States, the National Fire Protection Associa- includes the bed, bedside, and overbed tables, and the imme-
tion’s Life Safety Code also affects ICU size and design. The diate area occupied by clinicians when they provide care. The
Life Safety Code limits the size of any suite to 5000 square feet hygiene zone includes the patient’s toilet, sink, and activities
if it does not have intervening smoke partitions and fire-rated associated with hygiene. The staff zone includes the area just
doors.16 The area per bed of a unit ranges from 650 to 1200 inside or outside the entry to the patient’s room to support
square feet or more depending on function on the unit, so nursing and caregiver functions; this may include a writing
most units require smoke partitions or “hold open” smoke surface, provisions for hand hygiene, patient information,
doors. Hospitals and designers need to ensure that when any medication, and supplies. The family zone may include seating
one part of an ICU larger than 5000 square feet becomes or provisions for overnight stay, storage space, separate light-
unavailable in the event of fire or smoke breakout, the other ing, Internet access, and a writing surface, among others.
parts of the unit have the required components for patient and Defining patient room in terms of what is in a patient’s view
staff safety. and what is not can be important. Evidence suggests that
patients lying on an ICU bed are stressed when they see
medical equipment, accessories, and monitors.23 Therefore it
Unit Location is reasonable to keep these devices away from the patient’s
Important departmental relationships of the unit should be view, and instead put family space within the patient’s view.
carefully considered during the site selection process. Conve- Architectural treatment of areas within a patient’s view also
nient physical movement across departments may help reduce can be important because it appears that “positive distrac-
many safety risks associated with patient transfer. In general, tions” such as nature can reduce stress and pain3 and that
patients are transferred from one place to another as often as natural light can reduce analgesic use.24 Soothing color and
three to six times during their hospital stay to receive the care light, natural materials, and paintings of nature may be used
Section I—Background 17

in the area within the patient’s view to make his or her experi- and still maintain maximum visibility of patients and moni-
ence more comfortable. tors. Breakaway glass doors also allow maximum clearance to
move patients in and out of the room. In an emergency, break-
away glass doors allow the room to become more open than
Patient Room Size the other doors. However, with breakaway doors closed it may
The need for larger patient rooms is increasing in ICUs. Advo- be difficult to hear patient alarms, for example, ventilator
cates of infection prevention recommend that each patient alarms.
room should have dedicated patient care equipment to reduce
cross-infection with more resistant microbial strains. Others
also recommend that each room should have a dedicated Life Support Systems in a Patient Room
family space, with amenities to improve family integration Easy access to the patient’s head and the ability to move a
with patient care. As medical breakthroughs and advance- patient bed around during procedures are important in the
ments occur, more technology is brought into patient rooms, NCCU. Traditional head wall systems that have been used
and this requires more space. The increasing multidisciplinary since the 1970s to provide the life-support systems do not
nature of patient care in the NCCU also requires patient allow easy access to the patient’s head, nor do they allow clini-
rooms to accommodate larger medical teams. Additional cians to reorient the bed when needed. Headwall systems
space also may be needed in patient rooms to support research include power outlets and outlets for medical gases and
and the increasing number of procedural interventions that vacuum on one or both sides of a patient bed. Some installa-
now are performed in ICUs, such as bronchoscopy, echocar- tions also include wall-mounted monitors and equipment for
diography, and placement of external ventricular drains. a patient’s vitals.
The Guidelines stipulate, “Ward-type ICUs should allow at Power columns, whether rotating or static, are now used in
least 225 square feet of clear floor area per bed. ICUs with many ICUs to provide life support systems. Equipped with
individual patient modules should allow at least 250 square medical utilities, power outlets, and monitors, these power
feet per room [assuming one patient per room]. …”12 In a columns allow easy access to the patient’s head and may allow
survey of the best-practice ICUs in the United States built clinicians to reorient the bed. Two, instead of one, power
between 1993 and 2003, the average size of a patient room was columns—one on each side of the bed—can be installed to
250 square feet.6 However, since 2000, many best-practice help increase the symmetry of functions around the patient
ICUs have also used more than 250 square feet for patient bed. However, it can be difficult to work around power
rooms,25 suggesting that hospitals are aware of trends such as columns during a procedure. Other innovations—the ceiling-
the demand for family spaces in patient rooms. mounted boom, ceiling columns, or the Draeger Ponta
beam—help provide easy access and sufficient flexibility for
proper patient care in NCCUs and in particular provide
Privacy and Visibility in a Patient Room access to the patient’s entire body, especially to the head.
Privacy is an important factor associated with individual sat- These ceiling-mounted systems, however, add cost and often
isfaction in many environments including hospitals and even require additional structural support. These systems occa-
in moments of extreme crisis, privacy may be required to sionally can conflict with a patient lift system in an NCCU
preserve individual dignity. Patient and family surveys suggest patient room.
hospitals with more private rooms tend to have higher patient
satisfaction rates (www.pressganey.com). There are several
advantages to private rooms. First, they provide dedicated Toilets in a Patient Room
space for individualized care without disturbing other patients ICU patient rooms are not required to have toilets, but there
and can help reduce noise, improve patient sleep quality, and are many good reasons to have a toilet in the room for both
support staff-patient communication.21,26 Second, hospital- the patient and family. Toilets in patient rooms can be used to
acquired infection rates in ICUs with private rooms are less dump and clean bedpans, which can be a major source of
because of improved airflow, better ventilation, and more aerosol contaminants and infectious organisms,33 and con-
accessible handwashing facilities.1,27-30 Finally, the private tained systems often are used to reduce aerosols. The location,
room design allows for a patient care environment with more use, and design of a toilet or of other devices to eliminate
control over optimal environmental conditions.31 However, in waste have a significant effect on ICU design. A review of
private rooms patient visibility is often at a stake and patients current design practice shows that toilets are placed in many
are afraid of being left alone. From a clinical viewpoint it is different locations in relation to a patient room. Inboard
easier to make a case for open patient rooms in ICUs; this toilets are on the corridor side of patient rooms; outboard are
permits easy visual monitoring of the patient by the clinical on the window side. A third option is to place the toilets of
staff that can affect patient safety. ICU staff often prefer an two adjacent patient rooms next to each other in a wet zone.
open ICU to see everything that happens, to readily seek help Each location has advantages and disadvantages.6
from others in a crisis, and to act immediately in groups
without the constraints of walls of a private patient room.32
In private patient rooms, where glass often is used for patient Hospital-Acquired Infections
visibility, measures to ensure visual and acoustic privacy of In patient room design, air quality, single-bed patient rooms,
patients and their families when needed also are necessary. lighting conditions, noise level, and handwashing sink are
Hospitals often prefer breakaway glass doors because they can important because they can help reduce infections among
be closed for privacy, noise reduction, and infection control patients. Most studies show that (1) private patient rooms can
18 Section I—Background

reduce cross-infection among ICU patients; (2) single-bed hospitals include telephones, alarms, trolleys, ice machines,
patient rooms with high-quality high-efficiency particulate paging systems, nurse shift change, staff caring for other
air (HEPA) filters and with negative- or positive-pressure patients, doors, staff conversations, and patients crying out or
ventilation are more effective in preventing airborne patho- coughing.54,55 Hospital noise can be improved if proper design
gens; and (3) multibed rooms are more difficult to decon- and management measures are in place.
taminate and have more surfaces that act as a reservoir for
pathogens.2,30,34 The 2006 American Institute of Architects
Guidelines for Design and Construction of Healthcare Facili- Reducing Patient Falls and Fall Severity
ties therefore has adopted the single-bed room as the stan- The physical environment can be a root cause for patient
dard for all new construction in the United States.35 falls.56 Among specific interior design elements, flooring can
Infrequent handwashing by health care staff is associated contribute to the incidence of falls and the severity of injury
with hospital-acquired infections.36 Several design factors from a fall.57 Patients may suffer more injuries when they fall
may discourage handwashing, including: (1) poor sink loca- on vinyl floors than carpeted floors.58 Subfloors also may
tion, (2) poor visibility, (3) uncomfortable sink height, and influence the injury from falls; the risk of fracture is less for
a (4) lack of redundancy and wide spatial separation of wooden than concrete subfloors.59 Several design factors can
resources that are used sequentially in handwashing.36-39 help reduce the incidence of falls including decentralized
There are conflicting results on how physical design influ- observation units next to patient rooms rather than a central-
ences handwashing compliance.40-43 There is, however, a con- ized nursing station,17 patient lifts and other transfer devices,
sensus that a multi-strategy intervention that includes staff and innovative acuity adaptable patient rooms.
education, easy visual and physical access to sinks, standard
sink locations in all patient rooms, comfortable sink heights,
and alcohol-based dispensers can help increase handwashing Other Patient and Staff Injuries
compliance.36,37 Manual lifting of ICU patients poses a high risk of injury for
A review of current design practice shows that handwashing patients, such as dislodgement of invasive tubes and lines,
sinks are placed at many different locations in a patient room, shoulder dislocation, fracture of fragile bones, or being
including: (1) locations directly outside the room entrance- dropped.60 Skin tears and abrasions also may occur when
way, (2) immediately after the entranceway either on the foot- patients are pulled up or across beds, and manual patient
wall or on the head wall, (3) somewhere in the middle of the handling can contribute to pain in critically ill patients. Pain
footwall, and (4) at the far end of the room walls. Designers experienced by these patients during turning or repositioning
and hospitals must consider the advantages and disadvantages sometimes is greater than that experienced during tracheal
of each of these locations.6 (For a discussion on this topic, see suctioning, tube advancement, and wound dressing changes.60
Rashid.6) Staff also are at risk for work-related injuries when moving
patients. The use of ceiling lifts can help limit risks associated
with manual moving to both patient and staff. If possible, lifts
Psychosis Among Intensive Care should extend into the toilet room.3 If patient volumes and
Unit Patients staffing patterns allow, ICU rooms that can flex in acuity from
ICU patients who are confined to bed may suffer from delir- super-acute to step-down also may reduce patient transfers
ium, also known as “ICU psychosis.”44 There is growing evi- and discontinuity in care.
dence from non-ICU settings that both natural light and
outdoor views can help patients maintain sensory orientation
and circadian rhythm.45-49 This in turn can reduce the likeli- Staff Work Areas
hood of delirium. Consequently, the Guidelines recommend:
“Windows are an important aspect of sensory orientation,
and Support Spaces
and as many rooms as possible should have windows to rein- ICUs can be stressful workplaces that can endanger the physi-
force day/night orientation. … If windows cannot be pro- cal and mental health of the clinical staff. In the United States
vided in each room, an alternate option is to allow a remote there is a shortage of critical care nursing staff, and the current
view of an outside window or skylight.”12 Artificial lighting staff is older and has a high turnover rate. Staff turnover rate
conditions that mimic the variations in natural light also can in many institutions is greater in ICUs than other wards
help patients maintain sensory orientation and circadian because of the stressful working conditions. ICU design,
rhythm. Looking at a ceiling or a wall painted in solid colors however, can help relieve staff stress by reducing unnecessary
for a long period also may contribute to patient delirium. physical labor, by providing amenities, and by providing posi-
Warm colors and paintings of nature may improve the envi- tive distractions for staff physical and mental recovery.
ronment. Sometimes a calendar that shows the date or a
digital clock that shows date and time may help patients
adjust their internal physiologic rhythm. However, the monot- Staff Work and Support Area Location
onous clicking sound of an analog clock can cause distress in and Layout
ICU patients. There are three basic nursing unit configurations: (1) central-
ICU psychosis also may be associated with a high level of ized nursing station, (2) nursing substation, and (3) nursing
noise.50,51 Noise level in the ICU ranges from 50 to 75 dB, with observation unit. In older hospital units a centralized nursing
peaks up to 85 dB.52 This is much higher than World Health station is generally the main component of the staff work area.
Organization recommendations for noise levels in hospital Along with the patients’ records room, the central monitoring
patient rooms and units.53 Common sources of noise in station, and staff workstations for patient charting and medical
Section I—Background 19

recording, it serves as the hub of all unit functions. This type Nurses may spend between 15% and 25% of their time in
of nursing station usually has a centralized support and service charting.67-69 Computers for charting may be found at the
area next to it that accommodates medical and supply storage, nursing observation units next to the patient room, in the
pharmacy, conference rooms, administrative offices, and other patient room, on mobile carts, and/or at the central nursing
ancillary functions. In older units a centralized nursing station stations away from the patient room. When charting is not
is where clinical management, staff interaction, mentoring, done at or near the bedside nurses may spend more time away
and socialization occur. However, centralized nursing stations from the bedside to prepare and store charts at other locations
may contribute to errors and inefficiency because of noise, or make more mistakes in charting because of memory lapses
crowding, and considerable walking distance from patient between the time of information collection at the bedside and
rooms. putting it on the chart.
In hospital units built in the 1990s and 2000s, the central- Several factors contribute to staff stress in the ICU. Among
ized nursing station often is replaced with several decentral- them is noise that can be associated with emotional exhaus-
ized observation units with direct observation of one or two tion and burnout among ICU nurses.70,71 Excessively high
rooms and located either just outside or inside the patient noise levels in healthcare settings and frequent interruptions
room. Typical functions include a work surface for patient also can interfere with work.72 For example, in a recent time-
charting, a computer to record and access patient informa- and-motion study that included 40 doctors for more than 210
tion, and telecommunication services. There may or may not hours, Westbrook et al. observed that interruptions led doctors
be storage spaces for medication and supplies, handwashing to spend less time on the tasks they were working on and, in
facilities, and image retrieval systems. It is suggested that the nearly a fifth of cases, to give up on the task.73
decentralized observation units increase efficiency, but Patients in NCCUs often have numerous monitoring and
perhaps at the cost of staff “social life.” Team workstations, recording devices with alarms that may also contribute to staff
sometimes distributed throughout a unit, provide spaces for stress and fatigue. Nurses may become insensitive to alarms,
interdisciplinary teamwork, mentoring, clinical management, because every alarm may not need immediate attention. There-
and social functions that may not be feasible in the totally fore they can miss that important alarm which requires imme-
decentralized observations units. diate attention. For example, an investigation by the Boston
Hospitals may use different combinations of the basic Globe suggested that 216 deaths from 2005 to 2010 nationwide
nursing unit configurations in ICUs.6 The relation of the were associated with monitor alarm problems.74 Device-related
support or service areas to nursing units differs from unit to incidents may be underreported and it is possible that the
unit. In an exploratory study Zborowsky et al.61 investigated number of adverse events associated with alarm problems is
how nursing station design (i.e., centralized and decentralized higher. According to the Globe’s investigation, the deaths listed
nursing station layouts) affected nurses’ use of space, patient in the U. S. Food and Drug Administration (FDA) data were
visibility, noise levels, and perceptions of the work environ- linked to problems with alarms on patient monitors that track
ment. They concluded that the “hybrid” nursing design model heart functions, breathing and other vital signs. The problem
in which decentralized nursing stations are coupled with typically was not a broken device. Instead, most cases occurred
centralized meeting rooms for consultation between staff because medical staff did not notice an alarm or react with
members may strike a balance between the increase in com- urgency. This includes not hearing the alarm, ignoring an
puter duties and the ongoing need for communication and alarm, mis­programming complicated monitors or forgetting
consultation that addresses the conflicting demands of tech- to turn them on.
nology and direct patient care. In another recent study, Hen- The interface between technology and patients also can
drich et al. examined how nurses adopt distinct movement challenge NCCU staff and contribute to stress. Often, patient
strategies based on features of unit topology and nurse assign- monitor and access lines are built up with multiple ports and,
ments and observed that the spatial qualities of nurse assign- even with proper training and instructions it may difficult to
ments and unit layout affect nurse strategies for moving sort out where to put this or that connector. ICU nurses also
through units and affect how frequently nurses enter patient need to ensure that medications are compatible and so need
rooms and the nurse station.62 Therefore how various design to know how to control the stopcock. It is expected that tech-
features of nursing unit and support space configuration nology innovations will help overcome some of the these
affect staff stress and effectiveness, staff communication, and interface related problems, e.g., each port can be made unique
task performance meets the needs of a particular ICU need to to help eliminate mixing up of lines, devices can be created to
be considered. help identify incompatible drugs or integrated printers in
every patient room for prescriptions may help eliminate
medical errors. Together, several simple fail-safe devices may
Staff Stress and Effectiveness remove some of the cognitive load that contributes to stress
The location of supplies and equipment and of electronic among nurses.75 For example, Kobayashi et al. used simple
charting impact the time spent in patient care by nurses. human factors engineering principles to develop an experi-
Nurses spend a lot of time walking, which includes the time mental chart binder system with alternating color-based
to locate and gather supplies and equipment and to find other chart groupings, simple and prominent identifiers, and
staff members.63,64 Studies suggest that bringing staff and sup- embedded visual cues.76 This was associated with a significant
plies physically and visually closer to the patient reduces the reduction in chart binder location problems, so contributing
time nurses spend walking about the unit.65,66 Decentralized to safe patient care delivery. Similarly Blomkvist et al.,77 who
nurses’ stations and supplies’ servers next to patient rooms examined the effects of changing the acoustic conditions
allows nurses to spend more time in direct patient care (sound-absorbing versus sound-reflecting ceiling tiles) in a
activities.25-27 coronary ICU, observed that there were positive staff
20 Section I—Background

outcomes, including improved speech intelligibility and self-esteem, and (5) enhance positive relationships by offering
reduced perceived work demands, pressure and strain during love and comfort.97 In addition, families can help busy nurses
the periods of improved acoustic conditions. and physicians.98,99

Staff Communication Location of Family Space


Staff communication is a major variable in health care and The location of family waiting spaces in ICUs has both prac-
ICU safety and several studies demonstrate that a high degree tical and symbolic importance. Having family members
of involvement and interaction among caregivers can influ- nearby often helps to shift an ICU’s culture and make fami-
ence patient outcomes78 and length of stay.79 For example, lies a greater part of decision making (though sometimes this
Baggs et al.80 examined interdisciplinary collaboration and also requires clinicians to create structured ways of dealing
patient outcomes in a medical ICU and observed that patients with stressed family members). Symbolically the presence of
had a 5% chance of death or readmission where nurses family spaces located outside the unit may suggest that fami-
believed they had worked successfully with medical residents. lies are not integrated with patient care, whereas those pro-
The risk was tripled when the residents made decisions about vided within the patient room may indicate that families are
patient care without adequate nurse consultation. The value integrated with patient care. Depending on their accessibility
of better communication is strongest in the very sick, complex and comfort, family spaces provided within the unit but not
patients.81 Research in “Magnet” hospitals also indicates that in the patient room can suggest the family role is in a state of
healthy collaborative relationships among caregivers are pos- flux in the unit. These impressions can, at times, be wrong.
sible and appear linked to optimal patient outcome.82 A Some units may still not allow families to play an active role
growing body of literature in several environmental design in patient care even with families present in the patient
research areas including offices, laboratories, housing com- room. Hospitals should consider providing family spaces at
plexes, and acute care health facilities show that the physical several of these locations for different functions and ameni-
design of an environment may affect communication, interac- ties.6 (For a discussion on the possible sociologic implica-
tion, and/or or collaboration (For a review of the literature, tions of different locations of family spaces in ICUs, see
see Ulrich et al.3; Rashid83; Elsbach and Pratt84; Rashid and Rashid, 2006.6)
Zimring.85) When designing staff areas in ICUs, the following
should be considered: (1) Physical design can affect the quality
and the quantity of interaction.86 (2) Location of people and Family Waiting Area Layout
activity and physical distance among workers may be linked Family waiting areas may be broken down into zones with
to their informal communication.87 (3) Proximity of work- varying degrees of privacy and control similar to a home. The
spaces may predispose to the development of an informal number of ICU patient rooms, the availability of family space
group among compatible people as an outgrowth of the infor- in the patient room, the average length of patient stay, and the
mal communication associated with proximity.87 (4) Spatial types of amenities are some of the issues to consider to deter-
arrangement including the location of functions, walls, parti- mine the size of a common waiting space. Waiting areas should
tions, furnishings, and other barriers may affect cohesiveness be divided into sections to provide more intimate and quieter
and interaction among groups.88 (5) Visibility and accessibility resting spaces and relatively busy and noisy activity spaces.
play a powerful role in the way individuals perceive and use Solid partitions, dividers, glass walls, or planters can separate
workplaces and communicate within.89 (6) Time spent in each section according to the need of these spaces. Each
walking by staff may be related to time spent in patient care section should contain comfortable chairs or sofas for the
activities including nurse-physician interactions.17 family, and resting and activity spaces should include private
spaces or booths for telephone conversations. Activity spaces
should include: (1) computers with Internet access that allow
Lighting Conditions and Task Performance families to access these computers to stay up-to-date with
There are few studies on the effects of lighting conditions on patient status and the outside world and (2) study carrels with
task performance among ICU staff. Studies in other work set- health care information for families. There should be a media
tings show that staff may make more mistakes in inappropri- room to separate the television from the rest of the waiting
ate lighting conditions, and performance on visual tasks gets area, so families who wish to have quiet and solitude are not
better as light levels increase.90 For example, medication dis- disturbed. Families with children should be given spaces sepa-
pensing errors among hospital workers are more frequent rate from a “quiet zone” for adults only. A play area for chil-
when daylight hours are fewer91; these errors can be reduced dren within the direct visual reach of the adult family members
at an illumination level greater than the baseline level of 45 should be considered in the area designated for families with
foot candles.92 Studies in offices also indicate the importance children. Some hospitals provide family sleep rooms with
of appropriate lighting levels for complex tasks that require private bathrooms, kitchenettes, and laundry in their waiting
excellent vision.93 areas for family members who must stay at the hospital for an
extended period.
Spaces for Families
Many patients in the NCCU cannot communicate for them- Family Space in Patient Rooms
selves. Therefore family members have an important role as When possible, each patient room should include a well-
surrogate decision makers.94-96 Family members can also help defined family area to allow families to be present for shift
patients (1) perform daily functions, (2) understand concerns change report, teaching sessions, care-planning discussions,
about health, (3) foster a link to the environment, (4) reinforce and daily medical rounds. A family space within a patient’s
Section I—Background 21

room provides families a more comfortable environment for high-performance sound-absorbing materials can be used to
activities, and being able to sleep in the patient’s room pro- reduce reverberation time, sound propagation, and noise
vides social support and reassurance for the patient and family intensity levels. Storage areas, staff lounges, and utility rooms
members. also can be located away from patient rooms and family spaces
to reduce noise. Internal corridors between storage and utility
rooms can help clinical and support staff members perform
Furnishings and Finishes necessary tasks without disturbing patients or families.
Furnishings in family areas should include comfortable seating
for the family and the option of a wall-mounted fold-down
bed or foldout chair-bed. Flexible furniture arrangements that Music in Family Space
allow families to change furniture layout to meet their needs Sometimes music can be used to mask distressing environ-
are preferable, because seating arrangements that cannot be mental noise that cannot otherwise be eliminated. Sounds of
changed or chairs that cannot be moved may cause frustration nature accompanied by soft music also can be used in family
among families. Unnecessary sources of visual stimulation waiting areas to calm anxious families or visitors. However,
should be minimized and wall furnishings should not be of not all music can produce a desired calming effect. Music
bold patterns or colors that can be misperceived as threatening often evokes emotions and feelings that are rooted in an indi-
objects (e.g., bugs, animals) by patients or their families. Wall vidual’s past experiences and personal preferences.108 Thus it
coverings and colors should be soothing and relaxing. In is essential to respect music preferences of family members
general, the attractiveness of the physical environment in and provide them choices.
waiting areas has been shown to be significantly associated
with higher perceived quality of care, less anxiety, and higher
reported positive interaction with staff.100 Artwork in Family Space
Appropriate artwork can help reduce stress among patient
families.109 The choice of artwork needs to be sensitive to
Access to Patients and Caregivers culture, religion, the specific geographic area, and the interior
Family spaces should provide easy visual or physical access to design scheme. Hospitals should consider developing systems
patient rooms so family members can see the patient. Families that allow artwork to be changed by the patient family as easily
also should have easy access to caregivers when needed and as changing television channels. Because art varies enormously
know when caregivers are available in the unit. For patient in subject matter and style, not all artwork is suitable for high-
safety it is better for families to enter the unit through a sepa- stress health care spaces.
rate entry other than the one used by service and clinical staff.
Designers and hospitals need to ensure that such a system of
entrances does not make interfaces among families and care- Lighting in Family Space
givers difficult. If the ICU design restricts family-caregiver Appropriate lighting can influence mood or create a relaxed
interfaces, families may gather at places where they are likely ambience. Therefore attention should be given to make
to find caregivers. natural light available in all family spaces in the unit. When
the family space is within the patient room, it is necessary to
make sure that the intensity of natural light is comfortable
Staff-Family Communication to patients. The use of slightly tinted or reflective glass can
In ICUs, staff-family communication can provide emotional, reduce glare and heat production from sunlight. Vertical
informational, and tangible supports to family members, and blinds and other window treatments can be used to adjust
can facilitate family members’ involvement in patient care. light intensity as desired by the patient. For artificial light in
Hospitals should consider the following to help improve family spaces, it is important to consider multiple lighting
family-staff interactions and communication. (1) Central options that can be controlled by the family when appropri-
nursing stations and glass partitions around the staff area can ate. Where natural light is not an option (e.g., older ICUs),
limit family access to staff. (2) Decentralized nursing stations full-spectrum fluorescent lighting can be used for compara-
may provide more opportunities for a nurse to spend time in ble benefits. A dynamic lighting solution that allows the
patient rooms. These stations also can be used during medical color and temperature levels to be changed according to the
rounds by medical teams to retrieve patients’ records. (3) time of day may be suitable for a patient room or family
Private patient rooms and well-designed consultation rooms space.
may provide opportunities for confidential discussions. (4)
Within hallways, alcoves can provide private spaces for confi-
dential discussions. (5) Seating that is arranged side-by-side Odors and Aromas
along family space walls can discourage social interaction. (6) Pleasing aromas can help reduce blood pressure, slow the rate
Private and peaceful spaces can help improve communication. of respiration, lower pain perception levels, improve the
(7) In dim lighting conditions and in rooms with softer floor immune system, and help increase a sense of well-being
materials, people may interact longer.101-107 among family members who are under severe mental and
physical stress. In contrast, odors (“negative smells”) may
stimulate anxiety, fear, and stress.110 Hospitals, particularly
Noise Reduction in Family Space ICUs, are well known for their unpleasant odors or chemical
Carpeting in corridors next to family waiting spaces can smells. If possible, strong-smelling cleaning agents should be
reduce the sound of footsteps, rolling carts, staff member avoided near family areas. Aroma should be used with caution
conversations, and other common noises in ICUs, and in ICU family areas.
22 Section I—Background

high-quality care. Consequently a larger NCCU was proposed,


Nature, Spirituality, and Religion initially to appeal to the bottom line: if the unit had more beds
Nature can have a positive effect on physical and emotional and could attract more patients, it could generate more
well-being; hence it is preferable to design family areas with revenue and, most important, fulfill staff ’s mission as the hos-
windows to the outside. If possible these spaces need to be pital of last resort for many of these patients with complex
close to hospital gardens with plants, water, and other natural brain injury. In 2007 Emory Medical Center was in the early
objects. When family spaces do not have a view of or access to phase of planning a replacement hospital, and the administra-
nature, nature may be brought into the unit (e.g., potted tion did not want to spend a lot of money on a new ICU that
plants, sound of nature, and nature-related artwork). Hospi- would be demolished in 5 years when the new hospital opened.
tals also should consider outdoor labyrinths in gardens as a Early on, the new NCCU was described as the “throwaway”
focus for spirituality.111-113 NCCU, and the hospital agreed to meet the state and federal
A working knowledge of common cultural and religious requirements so that it could quickly open to meet the imme-
needs of patients and families is required to design family diate ICU bed shortages.
spaces. Sometimes a focus group with local spiritual and The initial design was for a 24-bed ICU with a “track”
ethnic leaders before ICU design or redesign may help identify around it; visitors would enter the patient rooms from the
common design concerns of these groups. It may help families back so as not to disrupt the central area used by the doctors
to have a community room for tai chi, yoga, and other spiri- and nurses. The rooms measured 200 square feet, as required
tual modalities and a chapel or a sanctuary close to the ICU by the state of Georgia, with no dedicated space for family
where religious services can take place. Sometimes these places members. This design essentially duplicated the current ICUs
are the only quiet refuge for families from the chaos of the at that time. In these units the typical patient room was so
hospital. Hospitals should also provide religious books, inspi- crowded with specialized equipment that it was difficult to get
rational texts, and texts on grief and coping written in mul- to the patient without tripping over cords or knocking out
tiple languages in the chapel or the community room, and invasive lines Figure 3.1 shows a comparison of the ICU
consider having space for common religious items used by before and after construction. It took time to respond to an
people of different faiths (e.g., rosaries, crucifixes, and holy emergency and to maintain sterility was near impossible.
water for Catholic users; clean clothing, prayer rugs, and com- During patient care rounds there was little room for the ICU
passes for Muslim users; Sabbath kits for Jewish users) in the team’s numerous members in crowded rooms and hallways.
chapel or the community room. When building a new unit, a In the central areas, nurses were crowded around desks with
tile marker could be inserted in all family spaces and patient charts spread all over tables; this increased the potential for
rooms to signify the direction of prayer. mistakes in documentation, record keeping, and medication
administration. Families were limited to dark, common spaces

Case Study: Emory University


Hospital Neuroscience Critical
Care Unit
In 2005 an architecture firm that worked with the Emory
Hospital in Atlanta, Georgia, presented a design to replace the
NCCU. The unit’s medical director (senior author Owen
Samuels, MD) was concerned that the intended design might
not address some of the issues described earlier that could
affect patient outcomes. Dr. Samuels approached Craig
Zimring of Georgia Institute of Technology School of Archi-
tecture about published evidence on the effects of ICU design.
Dr. Zimring agreed to have his graduate students, under the
guidance of Dr. Rashid, search the literature and develop
design concepts based on research findings. This exercise ulti-
mately led to an interdisciplinary design charrette with the
architectural firm and a revised NCCU design that included
the design implications from the literature. This section
reviews how the NCCU was designed primarily from the per-
spective of the medical director.

Evidence for a Better Way


Neurocritical care admissions at Emory increased from 587
patients in 1999 to more than 1400 patients in 2007. This
rapid growth meant that in 2007 the existing NCCU was out
of beds and patients with severe neurologic disorders were
cared for in three geographically separated small units (two
seven-bed units, and one nine-bed unit). This approach was Fig. 3.1  Top panel, Crowded initial conditions. Bottom panel, Completed
inefficient and compromised patient safety and delivery of project.
Section I—Background 23

in the outside hall away from patients and were restricted to


visiting during morning rounds. Discussions between doctor Table 3.1  Project Goals and Metrics
and families, including those about prognosis, brain death, for ICUD Design
organ donation, and end-of-life concerns took place either in Outcome
the cluttered patient rooms or in public hallways with no Design Drivers Design Response Measures
privacy. The new proposed space promised little more than Support families Family zone in Greater
some new converter chairs. patient room satisfaction on
There was concern that the proposed design was not ideal. Kid’s room Press-Ganey and
To convince the administration to pursue a completely new Lockers and Emory ICU
concept, designers focused on key people: the chief nursing showers surveys
Family quiet room Fewer complaints
officer for neurosciences and the chief executive officer of and litigation
Emory Healthcare. The design team told them that current
Support more Medical gas booms Fewer patient
ICUs were uncomfortable for families, the space was inher- procedures at Larger patient transfer
ently dangerous with a large potential for avoidable medical the bedside zone complications
mistakes that was largely unrecognized, and staff burnout was Improved and lower costs
common. Doctors and nurses make clinical decisions based in ergonomics Fewer errors
part on evidence from the literature. Shouldn’t such evidence Shorter stays
More time spent
also inform how hospitals and ICUs are designed? Although by the ICU staff
research in ICU design is an emerging field, there is a large in the ICU area
body of scientific evidence on how the physical environment Reduce infection Numerous rubs Improved
affects patient outcome, staff effectiveness, family well-being, and handwashing
and costs. The team therefore proposed a new design founded handwashing compliance
on an evidence-based approach for patient and family- stations Lower MSRA and
centered care supported by a healing environment but flexible hospital-
acquired
enough that it could continue to change in the future. Devel- infection rates
opers were confident that an improved design could reduce
Reduce medical Improved ceiling Fewer medical and
staff stress and enhance performance. As an academic institu- errors and tiles medication
tion, the team also wanted to study the effect of a new type increase patient Carpet where errors
of ICU. safety appropriate Less litigation
Charting niches Reduced
Zoned caregiver self-extubation
Emphasis About Family Involvement area Decreased falls
and injuries
Many factors other than technologic advancement can con- related to
tribute to patient outcome. When the ICU was designed, the patients leaving
team had several goals or “design drivers” (Table 3.1) in mind, beds
with accompanying measurable outcome variables to be ICU, Intensive care unit; MRSA, methicillin-resistant Staphylococcus aureus.
tracked. A primary driver for the new ICU was family support.
It was proposed to eliminate signs that restricted family visita-
tion, and the team was tempted to replace the sign “Physician
Rounds in Progress” with “Physician Rounds in Progress, American Association of Critical Care Nurses, and the Ameri-
Family Presence Encouraged.” The plan included a family zone can Institute of Architects jointly give this award. Next the
located within the patient’s room, as the “family studio,” a team partnered with the division of health care design at
children’s area located immediately outside the ICU, family Georgia Institute of Technology’s College of Architecture, led
lockers and showers, clothing washers and dryers, and a quiet by an environmental psychologist who specializes in the use
room for families and close friends. Outcome measures to be of architectural design as a healing tool. Several charrette ses-
collected included patient and family satisfaction scores, sions were held, which included key stakeholders from Emory,
hospital-acquired infections, nursing retention rates, ICU the architects (HKS, Inc.), and Georgia Tech.
length of stay, organ donation success, and number of litiga- A mock-up of the proposed ICU that included nurses’
tion filings. This family-centered approach was to be balanced station, patient rooms, wall-to-ceiling booms, and family areas
with the ability to turn the patient ICU room into a “mini- was created. Several procedures, including resuscitation, intu-
operating room” to reduce patient transfers and support more bation, intracranial monitor implantation, nursing shift-
procedures at the bedside. Other important design drivers change, and interactions between families and staff were
were reduction of medical errors; increased patient safety, and enacted through role-playing and recorded by videographers
staff satisfaction. Each goal had measurable outcomes to be for later analysis. A mock-up of the family studio allowed
tracked. analysis of functionality and family flow. Family members of
patients who had recently been discharged from or were still
in the ICU were involved with the unit’s design throughout
A Dynamic Design Process the process. As developers learned from such experiences, the
To determine factors such as patient room size and configura- design of the ICU was altered even as construction was under
tion and the design of family spaces, the design team analyzed way. It was originally planned to distribute the nurses’ stations
best practices of the prior 10 years’ winners of the ICU Design throughout the unit, but later it was decided to keep a com-
Citation Award. The Society of Critical Care Medicine, the munal area as well, because the team realized that nurses and
24 Section I—Background

2.6m
(8’7”)

8.5m 1.5m
(28’0”) (4’9”)
1.5m
(4’9”)
5.3m
(17’3”) 1.6m
(5’4”)

2.9m
(9’5”)

Fig. 3.2  Illustration of two adjacent ICU patient


rooms (shaded blue). The nurse’s station outside
the patient rooms and the adjoining family
areas (shaded green) also are visible. Room 4.5m (15’) 9.1m (30’)
dimensions are provided.

family studio has a table, chairs, comfortable sleeping arrange-


ments, flat-screen television, wireless Internet access, music,
and a white-noise system to blunt surrounding noises. The
new unit allows staff to do things that could not be done
before including hold a private conversation with a family
member when visiting a patient. Family members can leave
the room for some respite and still be just a stone’s throw away
from their loved one.
The following case that bridged the transition between
the old and new NCCU illustrates some of the advantages of
the new family-centered unit. David was a 31-year-old com-
puter programmer, the father of a 3-year-old girl, and about
to be married. He was admitted to the NCCU with a grade
IV subarachnoid hemorrhage and was in the old ICU for 4
days, and then moved to the new NCCU when it opened.
David later developed neurogenic pulmonary edema, severe
Fig. 3.3  Patient room, view from above. pneumonia, acute respiratory distress syndrome, and heart
failure and required induced coma for intracranial hyperten-
sion. He subsequently had an aneurysm rebleed and pro-
doctors at times need to be in the same area to better support gressed to brain death and his family decided to donate his
one another. organs.
His family, parents, and his fiancée kept a rotating vigil 24
hours a day. They always felt they were in the way in the old
Proposal Becomes Reality ICU, whereas they felt welcome in the new facility. The family
The new NCCU opened February 2007. The rooms are often stood at David’s bedside as the team explained the
between 345 and 450 square feet (Fig. 3.2) whereas the old purpose of the complex monitors and instruments and did
rooms were 120 to 200 square feet. Pneumatic booms lift not have to leave the bedside for discussions about brain death
equipment off the floor. This limits clutter and allows rapid or organ donation. The mother said, “This was our home for
360-degree access to the patient’s head in emergency situa- a month, and it got so that the nurses could tell when we
tions. The beds and doors are configured so that patients who needed a hug.”114 David’s father said, “No one ever misled us
are awake have a direct line of sight to the nurses’ station or a or told us anything but the truth … and most importantly, we
bay window. Each room is a suite that consists of the patient were there for everything.”115
room and a family area separated by a curved wall with large Hospital staff did everything they could for David, and
glass-block windows that lets in natural light (Fig. 3.3). The nothing could change his ultimate outcome. But the way
Section I—Background 25

someone dies is important. The circumstances of how David standardized protocols for integration of medical equipment
was treated probably helped allow the family to donate his and devices, many interface problems related to technology
organs and better come to terms with his death. They later that we see in ICUs today may be eliminated.
generously donated their time to help the NCCU develop the
family-centered approach by participating in many discus-
sions about their experiences. References
1. Bartley J, Streifel AJ. Design of the environment of care for safety of patients
and personnel: does form follow function or vice versa in the intensive care
Family-Centered Units unit? Crit Care Med 2010;38(Suppl. 8):388–98.
2. Ulrich RS, Zimring C, Joseph A, et al. The role of the physical environment
Pose Challenges in the hospital of the 21st century: a once-in-a-lifetime opportunity.
Concord, CA: The Center for Health Design; 2004.
The concept of care around the patient and their family makes 3. Ulrich RS, Zimring C, Zhu X, et al. A review of the research literature on
sense. However, there is resistance to it, even from the most evidence-based healthcare design. HERD 2008;1(3):61–125.
dedicated health care workers. The team at Emory spent about 4. Zimring C, Ulrich RS, Joseph A, et al. The environment’s impact on safety.
In: Marberry SO, editor. Improving healthcare with better building design.
a year and a half preparing for family-centered care; starting Chicago: Health Administration Press; 2006. p. 63–81.
the process was the real challenge. In a unit that is designed for 5. Cai H, Do EY-L, Zimring CM. Extended linkography and distance graph in
both patients and their families, how does one care for patients design evaluation: an empirical study of the dual effects of inspiration
and their families simultaneously? There are several challenges: sources in creative design. Design Studies 2010;31(2):146–68.
6. Rashid M. A decade of adult intensive care unit design: a study of the
 Team rounding: Doctors and nurses may be apprehensive physical design features of the best-practice examples. Crit Care Nurs Quart
about inviting families to rounds. Training doctors and 2006;29(4):282–311.
7. Kohlenberg A, Schwab F, Behnke M, et al. Screening and control of
nurses with families present is a real paradigm shift and methicillin-resistant Staphylococcus aureus in 186 intensive care units:
raises many controversial issues. different situations and individual solutions. Critical Care 2011;15:R285.
 Nurses in view: Imagine a nurse operating six or seven 8. Vincent JL, Bihari DJ, Suter PM, et al. The prevalence of nosocomial
intravenous pumps and trying to figure out medications infection in intensive care units in Europe: results of the European
while having a family member—or three or four Prevalence of Infection in Intensive Care (EPIC) Study. EPIC International
Advisory Committee. JAMA 1995;274(8):639–44.
members—continuously present. 9. Akansel N, Kaymakçi S. Effects of intensive care unit noise on patients: a
 Urgent or frightening treatment: How does staff deal with study on coronary artery bypass graft surgery patients. J Clin Nurs
resuscitation? What if the family is right by the bedside? 2008;17(12):1581−90.
Should they be asked to leave? What kind of support do 10. Piergeorge AR, Ceserano FL, Casanova DM. Designing the critical care unit:
a multidisciplinary approach. Crit Care Med 1983;11(7):541–45.
they need? 11. Society of Critical Care Medicine (SCCM). NIH consensus development
The authors do not have all the answers to such problems but conference on critical care medicine. Crit Care Med 1983;11:466–9.
12. Society of Critical Care Medicine (SCCM). Guidelines for intensive care unit
are currently studying them to figure out best practices. design. Crit Care Med 1995;23(3):582–8.
13. Stoddard JC. Design, staffing and equipment requirements for an intensive
Outcomes care unit. Int Anesthesiol Clin 1981;19(2):77–95.
14. Hamilton DK, editor. ICU 2010: ICU design for the future, a critical care
design symposium. Houston: Center for Innovation in Health Facilities;
Emory’s NCCU won the 2008 ICU Design Citation Award 2000.
from the Society of Critical Care Medicine, the American 15. Carthey J. Reinterpreting the hospital corridor: “wasted space” or essential
Association of Critical Care Nurses, and the American Insti- for quality multidisciplinary clinical care? HERD 2008;2(1):17–29.
tute of Architects Academy on Architecture for Health. The 16. National Fire Protection Association. Section 19.2.5.7.2.3. Quincy, MA; 2012.
authors have started to look at outcomes associated with the 17. Hendrich A. Case study: the impact of acuity adaptable rooms on future
designs, bottlenecks and hospital capacity. Paper presented at the Impact
NCCU. Patient and staff satisfaction have increased, according Conference on Optimizing the Physical Space for Improved Outcomes,
to self-assessments. Most recent Press-Ganey Family Satisfac- Satisfaction and the Bottom Line, Atlanta, 2003.
tion scores indicate greater than 85% overall satisfaction. 18. Hendrich A, Fay J, Sorrells A. Effects of acuity-adaptable rooms on flow of
Other variables of quality care benchmarks include significant patients and delivery of care. Am J Crit Care 2004;13(1):35–45.
19. Hendrich A, Lee N. Intra-unit patient transports: time, motion, and cost
decrease in pneumonia rates and central venous line blood- impact on hospital efficiency. Nurs Econ 2005;23(4):157–64.
stream infections, increased compliance with critical care 20. Cook RI, Render M, Woods DD. Gaps in the continuity of care and progress
pathways, increased nursing retention, and a trend toward on patient safety. BMJ 2000;320(7237):791–4.
decreased medical legal litigation. 21. Jastremski CA, Harvey M. Making changes to improve the intensive care unit
experience for patients and their families. New Horiz 1998;6(1):99–109.
22. Hamilton DK, Shepley MM. Design for critical care: an evidence based
Conclusion approach. Oxford: Elsevier Ltd; 2010.
23. Tanja-Dijkstra K. The impact of bedside technology on patients’ well being.
This chapter has briefly discussed environmental design crite- Health Environments Research & Design Journal 2011;5(1):43−51.
ria for the ICU, the patient room, staff and support areas, and 24. Walch JM, Rabin BS, Day R, et al. The effect of sunlight on post-operative
analgesic medication usage: a prospective study of spinal surgery patients.
family areas of ICUs to help improve patient, staff, and family Psychosom Med 2005;67(1):156–63.
outcomes. These processes can influence patient outcomes 25. Cadenhead C, Anderson D. Critical care unit design, the winners and future
and have an effect on how staff monitor individual patients trends: an investigative study. World Health Design Journal 2009;2:72–7.
and overall outcomes and quality in the ICU. Although some 26. Chaudhury H, Mahmood A, Valente M. Advantages and disadvantages of
of these issues will likely persist for the foreseeable future, single-versus multiple-occupancy rooms in acute care environments: a
review and analysis of the literature. Enviro Behav 2005;37:760–86.
others are being addressed by technical and process changes. 27. Levin PD, Golovanevski M, Moses AE, et al. Improved ICU design reduces
For example, as manufacturers of medical equipment and acquisition of antibiotic resistant bacteria: a quasi-experimental
supplies continue to merge and consolidate and develop observational study. Critical Care 2011;15:R21.
26 Section I—Background

28. Maki DG. Nosocomial infection in the intensive care unit. In: Parillo JE, 40. Kaplan LM, McGuckin M. Increasing handwashing compliance with more
Bone RC, editors. Critical care medicine-principles of diagnosis and accessible sinks. Infection Control 1986;7(8):408–10.
management. St. Louis: Mosby; 1995. p. 893–954. 41. Vernon MO, Trick WE, Welbel SF, et al. Adherence with hand hygiene: does
29. Shirani KZ, McManus AT, Vaughan GM, et al. Effects of environment on number of sinks matter? Infection control and hospital epidemiology
infection in burn patients. Arch Surg 1986;121(1):31–6. 2003;24(3):224–5.
30. Teltsch DY, Hanley J, Loo V, et al. Infection acquisition following intensive 42. Vietri NJ, Dooley DP, Davis CE, et al. The effect of moving to a new hospital
care unit room privatization. Arch Intern Med 2011;171(1):32–8. facility on the prevalence of methicillin-resistant Staphylococcus aureus.
31. Van Enk RA, Steinberg F. Comparison of private room with multiple-bed American Journal of Infection Control 2004;32(5):262–7.
ward neonatal intensive care unit environments. Health Environments 43. Trick W, Vernon M, Welbe LS, et al. Multicenter intervention program to
Research & Design Journal 2011;5(1):52–63. increase adherence to hand hygiene recommendations and glove use and to
32. France DJ, Throop P, Walczyk B, et al. Does patient-centered design reduce the incidence of antimicrobial resistance. Infection Control and
guarantee patient safety? Using human factors engineering to find a balance Hospital Epidemiology 2007;28(1):42–9.
between provider and patient needs. J Patient Saf 2005;1(3):145–53. 44. Dyson M. Intensive care unit psychosis, the therapeutic nurse-patient
33. Burrington M, Architects WHR. An alternative method for human waste relationship and the influence of the intensive care setting: analyses of
disposal in critical care. In Hamilton DK, editor. ICU 2010: ICU design for interrelating factors. Journal of Clinical Nursing 1999;8:284–90.
the future, a critical care design symposium. Houston: Center for Innovation 45. Beauchemin KM, Hays P. Sunny hospital rooms expedite recovery from
in Health Facilities; 2000. severe and refractory depressions. Journal of Affective 1996;40(1-2):
34. Mullin B, Rouget C, Clément C, et al. Association of private isolation rooms 49–51.
with ventilator-associated Acinetobacter baumanii pneumonia in a surgical 46. Benedetti F, Colombo C, Barbini B, et al. Morning sunlight reduces length of
intensive-care unit. Infection Control and Hospital Epidemiology hospitalization in bipolar depression. Journal of Affective Disorders
1997;18(7):499–503. 2001;62(3):221–3.
35. Facilities Guidelines Institute. Guidelines for design and construction of health 47. Lewy AJ, Bauer VK, Cutler NL, et al. Morning vs. evening light treatment of
care facilities. Washington, DC: The American Institute of Architects; 2006. patients with winter depression. Archives of General Psychiatry
36. Joseph A. The impact of the environment on infections in healthcare 1998;55(10):890–6.
facilities. Concord, CA: The Center for Health Design; 2006. 48. Ulrich RS. View through a window may influence recovery from surgery.
37. Boyce, J. Antiseptic technology: access, affordability, and acceptance. Science 1984;224(4647):420–1.
Emerging Infectious Diseases 2001;7(2):231–3. 49. Ulrich RS. Effects of interior design on wellness: theory and recent scientific
38. Lankford MG, Zembower TR, Trick WE, et al. Influence of role models and research. Journal of Health Care Design 1991;3:97–109.
hospital design on hand hygiene of healthcare workers. Emergency 50. Gelling L. Causes of ICU psychosis: the environmental factors. Nursing in
Infectious Diseases 2003;9(2):217–23. Critical Care 1999;4:22–6.
39. Larson E, Albrecht S, O’Keefe M. Hand hygiene behavior in a pediatric
emergency department and a pediatric intensive care unit: comparison of A complete list of references for this chapter can be found online at
use of 2 dispenser systems. American Journal of Critical Care www.expertconsult.com.
2005;14(4):304–11.
Section I—Background 26.e1

References 29. Shirani KZ, McManus AT, Vaughan GM, et al. Effects of environment on
infection in burn patients. Arch Surg 1986;121(1):31–6.
1. Bartley J, Streifel AJ. Design of the environment of care for safety of 30. Teltsch DY, Hanley J, Loo V, et al. Infection acquisition following intensive
patients and personnel: does form follow function or vice versa in the care unit room privatization. Arch Intern Med 2011;171(1):32–8.
intensive care unit? Crit Care Med 2010;38(Suppl. 8):388–98. 31. Van Enk RA, Steinberg F. Comparison of private room with multiple-bed
2. Ulrich RS, Zimring C, Joseph A, et al. The role of the physical environment ward neonatal intensive care unit environments. Health Environments
in the hospital of the 21st century: a once-in-a-lifetime opportunity. Research & Design Journal 2011;5(1):52–63.
Concord, CA: The Center for Health Design; 2004. 32. France DJ, Throop P, Walczyk B, et al. Does patient-centered design
3. Ulrich RS, Zimring C, Zhu X, et al. A review of the research literature on guarantee patient safety?: Using human factors engineering to find a
evidence-based healthcare design. HERD 2008;1(3):61–125. balance between provider and patient needs. J Patient Saf 2005;1(3):
4. Zimring C, Ulrich RS, Joseph A, et al. The environment’s impact on safety. 145–53.
In: Marberry SO, editor. Improving healthcare with better building design. 33. Burrington M, Architects WHR. An alternative method for human waste
Chicago: Health Administration Press; 2006. p. 63–81. disposal in critical care. In Hamilton DK, editor. ICU 2010: ICU design for
5. Cai H, Do EY-L, Zimring CM. Extended linkography and distance graph in the future, a critical care design symposium. Houston: Center for
design evaluation: an empirical study of the dual effects of inspiration Innovation in Health Facilities; 2000.
sources in creative design. Design Studies 2010;31(2):146–68. 34. Mullin B, Rouget C, Clément C, et al. Association of private isolation
6. Rashid M. A decade of adult intensive care unit design: a study of the rooms with ventilator-associated Acinetobacter baumanii pneumonia in a
physical design features of the best-practice examples. Crit Care Nurs Quart surgical intensive-care unit. Infection Control and Hospital Epidemiology
2006;29(4):282–311. 1997;18(7):499–503.
7. Kohlenberg A, Schwab F, Behnke M, et al. Screening and control of 35. Facilities Guidelines Institute. Guidelines for design and construction of
methicillin-resistant Staphylococcus aureus in 186 intensive care units: health care facilities. Washington, DC: The American Institute of Architects;
different situations and individual solutions. Critical Care 2011;15:R285. 2006.
8. Vincent JL, Bihari DJ, Suter PM, et al. The prevalence of nosocomial 36. Joseph A. The impact of the environment on infections in healthcare
infection in intensive care units in Europe: results of the European facilities. Concord, CA: The Center for Health Design; 2006.
Prevalence of Infection in Intensive Care (EPIC) Study. EPIC International 37. Boyce, J. Antiseptic technology: access, affordability, and acceptance.
Advisory Committee. JAMA 1995;274(8):639–44. Emerging Infectious Diseases 2001;7(2):231–3.
9. Akansel N, Kaymakçi S. Effects of intensive care unit noise on patients: a 38. Lankford MG, Zembower TR, Trick WE, et al. Influence of role models and
study on coronary artery bypass graft surgery patients. J Clin Nurs hospital design on hand hygiene of healthcare workers. Emergency
2008;17(12):1581−90. Infectious Diseases 2003;9(2):217–23.
10. Piergeorge AR, Ceserano FL, Casanova DM. Designing the critical care unit: 39. Larson E, Albrecht S, O’Keefe M. Hand hygiene behavior in a pediatric
a multidisciplinary approach. Crit Care Med 1983;11(7):541–45. emergency department and a pediatric intensive care unit: comparison of
11. Society of Critical Care Medicine (SCCM). NIH consensus development use of 2 dispenser systems. American Journal of Critical Care
conference on critical care medicine. Crit Care Med 1983;11:466–9. 2005;14(4):304–11.
12. Society of Critical Care Medicine (SCCM). Guidelines for intensive care 40. Kaplan LM, McGuckin M. Increasing handwashing compliance with more
unit design. Crit Care Med 1995;23(3):582–8. accessible sinks. Infection Control 1986;7(8):408–10.
13. Stoddard JC. Design, staffing and equipment requirements for an intensive 41. Vernon MO, Trick WE, Welbel SF, et al. Adherence with hand hygiene: does
care unit. Int Anesthesiol Clin 1981;19(2):77–95. number of sinks matter? Infection control and hospital epidemiology
14. Hamilton DK, editor. ICU 2010: ICU design for the future, a critical care 2003;24(3):224–5.
design symposium. Houston: Center for Innovation in Health Facilities; 42. Vietri NJ, Dooley DP, Davis CE, et al. The effect of moving to a new
2000. hospital facility on the prevalence of methicillin-resistant Staphylococcus
15. Carthey J. Reinterpreting the hospital corridor: “wasted space” or essential aureus. American Journal of Infection Control 2004;32(5):262–7.
for quality multidisciplinary clinical care? HERD 2008;2(1):17–29. 43. Trick W, Vernon M, Welbe LS, et al. Multicenter intervention program to
16. National Fire Protection Association. Section 19.2.5.7.2.3. Quincy, MA; increase adherence to hand hygiene recommendations and glove use and to
2012. reduce the incidence of antimicrobial resistance. Infection Control and
17. Hendrich A. Case study: the impact of acuity adaptable rooms on future Hospital Epidemiology 2007;28(1):42–9.
designs, bottlenecks and hospital capacity. Paper presented at the Impact 44. Dyson M. Intensive care unit psychosis, the therapeutic nurse-patient
Conference on Optimizing the Physical Space for Improved Outcomes, relationship and the influence of the intensive care setting: analyses of
Satisfaction and the Bottom Line, Atlanta, 2003. interrelating factors. Journal of Clinical Nursing 1999; 8:284–90.
18. Hendrich A, Fay J, Sorrells A. Effects of acuity-adaptable rooms on flow of 45. Beauchemin KM, Hays P. Sunny hospital rooms expedite recovery from
patients and delivery of care. Am J Crit Care 2004;13(1):35–45. severe and refractory depressions. Journal of Affective 1996;40(1-2):49–51.
19. Hendrich A, Lee N. Intra-unit patient transports: time, motion, and cost 46. Benedetti F, Colombo C, Barbini B, et al. Morning sunlight reduces length
impact on hospital efficiency. Nurs Econ 2005;23(4):157–64. of hospitalization in bipolar depression. Journal of Affective Disorders
20. Cook RI, Render M, Woods DD. Gaps in the continuity of care and 2001;62(3):221–3.
progress on patient safety. BMJ 2000;320(7237):791–4. 47. Lewy AJ, Bauer VK, Cutler NL, et al. Morning vs. evening light treatment of
21. Jastremski CA, Harvey M. Making changes to improve the intensive care patients with winter depression. Archives of General Psychiatry
unit experience for patients and their families. New Horiz 1998;6(1): 1998;55(10):890–6.
99–109. 48. Ulrich RS. View through a window may influence recovery from surgery.
22. Hamilton DK, Shepley MM. Design for critical care: an evidence based Science 1984;224(4647):420–1.
approach. Oxford: Elsevier Ltd; 2010. 49. Ulrich RS. Effects of interior design on wellness: theory and recent
23. Tanja-Dijkstra K. The impact of bedside technology on patients’ well being. scientific research. Journal of Health Care Design 1991;3:97–109.
Health Environments Research & Design Journal 2011;5(1):43−51. 50. Gelling L. Causes of ICU psychosis: the environmental factors. Nursing in
24. Walch JM, Rabin BS, Day R, et al. The effect of sunlight on post-operative Critical Care 1999;4:22–6.
analgesic medication usage: a prospective study of spinal surgery patients. 51. Hughes J. Hallucinations following cardiac surgery in a pediatric intensive
Psychosom Med 2005;67(1):156–63. care unit. Intensive and Critical Care Nursing 1994;10:209–11.
25. Cadenhead C, Anderson D. Critical care unit design, the winners and future 52. Gabor J, Cooper A, Crombach S, et al. Contribution of the intensive care
trends: an investigative study. World Health Design Journal 2009;2:72–7. unit environment to sleep disruption in mechanically ventilated patients
26. Chaudhury H, Mahmood A, Valente M. Advantages and disadvantages of and healthy subjects. American Journal of Respiratory and Critical Care
single-versus multiple-occupancy rooms in acute care environments: a Medicine 2003;167:708–15.
review and analysis of the literature. Enviro Behav 2005;37:760–86. 53. Busch-Vishniac I, West J, Barnhill C, et al. Noise levels in Johns Hopkins
27. Levin PD, Golovanevski M, Moses AE, et al. Improved ICU design reduces Hospital. Journal of the Acoustical Society of America 2005;118:
acquisition of antibiotic resistant bacteria: a quasi-experimental 3629–1645.
observational study. Critical Care 2011;15:R21. 54. Cropp A, Woods L, Raney D. Name that tone: the proliferation of alarms in
28. Maki DG. Nosocomial infection in the intensive care unit. In: Parillo JE, the intensive care unit. Chest 1994;105:1217–20.
Bone RC, editors. Critical care medicine-principles of diagnosis and 55. Ulrich RS, Lawson B, Martinez M. Exploring the patient environment: an
management. St. Louis: Mosby; 1995. p. 893–954. NHS estates workshop. London: The Stationery Office; 2003.
26.e2 Section I—Background

56. Joint Commission on Accreditation of Healthcare Organizations. Root 84. Elsbach KD, Pratt MG. The physical environment in organizations. The
causes of patient falls. Sentinel event statistics [Graph] 2006 [cited 2007 Academy of Management Annals 2007;1(1):181−224.
7/22/07]; Available from: http://www.jointcommission.org/NR/rdonlyres/ 85. Rashid M, Zimring C. On psychosocial constructs and office settings: a
FA5A080F-C259-47CC-AAC8-BAC3F5C37D84/0/se_rc_patient_falls.jpg review of the empirical literature. In Chaudhury H. editor. Proceedings of
57. Drahota A, Gal D, Windsor J. Flooring as an intervention to reduce injuries the Environmental Design Research Association (EDRA) 36 Conference.
from falls in healthcare settings. Quality in Ageing and Older Adults Vancouver, Canada: EDRA; 2005. p. 107–22.
2007;8(1):3–9. 86. Sundstrom E, Herbert RK, Brown DW. Privacy and communication in an
58. Healey F. Does flooring type affect risk of injury in older in-patients? open-plan office: a case study. Environment and Behavior 1982;1:379–92.
Nursing Times 1994;90(27):40–1. 87. Allen T. Managing the flow of technology. Cambridge, MA: MIT Press;
59. Simpson AH, Lamb S, Roberts PJ, et al. Does the type of flooring affect the 1977.
risk of hip fracture? Age and Ageing 2004;33(3):242–6. 88. Bobele HK, Buchanan PJ. Building a more productive environment.
60. Nelson AL, Matz M, Chen F, et al. Development and evaluation of a Management World 1979;8(1):8–10.
multifaceted ergonomics program to prevent injuries associated with 89. Hillier B, Penn A. Visible colleges: structure and randomness in the place of
patient handling tasks. Journal of International Nursing Studies 2006;43: discovery. Science in Context 1991;4(1):23–49.
717–33. 90. Boyce P, Hunter C, Howlett O. The benefits of daylight through windows.
61. Zborowsky T, Bunker-Hellmich L, Morelli A, et al. Centralized vs. Troy, New York: Rensselaer Polytechnic Institute; 2003.
decentralized nursing stations: effects on nurses’ functional use of space 91. Roseman C, Booker JM. A seasonal pattern of hospital medication errors in
and work environment. Health Environments Research & Design Journal Alaska. Psychiatry Research 1995;57(3):251–7.
2010;3(4):19−42. 92. Buchanan TL, Barker KN, Gibson JT, et al. Illumination and errors in
62. Hendrich A, Chow MP, Bafna S, et al. Unit-related factors that affect dispensing. American Journal of Hospital Pharmacy 1991;48(10):2137–45.
nursing time with patients: spatial analysis of the time and motion study. 93. Rashid M, Zimring C. A review of the empirical literature on the
Health Environments Research & Design Journal 2009;2(2):5–20. relationships between indoor environment and stress in healthcare and
63. Burgio L, Engel B, Hawkins A, et al. A descriptive analysis of nursing staff office settings: problems and prospects of sharing evidence. Environment
behaviors in a teaching nursing home: differences among NAs, LPNs and and Behavior 2008;40(3):151–90.
RNs. The Gerontologist 1990;30(1):107–12. 94. Bozett FW, Gibbons R. The nursing management of families in the critical
64. Tucker A, Spear S. Operational failures and interruptions in hospital care setting. Critical Care Update 1983;10(2):22–7.
nursing. Health Services Research 2006;41(3):643–62. 95. Byers, JF. Family discussion groups, a way to meet the needs of the ICU
65. Shepley MM. Predesign and postoccupancy analysis of staff behavior in a patient’s family. Critical Care Nurse 1983;3(1):16–20.
neonatal intensive care unit. Children’s Health Care 2002;31(3):237–53. 96. Rashid M. Environmental design for patient families in intensive care units.
66. Shepley MM, Davies K. Nursing unit configuration and its relationship to Journal of Healthcare Engineering 2010;1(3):367–97.
noise and nurse walking behavior: an AIDS/HIV unit case study. AIA 97. Simpson T. The family as a source of support for the critically ill adult.
Academy Journal 2003. Available from: http://www.aia.org/aah/journal/ AACN Clinical Issues in Critical Nursing 1991;2(2):229–35.
0401/article0404.asp. 98. Hodovanic BH, Reardon D, Reese W, et al. Family crisis intervention program
67. Deane D, McElroy MJ, Alden S. Documentation: meeting requirements in the medical intensive care unit. Heart and Lung 1984;13(3):243–9.
while maximizing productivity. Nursing Economics 1986;4(4):174–7. 99. Molter NC. Needs of relatives of critically ill patients: a descriptive study.
68. Pierpont GL, Thilgen D. Effect of computerized charting on nursing Heart and Lung 1979;8(2):332–9.
activity in intensive care. Critical Care Medicine 1995;23(6):1067–73. 100. Pati D, Nanda U. Influence of positive distractions on children in two clinic
69. Quist BD. Work sampling nursing units. Nursing Management 1992;23(9): waiting areas. Health Environments Research & Design Journal
50–1. 2011;4(3):124–40.
70. Morrison W, Haas E, Shaffner D, et al. Noise stress and annoyance in a 101. Gurascio-Howard L, Malloch K. Centralized and decentralized nurse
pediatric intensive care unit. Critical Care Medicine 2003;31(1):35–7. station design: an examination of caregiver communication, work activities,
71. Topf M, Dillon E. Noise-induced stress as a predictor of burnout in critical and technology. Health Environments Research and Design Journal
care nurses. Heart and Lung 1988;17:247–50. 2007;1(1):44–57.
72. Bayo MV, Garcia AM, Garcia A. Noise levels in an urban hospital and 102. Melin L, Gotestam KG. The effects of rearranging ward routines on
workers’ subjective responses. Archives of Environmental Health 1995; communication and eating behaviors of psychogeriatric patients. Journal of
50(3):247–51. Applied Behavior Analysis. 198;14(1):47–51.
73. Westbrook JI, Coiera E, Dunsmuir WT, et al. The impact of interruptions 103. Peterson R, Knapp T, Rosen J. The effects of furniture arrangement on the
on clinical task completion. Qual Saf Health Care 2010;19(4):284−9. behavior of geriatric patients. Behavior Therapy 1977;8:464–7.
74. Boston Globe. “Patient alarms often unheard, unheeded.” Feb. 11, 2011. 104. Holahan C. Seating patterns and patient behavior in an experimental
Available at: http://www.boston.com/lifestyle/health/articles/2011/02/13/ dayroom. Journal of Abnormal Psychology 1972;80(2):115–24.
patient_alarms_often_unheard_unheeded/ (accessed Feb. 29, 2012). 105. Sommer R, Ross H. Social interaction on a geriatrics ward. The
75. Rashid M. Technology and the future of ICU design. Critical Care Nursing International Journal of Social Psychiatry 1958;4(2):128–33.
Quarterly 2011;34(4):332–60. 106. Astedt-Kurki P, Paavilainen E, Tammentie T, et al. Interaction between adult
76. Kobayashi L, Boss RM , Gibbs FJ, et al. Color-coding and human factors patients’ family members and nursing staff on a hospital ward.
engineering to improve patient safety characteristics of paper-based Scandinavian Journal of Caring Sciences 2001;15(2):142–50.
emergency department clinical documentation. Health Environments 107. Miwa Y, Hanyu K. The effects of interior design on communication and
Research & Design Journal 2011;4(4):79−88. impressions of a counselor in a counseling room. Environment and
77. Blomkvist V, Eriksen C, Theorell T, et al. Acoustics and psychosocial Behavior 2006;38(4):484–502.
environment in intensive coronary care. Occupational and Environmental 108. Daykin N, Bunt L. Music as a Resource for Health and Well-Being. In:
Medicine 2005;62(3):1–8. Söderback Ingrid, editor. International Handbook of Occupational Therapy
78. Knaus WA, Draper EA, Wagner DP, et al. An evaluation of outcome from Interventions. New York: Springer; 2009. p. 453–46.
intensive care in major medical centers. Ann Intern Med 1986;104:410–8. 109. Ulrich RS, Gilpin L. Healing arts. In: Frampton SB, gilpin L, Charmel P,
79. Shortell SM, Zimmerman JE, Rousseau DM, et al. The performance of editors. Putting patients first: Designing and practicing patient-centered
intensive care units: does good management make a difference? Medical care. San Francisco: Jossey-Bass; 2003. p. 117–46.
Care 1994;32(5):508–25. 110. Cooke B, Ernst E. Aromatherapy: A systematic review. British Journal of
80. Baggs JG, Ryan SA, Phelps CE, et al. The association between General Practice 2000;50:493–6.
interdisciplinary collaboration and patient outcomes in a medical/intensive 111. Marcus CC, Barnes M, editors. Healing gardens: Therapeutic benefits and
care unit. Heart and Lung 1992;21(1):18–24. design recommendations. New York: Wiley; 1999.
81. Baggs JG, Schmitt M, Mushlin A, et al. Association between nurse-physician 112. O’Brien ME. Spirituality in Nursing. Sudbury, CT: Jones and Bartlett; 2003.
collaboration and patient outcomes in three intensive care units. Critical 113. Young C. Spiritually healing environments. In: Spirituality, Health, and
Care Medicine 1999;27(9):1991–8. Healing. Jones & Bartlett Publishers; 2006. p. 147–68.
82. Kramer M, Schmalenberg C. Securing “good” nurse physician relationship. 114. Landro L. ICUs’ new message: welcome, families. Wall St. J. 2007. (Retrieved
Nursing Management 2003;34(7):34–8. from http://oruine.wsj.com/article/SB118419577614963880.html/).
83. Rashid M. Hospital design and face-to-face interaction among clinicians: a 115. Mantone J. Building family-friendly lCUs. Wall St. J. 2007. <http://
theoretical model. Health Environments Research & Design Journal blogs.wsj.com/health/2007/07/12/building-familyfriendly-icus/>. (accessed
2009;2(4):62−84. 30.10.2008).
Chapter
4  
I

Informatics Infrastructure
for the Neurocritical
Care Unit
J. Michael Schmidt, David K. Vawdrey, and Richard S. Moberg

4. Do osmolar data suggest that additional osmotic therapy


Introduction would be dangerous to the patient?
S.P. is a 60-year-old female with a history of hypertension who
presented comatose with a left thalamic intracerebral hemor- Today clinicians generally are required to answer these ques-
rhage associated with intraventricular hemorrhage, midline tions in their heads, approximating when interventions were
shift, and localized mass effect. Emergently, an external ven- made by thumbing through the hourly recorded data, estimat-
tricular drain was placed into the left frontal horn, and on day ing each therapy’s impact on ICP, and manually checking
5 an additional drain was placed into the left temporal horn laboratory data to estimate the osmolar gap. Ideally other
for persistent left temporal dilation. Invasive multimodality aspects of ICP management protocols also should be evalu-
monitoring including intracranial pressure (ICP), and brain ated, for example, end-tidal carbon dioxide (CO2) and tem-
tissue oxygen tension (PbtO2), and cerebral microdialysis was perature. Further, analysis tools at the bedside should exist to
placed into the right frontal lobe. She received repeated doses evaluate the patient’s physiologic status, such as cerebral auto-
of mannitol and hypertonic saline for persistent elevated ICP. regulation, which would indicate how ICP would respond to
Managing patients with intracranial hypertension, like S.P., changes in blood pressure. This then may reduce the need to
is a mainstay in neurocritical care and involves a structured discuss how other invasive neuromonitoring data (e.g., cere-
treatment protocol1 to continually assess the effect of each bral blood flow, PbtO2, cerebral metabolism) may facilitate ICP
intervention. Tracking and quantifying physiologic measures management. Any attempt to understand these dynamic rela-
such as cerebral perfusion pressure (CPP) also is crucial and tionships among ICP, interventions, and other physiology is
needs to be put into the context of sedation level, osmothera- complicated, and yet this is only one aspect of patient care.
pies, ventilator settings, and temperature modulation among The promise of clinical information systems is to improve
many other interventions. In a digital world clinical staff patient care, reduce medical errors, reduce documentation
should be able to systematically, continually, and rapidly eval- time, and improve efficiency.2,3 Paper-based hospital medical
uate the effect of various treatments for increased ICP. For records are generally inefficient (e.g., illegible handwriting,
instance, when the impact of mannitol and hypertonic saline manual data entry) and have been shown to contribute to
administration in S.P. is evaluated, the neurocritical care unit medical errors.4,5 Billions of dollars are being devoted to the
(NCCU) staff should have immediate access to basic variables development and implementation of electronic health records
such as osmolality and ICP at the bedside (Fig. 4.1). As fun- (EHRs)6 and significant progress has been made to bring these
damental as this is, a plot that contains these core elements is data together for documentation purposes.4 Intensive care
difficult to create in real time and even retrospectively at most units (ICUs) have been migrating from paper-based to com-
institutions. But imagine being able to look at the patient’s puterized charting systems for more than a decade.7
display screen in the NCCU and to be able to answer the fol- In the ICU, however, problems that face clinicians and
lowing questions easily: nurses extend beyond the need for more efficient documenta-
tion. During rounds of critically ill patients each morning, a
1. When mannitol is administered, by how much (mm Hg) physician may be confronted with more than 200 variables,8,9
is ICP lowered; how quickly is ICP lowered, and how long and some clinical information systems acquire and store phys-
until ICP again increases to greater than 20 mm Hg? iologic variables and device parameters online at least every
2. Are repeated administrations of mannitol equally effective minute.10 People, however, are not able to judge the degree of
or does repeated administration fail to lower ICP as much, relatedness between more than two variables.11 The ability to
as quickly, or as long? store high-dimensional data far exceeds the intellectual capac-
3. Does adding hypertonic saline provide additional ICP ity to understand it unassisted12; this greatly contributes to
reduction? conditions of constant “information overload” that can lead
© Copyright 2013 Elsevier Inc. All rights reserved. 27
28 Section I—Background

ICP Osmolal-Serium 3% Saline Mannitol 20%

60
500
340 2500
40 400
2000

ICP (mm Hg)


320

75 mL/hr
20 300
1500
300
0 200 1000
280
–20 100 500

260
Fig. 4.1  Example of minimum integrated data display 0 0
to support intracranial pressure management. ICP, 00:00 00:00
Intracranial pressure. 1/9/2007 1/10/2007 1/11/2007

to preventable medical errors.11,13 A clinical informatics infra- endpoints or improve clinical conditions such as cerebral vaso-
structure in the NCCU that only supports documentation is spasm or sepsis. Second, patient data should provide under­
simply not sufficient. Ideally NCCU staff should be able to use standing of the patient’s physiologic status (e.g., dysautonomia,
clinical decision support systems to help understand what a cerebral autoregulation failure), and how such physiology
patient’s data is trying to tell them.14 impacts other metrics of brain health (see Chapter 40). Third,
Currently there is a paucity of clinical decision support complex relationships within patient data should be automati-
systems in use in the ICU,12 but this is understandable because cally processed to enable better understanding of prognosis
the data and infrastructure to support their development and and earlier diagnosis of secondary events before clinical symp-
use are generally not in place. When patient data is collected toms occur. This automatic processing of data further sup-
in digital form, it usually resides on isolated systems without ports clinical decision making, and can enable computerized
a way to bring this data together in meaningful ways.4 The implementation of clinical protocols16 (see Chapter 45) or
objective of this chapter is to provide a blueprint for work decision making on prognosis or outcome prediction if appro-
with hospital administration, information technology, bio- priate models are used.17,18
medical engineering, industry vendors, and clinical staff to
create an informatics infrastructure that provides decision
support. Many hospitals already have data-collection systems Clinical Computing Systems
in place for electronic documentation, laboratory results, and
physician-order systems but need strategies to integrate these The Blueprint
data with high-resolution bedside monitor data to support The goal is to create the informatics infrastructure necessary
clinical decision support systems. First, clinicians need to be to enable health care providers to leverage data from database
able to articulate a clear message as to why patient data must systems that contain physiologic, laboratory, pharmacy, and
be stored at a high resolution and be accessible in real time to other clinical data for the continued development and imple-
support clinical decision making. mentation of better clinical support tools to provide patients
the best care possible.3,4 There are many ways to accomplish
these goals, but it is essential to work closely with the institu-
The Justification: Why Clinical Data tion’s information technology (IT) department to devise the
right solution for the ICU and institution because it is often
Is Needed difficult to tell what is needed before the project starts. Regard-
Data are a commodity that has many purposes but in health less of the specifics of the solution, each will contain the same
care enable hospitals to provide medical care and meet associ- basic elements: (1) collection and storage of different kinds of
ated legal obligations. Patient data must be maintained securely patient data to a database system; (2) creation of a data ware-
over a long period of time and done so in a way that maintains house whereby patient data are integrated into a single data-
patient privacy. Storing data digitally can be more efficient, base with copies of all databases to support real-time analytics;
help reduce medical errors, improve medical device manage- and (3) use of software to perform various methods of analysis
ment, and reduce health care costs.3,14,15 It is important for that together create a clinical decision support system (Fig.
clinicians and nursing staff to voice how patient data can be 4.2). Chapters 40 and 45 describe some NCCU specific solu-
used to improve patient care. In the absence of that, other tions that have been implemented.
more mundane administrative concerns usually take prece-
dence. In the NCCU the clinical need for real-time access to
patient data and clinical decision support systems falls into Planning
three categories. First, NCCU staff need tools to evaluate the Informatics is a rapidly changing in field, and therefore a
effectiveness of treatment(s) meant to modify physiologic department should not be locked in any one system at an
Section I—Background 29

Patient room

Bedside devices
Monitor network

Patient monitor

Patient monitor mainframe

Hospital network

Serial-to-TCP/IP
A B Raid storage drive
Bedside data server

D C Data warehouse

Bedside Lab EHR EEG


data

Clinical decision support Decision support


analytics

Other data collection and


Knowledge advancement Clinical research storage systems

Fig. 4.2  Theoretical data collection infrastructure for the neurocritical care unit. A, Medical devices that monitor patient physiology can plug
into the bedside patient monitor or be transmitted over the network using a serial-to-TCP/IP converter. B, A data server dedicated to collecting and
storing physiologic data can receive data from the patient monitor mainframe or directly from a serial-to-TCP/IP converter that is installed on the
server as a COM port. C, An IT-managed data warehouse receives all forms of patient data and makes it available for multiple purposes. D, Clinical
decision support systems use the data warehouse to facilitate real-time clinical decision making in the intensive care unit. The data warehouse also
supports retrospective analysis of a patient for quality assurance, clinical research, and other aspects of knowledge advancement. COM, Communication;
EEG, electroencephalogram; EHR, electronic health record; IT, information technology; TCP/IP, transmission control protocol/Internet protocol.

infrastructure level. Data should be stored in an open non- emergency department (ED) settings.20 Decisions also should
proprietary format such as Structured Query Language (SQL) be made about whether to integrate the NCCU data with
(Open Database Connectivity [ODBC] compliant) database other ICUs, both in the same institution and other medical
that will allow any system to access the data. Many ICU direc- centers. Creation of such integrative databases may help facili-
tors strive to include an informatics infrastructure when tate syndrome surveillance, decision support, comparative
building a new ICU. This is a great time to do this because effectiveness research, and outcome research.21,22 Data collec-
there will be a capital budget, and essential items such as tion systems and the EHR, however, do not replace verbal
installing power and internet connectivity in patient rooms communication for efficient interdisciplinary exchange of
will be cheapest during the construction phase. An ICU con- patient information in the NCCU but can be used to supple-
struction project also will mean that health care providers will ment this.23
have the attention of hospital administrators and information
technology personnel. It also is vital to create an operating
budget to maintain the infrastructure over time and to Collecting Bedside Device Data
upgrade equipment19 and to decide whether remote data
viewing is important to put such a system in place. There are Overview
commercial products, such as Citrix MetaFrame, that securely Current EHR systems generally are adequate to store clinical
enable this functionality and are gaining favor in ICU and documentation and laboratory, pathology, and radiology
30 Section I—Background

information. However, most EHRs are not designed to capture


and store high-frequency physiologic measurements obtained
Collecting Data from Individual Bedside
from patient monitoring equipment. In the NCCU, invasive Patient Monitors or Devices
and noninvasive monitoring techniques provide continuous It may not always be possible to collect bedside device data
measurement of physiologic parameters, and this high- through a network portal, and many medical devices do not
frequency data can be vital to understand the course of plug into the patient monitor, or if a device does plug into the
critical illness and optimize treatment. Data documented in monitor, not all the data are transferred. Data then need to be
an EHR by the NCCU staff, typically on a hourly basis, are collected from the individual medical device or sometimes
not sufficient to fully represent this physiologic data stream, even each individual patient monitor. Most bedside devices
and therefore it is desirable to establish a thoughtful strategy (including the patient monitor) are equipped with RS-232
to collect and use data from bedside medical devices such (digital) and analog (waveform) data output ports for auto-
as ventilators, patient monitors, infusion pumps, and other matic data output capabilities. Specifications for data com-
neurospecific monitors. Bedside medical device data are munication are generally available in the device’s technical
generally the most difficult data to obtain, whereas continu- manual or can be obtained from the device manufacturer.
ous electroencephalographic and associated patient video Medical devices output data usually as a comma-delimited
data create the greatest demands on network and storage text string (e.g., 21, 15.6, 78.24, 12, 15) every few seconds. This
capacity. The informatics infrastructure needed to collect text string must be converted into data that fill specific fields
and store continuous electroencephalographic data in the in a database, which requires knowledge about what each
ICU environment is described in detail elsewhere.24 number in the text string represents and in what field in the
database it should be stored. This translation is referred to as
a device interface (a small bit of software that automatically
Collecting Bedside Patient Monitor Data converts data from the device into sensible data for a particu-
Via a Network Portal lar database). Provided it is known what data output is sent
Many bedside medical devices are plugged into the standard from the machine, what each number means, and where it
patient monitor, which serves to display all the digital and goes in the database, actually writing a device interface is not
waveform data on a single bedside display in real time. complicated.
Most clinical information systems from patient monitor One limitation of RS-232 for bedside device communica-
vendors store these data up to 72 hours for clinical review tion is that there are many different ways to send data over the
purposes before they are deleted forever. Some may transfer serial interface. Several different pin-out and connector styles
hourly values into an EHR for nurse verification. Networked exist, and there are multiple options for baud rate, parity, stop
patient monitors facilitate patient data display in areas other bits, handshaking, flow control, and signaling.27 For example,
than the patient’s room, such as a central station or the many bedside devices output the data string at a set time
nursing pod area, by allowing other systems to receive that frequency without prompting, whereas with other devices,
data from a central server. Networked systems represent one such as the patient bedside monitor, the software interface
potential strategy to collect all data on every bedside patient must send a coded request to receive data. Therefore the
monitor from a single networked portal at a high temporal device interface must be written to accommodate the specifics
resolution. of each device or patient monitor configuration. Some newer
Some medical device manufacturers, including Philips modes to output data from devices include Universal Serial
Healthcare (Andover, MA) and GE Healthcare (Chalfont St. Bus (USB), 802.3 (Ethernet), IEEE 11073, or even wireless
Giles, UK), have developed their own proprietary solutions (e.g., 802.11 b/g, Bluetooth) data communication capabilities.
for automated data acquisition through this single portal. Because the life span for some medical devices is 10 to 15
Customers rarely use these solutions because they tend to be years, it is unlikely that RS-232 ports will disappear in the
expensive, do not provide adequate data frequency, and near future.
sometimes do not integrate easily with other clinical infor- At Columbia University it was decided to use Bedmaster XA
mation systems. These systems do continue to improve, to collect bedside data in the NCCU, because the company
however, and it is sensible to investigate data collection and also wrote device interfaces for devices that did not connect
storage solutions from the institution’s patient monitor to the bedside monitor, or did so incompletely. For example,
vendor. Third-party products such as DataCaptor (Capsule it is possible to collect all 22 parameters outputted from the
Technologie, Paris, France) are designed to convert the non- Arctic Sun cooling device (Medivance, Louisville, CO). This
standard output from bedside devices into HL7 format, allows caregivers to automatically track the device’s water tem-
which can then be sent to EHR systems such as Cerner Mil- perature, which decreases when the machine works harder to
lennium (Cerner Corporation, Kansas City, MO) and Eclip- maintain body temperature (i.e., fever spike), and other
sys Sunrise Clinical Manager (Eclipsys Corporation, Atlanta, parameters such as ICP and PbtO2. There also is a device inter-
GA). Open-source efforts to collect bedside device data for face for the Camino ICP monitor (Integra Neuroscience,
both research and clinical care purposes is described else- Paramus, NJ), even though this monitor can plug into the
where.25,26 In the Columbia University Medical Center neuro- patient bedside monitor. Many brain injury patients may have
logical ICU, Bedmaster XA (Excel Medical Electronics, an external ventricular drain and a parenchymal ICP monitor.
Jupiter, FL) is used to collect all the bedside patient monitor It is important to distinguish each ICP from the other.
data including parameter data at least every 5 seconds and all However, there are limited methods to maintain the same data
visible waveform data at a resolution of 240 Hz. This system label for each device regardless of where it was plugged in on
works for both the General Electric and Philips patient the patient monitor. Instead the Camino is now to both the
monitors. patient monitor and the secondary system, where plugged in
Section I—Background 31

the data label can be controlled. It is important to understand medical equipment. It is recommended that testing be per-
how the patient monitor handles different device scenarios formed for EMI between mobile wireless communication and
when considering how to collect patient data. medical devices.34 Finally, with many devices and multiple
wireless technologies competing to use the same frequency
spectrum to transmit, crowding can occur that may decrease
Standards in Bedside Device Collection throughput and affect the reliability of data exchange. Device
Technical standards developed for medical device communi- manufacturers that use RF communication should provide an
cation are available. These standards are supposed to simplify analysis of data communication requirements, including esti-
the problem of getting device data into multiple data systems, mated bandwidth utilization at periods of peak and normal
but often are not adopted by device manufacturers. Efforts to usage.
establish a medical device connectivity standard began in the Data overload is one of the greatest informatics chal-
1980s with the Medical Information Bus (MIB). The MIB was lenges in the NCCU for both medical informaticians and
developed by representatives from medical device manufac- intensive care clinicians.35-39 For example, at Columbia Uni-
turers, health care institutions, and academic departments versity NCCU bedside monitors store approximately 200
who advocated “plug-and-play” data sharing among bedside megabytes (MB) per day per bed, whereas continuous elec-
devices irrespective of device vendor.28,29 In 1996 the MIB troencephalograms (EEGs) can generate approximately 1
specification was approved by the Institute of Electrical and gigabyte (GB) per day for EEG alone, and 20  GB per day
Electronic Engineers (IEEE) Standards Board under the name when there is video recording. When multiple systems in
“IEEE 1073 Standard for Medical Device Communica- multiple patient rooms record simultaneously, network per-
tions.”30-32 The IEEE 1073 standard continued to evolve, and formance can slow down or data loss is possible. Modern
in 2004 the International Organization for Standardization Ethernet networks that use 1  GB per second or greater
(ISO) formally ratified IEEE 1073 as “ISO 11073 Point-of- connections between switches and routers should be used
Care Medical Device Communication Standard. Despite this, throughout the network to help avoid this problem. It also
the ISO/IEEE 11073 standard still is not widely used by the is important to check for potential bottleneck areas where
medical device industry. Consequently, most hospitals have older systems that use 10 or 100 MB per second connec-
limited or no integration of bedside device data with their tions may exist (e.g., when networks span new and old
EHR systems and other clinical information systems. buildings).24
Regulatory concerns present another barrier to medical
device connectivity at least in the United States. The U.S. Food
and Drug Administration (FDA) requires a manufacturer of Linking Patients to Their Data
a data collection device to test the connections to each device Medical device connectivity is complex, and this extends
from which it receives data rather than just test to ensure beyond simply the physical connectors and communication
proper implementation of a standard (such as ISO/IEEE protocols. Perhaps more important is to establish patient
11073). This presents an unnecessary burden on the manufac- context and ensure that device(s) data are reliably associated
turer, similar to a computer manufacturer’s having to test the with the correct patient. A wired infrastructure simplifies
connection to every printer rather than just test to the USB patient association because each device can be connected
standard. The FDA realizes the benefits of medical device con- unambiguously to a particular location (i.e., room or bed
nectivity and the regulatory issues are being addressed as part number). Use of existing electronic admission/discharge/
of the mission of the Medical Device Plug and Play (MDPnP) transfer (ADT) systems to map a specific patient to a location
program.33 can link device data to a patient. However, mobile devices
(e.g., infusion pumps) and wireless data communication
introduce additional challenges for patient identification.
Transferring Data Over the Network Some devices allow a patient’s medical record number to be
The simplest configuration to transfer data from a medical entered into the device interface, or provide bar code scanning
device to a computer is to plug a RS-232 cable purchased from capabilities to accomplish the same task.
the local computer store into the RS-232 output of the medical It is essential to consider how data will be linked to a patient
device and then plug the other end into the nine-pin com- before device purchase. There may be an advantage to auto-
munication (COM) port of a regular personal computer. A matic data documentation directly to an electronic medical
serial-to-TCP/IP converter enables one to send data from record (EMR) from devices such as ventilators and infusion
multiple bedside devices over the hospital or local network to pumps; several studies show this is more accurate and saves
a server located in another room or building. Placed in the nursing time.33,40,41 In some ICUs ventilators are always con-
patient’s room and “installed” on the server, it creates multiple nected to the bedside monitor and their data automatically
COM ports that allow many devices to plug into it and trans- transmitted to an EMR for verification. Infusion pumps
fer its data over the network using standard TCP/IP protocols. should have the capability to send streaming data, but not all
These converters are available in many configurations includ- are able to do this; some that do, send anonymous drug and
ing virtual and wireless. dose data to a server to help biomedical departments with asset
Special care is needed when wireless communication is used management and device maintenance. Transmission of anon-
to acquire data from bedside medical devices. First, if mobile ymous data bypasses the Health Insurance Portability and
monitoring solutions rely on battery power, power consump- Accountability Act (HIPAA) and privacy concerns. This allevi-
tion issues must be considered. Second, wireless communica- ates potential headaches for biomedical engineering and infor-
tion devices are a cause of radio frequency (RF) electromagnetic matics departments; however, this does not help clinicians
interference (EMI) that may cause undesirable effects to manage patients. These various issues need to be addressed
32 Section I—Background

before making purchase decisions. No matter the mechanism


used to link device data to patients, local testing and periodic
Data Warehouse Configurations
validation should be performed to ensure accuracy. In this chapter the focus has been on how to get high-frequency
data (i.e., parameter data every 5 seconds) from the patient
monitor and other bedside medical devices to a “server.”
Time Synchronization Servers are simply a class of computers with large computa-
It is critical to synchronize data together from multiple tional and storage capacities that manage, store, and retrieve
devices for accurate bedside device data collection. Each data for other computers or devices.19 A data warehouse is a
device may maintain its own internal clock, and additional collection of decision support technologies to enable better
clocks may exist for the data acquisition system and the hos- and faster decisions42 that is composed of multiple servers and
pital clinical information system. When data that are used for networked data storage to support clinical decision making
time-sensitive analyses (e.g., when does an infused medica- and research.3,43
tion affects vital signs, when do alarms trigger, and what is A data warehouse is where laboratory, imaging, interven-
the duration?) are recorded, it is important to know exactly tion, physiologic, and all other patient databases reside. Tra-
the order and delay between events. The Network Time Proto- ditional data warehouses are set up to bring several different
col (NTP) is the most commonly used Internet time protocol kinds of data (e.g., laboratory and physiologic) together into
and can be used if utilities for time synchronization are oth- a unified database to be used by clinical support software
erwise unavailable. tools. Software programs translate data output from the device
into an intelligent form to allow the data to be added into a
database at medical device interfaces. Similarly to transfer data
Other Data Collection Considerations from one database to another database also requires a software
It is important to anticipate how the data may be used to interface. Database interfaces rely on standards such as HL7
prepare an effective strategy for physiologic data acquisition to streamline the process. An interface engine is software that
in the NCCU. Is the purpose of data collection to facilitate manages all the standardized “messages” being sent back and
clinical care, research, or both? If the data are to be used to forth between databases.44 An alternative and more flexible
provide patient care and there is an existing EHR system in strategy is to create a real-time replica of each database paired
use, it is important to decide whether bedside device data will with software that supports real-time and ad hoc queries from
be automatically transferred to the EHR, and if so, how this decision support software.43 The exact strategy to be used in
will be accomplished. Undoubtedly there will be workflow the NCCU will require discussion with the institutional IT
changes for care providers associated with automatic data col- department and vendors.
lection (e.g., documentation and data review processes). To There are several important considerations to link data-
help anticipate some of the sociotechnical challenges that may bases. First, performance bottlenecks are not specific to the
arise, both technologic and associated with personnel or network and can occur at the server level. There needs to be
workflow, the following questions should be addressed: adequate storage capacity for primary data collection of
patient monitors and medical devices, a backup of these data
 How often will data be transferred to the existing EHR
in case of hardware failure, and replicas or integrated data-
system?

bases that will support clinical support applications to prevent
Will manual verification be required before the data are
bottlenecks. Second, the file server and associated disk systems
included in the patient’s record?

need to be sufficiently robust to avoid performance slow-
For each data element that is currently documented in the
downs during data collection, data review, and other mainte-
patient’s record, does that element exist in the bedside
nance functions. Third, file servers used in critical care should
device’s output stream?

be capable of operating without interruption, despite common,
Will filters be applied to the data to reduce artifact
predictable component failures. Strategies such as redundant
(i.e., “noise” in the signal) or select representative measure-
power supplies and redundant arrays of independent disks
ments?

(RAIDs) rather than storing data on single hard drives may
Will the same type of measurement be obtained simulta-
prevent this. Though there are many variations of a RAID
neously from separate devices (e.g., respiratory rate col-
drive, in its basic form data are divided and written across
lected from a patient monitor and a mechanical ventilator)?
multiple devices simultaneously and redundantly. This can
How will these measurements be dealt with?

increase performance and provide fault tolerance.19,24
How will downtime procedures be created and enforced
(for the EHR system as well as the data collection system)?
Some clinical information systems may not be capable of han-
Clinical Decision Support
dling second-by-second or even minute-by-minute recording Research still is needed to determine what data should be col-
of physiologic variables. If data are to be stored in a secondary lected at the bedside, and how information is best summarized
or research database instead of or in addition to an EHR and presented to health care providers in the NCCU. Innova-
system, several other questions should be considered: tive graphic displays show particular promise but have yet to
be adopted on a large scale.37,45-49 Display of physiologic trends
 How will data be stored (e.g., flat files, relational database, also is important in critical care, perhaps more so than simple
XML database)? numeric displays or threshold alarms. Adoption of automatic
 How will data backups be performed? data acquisition from bedside devices will finally enable the
 How frequently can or should data be acquired and stored research and patient care environment that Peters and Stacy
(e.g., 500 Hz, 1 sec, 5 sec, 1 min, 15 min, 1 hr)? anticipated almost a half-century ago:
Section I—Background 33

“The ability to combine analogue and digital computation 5. Mador R, Shaw N. The impact of a critical care information system (CCIS)
on time spent charting and in direct patient care by staff in the ICU: a
… moves the field of analysis of physiologic data to that
review of the literature. Int J Med Inform 2009;78(7):435–45.
point where mathematical postulates can be set up and 6. Gill JM. EMRs for improving quality of care: promise and pitfalls.
tested in the real situation. The enhancement of this skill [comment] Fam Med 2009;41(7):513–15.
and propensity will permit us to take physiology out of the 7. Donati A, Gabbanelli V, Pantanetti S, et al. The impact of a clinical
data-collecting-for-data’s-sake stage to the more imagina- information system in an intensive care unit. J Clin Monit Comput 2008;
22(1):31–6.
tive methods now employed in physical sciences. This step 8. Morris G, Gardner R. Computer applications. In: Hall, J, Schmidt G,
will mean as much to the advancement of knowledge in Wood L, editors. Principles of critical care. McGraw-Hill: New York; 1992.
physiology as it has meant in the physical sciences.”50 p. 500–14.
9. Gather U, Imhoff M, Fried R. Graphical models for multivariate time series
from intensive care monitoring. Stat Med 2002;21(18):2685–2701.
Chapters 40 and 45 address these issues in further detail.
10. Imhoff M, Fried R, Gather U, et al. Dimension reduction for physiological
However, how the data will be used needs to be considered to variables using graphical modeling. AMIA Annu Symp Proc 2003:313–17.
collect it in a manner that enables proper use. Currently there 11. Imhoff M, Fried R, Gather U. Detecting relationships between physiological
is little regulatory guidance on clinical decision support variables using graphical modeling. Proc AMIA Symp 2002:340–4.
systems despite reports that have surfaced about their poten- 12. De Turck F, Decruyenaere J, Thysebaert P, et al. Design of a flexible platform
for execution of medical decision support agents in the intensive care unit.
tial for harm.51 Two independent organizations are address- Comput Biol Med 2007;37(1):97–112.
ing this52,53 and will hopefully provide some certification 13. Jennings D, Amabile T, Ross L. Informal assessments: data-based versus
guidelines. Thus the regulatory issues ahead are certain to theory-based judgments. In: Kahnemann D, Slovic P, Tversky A, editors.
become more stringent, but the actual outcome remains Judgments under uncertainty: heuristics and biases. Cambridge University
Press: New York; 1982. p. 211–30.
unpredictable.
14. Adhikari N, Lapinsky S. Medical informatics in the intensive care unit:
overview of technology assessment. J Crit Care 2003;18(1):41–7.
Conclusion 15. Clemmer TP. Computers in the ICU: where we started and where we are
now. J Crit Care 2004;19(4):201–7.
To create the infrastructure necessary for a successful neuro- 16. Campion TR Jr, Waitman LR, May AK, et al. Social, organizational, and
contextual characteristics of clinical decision support systems for intensive
monitoring and clinical informatics program in the NCCU is insulin therapy: a literature review and case study. Int J Med Inform 2010;
feasible, and there are enough commercial resources available 79(1):31–43.
to help with this process. The key to success is to recognize 17. Peelen L, de Keizer NF, Jonge E, et al. Using hierarchical dynamic Bayesian
that there are actually four problems to consider when the networks to investigate dynamics of organ failure in patients in the intensive
care unit. J Biomed Inform 2010;43(2):273–86.
infrastructure is developed: data collection, data visualization,
18. Ji SY, Smith R, Huynh T, et al. A comparative analysis of multi-level
data analysis, and decision support. The first and most impor- computer-assisted decision making systems for traumatic injuries. BMC Med
tant step is data collection, because without the data nothing Inform Decis Mak 2009;9:2.
else is possible. It is critical to clearly articulate to hospital 19. Chou D, Sengupta S. Infrastructure and security. In: Practical guide to
administrators and information technologists what data the clinical computing systems: design, operations, and infrastructure. Payne T,
editor. Burlington, MA: Academic Press; 2008.
NCCU needs, how it will be used, and what positive impact it 20. Payne T. Architecture of clinical computer systems. In Payne T, editor.
should have on patient care, for example, better outcomes or Practical guide to clinical computing systems: design, operations, and
reduced length of stay. Each hospital constituent group has infrastructure. Burlington, MA: Academic Press; 2008.
different needs for the same data, and unless the NCCU needs 21. Herasevich V, Pickering BW, Dong Y, et al. Informatics infrastructure for
syndrome surveillance, decision support, reporting, and modeling of critical
are understood, the required data may turn out to be unavail-
illness. Mayo Clin Proc 2010;85(3):247–54.
able or inaccessible. Therefore NCCU staff should work with 22. Platt R, Takvorian SU, Septimus E, et al. Cluster randomized trials in
the information technology group to make sure that network comparative effectiveness research: randomizing hospitals to test methods
and storage concerns are addressed up front and that there is for prevention of healthcare-associated infections. Med Care 2010;48
an equipment maintenance plan. The data should be stored (6 Suppl):S52–7.
23. Collins SA, Bakken S, Vawdrey DK, et al. Model development for EHR
in a manner such that it can be automatically exported in a interdisciplinary information exchange of ICU common goals. Int J Med
standard format to any number of viewing or analysis systems. Inform 2010; Oct 23. Epub ahead of print.
In addition, communication between different health care 24. Kull L, Emerson R. Continuous EEG monitoring in the intensive care unit:
providers (e.g., nurses, pharmacists, physicians) is necessary technical and staffing considerations. J Clin Neurophysiol 2005;22(2):
107–18.
to ensure a synchronized and efficient system.54 These steps
25. Kroth PJ, Belsito A, Overhage JM, et al. Bedside vital signs capture for the
will ensure that the infrastructure will withstand the test of non-ICU setting—an open source, PC-based solution. Proc AMIA Symp
time and subsequently enable leverage of whatever technolo- 2001:344–8.
gies emerge in the years to come. 26. Vawdrey DK, Hum RS. OpenMDC: an open-source framework for medical
device communication. In: AMIA Annu Symp Proc. Washington, DC; 2008.
27. TAL tech: Instrumental software solutions. [cited 1/2/2010]; Available from:
References www.taltech.com/resources/intro-sc.html.
28. Gardner RM, Hawley WL, East TD, et al. Real time data acquisition:
1. Mayer S, Chong J. Critical care management of increased intracranial recommendations for the Medical Information Bus (MIB). Int J Clin Monit
pressure. J Intensive Care Med 2002;17(2):55–67. Comput 1991;8(4):251–8.
2. Payne T. Introduction and overview of clinical computing systems within a 29. Shabot MM. Standardized acquisition of bedside data: the IEEE P1073
medical center. In: Payne T, editor. Practical guide to clinical computing medical information bus. Int J Clin Monit Comput 1989;6(4):197–204.
systems: design, operations, and infrastructure. Burlington, MA; Academic 30. Garnsworthy J. Standardizing medical device communications: the Medical
Press; 2008. Information Bus. Med Device Technol 1998;9(3):18–21.
3. Martich G, Waldmann C, Imhoff M. Clinical informatics in critical care. 31. Kennelly RJ. Improving acute care through use of medical device data. Int J
J Intensive Care Med 2004;19(3):154–163. Med Inf 1998;48(1-3):145–9.
4. Varon J, Marik PE. Clinical information systems and the electronic 32. Kennelly RJ, Gardner RM. Perspectives on development of IEEE 1073: the
medical record in the intensive care unit. Curr Opin Crit Care 2002;8(6): Medical Information Bus (MIB) standard. Int J Clin Monit Comput
616–24. 1997;14(3):143–9.
34 Section I—Background

33. Dalto JD, Johnson KV, Gardner RM, et al. Medical information bus usage for 42. Chaudhuri S, Dayal U. An overview of data warehousing and OLAP
automated IV pump data acquisition: evaluation of usage patterns. Int J Clin technology. ACM Sigmod Record 1997;26(1):65–74.
Monit Comput 1997;14(3):151–4. 43. Chelico J, Wajngurt D. Architectural design of a data warehouse to support
34. Bruns B, Dimantha S. Evaluating EMI in a multi-hospital facility. Biomed operational and analytical queries across disparate clinical databases. AMIA
Instrum Technol 2006;Suppl:40–2. Annu Symp Proc 2007;11:901.
35. Alberdi E, Gilhooly K, Hunter J, et al. Computerisation and decision making 44. Payne T, Hoath J. Creating and supporting interfaces. In Payne T, editor.
in neonatal intensive care: a cognitive engineering investigation. J Clin Monit Practical guide to clinical computing systems: design, operations, and
Comput 2000;16(2):85–94. infrastructure. Burlington, MA: Academic Press; 2008.
36. Calvelo D, Chambrin MC, Pomorski D, et al. Towards symbolization using 45. Cole WG, Stewart JG. Metaphor graphics to support integrated decision
data-driven extraction of local trends for ICU monitoring. Artif Intel Med making with respiratory data. Int J Clin Monit Comput 1993;10(2):91–100.
2000;19(3):203–23. 46. Horn W, Popow C, Unterasinger L. Support for fast comprehension of ICU
37. Clemmer TP, Gardner RM. Data gathering, analysis, and display in critical data: visualization using metaphor graphics. Methods Inf Med 2001;40(5):
care medicine. Respir Care 1985;30(7):586–601. 421–4.
38. Cole WG, Stewart JG. Human performance evaluation of a metaphor 47. Syroid ND, Agutter J, Drews FA, et al. Development and evaluation of a
graphic display for respiratory data. Methods Inf Med 1994;33(4):390–6. graphical anesthesia drug display. Anesthesiology 2002;96(3):565–75.
39. Sims AJ, Pay, DA, Watson BG. An architecture for the automatic acquisition 48. Powsner SM, Tufte ER. Graphical summary of patient status. Lancet 1994;
of vital signs by clinical information systems. IEEE Trans Inf Technol 344(8919):386–9.
Biomed 2000;4(1):74–5. 49. Drews FA, Westenskow DR. The right picture is worth a thousand numbers:
40. Sapo M, Wu S, Asgari S, et al. A comparison of vital signs charted by nurses data displays in anesthesia. Hum Factors 2006;48(1):59–71.
with automated acquired values using waveform quality indices. J Clin 50. Peters RM, Stacy RW. Automatized clinical measurement of respiratory
Monit Comput 2009;23(5):263–71. parameters. Surgery 1964;56:44–52.
41. Vawdrey D, Gardner RM, Evans RS, et al. Assessing data quality in
manual entry of ventilator settings. J Am Med Informat Assoc 2007;14(3): A complete list of references for this chapter can be found online at
295–303. www.expertconsult.com.
Section—Background 34.e1

References 26. Vawdrey DK, Hum RS. OpenMDC: an open-source framework for medical
device communication. In: AMIA Annu Symp Proc. Washington, DC; 2008.
1. Mayer S, Chong J. Critical care management of increased intracranial 27. TAL tech: Instrumental software solutions. [cited 1/2/2010]; Available from:
pressure. J Intensive Care Med 2002;17(2):55. www.taltech.com/resources/intro-sc.html.
2. Payne T. Introduction and overview of clinical computing systems within a 28. Gardner RM, Hawley WL, East TD, et al. Real time data acquisition:
medical center. In: Payne T, editor. Practical guide to clinical computing recommendations for the Medical Information Bus (MIB). Int J Clin Monit
systems: design, operations, and infrastructure. Academic Press: Burlington, Comput 1991;8(4):251–8.
MA;2008. 29. Shabot MM. Standardized acquisition of bedside data: the IEEE P1073
3. Martich G, Waldmann C, Imhoff M. Clinical informatics in critical care. medical information bus. Int J Clin Monit Comput 1989;6(4):197–204.
J Intensive Care Med 2004;19(3):154. 30. Garnsworthy J. Standardizing medical device communications: the Medical
4. Varon J, Marik PE. Clinical information systems and the electronic medical Information Bus. Med Device Technol 1998;9(3):18–21.
record in the intensive care unit. Curr Opin Crit Care 2002;8(6):616–24. 31. Kennelly RJ. Improving acute care through use of medical device data. Int J
5. Mador R, Shaw N. The impact of a critical care information system (CCIS) Med Inf 1998;48(1-3):145–9.
on time spent charting and in direct patient care by staff in the ICU: a 32. Kennelly RJ, Gardner RM. Perspectives on development of IEEE 1073: the
review of the literature. Int J Med Inform 2009;78(7):435–45. Medical Information Bus (MIB) standard. Int J Clin Monit Comput
6. Gill JM. EMRs for improving quality of care: promise and pitfalls. 1997;14(3):143–9.
[comment] Fam Med 2009;41(7):513–15. 33. Dalto JD, Johnson KV, Gardner RM, et al. Medical information bus usage for
7. Donati A, Gabbanelli V, Pantanetti S, et al. The impact of a clinical automated IV pump data acquisition: evaluation of usage patterns. Int J Clin
information system in an intensive care unit. J Clin Monit Comput Monit Comput 1997;14(3):151–4.
2008;22(1):31–6. 34. Bruns B, Dimantha S. Evaluating EMI in a multi-hospital facility. Biomed
8. Morris G, Gardner R. Computer applications. In: Hall, J, Schmidt G, Wood Instrum Technol 2006;Suppl:40–2.
L, editors. Principles of critical care. McGraw-Hill: New York; 1992. 35. Alberdi E, Gilhooly K, Hunter J, et al. Computerisation and decision making
p. 500–14. in neonatal intensive care: a cognitive engineering investigation. J Clin Monit
9. Gather U, Imhoff M, Fried R. Graphical models for multivariate time series Comput 2000;16(2):85–94.
from intensive care monitoring. Stat Med 2002;21(18):2685–2701. 36. Calvelo D, Chambrin MC, Pomorski D, et al. Towards symbolization using
10. Imhoff M, Fried R, Gather U, et al. Dimension reduction for physiological data-driven extraction of local trends for ICU monitoring. Artif Intel Med
variables using graphical modeling. AMIA Annu Symp Proc 2003:313–17. 2000;19(3):203–23.
11. Imhoff M, Fried R, Gather U. Detecting relationships between physiological 37. Clemmer TP, Gardner RM. Data gathering, analysis, and display in critical
variables using graphical modeling. Proc AMIA Symp 2002:340–4. care medicine. Respir Care 1985;30(7):586–601.
12. De Turck F, Decruyenaere J, Thysebaert P, et al. Design of a flexible platform 38. Cole WG, Stewart JG. Human performance evaluation of a metaphor
for execution of medical decision support agents in the intensive care unit. graphic display for respiratory data. Methods Inf Med 1994;33(4):390–6.
Comput Biol Med 2007;37(1):97–112. 39. Sims AJ, Pay, DA, Watson BG. An architecture for the automatic acquisition
13. Jennings D, Amabile T, Ross L. Informal assessments: data-based versus of vital signs by clinical information systems. IEEE Trans Inf Technol
theory-based judgments. In: Kahnemann D, Slovic P, Tversky A, editors. Biomed 2000;4(1):74–5.
Judgments under uncertainty: heuristics and biases. Cambridge University 40. Sapo M, Wu S, Asgari S, et al. A comparison of vital signs charted by nurses
Press: New York; 1982. p. 211–30. with automated acquired values using waveform quality indices. J Clin
14. Adhikari N, Lapinsky S. Medical informatics in the intensive care unit: Monit Comput 2009;23(5):263–71.
overview of technology assessment. J Crit Care 2003;18(1):41–7. 41. Vawdrey D, Gardner RM, Evans RS, et al. Assessing data quality in manual
15. Clemmer TP. Computers in the ICU: where we started and where we are entry of ventilator settings. J Am Med Informat Assoc 2007;14(3):295–303.
now. J Crit Care 2004;19(4):201–7. 42. Chaudhuri S, Dayal U. An overview of data warehousing and OLAP
16. Campion Jr TR, Waitman LR, May AK, et al. Social, organizational, and technology. ACM Sigmod Record 1997;26(1):65–74.
contextual characteristics of clinical decision support systems for intensive 43. Chelico J, Wajngurt D. Architectural design of a data warehouse to support
insulin therapy: a literature review and case study. Int J Med Inform operational and analytical queries across disparate clinical databases. AMIA
2010;79(1):31–43. Annu Symp Proc 2007;11:901.
17. Peelen L, de Keizer NF, Jonge E, et al. Using hierarchical dynamic Bayesian 44. Payne T, Hoath J. Creating and supporting interfaces. In: Payne T, editor.
networks to investigate dynamics of organ failure in patients in the intensive Practical guide to clinical computing systems: design, operations, and
care unit. J Biomed Inform 2010;43(2):273–86. infrastructure. Burlington, MA; Academic Press; 2008.
18. Ji SY, Smith R, Huynh T, et al. A comparative analysis of multi-level 45. Cole WG, Stewart JG. Metaphor graphics to support integrated decision
computer-assisted decision making systems for traumatic injuries. BMC Med making with respiratory data. Int J Clin Monit Comput 1993;10(2):91–100.
Inform Decis Mak 2009;9:S6. 46. Horn W, Popow C, Unterasinger L. Support for fast comprehension of ICU
19. Chou D, Sengupta S. Infrastructure and security. In: Practical guide to data: visualization using metaphor graphics. Methods Inf Med
clinical computing systems: design, operations, and infrastructure. Payne T, 2001;40(5):421–4.
editor. Burlington, MA: Academic Press; 2008. 47. Syroid ND, Agutter J, Drews FA, et al. Development and evaluation of a
20. Payne T. Architecture of clinical computer systems. In: Payne T, editor. graphical anesthesia drug display. Anesthesiology 2002;96(3):565–75.
Practical guide to clinical computing systems: design, operations, and 48. Powsner SM, Tufte ER. Graphical summary of patient status. Lancet
infrastructure. Burlington, MA: Academic Press; 2008. 1994;344(8919):386–9.
21. Herasevich V, Pickering BW, Dong Y, et al. Informatics infrastructure for 49. Drews FA, Westenskow DR. The right picture is worth a thousand numbers:
syndrome surveillance, decision support, reporting, and modeling of critical data displays in anesthesia. Hum Factors 2006;48(1):59–71.
illness. Mayo Clin Proc 2010;85(3):247–54. 50. Peters RM, Stacy RW. Automatized clinical measurement of respiratory
22. Platt R, Takvorian SU, Septimus E, et al. Cluster randomized trials in parameters. Surgery 1964;56:44–52.
comparative effectiveness research: randomizing hospitals to test methods 51. Tsai TL, Fridsma DB, Gatti G. Computer decision support as a source of
for prevention of healthcare-associated infections. Med Care 2010;48 interpretation error: the case of electrocardiograms. J Am Med Inform Assoc
(6 Suppl):S52–7. 2003;10:478–83.
23. Collins SA, Bakken S, Vawdrey DK, et al. Model development for EHR 52. Healthcare Information Technology Standards Panel (HITSP), New York:
interdisciplinary information exchange of ICU common goals. Int J Med American National Standards Institute.
Inform 2010; Oct 23. [Epub ahead of print] 53. Certification Commisssion for Health Information Technology (CCHIT),
24. Kull L, Emerson R. Continuous EEG monitoring in the intensive care unit: Chicago, IL.
technical and staffing considerations. J Clin Neurophysiol 54. Pingenot AA, Shanteau J, Sengstacke LT. Description of inpatient medication
2005;22(2):107–18. management using cognitive work analysis. Comput Inform Nurs
25. Kroth PJ, Belsito A, Overhage JM, et al. Bedside vital signs capture for the 2009;27(6):379–92.
non-ICU setting—an open source, PC-based solution. Proc AMIA Symp
2001:344–8.
Chapter
5  
I

Nursing and Education


DaiWai M. Olson

profession. Wreszinski4 translated the Ebbers papyrus such


Introduction that future English versions report “wet nurse” as the figure
Nurses and physicians come from two distinctly different pro- who provides milk to a child. Historians find no female ver-
grams. In the neurocritical care unit (NCCU) they work side sions of a nurse’s role as an attendant or assistant to the physi-
by side but may not have been provided with the opportunity cian. The nosocomii were men who were educated by
to understand each other’s roles. The purpose of this chapter physicians and taught how to run a hospital; the role of the
is to provide a common ground of understanding for nurses nosocomii (from which we derive nosocomial) is not fully
and physicians to help improve multimodal monitoring described but is thought to be the first ancestor of modern
systems by exploring the role of nursing in patient monitoring nursing.
and how nursing education influences that role. More modern times provide a few clues to the emerging
role of nursing. The physician’s role evolved over time and was
symbolized through Asclepius, the god of medicine and
Historical Perspective of Nursing healing. The rod of Asclepius often is shown with a coiled
snake. Of the five daughters of Asclepius, it is Hygeia, goddess
and Monitoring of health, from whom many believe the modern nurse descends
Almost a century ago George Santayana1 wrote, “Those who (Fig. 5.1). Drawings and sculptures of Hygeia often depict her
cannot remember the past are condemned to repeat it.” The holding a snake, cup, or both. Further symbolism of the rod
history of nursing parallels the development of patient moni- of Asclepius is the caduceus. This symbol, which has come to
toring and equally affords an insight to predict some of the represent medicine, shows twin snakes (or a two-headed
future directions in patient monitoring. Today nurses view the snake) coiled about the rod of Asclepius. The caduceus was
terms multitasking and multimodal monitoring not as new adopted in the early 1900s to represent medicine and often is
terms, but as evolving paradigms.2 In this chapter the histori- confused with the twin snakes and winged staff of Hermes (a
cal influence of medicine on the current role of the nurse symbol of commerce). Controversy exists whether the snake
serves as a mechanism to understand how nursing can represents the twin battles of life and death, or more directly
optimally contribute to emerging trends in neurologic the practice of removing worms from open wounds by
monitoring. winding them about a small staff.

Ancient Past Immediate Past


In ancient times nursing and medicine were intertwined, and During the middle ages the role of God in health care expanded
there were healers who brewed medicinal teas and tended to and in times of plague, men and women devoted to God began
injuries with a combination of mysticism, herbalism, and to house and care for the sick. Most historical scholars believe
remedies handed down through oral history.3 After each cup that the nun’s habit of the 15th and 16th centuries is the fore-
of “medicine” and after each broken bone was set, the healers runner to the nurses’ cap of the mid-20th century. Nursing
monitored their patients and observed their successes and care was a separated workforce, both by sex and time. Male
failures that they shared with other healers. When human nurses associated with religious brotherhoods and female
societies settled down and towns were established, medical nurses associated with sisterhoods often were separated by
care was regimented and evolved into a discipline. distances that were not easily crossed.5
As the base of medical knowledge expanded, humankind The end of the Middle Ages marked the onset of modern
began to document the effect of various treatments. Oral history and a further evolution in the healing arts. In particu-
history or direct observation no longer could adequately lar global commerce, increasing life span, growing population
transfer medical knowledge that now was shared across time centers, and war created new opportunities and challenges for
and distance in written form. Although physical evidence the medical team. During this time it was realized that a physi-
exists to support splinting of bones, herbal remedies, and even cian alone could not attend to the sick and wounded. Instead,
trepanation, much of known early medical history is attrib- nurses around the world took on this role. Florence Nightin-
uted to Egyptian papyrus writings.3 Several key writings gale provides one of the earliest and most resilient personifica-
provide clues to the role of nursing in emerging societies. tions of the nursing profession and the role of nursing care in
Except for midwifery, nursing was predominantly a male patient outcome. She had been educated in mathematics and
© Copyright 2013 Elsevier Inc. All rights reserved. 35
36 Section I—Background

histories and verbal reports of facts were replaced with short


notes. Notes become cumbersome and were placed in charts,
which became disorganized and were replaced with tab-
separated sections. Physicians wrote orders for nurses to
follow (being careful to press down hard enough to ensure
that their notes filtered through multiple layers of carbon-
copy paper. Electronic medical records (EMRs) now dominate
the field and provide the opportunity to think of data acquisi-
tion beyond traditional boundaries.
Changes in how we observe and record data have paral-
leled the changes in who records and observes data. After the
U.S. Civil War, nursing became a primarily female occupa-
tion. The “Angels of the Battlefield” became famous leaders.
Dorothea Dix became the first female superintendent of the
Union Army, Mary Todd Lincoln advocated for nursing
before and after her husband’s assassination, and Clara
Barton became the founder of the American Red Cross. With
each new war the face of nursing changed again. By the end
of World War II, nearly 60,000 women joined the military
nurse corps. With the advantage of rapid transport, medical
evacuation, and mobile army surgical hospital (MASH) tech-
nology, men became trained as corpsmen in subsequent
wars. Many of those corpsmen turned to nursing as a postwar
Fig. 5.1  Asclepius and Hygeia. profession.7
Education of nurses has evolved quickly to meet the growing
demand for what Nightingale predicted: “a really observing
through careful observation was able to demonstrate a signifi- nurse.” In 1873 Linda Richards became the first nurse to grad-
cant change in outcomes by modifying the postoperative care uate as a Nurse with a Diploma in the United States. Thirty
and environment of soldiers during the Crimean War. By war’s years later, North Carolina became the first state to require
end she had penned texts on care and conduct in both military nursing licensure. By 1956 Columbia University became the
and civilian hospitals and helped to establish a center for first school to award a master’s degree in nursing, and in 1965,
nursing training. After the war she wrote Notes on Nursing, the University of Colorado established the first nurse practi-
which provided details on how nurses should care for and tioner position. Although nurse educators and nurse scientists
monitor patients in their wards.6 In this book her words began entering academia with doctoral degrees in affiliated
provide a glimpse of the future: areas (e.g., biology, education, psychology), the first PhD pro-
grams specifically in nursing were established in 1954.7
“What you want are facts, not opinion—for what can have any Currently nurses have a wide range of programs and career
opinion of any value as to whether the patient is better or worse, paths that include certification and specialization that range
excepting the constant medical attendant, or the really observing from the licensed practical nurse (LPN) to the doctorally pre-
nurse?” F. Nightingale (1860) pared nurse. Table 5.1 provides a list of titles and descriptions
for professional nurses with advanced education to give a
This insight forecasts two needs in the establishment of fact general understanding of the primary role designated to each
and a foundation of monitoring in neurocritical care. First, discipline.
the phrase “constant medical attendant” implies a need to
obtain data without gaps across time. Second is the use of the
word “observing” as a method to obtain facts about the patient Nursing Theory—Practice
rather than provide opinions.
and Process
The discipline of nursing is a unique blend of art and science
Recent History that evolved over many years even before nursing education
In the last century nursing and medicine have witnessed an became compulsory.8,9 The various views on epistemology in
astounding evolution in the ability to acquire and store data. nursing are summarized by Walker and Avant.10 Empirical
Although nothing has replaced the utility of direct observa- knowing is guided by practice and observation. In the empiri-
tion, new tools have been developed to enhance observational cal realm, knowledge is created from an extension of the
skills and provide for continuous assessment. Feeling for the senses; it arises from experiment.11 The need for nursing
threadiness of a pulse was replaced with blood pressure aus- knowledge as a separate discipline was a major catalyst in the
cultation, then noninvasive blood pressure cuffs and subse- evolutionary process of nursing.12 As nurses became educated
quently with continuous intra-arterial blood pressure they contributed to the fundamental knowledge base of
monitoring and now an effort to use noninvasive blood flow nursing; nursing theory emerged as the driving force to test
monitoring. and explain nursing practice.13
Just as the tools to find facts have evolved, so have the The advantages to this approach are well described.14,15
methods to communicate and record these facts. Oral Nursing science provides the best evidence for nursing care of

Table 5.1.  A Short Description of Common Nursing Titles
Certified Registered Doctorate in
Registered Nurse Clinical Nurse Specialist Nurse Practitioner Nurse Anesthetist Nursing Practice Doctor of Philosophy
(RN) (CNS)* (NP)* (CRNA) (DNP) (PhD)
Career focus Diploma, associate Expert in clinical nursing, Midlevel provider of Midlevel provider of Leadership roles as Nursing research
or baccalaureate master’s degree direct care, master’s anesthesia care, an advanced-
degree in nursing degree master’s degree practice nurse
Degree Entry to practice as A CNS is an advanced- An advanced-practice Advanced-practice Nursing leaders in To prepare nurse
objectives a nursing practice nurse familiar with nurse who diagnoses nursing. CRNAs interdisciplinary scientists to develop
professional the theory and research and manages common provide anesthetics health care new knowledge for
related to a nursing acute and stable chronic to patients in every teams. Using the the science and
specialty area, such as health problems. NPs practice setting, and tools and skills practice of nursing.
critical care. Traditionally, a perform comprehensive for every type of specific to Graduates will lead
CNS influences patient physical exams, order surgery or procedure. translating interdisciplinary
outcomes through direct and interpret diagnostic They are the sole evidence gained research teams,
clinical practice, tests, obtain specialty anesthesia providers through nursing design, and conduct
collaboration, education, consults, and perform in two thirds of all research into research studies, and
research, consultation and and prescribe rural hospitals, and practice, they disseminate
system leadership. CNSs are therapeutic measures, the main provider of improve systems knowledge for nursing
uniquely prepared, by including most classes anesthesia to of care, and and related disciplines,
experience and education, of medications. NPs, expectant mothers measure particularly addressing
to manage the complexities individually accountable and to men and outcomes of trajectories of chronic
of negotiating complex for their practice and women serving in patient groups, illness and care
health care delivery decisions, collaborate the U.S. Armed populations, and systems.
systems. closely with physicians. Forces. communities.
Curriculum Basic nursing care Improve outcomes for Direct care provider with Direct care provider Translation of Independent researcher
focus patients by influencing prescriptive authority with prescriptive evidence to
systems level, team, and authority practice at the
patient change systems level
Core Nursing assessment, Applied statistics, research Applied statistics. Neurobiology, spinal Evidence-based Philosophy of science
courses† nutrition, design and methodology. Graduate-level and epidural practice, applied and theory
pharmacology, Graduate-level pharmacology, anesthesia, and statistics data development,
human growth pharmacology, assessment, assessment, and advanced airway driven health care advanced research
and development, and pathophysiology. CNS pathophysiology, and management. improvement, design, statistics and
psychology, students are required to courses in an elected Graduate-level financial data analysis
maternal/child, take specialty focus courses specialty (e.g., pharmacology. management and longitudinal and
microbiology, and advanced health management of CRNAs are required budget planning, qualitative research
communication, assessment. critically ill patients, for to achieve a number effective methods. PhD
anatomy and the acute care NP, of clinical internship leadership, health students are required
physiology, and versus child health in hours (varies by systems to take courses related
mental health family care, for the state). transformation to their intended area
nursing family NP) of research.
Prescriptive No No‡ Yes Yes No‡ No
authority
Point of High school BSN BSN BSN BSN or MSN BSN or MSN
entry diploma
Program 2-4 years 2-3 years 2-3 years 2-3 years 2-3 years 4-7 years
length§
Section I—Background

*There are a variety of subspecialties (i.e., family practice, acute care).



Course requirements vary by state and university. The core requirements are provided here only as a sample of general requirements.

May have prescriptive authority.
37

§
Full-time student.
38 Section I—Background

patients.16 Caring, although not solely in the domain of for action, and is able to recognize the entirety of the situa-
nursing, requires a theoretical basis, just as curing, not solely tion.19 To accomplish these performance aspects, the nurse
in the domain of medicine, requires a theoretical basis.17,18 must possess critical thinking skills and have acquired new
Theory provides a foundation of knowledge about a problem schema.21 The progress from novice nurse to expert nurse
or situation; what is known, what are the components, media- typically occurs over many years. As nurses gain experience
tors, moderators, and co-variables. Importantly, theory pro- they develop enhanced critical thinking skills, which leads to
vides the platform from which new hypotheses may be the development of new schema and they become more adept
generated and tested. Nursing knowledge is not limited to at recognizing and interpreting information.18,19
academic research or rigid theories. Theories that are not sup- A clinical example provides additional meaning to these
ported by scientific testing give way to those that hold up to five stages. It is 2 pm and the nurse prepares to turn her
such scrutiny. patient in bed. The novice states, “It is 2 pm; we turn patients
Nurses at all levels are responsible for generating and testing at 2 pm,” but does not recognize the implications of the
theories, and for participating in research. Most recently, the patient’s heart rate. The advanced beginner recognizes the
push for evidence-based practice (EBP) has emerged as a patient’s heart rate as she turns the patient, and then (having
dominant paradigm in nursing care. Understanding the role seen results in the past) administers analgesia. The competent
of nursing theory in nursing education and bedside care pro- nurse notes the heart rate before she turns her patient; she
vides insight for members of other disciplines that interact plans to administer analgesia and wait for a more normal
with nurses. One of the most widely explored theories in criti- heart rate before turning the patient. The proficient nurse
cal care setting is Benner’s Novice to Expert Theory.19 accepts the holism of the situation noting that the patient was
recently given a diuretic; she acquires additional data (blood
pressure, oxygen saturation) and follows the maxim that
The Novice to Expert Theory “fluid is the answer” before seeking a new medical order.
Benner’s Novice to Expert theory of nursing care is often cited Finally, the expert nurse is one who can step out of maxims,
in the critical care setting19 and describes five stages of profes- rules, and guidelines. Intuitively, the expert talks soothingly
sional development: (1) novice, (2) advanced beginner, (3) to the patient, touching gently, planning her approach to turn
competent, (4) proficient, and (5) expert. At first appearance the patient with assistance only after allowing family to visit
the NCCU is a chaotic environment filled with sights, sounds, and turning down the radio that inadvertently had been left
and even smells that rarely are encountered in daily life. In the playing.
midst of this chaos, nurses are focused on the task of patient
care. When nurses first come to the intensive care unit (ICU)
they enter at the novice level.19 When novices, nurses often
Care Models
may be unsuccessful in recognizing and interpreting key data. Nursing theory leads to the development of care models. An
The focus is on task completion, and novices are governed by information-inclusive model of nursing knowledge is pre-
rules (medical orders) such as “record B/P q1h, notify medical ferred to a model in which knowledge is limited by its point
doctor if SBP<180 mm Hg.” The novice has yet to gain the of origin. Jacobs-Kramer and Chinn22 modify Carper’s23
experience required to understand how the context of one model of nursing knowledge only to the extent of suggesting
situation varies from another situation. that the model become an active template to synthesize groups
Through a carefully orchestrated orientation most novices of knowledge from multiple disciplines. Nursing models that
are prepared to move to the stage of advanced beginner within embrace both empirical and nonempirical approaches to grow
1 year.19 Advanced beginners have developed the ability to and develop the knowledge base provide the greatest potential
quickly and accurately interpret much of the data, most of the to advance the contributions of nursing toward improving
time. The competent nurse is one who has been working in patient outcomes. The American Association of Critical-Care
the ICU for 3 to 5 years and has begun to link incoming data Nurses (AACN) embraces the synergy model for care of criti-
to long-term outcomes. This stage is embodied by the nurse cally ill patients.24 Additionally, many hospitals and universi-
who consciously plans a strategy to provide care but may lack ties have adopted various evidence-based nursing practice
the skills of more advanced professionals to quickly respond (EBNP) models.25
to change.
The nurse who can understand the holism of patient care
and quickly recognize how to acquire and interpret needed Evidence-Based Practice
data is considered proficient. Proficiency is not garnered by A sharp distinction exists between EBP and practice sup-
the number of years a nurse has worked or by his or her level ported by evidence. As the term implies, EBP is that which is
of education. Proficiency is demonstrated in actions that rooted in research.26 The implementation of EBP requires that
move the patient forward along a continuum of care. The the practitioner begin with a question and then evaluate all
proficient nurse relies on years of experience to provide care the available evidence that surrounds the clinical question;
guided by maxims with subtle nuances for patient-specific practice is thus driven from the results of this process.27 Prac-
situations. tice supported by evidence is a significantly less rigorous
Critical thinking skills have been directly linked to nurses process in which the practitioner has a practice pattern or
advancing from novice to expert.19,20 At the expert level nurses paradigm and then seeks evidence to support that paradigm.27
have accomplished performance aspects that have allowed Many health care systems have adopted the principles of EBP
them to progress to higher levels of professionalism. The and created more individualized models. The Iowa model is
expert nurse is one who has become an active participant in one such example that has been successfully adopted by dif-
the care of ICU patients, relies on experience as a foundation ferent health care systems.28,29
Section I—Background 39

Glasgow Coma Scale, National Institutes of Health [NIH]


Synergy Model stroke score, etc.) with varying degrees of success. In the
The synergy model focuses on the needs of the patients but NCCU such tools are commonplace. Incumbent on the cadre
describes a dyadic relationship between nurses’ competencies of seasoned nurses and physicians is the responsibility to
and patients’ characteristics.30 The model identifies eight explore and understand the interrater reliability of each tool
patient characteristics and eight dimensions of nursing prac- both across populations and within their individual units.
tice. Levels for the characteristics and dimensions are described During their primary training most nurses receive only a
in detail elsewhere.31 The nursing competencies are clinical few days of clinical experience in an ICU setting. Although
judgment, advocacy and moral agency, caring practices, col- most RNs likely will have been exposed to telemetry monitors,
laboration, systems thinking, response to diversity, facilitator noninvasive blood pressure monitors, and pulse oximetry, it
of learning, and clinical inquiry. The patient characteristics are is very likely that an RN will graduate without ever being
resiliency, vulnerability, stability, complexity, resource avail- inside an NCCU. Most hospitals provide a significant orienta-
ability, participation in care, participation in decision making, tion period during which an RN becomes familiar with the
and predictability.31 The health care environment is a vital care paradigms and equipment in their new unit. The RN who
element in the synergy model and can either promote or initiates and maintains noninvasive neuromonitoring most
hinder outcomes. For optimal synergy the environment needs likely will be exposed to that particular monitor for the first
to be one in which the nurse-patient dyad is supported. time during his or her unit-based orientation.
In the neurocritical care setting, nursing provides an
optimum contribution only through thoughtful collabora-
tions. Nursing is a vital component of any care delivery model, Invasive Monitoring
and it is primarily tasked with bedside decision making within Beginning with the moment a decision is made to initiate
the context of primarily physician-derived parameters.32 invasive monitoring, the nurse and physician should collabo-
Nurses benefit from an understanding of the history of the rate to provide optimum success. The major portion of inva-
medical model, and physicians benefit from an understanding sive monitoring falls within the category of physician-directed,
of the history and development of nursing theory and care nurse-driven care. Invasive monitoring takes many forms in
delivery models. the NCCU. Perhaps the most common example is intra-
arterial blood pressure monitoring. In this example the nurse
and physician must work together to initiate monitoring.
Monitoring Tools Although most state boards of nursing permit RNs to insert
A wide variety of monitoring tools exist and are discussed intra-arterial catheters, it is most common that either an
throughout this textbook. Because the purpose of this chapter advanced practice nurse (APN) or physician perform this
is to provide information about the role of the nurse in mul- activity. While the physician or APN inserts the catheter, the
timodal monitoring, noninvasive and invasive monitoring is RN must prepare the pressure line and tubing (Fig. 5.2) Often
only briefly discussed. More complete explanation of the the nurse becomes the eyes and ears for the medical team
many modes of monitoring, their benefits and limitations, are during bedside procedures to monitor the patient and provide
provided in other chapters. updates on the patient’s vital signs (Fig. 5.3).
Basic nursing education provides information about the A single monitoring device may be used for multimodal
use and interpretation of only a few of the monitoring tools monitoring. For example, an intra-arterial pressure line will
used in critical care. For critical care nurses, the term monitor provide a blood pressure reading, access to obtain blood to
is often used interchangeably in both the verb and noun sample biomarkers and blood gases, and the ability to provide
forms. Hence, a nurse may monitor the patient with a monitor! additional blood flow measurements such as cardiac output
Most nurses adhere to hospital-specific or unit-specific pro- and stroke volume. However, with a single monitor actions
tocols and policies. Within the United States these protocols or parameters can be mutually exclusive. For example, if
are developed based on each state’s board of nursing scope of
practice. Although most of the differences noted are small and
seemingly insignificant, when nurses change position or relo-
cate to a new NCCU, there is a need to review all of the policies
and procedures for that new position. Likewise, the physician
who relocates will benefit from understanding the nuances
associated with monitoring in any given unit.

Noninvasive Monitoring
There are many types of monitors described throughout this
book. Noninvasive monitors in general are those that do not
require that you break the skin or invade the body to obtain
data. Most nursing units allow that nurses with a registered
nurse (RN) degree or higher (see Table 5.1) may place nonin-
vasive monitors without a physician’s order.
The most basic and primary mode of noninvasive monitor-
ing has been and will remain the physical examination. Tools Fig. 5.2  Nurse monitoring vital signs while the physician performs a
have been developed to assist with this examination (e.g., procedure.
40 Section I—Background

100
HR ICP
80

60

40

20

0
3:00 3:15 3:30 3:45 4:00
3:00 PM 3:15 PM 3:30 PM 3:45 PM 4:00 PM
HR 64 66 68 69 74
ICP 12 13 11 13 12
Fig. 5.3  The nurse may become the eyes and ears for the medical team
during a bedside procedure. Fig. 5.4  Trends. The figure shows 1 hour of data. The graph (top)
indicates trends noted from heart rate (HR) and intracranial pressure (ICP)
sampled once per minute. The electronic medical record (EMR) data
(bottom) indicate values that were sampled and documented at
15-minute intervals.
an RN is directed to drain cerebrospinal fluid from an intra-
ventricular catheter, the ability to monitor intracranial pres-
sure (ICP) temporarily ceases. When this is the case, nurses
often are tasked to coordinate the timing during each compo- darker than normal urine, low volume of intravenous (IV)
nent of monitoring to calibrate monitors to maintain accu- fluid remaining in the primary infusion, soft rattle with each
racy, record data from monitors, interpret the meaning of the respiratory cycle, and the conspicuously close proximity of the
data, and if necessary take action on those findings.32 patient’s hand to his or her endotracheal tube. Nurses have
Nurses employed as RNs do not have prescriptive authority, indeed developed a unique skill set for multimodal monitor-
and it may be the case that the most appropriate course of ing. Nowhere is this more evident than in the NCCU. Just
action is to inform the physician or APN of their findings. as there are significant accomplishments of nursing, it should
When this is the case, the RN must decide if and when to leave be noted that since nurses are neither trained in, nor licensed
the bedside, a course of action that may leave the patient unat- as, diagnosticians, they are obligated to communicate their
tended and unmonitored. Systems designed to reduce the findings.37
period of time when patients are unattended are highly desir- Nurses have the luxury and obligation to make continual
able; delegating to technology is one such system.33 The sim- observations but have the tools to report only interval obser-
plest example of this is setting alarm parameters. More vations. When taking care of critically ill neuroscience patients,
complex methods are being developed and are discussed later the nurse often spends long periods of time in one room with
in this text (see Chapters 4, 40, 43, 45, 48). These technologic one patient. The nurse therefore may observe continual
developments and the ability to delegate the role of data acqui- changes in a patient’s vital signs or neurologic status. However,
sition to computers represent an evolution in the practice current documentation systems allow for the recording of
paradigm.34-36 Thus technology has allowed ICU nurses the only finite amounts of data at discrete intervals. Because the
ability to acquire more data points and to acquire data from continual observations are reported at intervals, there is a risk
more sources, but data are only useful if they are interpreted of ghosting or aliasing. Ghosting is the emergence of false
(correctly). frequencies and is related to sampling rate.38 In an example
showing 1 hour of data (Fig. 5.4) the graph at top is based on
HR and ICP sampled at 1-minute intervals and indicates that
Nursing and Monitoring the patient’s heart rate steadily decreases with periods of wide
In the NCCU, nurses monitor the monitors. Sandelowski11 fluctuation that appear to correspond to an increasing ICP. Yet
writes of the link between nursing and technology that “as the in this same example, the HR and ICP were sampled and
primary machine tenders in health care, nurses often acquire documented in the patient record at 15-minute intervals and
an understanding of how to apply, operate, and interpret the indicate an increasing HR and stable ICP.
products of devices that becomes an integral part of the tacit The purpose of the medical record (electronic or paper) is
know-how of clinical practice.” In this statement she identifies to provide an avenue of communication among health care
several vital components: (1) that the nurses are indeed the providers39—literally, to tell the patient’s story. Each event in
primary machine tenders, (2) that there is a knowledge basis the patient’s time in the ICU has meaning. “The primary role
required for nursing care, and (3) that nurses have acquired a of documentation should be to clearly describe and commu-
unique ability to interpret meaning from monitors. nicate what is going on with the patient.”39 Only by capturing
Nurses can, and have, walked into a patient’s room and and recording data from the many monitors can nurses
quickly gathered data on heart rate, systolic and diastolic provide physicians with the information they need. The
blood pressure, oxygen saturation, intracranial and cerebral duality of this obligation is noteworthy. Just as nurses are
perfusion pressure, ventilatory status, and temperature all obligated to collect and present a comprehensive and com-
while noting an increase in the patient’s level of agitation, plete dataset, physicians are obligated to ensure that they in
Section I—Background 41

turn evaluate each data element and if not, should not order
that data to be collected. Box 5.1  Considerations During Intrahospital
Transport
Monitoring During Transport  Pretransport communication
 Is this transport necessary?
The mantra of the nurse who monitors a patient during intra-
 Is the receiving area ready?
hospital transport is: “Plan for the worst—hope for the best—
 Transport personnel
be ready for anything.” Intrahospital transport, defined as any
 How many?
time when a patient is moved from the primary area of care
 Qualifications
to another location within the same facility, requires that the
 Do the transport personnel have coverage for other
patient receive the same level of care throughout the intrahos-
patients they may be caring for in the unit?
pital transport as would have been received had he or she
 Monitoring equipment
remained in the primary care area.40,41 The medical record
 The patient should receive the same minimum standard of
should indicate that the patient was monitored throughout
care during transport as was prescribed in the critical care
the transport and indicate the patient’s response during each
unit.
phase of transport. With most current systems of data acquisi-
 Battery life
tion, a loss of some patient data is inevitable, and in particular
 Physical location of the monitor during transport
this often may occur during patient transport.
 Time and distance
The very nature of critical care drives the need for addi-
 How long will the patient and nurse be involved in the
tional investigational and treatment options that exist only
intrahospital transport?
beyond the confines of the patient’s room. Nearly half of all
 What physical barriers exist between the critical care unit
patients admitted to critical care experience at least one intra-
and the transport destination?
hospital transport, and this most often occurs early during the
 Is the destination on a different floor?
critical care stay.42 It is well established that any decision to
 Is the destination readily accessible?
transport a patient must be made by balancing the potential
 Does the patient need to pass through high-access
risks and benefits (Box 5.1).41,43
areas?
For each episode of intrahospital transport there are three
 Resources
phases during which the patient is exposed to different risk
 Phase I and phase III
factors. Phase I begins with the decision that transport will
 Must be able to competently deliver the standard of
occur and ends when the patient reaches the intrahospital
care
destination. Phase II is a maintenance phase that begins when
 Emergency equipment
the patient arrives at a new destination and persists through-
 Airway and cardiac resuscitation equipment
out the patient’s stay at the new location. Phase III encom-
 Emergency medications
passes that period of time during which the patient is
 Phase II
transported back to the ICU and concludes only when the
 Must be equal to or greater than the standard of care
patient is stable or has returned to pretransport status.44 Spe-
in the critical care unit
cialization and education along with highly prepared staff are
 Documentation
associated with lower rates of adverse events during intrahos-
 Ability to acquire and record data
pital transport.45
 Ability to store and retrieve data

Phase I—From ICU to Destination


During phase I of the transport, most commonly it is the staff
nurse or a combination of staff nurse and respiratory therapist
who monitor the patient.45 The physician must clearly balance
the need to travel against the risk of problems during travel.
Phase II—Maintenance at the
Having made the decision that intrahospital transport is indi- Intrahospital Destination
cated, there is a joint responsibility to determine monitoring The degree of multimodal monitoring that takes place during
needs. The frequency of intrahospital transport may be a phase II of intrahospital transport often is determined by
factor in the purchase of new monitoring devices. Battery life, temporal parameters. Expert level and experienced nursing
portability, data storage and transfer, and even the weight of staff know to expect that a noncontrasted head computed
a monitor will factor into the decision of which monitors are tomography (CT) scan typically is performed in only a few
used during transport. minutes. The decision to re-level transducers, reconnect mon-
Loss of data during phase I of the transport may be associ- itors, or troubleshoot artifact is balanced against the risk of
ated with a combination of variables. Data may be lost if the keeping the patient flat for a prolonged period of time.47
signal quality is such that the data are uninterpretable; such is Several studies have begun to explore solutions to this conun-
the case with artifact because patient movement is a common drum.40,42,43 One such example is the development of a scoring
cause of artifact.46 Transport inevitably creates a variety of system to grade the level of acuity and risk associated with the
movement. Excessive artifact may result in data not being event.45 Higher-acuity patients require additional monitoring,
displayed or in displayed data that do not reflect the true resources, and staffing. Alternatively portable CT scanners in
patient condition. Few studies have explored data loss during the ICU can help reduce the need for some intrahospital
physical transport.40,41 transport.
42 Section I—Background

have the capacity to send and display some information from


Phase III—Returning to the ICU any one given patient to the room of another patient, few if
Phase III begins when the patient is transported back to the any monitors provide a full spectrum of readily available data
point of origin (the NCCU). The goals and concerns during to be displayed outside of a single patient room. A nurse who
this portion of phase III are the same as those during phase I. cares for two patients must collaborate with her peers to
On return to the NCCU the staff must simultaneously tend provide continual monitoring. This action may be a simple
the monitoring systems while assessing the patient. The nurse request to listen for alarms or as complicated as the shift hand­
should perform a comprehensive patient assessment that over. If one nurse needs help she asks other nurses to come to
includes ensuring that all access lines, drains, and life-support the bedside—they collaborate when one nurse makes a new
devices are secure, intact, and properly functioning. In addi- observation.
tion, the nurse should evaluate the effects of any medications
or interventions performed during the intrahospital transport
and should report any patient condition changes. Nurse and Midlevel Practitioner
The nurse must ensure that all monitoring devices func- Technology has provided increased levels of information to
tion properly. Devices that were taken with the patient during obtain and monitor critically ill patients. To meet this need,
transport may need to be recalibrated or re-leveled. Alarm the number of eyes and ears needed to run an ICU also has
parameters should be reviewed to ensure that any parameters increased. Many NCCUs now employ a variety of midlevel
that were reset during transport are now set to the desired practitioners (see Table 5.1). The APNs or PAs typically have
level, and any device that requires electricity again is plugged a minimum of a master’s degree. During the past decade mid-
into an outlet. Many NCCUs now use an EMR. Monitors level practitioners have begun to take more active roles in the
may need to be synchronized to central hubs such that daily functions of how an ICU runs. Because most midlevel
there is a transfer of data to the EMR. The final phase of an practitioners have at least a limited prescriptive authority, they
intrahospital transport is marked by a combination of two may be the first person the staff nurse (RN) approaches when
conditions: (1) the patient has been returned to the neuro- seeking a new order or seeking to clarify an existing order.
critical care unit, and (2) the patient has returned to a stable
condition.44
Nurse and Physician
Without active collaboration between the nurse who provides
Collaboration direct care and the physician who directs care, the system can
Collaboration is a key element in monitoring the patient with fall apart. Physicians must understand not only their own
a neurologic injury. Nurses who take care of patients must respective roles, but also must have an understanding of the
collaborate on a variety of levels. The nurse-to-nurse collabo- role, abilities, and limitations of their counterpart (the nurse).
ration takes place both within and between nursing shifts. Active collaboration on the part of the nurse means providing
Nurses collaborate with midlevel practitioners (APNs and the physician with the products of monitoring. This exchange
physician assistants [PAs]) as well as with medical residents may be in a more formal or ritualized pattern; for example,
and attending physicians to modify the patient’s plan of care. during morning rounds the nurse may report the patient’s
Nurses also may collaborate with ancillary department per- condition to the ICU team (Fig. 5.5). Other collaborations
sonnel (e.g., pharmacy, respiratory care, physical therapy) to may occur in less formal exchanges, when the nurse calls or
coordinate the timing of care delivery. Finally, the nurse will speaks with the physician or physician designee. Whether in
collaborate with the patient and family to determine the goals oral or written form, the physician requires data from which
of care and provide education. to make vital decisions. Equally, the physician must work
with the nursing staff to develop and continually refine the
plan of care.
Nurse-to-Nurse Collaboration
The nurse-to-nurse collaboration takes place on two fronts
and in particular during the nursing handover. The nursing
handover (also called shift report, or handoff) occurs when one
nurse ends her or his shift and hands over treatment of the
patient to the next nurse. During this time, the two nurses
communicate about the events and observations made during
the previous shift.48 Handover typically lasts from 15 to 30
minutes but currently is not a standardized process.49 The
oncoming nurse and the care nurse whose shift is ending work
jointly to review the patient’s medical history and events of
the day and develop a plan of care for the next shift.48,49
The second form of nurse-to-nurse collaboration occurs
during and throughout the nursing shift. Throughout a
nursing shift, the expert and proficient nurse provide informal
education to the novice, advanced beginner, and competent
nurses.19 It is not possible for any one nurse to be in more than
one place at one time, and often nurses are assigned to provide Fig. 5.5  The neurocritical care team discussing patient care during
care to multiple patients. Although most bedside monitors rounds.
Section I—Background 43

benefit of low production cost and ease of attendance. A hos-


Continuous Education pital employee (e.g., nurse educator, APN, experienced nurse,
Neurocritical care is a relatively new field. New treatment or hospital administrator), a physician faculty member or
options or strategies, medications, and interventions continu- invited guest, or an industry representative may present the
ally are being developed. New methods and tools available to inservice. The obvious advantages of inservicing should be
monitor patients also are being developed and introduced balanced against the risk of bias. It is possible that a presenta-
at an amazing pace. To maintain competence and provide tion could provide material that is out-of-date or be designed
the highest level of care, nurses (and physicians) must engage to provide only a single view of a topic. Repeated inservicing
in a program that encourages and provides continuing by a limited number of individuals may result in the same
education. material being delivered again and again and so prevent the
influx of new ideas.
Nursing Education
A nurse officially becomes a nurse upon graduation from an Patient and Family Education
accredited school of nursing. However, only after passing the Another aspect of education about new monitoring is the
licensure examination is a nurse registered with the state need to educate patients and families. The concept of open-
board of nursing and conferred the status of RN. RN status visiting, although not universally embraced, has become wide-
indicates that a nurse has met the minimum licensing stan- spread. Gone are the days when family members visited
dards and may begin professional practice. During nursing patients twice each day in 10-minute intervals. Today loved
school, few nurses have adequate exposure to the NCCU to ones are omnipresent and considered part of the care team
be considered competent enough to provide care indepen- (see Chapter 3).52 As members of the care team, patients and
dently. For this reason, the orientation period (discussed family members must be educated to understand the moni-
earlier) often is the first post-degree education that RNs tors. No two patients and no two families are alike, and edu-
receive. cational needs must be tailored to the individual. Promoting
Nationally, several programs have been developed that are patient and family education requires is a collaborative effort
geared to provide specific aspects of neuroscience knowledge between physicians and nurses.53
and critical care knowledge to the novice and advanced begin-
ner nurse. The Foundations of Neuroscience Nursing (FNN)
program was developed by the American Association of Neu-
Conclusion
roscience Nurses.50 The Essentials of Critical Care Orientation As a distinct discipline, neurocritical care is continually
(ECCO) was created by the AACN orientation program.51 expanding. New procedures and treatment options have pro-
The FNN program is an electronically distributed educa- vided patients with neurologic injury and illness the oppor-
tional package. The course is composed of 21 modules that tunity to lead full, productive lives long after their hospital
range between 30 and 150 minutes in length and covers a discharge. A vast array of monitoring options already exist
broad spectrum of topics related to the care of patients with that can be used to observe for changes in patient condi-
neurologic and neurosurgical injuries. It is offered to indi- tions, and newer devices are in development. To be optimally
viduals who wish to increase their knowledge and to institu- effective, these current and future monitoring devices will
tions that wish to provide the content to one or more require highly educated staff that specialize in the care and
employees. An option to negotiate a multisite contract also treatment of neurocritically ill patients. The role of multi-
exists. The target audience for the FNN course is nurses with modal monitoring in neurocritical care is expanding; the
less than 2 years of experience who provide care to neurosci- role of the nurse in critical care is equally expanding to meet
ence patients. The program is designed to be facilitated by an this need.
instructor (experienced nurse or nurse educator) from the
institution that purchased the product.50
The most recent version of the ECCO program is a series References
of more than 40 lessons in 10 modules. ECCO is specifically 1. Santayana G. The life of reason. New York: C. Scribner’s Sons; 1922.
targeted as an orientation package to nurses who enter a criti- 2. Olson DM. Multimodal neurological monitoring. In: Kaplow R, Hardin SR,
cal care unit and covers each of the major human biologic editors. Critical care nursing: synergy for optimal outcomes. Sudbury, Mass:
systems and the most common diseases found in the critical Jones & Bartlett; 2007. p. 359–74.
care setting. Completion of this Internet-based course pro- 3. Davidson GR. Medicine through the ages. London, New York: Methuen; Roy
Publishers; 1968.
vides the RN with 69 contact hours.51 4. Wreszinski W. Königliche Museen zu Berlin. Der grosse medizinische
In most states the board of nursing requires that nurses Papyrus des Berliner Museums (Pap. Berl. 3038) in Facsimile und Umschrift
demonstrate that they have maintained their nursing compe- mit Übersetzung, Kommentar und Glossar. Leipzig: J.C. Hinrichs; 1909.
tency by obtaining continuing education. Nurses obtain and 5. Mackintosh C. A historical study of men in nursing. J Adv Nurs 1997;26(2):
232–6.
maintain contact hours through a variety of venues. Local, 6. Nightingale F. Florence Nightingale’s notes on nursing. Skretkowicz V, editor.
regional, and national conferences offer a chance for nurses to Reviewed with additions ed. London: Scutari Press; 1992.
obtain continuing education units (CEUs). Although these 7. Shuttleworth A, O’Dowd A. 100 years of nursing. Nurs Times 2005;101(19):
annual events are popular, time, travel, and money limit the 17–28, 30–18, 40–18.
ability of many to attend. Hospital-based inservices often 8. Barrett EA. What is nursing science? Nurs Sci Q 2002;15(1):51–60.
9. Parse RR. Nursing: the discipline and the profession. Nurs Sci Q 1999;12(4):
provide a unique opportunity for low-cost education. 275–6.
Hospital- and unit-based inservices are a nearly universal 10. Walker LO, Avant KC. Strategies for theory construction in nursing. 3rd ed.
experience in the nursing lexicon. Nursing inservices offer the Norwalk, Conn: Appleton & Lange; 1995.
44 Section I—Background

11. Sandelowski M. Knowing and forgetting: the challenge of technology for a 33. Crocker C, Timmons S. The role of technology in critical care nursing. J Adv
reflexive practice science of nursing. In: Thorne SE, Hayes VE, editors. Nurs 2009;65(1):52–61.
Nursing praxis knowledge and action. Thousand Oaks, Calif: Sage 34. Cuthbertson BH, Boroujerdi M, Prescott G. The use of combined
Publications; 1997. p. 69–85. physiological parameters in the early recognition of the deteriorating acute
12. Donaldson SK, Crowley DM. The discipline of nursing. Nurs Outlook 1978; medical patient. J R Coll Physicians Edinb 2010;40(1):19–25.
26(2):113–20. 35. Stylianides N, Dikaiakos MD, Gjermundrod H, et al. Intensive care window:
13. Verran JA. The value of theory-driven (rather than problem-driven) real-time monitoring and analysis in the intensive care environment. IEEE
research. Semin Nurse Manag 1997;5(4):169–72. Trans Inf Technol Biomed 2011;15(1):26–32.
14. Chinn PL, Jacobs MK. Theory and nursing: a systematic approach. St. Louis: 36. Burgess LP, Herdman TH, Berg BW, et al. Alarm limit settings for early
CV Mosby; 1983. warning systems to identify at-risk patients. J Adv Nurs 2009;65(9):1844–52.
15. Sidani S, Braden CJ. Evaluating nursing interventions: a theory-driven 37. Lacey SR, Cox KS. Nursing: key to quality improvement. Pediatr Clin North
approach. Thousand Oaks, CA: Sage Publications; 1998. Am 2009;56(4):975–85.
16. Parse RR. Paradigms: a reprise. Nurs Sci Q 2000;13(4):275–6. 38. Webster JG, Clark JW. Medical instrumentation: application and design. 4th
17. Watson J, Smith MC. Caring science and the science of unitary human ed. Hoboken, NJ: John Wiley & Sons; 2010.
beings: a trans-theoretical discourse for nursing knowledge development. 39. Schiff GD, Bates DW. Can electronic clinical documentation help prevent
J Adv Nurs 2002;37(5):452–61. diagnostic errors? N Engl J Med 2010;362(12):1066–9.
18. Beeby JP. Intensive care nurses’ experiences of caring. Part 1: consideration 40. Day D. Keeping patients safe during intrahospital transport. Crit Care Nurse
of the concept of caring. Intensive Crit Care Nurs 2000;16(2):76–83. 2010;30(4):18–32; quiz 33.
19. Benner P. From novice to expert: excellence and power in clinical nursing 41. Warren J, Fromm Jr RE, Orr RA, et al. Guidelines for the inter- and
practice. Menlo Park, CA: Addison-Wesley, Nursing Division; 1984. intrahospital transport of critically ill patients. Crit Care Med 2004;32(1):
20. McKane CL, Schumacher L. Professional advancement model for critical care 256–62.
orientation. J Nurs Staff Dev 1997;13(2):88–91. 42. Voigt LP, Pastores SM, Raoof ND, et al. Review of a large clinical series:
21. Rashotte J, Thomas M. Incorporating educational theory into critical care intrahospital transport of critically ill patients: outcomes, timing, and
orientation. J Contin Educ Nurs 2002;33(3):131–7. patterns. J Intensive Care Med 2009;24(2):108–15.
22. Jacobs-Kramer MK, Chinn PL. Perspectives on knowing: a model of nursing 43. Jarden RJ, Quirke S. Improving safety and documentation in intrahospital
knowledge. In: Nicoll LH, editor. Perspectives on nursing theory. 3rd ed. transport: development of an intrahospital transport tool for critically ill
Philadelphia: Lippincott; 1997. p. 323–30. patients. Intensive Crit Care Nurs 2010;26(2):101–7.
23. Carper B. Fundamental patterns of knowing in nursing. ANS Adv Nurs Sci 44. Noa Hernandez JE, Gonzalez EC, Romero JM, et al. [Intrahospital
1978;1(1):13–23. transportation of the seriously ill patient. The need for an action guideline.].
24. Hardin SR, Kaplow R. Synergy for clinical excellence: the AACN synergy Enferm Intensiva 2011;22(2):74–7.
model for patient care. Sudbury, MA: Jones & Bartlett; 2005. 45. Kue R, Brown P, Ness C, et al. Adverse clinical events during intrahospital
25. Keele R. Nursing research and evidence-based practice: ten steps to success. transport by a specialized team: a preliminary report. Am J Crit Care 2011;
Sudbury, MA: Jones & Bartlett Learning; 2011. 20(2):153–61; quiz 162.
26. Youngblut JM, Brooten D. Evidence-based nursing practice: why is it 46. Jevon P. An introduction to electrocardiogram monitoring. Nurs Crit Care
important? AACN Clin Issues 2001;12(4):468–76. 2010;15(1):34–8.
27. Stillwell SB, Fineout-Overholt E, Melnyk BM, et al. Evidence-based practice, 47. Bekar A, Ipekoglu Z, Tureyen K, et al. Secondary insults during intrahospital
step by step: asking the clinical question: a key step in evidence-based transport of neurosurgical intensive care patients. Neurosurg Rev 1998;
practice. Am J Nurs 2010;110(3):58–61. 21(2-3):98–101.
28. Titler MG, Kleiber C, Steelman VJ, et al. The Iowa model of evidence-based 48. Chaboyer W, McMurray A, Johnson J, et al. Bedside handover: quality
practice to promote quality care. Crit Care Nurs Clin North Am 2001;13(4): improvement strategy to “transform care at the bedside.” J Nurs Care Qual
497–509. 2009;24(2):136–42.
29. Stetler CB, Ritchie JA, Rycroft-Malone J, et al. Institutionalizing evidence- 49. Manser T, Foster S. Effective handover communication: an overview of
based practice: an organizational case study using a model of strategic research and improvement efforts. Best Pract Res Clin Anaesthesiol 2011;
change. Implement Sci 2009;4:78. 25(2):181–91.
30. Curley MA. Patient-nurse synergy: optimizing patients’ outcomes. Am J Crit 50. Foundations of Neuroscience Nursing [computer program]: Helmick K,
Care 1998;7(1):64–72. Fitzsimmons B, Forsyth L, editors. Glenview, Ill: American Association of
31. Kaplow R, Hardin SR. Critical care nursing: synergy for optimal outcomes. Neuroscience Nurses; 2006.
Sudbury, MA: Jones & Bartlett; 2007.
32. Olson DM, Graffagnino C. Consciousness, coma, and caring for the A complete list of references for this chapter can be found online at
brain-injured patient. AACN Clin Issues 2005;16(4):441–55. www.expertconsult.com.
Section I—Background 44.e1

References 30. Curley MA. Patient-nurse synergy: optimizing patients’ outcomes. Am J Crit
Care 1998;7(1):64–72.
1. Santayana G. The life of reason. New York: C. Scribner’s Sons; 1922. 31. Kaplow R, Hardin SR. Critical care nursing: synergy for optimal outcomes.
2. Olson DM. Multimodal neurological monitoring. In: Kaplow R, Hardin SR, Sudbury, MA: Jones & Bartlett; 2007.
editors. Critical care nursing: synergy for optimal outcomes. Sudbury, Mass: 32. Olson DM, Graffagnino C. Consciousness, coma, and caring for the
Jones & Bartlett; 2007. p. 359–74. brain-injured patient. AACN Clin Issues 2005;16(4):441–55.
3. Davidson GR. Medicine through the ages. London, New York: Methuen; Roy 33. Crocker C, Timmons S. The role of technology in critical care nursing. J Adv
Publishers; 1968. Nurs 2009;65(1):52–61.
4. Wreszinski W. Königliche Museen zu Berlin. Der grosse medizinische 34. Cuthbertson BH, Boroujerdi M, Prescott G. The use of combined
Papyrus des Berliner Museums (Pap. Berl. 3038) in Facsimile und Umschrift physiological parameters in the early recognition of the deteriorating acute
mit Übersetzung, Kommentar und Glossar. Leipzig: J.C. Hinrichs; 1909. medical patient. J R Coll Physicians Edinb 2010;40(1):19–25.
5. Mackintosh C. A historical study of men in nursing. J Adv Nurs 1997;26(2): 35. Stylianides N, Dikaiakos MD, Gjermundrod H, et al. Intensive care window:
232–6. real-time monitoring and analysis in the intensive care environment. IEEE
6. Nightingale F. Florence Nightingale’s notes on nursing. Skretkowicz V, editor. Trans Inf Technol Biomed. 2011;15(1):26–32.
Reviewed with additions ed. London: Scutari Press; 1992. 36. Burgess LP, Herdman TH, Berg BW, et al. Alarm limit settings for early
7. Shuttleworth A, O’Dowd A. 100 years of nursing. Nurs Times 2005;101(19): warning systems to identify at-risk patients. J Adv Nurs 2009;65(9):1844–52.
17–28, 30–18, 40–18. 37. Lacey SR, Cox KS. Nursing: key to quality improvement. Pediatr Clin North
8. Barrett EA. What is nursing science? Nurs Sci Q 2002;15(1):51–60. Am 2009;56(4):975–85.
9. Parse RR. Nursing: the discipline and the profession. Nurs Sci Q 1999;12(4): 38. Webster JG, Clark JW. Medical instrumentation: application and design. 4th
275–6. ed. Hoboken, NJ: John Wiley & Sons; 2010.
10. Walker LO, Avant KC. Strategies for theory construction in nursing. 3rd ed. 39. Schiff GD, Bates DW. Can electronic clinical documentation help prevent
Norwalk, Conn: Appleton & Lange; 1995. diagnostic errors? N Engl J Med 2010;362(12):1066–9.
11. Sandelowski M. Knowing and forgetting: the challenge of technology for a 40. Day D. Keeping patients safe during intrahospital transport. Crit Care Nurse
reflexive practice science of nursing. In: Thorne SE, Hayes VE, editors. 2010;30(4):18–32; quiz 33.
Nursing praxis knowledge and action. Thousand Oaks, CA: Sage 41. Warren J, Fromm Jr RE, Orr RA, et al. Guidelines for the inter- and
Publications; 1997. p. 69–85. intrahospital transport of critically ill patients. Crit Care Med 2004;32(1):
12. Donaldson SK, Crowley DM. The discipline of nursing. Nurs Outlook 1978; 256–62.
26(2):113–20. 42. Voigt LP, Pastores SM, Raoof ND, et al. Review of a large clinical series:
13. Verran JA. The value of theory-driven (rather than problem-driven) intrahospital transport of critically ill patients: outcomes, timing, and
research. Semin Nurse Manag 1997;5(4):169–72. patterns. J Intensive Care Med 2009;24(2):108–15.
14. Chinn PL, Jacobs MK. Theory and nursing: a systematic approach. St. Louis: 43. Jarden RJ, Quirke S. Improving safety and documentation in intrahospital
C.V. Mosby; 1983. transport: development of an intrahospital transport tool for critically ill
15. Sidani S, Braden CJ. Evaluating nursing interventions: a theory-driven patients. Intensive Crit Care Nurs 2010;26(2):101–7.
approach. Thousand Oaks, CA: Sage Publications; 1998. 44. Noa Hernandez JE, Gonzalez EC, Romero JM, et al. [Intrahospital
16. Parse RR. Paradigms: a reprise. Nurs Sci Q 2000;13(4):275–6. transportation of the seriously ill patient. The need for an action guideline.].
17. Watson J, Smith MC. Caring science and the science of unitary human Enferm Intensiva 2011;22(2):74–7.
beings: a trans-theoretical discourse for nursing knowledge development. 45. Kue R, Brown P, Ness C, et al. Adverse clinical events during intrahospital
J Adv Nurs 2002;37(5):452–61. transport by a specialized team: a preliminary report. Am J Crit Care 2011;
18. Beeby JP. Intensive care nurses’ experiences of caring. Part 1: consideration 20(2):153–61; quiz 162.
of the concept of caring. Intensive Crit Care Nurs 2000;16(2):76–83. 46. Jevon P. An introduction to electrocardiogram monitoring. Nurs Crit Care
19. Benner P. From novice to expert: excellence and power in clinical nursing 2010;15(1):34–8.
practice. Menlo Park, CA: Addison-Wesley, Nursing Division; 1984. 47. Bekar A, Ipekoglu Z, Tureyen K, et al. Secondary insults during intrahospital
20. McKane CL, Schumacher L. Professional advancement model for critical care transport of neurosurgical intensive care patients. Neurosurg Rev
orientation. J Nurs Staff Dev 1997;13(2):88–91. 1998;21(2-3):98–101.
21. Rashotte J, Thomas M. Incorporating educational theory into critical care 48. Chaboyer W, McMurray A, Johnson J, et al. Bedside handover: quality
orientation. J Contin Educ Nurs 2002;33(3):131–7. improvement strategy to “transform care at the bedside.” J Nurs Care Qual
22. Jacobs-Kramer MK, Chinn PL. Perspectives on knowing: a model of nursing 2009;24(2):136–42.
knowledge. In: Nicoll LH, editor. Perspectives on nursing theory. 3rd ed. 49. Manser T, Foster S. Effective handover communication: an overview of
Philadelphia: Lippincott; 1997. p. 323–30. research and improvement efforts. Best Pract Res Clin Anaesthesiol 2011;
23. Carper B. Fundamental patterns of knowing in nursing. ANS Adv Nurs Sci 25(2):181–91.
1978;1(1):13–23. 50. Foundations of Neuroscience Nursing [computer program]: Helmick K,
24. Hardin SR, Kaplow R. Synergy for clinical excellence: the AACN synergy Fitzsimmons B, Forsyth L, editors. Glenview, Ill: American Association of
model for patient care. Sudbury, MA: Jones & Bartlett; 2005. Neuroscience Nurses; 2006.
25. Keele R. Nursing research and evidence-based practice: ten steps to success. 51. Kaddoura MA. Effect of the essentials of critical care orientation (ECCO)
Sudbury, MA: Jones & Bartlett Learning; 2011. program on the development of nurses’ critical thinking skills. J Contin
26. Youngblut JM, Brooten D. Evidence-based nursing practice: why is it Educ Nurs 2010;41(9):424–32.
important? AACN Clin Issues 2001;12(4):468–76. 52. Livesay S, Gilliam A, Mokracek M, et al. Nurses’ perceptions of open visiting
27. Stillwell SB, Fineout-Overholt E, Melnyk BM, et al. Evidence-based practice, hours in neuroscience intensive care unit. J Nurs Care Qual 2005;20(2):
step by step: asking the clinical question: a key step in evidence-based 182–9.
practice. Am J Nurs 2010;110(3):58–61. 53. Farahani MA, Sahragard R, Carroll JK, et al. Communication barriers to
28. Titler MG, Kleiber C, Steelman VJ, et al. The Iowa model of evidence-based patient education in cardiac inpatient care: a qualitative study of multiple
practice to promote quality care. Crit Care Nurs Clin North Am 2001;13(4): perspectives. Int J Nurs Pract 2011;17(3):322–8.
497–509.
29. Stetler CB, Ritchie JA, Rycroft-Malone J, et al. Institutionalizing evidence-
based practice: an organizational case study using a model of strategic
change. Implement Sci 2009;4:78.
Chapter
6  
I

Quality Assessment in the


Neurocritical Care Unit
Anoma Nellore, Peter D. le Roux, and David A. Horowitz

increase the likelihood of desired health outcomes and are


Introduction consistent with current professional knowledge.” Patient safety
Intensive care is one of the most expensive forms of care is integral to quality because it is difficult to provide successful
delivered by a hospital system. In 1993, intensive care repre- care when safety is compromised; that is, it is difficult to
sented 25% to 30% of acute hospital costs and constituted 1% examine quality and safety in isolation. The IOM defines
of the gross national product (GNP).1 It has become increas- safety as “the absence of clinical error, either by commission
ingly important in an era of public and private accountability [unintentionally doing the wrong thing] or omission [unin-
to measure and benchmark quality of intensive care unit tentionally not doing the right thing].” This is similar to the
(ICU) care; however, this is a complicated task. Quality World Health Organization (WHO) definition.
measures of health care are divided into three broad areas: The concept of quality in health care is central to clinical
(1) process (appropriate delivery of health care), (2) outcome practice and is as old as medicine. Indeed the phrase primum
(measured endpoints of care), and (3) structure (adequate non nocere (first do no harm) dates back to Hippocrates and is
resources to provide health care).2 Process measures include one of the principal tenets of medical ethics. In the early 20th
appropriate uses of stress ulcer and deep vein thrombosis century, Codman advocated public reporting of outcomes by
(DVT) prophylaxis, appropriate transfusion thresholds, daily both physicians and hospitals.3 Modern quality initiatives
interruption of sedation, appropriate glycemic control, and (QIs) started in other industries that realized that unexplained
use of aspirin and beta-blockers in acute myocardial infarc- variation leads to poor quality, and processes that decrease
tions among other benchmarks. Outcome measures include variation coupled with continuous evaluation can improve
such parameters as mortality, incidence of catheter-related quality and therefore profit. For example, in the 1950s Japa-
bloodstream infections, or rates of ventilator-associated pneu- nese car manufacturers applied Deming’s theory that improv-
monia (VAP). Typical structural quality indicators include the ing quality will reduce expenses while increasing productivity
presence of an ICU medical director, daily rounds by an inten- and market share; this led to an international demand for
sivist, appropriate ICU nurse-to-patient ratio, and multidisci- Japanese automobiles.4 For example, Toyota became a model
plinary rounds involving a pharmacist. of operational excellence, in part through kaizen, a Japanese
Use of each type of quality metric has its advantages and philosophy of continuous improvement. However, in 2009,
disadvantages. For example, although structural quality indi- there was a recall of more than 8 million vehicles and a decline
cators are easy to define and measure, they may not translate in stock value thought by some to be a subtle abandonment
to immediate improvement in care. Process quality indicators of these principles. In the 1980s Donabedian introduced the
may require tailoring to specific disease processes and may structure, process, and outcome paradigm to assess quality in
become cumbersome. Although the public (and health care health care but also cautioned that before assessment can
providers) are most interested in outcome metrics, data that begin it must be determined how quality is to be defined,
pertain to outcome measures may take a long time to obtain something that is still ongoing.5 During the past decade there
and there is little definition of appropriate standards. In addi- has been increased focus on quality and safety of medical care
tion, outcome measures often require risk factor stratification with the recognition of the high cost of health care, potential
for valid comparisons and although much work has been done for harm, and a perceived need for transparency about hospi-
in general critical care, there has been less research on bench- tals to the public.2,6-8 The report “To Err Is Human” by the
marking specific to neurocritical care. IOM in the United States in 1999 led to an increased focus on
safety and quality of care. In 2009 the European Society of
Intensive Care Medicine (ESICM) initiated a task force to
History of and Impetus for improve the safety and quality of care provided to ICU
patients, leading to a publication of prospectively defined
Qualitative Initiatives indicators to improve quality of critical care.9 QIs are increas-
The Institute of Medicine (IOM) defines quality as “the degree ingly a focus of hospitals because pay-for-performance
to which health services for individuals and populations (P4P) programs are under development by organizations that
© Copyright 2013 Elsevier Inc. All rights reserved. 45
46 Section I—Background

purchase health care such as the Leapfrog Group and the There was fair to moderate agreement in identifying ICU
Centers for Medicare & Medicaid Services (CMS). To measure quality outliers among the three scoring systems. However, the
quality a combination of process and outcome indicators may majority of ICUs in the study were classified as above average
be best,10 but measuring ICU performance can be a complex and so the authors questioned the utility of SMRs if high-
and time-consuming task11 because it depends on multiple performance ICUs cannot be reliably highlighted as role
domains such as patient- and family-centered outcomes, the model institutions and questioned the ability of the various
work environment, personnel satisfaction, and economic per- models to be used for benchmarking. More recent study from
formance among others. Europe using validation sets each of more than 50,000 patients
and from multiple ICUs suggest that the original APACHE IV
shows good discrimination and accuracy but poor calibra-
Process and Outcome tion,15 whereas a risk profile management using the new SAPS
II can be used to evaluate individual ICUs according to the
Quality Indicators specific risk for patients to die compared with a reference
Donabedian proposed that structure, process, and outcome be sample over the whole spectrum of hospital mortality.16
reviewed to improve quality of health care.5 Processes of care There are other limitations to the hospitalized SMR because
usually are the easiest to target initially, modify them if neces- it was developed for all patients admitted to a hospital with a
sary, and then measure outcomes associated with the process. primary diagnosis that is associated with 80% of all in-hospital
Outcomes such as mortality are easy to measure. However, mortality based on national outcomes.17 SMR, therefore, may
many variables and processes influence mortality, and so better reflect hospital rather than ICU mortality. Consistent
attributing variation in mortality to a single intervention or with this Brinkman et al., in a cohort study that included
process change is not always feasible. There thus can be some 66,564 ICU patients from 55 Dutch ICUs, found that a model
overlap between outcome and process in QIs. For example, based on clinical data (SAPS II) outperformed SMR.18 Hence
process can include use of available technology and staff edu- when comparing ICU performance it may be better to use a
cation and training, whereas outcome may include resource clinical (ICU) rather than an administrative or hospital model,
use and appropriate use of diagnostic or therapeutic proce- because there is great variation across ICUs in disease severity
dures. The following text reviews process and outcome indica- and hospitals (or ICUs) that admit more severely ill patients
tors together. are disadvantaged when compared with institutions that
admit less severely ill patients. Instead the SMR could be used
to indicate when performance may be poor rather than as a
Risk Adjustment quality indicator. Furthermore, although short-term mortality
Quality and benchmarking initiatives require accurate strati- may be sufficient to differentiate outcomes in general ICUs, it
fied risk adjustment. The main prognostic models used to is not an adequate measure to reflect recovery after many
assess overall illness severity of adults admitted to the ICU neurologic conditions that require ICU admission or to assess
are the Acute Physiology and Chronic Health Evaluation quality in a neurocritical care unit (NCCU) because long-term
(APACHE), Simplified Acute Physiology Score (SAPS), and functional recovery is more important in these patients.
Mortality Probability Model (MPM). The SAPs and MPM
have been updated to their third versions and APACHE to its
fourth version (see also Chapter 10).
Process Quality Indicators
Process indicators usually refer to how something is done
(or not). These indicators describe the interactions among
Mortality—Outcome patients, their families, and health care providers and the
Short-term (e.g., 30 days) mortality often is used to measure complex tasks performed by the ICU staff to achieve a speci-
success of management strategies in general ICU care. fied outcome. There often may be a gap between process
However, mortality as an outcome quality indicator has limi- quality indicator establishment and use in ICU care. For
tations. Crude mortality cannot be used to evaluate ICU care example, Pronovost et al. developed and tested ICU perfor-
because it does not adjust for underlying patient mix or mance measures in a study that involved 13 ICUs, including 3
disease state. The standardized mortality ratio (SMR) com- medical, 3 surgical, and 7 combined ICUs at nonteaching hos-
pares the observed to the expected mortality rate. The expected pitals.8 Three types of performance monitors—infection
mortality rate may be derived from a number of physiologic control forms, daily rounding forms, and team leader forms—
parameters including APACHE scoring, SAPS, and the MPM. were used to gather data. There was significant variation in
These models, however, all have limitations in external valida- compliance with quality measures among ICUs and within
tion series when applied to data sets outside their original individual ICUs. For example, elevated head-of-bed position
population.12-14 For example, Sirio et al. observed consistent to prevent VAP was observed in 67% (range 42%-99%) of the
overestimation of the SMR (actual/predicted mortality) devel- ICUs. Other quality measures—appropriate blood transfusion
oped from a national sample that was applied to a community- goals (9%-66%) appropriate peptic ulcer (71%-98%), or DVT
based sample of hospital ICUs in a metropolitan area.6 The prophylaxis (48%-98%)—also had a wide range. Based on
data further suggested that the methods used to describe stan- compliance with the process quality indicators the authors
dard outcome measures may require recalibration when suggest that 1604 excess ICU and 198 excess hospital days
applied to new samples and to reflect practice changes over per year were incurred. Detailed data derived from patients
time. Glance et al. compared the SMRs based on three differ- contained in the eICU Research Institute (eRI) data repository
ent scoring systems (APACHE II, SAPS II, and MPM II) to see who were discharged from the hospital during 200819 further
if each system could identify the same low-performance ICUs.7 describe adherence with critical care best practice including
Section I—Background 47

stress ulcer (90.8%) and venous thrombosis prophylaxis of ICU “quality” is debated for several reasons. First, VAP can
(86.8%), beta-blocker use (68%-79%), low tidal volume ven- be difficult to diagnose because of underlying cardiopulmo-
tilation (28% for <6 mL/kg), and glycemic control (56% in nary disorders (e.g., pulmonary contusion, acute respiratory
110-180 mg/dL) in current clinical practice in 271 ICUs in 188 distress syndrome, atelectasis) and the nonspecific radio-
institutions across diverse regions of the United States. The graphic and clinical signs associated with VAP. One third of
following text examines individual process QIs and suggests patients diagnosed with VAP have no evidence of VAP at
strategies toward their implementation.8 autopsy and one fourth of mechanically ventilated patients
who are not diagnosed with VAP actually have a VAP at
autopsy. Second, clinical criteria described by the American
End-of-Life Care Thoracic Society (ATS) lack specificity because other condi-
Quality end-of-life care, also sometimes referred to as pallia- tions may mimic VAP. For example, purulent secretions often
tive care, is an important aspect of ICU treatment because one are present in intubated patients because of poor mucociliary
in five U.S. deaths occurs in the ICU. Many of these deaths are clearance. Furthermore colonization of oral secretions can
in patients in whom treatment goals are changed to comfort confuse respiratory culture data, and surveillance for VAP
measures. Interviews with family members after a patient dies requires dedicated and skilled staff.27,28 Third, reporting of
using a 31-item Quality of Dying and Death (QODD) ques- VAP is variable. For example, Eggimann et al. examined dif-
tionnaire demonstrate that higher scores (i.e., quality) are ferent denominators (1000 patient-days, patient-days at risk,
associated with death at home or in a location of the patient’s ventilator-days, and ventilator-days at risk) used to report VAP
choice, lower symptom burden, satisfaction with symptom and found that the method of reporting VAP could underes-
management, communication about treatment preferences, timate its incidence, particularly if reported as ventilator-days
and satisfaction with communication from health care pro- or ventilator-days at risk.29 External reporting pressures also
viders.20 Other investigators have found that among ICU may encourage stricter interpretation of subjective signs and
patients high QODD scores are associated with documenta- other surveillance initiatives that then may artificially lower
tion of a living will, withdrawal of tube feeding, a family VAP rates.
conference to discuss the patient’s end-of-life care, family at In 2005 the ATS and the Infectious Diseases Society of
the bedside at the time of death, and absence of cardiopulmo- America (ATS/IDSA) published guidelines to manage hospital-
nary resuscitation (CPR).21 In particular, symptom control, acquired pneumonia (HAP), VAP, and health care–associated
preparation for death, perceived preservation of dignity and pneumonia (HCAP). There is consistent evidence that strate-
respect, and enough time to visit family members are associ- gies that affect the primary pathophysiologic mechanisms of
ated with high QODD scores.22 VAP reduce the incidence of the disease. Preventive measures
Quality indicators for palliative care have been developed. are best implemented when grouped into “bundles” to improve
However, most of these quality indicators cover one specific overall efficacy and sustain compliance.30-35 Several different
setting or target group, for example, cancer or patients who protocols (or bundles) to reduce VAP are described. For
are at home.23 Ghafoor et al have recently described a set of example, Lansford et  al. described use of head-of-bed eleva-
indicators for palliative care and for support of relatives tion, twice-daily chlorhexidine oral cleans, once-daily ventila-
before or after the patient’s death that appears applicable to tor wean, and conversion from nasogastric to orogastric tube
all settings where palliative care is provided to adult patients. in a trauma ICU; the VAP incidence decreased from 6.9
In addition, there are policy, standard orders, and quality- episodes per 1000 patient-days to 2.8 episodes per 1000
assurance monitoring described for palliative sedation patient-days.36 Cocanour et  al. described a ventilator bundle
therapy,24 but there are few validated outcome measures to (head-of-bed elevation, stress gastritis prophylaxis, DVT pro-
use in improving end-of-life care in the ICU. There are two phylaxis, and planned sedation weans).37 They found that the
main models to integrate ICU and palliative care: (1) “con- VAP incidence was not altered with use of the ventilator
sultative,” which increases the involvement of palliative care bundle alone. Instead a benefit was observed when compli-
consultants for ICU patients and their families, particularly ance with the ventilator bundle was audited daily with weekly
those patients at greatest risk for poor outcome; and (2) caregiver feedback. Other techniques that can be included
“integrative,” which embeds palliative care into daily practice into VAP bundles include proper hand hygiene, high nurse-
for all patients and families in the ICU. These models, to-patient ratio, avoidance of unnecessary transfer of ven­
however, are not mutually exclusive.25 tilated patients and manipulation of ventilator circuits,
incorporation of sedation control and weaning protocols into
patient care, use of noninvasive mechanical ventilation, main-
Ventilator-Associated Pneumonia tenance of adequate pressure of endotracheal cuffs, selective
VAP is a common event in the ICU (8%-28% of mechanically digestive tract decontamination, and use of oral rather than
ventilated patients) and can increase ICU length of stay and nasal intubation, among others. These techniques need to be
adversely affect outcome. For example, Yang et al. found that supplemented with appropriately educated and trained staff
traumatic brain injury (TBI) and stroke patients who develop and a daily checklist. Initiation of a VAP bundle generally is
VAP have greater hospital expenses, longer length of stay in associated with a reduced VAP incidence (on average from
the hospital and in the ICU, and a greater number of readmis- 10 cases/1000 ventilator-days to 3 or 4 cases/1000 ventilator
sions than those without VAP.26 Consequently the incidence days) and with cost savings, but it remains unclear if this
and efforts to prevent VAP often are used as a benchmark of reduces overall ICU mortality.38-40
quality of ICU care. However, the complexity and subjectivity A reduction in the number of days of mechanical ven­
of VAP surveillance can limit its value to assess and compare tilation is an important goal to reduce VAP. This may be
care for ICU patients, and so the use of VAP as a benchmark accomplished through use of protocol-driven weaning
48 Section I—Background

parameters by a multidisciplinary team, including nursing affect patient outcome. The U.S. Centers for Disease
staff and respiratory therapists.41-44 For example, Kollef et al. Control and Prevention (CDC) National Healthcare Safety
in a randomized trial of different weaning methods in medical Network (NHSN; formerly the National Nosocomial Infec-
and surgical ICUs observed that weaning protocols were asso- tions Sur­veillance [NNIS] group) collects national data on
ciated with significant reduction in duration of mechanical catheter-associated bloodstream infections (CA-BSIs). How
ventilation. The least success with a weaning protocol occurred the condition is defined may affect the incidence. For
in the ICU without an ICU director and critical care fellows.41 example, CA-BSIs and catheter-related bloodstream infec-
Use of a multidisciplinary weaning protocol also can help tions (CR-BSIs) differ in the degree of proof needed to
reduce ventilation time in patients ventilated greater than 7 show that the catheter “causes” the infection, so the CR-BSI
days.44 Inclusion of parameters such as assessment of sedation rate is significantly lower than the CA-BSI rate.52 In October
status using a standardized form and standard clinical criteria of 2008, Medicare enacted new regulations that reduced
for weaning eligibility including hemodynamic status, oxy- reimbursement for HAIs and CA-BSIs and in 2009 the IDSA
genation requirements, and minute ventilation can be used to published updated guidelines for the diagnosis and manage-
help clear patients for T-piece or pressure support weans.42 ment of all intravascular catheter-related infections.
Today many hospitals report VAP rates at or close to zero, Several factors such as central venous catheters (CVCs),
so health care–regulating bodies propose that HAP should not total parenteral nutrition (TPN) days, and hemodialysis cath-
be reimbursed and potentially should be a “never event.” eters are associated with nosocomial BSIs.53 Consequently a
However, VAP rates reported by hospital administrative variety of protocols and techniques now are in use to reduce
sources appear to be less accurate than physician-reported the incidence of infections associated with central lines,
rates and underestimate VAP incidence, that is, a never event that is, central line–associated bacteremia (CLAB) or central
may not be possible.45 It also is difficult to unravel the relative line–associated bloodstream infections (CLABSIs). First, is
contribution of care improvements rather than surveillance the use of chlorhexidine or silver sulfadiazine or antibiotic-
effects on the low VAP rates. Furthermore, whether a VAP rate impregnated CVCs54-56 that at the very least appear to reduce
of zero is obtainable in the ICU is unclear because many the risk of colonization and perhaps BSIs. These catheters can
patients who are admitted already intubated have evidence of be expensive and there is a concern that use of antibiotic-
early pneumonia or bacterial growth within 48 hours of impregnated catheters may be associated with an increased
arrival. This suggests that early infection or colonization risk of antimicrobial resistance. In addition, adherence to
occurred before admission.46,47 In the future, surveillance defi- standardization of the processes of care associated with CVC
nitions that use more objective criteria may better mirror and placement rather than use of coated CVCs (i.e., placement,
inform clinical practice.48 This includes objective alternatives dressing, and maintenance/removal protocols) appears to be
such as average duration of mechanical ventilation, length of a more important factor in reduction of CR-BSIs.57 Second is
stay, mortality, and antibiotic prescribing to estimate disease use of chlorhexidine-soaked dressings. For example, the Bio-
burden and guide quality improvement. patch dressing surrounds the hub of the CVC and is saturated
with chlorhexidine. Data from the manufacturer suggest that
its use is associated with a 60% decrease in BSIs and 44%
Pressure Ulcer decrease in local site infections (see www.jnjgateway.com).
The National Quality Forum has identified a pressure ulcer as Third, a reduction in the duration of central venous access and
a hospital-acquired condition (HAC) that is high cost and may peripherally inserted central catheter (PICC) rather than CVC
be prevented by using evidence-based guidelines. The CMS use may help reduce the incidence of CR-BSI particularly in
intends to no longer reimburse acute care facilities for the cost patients who are in the ICU a long time.58 Fourth, resident
associated with facility-acquired (FA) ulcers. ICU patients are oversight and credentialing policy for CVC placement can
at risk for pressure ulcer development because of comorbid help reduce CLABSIs.59 Finally and perhaps most important
diseases, immobility, and use of sedation and analgesia that is a bundle approach with physician and patient specific steps
reduce patient perception of high pressure points. In the Inter- and use of a checklist. Compliance with these protocols
national Pressure Ulcer Prevalence Survey (IPUPS), an obser- (bundles) has improved the safety of hospitalized patients,
vational, cross-sectional cohort study conducted in the United and better compliance appears to be associated with a reduc-
States during 2008 and 2009, FA pressure ulcer rates were tion in infection and CLABSIs.60 Bundle adherence and
highest in ICUs (~10%), although the incidence varied accord- CLABSI rates may depend on the institution; where compli-
ing to ICU type. In 2009, 3.3% of ICU patients developed ance is low the sites often lack a functional team, forcing
severe pressure ulcers (stage III, stage IV).49 There are several functions, and feedback systems.61
risk assessment scales for pressure ulcers, although often they
are not specific to ICU populations.50 The APACHE score also
correlates with pressure ulcer risk. Development of hospital- Glycemic Control (See Also Chapter 14)
wide protocols including early skin assessments and transfers Both hyperglycemia and hypoglycemia can adversely affect
to pressure-reducing mattresses51 can help reduce the inci- ICU outcome.62,63 Furthermore increased insulin administra-
dence of pressure ulcers or increase the length of time it takes tion and poor glycemic control in nondiabetic patients
for an ulcer to develop. may aggravate outcome, hence insulin and glycemic control
protocols have developed. The most successful protocols
include bedside glucose monitoring, nursing-driven proto-
Bloodstream Infections cols, and computerized decision-making algorithms.64-66
Hospital acquired infections (HAIs) and in particular Although there are many clinical and fiscal benefits to
bloodstream infections (BSIs), are common and adversely glucose control67-71 there are barriers to “tight” glucose
Section I—Background 49

control in the ICU that include lack of a defined target devices is subject to federal and state regulations, and accredi-
glucose range, health care provider fear of hypoglycemia, tation standards developed by the College of American
and changes to subcutaneous insulin, par­ticularly when Pathologists and The Joint Commission. Future research will
patients’ nutritional and clinical status is in flux.67-72 need to address whether computerized decision support
Hyperglycemia is common among ICU patients, and 90% systems and newer technologies that allow accurate and con-
of critically ill patients develop blood glucose concentrations tinuous or near-continuous BG measurements improves the
greater than 110 mg/dL (6.1 mmol/L). Van den Bergh’s land- quality of glycemic control.83
mark study published in 2001 supported the use of intensive
insulin therapy (IIT) to target normoglycemia (a blood
glucose concentration of 80-110 mg/dL [4.4-6.1 mmol/L]) in
Structural Quality Indicators
the critically ill and postoperative patient.70 However, other Structure in the ICU refers to the type and size of the ICU,
investigators have not been able to replicate these findings, and ICU design, availability of technology, and staffing among
a meta-analysis that included 29 randomized trials with 8432 others (see Chapter 3 for a review on ICU design). Interven-
patients showed that tight glucose control is not associated tions that influence structure usually take longer to implement
with reduced hospital mortality but is associated with an and are more expensive than changes to processes of care. The
increased risk of hypoglycemia in critically ill adult patients.73 following text discusses organization and management in
A paradigm shift in glucose control in the ICU occurred after critical care by reviewing ICU staffing, transport, handovers,
the publication in 2009 of the normoglycemia in intensive and telemedicine.
care evaluation survival using glucose algorithm regulation
(NICE-SUGAR).74 This trial demonstrated increased mortal-
ity and incidence of hypoglycemia in patients managed with Physician Staffing Models
IIT. Current recommendations are more moderate and target Models of physician staffing in the ICU include low intensity,
a blood glucose (BG) concentration between 144 mg/dL and or open, and high intensity, or closed. In low-intensity systems
180 mg/dL (8-10 mmol/L). These “moderate” insulin proto- there is either no or only partial intensivist direction. A
cols are in use in many ICUs and appear to avoid hyper­ patient’s primary attending physician controls the patient’s
glycemia and low glucose variability while rarely inducing admission to and discharge from the ICU. Intensivists may
hypoglycemia.75,76 These various studies were conducted serve as part of an acute resuscitation team or be consulted
largely in non-NCCU patients, and laboratory studies that for a particular management question. In a closed unit, a
indicate the deleterious effects of severe hyperglycemia in the patient’s attending physician yields control of patient care to
context of anaerobic conditions in the brain and clinical a critical care team. The team model that characterizes the
microdialysis studies in severe brain injury that show intersti- closed unit empowers medical and nursing administrators to
tial hypoglycopenia in NCCU patients with tight glucose effect standard protocols of care. In the United States, the
control77 underscore the still unsettled question on optimal model of ICU care historically has been a low-intensity
glucose management in NCCU patients. system.84,85 For example, in 2000 Angus et al. published a
Most hospitals have established glycemic control programs. survey of 393 ICUs; 23% were managed by a full-time inten-
Based on reports from the 2009 Remote Automated Labora- sivist; an intensivist consultant was available in 14%; and 29%
tory System (RALS) that provides trends in glycemic control had no access to an intensivist at all.84 High-intensity units
between 2006 and 2009 from 576 U.S. hospitals (including usually are associated with teaching hospitals, hospitals with
175 million BG results, 25% from the ICU) for many institu- large patient volume, surgical ICUs, or trauma ICUs and are
tions, glycemic control has improved: the mean range of BG more prevalent.85
results in ICU patients in 2009 was 121.1 to 217 mg/dL.78 Several studies describe a benefit to closed rather than open
Earlier RALS reports show that hospital hyperglycemia systems.86-98 For example, Pronovost et al. reviewed 27 ran-
(>180 mg/dL) prevalence was 46.0% in the ICU and 31.7% domized or observational trials that described the effect of
outside the ICU. Hospital hypoglycemia (<70 mg/dL) preva- physician staffing on mortality and length of stay in medical,
lence was 10.1% in the ICU and 3.5% for non-ICU readings.79 surgical, mixed medical and surgical, or pediatric ICUs. In
What is unclear is whether quality indicators for diabetes care nearly every study, decreased mortality was associated with a
are associated with patient outcomes. For example, Sidoren- closed ICU and in half decreased ICU length of stay was asso-
kov et al. conducted a systematic literature review of 24 studies ciated with a closed ICU system.1 The benefits of a high-
(but not limited to ICU patients) to test this relationship.80 intensity model may be greatest when patients are admitted at
There was insufficient evidence that quality indicators pre- night or during the weekend,88 patients admitted more than
dicted better patient outcomes. Consequently insulin infusion 48 hours, or among elderly patients with more comorbid dis-
protocols (IIPs) to achieve desired BG targets must be indi- eases.92 The benefits of closed systems on patient outcome,
vidualized and validated to the ICU and institution where they ventilator-days, length of stay, morbidity, cost, and discharge
are used. The implementation of IIPs is not a simple process disposition among other measures also are seen in various
but requires a carefully planned, inclusive, and continual team subspecialty ICUs including trauma, surgical, and neurocriti-
effort. Continual assessment of protocol errors, adverse events, cal care89-92,96,97 in patients with acute lung injury or who
staff satisfaction, and outcomes is vital to success, the goal of undergo specific high-risk procedures such as abdominal
which is effective glucose control while avoiding hyperglyce- aortic surgery or esophageal resection.93-95 For example,
mia, hypoglycemia, and glucose variability, all of which are Varelas et al. compared patient outcome and disposition using
detrimental.81,82 Accurate and efficient glucose monitoring historical controls in a university hospital NCCU that changed
devices, including automated point-of-care (POC) BG data from an open ICU to one with a dedicated neurointensivist.89
management software therefore are essential. Each of these Hospital and ICU length of stay were reduced and fewer
50 Section I—Background

patients were discharged to nursing homes when the NCCU trainees. However, a study by Prasad et al. suggests this is not
was led by a neurointensivist. Implementing a high-intensity so.110 Second, the ICU environment is complex and highly
staffing model including continuous (24 hour) onsite pres- dynamic and often requires that clinicians adjust and deviate
ence of a critical care specialist also is associated with improved from standard protocol when confronted with nonstandard
processes of care and staff satisfaction.98 circumstances. These deviations may be errors or innovations.
Observations in acute trauma care indicate that protocol devi-
ations by more experienced physicians are a combination of
Evidence-Based Protocols errors and innovations, whereas the deviations by less experi-
The mechanisms that underlie improved outcomes in high- enced practitioners are mostly errors.111 Finally, bundles of
intensity ICUs are unclear but may be associated with care often are implemented under pressure from hospital
increased use of evidence-based quality indicators and administrators or regulatory bodies with little regard for the
increased use of evidence-based guidelines or protocols.99 context in which the evidence was generated and without
These protocols include DVT prophylaxis, stress ulcer pro- understanding the difference between efficacy and effective-
phylaxis, sedation interruption, spontaneous breathing trials, ness;112 that is, the interventions could be implemented to a
and glycemic control, among others. Even just having an larger group of ICU patients that differ from those in whom
available intensivist can contribute to evidence-based proto- the evidence was gathered. The risk then is that the bundle
col enforcement.99 Other processes that may be associated tools or protocols can be used for quality control, performance
with better outcomes and are improved in a closed system evaluation, and legal and regulatory purposes out of context
include weaning by respiratory therapist protocol, daily and without robust validation. This will put patients and pro-
rounds with pharmacists, nurse-to-patient ratio of greater fessionals at risk of being harmed by the protocols or bundles
than 1 : 2, and ICU mortality and morbidity reviews.100 A of care.
mandatory bedside checklist can be a simple, cost-effective
method to prevent errors of omission and improve imple-
mentation of ICU best practices.101 Intensive Care Unit Physician
Introduction of evidence-based protocols requires an audit
of current practice, expert-led education sessions, and dis-
Staffing Plan
semination of algorithms (because adoption of protocols In the United States there still is a relative paucity of ICUs
with low baseline adherence changes clinical practice little).102 that are closed and have a board-certified intensivist available
Evidence-based guidelines create a base to allow change in at all times.84-95 These staffing issues are changing in part
health care practitioner behavior. However, they do not in because of the Leapfrog initiative or addressed through the
and of themselves effect change or outcome unless their use development of the eICU and telemedicine (see Chapters 42
is accompanied by conversion of a core of key goals in the through 44). The Leapfrog organization recommends all
guideline recommendations to quality indicators, and with ICUs have a board-certified intensivist available onsite during
regular feedback on performance, for example, weekly.103,104 daytime hours and be immediately reachable through a pager
In addition, the protocols must be updated to incorporate during nighttime hours with a critical care physician or “phy-
new guidelines and evidence if these new guidelines and evi- sician extender” available within 5 minutes of the ICU at
dence meet rigorous critical appraisal. Most appraisal systems night. These rules formulate the Leapfrog’s 2002 ICU Phy­
consider randomized controlled trials (RCTs) the gold stan- sician (IPS) Standard (www.leapfroggroup.org).113 There
dard given their ability to limit bias. However, many RCTs in are, however, several obstacles to the Leapfrog standard,
critical care have failed,105,106 or a number of trials have had including (1) primary physician resistance to yield control
results that are initially positive and then subsequent trials over patient care, (2) perceived loss of continuity of patient
are less positive.107 This has led to uncertainty about the sci- care, (3) shortage of critical care physicians to staff ICUs,
entific approach to ICU research and that guideline develop- (4) limited fiscal incentive for critical care–certified physi-
ment may need reevaluation. In part this may be explained by cians, and (5) poor fiscal incentive for hospitals with small
the heterogeneous nature of ICU patients, who all have dif- ICUs to hire full-time ICU physicians. These factors are
fering physiologic requirements over time. Consequently, greater in smaller nonacademic hospitals where there may
ICU clinicians routinely adjust treatment dose and care based not even be an ICU director.100,114 To overcome some of these
on multiple factors that can be difficult to simulate in a clini- obstacles, many ICU directors recommend financial incentive
cal trial.108 Procedure or disease-specific registries, observa- from either a hospital, the government, or a third party to
tional cohorts, or studies that aim to understand variation ensure compliance with the Leapfrog initiative.114 Consistent
in performance and delivery of care and how to reduce with this a survey of ICU directors as well as chief medical
individual and organizational underperformance may help officers suggest that adoption of the IPS standard was facili-
answer some of the limitations observed with RCTs in critical tated by intensivist salary support.115
care.105,107 The past decade also has seen the bringing together What is the cost of achieving the Leapfrog IPS standard?
of positive studies and interventions into bundles of Hypothetical financial models have been applied to 6-, 12-,
care (goal-directed therapy, protocolized care). While these and 18-bed ICUs. The variables in the model include annual
bundles appear to be associated with improved outcomes, admissions, intensivist salary, revenue from intensivist billing,
there needs to be a balance between individualized versus and salary of critical care physician extenders among others.
protocolized care.109 Savings, because of improved patient outcomes and shorter
There are several concerns with protocols in ICU care. First, ICU length of stays, range between $510,000 and $3.3
the use of clinical protocols may negatively affect medical million.116 The greatest savings occur in larger ICUs and are
education by eliminating clinical decision making from associated with ICU beds, length of stay, and cost of ICU bed
Section I—Background 51

per day. However, under worst-case scenario conditions, costs clinical handover practices (i.e., mechanisms to transfer
may range from $890,000 to $1.3 million. Small ICUs may be information, responsibility, and authority) and many new
disproportionately more vulnerable to worst-case conditions doctors feel unprepared for the handover process.129 Studies
and experience net loss more frequently under the IPS in ICU care also show that medical members of the
standard.114 ICU team provide data that can differ from the medical
The final obstacle is having enough staff. The Committee record when asked to recall clinical information discussed
on Manpower for Pulmonary and Critical Care Societies at handover.130
(COMPACCS), commissioned by the American College of Efforts to improve handover safety and effectiveness are
Chest Physicians, the ATS, and the Society of Critical Care ongoing. Multidisciplinary teams administer critical care, and
Medicine, assessed the future supply and demand for intensiv- it is conceivable that behavioral methods to enhance team
ists until 2030. COMPACCS found that the supply of intensiv- performance may affect the quality of care. This can be
ists likely will be static until 2030, whereas the demand for achieved in development of safe handover practices that
critical care services, particularly by older patients, will increase requires (1) awareness of handover problems and opportuni-
as the baby boomer generation ages.117,118 Other critical care ties; (2) identification of solutions; (3) adaptation of local best
workforce analyses estimate a 35% shortage of intensivists practices, for example, structured handovers that include both
by 2020, particularly in surgical critical care in the United written and verbal components to improve information
States.119 The likely intensivist workforce shortage calls for exchange; (4) multidisciplinary education about the clinical
creative staffing solutions. Among these are (1) use of “novel” handover process to encourage teamwork and a common lan-
critical care personnel including critical care–certified physi- guage among different services in the hospital; and (5) insti-
cian assistants and nurse practitioners and physicians from tutionalizing practice changes.131 Three factors—information
emergency medicine and anesthesiology who receive ICU transfer, shared understanding, and working atmosphere—
training; (2) regionalization; and (3) telemedicine. In many predict handover quality.132 However, handover is not only a
countries around the world anesthesiologists provide critical skills-based task; there also are complex interactions between
care but in the United States in 2006, less than 4% of board- individual and systems factors. A full understanding of team
certified anesthesiologists were also certified in critical care. principles including leadership, psychological safety, transac-
There are two major barriers to dual certification: a paucity of tive memory, and accountability all are important to the han-
programs that offer such certification, and surgeons and anes- dover process (and to critical care) and can be used to improve
thesiologists who practice critical care tend to take a pay cut.117 patient care.133 These same team principles also can be applied
An alternative may be increased use of acute care nurse prac- to rapid response systems, in which there are few benchmark-
titioners (ACNPs). In a “semiclosed” surgical intensive care ing tools to allow comparison for quality assurance, and car-
unit (SICU) the presence of ACNPs increases compliance with diopulmonary resuscitation (CPR).134,135
clinical practice guidelines.120
In 2004 the American Association of Critical-Care Nurses,
the American College of Chest Physicians, the ATS, and the
Telemedicine
Society of Critical Care Medicine, convened the Critical Care Telemedicine is described in Chapters 42 and 44 but is briefly
Workforce Partnership and published a white paper titled discussed here to address physician staffing. Since the concept
“FOCCUS—Framing Options for Critical Care in the United was introduced in 1977, several publications have described
States.” The conference highlighted that although trauma its use. In particular, the presence of an off-site intensivist who
patients are classified by need into level of trauma service, monitors patient care remotely using cameras and data trans-
there is no tiered critical care delivery system. Such a regional- mission equipment and then communicates his or her recom-
ized and standardized triage system may appropriately direct mendations to the onsite personnel either during formal
intensivist workforce demand to higher-level, higher-acuity rounds or during informal communication with the onsite
centers.117 attending staff is associated with decreased ICU and hospital
mortality and decreased ICU length of stay and costs.136 These
same benefits may be seen when a single intensivist team
Clinical Handovers monitors multiple ICU sites. Together these early observa-
Staffing requirements and transport of ICU patients for diag- tional data suggest that telemedicine could be used to stem the
nostic or therapeutic procedures mean clinical handover is future demand for intensivists.137
a necessary process in ICU care. There is a well-described In 2003 VISICU, Inc was founded to provide ICU telemedi-
association between intrahospital transport of critically ill cine. These sites are typically multihospital systems, and
patients and complications including exacerbation of pulmo- several are associated with large academic institutions. In
nary function and intracranial physiology.121-124 The rate of smaller networks (fewer than 70 beds), a remote team consists
adverse events during patient transport, both intra- and of one critical care nurse and one critical care physician. In
interhospital, which by definition include transitions of care, larger networks (more than 120 ICU beds), two critical care
can be reduced use of a specialized transport team and struc- physicians and three critical care nurses remotely monitor
tured process.125,126 In addition, deficiencies in the handover patient care. The hours of remote monitoring vary by site and
process can introduce error and discontinuities in care and need (e.g., some sites only use VISICU at nights and week-
compromise patient care, whereas distractions during hando- ends). There are several challenges to implement telemedicine
ver can adversely affect information transfer.127 By contrast, a in the ICU, including (1) cost, because ICU telemedicine does
formal, structured handover process, including during transi- not command separate billing and so health systems assume
tions of care can reduce medical errors and improve team- the costs of salary and infrastructure; (2) restructuring of
work.128 However, many hospitals lack safe and effective rounds to multidisciplinary rounds when the remote team can
52 Section I—Background

inform the onsite team; (3) staff training to use the remote
system; (4) maintenance costs for software or hardware down- Which Indicators Can Drive
time, which may have real clinical implications if an ICU is Safety and Quality in the
entirely dependent on remote monitoring; (5) privacy of
patient information and confidentiality; and (6) different
Intensive Care Unit?
management policies and protocols between the “remote” Quality assurance in health care continues to mature and
intensivist and onsite personnel.138 benchmarks are used more frequently to compare perfor-
Despite these challenges ICU telemedicine may help address mance between institutions. A critical assessment of the diver-
the shortage of board-certified critical care physicians to staff sity in critical care practice and organization and their
ICUs. In addition, new standards for supervision and duty association with outcomes can help inform the development
hours for residency programs released by the Accreditation of evidence-based guidelines. This is important given the
Council for Graduate Medical Education (ACGME) were increasing use of critical care services and the significant cost
introduced in July 2011. In ICUs that depend on residents for of ICU care. However, the robustness of this comparison
portions of patient care, telemedicine, among other options depends on which quality indicators or benchmarks are used.
such as increased use of nurse practitioners and physician Other important considerations in benchmark use include
assistants or partnerships with hospitalists, may offer a solu- definition, time frame, case-mix adjustment, and the context
tion to the change in workflow. However, further research is in which the original benchmark was described and then is
needed to examine how the new duty hours influence patient used. In 2009 the ESICM formed a Safety and Quality Task
safety, continuity of care, resident learning, and ICU staff- Force with the intent of improving the safety and quality of
ing.139 The University Health System Consortium ICU Tele- ICU care. This task force developed a directive for change
medicine Task Force, an independent organization with no signed by 57 national and international critical care organiza-
financial interest in VISICU, has issued recommendations for tions in the Declaration of Vienna141 and next identified a set
institutions that are interested in a remote ICU program. of indicators that could be used to measure the quality of
Among these recommendations are warnings that a telemedi- ICU care in a prospective fashion. To do this the Safety and
cine program does not obviate the need for onsite full or Quality Task Force used 18 nominated experts from 9 coun-
part-time intensivists and that such a program does not serve tries and a modified Delphi process. A final set of 9 indicators,
hospitals with small ICU capacity.140 These cautions serve as each of which had a greater than 90% agreement rate, from
important reminders that the challenge of appropriate physi- 111 potential indicators, were agreed upon (Table 6.1).9
cian staffing to maintain quality in ICU care still need to be Similar efforts have been made in stroke research to ensure
resolved. common standards to define quality indicators, collect infor-
mation, and promote valid comparisons of acute stroke care
between institutions.142,143
Benchmarking and Quality
Improvement in the Future
The Practice of Neurocritical Care Does Benchmarking
To improve quality (of therapy), it is important to know, eval-
Influence Outcome?
uate, and make transparent daily ICU processes. Much of the An ICU benchmark may be defined as a quantitative, stan-
literature and research associated with quality assurance and dardized measurement to compare the performance of differ-
benchmarking is, in general, medical or surgical critical care ent ICUs. Industries other than health care have used
and there is little that is specific to neurocritical care. Can
these results be extrapolated to the NCCU or are there differ-
ences that suggest NCCU specific research is needed? Large
databases that describe the range of critical care practice Table 6.1  Prospectively Defined Indicators
matched to acuity-adjusted outcomes have only recently to Improve the Safety and Quality of Care
become available. For example, Lilly et al. (2011) provided for Critically Ill Patients*
benchmark data from 243,553 adult admissions, including Intensive care unit (ICU) fulfils national requirements to
7692 patients with neurologic disorders from 271 ICUs all provide intensive care
supported with ICU telemedicine monitoring.19 There were 24-hour availability of a consultant level intensivist
Adverse event reporting system
important patient demographic (age) and clinical differences Presence of routine multidisciplinary clinical ward rounds
(APACHE IV) between NCCUs and other ICUs. The average Standardized handover procedure for discharging patients
length of stay in the ICU was just over 3 days. However, neu- Reporting and analysis of standardized mortality ratio (SMR)
roscience and trauma patients had longer stays, and NCCU ICU readmission rate within 48 hours of ICU discharge
patients more frequently required rehabilitation or chronic The rate of central venous catheter–related bloodstream
infection
care than other ICU types. Unadjusted mortality rates were The rate of unplanned endotracheal extubations
less for nontrauma surgical or mixed ICUs than medical or
neuroscience ICUs (hospital 10.1%, ICU 6.4%). The APACHE Modified from Rhodes A, Moreno RP, Azoulay E, et al. Prospectively defined
indicators to improve the safety and quality of care for critically ill patients:
IV model overpredicted hospital and ICU mortality for all a report from the Task Force on Safety and Quality of the European Society
ICU types. These data suggest that there may be enough dif- of Intensive Care Medicine (ESICM). Intensive Care Med 2012; Jan 26. Epub
ferences between NCCUs and other ICUs that specific NCCU ahead of print.
*Final set of prospectively defined indicators agreed upon by the Task Force
safety, and quality assurance may better inform select practices on Safety and Quality of the European Society of Intensive Care Medicine.
in neurocritical care.
Section I—Background 53

benchmarks for many years in large part to compare their


performance against competitors and so gain greater market How to Ensure a Successful
share or reduce costs. The primary goal is to increase efficiency Quality Assurance or
and reduce variability that in turn should increase quality.
Benchmarking has become more frequent in health care,
Benchmarking Initiative
driven in large part by a need to deliver care at lower costs Several prerequisites are necessary to ensure a successful
(i.e., increased efficiency) and to standardize care (i.e., vari- quality assurance or benchmarking initiative: (1) a willingness
ability reduction). When the standards, including clinical to compare ICUs, (2) intent to act on the results with quality
guidelines, order sets, and care bundles are evidence based it improvement programs, (3) accurate clinical documentation
is postulated that the reduced variability will improve the with physician buy-in,152,153 (4) mature data collection tools,
quality of care and so enhance outcome. However, it is difficult and (5) a synergistic effort between the hospital administra-
to demonstrate that benchmarking is associated with improved tors and the medical staff with assembly of a multidisciplinary
outcomes.144-147 For example, Hernandez et al. linked Medi- team.154 Within each ICU or department the foundation
care claims with the Get With the Guidelines-Heart Failure needed to measure and enhance quality is good team spirit,
registry of the American Heart Association for heart failure communication, and cooperation.155 Program implementa-
patients greater than 65 years old and admitted to hospital.146 tion requires (1) choosing the right tool to measure perfor-
The agreement between different methods to rank hospital- mance and outcome and identification of clinical processes
based quality of care and 30-day mortality or readmission was that need to be changed; (2) standard measurement defini-
poor. Similarly Sciacovelli et al., who collected data on quality tions, a statement of desired goals, and a clear timeline; (3)
indicators over a 3-year period from a clinical laboratory, teaching sessions and daily goal sheets or checklists; (4) con-
observed that processes under laboratory control but not pro- sistent measurement and feedback practices of both process
cesses that required cooperation between the patient care pro- compliance and outcome data156; and (5) use of human factor
viders and the laboratory improved (i.e., more effective analysis that can provide insights into how interactions
laboratory function did not necessarily mean better patient between organizations, tasks, and the individual worker influ-
care).147 On the other hand, Woodhouse et al. in a benchmark- ence behavior and affect systems reliability because unreliable
ing program in eight ICUs in the Netherlands observed a 3% delivery of best practice is a contributor to medical error. Data
decline in the SMR each year over an 8-year period.144 There that are collected particularly for benchmarking should be (1)
were three main components to their benchmarking: (1) incorporated into the daily routine with minimal added work;
quality (e.g., SMR), (2) availability (e.g., was a bed available (2) clinically relevant; (3) comparable across various ICUs
for a patient who required ICU care), and (3) efficiency (e.g., with risk adjustment; (4) valid (i.e., a measurement change
length of stay and ventilator duration). They expected bench- results in a change in the outcome of interest); (5) robust; (6)
marking would lead to a convergence of “best practices” standardized; and (7) anonymous.144 The collected data can
between hospitals. However, they found that over time all the then be used to guide quality improvement. This may be best
ICUs improved in such a way that the gap between the best achieved through small tests of change in an incremental
and worst performing ICUs remained. process using repeat evaluation and refinement such as a Plan-
It has become increasingly common to release information Do-Study-Act (PDSA) cycle. Pronovost et al. have suggested a
about the performance of hospitals, health care providers, and five-phase translational (T) framework to develop programs
health care organizations into the public domain. Ideally to reduce preventable harm or enhance quality157 (Table 6.2).
public reporting should be focused on the program rather It should be remembered that sustained high performance is
than individual health care provider because modern medi- not a permanent state that an institution or ICU reaches but
cine including critical care or neurosurgery requires a rather a dynamic balance and one that is constantly moving
team-based approach. However, it is unclear whether public forward.158
reporting affects the performance of health care professionals,
improves quality, or alters behavior of consumers. A Cochrane
analysis148 found a small effect for coronary bypass surgery
and low-complication outliers for lumbar diskectomy. These
effects disappeared within 2 months after public release.
Table 6.2  Five-Phase Translational (T)
However, overall the analysis concluded that public release of
Framework to Develop Programs to Reduce
performance data does not change consumer behavior or
Preventable Harm or Enhance Quality
improve care. Similarly, Ryan and Blustein, who examined the
Massachusetts Medicaid’s (MassHealth) hospital-based pay- Phase T0: discover opportunities and approaches to prevent
for-performance program, did not find improved quality adverse health care events
despite substantial financial incentives.149 Furthermore, rather Phase T1: use T0 discoveries to develop and test interventions
on a small scale
than outcome, it appears that improvements in process and Phase T2: broaden and strengthen the evidence base for
structure are most likely to increase a patient’s overall evalu- promising interventions to develop evidence-based
ation of the quality of care.150 Physicians also need to be guidelines
mindful of unintended negative consequences associated with Phase T3: translate guidelines into clinical practice
Phase T4: implement and evaluate T3 work on a national and
performance measurement reporting. For example, many international scale
payers and regulatory bodies measure hospital quality of care
by the rate of HAP. Public reporting of these rates may then Modified from Pronovost PJ, Cardo DM, Goeschel CA, et al. A research
framework for reducing preventable patient harm. Clin Infect Dis
drive increased use of antibiotics and so lead to bacterial 2011;52(4):507–13.
resistance.151
54 Section I—Background

II and Simplified Acute Physiology Score II. J Crit Care 2011;26(1):


Conclusion 105.e11–18.
16. Moreno RP, Bauer P, Metnitz PG. Characterizing performance profiles of
Measurement of hospitals’ performance has become more ICUs. Curr Opin Crit Care 2010;16(5):477–81.
commonplace in part to inform patient choices and payer 17. Jarman B, Aylin P: Death rates in England and Wales and the United States:
reimbursement decisions including pay-for-performance ini- variation with age, sex, and race. BMJ 2004;329:1367.
tiatives and potential nonpayments for readmissions and 18. Brinkman S, Abu-Hanna A, van der Veen A, et al. A comparison of the
performance of a model based on administrative data and a model based on
adverse events. Consequently, many institutions have replaced clinical data: effect of severity of illness on standardized mortality ratios of
revenue growth models of external market and service expan- intensive care units. Crit Care Med 2012;40(2):373–8.
sion with cost management and enhancement of institutional 19. Lilly CM, Zuckerman IH, Badawi O, et al. Benchmark data from more than
capabilities to ensure fiscal stability. The significant cost of 240,000 adults that reflect the current practice of critical care in the United
States. Chest 2011;140(5):1232–42.
critical care services has prompted the medical community
20. Curtis JR, Patrick DL, Engelberg RA, et al. A measure of the quality of dying
and regulatory agencies to examine the range of organization and death: initial validation using after-death interviews with family
and practice in critical care and how this is associated with members. J Pain Symptom Manage 2002;24(1):17–31.
outcomes. This provides opportunities to improve delivery of 21. Glavan BJ, Engelberg RA, Downey L, et al. Using the medical record to
care and enhance quality of care particularly when coupled evaluate the quality of end-of-life care in the intensive care unit. Crit Care
Med 2008;36(4):1138–46.
with evidence-based guidelines. Much of the focus has been 22. Mularski RA, Heine CE, Osborne ML, et al. Quality of dying in the ICU:
on preventable complications (e.g., medical errors, health ratings by family members. Chest 2005;128(1):280–7.
care–associated infections, and venous thromboembolism) 23. Claessen SJ, Francke AL, Belarbi HE, et al. A new set of quality indicators for
that are common in hospitalized and ICU patients in general. palliative care: process and results of the development trajectory. J Pain
Symptom Manage. 2011;42(2):169–82.
A large body of scientific evidence exists from clinical trials to
24. Ghafoor VL, Silus LS. Developing policy, standard orders, and quality-
provide evidence-based guidelines to manage many of these assurance monitoring for palliative sedation therapy. Am J Health Syst
conditions. However, whether these recommendations apply Pharm 2011;68(6):523–7.
to all patients in the NCCU or how best to enhance outcome 25. Nelson JE, Bassett R, Boss RD, et al. Improve Palliative Care in the Intensive
through quality assurance initiatives in the NCCU still requires Care Unit Project. Models for structuring a clinical initiative to enhance
palliative care in the intensive care unit: a report from the IPAL-ICU project
many questions to be answered. (Improving Palliative Care in the ICU). Crit Care Med 2010;38(9):1765–72.
26. Yang CC, Shih NC, Chang WC, et al. Long-term medical utilization
References following ventilator-associated pneumonia in acute stroke and traumatic
brain injury patients: a case-control study. BMC Health Serv Res 2011;
1. Pronovost PJ, Angus DC, Dorman TM, et al. Physician staffing patterns and 11:289.
clinical outcomes in critically ill patients: a systematic review. JAMA 2002; 27. Klompas MM, Platt RM. Ventilator-associated pneumonia: the wrong quality
288(17):2151–62. measure for benchmarking. Ann Intern Med 2007;147(11):803–5.
2. Pronovost PJ, Miller MR, Dorman TM, et al. Developing and implementing 28. Uckay I, Ahmed QA, Sax H, et al. Ventilator-associated pneumonia as a
measures of quality of care in the intensive care unit. Curr Opin Crit Care quality indicator for patient safety? Clin Infect Dis 2008;46(4):557–63.
2001;7(4):297–303. 29. Eggimann P, Hugonnet S, Sax H, et al. Ventilator-associated pneumonia:
3. Mallon WJ. Earnest Amory Codman: the end result of a life in medicine. caveats for benchmarking. Intensive Care Med 2003;29(11):2086–9.
Philadelphia: Saunders; 2000. 30. Mangino JE, Peyrani P, Ford KD, et al. Development and implementation of
4. Deming WE. Out of the crisis. Cambridge, MA: MIT Press; 1986. a performance improvement project in adult intensive care units: overview
5. Donabedian A. The quality of care: how can it be assessed? JAMA 1988; of the Improving Medicine Through Pathway Assessment of Critical Therapy
260(12):1743–8. in Hospital-Acquired Pneumonia (IMPACT-HAP) study. Crit Care 2011;
6. Sirio CA, Shepardson LB, Rotondi AJ, et al. Community-wide assessment of 15(1):R38.
intensive care outcomes using a physiologically based prognostic measure: 31. Ramirez P, Bassi GL, Torres A. Measures to prevent nosocomial infections
implications for critical care delivery from Cleveland health quality choice. during mechanical ventilation. Curr Opin Crit Care 2012;18(1):86–92.
Chest 1999;115(3):793–801. 32. Rello J, Lode H, Cornaglia G, et al. VAP care bundle contributors:
7. Glance LG, Osler TM, Dick AP. Rating the quality of intensive care units: is a European care bundle for prevention of ventilator-associated pneumonia.
it a function of the intensive care unit scoring system? Crit Care Med 2002; Intensive Care Med 2010;36(5):773–80.
30(9):1976–82. 33. Roberts N, Moule P. Chlorhexidine and tooth-brushing as prevention
8. Pronovost PJ, Berenholtz SM, Ngo K, et al. Developing and pilot testing strategies in reducing ventilator-associated pneumonia rates. Nurs Crit Care
quality indicators in the intensive care unit. J Crit Care 2003;18(3): 2011;16(6):295–302.
145–55. 34. Labeau SO, Van de Vyver K, Brusselaers N, et al. Prevention of ventilator-
9. Rhodes A, Moreno RP, Azoulay E, et al. Prospectively defined indicators to associated pneumonia with oral antiseptics: a systematic review and
improve the safety and quality of care for critically ill patients: a report from meta-analysis. Lancet Infect Dis 2011;11(11):845–54.
the Task Force on Safety and Quality of the European Society of Intensive 35. Li Bassi G, Torres A. Ventilator-associated pneumonia: role of positioning.
Care Medicine (ESICM). Intensive Care Med 2012; Jan 26. Epub ahead of Curr Opin Crit Care 2011;17(1):57–63.
print. 36. Lansford T, Moncure M, Carlton E, et al. Efficacy of a pneumonia prevention
10. Lilford R, Pronovost PJ. Using hospital mortality rates to judge hospital protocol in the reduction of ventilator-associated pneumonia in trauma
performance: a bad idea that just won’t go away. BMJ 2010;340:955–7. patients. Surg Infect 2007;8(5):505–10.
11. Huang DT, Clermont G, Kong L, et al. Intensive care unit safety culture and 37. Cocanour CS, Peninger M, Li T, et al. Decreasing ventilator-associated
outcomes: a US multicenter study. Int J Qual Health Care 2010;22(3): pneumonia in a trauma ICU. J Trauma 2006;61(1):122–9.
151–61. 38. Bird D, Zambuto A, O’Donnell C, et al. Adherence to ventilator-associated
12. Sakr Y, Marques J, Mortsch S, et al. Is the SAPS II score valid in surgical pneumonia bundle and incidence of ventilator-associated pneumonia in the
intensive care unit patients? J Eval Clin Pract 2010;Sep 22. Epub ahead of surgical intensive care unit. Arch Surg 2010;145(5):465–70.
print. 39. Stone Jr ME, Snetman D, O’ Neill A, et al. Daily multidisciplinary rounds to
13. Poole D, Rossi C, Anghileri A, et al. External validation of the Simplified implement the ventilator bundle decreases ventilator-associated pneumonia
Acute Physiology Score (SAPS) 3 in a cohort of 28,357 patients from 147 in trauma patients: but does it affect outcome? Surg Infect 2011;12(5):373–8.
Italian intensive care units. Intensive Care Med 2009;35(11):1916–24. 40. Sundar KM, Nielsen D, Sperry P. Comparison of ventilator-associated
14. Niskanen M, Reinikainen M, Pettilä V. Case-mix-adjusted length of stay and pneumonia (VAP) rates between different ICUs: implications of a zero VAP
mortality in 23 Finnish ICUs. Intensive Care Med 2009;35(6):1060–7. rate. J Crit Care 2011;Jul 5. Epub ahead of print.
15. Brinkman S, Bakhshi-Raiez F, Abu-Hanna A, et al. External validation of 41. Kollef MH, Shapiro SD, Silver P, et al. A randomized, controlled trial of
Acute Physiology and Chronic Health Evaluation IV in Dutch intensive care protocol-directed versus physician-directed weaning from mechanical
units and comparison with Acute Physiology and Chronic Health Evaluation ventilation. Crit Care Med 1997;25(4):567–74.
Section I—Background 55

42. Dries DJ, McGonigal MD, Malian MS, et al. Protocol-driven ventilator 47. Bouadma L, Deslandes E, Lolom I, et al. Long-term impact of a multifaceted
weaning reduces use of mechanical ventilation, rate of early reintubation, prevention program on ventilator-associated pneumonia in a medical
and ventilator-associated pneumonia. J Trauma Inj Infect Crit Care 2004; intensive care unit. Clin Infect Dis 2010;51(10):1115–22.
56(5):943–52. 48. Klompas M. Is a ventilator-associated pneumonia rate of zero really possible?
43. Henneman EA, Dracup KA, Ganz T, et al. Effect of a collaborative weaning Curr Opin Infect Dis 2012;Jan 13. Epub ahead of print.
plan on patient outcome in the critical care setting. Crit Care Med 2001; 49. VanGilder C, Amlung S, Harrison P, et al. Results of the 2008-2009
29(2):297–303. International Pressure Ulcer Prevalence Survey and a 3-year, acute care,
44. Henneman EA, Dracup KA, Ganz T, et al. Using a collaborative weaning unit-specific analysis. Ostomy Wound Manage 2009;55(11):39–45.
plan to decrease duration of mechanical ventilation and length of stay in the 50. Paul JA, Keller JW, van Ramshorst B, et al. Pressure ulcers in intensive care
intensive care unit for patients receiving long-term ventilation. Am J Crit patients: a review of risks and prevention. Intensive Care Med
Care 2002;11(2):132–40. 2002;28:1379–88.
45. Thomas BW, Maxwell RA, Dart BW, et al. Errors in administrative-reported
ventilator-associated pneumonia rates: are never events really so? Am Surg A complete list of references for this chapter can be found online at
2011;77(8):998–1002. www.expertconsult.com.
46. Wahl WL, Zalewski C, Hemmila MR. Pneumonia in the surgical intensive
care unit: is every one preventable? Surgery 2011;150(4):665–72.
Section I—Background 55.e1

References brain injury patients: a case-control study. BMC Health Serv Res
2011;11:289.
1. Pronovost PJ, Angus DC, Dorman TM, et al. Physician staffing patterns and 27. Klompas MM, Platt RM. Ventilator-associated pneumonia: the wrong
clinical outcomes in critically ill patients: a systematic review. JAMA 2002; quality measure for benchmarking. Ann Intern Med 2007;147(11):803–5.
288(17): 2151–62. 28. Uckay I, Ahmed QA, Sax H, et al. Ventilator-associated pneumonia as a
2. Pronovost PJ, Miller MR, Dorman TM, et al. Developing and implementing quality indicator for patient safety? Clin Infect Dis 2008;46(4):557–63.
measures of quality of care in the intensive care unit. Curr Opin Crit Care 29. Eggimann P, Hugonnet S, Sax H, et al. Ventilator-associated pneumonia:
2001;7(4):297–303. caveats for benchmarking. Intensive Care Med 2003;29(11):2086–9.
3. Mallon WJ. Earnest Amory Codman: the end result of a life in medicine. 30. Mangino JE, Peyrani P, Ford KD, et al. Development and implementation
Philadelphia: Saunders; 2000. of a performance improvement project in adult intensive care units:
4. Deming WE. Out of the crisis. Cambridge, MA: MIT Press; 1986. overview of the Improving Medicine Through Pathway Assessment of
5. Donabedian A. The quality of care: how can it be assessed? JAMA 1988; Critical Therapy in Hospital-Acquired Pneumonia (IMPACT-HAP) study.
260(12):1743–8. Crit Care 2011;15(1):R38.
6. Sirio CA, Shepardson LB, Rotondi AJ, et al. Community-wide assessment of 31. Ramirez P, Bassi GL, Torres A. Measures to prevent nosocomial infections
intensive care outcomes using a physiologically based prognostic measure: during mechanical ventilation. Curr Opin Crit Care 2012;18(1):86–92.
implications for critical care delivery from Cleveland health quality choice. 32. Rello J, Lode H, Cornaglia G, et al. VAP care bundle contributors:
Chest 1999;115(3):793–801. a European care bundle for prevention of ventilator-associated pneumonia.
7. Glance LG, Osler TM, Dick AP. Rating the quality of intensive care units: is Intensive Care Med 2010;36(5):773–80.
it a function of the intensive care unit scoring system? Crit Care Med 2002; 33. Roberts N, Moule P. Chlorhexidine and tooth-brushing as prevention
30(9):1976–82. strategies in reducing ventilator-associated pneumonia rates. Nurs Crit Care
8. Pronovost PJ, Berenholtz SM, Ngo K, et al. Developing and pilot testing 2011;16(6):295–302.
quality indicators in the intensive care unit. J Crit Care 2003;18(3):145–55. 34. Labeau SO, Van de Vyver K, Brusselaers N, et al. Prevention of ventilator-
9. Rhodes A, Moreno RP, Azoulay E, et al. Prospectively defined indicators associated pneumonia with oral antiseptics: a systematic review and
to improve the safety and quality of care for critically ill patients: a report meta-analysis. Lancet Infect Dis 2011;11(11):845–54.
from the Task Force on Safety and Quality of the European Society of 35. Li Bassi G, Torres A. Ventilator-associated pneumonia: role of positioning.
Intensive Care Medicine (ESICM). Intensive Care Med 2012; Jan 26. Epub Curr Opin Crit Care 2011;17(1):57–63.
ahead of print. 36. Lansford T, Moncure M, Carlton E, et al. Efficacy of a pneumonia
10. Lilford R, Pronovost PJ. Using hospital mortality rates to judge hospital prevention protocol in the reduction of ventilator-associated pneumonia in
performance: a bad idea that just won’t go away. BMJ 2010;340:955–7. trauma patients. Surg Infect 2007;8(5):505–10.
11. Huang DT, Clermont G, Kong L, et al. Intensive care unit safety culture 37. Cocanour CS, Peninger M, Li T, et al. Decreasing ventilator-associated
and outcomes: a US multicenter study. Int J Qual Health Care 2010;22(3): pneumonia in a trauma ICU. J Trauma 2006;61(1):122–9.
151–61. 38. Bird D, Zambuto A, O’Donnell C, et al. Adherence to ventilator-associated
12. Sakr Y, Marques J, Mortsch S, et al. Is the SAPS II score valid in surgical pneumonia bundle and incidence of ventilator-associated pneumonia in
intensive care unit patients? J Eval Clin Pract 2010;Sep 22. Epub ahead of the surgical intensive care unit. Arch Surg 2010;145(5):465–70.
print. 39. Stone Jr ME, Snetman D, O’ Neill A, et al. Daily multidisciplinary rounds to
13. Poole D, Rossi C, Anghileri A, et al. External validation of the Simplified implement the ventilator bundle decreases ventilator-associated pneumonia
Acute Physiology Score (SAPS) 3 in a cohort of 28,357 patients from 147 in trauma patients: but does it affect outcome? Surg Infect 2011;12(5):
Italian intensive care units. Intensive Care Med 2009;35(11):1916–24. 373–8.
14. Niskanen M, Reinikainen M, Pettilä V. Case-mix-adjusted length of stay 40. Sundar KM, Nielsen D, Sperry P. Comparison of ventilator-associated
and mortality in 23 Finnish ICUs. Intensive Care Med 2009;35(6):1060–7. pneumonia (VAP) rates between different ICUs: implications of a zero VAP
15. Brinkman S, Bakhshi-Raiez F, Abu-Hanna A, et al. External validation of rate. J Crit Care 2011;Jul 5. Epub ahead of print.
Acute Physiology and Chronic Health Evaluation IV in Dutch intensive care 41. Kollef MH, Shapiro SD, Silver P, et al. A randomized, controlled trial of
units and comparison with Acute Physiology and Chronic Health protocol-directed versus physician-directed weaning from mechanical
Evaluation II and Simplified Acute Physiology Score II. J Crit Care 2011; ventilation. Crit Care Med 1997;25(4):567-74.
26(1):105.e11–18. 42. Dries DJ, McGonigal MD, Malian MS, et al. Protocol-driven ventilator
16. Moreno RP, Bauer P, Metnitz PG. Characterizing performance profiles of weaning reduces use of mechanical ventilation, rate of early reintubation,
ICUs. Curr Opin Crit Care 2010;16(5):477–81. and ventilator-associated pneumonia. J Trauma Inj Infect Crit Care
17. Jarman B, Aylin P: Death rates in England and Wales and the United States: 2004;56(5):943–52.
variation with age, sex, and race. BMJ 2004;329:1367. 43. Henneman EA, Dracup KA, Ganz T, et al. Effect of a collaborative weaning
18. Brinkman S, Abu-Hanna A, van der Veen A, et al. A comparison of the plan on patient outcome in the critical care setting. Crit Care Med
performance of a model based on administrative data and a model based 2001;29(2):297–303.
on clinical data: effect of severity of illness on standardized mortality ratios 44. Henneman EA, Dracup KA, Ganz T, et al. Using a collaborative weaning
of intensive care units. Crit Care Med 2012;40(2):373–8. plan to decrease duration of mechanical ventilation and length of stay in
19. Lilly CM, Zuckerman IH, Badawi O, et al. Benchmark data from more than the intensive care unit for patients receiving long-term ventilation. Am J
240,000 adults that reflect the current practice of critical care in the United Crit Care 2002;11(2):132–40.
States. Chest 2011;140(5):1232–42. 45. Thomas BW, Maxwell RA, Dart BW, et al. Errors in administrative-reported
20. Curtis JR, Patrick DL, Engelberg RA, et al. A measure of the quality of ventilator-associated pneumonia rates: are never events really so? Am Surg
dying and death: initial validation using after-death interviews with family 2011;77(8):998–1002.
members. J Pain Symptom Manage 2002;24(1):17–31. 46. Wahl WL, Zalewski C, Hemmila MR. Pneumonia in the surgical intensive
21. Glavan BJ, Engelberg RA, Downey L, et al. Using the medical record to care unit: is every one preventable? Surgery 2011;150(4):665–72.
evaluate the quality of end-of-life care in the intensive care unit. Crit Care 47. Bouadma L, Deslandes E, Lolom I, et al. Long-term impact of a
Med 2008;36(4):1138–46. multifaceted prevention program on ventilator-associated pneumonia in a
22. Mularski RA, Heine CE, Osborne ML, et al. Quality of dying in the ICU: medical intensive care unit. Clin Infect Dis 2010;51(10):1115–22.
ratings by family members. Chest 2005;128(1):280–7. 48. Klompas M. Is a ventilator-associated pneumonia rate of zero really
23. Claessen SJ, Francke AL, Belarbi HE, et al. A new set of quality indicators possible? Curr Opin Infect Dis 2012;Jan 13. Epub ahead of print.
for palliative care: process and results of the development trajectory. J Pain 49. VanGilder C, Amlung S, Harrison P, et al. Results of the 2008-2009
Symptom Manage. 2011;42(2):169–82. International Pressure Ulcer Prevalence Survey and a 3-year, acute care,
24. Ghafoor VL, Silus LS. Developing policy, standard orders, and quality- unit-specific analysis. Ostomy Wound Manage 2009;55(11):39–45.
assurance monitoring for palliative sedation therapy. Am J Health Syst 50. Paul JA, Keller JW, van Ramshorst B, et al. Pressure ulcers in intensive care
Pharm 2011;68(6):523–7. patients: a review of risks and prevention. Intensive Care Med 2002;28:
25. Nelson JE, Bassett R, Boss RD, et al. Improve Palliative Care in the Intensive 1379–88.
Care Unit Project. Models for structuring a clinical initiative to enhance 51. de Laat EH, Pickkers P, Schoonhoven L, et al. Guideline implementation
palliative care in the intensive care unit: a report from the IPAL-ICU results in a decrease of pressure ulcer incidence in critically ill patients. Crit
project (Improving Palliative Care in the ICU). Crit Care Med 2010;38(9): Care Med 2007;35(3):815–20.
1765–72. 52. Sihler KC, Chenoweth C, Zalewski C, et al. Catheter-related vs. catheter-
26. Yang CC, Shih NC, Chang WC, et al. Long-term medical utilization associated blood stream infections in the intensive care unit: incidence,
following ventilator-associated pneumonia in acute stroke and traumatic microbiology, and implications. Surg Infect 2010;11(6):529–34.
55.e2 Section I—Background

53. Taylor GD, McKenzie M, Kirkland T. The impact of health care 79. Cook CB, Kongable GL, Potter DJ, et al. Inpatient glucose control: a
restructuring on nosocomically acquired blood stream infectious. Can J glycemic survey of 126 U.S. hospitals. J Hosp Med 2009;4(9):E7–E14.
Infect Dis 2000;11(1):29–32. 80. Sidorenkov G, Haaijer-Ruskamp FM, de Zeeuw D, et al. Review: relation
54. Douglas JE, Schuerer JEZ, Thomas J, et al. Effect of chlorhexidine/silver between quality-of-care indicators for diabetes and patient outcomes: a
sulfadiazine-impregnated central venous catheters in an intensive care unit systematic literature review. Med Care Res Rev 2011;68(3):263–89. Epub
with a low blood stream infection rate after implementation of an 2011, May 2.
educational program: a before–after trial. Surg Infect 2007;8(4):445–54. 81. Duning T, van den Heuvel I, Dickmann A, et al. Hypoglycemia aggravates
55. Rupp ME, Lisco SJ, Lipsett PA, et al. Effect of a second-generation venous critical illness-induced neurocognitive dysfunction. Diabetes Care
catheter impregnated with chlorhexidine and silver sulfadiazine on central 2010;33(3):639–44. Epub 2009, Dec 23.
catheter-related infections: a randomized, controlled trial. Ann Intern Med 82. Hermanides J, Vriesendorp TM, Bosman RJ, et al. Glucose variability is
2005;143(8):570–80. associated with intensive care unit mortality. Crit Care Med 2010;38(3):
56. Darouiche RO, Raad II, Heard SO, et al. A comparison of two 838–42.
antimicrobial-impregnated central venous catheters. N Engl J Med 1999; 83. Meyfroidt G, Wouters P, De Becker W, et al. Impact of a computer-
340(1):1–8. generated alert system on the quality of tight glycemic control. Intensive
57. Cherry-Bukowiec JR, Denchev K, Dickinson S, et al. Prevention of Care Med 2011;37(7):1151–7. Epub 2011, Mar 3.
catheter-related blood stream infection: back to basics? Surg Infect 2011; 84. Angus DC, Kelley MA, Schmitz RJ, et al. Current and projected workforce
12(1):27–32. Epub 2010, Dec 20. requirements for care of the critically ill and patients with pulmonary
58. Gunst M, Matsushima K, Vanek S, et al. Peripherally inserted central disease: can we meet the requirements of an aging population? JAMA 2000;
catheters may lower the incidence of catheter-related blood stream 284(21):2762–70.
infections in patients in surgical intensive care units. Surg Infect 85. Angus DC, Shorr AF, White A, et al. Critical care delivery in the United
2011;12(4):279–82. Epub 2010, Jul 14. States: distribution of services and compliance with leapfrog
59. Cherry RA, West CE, Hamilton MC, et al. Reduction of central venous recommendations. Crit Care Med 2006;34(4):1016–24
catheter associated blood stream infections following implementation of a 86. Carlson RW, Weiland DE, Srivathsan K. Does a full-time, 24-hour
resident oversight and credentialing policy. Patient Saf Surg 2011;5(1):15. intensivist improve care and efficiency? Crit Care Clin 1996;12(3):525–51.
60. Seddon ME, Hocking CJ, Mead P, et al. Aiming for zero: decreasing central 87. Burchardi H, Moerer O. Twenty-four hour presence of physicians in the
line associated bacteraemia in the intensive care unit. N Z Med J 2011; ICU. Crit Care 2001;5(3):131–7.
124(1339):9–21. 88. Arabi YM, Alshimemeri AM, Taher SM. Weekend and weeknight
61. Render ML, Hasselbeck R, Freyberg RW, et al. Reduction of central line admissions have the same outcome of weekday admissions to an intensive
infections in Veterans Administration intensive care units: an observational care unit with onsite intensivist coverage. Crit Care Med 2006;34(3):605–11.
cohort using a central infrastructure to support learning and improvement. 89. Varelas P, Varelas PN, Schultz L, et al. The impact of a neuro-intensivist on
BMJ Qual Saf 2011;20(8):725–32. Epub 2011, Apr 2. patients with stroke admitted to a neurosciences intensive care unit.
62. Finney SJ, Zekveld C, Elia A, et al. Glucose control and mortality in Neurocrit Care 2008;9(3):293–9.
critically ill patients. JAMA 2003;290(15):2041–7. 90. Ghorra SMD, Reinert SE, Cioffi W, et al. Analysis of the effect of conversion
63. Gale SC, Sicoutris CP, Reilly PM, et al. Poor glycemic control is associated from open to closed surgical intensive care unit. Ann Surg 1999;229(2):
with increased mortality in critically ill trauma patients. Am Surg 2007; 163–71.
73(5):454–60. 91. Hanson III CW, Deutschman CS, Anderson III HL, et al. Effects of an
64. Shulman R, Finney SJ, O’Sullivan C, et al. Tight glycaemic control: a organized critical care service on outcomes and resource utilization: a
prospective observational study of a computerised decision-supported cohort study. Crit Care Med 1999;27(2):270–4.
intensive insulin therapy protocol. Crit Care 2007;11(4):R75. 92. Nathens AB, Rivara FP, MacKenzie EJ, et al. The impact of an intensivist-
65. Meynaar IA, Dawson L, Tangkau PL, et al. Introduction and evaluation of model ICU on trauma-related mortality. Ann Surg 2006;244(4):545–54.
a computerised insulin protocol. Intensive Care Med 2007;33(4):591–6. 93. Pronovost PJ, Jenckes MW, Dorman TM, et al. Organizational
66. Quinn JA, Berghoff JL, Colombo CS, et al. A practical approach to characteristics of intensive care units related to outcomes of abdominal
hyperglycemia management in the intensive care unit: evaluation of an aortic surgery. JAMA 1999;281(14):1310–17.
intensive insulin infusion protocol. Pharmacotherapy 2006;26(10):1410–20. 94. Dimick JB, Pronovost PJ, Heitmiller RF, et al. Intensive care unit physician
67. Krinsley JS, Jones RL. Cost analysis of intensive glycemic control in staffing is associated with decreased length of stay, hospital cost, and
critically ill adult patients. Chest 2006;129(3):644–50. complications after esophageal resection. Crit Care Med 2001;29(4):
68. Schmeltz LR, DeSantis AJ, Thiyagarajan V, et al. Reduction of surgical 753–8.
mortality and morbidity in diabetic patients undergoing cardiac surgery 95. Treggiari MM, Martin DP, Yanez ND, et al. Effect of intensive care unit
with a combined intravenous and subcutaneous insulin glucose organizational model and structure on outcomes in patients with acute
management strategy. Diabetes Care 2007;30(4):823–8. lung injury. Am J Respir Crit Care Med 2007;176(7):685–90.
69. Van den Berghe G, Wilmer A, Hermans G, et al. Intensive insulin therapy in 96. Chittawatanarat K, Pamorsinlapathum T. The impact of closed ICU model
the medical ICU. N Engl J Med 2006;354(5):449–61. on mortality in general surgical intensive care unit. J Med Assoc Thai
70. Krinsley JS. Effect of an intensive glucose management protocol on the 2009;92(12):1627–34.
mortality of critically ill adult patients. Mayo Clin Proc 2004;79(8): 97. Treggiari MM, Martin DP, Yanez ND, et al. Effect of intensive care unit
992–1000. organizational model and structure on outcomes in patients with acute
71. van der Voort PHJ, Feenstra RA, Bakker AJ, et al. Intravenous glucose lung injury. Am J Respir Crit Care Med 2007;176(7):685–90. Epub 2007,
intake independently related to intensive care unit and hospital mortality: Jun 7.
an argument for glucose toxicity in critically ill patients. Clin Endocrinol 98. Gajic O, Afessa B, Hanson AC, et al. Effect of 24-hour mandatory versus
(Oxf) 2006;64(2):141-5. on-demand critical care specialist presence on quality of care and family
72. Anger KE. Barriers to glucose control in the intensive care unit. and provider satisfaction in the intensive care unit of a teaching hospital.
Pharmacotherapy 2006;26(2):214–28. Crit Care Med. 2008;36(1):36–44.
73. Wiener RS, Wiener DC, Larson RJ. Benefits and risks of tight glucose 99. Kahn JM, Brake H, Steinberg KP. Intensivist physician staffing and the
control in critically ill adults: a meta-analysis. JAMA 2008;300(8):933–44. process of care in academic medical centres. Qual Saf Health Care
74. NICE-SUGAR Study Investigators, Finfer S, Chittock DR, et al. Intensive 2007;16(5):329–33.
versus conventional glucose control in critically ill patients. N Engl J Med 100. Nathens AB, Maier RV, Jurkovich GJ, et al. The delivery of critical care
2009;360(13):1283–97. Epub 2009, Mar 24. services in us trauma centers: is the standard being met? J Trauma
75. Egi M, Finfer S, Bellomo R. Glycemic control in the ICU. Chest 2011; 2006;60(4):773–84.
140(1):212–20. 101. Byrnes MC, Schuerer DJ, Schallom ME, et al. Implementation of a
76. Kutcher ME, Pepper MB, Morabito D, et al. Finding the sweet spot: mandatory checklist of protocols and objectives improves compliance with
identification of optimal glucose levels in critically injured patients. a wide range of evidence-based intensive care unit practices. Crit Care Med
J Trauma 2011;71(5):1108–14. 2009;37(10):2775–81.
77. Oddo M, Schmidt JM, Carrera C, et al. Impact of tight glycemic control on 102. Scales DC, Dainty K, Hales B, et al A multifaceted intervention for quality
cerebral glucose metabolism after severe brain injury: a microdialysis study. improvement in a network of intensive care units: a cluster randomized
Crit Care Med 2008;36:3233–8. trial. JAMA 2011;305(4):363–72. Epub 2011, Jan 19.
78. Anderson M, Zito D, Kongable G. Benchmarking glucose results through 103. Schramm GE, Kashyap R, Mullon JJ, et al. Septic shock: a multidisciplinary
automation: the 2009 remote automated laboratory system report. response team and weekly feedback to clinicians improve the process of
J Diabetes Sci Technol 2010;4(6):1507–13. care and mortality. Crit Care Med 2011;39(2):252–8.
Section I—Background 55.e3

104. Daniels R. Surviving the first hours in sepsis: getting the basics right (an 131. Clarke CM, Persaud DD. Leading clinical handover improvement: a change
intensivist’s perspective). J Antimicrob Chemother 2011;66(Suppl 2): strategy to implement best practices in the acute care setting. J Patient Saf
ii11–23. 2011;7(1):11–18.
105. Vincent JL. We should abandon randomized controlled trials in the 132. Manser T, Foster S, Gisin S, et al. Assessing the quality of patient handoffs
intensive care unit. Crit Care Med 2010;38(10 Suppl):S534–8. at care transitions. Qual Saf Health Care 2010;19(6):e44.
106. Ospina-Tascon GA, Buchele GL, Vincent JL: Multicenter, randomized, 133. Manthous CA, Hollingshead AB. Team science and critical care. Am J
controlled trials evaluating mortality in intensive care: doomed to fail? Crit Respir Crit Care Med 2011;184(1):17–25. Epub 2011, Mar 18.
Care Med 2008;36:1311–22. 134. Oglesby KJ, Durham L, Welch J, et al. ‘Score to door time’, a benchmarking
107. Walker S, Brett S. Oiling the wheels of intensive care to reduce “machine tool for rapid response systems: a pilot multi-centre service evaluation. Crit
friction”: the best way to improve outcomes? Crit Care Med 2010;38(10 Care 2011;15(4):R180.
Suppl):S642–8. 135. Hunziker S, Johansson AC, Tschan F, et al. Teamwork and leadership in
108. Tobin MJ. Counterpoint: evidence-based medicine lacks a sound scientific cardiopulmonary resuscitation. J Am Coll Cardiol 2011;57(24):2381–8.
base. Chest 2008;133(5):1071–4; discussion 1074–7. 136. Rosenfeld BA, Dorman TM, Breslow MJ, et al. Intensive care unit
109. Wendon J. Critical care “normality”: individualized versus protocolized telemedicine: alternate paradigm for providing continuous intensivist care.
care. Crit Care Med 2010;38(10 Suppl):S590–9. Crit Care Med 2000;28(12):3925–31.
110. Prasad M, Holmboe ES, Lipner RS, et al. Clinical protocols and trainee 137. Breslow MJ, Rosenfeld BA, Doerfler M, et al. Effect of a multiple-site
knowledge about mechanical ventilation. JAMA 2011;306(9):935–41. intensive care unit telemedicine program on clinical and economic
111. Kahol K, Vankipuram M, Patel VL, et al. Deviations from protocol in a outcomes: an alternative paradigm for intensivist staffing. Crit Care Med
complex trauma environment: errors or innovations? J Biomed Inform 2004;32(1):31–8.
2011;44(3):425–31. Epub 2011, Apr 13. 138. Breslow MJ. Remote ICU care programs: current status. J Crit Care
112. Moreno R, Rhodes A. Evidence should not be viewed in isolation. Crit Care 2007;22(1):66–76.
Med 2010;38(10 Suppl):S528–33. 139. Pastores SM, O’Connor MF, Kleinpell RM, et al. The Accreditation Council
113. Gasperino J. The Leapfrog initiative for intensive care unit physician for Graduate Medical Education resident duty hour new standards: history,
staffing and its impact on intensive care unit performance: a narrative changes, and impact on staffing of intensive care units. Crit Care Med
review. Health Policy 2011;102(2-3):223–8. Epub 2011, Mar 24. 2011;39(11):2540–9.
114. Kahn JM, Matthews FA, Angus DC, et al. Barriers to implementing the 140. Cummings J, Krsek C, Vermoch K, et al. Intensive care unit telemedicine:
Leapfrog group recommendations for intensivist physician staffing: a survey review and consensus recommendations. Am J Med Qual 2007;22(4):
of intensive care unit directors. J Crit Care 2007;22(2): 97–103. 239–50.
115. Pronovost PJ, Thompson DA, Holzmueller CG, et al. Impact of the 141. Moreno RP, Rhodes A, Donchin Y. Patient safety in intensive care medicine:
Leapfrog group’s intensive care unit physician staffing standard. J Crit Care the Declaration of Vienna. Intensive Care Med 2009;35:1667–72.
2007;22(2):89–96.e24. 142. Whiteman R, Gould L, Oczkowski W, et al. Using a quality improvement
116. Pronovost PJ, Needham DM, Waters HR, et al. Intensive care unit physician process to create measurable improvement in care delivery for acute stroke.
staffing: Financial modeling of the Leapfrog standard. Crit Care Med 2004; Healthc Quart 2011;14(3):75–9.
32(6):1247–53. 143. Wiedmann S, Norrving B, Nowe T, et al. Variations in quality indicators of
117. Kozar RA, Shackford SR, Cocanour CS. Challenges to the care of the acute stroke care in 6 European countries: the European Implementation
critically ill: novel staffing paradigms. J Trauma 2008;64(2):366–73. Score (EIS) collaboration. Stroke 2012;43(2):458–63. Epub 2011, Dec 8.
118. Pronovost PJ, Waters HP, Dorman TM. Impact of critical care physician 144. Woodhouse D, Berg M, van der Putten J, et al. Will benchmarking ICUs
workforce for intensive care unit physician staffing. Curr Opin Crit Care improve outcome? Curr Opin Crit Care 2009;15(5):450–5. Review.
2001;7(6):456–9. 145. Kardooni S, Haut ER, Chang DC, et al. Hazards of benchmarking
119. Napolitano LM, Fulda GJ, Davis KA, et al. Challenging issues in surgical complications with the National Trauma Data Bank: numerators in search
critical care, trauma, and acute care surgery: a report from the critical care of denominators. J Trauma. 2008;64(2):273–7.
committee of the American Association for the Surgery of Trauma. 146. Hernandez AF, Fonarow GC, Liang L, et al. The need for multiple measures
J Trauma 2010;69(6):1619–33. of hospital quality: results from the Get with the Guidelines-Heart Failure
120. Gracias VH, Sicoutris CP, Stawicki SP, et al. Critical care nurse practitioners Registry of the American Heart Association. Circulation 2011;124(6):
improve compliance with clinical practice guidelines in “semiclosed” 712–19. Epub 2011, Jul 25.
surgical intensive care unit. J Nurs Care Qual 2008;23(4):338–44. 147. Sciacovelli L, Sonntag O, Padoan A, et al. Monitoring quality indicators in
121. Lovell MA, Mudaliar MY, Klineberg PL. Intrahospital transport of critically laboratory medicine does not automatically result in quality improvement.
ill patients: complications and difficulties. Anaesth Intensive Care 2001; Clin Chem Lab Med 2011;Dec 13. Epub ahead of print.
29(4):400–5. 148. Ketelaar NA, Faber MJ, Flottorp S, et al. Public release of performance data
122. Warren J, Fromm RE Jr, Orr RA, et al. Guidelines for the inter- and in changing the behaviour of healthcare consumers, professionals or
intrahospital transport of critically ill patients. Crit Care Med organisations. Cochrane Database Syst Rev 2011;11:CD004538.
2004;32(1):256–62. 149. Ryan AM, Blustein J. The effect of the MassHealth hospital pay-for-
123. Bercault N, Wolf M, Runge I, et al. Intrahospital transport of critically ill performance program on quality. Health Serv Res 2011;46(3):712–28.
ventilated patients: a risk factor for ventilator-associated pneumonia—a doi: 10.1111/j.1475–6773.2010.01224.x. Epub 2011, Jan 6.
matched cohort study. Crit Care Med 2005;33(11):2471–8. 150. Rademakers J, Delnoij D, de Boer D. Structure, process or outcome: which
124. Swanson E, Mascitelli J, Stiefel M, et al. The effect of patient transport on contributes most to patients’ overall assessment of healthcare quality? BMJ
brain oxygen in comatose patients. Neurosurgery 2010;66:925–32. Qual Saf 2011;20(4):326–31. Epub 2011, Feb 21.
125. Kue R, Brown P, Ness C, et al. Adverse clinical events during intrahospital 151. Metersky ML. Should management of pneumonia be an indicator of
transport by a specialized team: a preliminary report. Am J Crit Care quality of care? Clin Chest Med 2011;32(3):575–89.
2011;20(2):153–61; quiz 162. 152. Breuer S, Arquilla V. Clinical documentation improvement: focus on
126. Wiegersma JS, Droogh JM, Zijlstra JG, et al. Quality of interhospital quality. Healthc Financ Manage 2011;65(8):84–6, 88, 90.
transport of the critically ill: impact of a mobile intensive care unit 153. Zegers M, de Bruijne MC, Spreeuwenberg P, et al. Quality of patient record
with a specialized retrieval team. Crit Care 2011;15(1):R75. Epub 2011, keeping: an indicator of the quality of care? BMJ Qual Saf 2011;20(4):
Feb 28. 314–18. Epub 2011, Feb 8.
127. Lyons MN, Standley TD, Gupta AK. Quality improvement of doctors’ 154. Goeschel CA, Wachter RM, Pronovost PJ. Responsibility for quality
shift-change handover in neuro-critical care. Qual Saf Health Care improvement and patient safety: hospital board and medical staff
2010;19(6):e62. Epub 2010, Apr 27. leadership challenges. Chest 2010;138(1):171–8.
128. Joy BF, Elliott E, Hardy C, et al. Standardized multidisciplinary protocol 155. Gisvold SE, Fasting S. How do we know that we are doing a good job—can
improves handover of cardiac surgery patients to the intensive care unit. we measure the quality of our work? Best Pract Res Clin Anaesthesiol
Pediatr Crit Care Med 2011;12(3):304–8. 2011;25(2):109–22.
129. Cleland JA, Ross S, Miller SC, et al. “There is a chain of Chinese 156. Pinto A, Burnett S, Benn J, et al. Improving reliability of clinical care
whispers …”: empirical data support the call to formally teach practices for ventilated patients in the context of a patient safety
handover to prequalification doctors. Qual Saf Health Care 2009;18(4): improvement initiative. J Eval Clin Pract 2011;17(1):180–7.
267–71. 157. Pronovost PJ, Cardo DM, Goeschel CA, et al. A research framework for
130. Pickering BW, Hurley K, Marsh B. Identification of patient information reducing preventable patient harm. Clin Infect Dis 2011;52(4):507–13.
corruption in the intensive care unit: using a scoring tool to direct quality 158. Wolf JA. Sustaining high performance: dynamic balancing in an otherwise
improvements in handover. Crit Care Med 2009;37(11):2905–12. unbalanced system. Adv Health Care Manage 2011;10:367–84.
I
Chapter
7  

Brain Monitoring Issues


in Pediatrics
Anthony A. Figaji

used. However, the GCS as a monitoring tool is limited by


Introduction prehospital and ICU sedation16 and because children with
Neuromonitoring in the intensive care unit (ICU) is under- higher GCS may deteriorate suddenly.17 Methods of assessing
taken for several acute neurologic conditions in children. initial clinical severity, such as the GCS and acute illness
Traumatic brain injury (TBI) constitutes the most common scores, do not predict which patients are at risk for all types
indication for neuromonitoring because injury is the leading of secondary injury.18
cause of death in children over the age of 1 year,1,2 and those
who sustain TBI have the highest chance of dying or being
permanently disabled.3,4 Early treatment to prevent second- Prehospital Hypotension
ary insults may avoid as much as 30% of pediatric TBI
deaths.5 Therefore, monitoring for these potential secondary
and Hypoxia
insults is the foundation of management for children. Polytrauma is common in children with severe TBI,19,20 and
Importantly, experimental studies demonstrate that neuro- the addition of extracranial injury increases the risk of hypo-
monitoring findings, including intracranial pressure (ICP), tension and hypoxia.21 Hypoxia and hypotension occur in
brain oxygen, and microdialysis correlate with neuropathy 31% to 68% of children who sustain severe TBI and are associ-
of animal models of pediatric TBI.6 The aim of this chapter ated with poor outcome, especially hypotension.12,21 Although
is not to provide a comprehensive summary of monitoring the diagnosis of hypoxia in children is standard, and consis-
options, but rather to highlight some differences between tent with that in adults, the definition of hypotension varies.
adults and children with respect to neuromonitoring. In Systolic blood pressure (SBP) is commonly used to define
addition, the reader is reminded that neuromonitoring may hypotension, either as SBP less than 90 mm Hg or less than
be valuable in children and adolescents with nontraumatic the fifth percentile for age,22,23 but these approaches have limi-
coma (e.g., meningitis, hepatic encephalopathy, stroke, near tations. First, mean arterial pressure (MAP) is more appropri-
drowning, and hypoxic-ischemic injury).7-10 ate for determining likely cerebral perfusion pressure (CPP).
Second, height influences the normal range of blood pressure
(BP) in children, but is rarely considered. Height is related to
Children and Adults Are Different BP independently of age, so there are significant differences
Potential secondary insults in children are similar to those in across height-for-age percentile blood pressure ranges.24,25
adults; however, children have different physical dimensions
and physiologic responses. They are rapidly growing and
developing, and normal ranges and thresholds for treatment
Cerebral Blood Flow
not only are different from adults, but also vary between age There are important differences in cerebral blood flow (CBF)
groups. For these reasons, and because of the smaller research values between adults and children in health and disease.
base, the best evidence-based recommendations for specific Specifically, the role of hyperemia as the predominant cause
aspects of clinical management in pediatric TBI are still mostly of diffuse swelling observed after TBI in children has been
set at the level of options.11 debated.26-28 When CBF values in children are compared with
mean values for age, the incidence of true hyperemia appears
to be much lower than previously believed.28 CBF values tend
Glasgow Coma Scale to increase from the lowest values at birth to a peak at age
The Glasgow Coma Scale (GCS) is the standard method of 3 to 5 years and then decrease to adult levels.28,29 Therefore,
assessing and monitoring the level of consciousness in pedi- when interpreting high CBF values in children with TBI (high
atric TBI.12,13 In children the utility of the GCS is limited in and low), age-specific ranges must be considered. Low CBF is
younger patients; therefore, modifications of the GCS such as common early after injury and is associated with worse
the Pediatric Coma Scale14 and the Children’s Coma Scale15 are outcome; however, data on practical CBF monitoring for
56 © Copyright 2013 Elsevier Inc. All rights reserved.
Section I—Background 57

children in critical care are limited.30,31 Ideally, absolute CBF of the patient, the absolute ICP value, and the underlying
values should be interpreted with metabolic demand and cause of the increased ICP. Although it is important to treat
flow-metabolism coupling. elevated ICP, additional data are required to guide sound
clinical decisions. Therefore, a multimodal approach has par-
Intracranial Pressure: Indications, ticular value.
Methods, and Thresholds
Indications for intracranial pressure (ICP) monitoring in chil-
Cerebral Perfusion Pressure
dren vary among different institutions.3,32 Current recommen- Low CPP is known to be associated with poor outcome;
dations support ICP monitoring for children with severe TBI, however, this may be a surrogate marker for high ICP. However,
regardless of whether fontanels and sutures are open.11 Still, some argue that absolute ICP values are of lesser importance
in the absence of randomized controlled trials, some question if CPP is adequate. Therefore, as in adults, the merits of ICP-
whether ICP monitoring is beneficial in pediatric TBI33; the targeted treatment versus CPP-targeted treatment are also
reasons for this are complex.34 In the United Kingdom only debated in children.41 Higher CPP targets in adult TBI have
about 60% of children presenting with severe TBI undergo been recommended42 but may increase the risk of acute respi-
ICP monitoring, with large among-center variation,32 and ratory distress syndrome,43 and if autoregulation is impaired
many patients receive ICP-targeted therapy without an ICP it may increase ICP44 and tissue edema.45,46 The Lund approach
monitor in situ. But adherence to published guidelines appears using brain volume regulation and microdialysis monitoring
to be increasing.35 Indications for ICP monitoring in children to accept lower CPP levels also has been described in chil-
are based on less evidence than in adults because of the dren47 but is not without controversy and risks.48
paucity of studies.11 Because computed tomography (CT) It has not been established that active modulation of CPP,
signs cannot reliably exclude intracranial hypertension,36 it is beyond avoiding hypotension, benefits pediatric TBI patients,
recommended to monitor all patients with severe TBI who are so there is little agreement on what constitutes an appropriate
not likely to be extubated within 12 hours. CPP target for age. Chambers et al.49 recommended CPP
Standard methods for ICP monitoring in adults are also thresholds of 48, 54, and 58 for age-groups 2 through 6,
used in children; intraparenchymal devices are most com- 7 through 10, and 11 through 15 years, respectively, and
monly used and complications are rare.37 A ventriculostomy 45 mm Hg overall.50 Prabhakaran et al.41 examined ICP- and
can be useful in some patients38 but is less commonly used,32 CPP-targeted approaches but reached no definitive conclu-
most likely because of difficulties in cannulating small ven- sion. Vavilala et al.51 found that all children who had an admis-
tricles in the typical child with diffuse swelling. For practical sion SBP less than 90 mm Hg had a poor outcome, but that
purposes the bolt system for intracranial catheters in pediat- the 75th percentile of SBP for age was a better predictor for
rics is usually suitable even in younger children, but the very outcome. Jones et al.52 and Hackbarth et al.20 recommended
young may require a formal burr hole and subgaleal tunneling CPP greater than 50 to 60 mm Hg, but Downard et al.53 found
if the skull is particularly thin. that CPP greater than 50 mm Hg conferred no survival benefit
In children the definition of elevated ICP is less clear and argued that benefits of CPP elevation may simply be a
because of changing physiologic norms with age, the differ- proxy for the avoidance of hypotension.
ent pressure-volume relationship of the brain in children,38 None of the these studies considered height for age in the
and uncertainty about whether the autoregulatory curve in evaluation of MAP or CPP. Furthermore, all recommenda-
children is similar to that of adults.39 Lower thresholds than tions for specific targets may not be appropriate given
those recommended for treatment in adults are often used that impaired cerebral autoregulation is common in children
for younger children, but there are no reliable data to support with severe TBI.39 Nevertheless, current recommendations
this. The treatment threshold of 20 mm Hg is recommended for pediatric TBI support the avoidance of CPP less than
at the level of an option for children.11 If the anatomic char- 40 mm Hg in all children and targeting CPP greater than
acteristics of the brain that determine the point at which a 50 mm Hg as an option. These have no clear recommenda-
pediatric brain undergoes herniation are similar to that of an tions for an age-related approach.11 It is likely that a single CPP
adult brain, and the autoregulatory curve is similar,39 then value is not appropriate for all patients given differences in age
targeting a similar threshold appears sensible, so long as an and metabolic needs, the tissue pressure effect of increased
adequate CPP is maintained. This ICP threshold for treat- ICP (rather than just the CPP-lowering effect thereof), the
ment is supported by the findings in children of an inverse status of pressure autoregulation, and the variable coupling
relationship between CBF and ICP greater than 20 mm Hg27 between metabolism and CBF.40,54,55
and significant changes in the pressure-volume index when
ICP is greater than 20 mm Hg.38 However, the relationship
between ICP and CBF or oxygenation of the brain is com-
Brain Tissue Oxygen
plex. Several pathophysiologic mechanisms may underlie Several studies have examined the relationship between brain
an increase in ICP in individual children, including classic tissue oxygen tension (PbtO2) and outcome in adults,56 but
cerebral edema, seizures, impaired pressure autoregulation, few in children.57-61 Narotam et al.57 (n = 16) found that no
hyperemia (uncoupled flow-metabolism coupling), increased children with normal initial PbtO2 died. Stiefel et al.58 (n = 6)
CO2, and so on. Therefore, the precise relationship between found PbtO2 was significantly lower if ICP was greater than
ICP and perfusion of the brain depends not only on the 20 mm Hg and CPP was less than 40 mm Hg. Figaji et al.61
effect of ICP on perfusion, but also on the reason for the (n = 52) found that low PbtO2 was an independent predictor
increased ICP.40 In the author’s experience, ICP affects oxy- of poor outcome. Poor outcome was linked to the depth and
genation of the brain at different levels, depending on the age duration of brain hypoxia, especially when PbtO2 was less than
58 Section I—Background

60 Jugular Bulb Venous


50 Oxygen Saturation
40
Intracranial pressure
30 There are fewer reports of SjvO2 monitoring in children com-
20 pared with adults.80,81 The technique in children has been
10 described.82 Perez et al.81 found that poor outcome in pediatric
0
TBI was associated with episodes of SjvO2 less than 55% and
35
30 pathologic arteriovenous difference in lactate. The technical
25 limitations of SjvO2 have not been specifically described in
20 Brain tissue oxygen children, but are probably similar to those in adults.83,84
15
10
0 Transcranial Doppler
Fig. 7.1  Graph of continuous monitoring of intracranial pressure (ICP) Transcranial Doppler (TCD) monitoring has been used as a
and brain tissue oxygen tension (PbtO2) in a 7-year-old child, recorded noninvasive monitoring tool in pediatric TBI, hydrocephalus,
over a 3-hour period, showing dynamic changes in ICP and its effect on and meningitis.85-87 Insonation of the basal vessels is usually
PbtO2. easier in children because of the thinner temporal bone. The
optimal depth of insonation of the middle cerebral artery
(MCA), is shorter in children than in adults and depends on
age and other physical factors such as scalp swelling after
10 mm Hg. In addition, episodes of critical brain hypoxia trauma. Normal values for TCD-derived flow velocity (FV) in
(PbtO2 <10 mm Hg) occurred in almost one third of patients the MCA (FVMCA) depend on age. FVMCA is approximately
despite recommended treatment targets being met for ICP, 24 cm/second in newborns, increases with age to peak at
CPP, arterial oxygenation, and hemoglobin concentration.59 97 cm per second at 6 to 9 years old, and then decreases to adult
These episodes appear to be not only outcome predictors values of about 50 cm per second.39 The reported age for peak
but also a manifestation of ongoing or secondary injury.62 mean FVMCA in children is slightly higher than the age reported
In children, low PbtO2 is associated with poor outcome, is a for peak CBF.28,29 There also appear to be slight gender differ-
manifestation of secondary injury, and responds to global ences in anterior and posterior circulation FV in children.88
changes in blood pressure, ICP (Fig. 7.1), and systemic The TCD-derived pulsatility index (PI) may be useful for
oxygenation.18,54,55,61,63-66 the noninvasive estimation of ICP and CPP in adults.89,90
Although this is not as well defined in children, the PI may be
associated with outcome after pediatric TBI91,92 and may be
Near-Infrared Spectroscopy useful as a guide in treating hydrocephalus.93 The Lindegaard
Theoretically, near-infrared spectroscopy (NIRS) may be index may be helpful in monitoring vasospasm in adult TBI.94
more promising in young children and neonates67 in whom Although vasospasm appears to be uncommon after pediatric
transillumination is easier. Although many report the useful- severe TBI,87,95 monitoring for vasospasm may be useful for
ness of NIRS in adults and children when there is no subarachnoid hemorrhage or meningitis.85
primary brain injury, as in carotid endarterectomy and pedi-
atric cardiac anesthesia,68,69 studies of its utility in the pedi-
atric TBI population are limited.66 Although some suggest
Pressure Autoregulation
promising results,70 technical factors that interfere with Pressure autoregulation (PAR) may be impaired in about 40%
signal reliability, especially in head trauma, may limit the of children with severe TBI.39,96 Although this has implications
utility of the method with current technology. Reduced cor- for clinical management, PAR is not commonly measured in
relation between NIRS values and other markers of oxygen- children with TBI. If PAR is impaired, maintenance of ade-
ation, such as jugular venous oxygen saturation (SjvO2) and quate CBF may require increased CPP, but higher CPP may
PbtO2, has been reported,71-73 and normal NIRS values have also increase cerebral blood volume and ICP.44 Impaired PAR
been found with complete ischemia.74 However, the develop- is associated with poor outcome in adults and children.97,98
ment of newer technologies may reduce these technical limi- Testing of PAR has been well described99 and bedside methods
tations. NIRS may be used as a noninvasive tool for have been validated.100,101 TCD-based methods are the most
measuring physiologic phenomena, such as autoregulation common bedside techniques used in children. The strength of
and the effect of ICP.75,76 PAR in children may vary between the two hemispheres and
may change over time.102,103 Continuous measurement of the
strength of PAR over time104 has also been described in chil-
Microdialysis dren.105 The status of PAR has profound consequences for the
Very few cerebral microdialysis studies have been performed effect of BP changes on ICP and perfusion of the brain in
in children,77-79 but these have highlighted possible patho- pediatric TBI.54 Ideally, the status of PAR should be known
physiologic differences between adult and pediatric TBI. In when making decisions about what BP target is optimal in
general, the principles are expected to be largely similar in individual patients. Important issues that still need to be clari-
children, as would be the limitations. The expense, infrastruc- fied in children include whether the autoregulation curve is
ture required, intermittent sampling technique, and lack of an similar to that of adults39 and whether information from
“early warning system” have limited the more widespread use testing PAR leads to therapeutic decisions that benefit the
of microdialysis in pediatric ICUs. patient.
Section I—Background 59

modifiable factor that causes secondary injury or is merely a


Computed Tomography Scanning marker of injury severity remains uncertain. Furthermore,
Head CT is used for diagnosis, to estimate prognosis, to strict control of glucose may significantly reduce cerebral
monitor progress, and as a guide to treatment. Unlike adult glucose, increase markers of tissue injury, and worsen
TBI, diffuse injury is more common than focal injury in chil- outcome.127,128 No studies have examined the impact on
dren with severe TBI106 and mass lesions requiring surgical outcome of strict glycemic control in children with severe TBI.
removal are less common. Diffuse brain swelling has been Core temperature is routinely monitored in pediatric criti-
reported to be the most common CT finding in children with cal care, and hyperthermia should be avoided. Induced hypo-
severe TBI107 and increases the likelihood of poor outcome.108 thermia remains controversial for pediatric TBI,129,130 but is
Subarachnoid hemorrhage on head CT has also been associ- better established as a therapy for other conditions, such as
ated with poor outcome in pediatric TBI.106 In the presence of perinatal asphyxia.131
brain swelling, often the basal cisterns on head CT are effaced
or obliterated; however, in children increased ICP can occur
in the presence of completely normal-appearing cisterns.36
Other
There is increasing concern about the effects of head CT on Although not strictly a monitoring modality, imaging of the
the developing brain. Careful thought should be given to chil- brain has evolved substantially and has become a powerful
dren who require multiple CT scans, including those of other tool for research and outcome prediction in TBI. Tools
body parts and studies such as CT-perfusion or angiography, such as diffusion-weighted imaging, susceptibility-weighted
and alternative examinations sought, if feasible, to reduce the imaging, magnetic resonance spectroscopy, and diffusion
total radiation dose and the potential for cumulative deleteri- tensor imaging have contributed to the appreciation of edema,
ous effects.109-115,115a For example, susceptibility-weighted microhemorrhages, ischemia, and white matter injury in adult
imaging (SWI), a relatively novel MRI sequence may be useful and pediatric TBI.132 Continuous electroencephalographic
in select patients to detect hemorrhagic lesions.116 However, monitoring can be used to diagnose convulsive and noncon-
its clinical usefulness in children is still to be elucidated. vulsive epileptiform activity and monitor the depth of barbi-
turate sedation. Seizures may be seen in as many as one third
of children in the critical care environment, with the great
Serum Biomarkers majority of these being nonconvulsive.133 Cortical spreading
Various serum biomarkers have been examined in adult and depolarization events happen frequently in adult TBI, and are
pediatric TBI for prognosticating and predicting the need associated with metabolic disturbances and poor outcome.134
for imaging. Berger et al.117 found that biomarkers (neuron- The role of these events in pediatric neurocritical care has not
specific enolase, myelin-basic protein and S100-B) were pre- yet been determined.
dictive of outcome. Specifically, the prediction of good
outcome (97%) was better than the prediction of poor
outcome (75%), and peak concentrations correlated better
Conclusion
with outcome than initial concentrations. However, the rela- There are fewer reports that examine neuromonitoring tools
tive strengths of association between biomarkers and outcome in critically ill children than in adults, so the evidence base for
for different childhood age groups still need further clarifica- making clinical decisions is small. This is compounded by the
tion,117,118 and it is unclear whether biomarker levels will influ- increased complexity of changing physiology with age in chil-
ence acute management or be used primarily to predict dren, and so the choice of monitoring tools and appropriate
outcome.119 targets is arguably more challenging than in adults. It is
important to remember that the rules governing monitoring
issues in adults may not apply to children, that the nature of
Glucose, Hemoglobin, and the pathology may be different, and that age-related physical
and physiologic issues must be taken into account when treat-
Temperature Monitoring ing children. Given the increased complexity of brain injury
Global trends have favored more conservative thresholds for pathophsyiology in children it is arguable that a multimodal
blood transfusion in critically ill children120; however, acute approach has even greater value than in adults.
anemia may have particular risks in TBI patients.121 Studies
examining the effect of blood transfusion on brain oxygen-
ation in adults and children65,122 demonstrate increased oxy- References
genation with transfusion; however, this tends to be temporary. 1. Segui-Gomez M, MacKenzie EJ. Measuring the public health impact of
Transfusion does not universally increase oxygenation, and injuries. Epidemiol Rev 2003;25:3–19.
the prediction of the effect based on pretransfusion character- 2. Arias E, Anderson RN, Kung HC, et al. Deaths: final data for 2001. Natl Vital
istics is incomplete. Therefore, optimal hemoglobin concen- Stat Rep 2003;52(3):1–116.
3. Cantais E, Paut O, Giorgi R, et al. Evaluating the prognosis of multiple,
trations, or transfusion triggers, in critically ill children with severely traumatized children in the intensive care unit. Intensive Care Med
neurologic pathology remain debated. It may be that the 2001;27:1511–17.
optimal level depends on the underlying severity of brain 4. Mayer T, Walker ML, Johnson DG, et al. Causes of morbidity and mortality
injury and risk of ischemia. in severe pediatric trauma. JAMA 1981;245:719–21.
Glucose control is similarly controversial. Hyperglycemia is 5. Sharples PM, Storey A, Aynsley-Green A, et al. Avoidable factors contributing
to death of children with head injury. BMJ 1990;300:87–91.
associated with worse outcome in children with TBI,123-125 and 6. Friess SH, Ralston J, Eucker SA, et al. Neurocritical care monitoring
strict control of glucose in pediatric trauma patients has correlates with neuropathology in a swine model of pediatric traumatic
been recommended.126 However, whether hyperglycemia is a brain injury. Neurosurgery 2011;69(5):1139–47; discussion 1147.
60 Section I—Background

7. Seshia SS, Bingham WT, Kirkham FJ, et al. Nontraumatic coma in children 30. Figaji AA. Practical aspects of bedside cerebral hemodynamics monitoring in
and adolescents: diagnosis and management. Neurol Clin. pediatric TBI. Childs Nerv Syst 2010;26:431–9.
2011;29(4):1007–43. 31. Adelson PD, Srinivas R, Chang Y, et al. Cerebrovascular response in
8. Odetola FO, Clark SJ, Lamarand KE, et al. Intracranial pressure monitoring children following severe traumatic brain injury. Childs Nerv Syst
in childhood meningitis with coma: A national survey of neurosurgeons in 2011;27:1465–76.
the United States. Pediatr Crit Care Med 2011;12(6):e350–6. 32. Morris KP, Forsyth RJ, Parslow RC, et al. Intracranial pressure complicating
9. Kamat P, Kunde S, Vos M, et al. Invasive intracranial pressure monitoring is severe traumatic brain injury in children: monitoring and management.
a useful adjunct in the management of severe hepatic encephalopathy Intensive Care Med 2006;32:1606–12.
associated with pediatric acute liver failure. Pediatr Crit Care Med 33. Plotz FB, Kneyber M, van Heerde M, et al. Traumatic pediatric brain injury
2012;13(1):e33–8. and intracranial pressure monitoring: does it really improve outcome?
10. Allen BB, Hoffman CE, Traube CS, et al. Continuous brain tissue Intensive Care Med 2007;33:1675.
oxygenation monitoring in the management of pediatric stroke. Neurocrit 34. Figaji AA, Adelson PD. Does ICP monitoring in children with severe head
Care. 2011;15(3):529–36. injuries make a difference? Am Surg 2009;75:441–2.
11. Kochanek PM, Carney N, Adelson PD, et al. Guidelines for the acute medical 35. Dean NP, Boslaugh S, Adelson PD, et al. Physician agreement with
management of severe traumatic brain injury in infants, children, and evidence-based recommendations for the treatment of severe traumatic
adolescents—second edition. Pediatr Crit Care Med 2012;13(Suppl. 1): brain injury in children. J Neurosurg 2007;107:387–91.
S1–82. 36. Kouvarellis AJ, Rohlwink UK, Sood V, et al. The relationship between basal
12. Ducrocq SC, Meyer PG, Orliaguet GA, et al. Epidemiology and early cisterns on CT and time-linked intracranial pressure in paediatric head
predictive factors of mortality and outcome in children with traumatic injury. Childs Nerv Syst 2011;27:1139–44.
severe brain injury: experience of a French pediatric trauma center. Pediatr 37. Jensen RL, Hahn YS, Ciro E. Risk factors of intracranial pressure monitoring
Crit Care Med 2006;7:461–7. in children with fiberoptic devices: a critical review. Surg Neurol 1997;47:
13. Salorio CF, Slomine BS, Guerguerian AM, et al. Intensive care unit variables 16–22.
and outcome after pediatric traumatic brain injury: a retrospective study of 38. Shapiro K, Marmarou A. Clinical applications of the pressure-volume index
survivors. Pediatr Crit Care Med 2008;9:47–53. in treatment of pediatric head injuries. J Neurosurg 1982;56:819–25.
14. Simpson D, Reilly P. Pediatric coma scale. Lancet 1982;2:450. 39. Udomphorn Y, Armstead WM, Vavilala MS. Cerebral blood flow and
15. Hahn YS, McLone DG. Risk factors in the outcome of children with minor autoregulation after pediatric traumatic brain injury. Pediatr Neurol 2008;38:
head injury. Pediatr Neurosurg 1993;19:135–42. 225–34.
16. Lu J, Murray GD, Steyerberg EW, et al. Effects of Glasgow outcome scale 40. Rohlwink UK, Zwane E, Fieggen AG, et al. The relationship between
misclassification on traumatic brain injury clinical trials. J Neurotrauma intracranial pressure and brain oxygenation in children with severe
2008;25:641–51. traumatic brain injury. Neurosurgery 2012;70:1220–30; discussion 1231.
17. Lobato RD, Rivas JJ, Gomez PA, et al. Head-injured patients who talk and 41. Prabhakaran P, Reddy AT, Oakes WJ, et al. A pilot trial comparing cerebral
deteriorate into coma. Analysis of 211 cases studied with computerized perfusion pressure-targeted therapy to intracranial pressure-targeted
tomography. J Neurosurg 1991;75:256–61. therapy in children with severe traumatic brain injury. J Neurosurg 2004;
18. Figaji AA, Zwane E, Fieggen AG, et al. Acute clinical grading in pediatric 100:454–9.
severe traumatic brain injury and its association with subsequent 42. Rosner MJ, Rosner SD, Johnson AH. Cerebral perfusion pressure:
intracranial pressure, cerebral perfusion pressure, and brain oxygenation. management protocol and clinical results. J Neurosurg 1995;83:949–62.
Neurosurg Focus 2008;25:E4. 43. Robertson CS, Valadka AB, Hannay HJ, et al. Prevention of secondary
19. Feickert HJ, Drommer S, Heyer R. Severe head injury in children: impact of ischemic insults after severe head injury. Crit Care Med 1999;27:2086–95.
risk factors on outcome. J Trauma 1999;47:33–8. 44. Bouma GJ, Muizelaar JP, Bandoh K, et al. Blood pressure and intracranial
20. Hackbarth RM, Rzeszutko KM, Sturm G, et al. Survival and functional pressure-volume dynamics in severe head injury: relationship with cerebral
outcome in pediatric traumatic brain injury: a retrospective review and blood flow. J Neurosurg 1992;77:15–9.
analysis of predictive factors. Crit Care Med 2002;30:1630–5. 45. Durward QJ, Del Maestro RF, Amacher AL, et al. The influence of systemic
21. Tepas 3rd JJ, DiScala C, Ramenofsky ML, et al. Mortality and head injury: arterial pressure and intracranial pressure on the development of cerebral
the pediatric perspective. J Pediatr Surg 1990;25:92–5. vasogenic edema. J Neurosurg 1983;59:803–9.
22. Coates BM, Vavilala MS, Mack CD, et al. Influence of definition and location 46. Nordstrom CH. Assessment of the optimal cerebral perfusion pressure in
of hypotension on outcome following severe pediatric traumatic brain head-injured patients. Anesth Analg 2005;101(1):299–300.
injury. Crit Care Med 2005;33:2645–50. 47. Wahlstrom MR, Olivecrona M, Koskinen LO, et al. Severe traumatic brain
23. Adelson PD, Bratton SL, Carney NA, et al. Guidelines for the acute medical injury in pediatric patients: treatment and outcome using an intracranial
management of severe traumatic brain injury in infants, children, and pressure targeted therapy—the Lund concept. Intensive Care Med 2005;31:
adolescents. Pediatr Crit Care Med 2003;4:S12–8. 832–9.
24. Haque IU, Zaritsky AL. Analysis of the evidence for the lower limit of 48. Andrews PJ, Citerio G. Lund therapy—pathophysiology-based therapy or
systolic and mean arterial pressure in children. Pediatr Crit Care Med contrived over-interpretation of limited data? Intensive Care Med 2006;32:
2007;8:138–44. 1461–3.
25. Rosner B, Prineas RJ, Loggie JM, et al. Blood pressure nomograms for 49. Chambers IR, Jones PA, Lo TY, et al. Critical thresholds of intracranial
children and adolescents, by height, sex, and age, in the United States. pressure and cerebral perfusion pressure related to age in paediatric head
J Pediatr 1993;123:871–86. injury. J Neurol Neurosurg Psychiatry 2006;77:234–40.
26. Bruce DA, Raphaely RC, Goldberg AI, et al. Pathophysiology, treatment 50. Chambers IR, Treadwell L, Mendelow AD. Determination of threshold levels
and outcome following severe head injury in children. Childs Brain of cerebral perfusion pressure and intracranial pressure in severe head injury
1979;5:174–91. by using receiver-operating characteristic curves: an observational study in
27. Sharples PM, Stuart AG, Matthews DS, et al. Cerebral blood flow and 291 patients. J Neurosurg 2001;94:412–16.
metabolism in children with severe head injury. Part 1: relation to age,
Glasgow coma score, outcome, intracranial pressure, and time after injury.
J Neurol Neurosurg Psychiatry 1995;58:145–52. A complete list of references for this chapter can be found online at
28. Zwienenberg M, Muizelaar JP. Severe pediatric head injury: the role of www.expertconsult.com.
hyperemia revisited. J Neurotrauma 1999;16:937–43.
29. Chiron C, Raynaud C, Maziere B, et al. Changes in regional cerebral blood
flow during brain maturation in children and adolescents. J Nucl Med 1992;
33:696–703.
Section I—Background 60.e1

References 27. Sharples PM, Stuart AG, Matthews DS, et al. Cerebral blood flow and
metabolism in children with severe head injury. Part 1: relation to age,
1. Segui-Gomez M, MacKenzie EJ. Measuring the public health impact of Glasgow coma score, outcome, intracranial pressure, and time after injury.
injuries. Epidemiol Rev 2003;25:3–19. J Neurol Neurosurg Psychiatry 1995;58:145–52.
2. Arias E, Anderson RN, Kung HC, et al. Deaths: final data for 2001. Natl 28. Zwienenberg M, Muizelaar JP. Severe pediatric head injury: the role of
Vital Stat Rep 2003;52(3):1–116. hyperemia revisited. J Neurotrauma 1999;16:937–43.
3. Cantais E, Paut O, Giorgi R, et al. Evaluating the prognosis of multiple, 29. Chiron C, Raynaud C, Maziere B, et al. Changes in regional cerebral blood
severely traumatized children in the intensive care unit. Intensive Care Med flow during brain maturation in children and adolescents. J Nucl Med
2001;27:1511–17. 1992;33:696–703.
4. Mayer T, Walker ML, Johnson DG, et al. Causes of morbidity and mortality 30. Figaji AA. Practical aspects of bedside cerebral hemodynamics monitoring
in severe pediatric trauma. JAMA 1981;245:719–21. in pediatric TBI. Childs Nerv Syst 2010;26:431–9.
5. Sharples PM, Storey A, Aynsley-Green A, et al. Avoidable factors 31. Adelson PD, Srinivas R, Chang Y, et al. Cerebrovascular response in
contributing to death of children with head injury. BMJ 1990;300:87–91. children following severe traumatic brain injury. Childs Nerv Syst
6. Friess SH, Ralston J, Eucker SA, et al. Neurocritical care monitoring 2011;27:1465–76.
correlates with neuropathology in a swine model of pediatric traumatic 32. Morris KP, Forsyth RJ, Parslow RC, et al. Intracranial pressure complicating
brain injury. Neurosurgery 2011;69(5):1139–47; discussion 1147. severe traumatic brain injury in children: monitoring and management.
7. Seshia SS, Bingham WT, Kirkham FJ, et al. Nontraumatic coma in children Intensive Care Med 2006;32:1606–12.
and adolescents: diagnosis and management. Neurol Clin. 33. Plotz FB, Kneyber M, van Heerde M, et al. Traumatic pediatric brain injury
2011;29(4):1007–43. and intracranial pressure monitoring: does it really improve outcome?
8. Odetola FO, Clark SJ, Lamarand KE, et al. Intracranial pressure monitoring Intensive Care Med 2007;33:1675.
in childhood meningitis with coma: A national survey of neurosurgeons in 34. Figaji AA, Adelson PD. Does ICP monitoring in children with severe head
the United States. Pediatr Crit Care Med 2011;12(6):e350–6. injuries make a difference? Am Surg 2009;75:441–2.
9. Kamat P, Kunde S, Vos M, et al. Invasive intracranial pressure monitoring is 35. Dean NP, Boslaugh S, Adelson PD, et al. Physician agreement with
a useful adjunct in the management of severe hepatic encephalopathy evidence-based recommendations for the treatment of severe traumatic
associated with pediatric acute liver failure. Pediatr Crit Care Med brain injury in children. J Neurosurg 2007;107:387–91.
2012;13(1):e33–8. 36. Kouvarellis AJ, Rohlwink UK, Sood V, et al. The relationship between basal
10. Allen BB, Hoffman CE, Traube CS, et al. Continuous brain tissue cisterns on CT and time-linked intracranial pressure in paediatric head
oxygenation monitoring in the management of pediatric stroke. Neurocrit injury. Childs Nerv Syst 2011;27:1139–44.
Care. 2011;15(3):529–36. 37. Jensen RL, Hahn YS, Ciro E. Risk factors of intracranial pressure
11. Kochanek PM, Carney N, Adelson PD, et al. Guidelines for the acute monitoring in children with fiberoptic devices: a critical review. Surg
medical management of severe traumatic brain injury in infants, children, Neurol 1997;47:16–22.
and adolescents—second edition. Pediatr Crit Care Med 2012;13(Suppl. 1): 38. Shapiro K, Marmarou A. Clinical applications of the pressure-volume index
S1–82. in treatment of pediatric head injuries. J Neurosurg 1982;56:819–25.
12. Ducrocq SC, Meyer PG, Orliaguet GA, et al. Epidemiology and early 39. Udomphorn Y, Armstead WM, Vavilala MS. Cerebral blood flow and
predictive factors of mortality and outcome in children with traumatic autoregulation after pediatric traumatic brain injury. Pediatr Neurol
severe brain injury: experience of a French pediatric trauma center. Pediatr 2008;38:225–34.
Crit Care Med 2006;7:461–7. 40. Rohlwink UK, Zwane E, Fieggen AG, et al. The relationship between
13. Salorio CF, Slomine BS, Guerguerian AM, et al. Intensive care unit variables intracranial pressure and brain oxygenation in children with severe
and outcome after pediatric traumatic brain injury: a retrospective study of traumatic brain injury. Neurosurgery 2012;70:1220–30; discussion
survivors. Pediatr Crit Care Med 2008;9:47–53. 1231.
14. Simpson D, Reilly P. Pediatric coma scale. Lancet 1982;2:450. 41. Prabhakaran P, Reddy AT, Oakes WJ, et al. A pilot trial comparing cerebral
15. Hahn YS, McLone DG. Risk factors in the outcome of children with minor perfusion pressure-targeted therapy to intracranial pressure-targeted
head injury, Pediatr Neurosurg 1993;19:135–42. therapy in children with severe traumatic brain injury. J Neurosurg
16. Lu J, Murray GD, Steyerberg EW, et al. Effects of Glasgow outcome scale 2004;100:454–9.
misclassification on traumatic brain injury clinical trials. J Neurotrauma 42. Rosner MJ, Rosner SD, Johnson AH. Cerebral perfusion pressure:
2008;25:641–51. management protocol and clinical results. J Neurosurg 1995;83:949–62.
17. Lobato RD, Rivas JJ, Gomez PA, et al. Head-injured patients who talk and 43. Robertson CS, Valadka AB, Hannay HJ, et al. Prevention of secondary
deteriorate into coma. Analysis of 211 cases studied with computerized ischemic insults after severe head injury. Crit Care Med 1999;27:2086–95.
tomography. J Neurosurg 1991;75:256–61. 44. Bouma GJ, Muizelaar JP, Bandoh K, et al. Blood pressure and intracranial
18. Figaji AA, Zwane E, Fieggen AG, et al. Acute clinical grading in pediatric pressure-volume dynamics in severe head injury: relationship with cerebral
severe traumatic brain injury and its association with subsequent blood flow. J Neurosurg 1992;77:15–9.
intracranial pressure, cerebral perfusion pressure, and brain oxygenation. 45. Durward QJ, Del Maestro RF, Amacher AL, et al. The influence of systemic
Neurosurg Focus 2008;25:E4. arterial pressure and intracranial pressure on the development of cerebral
19. Feickert HJ, Drommer S, Heyer R. Severe head injury in children: impact of vasogenic edema. J Neurosurg 1983;59:803–9.
risk factors on outcome. J Trauma 1999;47:33–8. 46. Nordstrom CH. Assessment of the optimal cerebral perfusion pressure in
20. Hackbarth RM, Rzeszutko KM, Sturm G, et al. Survival and functional head-injured patients. Anesth Analg 2005;101:(1):299–300.
outcome in pediatric traumatic brain injury: a retrospective review and 47. Wahlstrom MR, Olivecrona M, Koskinen LO, et al. Severe traumatic brain
analysis of predictive factors. Crit Care Med 2002;30:1630–5. injury in pediatric patients: treatment and outcome using an intracranial
21. Tepas 3rd JJ, DiScala C, Ramenofsky ML, et al. Mortality and head injury: pressure targeted therapy—the Lund concept. Intensive Care Med
the pediatric perspective. J Pediatr Surg 1990;25:92–5. 2005;31:832–9.
22. Coates BM, Vavilala MS, Mack CD, et al. Influence of definition and 48. Andrews PJ, Citerio G. Lund therapy—pathophysiology-based therapy
location of hypotension on outcome following severe pediatric traumatic or contrived over-interpretation of limited data? Intensive Care Med
brain injury. Crit Care Med 2005;33:2645–50. 2006;32:1461–3.
23. Adelson PD, Bratton SL, Carney NA, et al. Guidelines for the acute medical 49. Chambers IR, Jones PA, Lo TY, et al. Critical thresholds of intracranial
management of severe traumatic brain injury in infants, children, and pressure and cerebral perfusion pressure related to age in paediatric head
adolescents. Pediatr Crit Care Med 2003;4:S12–8. injury. J Neurol Neurosurg Psychiatry 2006;77:234–40.
24. Haque IU, Zaritsky AL. Analysis of the evidence for the lower limit of 50. Chambers IR, Treadwell L, Mendelow AD. Determination of threshold
systolic and mean arterial pressure in children. Pediatr Crit Care Med levels of cerebral perfusion pressure and intracranial pressure in severe
2007;8:138–44. head injury by using receiver-operating characteristic curves: an
25. Rosner B, Prineas RJ, Loggie JM, et al. Blood pressure nomograms for observational study in 291 patients. J Neurosurg 2001;94:412–16.
children and adolescents, by height, sex, and age, in the United States. 51. Vavilala MS, Bowen A, Lam AM, et al. Blood pressure and outcome after
J Pediatr 1993;123:871–86. severe pediatric traumatic brain injury. J Trauma 2003;55:1039–44.
26. Bruce DA, Raphaely RC, Goldberg AI, et al. Pathophysiology, treatment 52. Jones PA, Andrews PJ, Easton VJ, et al. Traumatic brain injury in
and outcome following severe head injury in children. Childs Brain childhood: intensive care time series data and outcome. Br J Neurosurg
1979;5:174–91. 2003;17:29–39.
60.e2 Section I—Background

53. Downard C, Hulka F, Mullins RJ, et al. Relationship of cerebral perfusion 77. Tolias C, Richards D, Bowery N, et al. Investigation of extracellular amino
pressure and survival in pediatric brain-injured patients. J Trauma acid release in children with severe head injury using microdialysis. A pilot
2000;49(4):654–8; discussion 658–9. study. Acta Neurochir Suppl 2002;81:377–9.
54. Figaji AA, Zwane E, Fieggen AG, et al. Pressure autoregulation, intracranial 78. Tolias CM, Richards DA, Bowery NG, et al. Extracellular glutamate in the
pressure, and brain tissue oxygenation in children with severe traumatic brains of children with severe head injuries: a pilot microdialysis study.
brain injury. J Neurosurg Pediatr 2009;4:420–8. Childs Nerv Syst 2002;18:368–74.
55. Figaji AA, Zwane E, Thompson C, et al. Brain tissue oxygen tension 79. Richards DA, Tolias CM, Sgouros S, et al. Extracellular glutamine to
monitoring in pediatric severe traumatic brain injury: part 2: relationship glutamate ratio may predict outcome in the injured brain: a clinical
with clinical, physiological, and treatment factors. Childs Nerv Syst microdialysis study in children. Pharmacol Res 2003;48:101–9.
2009;25:1335–43. 80. Nakamura T, Nagao S, Kawai N, et al. Significance of multimodal cerebral
56. Maloney-Wilensky E, Gracias V, Itkin A, et al. Brain tissue oxygen and monitoring under moderate therapeutic hypothermia for severe head
outcome after severe traumatic brain injury: a systematic review. Crit Care injury. Acta Neurochir Suppl 1998;71:85–7.
Med 2009;37:2057–63. 81. Perez A, Minces PG, Schnitzler EJ, et al. Jugular venous oxygen
57. Narotam PK, Burjonrappa SC, Raynor SC, et al. Cerebral oxygenation in saturation or arteriovenous difference of lactate content and outcome in
major pediatric trauma: its relevance to trauma severity and outcome. children with severe traumatic brain injury. Pediatr Crit Care Med 2003;4:
J Pediatr Surg 2006;41:505–13. 33–8.
58. Stiefel MF, Udoetuk JD, Storm PB, et al. Brain tissue oxygen monitoring 82. Gayle MO, Frewen TC, Armstrong RF, et al. Jugular venous bulb
in pediatric patients with severe traumatic brain injury. J Neurosurg catheterization in infants and children. Crit Care Med 1989;17:385–8.
2006;105:281–6. 83. Dearden NM, Midgley S. Technical considerations in continuous jugular
59. Figaji AA, Fieggen AG, Argent AC, et al. Does adherence to treatment venous oxygen saturation measurement. Acta Neurochir Suppl (Wien)
targets in children with severe traumatic brain injury avoid brain hypoxia? 1993;59:91–7.
A brain tissue oxygenation study. Neurosurgery 2008;63(1):83–91; 84. Kiening KL, Unterberg AW, Bardt TF, et al. Monitoring of cerebral
discussion 91–2. oxygenation in patients with severe head injuries: brain tissue PO2 versus
60. Figaji AA, Zwane E, Thompson C, et al. Brain tissue oxygen tension jugular vein oxygen saturation. J Neurosurg 1996;85:751–7.
monitoring in pediatric severe traumatic brain injury: part 2: relationship 85. Kilic T, Elmaci I, Ozek MM, et al. Utility of transcranial Doppler
with clinical, physiological, and treatment factors. Childs Nerv Syst ultrasonography in the diagnosis and follow-up of tuberculous
2009;25(10):1335–43. meningitis-related vasculopathy. Childs Nerv Syst 2002;18:142–6.
61. Figaji AA, Zwane E, Thompson C, et al. Brain tissue oxygen tension 86. Nadvi SS, Du Trevou MD, Van Dellen JR, et al. The use of transcranial
monitoring in pediatric severe traumatic brain injury: part 1: relationship Doppler ultrasonography as a method of assessing intracranial pressure in
with outcome. Childs Nerv Syst 2009;25:1325–33. hydrocephalic children. Br J Neurosurg 1994;8:573–7.
62. Figaji AA, Zwane E, Fieggen AG, et al. Acute clinical grading in pediatric 87. Mandera M, Larysz D, Wojtacha M. Changes in cerebral hemodynamics
severe traumatic brain injury and its association with subsequent assessed by transcranial Doppler ultrasonography in children after head
intracranial pressure, cerebral perfusion pressure, and brain oxygenation. injury. Childs Nerv Syst 2002;18:124–8.
Neurosurg Focus 2008;25:E4. 88. Vavilala MS, Kincaid MS, Muangman SL, et al. Gender differences in
63. Figaji AA, Zwane E, Graham Fieggen A, et al. The effect of increased cerebral blood flow velocity and autoregulation between the anterior and
inspired fraction of oxygen on brain tissue oxygen tension in children with posterior circulations in healthy children. Pediatr Res 2005;58:574–8.
severe traumatic brain injury. Neurocrit Care 2010;12:430–7. 89. Chan KH, Miller JD, Dearden NM, et al. The effect of changes in cerebral
64. Figaji AA, Kent SJ. Brain tissue oxygenation in children diagnosed with perfusion pressure upon middle cerebral artery blood flow velocity and
brain death. Neurocrit Care 2010;12:56–61. jugular bulb venous oxygen saturation after severe brain injury. J Neurosurg
65. Figaji AA, Zwane E, Kogels M, et al. The effect of blood transfusion on 1992;77:55–61.
brain oxygenation in children with severe traumatic brain injury. Pediatr 90. Bellner J, Romner B, Reinstrup P, et al. Transcranial Doppler sonography
Crit Care Med 2010;11:325–31. pulsatility index (PI) reflects intracranial pressure (ICP). Surg Neurol
66. Rohlwink UK, Figaji AA. Methods of monitoring brain oxygenation. Childs 2004;62(1):45–51; discussion 51.
Nerv Syst 2010;26:453–64. 91. Meyer PG, Ducrocq S, Rackelbom T, et al. Surgical evacuation of acute
67. Wardle SP, Yoxall CW, Weindling AM. Determinants of cerebral fractional subdural hematoma improves cerebral hemodynamics in children: a
oxygen extraction using near infrared spectroscopy in preterm neonates. transcranial Doppler evaluation. Childs Nerv Syst 2005;21:133–7.
J Cereb Blood Flow Metab 2000;20:272–9. 92. Trabold F, Meyer PG, Blanot S, et al. The prognostic value of transcranial
68. Abdul-Khaliq H, Troitzsch D, Berger F, et al. Regional transcranial oximetry Doppler studies in children with moderate and severe head injury. Intensive
with near infrared spectroscopy (NIRS) in comparison with measuring Care Med 2004;30:108–12.
oxygen saturation in the jugular bulb in infants and children for 93. Vajda Z, Buki A, Veto F, et al. Transcranial Doppler-determined pulsatility
monitoring cerebral oxygenation. Biomed Tech (Berl) 2000;45:328–32. index in the evaluation of endoscopic third ventriculostomy (preliminary
69. Shimizu N, Gilder F, Bissonnette B, et al. Brain tissue oxygenation index data). Acta Neurochir (Wien) 1999;141:247–50.
measured by near infrared spatially resolved spectroscopy agreed with 94. Lindegaard KF, Nornes H, Bakke SJ, et al. Cerebral vasospasm diagnosis by
jugular bulb oxygen saturation in normal pediatric brain: a pilot study. means of angiography and blood velocity measurements. Acta Neurochir
Childs Nerv Syst 2005;21:181–4. (Wien) 1989;100:12–24.
70. Adelson PD, Nemoto E, Colak A, et al. The use of near infrared 95. Figaji AA, Zwane E, Fieggen AG, et al. Transcranial Doppler pulsatility
spectroscopy (NIRS) in children after traumatic brain injury: a preliminary index is not a reliable indicator of intracranial pressure in children with
report. Acta Neurochir Suppl 1998;71:250–4. severe traumatic brain injury. Surg Neurol 2009;72(4):389–94.
71. Buchner K, Meixensberger J, Dings J, et al. Near-infrared spectroscopy—not 96. Vavilala MS, Lee LA, Boddu K, et al. Cerebral autoregulation in pediatric
useful to monitor cerebral oxygenation after severe brain injury. Zentralbl traumatic brain injury. Pediatr Crit Care Med 2004;5:257–63.
Neurochir 2000;61:69–73. 97. Vavilala MS, Muangman S, Tontisirin N, et al. Impaired cerebral
72. Holzschuh M, Woertgen C, Metz C, et al. Dynamic changes of cerebral autoregulation and 6-month outcome in children with severe traumatic
oxygenation measured by brain tissue oxygen pressure and near infrared brain injury: preliminary findings. Dev Neurosci 2006;28:348–53.
spectroscopy. Neurol Res 1997;19:246–8. 98. Panerai RB, Kerins V, Fan L, et al. Association between dynamic cerebral
73. Nagdyman N, Fleck T, Schubert S, et al. Comparison between cerebral autoregulation and mortality in severe head injury. Br J Neurosurg 2004;18:
tissue oxygenation index measured by near-infrared spectroscopy and 471–9.
venous jugular bulb saturation in children. Intensive Care Med 2005;31: 99. Panerai RB. Assessment of cerebral pressure autoregulation in humans—a
846–50. review of measurement methods. Physiol Meas 1998;19:305–38.
74. Schwarz G, Litscher G, Kleinert R, et al. Cerebral oximetry in dead subjects. 100. Newell DW, Aaslid R, Lam A, et al. Comparison of flow and velocity during
J Neurosurg Anesthesiol 1996;8:189–93. dynamic autoregulation testing in humans. Stroke 1994;25:793–7.
75. Weerakkody RA, Czosnyka M, Zweifel C, et al. Near infrared spectroscopy 101. Steiner LA, Coles JP, Johnston AJ, et al. Assessment of cerebrovascular
as possible non-invasive monitor of slow vasogenic ICP waves. Acta autoregulation in head-injured patients: a validation study. Stroke 2003;34:
Neurochir Suppl 2012;114:181–5. 2404–9.
76. Zweifel C, Castellani G, Czosnyka M, et al. Noninvasive monitoring of 102. Vavilala MS, Tontisirin N, Udomphorn Y, et al. Hemispheric differences in
cerebrovascular reactivity with near infrared spectroscopy in head-injured cerebral autoregulation in children with moderate and severe traumatic
patients. J Neurotrauma 2010;27:1951–8. brain injury. Neurocrit Care 2008;9(1):45–54.
Section I—Background 60.e3

103. Tontisirin N, Armstead W, Waitayawinyu P, et al. Change in cerebral 118. Shore PM, Berger RP, Varma S, et al. Cerebrospinal fluid biomarkers versus
autoregulation as a function of time in children after severe traumatic glasgow coma scale and glasgow outcome scale in pediatric traumatic brain
brain injury: a case series. Childs Nerv Syst 2007;23:1163–9. injury: the role of young age and inflicted injury. J Neurotrauma 2007;24:
104. Czosnyka M, Smielewski P, Piechnik S, et al. Continuous assessment of 75–86.
cerebral autoregulation—clinical verification of the method in head injured 119. Sandler SJ, Figaji AA, Adelson PD. Clinical applications of biomarkers in
patients. Acta Neurochir Suppl 2000;76:483–4. pediatric traumatic brain injury. Childs Nerv Syst 2010;26:205–13.
105. Brady KM, Shaffner DH, Lee JK, et al. Continuous monitoring of 120. Lacroix J, Hebert PC, Hutchison JS, et al. Transfusion strategies for patients
cerebrovascular pressure reactivity after traumatic brain injury in children. in pediatric intensive care units. N Engl J Med 2007;356:1609–19.
Pediatrics 2009;124:e1205–12. 121. Hare GM, Mazer CD, Hutchison JS, et al. Severe hemodilutional anemia
106. Pillai S, Praharaj SS, Mohanty A, et al. Prognostic factors in children with increases cerebral tissue injury following acute neurotrauma. J Appl Physiol
severe diffuse brain injuries: a study of 74 patients. Pediatr Neurosurg 2007;103:1021–9.
2001;34:98–103. 122. Smith MJ, Stiefel MF, Magge S, et al. Packed red blood cell transfusion
107. Levin HS, Aldrich EF, Saydjari C, et al. Severe head injury in children: increases local cerebral oxygenation. Crit Care Med 2005;33:1104–8.
experience of the Traumatic Coma Data Bank. Neurosurgery 1992;31:435, 123. Michaud LJ, Rivara FP, Longstreth Jr WT, et al. Elevated initial blood
443. glucose levels and poor outcome following severe brain injuries in children.
108. Aldrich EF, Eisenberg HM, Saydjari C, et al. Diffuse brain swelling in J Trauma 1991;31:1356–62.
severely head-injured children. A report from the NIH Traumatic Coma 124. Cochran A, Scaife ER, Hansen KW, et al. Hyperglycemia and outcomes
Data Bank. J Neurosurg 1992;76:450–4. from pediatric traumatic brain injury. J Trauma 2003;55:1035–8.
109. Dougeni E, Faulkner K, Panayiotakis G. A review of patient dose and 125. Smith RL, Lin JC, Adelson PD, et al. Relationship between hyperglycemia
optimisation methods in adult and paediatric CT scanning. Eur J Radiol and outcome in children with severe traumatic brain injury. Pediatr Crit
2012;81(4):e665–83. Care Med 2012;13:85–91.
110. Guillerman RP. Newer CT applications and their alternatives: what is 126. Tuggle DW, Kuhn MA, Jones SK, et al. Hyperglycemia and infections in
appropriate in children? Pediatr Radiol 2011;41(Suppl 2):534–48. pediatric trauma patients. Am Surg 2008;74:195–8.
111. Mahesh M. Advances in CT technology and application to pediatric 127. Strong AJ, Boutelle MG, Vespa PM, et al. Treatment of critical care patients
imaging. Pediatr Radiol 2011;41(Suppl 2):493–7. with substantial acute ischemic or traumatic brain injury. Crit Care Med
112. Mueller DL, Hatab M, Al-Senan R, Cohn SM, et al. Pediatric radiation 2005;33(9):2147–9.
exposure during the initial evaluation for blunt trauma. J Trauma 128. Vespa P, Boonyaputthikul R, McArthur DL, et al. Intensive insulin therapy
2011;70(3):724–31. reduces microdialysis glucose values without altering glucose utilization or
113. Kuppermann N, Holmes JF, Dayan PS, et al, Pediatric Emergency Care improving the lactate/pyruvate ratio after traumatic brain injury. Crit Care
Applied Research Network (PECARN). Identification of children at very Med 2006;34:850–6.
low risk of clinically important brain injuries after head trauma: a 129. Hutchison JS, Ward RE, Lacroix J, et al. Hypothermia therapy after
prospective cohort study. Lancet 2009;374(9696):1160–70. traumatic brain injury in children. N Engl J Med 2008;358:2447–56.
114. Brenner DJ, Hall EJ. Computed tomography—an increasing source of 130. Kochanek PM, Bell MJ, Adelson P. Hypothermia therapy after traumatic
radiation exposure. N Engl J Med 2007;357(22):2277–84. brain injury in children. N Engl J Med 2008;359:1179.
115. Scaife ER, Rollins MD. Managing radiation risk in the evaluation of the 131. Jacobs S, Hunt R, Tarnow-Mordi W, et al. Cooling for newborns with
pediatric trauma patient. Semin Pediatr Surg 2010;19(4):252–6. hypoxic ischaemic encephalopathy. Cochrane Database Syst Rev 2007;(4):
115a.  Nievelstein RA, van Dam IM, van der Molen AJ. Multidetector CT in CD003311.
children: current concepts and dose reduction strategies. Pediatr Radiol 132. Friess SH, Kilbaugh TJ, Huh JW. Advanced neuromonitoring and imaging
2010;40(8):1324–44. in pediatric traumatic brain injury. Crit Care Res Pract 2012;361310.
116. Beauchamp MH, Ditchfield M, Babl FE, et al. Detecting traumatic brain 133. McCoy B, Sharma R, Ochi A, et al. Predictors of nonconvulsive seizures
lesions in children: CT versus MRI versus susceptibility weighted imaging among critically ill children. Epilepsia 2011;52(11):1973–8.
(SWI). J Neurotrauma 2011;28(6):915–27. 134. Hartings JA, Bullock MR, Okonkwo DO, et al. Spreading depolarisations
117. Berger RP, Beers SR, Richichi R, et al. Serum biomarker concentrations and and outcome after traumatic brain injury: a prospective observational
outcome after pediatric traumatic brain injury. J Neurotrauma 2007;24: study. Lancet Neurol 2011;10(12):1058–64.
1793–801.
Chapter
8  
I

Bioethics and the Family


Patricia D. Scripko and David M. Greer

“Medical technology has effectively created a twilight zone consciousness and of death that are useful in this discussion.
of suspended animation where death commences while life, The text continues with the utility of neuromonitoring, spe-
in some form, continues.” cifically in preventing futile care and its limitations. Finally,
—Justice William Brennan, 1990, regarding the case of ethical dilemmas in which families are engaged in conversa-
Nancy Cruzan v. The State of Missouri tions about the concepts of neuromonitoring, death, and futil-
ity are discussed.
Introduction
Medicine’s purpose is to improve and lengthen life. When a Brain Death: A Definition
person dies, therefore, medicine, in a sense, has failed. These of Human Death
simple truths lead to the far more complex and contentious
question of when that failure has occurred. Philosophers, History
doctors, and lawyers have debated the question of how to Although the terminology is misleading, brain death in the
define death for centuries. The answer has impli­cations for United States and most countries worldwide legally is equated
political and medical issues such as organ donation. to death of the person, not simply the death of the brain in an
In recent times, the issue has been complicated further otherwise living person. The ambiguous nomenclature is an
by steady progress in medical technology. Some of this artifact of historical changes to clinically identify and define
technology directly affects clinical actions to prevent and death (Table 8.2). Before the 19th century, cessation of
diagnose death by improving the assessment of it, inhibiting breathing, an easily observable function, equated human
its progress, or altering the understanding of death alto- death. With the invention of the stethoscope in the 1800s,
gether. Technologies that aid the ability to reverse certain cessation of an audible heartbeat became the more consid-
pathologic processes and thereby help avoid raising the ered definition. Once mechanical ventilation and resuscita-
white flag to death, also pose the related question: Is use tion became routine in critical care ICUs in the 1950s and
of these technologies in a particular patient futile? Futility 1960s1,2 and so allowed for continued ventilation and circula-
is a highly debated concept that concerns irreversible states tory support in patients in the absence of neurologic activ-
with depreciated quality or an inevitable progression to the ity,* brain death arose as an alternative definition for death in
end of life. The question of futility is both unavoidable, patients after catastrophe and with absent neurologic func-
in that it affects decisions in critical care, and ethically tion.1,3 The Harvard Commission clarified brain death crite-
complicated, because it raises questions of patient auton- ria in 1968;4 although altered slightly on multiple occasions
omy in end-of-life decision making and proper resource since (see reference 1 for review),5-10 the criteria typically
allocation. include coma, or total unawareness of external and internal
Neuromonitoring encompasses a specific set of technologic stimuli; cranial areflexia; no spontaneous breathing (apnea);
tools that may have an effect on current clinical protocols for and a flat electroencephalogram (EEG), all observed in the
patients with critical neurologic illnesses. It is worth consider- absence of alternative explanations for the state, including
ing how the potential of these tools may be realized, and what intoxication, metabolic disorders, or hypothermia.11 Legal
ethical implications they may have in the intensive care definitions depend on medical criteria, with vague terminol-
unit (ICU). This chapter attempts to integrate medical ogy that allows room for these criteria to change and still
and legal concepts into a discussion on the ethics of neuro- coincide with the law. For example, as written by the
monitoring in the ICU, specifically focusing on its role in cases American Bar Association (ABA) in 1975, “For all legal pur-
of perceived medical or neurologic futility. We begin by poses, a human body with irreversible cessation of total brain
defining human death, for to understand what is futile, function … shall be considered dead.”12 Specifics on what
one must understand what defines irreversible versus revers-
ible neurologically. Table 8.1 lists definitions of states of *Albeit in a disintegrated fashion, according to the 1981 President’s Commission Report.

© Copyright 2013 Elsevier Inc. All rights reserved. 61


62 Section I—Background

Table 8.1  Relevant Terms to Discuss Human Life and Death


Term Definition References
Locked-in syndrome Caused by a specific lesion in the pons. This syndrome consists of anarthria and 41
quadriplegia with preservation of cognition and conscious awareness. Patients typically
can respond only by blinking or with vertical eye movements. This syndrome must be
distinguished from states of impaired consciousness (i.e., MCS, PVS, coma).
Minimally conscious Identified by partial consciousness; sleep-wake cycles are present and the patient is able 62
state (MCS) to localize both noxious stimuli and location of sound origin. Communication consists
of intelligible verbalization and/or contingent vocalization.
Persistent vegetative For at least 1 month no conscious awareness or communicative ability. Sleep-wake cycles, 110
state (PVS) spontaneous, nonpurposeful movement, and stimulated arousal are present.
Permanent For at least 1 year after traumatic brain injury, or 3 months following ischemic injury (a), (a) 51
vegetative state the patient has no conscious awareness or communicative ability. Sleep-wake cycles (b) 110
spontaneous, nonpurposeful movements and stimulated arousal are present (b).
Coma No conscious awareness, no communicative ability, and no sleep-wake cycles, with an 41
inability to be aroused, even by noxious stimuli
Brain death Equated with human death, but complicated by differing specifics in its definition, which 111
depends on certain clinical diagnostic criteria
Irreversible Two connotations: a strong one, which means that the function can never, not even in 16
the future, be restored, and a weak one that means given the present tools and
situation, function cannot be restored.
Permanent “Lasting or intended to last or remain unchanged indefinitely” 15
“Lasting or continuing without interruption”
Human A being with specific traits identified as necessary and sufficient to declare one
“human.” These traits span from human deoxyribonucleic acid (DNA) to conscious
awareness, and are controversial.
Person Often associated with traits specific to an individual human, but, in this paper, is equated
with human.
Higher brain “Capacity to think, to perceive and to respond” (a) (a) 13
Sentience: “able to perceive or feel” (b) (b) 15
Lower brain The capacity to “integrate bodily functions” 13, 112
Whole brain Both higher and lower brain function 14, 32

constitutes “irreversible,” “total,” and “function” were not pro- something will not be recovered now and cannot ever be
vided; these are discussed in the following text. recovered later. The weaker definition refers to presently unre-
coverable objects or states that may or may not be recoverable
in the future (with better technology).16 Aulisio et al.17 sug-
A Closer Look at the Definition: “Total” gested an alternative weak interpretation of the word: an
and “Irreversible” inability to restore without intervention. These weaker defini-
Brain death carries a legal definition in accordance with whole tions are in line with the concept of the contextual depen-
brain theorists, or those persons who believe that the irrevers- dency of death that becomes very relevant in the neurocritical
ible cessation of both higher and lower brain function are care environment. For example, Ropper recognized that death
individually necessary and together sufficient to declare depends on technologic opportunities to preserve human life
someone a deceased human. Higher and lower brain functions that are available at a given time.18 This means that so long as
are defined, respectively, as the “capacity to think, to perceive there is no machine at the present to substitute for, or restore
and to respond” (cortical function) and the capacity to “inte- higher and lower brain function, then brain death is human
grate bodily functions” (brainstem function).13 Several authors death. A historical example of Ropper’s view is evident in the
support “whole brain” death criteria and claim its conservative impact of mechanical ventilation and resuscitation that elimi-
nature is necessary to protect against misdiagnosis of a person nated cardiac death as brain death (see Table 8.2). Further
as brain dead who actually is transitioning to an improved expanding the contextual dependency of irreversible, under
neurologic state or has the potential for such a change. To many state statutes, such as the often-cited Capron and Kass
fulfill these criteria, destruction of neuronal tissue must be model,19 physicians are bestowed the power to define and
so widespread that there is no doubt the person’s state is identify “irreversible” and “relevant” (brain) functions.†20
irreversible.14 These statutes call for diagnostic decisions to be based on best
Irreversible is itself an inconclusive term, with both strong
and weak interpretations,15 as noted by Bernat.14 According to †
The Uniform Determination of Death Act also reads that “determination of death must
Cole, under the stronger definition, irreversible means that be made in accordance with accepted medical standards.”
Section I—Background 63

Table 8.2  Important Events in Defining Human Life and Death By Chronologic Order
Year Event References
Pre-1800s Death assessed by cessation of breathing, a readily observable event 3
1800s Stethoscope is invented, leading physicians to identify death by the now-observable cessation of 3
beating of the heart (“cardiac death”)
1950s Mechanical ventilation and resuscitation are staples in the intensive care unit (ICU), leading physicians 1
to shift to a neurologic base for death criteria (“brain death”)
1968 Harvard criteria are written and define brain death as (1) deep coma/no withdrawal to painful 4
stimuli; (2) cranial and spinal arreflexia; (3) apnea when the ventilator is disconnected for 3
minutes; (4) a flat electroencephalogram (EEG); (5) exclusion of hypothermia and drugs as a cause
for the findings; and (6) repeat examination in 24 hours.
1969 Luis Kutner conceived advance directives as a practical concept. He proposed legislature in 1968. 96
1971 Minnesota criteria are published. Criteria were similar to the Harvard criteria, except that a requisite 5
flat EEG was omitted, the repeat examination was to occur at 12 hours, and an “irrefutable
intracranial lesion” was identified in the patient.
1973 The case of Roe v. Wade ruled in favor of abortion before the fetus is “viable,” at which point the 108
fetus is granted legal rights as a person, a living human. This had a great impact on how society
viewed human life.
1973 The case of Edelin: A chief obstetrics resident performed a late second-term abortion at the request 57
of a teenage girl and her mother. A 2-year trial culminated in Edelin’s conviction of criminal
negligence (but notably, not murder or manslaughter) for not attempting to save a human life (the
human fetus). His publicized conviction was given a few weeks before the Quinlan case began.
1975 The case of Karen Quinlan: A 21-year-old in New Jersey went into a coma, possibly induced by 57
barbiturates. Family asked to withdraw mechanical ventilation, but the Catholic hospital, St Clare’s,
would not. The New Jersey Supreme Court voted in favor of the parents’ wish, and 10 years later,
Karen died. The American Medical Association (AMA) and the Catholic church at this time did not
recognize a difference between actively killing and withdrawing life support. This case is
monumental about decisions to dissociate life maintained by machines with life maintained by the
person alone.
1975 Since there were several state statutes that defined brain death, the American Bar Association (ABA) 12
announced in a position statement that: “for all legal purposes, a human body with irreversible
cessation of total brain function…shall be considered dead.”
1976 Barry Keene got the first advance directive legislation passed and California became the first state to 109
have such legislation (see “Natural Death Act”).
1976 United Kingdom code eliminated the repeat examination as necessary for brain death, and required a 6, 7
specific CO2 level during an apnea trial rather than time to determine that the apnea test did not
elicit medullary-driven respirations in the patient.
1977 U.S. Collaborative Study eliminated the apnea test and reintroduced a flat EEG and the repeat 8
examination, now at 30 to 60 minutes.
1980 Uniform Determination of Death Act is approved for the United States. It holds that irreversible 20
cessation of all functioning of the brain, of whole brain death, is human death, as is irreversible
cessation of circulatory and respiratory function.
1981 U.S. President’s Commission (a) supported the Uniform Determination of Death Act (b) and (a) 9
reinstituted the apnea test and a repeat examination. Also only required cerebral blood flow (b) 20
assessment if necessary to determine brain death
1983 The case of Elizabeth Bouvier: A 25-year-old paraplegic who also had little function in her arms, Ms. 57
Bouvier sought suicide by self-starvation and became a media-popularized figure of a daunting
struggle with life and wish for a right to die. She was force-fed by a court order, which she fought.
At the end of the fight, the court sympathized with Ms. Bouvier and made the notable statement:
“It is incongruous, if not monstrous for medical practitioners to assert their right to preserve a life
that someone else must…endure. No criminal or civil liability attaches to honoring a competent,
informed patient’s refusal for medical service.”
1990 AMA supports withdrawal of care for patients in Persistent Vegetative State (PVS). 58
1988 American Academy of Neurology (AAN) position on withdrawal of care in PVS cases, specifically noted 30
that physicians, unless an advance directive requests otherwise, have no obligation to administer
medical treatment, including artificial nutrition and hydration, if they cannot improve the patient’s
neurologic state.
1989 The case of Carrie Coons: An 86-year-old in New York regained consciousness after being in a 35, Ch 1
supposed state of irreversible unconsciousness (PVS). She “awoke” just before a court-ordered
removal of her feeding tube took place. This laid doubt on the accuracy of the PVS diagnosis.

Continued
64 Section I—Background

Table 8.2  Important Events in Defining Human Life and Death By Chronologic Order—cont’d
Year Event References
1989 The case of Nancy Cruzan: The first “right to death” case heard by the U.S. Supreme Court. A patient 95
in PVS, Cruzan’s family asked for her feeding tube to be removed but were voted down by the
Missouri Supreme Court based on the role of the “parens patria” of the state or, the guardian of
the incompetent, which must err on the side of life if it must err. The U.S. Supreme Court decided
that there was no evidence that Ms. Cruzan would have wanted to endure what the state of
Missouri was defining for her as her “life.”
1991 The state of New Jersey enacted a law that provided citizens with a religious exemption from brain 44
death determination.
1995 AAN criteria published that eliminated a flat EEG as necessary to declare brain death, and added 10
strict testing details for the apnea test, such as delivery of 100% oxygen to avoid harm from the
test itself.
1990-2005 The case of Terri Schiavo: After suffering from a cardiac arrest, Ms. Schiavo was comatose, 49
transitioning to PVS later. Her husband, based on his understanding of Ms Schiavo’s wishes, fought
against her parents to have her feeding tube removed. The court voted in favor of removing the
tube. The case reignited questions over what is sufficient to declare a being a living human, and
how proxies should be identified for the neurologically incompetent.

hypotheses, and recognize that absolute facts are not a reality. The distinctions made between cellular and clinical func-
Overall, the subjectivity of irreversible to time, technology, tion in brain death criteria point to an important fact some
and physicians’ best hypotheses leaves room for doubt, debate, still dispute: being alive is different from being a living human.
and fear of being incorrect. To elaborate, persons may die, but their bodies and the discon-
nected parts of their brain may still live, metabolize, grow, and
even carry out previously initiated human reproduction, as in
A Closer Look at the Definition: “Function” the case of brain-dead mothers.22,23 Bartlett and Youngner
In the ABA’s statement that “for all legal purposes, a human note that “life may go on after a person has died … [but death
body with irreversible cessation of total brain function…shall of a person] is a question of when we no longer [are] … a
be considered dead,”12 it is brain function rather than the living human being… the death of the organism [may] outlive
presence and viability of the brain or its cellular components the death of a person.”24 This, along with insufficient evalua-
that is cited as the necessary criterion for human life. This is tion of neurologic status25 has led to some confusion made
evident in medical criteria, as well. What is not obvious in obvious by Alan Shewmon’s declarations of “evidence” (i.e.,
this statement, but is in others and in medical brain-death organ survival) of human life (rather than just life) persisting
criteria, is that this loss of brain function refers to a loss of when brain-death criteria are “met.”26,27
clinical function, not cellular function. Certain statutes, such Through this chapter, book, and in day-to-day clinical prac-
as that modeled by Capron and Kass, speak of “relevant” brain tice, the reader is asked to keep in mind the difference between
function,19 making this point clear. Viable, functional cells no cellular and integrated, clinical function. Sometimes neuro-
doubt persist in cases of brain death, but the integration of monitoring provides output about one, the other, or both, and
these cells’ functions that confers the “relevant” brain activity thus must be appropriately weighted when considering its
sufficient to declare the patient a living human, does not.21 value in providing insight about the prognosis of maintaining
Bedside assessments can distinguish cellular from clinical meaningful human life.
function. In contrast to the use of in vitro analytical tools and
EEG to determine the viability and activity of cells, clinical Loosening Brain Death Criteria to Include
function is assessed by such things as pupil reactivity, other
brainstem reflexes, medullary response in the apnea test, and Other Disorders of Consciousness: Issues in
withdrawal from painful stimuli. Distinctions between clinical Diagnostics and Prognostics
function and neuronal function were made by the President’s Some clinical conditions with no or significantly impaired
Commission in 1981,9 because it was recognized that clinical higher brain function, or consciousness, do not fulfill the
functions are observable demonstrations of coordinated brain Harvard and similar criteria for brain death, but often are
activity that may irreversibly be lost even when particular areas argued as equivalent to human death or to irreversibly dying
of the brain remain viable. This report called for all U.S. juris- (see reference 28 for review).28 One such condition is the per-
dictions to accept the Uniform Determination of Death Act, manent or persistent vegetative state (PVS), in which higher
which equated irreversible cessation of all clinical function of cortical functions (including consciousness) are absent and
the brain with brain death.20 Consistent with this the American cannot be restored.‡29 Brainstem function remains intact. In
Academy of Neurology (AAN) criteria in 1995 eliminated a 1988, the AAN,30 echoed by the American Medical Association
flat EEG, a measure of cellular function, as necessary to declare
brain death.10 In one review, the Quality Standards Subcom- ‡
Posner suggests that no restoration is possible in the case of permanent (>1 year) vegeta-
tive state, but restoration may occur when the vegetative state is persistent (>1 month
mittee of the AAN found no published reports of recovery in <1 year). Some regard the use of “permanent” and “persistent” as confusing, and suggest
neurologic function in adults who fulfilled the diagnosis of these adjectives be replaced by a physician’s statement on the prognosis of the vegetative
brain death outlined by the 1995 AAN criteria.11 state.
Section I—Background 65

(AMA) in 1990,31 announced approval of withdrawing care increase prognostic accuracy. It also may improve diagnostic
from patients who met criteria for PVS, but did not specifi- accuracy and serve as a confirmatory test, which is deemed
cally endorse equating PVS to brain death. Hesitation to necessary by some neurologists.14 Specifically, neuromonitor-
extend the clinical definition of brain death to include PVS ing could identify events, such as cessation of cerebral blood
arises from one of two origins: (1) a belief that consciousness flow, that are indicative of severe brain dysfunction or hernia-
is not necessary for human life to persist, declaring brainstem tion and not compatible with life41 and other means of
function, with its role in physiologic homeostasis, as necessary irreversible neuronal destruction. Furthermore, because neu-
(and possibly sufficient) for human life; and (2) a fear that romonitoring can improve diagnostic and prognostic abili-
such an extension would increase the number of false-positive ties, it may simplify critical care ethics by relieving the
brain-dead diagnoses. The latter is the focus here because the physician of the burden of doubt. As prognostic and diagnos-
concept of consciousness being necessary for human life is tic tools are improved, however, caution is needed not to
discussed in detail elsewhere.14,24,32 veer too far from conservative criteria that add extra protec-
A fear of false positives is understandable given media- tion against false positives of brain death. For the moment
popularized stories of “miracles” in the ICU; a patient the concept of imminent brain death is based on clinical cri-
declared brain dead awakens to life. This leaves the patient’s teria including: (1) mechanically ventilated deeply comatose
loved ones distrustful of medical diagnostic criteria for life, patient, admitted to an ICU, with (2) irreversible catastrophic
and elicits inquiries from readers across the nation as to brain damage of known origin, and (3) a Glasgow Coma
whether their own deceased loved ones were truly, irreversibly Scale score of 3 and the progressive absence of at least three
dead. Unfortunately, no media story begins with “this is rarely of six brainstem reflexes or a Full Outline of UnResponsive-
the case, and is likely due to an error in diagnosis rather than ness (FOUR) score of Eyes (E), Motor (M), Brainstem (B),
a miracle, but … .” People recall the cases of individuals like Respiration (R).42 This definition may be useful in identifying
Carrie Coons, who in 1989 regained consciousness before a potential organ donors.
court-ordered act to remove her feeding tube took place.33
Similar stories are present in academic literature. In 2006,
Voss et al. wrote of a 39-year old male shown to be cortically
Futility
dead, but not dead by whole brain or brainstem criteria, who The end of human life has numerous conceptual and clinical
transitioned to the minimally conscious state (MCS).34 This definitions, but these definitions do not clarify the irreversible
may suggest that rather than improved accuracy to diagnose points that lead to this inevitable end. The theoretical concept
cortical death, conservative criteria that guarantee the pres- of futility seeks to identify these points, but there is no con-
ence of widespread, irreversible destruction, such as the sensus on a practical delineation of states or measures that
current whole brain death criteria,14 are necessary because strictly define futility. Differences in these practical definitions
patients in MCS may recover.35 In addition, studies using often depend on the religious beliefs and cultural practices
advanced magnetic resonance imaging (MRI) techniques that assign particular restrictions, definitions, and priorities to
including functional MRI (fMRI) have reframed questions life, death, and medical intervention.43 Governing bodies in
about consciousness and coma and the capacity of recovery the United States tend to be sensitive to these differences. In
in severe brain injury.36-38 1991 the state of New Jersey enacted a law that provided citi-
Conservative criteria may only mask the true problem; zens with a religious exemption from brain death determina-
diagnostic accuracy needs improvement. This problem was tion.44 In these cases only cardiopulmonary arrest is an
addressed by Andrews et al.39 who reported a misdiagnosis of acceptable criterion for death. Similarly, New York hospitals
40% of their 40 patients who were diagnosed as being in PVS must have a protocol that allows for a “reasonable accommo-
(they were actually in MCS). Patient blindness or severe motor dation [to] religious or moral objections” to neurologically
disability or insufficient patient observation, confusion with determined death, although, unlike in New Jersey, patients are
terminology used, and evaluation by inexperienced physicians not ubiquitously given power to veto brain-death determina-
were the most common reasons for the misdiagnosis. These tions.45 Although such culturally sensitive policies protect citi-
issues become relevant in the neurocritical care unit (NCCU) zens’ First Amendment right to free exercise of religion
where consciousness and neurologic function often may be (applicable to state law through the due process clause of the
altered by therapies designed to correct an abnormality 14th constitutional amendment),46,47 they limit physicians’
detected by a monitor. Inexperience with the neuromonitor ability to avoid futile measures.
can lead to potentially serious errors if something such as the In medicine, futility is most simply understood as any
absence of intracranial blood flow is confused with simply measure that will not sufficiently restore quality or quantity
decreased blood flow.40 In addition, it is unlikely that informa- of life-years. ||48 The AAN set forth three key considerations
tion provided by a monitor presently used in the NCCU, even for physicians when treatment may be considered futile (spe-
if that information is consistent with current clinical criteria cifically in the setting of PVS). They are as follows:
for brain death, will replace that criteria because monitors for
the most part assess one aspect of brain function. The infor- 1. A patient’s right to self-determination is central to the
mation from a monitor may be useful to decide when a medical, ethical, and legal principles relevant to medical
“brain-death” examination is necessary. treatment decisions.
If death is considered to be an event, not a process,32
neuromonitoring may lend insight to the intricacies of dying ||
It differs from rationing in that in determining whether a treatment is futile, the physi-
and the process that precedes the event of death.§ This could cian asks whether it will achieve a beneficial end, rather than if it is worth using a treat-
ment knowing its likely result weighed against its financial and resource consumption
§
Note that some view death itself as a process, rather than an event. (i.e., hospital beds) costs.
66 Section I—Background

2. A physician also must attempt to promote the patient’s legal jurisdiction in end-of-life decision making, and aim to
well-being, either by relieving suffering or addressing or avoid futile treatment.54 Although the legal system does not
reversing a pathologic process. Where medical treatment entirely coincide with these position statements and leaves
fails to promote a patient’s well-being, there is no longer loopholes for vetoing physicians’ and families’ decisions to
an ethical obligation to provide it. withdraw care if clear advance directives or unified proxy
3. Treatments that provide no benefit to the patient or the support for withdrawal are not available, historically, courts
family may be discontinued. Medical treatment, including have voted in favor of such withdrawal, so long as clear oppos-
the medical provision of artificial nutrition and hydration ing directives also are not available. This is consistent with
(ANH), provides no benefit to patients in a persistent veg- Americans’ constitutional First Amendment rights, which
etative state, once the diagnosis has been established to a grant a coveted luxury of society: autonomy, even when
high degree of medical certainty.30 autonomous wishes are known only from loved ones and sur-
rogates (see reference 55 for discussion).55 The Cruzan case
The second consideration is most important; physicians who was an important national precedent in establishing this right
cannot reduce suffering or stop a pathologic process are freed and is summarized in Table 8.2.
of an ethical “obligation” to treat the underlying pathologic Futility can be considered in relation to autonomous indi-
process (symptomatic or comfort-directed treatment, however, viduals, but it also has important implications for societal
is still appropriate)30 because such treatment would be futile. good. What is futile for one may be valuable to others. For
Physicians carry the burden to determine whether feeding or example, when life-sustaining ICU measures are considered
other means of support actually prolong death and not life moral practices, some futile interventions may offer ritualistic
(i.e., are futile).49 The only exception to this “rule” occurs if a benefit to patients, families, and ICU staff and so help facili-
patient’s advance directive, or known previously communi- tate the process of dying.56 An individual’s outcome may not
cated opinion, indicates a wish for continued treatment in the be improved by monitoring in the NCCU, but monitoring
setting of inevitable death (futile treatment). In this case, the may elucidate pathologic processes that underlie inevitable
patient’s wish must be upheld to protect his or her autonomy, death or a highly morbid state, and so could lead to improved
and here the importance of the physician-patient relationship treatment regimens and drug discovery that will benefit
and physician honesty must be emphasized.50 The AMA society. A comparison may be drawn to clinical drug trials in
echoed these positions a year after the AAN voiced them.31 oncology when death of the patient is inevitable, or, more
Because brain death defines an irreversible state given the closely, to body donation for medical education. Although
technology available, no current medical intervention can such use of neuromonitoring may be ethically sound so long
“improve” a brain-dead person’s well-being. Thus all medical as the proper intents and consent support it, caution must be
measures used in a brain-dead patient, including neuromoni- exhibited to not take an entirely utilitarian position. Such a
toring, are futile. Similarly, in PVS patients, it is assumed that position is antagonistic to John Stuart Mill’s belief that the
the patient will not regain consciousness, and so treatment is goal of moral actions is to create freedom and autonomy, not
futile.¶39,51 The war against futility often drives policy and posi- to further the good of any particular division of society.57 It
tion statements. This has been true for some time but contin- also is antagonistic to both the AMA’s policy (E-8.081), which
ues to stir controversy. An early example of academia that states that “any quality of life considerations should be mea-
convened to combat futility was when the Harvard Commis- sured as the worth to the individual whose course of treatment
sion put forth their set of brain dead criteria.4 Other examples is in question, and not as a measure of social worth,”58 and
include the AAN and AMA statements that support with- most importantly, to Americans’ First Amendment, constitu-
drawal of care when futile,30,31 all of which arose from intents tional rights.46
to avoid futile measures in PVS patients. Futile measures carry financial, emotional, and ethical costs
Several recent legislative bills, antagonistic to efforts to that neuromonitoring may either exacerbate by being futile
minimize futility, were drafted and introduced because of the itself or relieve by helping to identify futile treatments. It must
Terri Schiavo case.49,52 These bills aim to reverse the policy that be stressed that physicians are not obligated to take futile
favors withdrawal of artificial nutrition and hydration and measures, but that they may be pushing the ethical envelopes
other treatments when a physician believes medical measures if they do so. For example, hospital or ICU beds used by
are futile, and no advance directive dictates otherwise.53 This patients in whom measures are futile are not available to other
would transfer decisive power from physicians and families patients who require care. Thus rationing of care in the ICU
who have insight to the patient’s wishes to a detached govern- is necessary to maintain an ethical practice.59 Even in cases in
ing body, and would set the default to administering poten- which society may benefit from a futile action (e.g., neuro-
tially futile treatments. The AAN does not support this, as monitoring in a PVS patient), both lack of patient consent for
evidenced by its 1988 position statement,30 and its public continued care and a lack of financial capacity ethically disfa-
opposition in 2005 to “all state and federal legislation that vor futile treatment. Neuromonitoring in this case includes
would presume to prescribe the patient’s preferences for arti- continuous physiologic data but may also include serial “bio-
ficial hydration and nutrition in situations where the patient markers” collected over several days or sophisticated imaging
lacks an advance directive or living will.”31 The AMA adopted techniques (reviewed in Chapters 18 and 28). A balance is
this latter position to complement their policies that limit necessary because this information can help decide when
management is futile or help “predict” which patient may have

a “good” outcome or a “poor” outcome, the definition of
It is possible that the rare reports of patients diagnosed as in PVS, who later recovered which may depend on whether a caregiver or family member
consciousness outside of the probable timeframes for recovery of 3 months (nontrau-
matic) and 12 months (traumatic) originally may have been in MCS, and simply were is asked. For example, a family member may regard a good
misdiagnosed. outcome as recovery of functional communication even
Section I—Background 67

though the individual may remain physically dependant. NCCU, monitoring also may have the potential to provide
Functional communication recovery can be assessed through information about a condition that is still reversible and
the Coma Recovery Scale-Revised (CRS-R).60,61 before it becomes irreversible.
A feasible way to decrease the financial burden of patients Monitoring should, in theory, yield information on the
with disorders of consciousness requires improved diagnostic patient’s status that will direct surgical, medical, and personal
and prognostic assessments.62 This particularly benefits deci- management to maximize benefit tailored to the patient’s
sion making for those patients who meet criteria for the MCS individual wants. This should improve outcomes as viewed by
due to any etiology, and patients with a traumatic brain injury the patient and physician. The data, however, on how moni-
(TBI) that results in “stunned” neuronal tissue.63 Unlike brain toring in the NCCU affects outcome are mixed. Most of these
dead and PVS cases, MCS and TBI patients may have a pos- studies have addressed the role of intracranial pressure (ICP)
sible, but ill-defined chance of recovery28,51,62 that calls into monitors in TBI; some studies suggest outcome is not
question assessments of futility. Perhaps with improved prog- improved by monitoring.69-72 However, a meta-analysis and
nostic tools, the probability of such transitions with particular literature review that included more than 100,000 patients
interventions will be easier to assess. Neuromonitoring may suggests that adherence to guidelines for severe TBI and use
harbor such prognostic potential that would not only classify of an ICP monitor is associated with better outcome after
monitoring as valuable in MCS and TBI, rather than as a futile severe TBI.73,74 Consistent with this Whitmore et al.75 using a
measure in these cases, but also as a tool to avoid other futile decision-analytical model observed that “aggressive” care,
measures. These tools also may include sophisticated and including use of invasive monitoring, for TBI is associated
advanced neuroimaging techniques or biomarkers.64-67 This with better outcomes and is more cost effective than “routine”
would lighten the economic and thus ethical loads carried by care and even for older patients. In addition, potential bene-
potentially futile care. fits to use of a monitor may depend on the use of more than
Contrary to the beliefs of some, actions to reduce futility one monitor (i.e., better targeting of therapy).76 For example,
should not be viewed as mere attempts to increase organ in severe TBI information from a brain oxygen monitor
donation. Neuromonitoring should have the same aim to appears to provide valuable information about outcome
combat futility, but it should be recognized that even with this beyond that provided by an ICP monitor, both in adults and
as its aim, some people may perceive it as a tool used to children,77-79 and when the two monitors are used together,
achieve earlier and increased organ donation from patients. therapy based on that information may provide further
Physicians must explain with sincerity to patients, families, improvements in outcome.80 The role of monitoring in other
and the media that this is not its purpose. The recent birth of conditions treated in the NCCU is less well studied than for
this technology and society’s unfamiliarity with it may further TBI, in which again the combination of monitors may be
contribute to the family’s suspicion and hesitation to condone more useful than a single monitor alone. For example,
its use. Dohmen et al. and Heiss et al.81,82 observed that although use
of microdialysis, ICP, and tissue oxygen pressure could classify
the pathophysiology of a malignant middle cerebral artery
The Utility of Monitoring infarct, this monitoring did not predict it in time to perform
a hemicraniectomy, whereas positron emission tomography
Ethically Weighted by Prognostic (PET) imaging did.
Ambiguity and Futility These mixed results associated with how monitoring affects
The early evolution of neurocritical care as a distinct subspe- NCCU care may reflect several issues, including: (1) neuro-
cialty has occurred in parallel with the ability to monitor and monitoring only assesses a small portion of brain tissue82 and
image physiologic function of the human brain. What role thus may suggest a generalization that misrepresents the
does monitoring in the NCCU play in futility decisions? This patient’s condition (or it provides global information and
section discusses the practicality to avoid futile measures, misses the regional data that is more important); (2) the risks
while promoting individual patient’s wishes. The text first of an invasive device or how information provided by the
addresses the utility and limits of monitoring in the NCCU to device is used may outweigh the benefits; (3) user dependency
provide a sense of its potential (although this is discussed in or the learning curve may further skew the risk-to-benefit
greater depth in the chapters about each monitor), then con- ratio;83 (4) inexperience with what to do with the information
tinues with a conversation about shared decision making with obtained from neuromonitoring may lead to errors;40
the patient’s family in cases that may involve neuromonitor- (5) more information (that may mean more treatment) does
ing. At the outset it is important to realize that use of a not always mean better outcome; or (6) the information pro-
monitor depends primarily on the accuracy of interpretation vided by a monitor may indicate a treatment that is useful but
of the data provided and whether effective therapies or deci- the treatment then has unexpected adverse effects.84,85
sions based on that information exist and are validated. To date no level I (i.e., a randomized controlled trial) evi-
Monitoring in critical care may be of most use in the fol- dence demonstrates that a monitor (or monitors) is helpful in
lowing situations, suggested by Masdeu as indicative of a need the NCCU. This may be important because randomized
for ancillary medical technology and confirmatory tests: (1) studies in general critical care showed that pulmonary artery
when clinical diagnosis is uncertain, (2) when metabolic catheter use did not always contribute to better patient
derangements are present, (3) when the brainstem is selec- outcomes.86-88 However, it can be argued that randomized
tively damaged, (4) when evaluation of brainstem function is trials may not be the gold standard answer to clinical ques-
hindered (i.e., by facial trauma), (5) in young children, and tions in critical care,89,90 because much of what is done during
(5) as a confirmatory test for brain death.68 Studies are under day-to-day practice is based on understanding pathophysiol-
way to assess the prognostic value of neuromonitoring. In the ogy and how treatment may influence that pathophysiology.
68 Section I—Background

In many conditions managed in the NCCU and for many 16 years later in the Cruzan case, and continue to maintain a
therapies, this knowledge about pathophysiology is only presence in discussions.95
beginning to be elucidated in part through the use of neuro- How may providers ensure that neuromonitoring devices
monitoring. Hence an argument could be made for neuro- are viewed as simply ancillary tools for physicians rather
monitoring both for the patient being treated and for patients than as hardwired controllers of life? The first step is to
likely to be treated in the future. However, any technologic define the potential value of these tools. Second, clarification
advance can have unintended consequences and so create a about the purpose and value of these tools as they pertain to
pseudodisease that requires “treatment.”91 How these conse- the individual patient must be provided to proxies. Finally,
quences are viewed legally differs from how they are seen in quality assessment of the ethical conduct displayed when
health care: in medicine results, conclusions, and recommen- neuromonitoring-related decisions are made, as is suggested
dations about care are subject to continuing review—they can for all practices in medicine,55 may motivate ethical behavior
change over time. By contrast, the courts make conclusions and boost confidence in the ethical use of these technolo-
only once, at trial.92 gies. These efforts toward individualizing patient care must
Because the prognostic and diagnostic value of some aspects be conducted with the overarching goal to avoid all mea-
of neuromonitoring still remains to be clearly defined, and sures the patient would deem futile, and pursue all measures
because neuromonitoring may be a futile measure in some the patient would deem worthwhile.
patients, the utility of neuromonitoring must be empirically In the NCCU, a patient’s wishes most often are unknown.
studied, and families must be well informed of its potential These preferences need to be considered when neuromoni-
limits and benefits. Physicians are more apt to individually toring is used for personalized end-of-life care. These prefer-
make decisions with little to no invited input from families ences are difficult to determine, in part because this is a new
and patients when the ambiguity of the utility of a medical set of technologies that yields a new set of data and more
treatment or tool is great.93 This is yet another motivator to generally because few persons have advance directives even
clarify the value and use of neuromonitoring: to allow a bal- though they were conceptually conceived in 1969.55,96 Even
anced conversation to occur between families and physicians when advance directives are available, their specifications can
to decide on treatments for loved ones.94 be cryptic and sometimes they are disregarded. State-to-state
variability in legal regulation of directives can create addi-
tional confusion for patients and physicians. Efforts to publi-
When and How to Use cize and standardize the collection and use of advance
Neuromonitoring in the Context directives49 are therefore worthwhile. A federal statute, the
of Patients’ and Their Families’ Patient Self-Determination Act that mandates that covered
medical institutions actively distribute, collect, and honor
Needs and Wants information on their protocol for handling advance direc-
Because neuromonitoring may provide more insight about a tives, has not been sufficient. This suggests there is a need to
patient’s prognosis, the family and treatment team may make fix the protocol, not just publicize it.55 The Uniform Health-
a more informed, patient-centric decision. A few issues can Care Decisions Act of 1993 represents an effort worth atten-
complicate this promising thought. Neuromonitoring itself tion.97 This act lends clarity, not just motivation, to the
may seem to represent a shift from personal, patient-oriented proper collection use of advance directives consent, advance
care to generalized, machine-controlled care. Since ventilators directives, and appointment of durable power of attorneys in
were introduced to the ICU in the 1950s and 1960s, talk of a manner that likely will best promote the patient’s presumed
machines, not humans, carrying out life has permeated ethical wishes. Both the AARP (formerly the American Association
literature. The impact of machines on the definitions, assess- of Retired Persons) and the ABA endorsed this act, but
ment, and perceptions of life and death is obviously great (see only a fraction of states have adopted it (see discussion in
Table 8.2). reference 49).49
The recognition of the technologic dependency of life and As our legal system progresses to meet patients’ needs, spe-
death is evident in legal documents and cases, including the cifically those prominent in disorders of consciousness, direct
statement by a Florida Court of Appeals judge of the Cruzan reference to decisions that involve neuromonitoring and the
case,95 who noted that “it may be determined by reason of the data it provides should perhaps be included. This is worth
advanced scientific and medical technologies of this day that careful thought, because the system to identify suitable
Life has, through causes beyond human control, reached the proxies for these patients is often insufficient and how sur-
unconscious and vegetative state where all that remains is the rogates view futility or decide about care may have implica-
forced function of the body’s vital functions.” Many ethically tions for health care providers. For example, one study
weighted court cases (see reference 57, case of Karen Quinlan, indicates that proxies did no better at fulfilling the neuro­
as state precedent; reference 95, case of Nancy Cruzan as logically compromised patient’s wishes than chance alone,98
national precedent; and reference 55, for summary) that fol- whereas other studies indicate that up to two thirds of
lowed publication of the Harvard Criteria4 for brain death surrogates express doubts about the accuracy of futility
often centered on what should be done with these life- predictions.99
supporting (or death-prolonging, so as not to suggest bias) Many legal and ethical obstacles complicate the process to
machines. The case of Karen Quinlan,57 a young woman in a choose a sufficient proxy for consent of three areas that
PVS whose parents desired to withdraw machine support may concern neuromonitoring: treatment, research, or with-
(see Table 8.2), was specifically referenced as evidence of dep- drawal of care. First, potential proxies must be deemed suit-
ersonalized, machine-controlled medicine.57 Perceptions of able to act as the legal representative of the impaired person,
machine-dependent medicine and life permeated the dialogue and be willing to do so. In a study by Mason et al., it was
Section I—Background 69

found that a large number of impaired persons did not have published resources that the family can use to expand their
a suitable proxy, and among those who did, the majority of knowledge.
the potential proxies were not willing to fill this role.100 To
claim a proxy as “suitable” a person must determine the An inability of physicians to clearly and fully address futility
motives behind the proxy’s decision, and ensure their choice to families means families may insist on increased end-of-life
is based on their best estimation of what the patient would care, prolong emotional suffering by the family, and render a
want. This assumes that the proxy understands the concep- poor reputation to the hospital as one that carelessly spends
tual basis for the treatment, comfort measures only (CMO) patients’ money without an impact on outcomes. This is par-
status, experimental therapies, or other routes being pursued. ticularly concerning because Consumer Reports now lists such
Second, it must be confirmed that the proxy will not acquire figures as average cost and intensity of end-of-life care for the
a secondary gain by prolonging or hastening an inevitable public to compare hospitals.104 In end-of-life care, mediation
death (e.g., financial, property, capitalizing on family dis- between families and the physician usually is successful in
agreements). In the case of multiple proxies, “disagreements resolving conflict if it exists, although it still is to be fully
among proxies with equal priorities” may arise101 and pose elucidated on how to optimally use an ethics consult in neuro-
yet another obstacle in this process. Finally, the legal specifics critical care.105,106 This can be important because there are
of proxy consent are similar to brain death criteria32,55,56 differences in how the public and health care professionals
in that they are under the jurisdiction of the state, and view futility. For example, Jacobs et al.107 in a survey of the
there is much heterogeneity between and confusion within general public and of trauma professionals found that more
states.55,102 Similarly there is significant variability in institu- of the public (57.4%) than the professionals (19.5%) believe
tional review board surrogate consent practices and what that divine intervention may save a person when treatment
risk is acceptable in research on incapacitated adults.103 was thought to be futile.
Indeed, there are limits#55 to studies that investigate the A final communicative issue that is more specific to neuro-
success of proxy appointments, but what is consistent with monitoring than other medical modalities used near the end
most of them is that the decision-making process for the of life concerns families and legal authorities who may misin-
neurologically incompetent patient is inadequate. Still, terpret data from neuromonitoring. This misinterpretation
current protocols for decision making in futile cases (see ref- may result in confusion, anger, and poor substituted judgment
erence 49 for review) offer a usable framework for decisions by the proxy. Physicians should be clear and complete in
about neuromonitoring. explaining the output of the monitors to prevent this confu-
There likely will be times when the treatment team believes sion. If misinterpretation-driven frustration still arises, the
that neuromonitoring is not indicated and views monitoring physician should again make a personal attempt to clarify the
as futile, but the family insists on its use (i.e., there is dis- situation and, if need be, call on a hospital ethicist if his or her
agreement with the perception of futility). In these circum- attempt fails.
stances, neuromonitoring may be viewed in the same light as
artificial nutrition and hydration, antibiotics, and other
medical treatments. Only so long as information provided by
Conclusion
monitoring can help prolong or improve life, in the physi- In any conversations about neuromonitoring, the relation of
cian’s best opinion, is the physician obligated to use it, unless these tools to ethical issues relevant to critical care must be
a patient’s advance directive or informally expressed wishes addressed. The most pertinent is futility. This chapter defined
request otherwise.49 The origin of the family’s request also futility, describing irreversible and reversible points that may
must be acknowledged and responded to in the following progress to the ultimate irreversible end—human death, with
cases: a definition that is still evolving. It also addressed the ethical
context of futile measures in medicine, bound by economical
1. If the family seems to harbor false hope for a recovery, or limits, and ethically weighted by how these limits restrict
if the family seems to be in denial, the physician should medical care for other patients. It recognized that neuromon-
clearly reiterate the prognosis and diagnosis. itoring, like other technologies in critical care, is ethically
2. If the family is simply unwilling to “give up,” the physician complicated and entwined with issues of brain death and
must assure them that they have done all they can, and futility given its impact on the quality and length of human
that further efforts are not in the best interest of the life. The potential for neuromonitoring to lighten ethical
patient. The physician also should comfort the family by burdens also was discussed, specifically, that brain death,
telling them that by withdrawing monitors, the family is along with states of less severe neurologic dysfunction (i.e.,
not giving up on their loved one, but rather, accepts that PVS and MCS) may be more accurately predicted, assessed,
the monitors simply do not provide data to benefit the and diagnosed with neuromonitoring. This could help
patient. prevent costly futile measures, or premature withdrawal of
3. If the family appears to have a desire for knowledge about care. Finally, it was stressed that the full impact of neuro-
the etiology of the patient’s state, physicians should help monitoring has yet to be realized. It is particularly important
them find closure in some other knowledge-based way. An that as this realization comes to fruition, as the prognostic
example of this is to share the most complete information value of neuromonitoring improves, caution be expressed.
available that is relevant to the patient’s condition, offer Overconfidence in neuromonitoring’s ability to predict a
one’s contact information for further questions, and then poor outcome may lead to hasty decisions that surrender
direct families to support and educational groups that have human life before its end is inevitable. This possibility is
expressed by the court-appointed guardian for Karen
#
Limits include variable methodology in data collection and assessment. Quinlan, Coburn, who said, “One human being, by conduct
70 Section I—Background

or lack of conduct, is going to cause the death of another 21. Bernat J. Philosophical and ethical aspects of brain death. In: Wijdicks EFM,
editor. Brain death. Philadelphia: Lippincott Williams & Wilkins; 2001. p.
human being.”57 Overconfidence also could lead to misguided
171–88.
rationing, and physicians may suggest against care they incor- 22. Brain dead mother is taken off life support. Available at: www.cbsnews.com/
rectly deem futile based on data from a monitor. This may be stories/2005/06/17/health/main702568.shtml and www.washingtonpost.com/
less likely when multimodality monitoring is used. On the wp-dyn/content/article/2005/08/03/AR2005080300224.html (accessed
other hand, inexperience with monitoring increases these September 2009).
23. Posner J. Alleged awakenings from prolonged coma and brain death and
risks,‡‡39 and a lack of clear patient directives about end-of- delivery of live babies from brain-dead mothers do not negotiate brain
life care complicates decision making. death. In: Marcelo Sanchez Sorondo HE, editor. The signs of death: the
Finally, physicians must understand patient and family proceedings of the working group. Scripta varia 110. Rome: Pontificia
values, concerns, and views of monitoring in the NCCU. Par- Academia Sciantiarvm; 2006.
24. Bartlett E, Youngner S. Human death and the destruction of the neocortex.
ticular attention must be paid to ensure that patients are not
In: Zaner RN, editor. Death: beyond whole-brain criteria. Dordrecht, NL:
viewed as hostages to machines, but rather that these tools Kluwer Academic Publishers; 1988. p. 199–215.
enhance patient-centric care and decision making. 25. Wang MY, Wallace P, Gruen JB. Brain death documentation: analysis and
issues. Neurosurgery 2002;51:731–5.
References 26. Shewmon DA. Chronic ‘brain death’ meta-analysis and conceptual
consequences. Neurology 1998;51:1538–45.
1. Daroff R. The historical evolution of brain death from former definitions of 27. Repertinger S, et al. Long survival following bacterial meningitis-associated
death: the Harvard Criteria to the present. In: Marcelo Sanchez Sorondo HE, brain destruction. J Child Neurol 2006;21(7):591–5.
editor. The signs of death: the proceedings of the working group. Scripta 28. Owen A. Disorders of consciousness. Ann N Y Acad Sci 2008;1124:
varia 110. Rome: Pontificia Academia Sciantiarvm; 2006. p. 217–21. 225–38.
2. Hacke W. Brain death: an artifact created by critical care medicine or the 29. Group discussion. In: Marcelo Sanchez Sorondo HE, editor. The signs of
death of the brain has always been the death of the individuum. In: Marcelo death: the proceedings of the working group. Scripta varia 110. Rome:
Sanchez Sorondo HE, editor. The signs of death: the proceedings of the Pontificia Academia Sciantiarvm; 2007. p. 21–3.
working group. Scripta varia 110. Rome: Pontificia Academia Sciantiarvm; 30. Certain aspects of the care and management of the persistent vegetative state
2006. p. 84–91. patient. AAN position statement adopted on April 21, 1988. Available at
3. Jennett B. Foreword. In: Wijdicks EFM, editor. Brain death. Philadelphia: www.aan.com/about/ethics/109556.pdf.
Lippincott Williams & Wilkins; 2001. p. IX–X. 31. Persistent vegetative state and the decision to withdraw or withhold life
4. Ad Hoc Committee of the Harvard Medical School. A definition of support. Council on Scientific Affairs and Council on Ethical and Judicial
irreversible coma. JAMA 1968;205:337–40. Affairs. JAMA 1990;263(3):426–30.
5. Mohandas A, Chou SN. Brain death—a clinical and pathologic study. 32. Bernat J, Culver CM, Gert B. On the definition and criterion of death. Ann
J Neurosurg 1971;35:211–18. Intern Med 1981;94:389–94.
6. Conference of Medical Royal Colleges and Their Facilities in the United 33. Steinbock B. Recovery from the persistent vegetative state? The case of Carrie
Kingdom. Diagnosis of brain death. BMJ 1976:1187–8. Coons. Hastings Cent Rep 1989;19:14–5.
7. Conference of Medical Royal Colleges and Their Facilities in the United 34. Voss H, Uluc AM, Dyke JP. Possible axonal regrowth in late recovery from
Kingdom. Diagnosis of brain death. Lancet 1976:1069–70. the minimally conscious state. J Clin Invest 2006;116(7):2005–11.
8. U.S. Collaborative Study. An appraisal of the criteria of cerebral death. JAMA 35. Lammi MH, Smith VH, Tate RL, et al. The minimally conscious state and
1977;237:982–6. recovery potential: a follow-up study 2 to 5 years after traumatic brain
9. U.S. President’s Commission. Guidelines for the determination of death. injury. Arch Phys Med Rehabil 2005;86(4):746–54.
JAMA 1981;246:2184–6. 36. Laureys S, Schiff ND. Coma and consciousness: paradigms (re)framed by
10. American Academy of Neurology. Practice parameters for determining brain neuroimaging. Neuroimage 2011;Dec 27. Epub ahead of print.
death in adults. Neurology 1995;45:1012–14. 37. Fins JJ, Schiff ND, Foley KM. Late recovery from the minimally conscious
11. Wijdicks EF, Varelas PN, Gronseth GS, et al. Evidence-based guideline state: ethical and policy implications. Neurology 2007;68(4):304–7.
update: determining brain death in adults: report of the Quality Standards 38. Giacino JT, Schnakers C, Rodriguez-Moreno D, et al. Behavioral assessment
Subcommittee of the American Academy of Neurology. Am Acad Neurol in patients with disorders of consciousness: gold standard or fool’s gold?
2010;74(23):1911–18. Prog Brain Res 2009;177:33–48.
12. House of Delegates redefines death, urges redefinition of rape, and undoes 39. Andrews K. Recovery of patients after four months or more in the persistent
the Houston amendments. Am Bar Assoc J 1975;61:463–4. vegetative state. BMJ 1993;306(6892):1597–600.
13. Veith FJ, Fein JM, Tendler MD. Brain death: a status report of medical and 40. Coimbra CG. Implications of ischemic penumbra for the diagnosis of brain
ethical considerations. JAMA 1977;238:1651–5. death. Braz J Med Biol Res 1999;32:1479–87.
14. Bernat J. How do physicians prove irreversibility in the determination of 41. Plum F, Posner JB. The diagnosis of stupor and coma. 3rd ed. Philadelphia:
death? In: Marcelo Sanchez Sorondo HE, editor. The signs of death: the FA Davis Co; 1980. p. 88–101.
proceedings of the working group. Scripta varia 110. Rome: Pontificia 42. de Groot YJ, Jansen NE, Bakker J, et al. Imminent brain death: point of
Academia Sciantiarvm; 2006. p. 159–76. departure for potential heart-beating organ donor recognition. Intensive
15. Oxford English Dictionary. 2nd ed. Oxford: Oxford University Press. 2006. Care Med 2010;36(9):1488–94. Epub 2010, Mar 16.
16. Cole D. Statutory definitions of death and the management of terminally ill 43. Gallagher C, Wijdicks EFM. Religious and cultural aspects of brain death. In:
patients who may become organ donors after death. Kennedy Inst Ethics J Wijdicks EFM, editor. Brain death. Philadelphia: Lippincott Williams &
1993;3(2):145–55. Wilkins; 2001. p. 135–50.
17. Aulisio MP, Devita M, Luebke D. Taking values seriously: ethical challenges 44. Olick RS. Brain death, religious freedom, and public policy: New Jersey’s
in organ donation and transplantation for critical care professionals. Crit landmark legislative initiative. Kennedy Inst Ethics J 1991:275–88.
Care Med 2007;35(2 Suppl):S95–101. 45. NY comp codes, rules & regs, Title 10, section 400.16(d), (e)(3) (1992)
18. Ropper A. The apnea test and rationale for brain death as death. In: Marcelo 46. U.S. Const. Amend 1
Sanchez Sorondo HE, editor. The signs of death: the proceedings of the 47. U.S. Const. Amend 14
working group. Scripta varia 110. Rome: Pontificia Academia Sciantiarvm; 48. Truog R, Burns J. Futility: a concept in evolution. Chest 2007;132(6):
2006. p. 237–49. 1987–93.
19. Capron AM, Kass LR. A statutory definition of the standards for determining 49. Bacon D, Williams MA, Gordon J. Position statement on laws and
human death: an appraisal and a proposal. Univ Penn Law Rev regulations concerning life-sustaining treatment, including artificial
1972;121:87–118. nutrition and hydration, for patients lacking decision-making capacity.
20. Uniform Determination of Death Act, Uniform Laws Annot 12A:589 Neurology 2007;68:1097–100.
www.law.upenn.edu/bll/archives/ulc/fnact99/1980s/udda80.htm. 50. Bitsori M, Georgopoulos D, Galanakis E. The question of futility and Roger
C. Bone. 2009;12(4):477–81. Epub 2009, Mar 4.

‡‡
Inexperience was one of the top five reasons for the misdiagnosis of PVS in a set of A complete list of references for this chapter can be found online at
MCS patients. www.expertconsult.com.
Section I—Background 70.e1

References 29. Group discussion. In: Marcelo Sanchez Sorondo HE, editor. The signs of
death: the proceedings of the working group. Scripta varia 110. Rome:
1. Daroff R. The historical evolution of brain death from former definitions Pontificia Academia Sciantiarvm; 2007. p. 21–3.
of death: the Harvard Criteria to the present. In: Marcelo Sanchez Sorondo 30. Certain aspects of the care and management of the persistent vegetative
HE, editor. The signs of death: the proceedings of the working group. state patient. AAN position statement adopted on April 21, 1988. Available
Scripta varia 110. Rome: Pontificia Academia Sciantiarvm; 2006. p. 217–21. at www.aan.com/about/ethics/109556.pdf.
2. Hacke W. Brain death: an artifact created by critical care medicine or the 31. Persistent vegetative state and the decision to withdraw or withhold life
death of the brain has always been the death of the individuum. In: support. Council on Scientific Affairs and Council on Ethical and Judicial
Marcelo Sanchez Sorondo HE, editor. The signs of death: the proceedings Affairs. JAMA 1990;263(3):426–30.
of the working group. Scripta varia 110. Rome: Pontificia Academia 32. Bernat J, Culver CM, Gert B. On the definition and criterion of death. Ann
Sciantiarvm; 2006. p. 84–91. Intern Med 1981;94:389–94.
3. Jennett B. Foreword. In: Wijdicks EFM, editor. Brain death. Philadelphia: 33. Steinbock B. Recovery from the persistent vegetative state? The case of
Lippincott Williams & Wilkins; 2001. p. IX–X. Carrie Coons. Hastings Cent Rep 1989;19:14–5.
4. Ad Hoc Committee of the Harvard Medical School. A definition of 34. Voss H, Uluc AM, Dyke JP. Possible axonal regrowth in late recovery from
irreversible coma. JAMA 1968;205:337–40. the minimally conscious state. J Clin Invest 2006;116(7):2005–11.
5. Mohandas A, Chou SN. Brain death—a clinical and pathologic study. 35. Lammi MH, Smith VH, Tate RL, et al. The minimally conscious state and
J Neurosurg 1971;35:211–18. recovery potential: a follow-up study 2 to 5 years after traumatic brain
6. Conference of Medical Royal Colleges and Their Facilities in the United injury. Arch Phys Med Rehabil 2005;86(4):746–54.
Kingdom. Diagnosis of brain death. BMJ 1976:1187–8. 36. Laureys S, Schiff ND. Coma and consciousness: paradigms (re)framed by
7. Conference of Medical Royal Colleges and Their Facilities in the United neuroimaging. Neuroimage 2011;Dec 27. Epub ahead of print.
Kingdom. Diagnosis of brain death. Lancet 1976:1069–70. 37. Fins JJ, Schiff ND, Foley KM. Late recovery from the minimally conscious
8. U.S. Collaborative Study. An appraisal of the criteria of cerebral death. state: ethical and policy implications. Neurology 2007;68(4):304–7.
JAMA 1977;237:982–6. 38. Giacino JT, Schnakers C, Rodriguez-Moreno D, et al. Behavioral assessment
9. U.S. President’s Commission. Guidelines for the determination of death. in patients with disorders of consciousness: gold standard or fool’s gold?
JAMA 1981;246:2184–6. Prog Brain Res 2009;177:33–48.
10. American Academy of Neurology. Practice parameters for determining 39. Andrews K. Recovery of patients after four months or more in the
brain death in adults. Neurology 1995;45:1012–14. persistent vegetative state. BMJ 1993;306(6892):1597–600.
11. Wijdicks EF, Varelas PN, Gronseth GS, et al. Evidence-based guideline 40. Coimbra CG. Implications of ischemic penumbra for the diagnosis of brain
update: determining brain death in adults: report of the Quality Standards death. Braz J Med Biol Res 1999;32:1479–87.
Subcommittee of the American Academy of Neurology. Am Acad Neurol 41. Plum F, Posner JB. The diagnosis of stupor and coma. 3rd ed. Philadelphia:
2010;74(23):1911–18. FA Davis Co; 1980. p. 88–101.
12. House of Delegates redefines death, urges redefinition of rape, and undoes 42. de Groot YJ, Jansen NE, Bakker J, et al. Imminent brain death: point of
the Houston amendments. Am Bar Assoc J 1975;61:463–4. departure for potential heart-beating organ donor recognition. Intensive
13. Veith FJ, Fein JM, Tendler MD. Brain death: a status report of medical and Care Med 2010;36(9):1488–94. Epub 2010, Mar 16.
ethical considerations. JAMA 1977;238:1651–5. 43. Gallagher C, Wijdicks EFM. Religious and cultural aspects of brain death.
14. Bernat J. How do physicians prove irreversibility in the determination of In: Wijdicks EFM, editor. Brain death. Philadelphia: Lippincott Williams &
death? In: Marcelo Sanchez Sorondo HE, editor. The signs of death: the Wilkins; 2001. p. 135–50.
proceedings of the working group. Scripta varia 110. Rome: Pontificia 44. Olick RS. Brain death, religious freedom, and public policy: New Jersey’s
Academia Sciantiarvm; 2006. p. 159–76. landmark legislative initiative. Kennedy Inst Ethics J 1991:275–88.
15. Oxford English Dictionary. 2nd ed. Oxford: Oxford University Press. 2006. 45. NY comp codes, rules & regs, Title 10, section 400.16(d), (e)(3) (1992)
16. Cole D. Statutory definitions of death and the management of terminally ill 46. U.S. Const. Amend 1
patients who may become organ donors after death. Kennedy Inst Ethics J 47. U.S. Const. Amend 14
1993;3(2):145–55. 48. Truog R, Burns J. Futility: a concept in evolution. Chest 2007;132(6):
17. Aulisio MP, Devita M, Luebke D. Taking values seriously: ethical challenges 1987–93.
in organ donation and transplantation for critical care professionals. Crit 49. Bacon D, Williams MA, Gordon J. Position statement on laws and
Care Med 2007;35(2 Suppl):S95–101. regulations concerning life-sustaining treatment, including artificial
18. Ropper A. The apnea test and rationale for brain death as death. In: nutrition and hydration, for patients lacking decision-making capacity.
Marcelo Sanchez Sorondo HE, editor. The signs of death: the proceedings Neurology 2007;68:1097–100.
of the working group. Scripta varia 110. Rome: Pontificia Academia 50. Bitsori M, Georgopoulos D, Galanakis E. The question of futility and Roger
Sciantiarvm; 2006. p. 237–49. C. Bone. 2009;12(4):477–81. Epub 2009, Mar 4.
19. Capron AM, Kass LR. A statutory definition of the standards for 51. Medical aspects of the persistent vegetative state. The Multi-Society Task
determining human death: an appraisal and a proposal. Univ Penn Law Rev Force on PVS. N Engl J Med 1994;330(22):1572–9.
1972;121:87–118. 52. Terri Schiavo case. In: Phelps S, editor. Gale encyclopedia of everyday
20. Uniform Determination of Death Act, Uniform Laws Annot 12A:589 law. Gale Group, 2003. eNotes.com. Available at http://www.enotes.com/
www.law.upenn.edu/bll/archives/ulc/fnact99/1980s/udda80.htm. topic/Terri_Schiavo_case. Accessed July 26, 2012 (accessed September
21. Bernat J. Philosophical and ethical aspects of brain death. In: Wijdicks 2009).
EFM, editor. Brain death. Philadelphia: Lippincott Williams & Wilkins; 53. MergerWatch. Summary of state bills restricting end-of-life choices
2001. p. 171–88. introduced in 2005 and 2006. March 28. Available online at
22. Brain dead mother is taken off life support. Available at: www.mergerwatch.org/pdfs/fs_end_of_life_legislation.pdf, 2006 (accessed
www.washingtonpost.com/wp-dyn/content/article/2005/08/03/ September 2009).
AR2005080300224.html (accessed September 2009). 54. Resolution 209 (A-05), as introduced at the 2005 Annual Meeting of the
23. Posner J. Alleged awakenings from prolonged coma and brain death and American Medical Association. Available online at www.ama-assn.org/
delivery of live babies from brain-dead mothers do not negotiate brain meetings/public/annual05/209a05.doc.
death. In: Marcelo Sanchez Sorondo HE, editor. The signs of death: the 55. Wolf SM, et al. Sources of concern about the Patient Self-Determination
proceedings of the working group. Scripta varia 110. Rome: Pontificia Act, 325. New Engl J Med 1991;1666–71.
Academia Sciantiarvm; 2006. 56. Mohammed S, Peter E. Rituals, death and the moral practice of medical
24. Bartlett E, Youngner S. Human death and the destruction of the neocortex. futility. Nurs Ethics 2009;16(3):292–302.
In: Zaner RN, editor. Death: beyond whole-brain criteria. Dordrecht, NL: 57. Pence GE. Classical cases in medical ethics: accounts of the cases that have
Kluwer Academic Publishers; 1988. p. 199–215. shaped medical ethics with philosophical, legal, and historical backgrounds.
25. Wang MY, Wallace P, Gruen JB. Brain death documentation: analysis and New York: McGraw-Hill, 1990. Ch 1-2.
issues. Neurosurgery 2002;51:731–5. 58. YPS delegate’s report, AMA Annual Meeting, June 17–22, 2005, Chicago, IL.
26. Shewmon DA. Chronic ‘brain death’ meta-analysis and conceptual Available at www.ama-assn.org/ama1/pub/upload/mm/17/
consequences. Neurology 1998;51:1538–45. a05delegatesreport.pdf.
27. Repertinger S, et al. Long survival following bacterial meningitis-associated 59. Truog RD, Brock DW, Cook DJ, et al. For the Task Force on Values, Ethics,
brain destruction. J Child Neurol 2006;21(7):591–5. and Rationing in Critical Care (VERICC). Rationing in the intensive care
28. Owen A. Disorders of consciousness. Ann N Y Acad Sci 2008;1124:225–38. unit. Crit Care Med 2006;34(4):958–63.
70.e2 Section I—Background

60. Giacino JT, Kalmar K, Whyte J. The JFK Coma Recovery Scale-Revised: 85. Contant CF, Valadka AB, Gopinath SP, et al. Adult respiratory distress
measurement characteristics and diagnostic utility. Arch Phys Med Rehabil syndrome: a complication of induced hypertension after severe head injury.
2004;85:2020–9. J Neurosurg 2001;95(4):560–8.
61. Maas AI, Harrison-Felix CL, Menon D, et al. Common data elements for 86. Harvey S, Harrison DA, Singer M, et al. Assessment of the clinical
traumatic brain injury: recommendations from the interagency working effectiveness of pulmonary artery catheters in management of patients in
group on demographics and clinical assessment. Arch Phys Med Rehabil intensive care (PAC-Man): a randomised controlled trial. Lancet
2010;91:1641–9. 2005;366(9484):472–7.
62. Giacino JT. The minimally conscious state: definition and diagnostic 87. Richard C, Warszawski J, Anguel N, et al. Early use of the pulmonary artery
criteria. Neurology 2002;58:349–53. catheter and outcomes in patients with shock and acute respiratory distress
63. Hennerici W. Surviving areas of brain tissue in brain death: is the whole syndrome: a randomized controlled trial. JAMA 2003;290(20):2713–20.
more than the sum of its parts? In: Marcelo Sanchez Sorondo HE, editor. 88. Sandham JD, Hull RD, Brant RF, et al. A randomized, controlled trial of the
The signs of death: the proceedings of the working group. Scripta varia 110. use of pulmonary-artery catheters in high-risk surgical patients. N Engl J
Rome: Pontificia Academia Sciantiarvm; 2006. p. 101–14. Med 2003;348(1):5–14.
64. Lescot T, Galanaud D, Puybasset L. Exploring altered consciousness states 89. Ospina-Tascon GA, Buchele GL, Vincent JL. Multicenter, randomized,
by magnetic resonance imaging in brain injury. Ann N Y Acad Sci controlled trials evaluating mortality in intensive care: doomed to fail? Crit
2009;1157:71–80. Care Med 2008;36:1311–22.
65. Galanaud D, Puybasset L. Cardiac arrest: has the time of MRI come? Crit 90. Vincent JL. We should abandon randomized controlled trials in the
Care 2010;14(2):135. Epub 2010, Mar 26. intensive care unit. Crit Care Med 2010;38(Suppl):S534–8.
66. Tshibanda L, Vanhaudenhuyse A, Galanaud D, et al. Magnetic resonance 91. Freeman JM. Beware: the misuse of technology and the law of unintended
spectroscopy and diffusion tensor imaging in coma survivors: promises and consequences. Neurotherapeutics 2007;4(3):549–54.
pitfalls. Prog Brain Res 2009;177:215–29. 92. Dickens BM, Cook RJ. The legal effects of fetal monitoring guidelines. Int J
67. Siman R, Toraskar N, Dang A, et al. A panel of neuron-enriched proteins as Gynaecol Obstet 2010;108(2):170–3. Epub 2009, Nov 22.
markers for traumatic brain injury in humans. J Neurotrauma 93. Parascandola M, Hawkins J, Danis M. Patient autonomy and the challenge
2009;26(11):1867–77. of clinical uncertainty. Kennedy Inst Ethics J 2002;12(3):245–64.
68. Masdeu J. Neuroimaging: a window into total brain destruction and the 94. Bernat J. Ethical aspects of determining and communicating prognosis in
unresponsive states. In: Marcelo Sanchez Sorondo HE, editor. The signs of critical care. Neurocritical Care 2004;1(1):L107–17.
death: the proceedings of the working group. Scripta varia 110. Rome: 95. Cruzan, by her parents and co-guardians, Cruzan et ux. v. Director,
Pontificia Academia Sciantiarvm; 2006. p. 222–34. Missouri Department of Health, et al. No. 88-1503 Supreme Court of the
69. Idris Z, Ghani RI, Musa KI, et al. Prognostic study of using different United States497 U.S. 261; 110 S. Ct. 2841; 111 L. Ed. 2d 224; 1990 U.S.
monitoring modalities in treating severe traumatic brain injury. Asian J LEXIS 3301; 58 U.S.L.W. 4916
Surg 2007;30(3):200–8. 96. Brown BA. The history of advance directives: a literature review. J Gerontol
70. Steiner LA, Andrews PJ. Monitoring the injured brain: ICP and CBF. Br J Nurs 2003;29(9):4–14.
Anaesth 2006;97(1):26–38. 97. Franck M, Fry TP, Hill MK, et al. Uniform Health Care Decisions Act.
71. Cremer OL, van Dijk GW, van Wensen E, et al. Effect of intracranial Available at: www.law.upenn.edu/bll/archives/ulc/fnact99/1990s/
pressure monitoring and targeted intensive care on functional outcome uhcda93.htm (accessed July 26, 2012).
after severe head injury. Crit Care Med 2005;33(10):2207–13. 98. Seckler AB, et al. Substituted judgment: how accurate are proxy predictions?
72. Shafi S, Diaz-Arrastia R, Madden C, et al. Intracranial pressure monitoring Ann Intern Med 1991;115(2):92–8.
in brain-injured patients is associated with worsening of survival. J Trauma 99. Zier LS, Burack JH, Micco G, et al. Surrogate decision makers’ responses to
2008;64(2):335–40. physicians’ predictions of medical futility. Chest 2009;136(1):110–7.
73. Stein SC, Georgoff P, Meghan S, et al. Relationship of aggressive monitoring 100. Mason S, et al. Brief report on the experience of using proxy consent for
and treatment to improved outcomes in severe traumatic brain injury. incapacitated adults. J Med Ethics 2006;32:61.
J Neurosurg 2010;112(5):1105–12. 101. Alzheimer’s Association. Research consent for cognitively impaired adults:
74. Stein SC, Georgoff P, Meghan S, et al. 150 years of treating severe traumatic recommendations for IRB and investigators. Alzheimer Dis Assoc Disord
brain injury: a systematic review of progress in mortality. J Neurotrauma 2004;18:171.
2010;27(7):1343–53. 102. Kim SY, et al. Proxy and surrogate consent in geriatric neuropsychiatric
75. Whitmore RG, Thawani JP, Grady MS, et al. Is aggressive treatment of research: update and recommendations. Am J Psychiatry 2004;162:777.
traumatic brain injury cost-effective? J Neurosurg 2012;Mar 6. Epub ahead 103. Gong MN, Winkel G, Rhodes R, et al. Surrogate consent for research
of print. involving adults with impaired decision making: survey of Institutional
76. Wartenberg KE, Schmidt JM, Mayer SA. Multimodality monitoring in Review Board practices. Crit Care Med 2010;38(11):2146–54.
neurocritical care. Crit Care Clin 2007;23(3):507–38. 104. Best Hospitals. Available at: www.consumerreports.org/health/doctors-and-
77. Oddo M, Levine JM, Mackenzie L, et al. Brain hypoxia is associated with hospitals/hospital-selector.htm (accessed July 26, 2012).
short-term outcome after severe traumatic brain injury independently of 105. Boissy AR, Ford PJ, Edgell RC, et al. Ethics consultations in stroke and
intracranial hypertension and low cerebral perfusion pressure. neurological disease: a 7-year retrospective review. Neurocrit Care
Neurosurgery 2011;69(5):1037–45; discussion 1045. 2008;9(3):394–9.
78. Figaji AA, Zwane E, Thompson C, et al. Brain tissue oxygen tension 106. Luce JM. A history of resolving conflicts over end-of-life care in intensive
monitoring in pediatric severe traumatic brain injury. Part 1: relationship care units in the United States. Crit Care Med 2010;38(8):1623–9.
with outcome. Childs Nerv Syst 2009;25(10):1325–33. 107. Jacobs LM, Burns K, Bennett Jacobs B. Trauma death: views of the public
79. Maloney-Wilensky E, Gracias V, Itkin A, et al. Brain tissue oxygen and and trauma professionals on death and dying from injuries. Arch Surg
outcome after severe traumatic brain injury: a systematic review. Crit Care 2008;143(8):730–5.
Med 2009;37(6):2057–63. 108. Roe et al. v. Wade, district attorney of Dallas County no. 70-18 Supreme
80. Nangunoori R, Maloney-Wilensky E, Stiefel MD, et al. Brain tissue oxygen Court of the United States 410 u.s. 113; 93 S. Ct. 705; 35 L. Ed. 2d 147;
based therapy and outcome after severe traumatic brain injury: a systematic 1973 U.S. LEXIS 159
literature review. Neurocrit Care 2011;Aug 16. Epub ahead of print. 109. California Health & Safety Code 7185 to 7194.5 (West Supp. 1994)
81. Dohmen C, Bosche B, Graf R, et al. Prediction of malignant course in MCA 110. Jennett B, Plum F. Foreword in: Persistent vegetative state after brain
infarction by PET and microdialysis. Stroke 2003;34(9):2152–8. damage: a syndrome in search of a name. Lancet 1972;1:734–7, pp ix-x.
82. Heiss WD, Dohmen C, Sobesky J, et al. Identification of malignant brain 111. Jennett B. Foreward. In Wijdicks EFM, editor. Brain death. Philadelphia:
edema after hemispheric stroke by PET-imaging and microdialysis. Acta Lippincott Williams & Wilkins; 2001, pp. ix–x.
Neurochir 2003;86:237–40. 112. Pallis C. On the brainstem criterion of death. In: Youngner S, Arnold RM,
83. Sahuquillo J. Does multimodality monitoring make a difference in Shapiro R, editors. The definition of death: contemporary controversies.
neurocritical care? Eur J Anaesthesiol 2008;42:83–6. Baltimore: Johns Hopkins University Press; 1999. p. 93–100.
84. Robertson CS, Valadka AB, Hannay HJ, et al. Prevention of secondary
ischemic insults after severe head injury. Crit Care Med 1999;27(10):2086–95.
Chapter
9  
I

Monitors During Anesthesia:


Effects of Anesthetic Agents
on Monitors
Jonathan McEwen, K.T. Henrik Huttunen, and Arthur M. Lam

and is primarily used for carotid endarterectomy or func-


Introduction tional neurosurgery where sedatives usually are omitted.
3. Monitored anesthesia care. Only light sedation is used in
Need for Anesthesia and Monitoring these cases to supplement local anesthetic at the surgical
During Surgery site. This technique may apply to epilepsy surgery or tumor
Monitoring is an essential component of patient care in the surgery that encroaches on eloquent areas where patient
operating room, and standard monitoring mandated by the cooperation is required. It also is used commonly in neu-
American Society of Anesthesiology includes blood pressure, roradiologic procedures, some endovascular procedures,
electrocardiogram, pulse oximetry, end-tidal CO2 (Et CO2), or peripheral nerve surgery.
and temperature. However, for the neurosurgical patient,
either the surgical procedure or the patient’s pathology may
necessitate specific monitoring of the central nervous system.
Anesthetic Agents
The interpretation of these monitored variables is potentially Anesthetic agents include hypnotics, analgesics, neuromuscu-
subjected to the following influences: (1) pathophysiologic lar blockers, and other adjuncts. Their effects on interpreta-
changes (related to the patient’s disease or surgical manipula- tion of monitors are mediated by (1) direct effect—the specific
tion), (2) physiologic influences (related to changes that are intrinsic influence of each anesthetic agent on the physiologic
within “physiologic range”), and (3) anesthetic influences activity of the brain (i.e., blood flow, metabolism, or func-
(changes that are anesthesia related). Pathophysiologic inter- tion), or (2) indirect effect—changes in monitored variables
pretation of these monitors is covered elsewhere in this text- secondary to decreased perfusion of the brain secondary to
book, and “physiologic changes” are relatively minor. The the systemic/cerebral effects of the anesthetic. The influence
focus of this chapter is on the potential interactions and influ- is unique to the anesthetic agent and to the monitored vari-
ence of anesthetic agents on these monitors. able, and is discussed under the monitored modalities later.
The common anesthetic agents in current practice include the
following:
Common Anesthetic Techniques Hypnotics
Although most neurosurgical procedures are performed under Inhaled agents: desflurane, sevoflurane, isoflurane,
general anesthesia, some procedures such as functional neu- halothane, and nitrous oxide (N2O)
rosurgery require either a cooperative patient or one with Intravenous agents: propofol, thiopental, benzodiaze-
intrinsic cortical or subcortical electroencephalographic activ- pines, ketamine, and etomidate
ity unaltered by any central nervous system depressants. In Analgesics
general the anesthetic techniques can be classified into three Morphine and other synthetic opiates, nonsteroidal
categories: anti-inflammatory drugs, gabapentin, and pregabalin.
Neuromuscular blockers
1. General anesthesia. The patient is rendered unconscious Short-acting agents, such as succinylcholine; inter­
in the conventional manner with the use of hypnotic/ mediate-duration agents, such as rocuronium and
potential inhaled or intravenous agents/narcotics with or vecuronium; and long-acting agents, such as pan-
without neuromuscular blocking agents. curonium
2. Regional anesthesia. A field block or regional block with Other adjuncts
local anesthetic with and without use of sedative is used. Neuroleptics and α-2 agonists, such as dexmedetomi-
This has limited application in neurosurgical procedures, dine
© Copyright 2013 Elsevier Inc. All rights reserved. 71
72 Section I—Background

applications for TCD that are described in detail in Chapter


Monitors and the Influence 30. This chapter addresses the use of TCD as an intraoperative
of Anesthetic Agents monitor and effects of anesthetic agents on TCD.
TCD has the ability to noninvasively assess local CBF veloci-
Cerebral Blood Flow and Surrogates ties in the major intracranial arteries. This determination of
Continuous direct measurement of cerebral blood flow (CBF) cerebral blood velocities allows for an indirect assessment of
continues to be an elusive goal, although laser Doppler1 and CBF, and TCD can quantitatively measure relative changes in
thermodilution flow meters2 are available. They can be diffi- CBF. This property makes TCD an appealing intraoperative
cult to use and are not widely used during surgical procedures. monitor, in that maintenance of adequate cerebral perfusion
Anesthetic agents do not directly influence the function of is a goal in any operative procedure.
these monitors, but can alter CBF (see Transcranial Doppler Bilateral insonation of the middle cerebral arteries (MCAs)
Ultrasonography). through the temporal bone (temporal window) is the most
commonly used intraoperative application of TCD. This
allows for comparison of blood flow velocities between
Jugular Venous Oximetry hemispheres, emboli monitoring, assessment of cerebral auto-
Jugular venous oxygen saturation reflects the balance between regulation, and CO2 reactivity. However, TCD has several
CBF and cerebral oxygen metabolism. Thus a higher jugular limitations that prevent routine application of it as an intra-
venous oxygen saturation (>70%) suggests flow in excess of operative monitor:
metabolic need, and conversely, a low saturation (<50%) indi-
cates increased extraction and is suggestive of ischemia. Anes- 1. In neurosurgical patients there is limited access to acoustic
thetic agents can influence this balance between flow and windows required for accurate flow velocity measurements
metabolism. Inhaled agents tend to result in higher jugular because of the surgical incision sites and the sterile surgical
venous saturation, whereas intravenous agents such as propo- field during a craniotomy. This limitation, however, may
fol result in lower saturation.3 More important than the anes- not apply during a carotid endarterectomy.
thetic influence is the cerebrovascular response to change in 2. Patient positioning complicates placement of the ultra-
carbon dioxide (CO2) tension; thus hyperventilation can sound transducers in stable positions. For reliable, con-
result in low jugular venous saturation, and hypoventilation tinuous assessment of cerebral BFV, TCD probes require a
may have the opposite effect. It is generally considered prudent constant insonation angle (CBF velocity is proportional to
to maintain jugular venous saturation greater than 50%. the cosine of the angle of insonation). This requires the
Although not a widely adopted practice, the use of jugular use of a fixation device, such as the LAM-Rack, to stabilize
venous oximetry is considered a useful intraoperative monitor the ultrasound transducers, because handholding tech-
for craniotomy at some centers where hyperventilation niques or intermittent measurements are unreliable.
is used.4 3. About 20% of patients have inadequate acoustic windows
because of temporal bone hyperostosis.
Intracranial Pressure and Despite its limitations, TCD can be a valuable intraoperative
Cerebral Perfusion Pressure Monitor monitor. It is particularly useful during carotid endarterec-
Monitoring of intracranial pressure (ICP) provides useful tomy,8-11 in which it can be more accurate than near infrared
information about the status of the intracranial compartment, spectroscopy (NIRS), stump pressure and somatosensory
and just as importantly the net perfusion pressure to the brain evoked potentials (SSEP) in patients at risk for intraoperative
(cerebral perfusion pressure = mean arterial pressure − ICP). cerebral hypoperfusion or emboli.12 TCD also allows for
Anesthetic agents do not per se interfere with the measure- assessment of postoperative embolic phenomenon13-19 and
ment of ICP, but they can alter ICP by causing vasodilation or hyperperfusion.20-25 Both of these conditions are associated
vasoconstriction. In general, inhaled agents cause either no with postoperative neurologic insult and stroke.26-27
change or a dose-related increase in CBF, whereas intravenous
agents such as propofol cause vasoconstriction and so a reduc-
tion in ICP (ketamine is an exception). The Prx index, that is, Anesthetic Effects on Blood Flow Velocity
the moving correlation coefficient between mean ICP and Unlike other neurophysiologic monitoring techniques, TCD
mean arterial blood pressure, can provide information about is relatively resistant to interference from anesthetic agents.
autoregulation.5 However, the effects of anesthetics on Prx However, changes observed in CBF under anesthesia may
remain unclear. reflect physiologic changes of the patient associated with anes-
thetic influence. Consequently, TCD has been used to study
neurophysiologic effects of anesthetic agents.28 Table 9.1 sum-
Transcranial Doppler Ultrasonography marizes these anesthetic physiologic effects measured by TCD.
Although not widely used in the operating room because of
technical and labor-intensive demands, transcranial Doppler
(TCD), when properly applied, can be a useful continuous Inhalational Anesthetics
monitor of relative changes in CBF.6 CBF-metabolism coupling is generally preserved during inha-
TCD is a noninvasive technique that assesses blood flow lation anesthesia, and has been extensively studied using TCD,
velocity (BFV) and direction in the cerebral arteries by means because changes in cerebral blood volume reflect correspond-
of a pulsed ultrasonic beam. Aaslid et al first described the ing CBF changes. Although flow-metabolism coupling is pre-
TCD technique in 1982.7 Today there are multiple clinical served with volatile anesthetics29; the net effect of a volatile
Section I—Background 73

Table 9.1  Anesthetic Effects on Cerebral Hemodynamics Measured by Transcranial Doppler


Anesthetic Agent CBFV Autoregulation CO2 Reactivity References
N2O ↑ Impaired Preserved 38-41
Isoflurane (0.5-1.5 MAC)  Preserved at 1 MAC,  >1MAC Preserved at 1 MAC 26, 28, 35, 37
Desflurane (0.5-1.5 MAC)  Preserved at 1 MAC, Impaired Preserved at 1 MAC 32-34
at 1.5 MAC
Sevoflurane (0.5-2 MAC)  Preserved at 1.5 MAC Preserved at 2 MAC 27-31
Midazolam (0.15 mg/kg bolus) ↔ 45
Thiopental (5 mg/kg bolus) ↓ 45
Propofol (2 mg/kg bolus) ↓ 45
Propofol (25-200 mcg/kg/min)  Preserved Preserved 42, 43, 44, 100
Etomidate (0.3 mg/kg bolus) ↓ Preserved 51-53
Ketamine (1.5 mg/kg bolus) ↑/↔ Preserved 45, 47, 48
Dexmedetomidine (various doses)  Impaired Impaired 49, 50
Fentanyl (various doses) ↔ Preserved 45, 55
Remifentanil (various doses) ↔ Preserved Preserved 42, 56
Sufentanil (0.1-0.2 vs 6 mcg/kg) ↔/ 54, 55

CBFV, Cerebral blood flow velocity; ↑, increased; ↓, decreased; , dose dependent increase; , dose dependent decrease; ↔, minimal effect; MAC, minimum alveolar
concentration.

anesthetic on CBF depends on the balance between the agent’s dose-dependent cerebral vasodilatory properties.32 When
direct vasodilatory effect and flow-metabolism coupling- unopposed by flow-metabolism coupling-mediated vasocon-
mediated vasoconstriction.30 striction, isoflurane, in concentrations of 0.5 to 1.5 MAC,
Sevoflurane is a commonly used volatile anesthetic during causes significant increases to cerebral BFVs.30 The effect of
neurosurgical procedures because it has relatively low blood- isoflurane on cerebral autoregulation is dose dependent, and
gas solubility that allows rapid titration and emergence, and this homeostatic mechanism is preserved up to concentrations
has favorable cerebral hemodynamic effects compared with of 1 MAC,39 beyond which it is impaired. However, this
other volatile anesthetics. Sevoflurane, similar to other volatile depression of cerebral autoregulation can be restored with
anesthetics, is a central nervous system depressant that can hyperventilation.40 CO2 reactivity is maintained at 1 MAC.41
decrease the cerebral metabolism. This depressant effect N2O is most commonly used as an adjuvant to other anes-
causes TCD-measured cerebral BFV to decrease secondary to thetic agents, because hyperbaric conditions are required to
flow-metabolism coupling.31 As concentrations of sevoflurane reach anesthetic concentrations with N2O alone. N2O is a
are increased, TCD flow velocities tend to increase, but to a potent cerebral vasodilator, and results in increased cerebral
lesser extent than other volatile agents32; this reflects the influ- BFVs.42 N2O also increases cerebral metabolism and can
ence of the intrinsic cerebrovasodilatory effect of sevoflurane33 increase CBF through vasodilation.43 Furthermore, cerebral
and can result in luxury perfusion states with concentrations autoregulation is impaired with N2O.44 Together these factors
greater than 1.5 minimum alveolar concentration (MAC). It make N2O an undesirable agent in patients with limited intra-
is unusual that clinical concentrations are required above cranial compliance. However, cerebrovasomotor reactivity to
this level. Furthermore, at 1.5 to 2 MAC, cerebral autoregula- CO2 is preserved.45
tion mechanisms34 and cerebral reactivity to CO2 are well
preserved.35
Desflurane has the lowest blood-gas solubility coefficient of Intravenous Anesthetics
the halogenated ether anesthetics, making it desirable for use Propofol is commonly used as an anesthetic induction
in neuroanesthesia because it facilitates a rapid emergence. agent and to maintain anesthesia. It is easily and rapidly titrat-
However, cerebral physiologic effects of desflurane are not as able and well tolerated for use during evoked potential moni-
favorable as for sevoflurane.36 Desflurane causes a dose- toring, and its cerebrovascular effects are favorable in the
dependent increase in cerebral BFVs; this takes effect at much neurosurgical patient. Propofol causes a dose-dependent
lower MAC values than for sevoflurane. To reduce the luxury decrease in cerebral BFVs,46 supporting its preservation of
perfusion, increased cerebral blood volume and potential flow-metabolism coupling. Propofol also has direct cerebral
increase in ICP that can be caused by desflurane, concentra- vasoconstricting properties that contribute to the observed
tions should be limited to 1 MAC. Cerebral autoregulation BFV decreases. Cerebral autoregulation and CO2 reactivity are
and CO2 reactivity are preserved at this clinically relevant well preserved.47,48
concentration.37,38 Thiopental is a barbiturate anesthetic, which like propofol,
Isoflurane has a long history of use as a neuroanesthetic can be used for both induction and maintenance of anesthe-
agent. Similar to other volatile agents, isoflurane has intrinsic sia. However, thiopental’s pharmacodynamic profile can be
74 Section I—Background

undesirable in the neurosurgical patient in that it delays emer- correlates or derivatives are influenced by anesthetic agents.
gence. Nevertheless, thiopental is frequently used as a neuro- Both intravenous anesthetics and inhalation anesthetics cause
protective agent during aneurysm surgery (e.g., during dose-related depression that ultimately may result in electrical
temporary arterial occlusion), because it decreases cerebral silence. Low-dose inhalation agents can cause initial stimula-
metabolism with the induction of burst suppression. Induc- tion with an increase in EEG activity in the alpha band. With
tion doses of 5 mg/kg decrease cerebral BFV.49 progressive increase in depth, there is an increase in delta
Ketamine is an intravenous anesthetic agent with dissocia- wave, and disappearance of beta and alpha bands. With further
tive anesthetic properties. It has a controversial history in the increase in anesthetic depth, burst-suppression progressing to
neurosurgical patient in that older evidence showed ketamine electrical silence will occur. The dose-response curve is differ-
could increase CBF velocities, cerebral metabolism, and ICP.50 ent among the inhaled anesthetics. Isoflurane will cause burst-
Ketamine increases cerebral BFV during induction,49 but as an suppression at 1.5 MAC and electrical silence at 2 MAC,
adjuvant to an anesthetic under controlled ventilation, ket- whereas sevoflurane causes burst-suppression at 2 MAC and
amine does not increase cerebral BFV.51 When used with pro- electrical silence at 2.5 MAC.61,62 There also is an age-anesthetic
pofol as part of a total intravenous technique, ketamine effect interaction, with lower anesthetic doses needed to
preserves cerebral autoregulation.52 achieve burst suppression in older patients.63 N2O paradoxi-
Dexmedetomidine is a selective α-2 agonist used as a seda- cally can cause an increase in EEG activity when added to an
tive in intensive care units and in the operating room for inhalation anesthetic. Seizure activity also has been reported
procedures such as awake craniotomies and carotid endarter- with most anesthetic agents.64-66 Even though some agents
ectomies. It has sedative properties with minimal respiratory have a higher propensity to cause epileptogenic activity, the
depression that makes it appealing for these applications. Dex- clinical significance in unclear. For example, inhalation anes-
medetomidine decreases cerebral BFVs in a dose-dependent thetics including sevoflurane and enflurane can cause epilep-
manner,53,54 which appears to result from decreased cerebral togenic spikes in healthy patients without a history of
metabolic rate with preserved flow-metabolism coupling.54 epilepsy.67 Seizure activity can interfere with monitoring of
However, at sedative doses, dexmedetomidine may cause a computer-processed EEG such as the Bispectral Index. Intra-
slight decrease in both dynamic cerebral autoregulation53 and venous agents in general do not cause stimulation and slow
CO2 reactivity.54 waves develop early. Both thiopental and propofol at increas-
Etomidate is an imidazole derivative intravenous anesthetic ing doses will result in burst suppression pattern followed by
agent commonly used as an induction agent, particularly in electrical silence.68 This is used often for brain protection
trauma patients or those with limited cardiac reserve, because during periods of potential ischemia.
of its hemodynamic stability. Induction doses of etomidate Because anesthetic agents can have a profound influence on
lead to decreased middle cerebral artery BFVs.55,56 CO2 reactiv- EEG, anesthetic depth must be maintained at a relatively steady
ity is preserved during etomidate infusion57; however, infu- state to reliably monitor cerebral ischemia, to allow meaning-
sions are not recommended because there may be adrenal ful interpretation of changes. These agents and benzodiaze-
suppression and propylene glycol toxicity. pines must be avoided during functional surgery when
Midazolam is a commonly used intravenous benzodiaze- mapping of electrical activity or epileptic foci is important. For
pine. It is used frequently as a preoperative anxiolytic or as an both deep brain stimulator implantation and resection of epi-
infusion in intensive care units as a sedative. There is conflict- leptic foci in patients who are otherwise awake, dexmedetomi-
ing evidence on the effects of midazolam on cerebral BFVs. In dine as the sole sedative agent has been used with some success,
one study midazolam at 0.15 mg/kg with fentanyl did not without interfering with electrophysiologic recording.69,70
affect MCA BFVs.49 In other studies, however, at a much lower
dose of 20 and 40 µg/kg, cerebral BFVs decreased about 20%
but returned to baseline in 5 minutes.58 Evoked Potentials
Opioids are used frequently during neurosurgery for mul- Multimodality evoked potentials increasingly are used intra-
tiple reasons: to blunt sympathetic responses to stimulation, operatively to monitor and help preserve integrity of the
to treat pain, and as adjuvants to help reduce the doses of central nervous system structures placed at risk by the surgical
other anesthetic drugs. In clinically relevant concentrations procedure. Anesthetic agents have unique and profound influ-
with controlled ventilation, opioids have limited effects on ence on evoked potentials, and these effects must be under-
cerebral physiology. Fentanyl and remifentanil do not signifi- stood to allow meaningful recording and interpretation of
cantly change cerebral BFVs. CO2 reactivity and cerebral auto- these signals. The modalities include SSEP, motor evoked
regulation also remain intact.46,49,59,60 Low doses of sufentanil potentials (MEP) and cranial nerves with motor components
(0.1 to 0.2 µg/kg) do not affect cerebral BFVs; however, (V, VII, IX, X, XII).
increasing doses can decrease cerebral BFV in a dose-
dependent fashion.58,59
Somatosensory Evoked Potential Monitoring
An important clinical decision when using SSEP monitoring
Cerebral Function is the choice between inhalational and intravenous anesthetic.
Even though SSEPs are not degraded to the same extent as
Electroencephalography (Unprocessed MEPs by inhalational anesthetics, they exhibit a dose-related
and Computer Processed) increase in latency and decrease in amplitude under volatile
Electroencephalogram (EEG) activity can be monitored intra- anesthesia. This is particularly true for cortical SSEPs that
operatively for a variety of reasons, such as to detect cerebral involve multiple synapses. Brainstem and spinal components
ischemia or to localize seizure activity. However, all EEG are relatively resistant to anesthetic effects. When volatile
Section I—Background 75

Table 9.2  Effects of Anesthetic Agents on Brainstem Auditory Evoked Potentials and
Somatosensory Evoked Potentials
BAEP SSEP
Latency Amplitude Latency Amplitude
Thiopental
Low dose ↔ ↔ ↑ ↓
High dose
↑ ↓ ↑ 

Propofol ↑ ↔ ↑ ↔ or ↓
Dexmedetomidine ? ? ↔ ↔
Midazolam ? ? ↔ ↓
Etomidate ↔ ↔ ↑ 

Ketamine ↔ ↔ ↔ 

Fentanyl ↔ ↔ ↑ ↓
Remifentanil ↔ ↔ ↔ ↔
Halogenated volatile agents
0.5 Mac
↑ ↔ ↔ or ↑ ↔ or ↓
>1 Mac
 

Nitrous oxide ↔ ↔ ↔ 

The relative influence of anesthetics on brainstem auditory evoked potentials (BAEP) and somatosensory evoked potentials (SSEP) is depicted. Halogenated volatile
agents referenced include desflurane, sevoflurane, and isoflurane.
↔, No change; ↑, clinically insignificant increase; ↓, clinically insignificant decrease; , clinically significant increase; , clinically significant decrease; ?, no
information.

agents are used, the dose should be kept below 1 MAC to anesthetic technique. Inhalation agents can increase the
maintain SSEP signal quality. This may be facilitated by the latency but do not abolish BAEPs.72 The effects of anesthetic
coadministration of potent opiates, which do not affect signal agents on BAEP and SSEP are summarized in Table 9.2.
quality. The addition of N2O typically is avoided, because this
further degrades the signal.71 In addition, the anesthetic con-
centration should be kept constant during critical periods to Motor Evoked Potentials
reduce anesthetic influence on the interpretation of changes. MEPs are electrical potentials monitored along the motor
The use of intravenous anesthetics, such as propofol, thiopen- pathway after stimulation of the motor cortex or spinal cord.
tal, or midazolam infusion, often results in better signal quality MEPs are elicited by direct or transcranial stimulation with
than with use of volatile agents. Ketamine and etomidate, in either electrical (TcEMEP) or magnetic (TcMMEP) tech-
particular, have been reported to increase the amplitude of niques. Responses can be recorded over the spinal cord from
SSEPs, although the effect that this has on sensitivity and electrodes placed in the epidural space (spinal D waves or
utility remains a matter of speculation. Regardless of the anes- epidural MEP), along peripheral motor nerves (neurogenic
thetic regimen, it is important to avoid bolus doses or rapid MEP), or as compound muscle action potentials (CMAPs)
changes in anesthetic depth during key portions of an inter- over muscle (myogenic MEP). Myogenic MEPs are large
vention, such as vessel occlusion or manipulation of the spine, amplitude biphasic potentials in response to motor unit depo-
to avoid confounding influences on signal integrity at a critical larization, and so, unlike SSEPs, signal averaging is not
time when the monitoring modality is most useful. required. This allows for near instant motor pathway feedback.
Other factors, such as blood pressure, temperature, and TcMMEPs are exquisitely sensitive to anesthetic agents and so
patient position, may influence evoked potential monitoring. are unsuitable for intraoperative monitoring.
Nerve injury associated with patient position, such as crush The entire motor pathway from cerebral motor cortex
injury at pressure points, or traction injury must be consid- through brainstem, corticospinal tracks, anterior horn,
ered. A decrease in blood pressure may also alter SSEPs and peripheral nerve, and neuromuscular junction to skeletal
even mild hypothermia can increase latency and decrease the muscle can be assessed using transcranial stimulation with
amplitude of evoked potentials because thermoregulatory myogenic MEP monitoring. Motor pathway injury can result
behaviors and physiologic responses are blunted in the anes- from ischemia, or disruption, retraction, compression, or dis-
thetized patient. traction of neural tissue. Each of these insults can cause func-
tional alterations that may be neurophysiologically evident.73
MEPs therefore are an appealing intraoperative monitor and
Brainstem Auditory Evoked are applicable to any surgical procedure which places the
Potential Monitoring motor pathway at risk for injury: craniotomy, spinal surgery,
Brainstem auditory evoked potentials (BAEPs) are resistant to or thoracoabdominal aortic aneurysm repair. Intraoperatively,
anesthetic effects and can be recorded with virtually any MEPs are usually monitored with SSEPs.
76 Section I—Background

1 stimulus 3 stimuli 5 stimuli

Isoflurane 0%

0.2%

0.4%

Fig. 9.1  Graphs that illustrate augmentation of


0.5 mV
motor evoked potentials (MEP) responses when
using multipulse stimuli with increasing isoflurane
doses. (Ubags LH, Kalkman CJ, Been HD. Influence of 0.6%
isoflurane on myogenic motor evoked potentials to
single and multiple transcranial stimuli during nitrous
oxide/opioid anesthesia. Neurosurgery. 1998;43(1):
90–4.) 0 100 Time (msec) 0 100 Time (msec) 0 100 Time (msec)

MEPs are useful to monitor for ischemia, particularly of energy may attenuate anesthetic fade, but increases the risk for
the anterior horn cells (alpha motor neurons), which are potential false-positive signal changes.
thought to be most sensitive to ischemic insults.73 Many pub- Volatile anesthetics are used commonly to maintain anesthe-
lications describe the use of MEP for intraoperative monitor- sia. All currently used halogenated anesthetics cause signifi-
ing, although there are no clearly defined thresholds for cant dose-dependent depression of myogenic MEP signals.80-85
latency or amplitude changes that correlate with risk of This depression is believed to result from volatile anesthetic-
motor pathway injury. In general, latency is robust to anes- induced inhibition of pyramidal activation of spinal motor
thetic or surgical insults; therefore amplitude changes are neurons.86 At full anesthetic concentrations of 1 MAC, multi-
assessed more commonly. The threshold considered concern- pulse TcEMEPs with isoflurane are only recordable in 8% of
ing for injury is the complete loss of signal rather than a fixed subjects.82 TcEMEP in response to paired TcE stimulation
percentage decrease in amplitude (e.g., 50%). Other authors during sevoflurane anesthesia at 1 MAC is unrecordable,84
consider the increase in stimulation energy required to repro- whereas with a four-pulse TcE stimulation this result improves
duce baseline signals as the marker for injury.74 The safety of to more than 80%.85 However, this is still inadequate for clini-
MEPs is well known.75 However, induction of seizures is a cal use. With desflurane or sevoflurane at 0.5 MAC with either
potential risk with TcEMEPs. N2O or a narcotic infusion, MEPs are recordable in 100% of
patients,81,83 and with isoflurane MEP monitoring is possible
in 90%.78 Total intravenous anesthetic regimens therefore
Anesthetic Effects seem preferable and only one study to date has shown ade-
MEPs are sensitive to anesthetics; therefore a tailored approach quate clinical recording conditions with a volatile anesthetic
is required to select the most appropriate anesthetic regimen.76 compared to total IV technique.81
Transcranial magnetic stimulation–generated MEPs are more N2O is used frequently as an anesthetic adjuvant. When
sensitive to anesthetics than transcranial electrical (TcE) stim- 50% N2O is used with intravenous infusion of narcotic with
ulation, and usually are abolished with induction, regardless or without propofol, multipulse TcEMEP amplitudes are
of the agents used. TcE stimulation therefore is the technique reduced by about half, but can be consistently recorded.87,88
used exclusively during intraoperative monitoring. Transcra- With concentrations ranging from 20% to 60%, N2O is associ-
nial magnetic stimulation currently has a very limited role in ated with a dose-dependent decrease in MEP amplitude, yet
the operating room. Early work with TcEMEPs showed that signals remain recordable in 100% of subjects.89 Given these
single-pulse signals were extremely sensitive to volatile agents findings, N2O is an acceptable anesthetic adjuvant during
even at subclinical doses.77,78 Subsequently, multipulse stimu- MEP monitoring when administered with other agents that
lation techniques have been shown to improve both low- have limited depressive effects.
amplitude baseline signals and MEP signals depressed by Propofol is currently the most popular anesthetic agent to
anesthesia (Fig. 9.1). Table 9.3 summarizes how different maintain anesthesia during MEP monitoring in that it is easily
anesthetic agents affect multipulse TcEMEPs, which is the titratable and has a favorable pharmacokinetic profile. Titrat-
current standard during intraoperative monitoring. “Anes- ing propofol infusion rate intraoperatively to achieve clinically
thetic fade,” a time-dependent decrease in MEP signal ampli- relevant concentrations does not affect MEP responses in
tude proportionate to the length of surgery regardless of the a clinically significant fashion.90 However, dose-dependent
anesthetic regimen can occur.79 Increasing the stimulation depression of myogenic MEPs can be demonstrated at higher
Section I—Background 77

Table 9.3  Anesthetic Effects on Transcranial Electrical Stimulation on Motor Evoked


Potential Responses
Change in Amplitude % of Subjects with
Anesthetic Agent TcE Pulses from Baseline Response References
Isoflurane (0.17 MAC) 5 ↓↓ 100% 81
Isoflurane (0.33 MAC) 5 ↓↓ 100% 81
Isoflurane (0.5 MAC) 5 ↓↓↓ 90% 81
Isoflurane (0.75 MAC) 4 NA 61% 82
Isoflurane (1 MAC) 5 NA 8% 83
Desflurane (0.5 MAC) 5 NA 100% 84
Sevoflurane (0.25 %) 2 ↓↓↓ 85% 85
Sevoflurane (0.5 MAC) 2/4 ø/NA 55%/100% 85, 86
Sevoflurane (0.75 MAC) 2/4 ø/NA 10%/90%–100% 85, 86
Sevoflurane (1 MAC) 2/4 ø/NA 0%/80%–90% 85, 86
N2O (50%) 2 ↔ 100% 88, 89
N2O (20%, 40%, 60%) 6  100% 90
Midazolam (0.05 mg/kg bolus) 1 ↓↓↓ 100% 92
Thiopental  NA 101
Propofol (67 µg/kg/min) 5 NA 100% 95
Propofol (>100 µg/kg/min) 4 97% 82
Etomidate (0.1 mg/kg bolus) 3 ↓ 100% 94
Etomidate (5-10 µg/kg/min) 1 100% 96
Ketamine (0.5 mg/kg bolus) 3 ↔ 100% 94
Ketamine (1 mg/kg/hr) 5 NA 100% 96
Dexmedetomidine (1 µg/kg bolus) ↔ 100% 97
Dexmedetomidine (0.15 µg/kg/hr) 7 ↔ 100% 98
Dexmedetomidine (~0.3 µg/kg/hr) 7 ↔ 100% 98
Fentanyl (3 µg/kg bolus) 1 ↔ 100% 92
Remifentanil (9 ng/mL)  100% 101
Sufentanil (0.5 µg/kg/hr) 3 100% 81
Neuromascular blockade  /ø
T1 45%–55% of baseline 6 ↔ 100% 102
T1 5%–15% of baseline 6 ↓↓↓ 100% 102

↔, Minimal/no change; ↓, decrease 5% to 30% ; ↓↓, decrease 30% to -60%; ↓↓↓, decrease greater than 60%; ø, absent or less than 10%; , dose-dependent
decrease; MAC, minimum alveolar concentration; blank indicates no reliable data.

doses.76 Bolus administration of propofol can lead to MEP Ketamine can be used in boluses (0.5 mg/kg) or as continu-
signal loss. However, since propofol has rapid redistribution ous infusions (1 mg/kg/hr) without altering MEP responses.92-94
and metabolism, signals return within minutes91 (Fig. 9.2). Excellent recording conditions are achieved in all cases.
Therefore during intraoperative MEP monitoring propofol is However, ketamine has dissociative anesthetic side effects
preferable to many of the other intravenous agents because of and potentially undesirable or controversial neurophysiologic
its desirable neurophysiologic effects and pharmacokinetic effects. These factors need to be balanced with the desire for
profile. reliable intraoperative MEP signals.
Midazolam boluses of 50 µg/kg cause significant depression Etomidate has appealing hemodynamic stability for anes-
of single-pulse TcEMEP signals; the signals, however, are still thesia induction. It also can be used as a continuous infusion
recordable in all patients.91 Infusions of 0.1 mg/kg/hr of mid- to maintain sedation. However, it may contribute to adrenal
azolam do not depress TcMMEP signals.76 Studies have not insufficiency and propylene glycol toxicity. Nevertheless,
been done in humans with multipulse TcE, but one would etomidate allows for excellent recording conditions for MEPs,
expect signals to be better than those described for TcM or with only a short duration of suppression following bolus
single pulse TcE. Other benzodiazepines have similar effects administration and stable recording conditions when given as
as midazolam. an infusion.91,93,95
78 Section I—Background

Propofol Midazolam Etomidate Fentanyl


+
2 mV

Control

2 min

5 min

10 min
Fig. 9.2  The effect of intravenous boluses of
anesthetic agents on motor evoked potentials 30 min
(MEPs). (Kalkman CJ, Drummond JC, Ribberink AA,
et al. Effects of propofol, etomidate, midazolam, and
fentanyl on motor evoked responses to transcranial
0 20 40 60 80 100 0 20 40 60 80 100 0 20 40 60 80 100 0 20 40 60 80 100
electrical or magnetic stimulation in humans. Anesthe-
siology. 1992;76(4):502–9.) Time (msec)

Dexmedetomidine is a relatively new sedative and anesthetic neuromuscular blockade or rebolusing can limit interpreta-
agent. The effects of dexmedetomidine on MEP amplitudes tion of MEPs and should be avoided.
have been studied in patients undergoing spine surgery.96-98
Earlier studies showed that a standard infusion rate of dexme-
detomidine has little effect on MEP amplitude and signals are
Cerebral Metabolism
recordable in 100% of subjects.96,97 However, a more recent Two modalities can be used intraoperatively to estimate cere-
study suggests that dexmedetomidine may cause a clinically bral oxygen metabolism: jugular venous oximetry (see also
significant dose-related decrease in MEP amplitude98 and loss Chapter 32) and transcranial NIRS (see also Chapter 33). Both
of signals has been described in pediatric spine surgery.99 modalities provide information on the balance between flow
Dexmedetomidine has propofol-sparing effects; therefore a and metabolism rather than on cerebral oxygen metabolism
low dose infusion may be a useful adjunct during propofol itself. Jugular venous oximetry is discussed in the section on
anesthesia when MEPs are monitored, but the effects of chang- CBF in this chapter.
ing infusion rate/plasma concentration must be taken into
account.
Opioids are administered commonly with other anesthetic Near Infrared Spectroscopy
agents. Opioids frequently are used when MEP monitoring is NIRS is a noninvasive technique that can measure cerebral
required because they have minimal effects. A single bolus of oxygen saturation, cerebral blood volume, and even mitochon-
fentanyl at 3 µg/kg does not decrease MEP amplitude.91 Suf- drial redox states using differential absorption of near infrared
entanil at an infusion rate of 0.5 µg/kg/hr allows reliable light (700 to 1000 nm).104 It is an evolving technology that is
recordings in 100% of patients.80 Other investigations have starting to be adopted for intraoperative neurophysiologic
shown that opioids decrease myogenic MEPs in a dose- monitoring, and so its role and utility is only beginning to be
dependent manner; however, at target-controlled plasma con- elucidated. NIRS is used most commonly in cardiac surgery
centrations remifentanil has the least effect.100 during cardiopulmonary bypass and during carotid endarter-
Neuromuscular blockade generally is used to facilitate tra- ectomy. In carotid endarterectomy it is used as an ischemia
cheal intubation or provide an immobile patient optimal sur- monitor during carotid cross clamping and appears to be
gical exposure. However, boluses of muscle relaxants effective and comparable to other commonly used monitors.
inevitably abolish MEPs, in that they block signal transduc- However, relative and absolute thresholds for changes in
tion at the neuromuscular junction. Careful infusions of neu- regional cerebral oxygen saturation remain to be defined.105
romuscular blockers to achieve 20% to 50% maintenance of NIRS also has been used during neuroendovascular proce-
single twitch height have been shown to be compatible with dures for cerebral aneurysm embolization106 and preopera-
reliable MEP recordings when used with a minimally sup- tively to assess patients with skull base tumors and giant
pressant anesthetic and multipulse TcE. However, deeper aneurysms to determine cerebral oxygen reserve during vessel
blockade leads to unsatisfactory signals.101 To improve MEP test occlusion.107 NIRS has several theoretical advantages that
amplitude, a posttetanic stimulation can be used.102,103 make it an appealing intraoperative monitor: (1) it is noninva-
Caution is necessary in patients with preoperative neurologic sive, (2) it allows for continuous assessment, (3) NIRS machines
deficits because they appear to be more sensitive to the are portable, and (4) spectroscopy is unaffected by anesthetics.
depressant effects of neuromuscular blocking drugs.95 It gen- However, there also are several disadvantages: (1) it can only be
erally is acceptable and reasonable to use a single dose of a applied to the forehead, (2) the signal is contaminated by
short-acting neuromuscular blocking agent at the time of extracranial absorption and scattering, and (3) the signals can
induction because its effects should wear off by the time be affected by ambient light. NIRS is a promising technology,
intraoperative MEP recording begins. The exception is when but further research is required to demonstrate its clinical effi-
a baseline recording is desired. However, maintenance of cacy and justify its routine use during anesthesia.108
Section I—Background 79

Anesthetic Effects Volatile Anesthetics


Measurement of oxyhemoglobin and deoxyhemoglobin by The observed NIRS changes seen with the halogenated inhala-
spectroscopy is unaffected by anesthetics. However, anesthetics tional agents support findings associated with the neurophysi-
can affect cerebral metabolism and CBF. Therefore indirect ologic effects of these agents. Desflurane, sevoflurane, and
changes can be observed in NIRS monitoring that result from isoflurane have all been examined at clinically relevant concen-
the pharmacodynamic and consequent physiologic effects of trations and at 1 MAC do not affect rSO2112-114 despite decreases
each anesthetic agent. Because NIRS is a relatively new and in blood pressure and cardiac output. These results demon-
evolving monitoring modality, there is limited evidence about strate that cerebral autoregulation is maintained at these con-
observed NIRS changes that occur with different anesthetic centrations. Both desflurane and sevoflurane can result in a
agents. The known effects are summarized in Table 9.4. Intra- dose-dependent increase in rSO2113 suggesting either a shift in
operative use of intravenous dyes such as indigo carmine109 the flow-metabolism coupling or cerebrovasodilation at higher
and indocyanine green110,111 can cause low regional cerebral concentrations. Sevoflurane, at inhalational induction concen-
oxygen saturation (rSO2), similar to that observed with pulse trations of 8%, has been shown to result in cerebral hyperemia,
oximetry. although these results are confounded by the use of N2O.115
The effects of N2O on NIRS have not been specifically studied;
however, N2O has been used as an anesthetic adjuvant in many
Table 9.4  Observed Changes in Regional of the studies published on NIRS and may be a confounding
Cerebral Oxygen Saturation Under Various factor.
Anesthetics
Anesthetic Agent Change in rSO2 References Intravenous Anesthetics
Isoflurane (~IMAC) ↔ 114 Because of the ability to rapidly and fairly predictably achieve
Isoflurane (~IMAC) ↔ 113 a state of unconsciousness, bolus administration of intrave-
nous anesthetic commonly facilitates induction of anesthesia.
Desflurane (>IMAC)  113
However, limited information is available on the effect of dif-
Sevoflurane (~IMAC) ↔ 112 ferent intravenous agents on rSO2. There is some evidence that
Sevoflurane (>IMAC) 113,115 sedative doses of midazolam (0.05 mg/kg), do not change

rSO2 .113 One study examined the induction effects of etomi-
Midazolam (0.05 mg/kg ↔ 114 date, propofol, and thiopental on cerebral oxygenation and
bolus)
found that propofol and thiopental slightly increased rSO2
Thiopental (6 mg/kg bolus) ↑ 116 while etomidate caused a small decrease in cerebral oxygen-
Propofol (3 mg/kg bolus) ↑ 116 ation116 (Fig. 9.3). These differences were explained by varying
Etomidate (0.3 mg/kg ↓ 116 flow-metabolism coupling and cerebral autoregulation prop-
bolus) erties of the agents. Surprisingly, induction with etomidate
resulted in decreased cerebral oxygenation. This observation
↔, Equivocal/no change; ↑, increase;  ,dose-dependent increase;
↓, decrease; MAC, minimum alveolar concentration.
supports animal work that shows etomidate decreases CBF
despite maintenance of mean blood pressure before its

20

Thiopental

10
∆[HbO2] (µMolar)

Propofol

0
Fig. 9.3  Line graphs that illustrate relative
changes in oxyhemoglobin (HbO2) measured
using near infrared spectroscopy (NIRS) after
induction of anesthesia with three intravenous
Etomidate anesthetic agents. (Lovell AT, Owen-Reece H, Elwell
CE, et al. Continuous measurement of cerebral
–10
oxygenation by near infrared spectroscopy during
–2 –1 0 1 2 3
induction of anesthesia. Anesth Analg 1999;88(3):
Time (min) 554–8.)
80 Section I—Background

sedative effects can decrease metabolism. How maintenance 11. Halsey JH. Risks and benefits of shunting in carotid endarterectomy. The
International Transcranial Doppler Collaborators. Stroke 1992;23(11):
or steady-state effects of intravenous anesthetic agents affect
1583–7.
NIRS is not clear. 12. Moritz S, Kasprzak P, Arlt M, et al. Accuracy of cerebral monitoring in
detecting cerebral ischemia during carotid endarterectomy: a comparison of
transcranial Doppler sonography, near-infrared spectroscopy, stump
Extracranial Monitors pressure, and somatosensory evoked potentials. Anesthesiology
2007;107(4):563–9.
Extracranial monitors commonly used in the operating room 13. Ackerstaff RGA. Cerebral circulation monitoring in carotid
include the ECG, noninvasive and invasive blood pressure, endarterectomy and carotid artery stenting. Front Neurol Neurosci 2006;
central venous pressure, pulse oximetry, and EtCO2. Anes- 21229–38.
thetic agents do not have any direct influence on the function 14. Gossetti B, Gattuso R, Irace L, et al. Embolism to the brain during carotid
stenting and surgery. Acta Chir Belg 2007;107(2):151–4.
of these monitors. However, the various anesthetic agents may
15. Telman G, Kouperberg E, Eran A, et al. Microemboli in MCA ipsilateral to
affect the physiologic variables recorded by these monitors by occluded common carotid artery: an observation and short review of the
virtue of their effects on the cardiovascular and respiratory literature. Neurol Res 2008;30(7):684–6.
system. The systemic effects of anesthetic agents on these vari- 16. Imray CHE, Pattinson KTS. Potential role for TCD-directed antiplatelet
ables are reviewed in standard anesthesia texts (e.g., Miller’s agents in symptomatic carotid artery dissection. Stroke 2006;37(3):767.
17. Zuromskis T, Wetterholm R, Lindqvist JF, et al. Prevalence of micro-emboli
Anesthesia, 7th edition, 2010, Elsevier, or Clinical Anesthesia, in symptomatic high grade carotid artery disease: a transcranial Doppler
6th edition, 2009, Lippincott Williams & Wilkins). study. Eur J Vasc Endovasc Surg 2008;35(5):534–40.
18. Alexandrov AV, Babikian VL, Adams RJ, et al. The evolving role of
transcranial Doppler in stroke prevention and treatment. J Stroke
Conclusion Cerebrovasc Dis 1998;7(2):101–4.
19. Roy J, Akhtar N, Watson T, et al. Transcranial Doppler microembolic signal
Advanced neuromonitoring frequently is used during neuro- monitoring is useful in diagnosis and treatment of carotid artery dissection:
surgical procedures to guide surgical decisions and help two case reports. J Neuroimaging 2007;17(4):350–2.
improve patient safety. The successful deployment of these 20. Sfyroeras GS, Karkos CD, Arsos G, et al. Cerebral hyperperfusion after
techniques requires a team approach that involves the neuro- carotid stenting: a transcranial doppler and spect study. Vasc Endovascular
Surg 2009;43:150–6.
surgeon, the anesthesiologist, and the neurophysiologist. The 21. Adhiyaman V, Alexander S. Cerebral hyperperfusion syndrome following
rational use of these monitoring modalities and how the carotid endarterectomy. Q J Med 2007;100(4):239–44.
information is interpreted depends on use of an optimal anes- 22. Ogasawara K, Sakai N, Kuroiwa T, et al. Intracranial hemorrhage associated
thetic regimen. This is only possible with a full understanding with cerebral hyperperfusion syndrome following carotid endarterectomy
and carotid artery stenting: retrospective review of 4494 patients. J
of how anesthetic agents influence these various monitors.
Neurosurg 2007;107(6):1130–6.
This chapter provides the basic information and the frame- 23. Jansen C, Sprengers AM, Moll FL, et al. Prediction of intracerebral
work to understand how anesthetic agents affect monitors haemorrhage after carotid endarterectomy by clinical criteria and
both directly or indirectly because of physiologic effects. This intraoperative transcranial Doppler monitoring. Eur J Vasc Surg 1994;8(3):
knowledge is valuable in both the operating room and neuro- 303–8.
24. Jansen C, Sprengers AM, Moll FL, et al. Prediction of intracerebral
critical care unit. haemorrhage after carotid endarterectomy by clinical criteria and
intraoperative transcranial Doppler monitoring: results of 233 operations.
Eur J Vasc Surg 1994;8(2):220–5.
References 25. Magee TR, Davies AH, Horrocks M. Transcranial Doppler evaluation of
cerebral hyperperfusion syndrome after carotid endarterectomy. Eur J Vasc
1. Fukui K, Morioka T, Hashiguchi K, et al. Relationship between regional Surg 1994;8(1):104–6.
cerebral blood flow and electrocorticographic activities under sevoflurane 26. Riles TS, Imparato AM, Jacobowitz GR, et al. The cause of perioperative
and isoflurane anesthesia. J Clin Neurophysiol 2010;27(2):110–5. stroke after carotid endarterectomy. J Vasc Surg 1994;19(2):206–16.
2. Mielck F, Brauer A, Radke O, et al. Changes of jugular venous blood 27. Spencer MP. Transcranial Doppler monitoring and causes of stroke from
temperature associated with measurements of cerebral blood flow using the carotid endarterectomy. Stroke 1997;28(4):685–91.
transcerebral double-indicator dilution technique. Eur J Anaesthesiol 28. Fodale V, Schifilliti D, Conti A, et al. Transcranial Doppler and anesthetics.
2004:289–95. Acta Anaesthesiol Scand 2007;51(7):839–47.
3. Yoshitani K, Kawaguchi M, Iwata M, et al. Comparison of changes in jugular 29. Lam AM, Matta BF, Mayberg TS, et al. Change in cerebral blood flow
venous bulb oxygen saturation and cerebral oxygen saturation during velocity with onset of EEG silence during inhalation anesthesia in humans:
variations of haemoglobin concentration under propofol and sevoflurane evidence of flow-metabolism coupling? J Cereb Blood Flow Metab
anaesthesia. Br J Anaesth 2005;94(3):341–6. 1995;15(4):714–7.
4. Matta BF, Lam AM, Mayberg TS, et al. A critique of the intraoperative use of 30. Matta BF, Mayberg TS, Lam AM. Direct cerebrovasodilatory effects of
jugular venous bulb catheters during neurosurgical procedures. Anesth Analg halothane, isoflurane, and desflurane during propofol-induced isoelectric
1994;79(4):745–50. electroencephalogram in humans. Anesthesiology 1995;83(5):980–5.
5. Czosnyka M, Smielewski P, Kirkpatrick P, et al. Continuous assessment of 31. Molnár C, Settakis G, Sárkány P, et al. Effect of sevoflurane on cerebral
the cerebral vasomotor reactivity in head injury. Neurosurgery 1997;41:11–7. blood flow and cerebrovascular resistance at surgical level of anaesthesia: a
6. Lam AM, Newell DW. Intraoperative use of transcranial Doppler transcranial Doppler study. Eur J Anaesthesiol 2006;24(02):179–84.
ultrasonography. Neurosurg Clin N Am 1996;7:709–22. 32. Matta BF, Heath KJ, Tipping K, et al. Direct cerebral vasodilatory effects of
7. Kincaid MS. Transcranial Doppler ultrasonography: a diagnostic tool of sevoflurane and isoflurane. Anesthesiology 1999;91(3):677–80.
increasing utility. Curr Opin Anaesthesiol 2008;21(5):552–9. 33. Bundgaard H, von Oettingen G, Larsen KM, et al. Effects of sevoflurane on
8. Schatlo B, Pluta RM. Clinical applications of transcranial Doppler intracranial pressure, cerebral blood flow and cerebral metabolism:
sonography. Rev Rec Clin Trials 2007;2(1):49–57. a dose-response study in patients subjected to craniotomy for cerebral
9. Ogasawara K, Suga Y, Sasaki M, et al. Intraoperative microemboli and low tumours. Acta Anaesthesiol Scand 1998;42(6):621–7.
middle cerebral artery blood flow velocity are additive in predicting 34. Wong GT, Luginbuehl I, Karsli C, et al. The effect of sevoflurane on cerebral
development of cerebral ischemic events after carotid endarterectomy. Stroke autoregulation in young children as assessed by the transient hyperemic
2008;39:3088–91. response. Anesth Analg 2006;102(4):1051–5.
10. Ackerstaff RG, Moons KG, van de Vlasakker CJ, et al. Association of 35. Nishiyama T, Matsukawa T, Yokoyama T, et al. Cerebrovascular carbon
intraoperative transcranial Doppler monitoring variables with stroke from dioxide reactivity during general anesthesia: a comparison between
carotid endarterectomy. Stroke 2000;31(8):1817–23. sevoflurane and isoflurane. Anesth Analg 1999;89(6):1437–41.
Section I—Background 81

36. Bedforth NM, Hardman JG, Nathanson MH. Cerebral hemodynamic 45. Aono M, Sato J, Nishino T. Nitrous oxide increases normocapnic cerebral
response to the introduction of desflurane: a comparison with sevoflurane. blood flow velocity but does not affect the dynamic cerebrovascular response
Anesth Analg 2000;91(1):152–5. to step changes in end-tidal P(CO2) in humans. Anesth Analg
37. Bedforth NM, Girling KJ, Skinner HJ, et al. Effects of desflurane on cerebral 1999;89(3):684–9.
autoregulation. Br J Anaesth 2001;87(2):193–7. 46. Conti A, Iacopino DG, Fodale V, et al. Cerebral haemodynamic changes
38. Brenet O, Granry JC, Poirier N, et al. The effect of desflurane on cerebral during propofol-remifentanil or sevoflurane anaesthesia: transcranial
blood flow velocity and cerebrovascular reactivity to CO2 in children. Doppler study under bispectral index monitoring. Br J Anaesth
Ann Fr Anesth Reanim 1998;17(3):227–33. 2006;97(3):333–9.
39. Summors AC, Gupta AK, Matta BF. Dynamic cerebral autoregulation during 47. Strebel S, Lam AM, Matta B, et al. Dynamic and static cerebral
sevoflurane anesthesia: a comparison with isoflurane. Anesth Analg autoregulation during isoflurane, desflurane, and propofol anesthesia.
1999;88(2):341–5. Anesthesiology 1995;83(1):66–76.
40. McCulloch TJ, Boesel TW, Lam AM. The effect of hypocapnia on the 48. Matta BF, Lam AM, Strebel S, et al. Cerebral pressure autoregulation and
autoregulation of cerebral blood flow during administration of isoflurane. carbon dioxide reactivity during propofol-induced EEG suppression. Br J
Anesth Analg 2005;100(5):1463–7. Anaesth 1995;74(2):159–63.
41. Leon JE, Bissonnette B. Cerebrovascular responses to carbon dioxide in 49. Thiel A, Zickmann B, Roth H, et al. Effects of intravenous anesthetic agents
children anaesthetized with halothane and isoflurane. Can J Anaesth on middle cerebral artery blood flow velocity during induction of general
1991;38(7):817–25. anesthesia. J Clin Monit 1995;11(2):92–8.
42. Lam AM, Mayberg TS, Eng CC, et al. Nitrous oxide-isoflurane anesthesia 50. Kochs E, Werner C, Hoffman WE, et al. Concurrent increases in brain
causes more cerebral vasodilation than an equipotent dose of isoflurane in electrical activity and intracranial blood flow velocity during low-dose
humans. Anesth Analg 1994;78(3):462–8. ketamine anaesthesia. Can J Anaesth 1991;38(7):826–30.
43. Matta BF, Lam AM. Nitrous oxide increases cerebral blood flow velocity
during pharmacologically induced EEG silence in humans. J Neurosurg A complete list of references for this chapter can be found online at
Anesthesiol 1995;7(2):89–93. www.expertconsult.com.
44. Iacopino DG, Conti A, Battaglia C, et al. Transcranial Doppler ultrasound
study of the effects of nitrous oxide on cerebral autoregulation during
neurosurgical anesthesia: a randomized controlled trial. J Neurosurg
2003;99(1):58–64.
Section I—Background 81.e1

References 25. Magee TR, Davies AH, Horrocks M. Transcranial Doppler evaluation of
cerebral hyperperfusion syndrome after carotid endarterectomy. Eur J Vasc
1. Fukui K, Morioka T, Hashiguchi K, et al. Relationship between regional Surg 1994;8(1):104–6.
cerebral blood flow and electrocorticographic activities under sevoflurane 26. Riles TS, Imparato AM, Jacobowitz GR, et al. The cause of perioperative
and isoflurane anesthesia. J Clin Neurophysiol 2010;27(2):110–5. stroke after carotid endarterectomy. J Vasc Surg 1994;19(2):206–16.
2. Mielck F, Brauer A, Radke O, et al. Changes of jugular venous blood 27. Spencer MP. Transcranial Doppler monitoring and causes of stroke from
temperature associated with measurements of cerebral blood flow using the carotid endarterectomy. Stroke 1997;28(4):685–91.
transcerebral double-indicator dilution technique. Eur J Anaesthesiol 28. Fodale V, Schifilliti D, Conti A, et al. Transcranial Doppler and anesthetics.
2004:289–95. Acta Anaesthesiol Scand 2007;51(7):839–47.
3. Yoshitani K, Kawaguchi M, Iwata M, et al. Comparison of changes in 29. Lam AM, Matta BF, Mayberg TS, et al. Change in cerebral blood flow
jugular venous bulb oxygen saturation and cerebral oxygen saturation velocity with onset of EEG silence during inhalation anesthesia in humans:
during variations of haemoglobin concentration under propofol and evidence of flow-metabolism coupling? J Cereb Blood Flow Metab
sevoflurane anaesthesia. Br J Anaesth 2005;94(3):341–6. 1995;15(4):714–7.
4. Matta BF, Lam AM, Mayberg TS, et al. A critique of the intraoperative use 30. Matta BF, Mayberg TS, Lam AM. Direct cerebrovasodilatory effects of
of jugular venous bulb catheters during neurosurgical procedures. Anesth halothane, isoflurane, and desflurane during propofol-induced isoelectric
Analg 1994;79(4):745–50. electroencephalogram in humans. Anesthesiology 1995;83(5):980–5.
5. Czosnyka M, Smielewski P, Kirkpatrick P, et al. Continuous assessment of 31. Molnár C, Settakis G, Sárkány P, et al. Effect of sevoflurane on cerebral
the cerebral vasomotor reactivity in head injury. Neurosurgery 1997; blood flow and cerebrovascular resistance at surgical level of anaesthesia: a
41:11–7. transcranial Doppler study. Eur J Anaesthesiol 2006;24(02):179–84.
6. Lam AM, Newell DW. Intraoperative use of transcranial Doppler 32. Matta BF, Heath KJ, Tipping K, et al. Direct cerebral vasodilatory effects of
ultrasonography. Neurosurg Clin N Am 1996;7:709–22. sevoflurane and isoflurane. Anesthesiology 1999;91(3):677–80.
7. Kincaid MS. Transcranial Doppler ultrasonography: a diagnostic tool of 33. Bundgaard H, von Oettingen G, Larsen KM, et al. Effects of sevoflurane on
increasing utility. Curr Opin Anaesthesiol 2008;21(5):552–9. intracranial pressure, cerebral blood flow and cerebral metabolism:
8. Schatlo B, Pluta RM. Clinical applications of transcranial Doppler a dose-response study in patients subjected to craniotomy for cerebral
sonography. Rev Rec Clin Trials 2007;2(1):49–57. tumours. Acta Anaesthesiol Scand 1998;42(6):621–7.
9. Ogasawara K, Suga Y, Sasaki M, et al. Intraoperative microemboli and low 34. Wong GT, Luginbuehl I, Karsli C, et al. The effect of sevoflurane on
middle cerebral artery blood flow velocity are additive in predicting cerebral autoregulation in young children as assessed by the transient
development of cerebral ischemic events after carotid endarterectomy. hyperemic response. Anesth Analg 2006;102(4):1051–5.
Stroke 2008;39:3088–91. 35. Nishiyama T, Matsukawa T, Yokoyama T, et al. Cerebrovascular carbon
10. Ackerstaff RG, Moons KG, van de Vlasakker CJ, et al. Association of dioxide reactivity during general anesthesia: a comparison between
intraoperative transcranial Doppler monitoring variables with stroke from sevoflurane and isoflurane. Anesth Analg 1999;89(6):1437–41.
carotid endarterectomy. Stroke 2000;31(8):1817–23. 36. Bedforth NM, Hardman JG, Nathanson MH. Cerebral hemodynamic
11. Halsey JH. Risks and benefits of shunting in carotid endarterectomy. The response to the introduction of desflurane: a comparison with sevoflurane.
International Transcranial Doppler Collaborators. Stroke Anesth Analg 2000;91(1):152–5.
1992;23(11):1583–7. 37. Bedforth NM, Girling KJ, Skinner HJ, et al. Effects of desflurane on cerebral
12. Moritz S, Kasprzak P, Arlt M, et al. Accuracy of cerebral monitoring in autoregulation. Br J Anaesth 2001;87(2):193–7.
detecting cerebral ischemia during carotid endarterectomy: a comparison 38. Brenet O, Granry JC, Poirier N, et al. The effect of desflurane on cerebral
of transcranial Doppler sonography, near-infrared spectroscopy, stump blood flow velocity and cerebrovascular reactivity to CO2 in children. Ann
pressure, and somatosensory evoked potentials. Anesthesiology 2007; Fr Anesth Reanim 1998;17(3):227–33.
107(4):563–9. 39. Summors AC, Gupta AK, Matta BF. Dynamic cerebral autoregulation
13. Ackerstaff RGA. Cerebral circulation monitoring in carotid endarterectomy during sevoflurane anesthesia: a comparison with isoflurane. Anesth Analg
and carotid artery stenting. Front Neurol Neurosci 2006;21229–38. 1999;88(2):341–5.
14. Gossetti B, Gattuso R, Irace L, et al. Embolism to the brain during carotid 40. McCulloch TJ, Boesel TW, Lam AM. The effect of hypocapnia on the
stenting and surgery. Acta Chir Belg 2007;107(2):151–4. autoregulation of cerebral blood flow during administration of isoflurane.
15. Telman G, Kouperberg E, Eran A, et al. Microemboli in MCA ipsilateral to Anesth Analg 2005;100(5):1463–7.
occluded common carotid artery: an observation and short review of the 41. Leon JE, Bissonnette B. Cerebrovascular responses to carbon dioxide in
literature. Neurol Res 2008;30(7):684–6. children anaesthetized with halothane and isoflurane. Can J Anaesth
16. Imray CHE, Pattinson KTS. Potential role for TCD-directed antiplatelet 1991;38(7):817–25.
agents in symptomatic carotid artery dissection. Stroke 2006;37(3):767. 42. Lam AM, Mayberg TS, Eng CC, et al. Nitrous oxide-isoflurane anesthesia
17. Zuromskis T, Wetterholm R, Lindqvist JF, et al. Prevalence of micro-emboli causes more cerebral vasodilation than an equipotent dose of isoflurane in
in symptomatic high grade carotid artery disease: a transcranial Doppler humans. Anesth Analg 1994;78(3):462–8.
study. Eur J Vasc Endovasc Surg 2008;35(5):534–40. 43. Matta BF, Lam AM. Nitrous oxide increases cerebral blood flow velocity
18. Alexandrov AV, Babikian VL, Adams RJ, et al. The evolving role of during pharmacologically induced EEG silence in humans. J Neurosurg
transcranial Doppler in stroke prevention and treatment. J Stroke Anesthesiol 1995;7(2):89–93.
Cerebrovasc Dis 1998;7(2):101–4. 44. Iacopino DG, Conti A, Battaglia C, et al. Transcranial Doppler ultrasound
19. Roy J, Akhtar N, Watson T, et al. Transcranial Doppler microembolic signal study of the effects of nitrous oxide on cerebral autoregulation during
monitoring is useful in diagnosis and treatment of carotid artery dissection: neurosurgical anesthesia: a randomized controlled trial. J Neurosurg
two case reports. J Neuroimaging 2007;17(4):350–2. 2003;99(1):58–64.
20. Sfyroeras GS, Karkos CD, Arsos G, et al. Cerebral hyperperfusion after 45. Aono M, Sato J, Nishino T. Nitrous oxide increases normocapnic cerebral
carotid stenting: a transcranial doppler and spect study. Vasc Endovascular blood flow velocity but does not affect the dynamic cerebrovascular
Surg 2009;43:150–6. response to step changes in end-tidal P(CO2) in humans. Anesth Analg
21. Adhiyaman V, Alexander S. Cerebral hyperperfusion syndrome following 1999;89(3):684–9.
carotid endarterectomy. Q J Med 2007;100(4):239–44. 46. Conti A, Iacopino DG, Fodale V, et al. Cerebral haemodynamic changes
22. Ogasawara K, Sakai N, Kuroiwa T, et al. Intracranial hemorrhage associated during propofol-remifentanil or sevoflurane anaesthesia: transcranial
with cerebral hyperperfusion syndrome following carotid endarterectomy Doppler study under bispectral index monitoring. Br J Anaesth
and carotid artery stenting: retrospective review of 4494 patients. J 2006;97(3):333–9.
Neurosurg 2007;107(6):1130–6. 47. Strebel S, Lam AM, Matta B, et al. Dynamic and static cerebral
23. Jansen C, Sprengers AM, Moll FL, et al. Prediction of intracerebral autoregulation during isoflurane, desflurane, and propofol anesthesia.
haemorrhage after carotid endarterectomy by clinical criteria and Anesthesiology 1995;83(1):66–76.
intraoperative transcranial Doppler monitoring. Eur J Vasc Surg 48. Matta BF, Lam AM, Strebel S, et al. Cerebral pressure autoregulation and
1994;8(3):303–8. carbon dioxide reactivity during propofol-induced EEG suppression. Br J
24. Jansen C, Sprengers AM, Moll FL, et al. Prediction of intracerebral Anaesth 1995;74(2):159–63.
haemorrhage after carotid endarterectomy by clinical criteria and 49. Thiel A, Zickmann B, Roth H, et al. Effects of intravenous anesthetic agents
intraoperative transcranial Doppler monitoring: results of 233 operations. on middle cerebral artery blood flow velocity during induction of general
Eur J Vasc Surg 1994;8(2):220–5. anesthesia. J Clin Monit 1995;11(2):92–8.
81.e2 Section I—Background

50. Kochs E, Werner C, Hoffman WE, et al. Concurrent increases in brain 76. Lotto ML, Banoub M, Schubert A. Effects of anesthetic agents and
electrical activity and intracranial blood flow velocity during low-dose physiologic changes on intraoperative motor evoked potentials. J Neurosurg
ketamine anaesthesia. Can J Anaesth 1991;38(7):826–30. Anesthesiol 2004;16(1):32–42.
51. Mayberg TS, Lam AM, Matta BF, et al. Ketamine does not increase cerebral 77. Zentner J, Albrecht T, Heuser D. Influence of halothane, enflurane, and
blood flow velocity or intracranial pressure during isoflurane/nitrous oxide isoflurane on motor evoked potentials. Neurosurgery 1992;31(2):298–305.
anesthesia in patients undergoing craniotomy. Anesth Analg 78. Haghighi SS, Green KD, Oro JJ, et al. Depressive effect of isoflurane
1995;81(1):84–9. anesthesia on motor evoked potentials. Neurosurgery 1990;26(6):993–7.
52. Engelhard K, Werner C, Möllenberg O, et al. S(+)-ketamine/propofol 79. Lyon R, Feiner J, Lieberman JA. Progressive suppression of motor evoked
maintain dynamic cerebrovascular autoregulation in humans. Can J potentials during general anesthesia: the phenomenon of “anesthetic fade.”
Anaesth 2001;48(10):1034–9. J Neurosurg Anesthesiol 2005;17(1):13–19.
53. Ogawa Y, Iwasaki K, Aoki K, et al. Dexmedetomidine weakens dynamic 80. Ubags LH, Kalkman CJ, Been HD. Influence of isoflurane on myogenic
cerebral autoregulation as assessed by transfer function analysis and the motor evoked potentials to single and multiple transcranial stimuli during
thigh cuff method. Anesthesiology 2008;109(4):642–50. nitrous oxide/opioid anesthesia. Neurosurgery 1998;43(1):90–4.
54. Drummond JC, Dao AV, Roth DM, et al. Effect of dexmedetomidine on 81. Pelosi L, Stevenson M, Hobbs GJ, et al. Intraoperative motor evoked
cerebral blood flow velocity, cerebral metabolic rate, and carbon dioxide potentials to transcranial electrical stimulation during two anaesthetic
response in normal humans. Anesthesiology 2008;108(2):225–32. regimens. Clin Neurophysiol 2001;112(6):1076–87.
55. Kofke WA, Dong ML, Bloom M, et al. Transcranial Doppler 82. Pechstein U, Nadstawek J, Zentner J, et al. Isoflurane plus nitrous oxide
ultrasonography with induction of anesthesia for neurosurgery. versus propofol for recording of motor evoked potentials after high
J Neurosurg Anesthesiol 1994;6(2):89–97. frequency repetitive electrical stimulation. Electroencephalogr Clin
56. Thiel A, Zickmann B, Roth H, et al. Effects of intravenous anesthetic agents Neurophysiol 1998;108(2):175–81.
on middle cerebral artery blood flow velocity during induction of general 83. Lo Y, Dan Y, Tan YE, et al. Intraoperative motor-evoked potential
anesthesia. J Clin Monit 1995;11(2):92–8. monitoring in scoliosis surgery: comparison of desflurane/nitrous oxide
57. Cold GE, Eskesen V, Eriksen H, et al. CBF and CMRO2 during continuous with propofol total intravenous anesthetic regimens. J Neurosurg
etomidate infusion supplemented with N2O and fentanyl in patients with Anesthesiol 2006;18(3):211–4.
supratentorial cerebral tumour: a dose-response study. Acta Anaesthesiol 84. Kawaguchi M, Inoue S, Kakimoto M, et al. The effect of sevoflurane on
Scand 1985;29(5):490–4. myogenic motor-evoked potentials induced by single and paired
58. Cheng MA, Hoffman WE, Baughman VL, et al. The effects of midazolam transcranial electrical stimulation of the motor cortex during nitrous oxide/
and sufentanil sedation on middle cerebral artery blood flow velocity in ketamine/fentanyl anesthesia. J Neurosurg Anesthesiol 1998;10(3):131–6.
awake patients. J Neurosurg Anesthesiol 1993;5(4):232–6. 85. Reinacher PC, Priebe H, Blumrich W, et al. The effects of stimulation
59. Hänel F, Werner C, von Knobelsdorff G, et al. The effects of fentanyl and pattern and sevoflurane concentration on intraoperative motor-evoked
sufentanil on cerebral hemodynamics. J Neurosurg Anesthesiol potentials. Anesth Analg 2006;102(3):888–95.
1997;9(3):223–7. 86. Rampil IJ, King BS. Volatile anesthetics depress spinal motor neurons.
60. Fodale V, Schifilliti D, Praticò C, et al. Remifentanil and the brain. Acta Anesthesiology 1996;85(1):129–34.
Anaesthesiol Scand 2008;52(3):319–26. 87. Ubags LH, Kalkman CJ, Been HD, et al. Differential effects of nitrous oxide
61. Frost EA. Inhalation anaesthetic agents in neurosurgery. Br J Anaesth and propofol on myogenic transcranial motor evoked responses during
1984;56(Suppl 1):47S-56S. sufentanil anaesthesia. Br J Anaesth 1997;79(5):590–4.
62. Kennedy RR, Minto C, Seethepalli A. Effect-site half-time for burst 88. van Dongen EP, ter Beek HT, Schepens MA, et al. The influence of nitrous
suppression is longer than for hypnosis during anaesthesia with oxide to supplement fentanyl/low-dose propofol anesthesia on transcranial
sevoflurane. Br J Anaesth 2008;100(1):72–7. myogenic motor-evoked potentials during thoracic aortic surgery. J
63. Schwartz AE, Tuttle RH, Poppers PJ. Electroencephalographic burst Cardiothorac Vasc Anesth 1999;13(1):30–4.
suppression in elderly and young patients anesthetized with isoflurane. 89. van Dongen EP, ter Beek HT, Schepens MA, et al. Effect of nitrous oxide on
Anesth Analg 1989;68(1):9–12. myogenic motor potentials evoked by a six pulse train of transcranial
64. Voss LJ, Seleigh JW, Barnard JPM, et al. The howling cortex: seizures and electrical stimuli: a possible monitor for aortic surgery. Br J Anaesth
general anesthetic drugs. Anesth Analg 2008;107:1689–703. 1999;82(3):323–8.
65. Kofke WA. Anesthetic management of the patient with epilepsy or prior 90. Scheufler KM, Reinacher PC, Blumrich W, et al. The modifying effects of
seizures. Curr Opin Anaesthesiol 2010;23:391–9. stimulation pattern and propofol plasma concentration on motor-evoked
66. Mäkelä JP, Iivanainen M, Pieninkeroinen IP, et al. Seizures associated with potentials. Anesth Analg 2005;100(2):440–7.
propofol anesthesia. Epilepsia 1993;34(5):832–5. 91. Kalkman CJ, Drummond JC, Ribberink AA, et al. Effects of propofol,
67. Jääskeläinen SK, Kaisti K, Suni L, et al. Sevoflurane is epileptogenic in etomidate, midazolam, and fentanyl on motor evoked responses to
healthy subjects at surgical levels of anesthesia. Neurology 2003; transcranial electrical or magnetic stimulation in humans. Anesthesiology
61(8):1073–8. 1992;76(4):502–9.
68. Mustola ST, Baer GA, Toivonen JK, et al. Electroencephalographic burst 92. Inoue S, Kawaguchi M, Kakimoto M, et al. Amplitudes and intrapatient
suppression versus loss of reflexes anesthesia with propofol or thiopental: variability of myogenic motor evoked potentials to transcranial electrical
differences of variance in the catecholamine and cardiovascular response to stimulation during ketamine/N2O- and propofol/N2O-based anesthesia.
tracheal intubation. Anesth Analg 2003;97(4):1040–5. J Neurosurg Anesthesiol 2002;14(3):213–7.
69. Souter MJ, Rozet I, Ojemann JG, et al. Dexmedetomidine sedation during 93. Ubags LH, Kalkman CJ, Been HD, et al. The use of ketamine or etomidate
awake craniotomy for seizure resection: effects on electrocorticography. to supplement sufentanil/N2O anesthesia does not disrupt monitoring of
J Neurosurg Anesthesiol 2007;19(1):38–44. myogenic transcranial motor evoked responses. J Neurosurg Anesthesiol
70. Rozet I, Muangman S, Vavilala MS, et al. Clinical experience with 1997;9(3):228–33.
dexmedetomidine for implantation of deep brain stimulators in Parkinson’s 94. Zaarour C, Engelhardt T, Strantzas S, et al. Effect of low-dose ketamine on
disease. Anesth Analg 2006;103:1244–8. voltage requirement for transcranial electrical motor evoked potentials in
71. Lam AM, Sharar SR, Mayberg TS, et al. Isoflurane compared with nitrous children. Spine 2007;32(22):E627–30.
oxide anaesthesia for intraoperative monitoring of somatosensory-evoked 95. Lang EW, Beutler AS, Chesnut RM, et al. Myogenic motor-evoked potential
potentials. Can J Anaesth 1994;41(4):295–300. monitoring using partial neuromuscular blockade in surgery of the spine.
72. Manninen PH, Lam AM, Nicholas JF. The effects of isoflurane and Spine 1996;21(14):1676–86.
isoflurane-nitrous oxide anesthesia on brainstem auditory evoked 96. Tobias JD, Goble TJ, Bates G, et al. Effects of dexmedetomidine on
potentials in humans. Anesth Analg 1985;64(1):43–7. intraoperative motor and somatosensory evoked potential monitoring
73. de Haan P, Kalkman CJ. Spinal cord monitoring: somatosensory- and during spinal surgery in adolescents. Paediatr Anaesth 2008;18:1082–8.
motor-evoked potentials. Anesthesiol Clin N Am 2001;19(4):923–45. 97. Bala E, Sessler DI, Nair DR, et al. Motor and somatosensory evoked
74. Calancie B, Harris W, Brindle GF, et al. Threshold-level repetitive potentials are well maintained in patients given dexmedetomidine during
transcranial electrical stimulation for intraoperative monitoring of central spine surgery. Anesthesiology 2008;109(3):417–25.
motor conduction. J Neurosurg 2001;95(2 Suppl):161–8. 98. Mahmoud M, Sadhasivam S, Salisbury S, et al. Susceptibility of transcranial
75. MacDonald DB. Safety of intraoperative transcranial electrical stimulation electric motor-evoked potentials to varying targeted blood levels of
motor evoked potential monitoring. J Clin Neurophysiol 2002;19(5): dexmedetomidine during spine surgery. Anesthesiology 2010;
416–29. 112(6):1364–73.
Section I—Background 81.e3

99. Mahmoud M, Sadhasivam S, Sestokas AK, et al. Loss of transcranial electric 107. Dujovny M, Slavin KV, Hernandez G, et al. Use of cerebral oximetry to
motor evoked potentials during pediatric spine surgery with monitor brain oxygenation reserves for skull base surgery. Skull Base Surg
dexmedetomidine. Anesthesiology 2007;106(2):393–6. 1994;4(3):117–21.
100. Scheufler K, Zentner J. Total intravenous anesthesia for intraoperative 108. Kakihana Y, Matsunaga A, Yasuda T, et al. Brain oxymetry in the operating
monitoring of the motor pathways: an integral view combining clinical and room: current status and future directions with particular regard to
experimental data. J Neurosurg 2002;96(3):571–9. cytochrome oxidase. J Biomed Opt 2008;13(3):033001.
101. van Dongen EP, ter Beek HT, Schepens MA, et al.Within-patient variability 109. McDonagh DL, McDaniel MR, Monk TG. The effect of intravenous indigo
of myogenic motor-evoked potentials to multipulse transcranial electrical carmine on near-infrared cerebral oximetry. Anesth Analg 2007;105(3):
stimulation during two levels of partial neuromuscular blockade in aortic 704–6.
surgery. Anesth Analg 1999;88(1):22–7. 110. Patel J, Marks K, Roberts I, et al. Measurement of cerebral blood flow in
102. Hayashi H, Kawaguchi M, Yamamoto Y, et al. The application of tetanic newborn infants using near infrared spectroscopy with indocyanine green.
stimulation of the unilateral tibial nerve before transcranial stimulation can Pediatr Res 1998;43(1):34–9.
augment the amplitudes of myogenic motor-evoked potentials from the 111. McCormick PW, Stewart M, Lewis G, et al. Intracerebral penetration of
muscles in the bilateral upper and lower limbs. Anesth Analg 2008; infrared light. Technical note. J Neurosurg 1992;76(2):315–8.
107(1):215–20. 112. Nishikawa K, Kanemaru Y, Hagiwara R, et al. The influence of sevoflurane
103. Yamamoto Y, Kawaguchi M, Hayashi H, et al. The effects of the on the bispectral index, regional cerebral oxygen saturation, and propofol
neuromuscular blockade levels on amplitudes of posttetanic motor-evoked concentration during propofol/N2O anesthesia. J Clin Monit Comput
potentials and movement in response to transcranial stimulation in 2006;20(6):415–20.
patients receiving propofol and fentanyl anesthesia. Anesth Analg 113. Fassoulaki A, Kaliontzi H, Petropoulos G, et al. The effect of desflurane and
2008;106(3):930–4. sevoflurane on cerebral oximetry under steady-state conditions. Anesth
104. Smith M. Perioperative uses of transcranial perfusion monitoring. Analg 2006;102(6):1830–5.
Anesthesiol Clin 2007;25(3):557–77. 114. Kanemaru Y, Nishikawa K, Goto F. Bispectral index and regional cerebral
105. Duffy CM, Manninen PH, Chan A, et al. Comparison of cerebral oximeter oxygen saturation during propofol/N2O anesthesia. Can J Anaesth.
and evoked potential monitoring in carotid endarterectomy. Can J Anaesth 2006;53(4):363–9.
1997;44(10):1077–81. 115. Iwasaki K, Nomoto Y, Ishiwata M, et al. Vital capacity induction with 8%
106. Bhatia R, Hampton T, Malde S, et al. The application of near-infrared sevoflurane and N2O causes cerebral hyperemia. J Anesth 2003;17(1):3–7.
oximetry to cerebral monitoring during aneurysm embolization: a 116. Lovell AT, Owen-Reece H, Elwell CE, et al. Continuous measurement of
comparison with intraprocedural angiography. J Neurosurg Anesthesiol cerebral oxygenation by near infrared spectroscopy during induction of
2007;19(2):97–104. anesthesia. Anesth Analg 1999;88(3):554–8.
II
Chapter
10  

Clinical Assessment in the


Neurocritical Care Unit
Ramani Balu, John A. Detre, and Joshua M. Levine

Because brainstem arousal centers are close to other impor-


Introduction tant nuclei and tracts that control eye movements and breath-
Many patients in the neurocritical care unit (NCCU) are in ing, the location of lesions causing global impairments in
coma, have altered consciousness, or have a disease process consciousness often can be readily determined by careful
that may lead to altered consciousness. Coma and other dis- physical examination.
orders of consciousness are common clinical problems that In the early 20th century Baron Constantin von Economo
indicate a severe disturbance of brain function. Monitoring first proposed the concept that specific brainstem nuclei
and management of patients with disturbed consciousness control arousal and alertness after his detailed clinicopatho-
depend in large part on the underlying etiology. The princi- logic study of patients with encephalitis lethargica.1 The vast
pal means of monitoring the comatose patient are clinical majority of these patients had profound somnolence along
neurologic examinations, the use of rating scales, neuroimag- with parkinsonism and focal oculomotor abnormalities, and
ing, and electrophysiologic tests. In addition, a variety of spent only a few hours awake each day. Postmortem analysis
invasive and noninvasive monitors of brain function is avail- of these patients demonstrated damage to the paramedian
able and discussed in other chapters of this book. Goals of reticular formation near the junction between the midbrain
monitoring include establishing an etiologic diagnosis and and the diencephalon. A small subset of patients who suffered
excluding coma mimics, determining the location and sever- from debilitating insomnia in contrast had damage to the
ity of injury, assessing response to therapy, and determining anterior hypothalamus. Based on these results, von Economo
prognosis. This chapter provides an overview of the anatomy proposed that a specific arousal-promoting center exists in the
of consciousness, the general clinical approach to the coma midbrain, and a separate sleep-promoting center is in the
patient, and neurologic assessment, with a focus on the clini- hypothalamus.
cal examination, quantitative coma scales, and electrophysio- Studies by Morruzi and Magoun in the 1940s2 showed that
logic and imaging studies that may provide insight into the selective lesions of the midbrain paramedian reticular forma-
prognosis of coma. In addition, other critical care scores that tion in cats caused electroencephalographic slowing and
provide insight into extracerebral pathophysiology are briefly behavioral unresponsiveness similar to sleep. Lesions in the
discussed. midbrain that contained ascending sensory pathways caused
defects in somatosensory and auditory-evoked responses but
had no effect on the normal desynchronized electroencepha-
Anatomy of Brain Circuits lographic pattern seen in awake animals. These investigators
Involved in Arousal, Alertness, coined the term ascending reticular activating system (ARAS)
to describe the critical function of the paramedian reticular
and Conscious Behavior formation in alertness. Subsequent lesion studies in humans
Level of arousal (alertness) depends on the coordinated activ- have confirmed the importance of the upper brainstem to
ity of multiple brainstem nuclei that send projections to promote and maintain behavioral arousal. Specifically, injury
diencephalic targets in the thalamus and hypothalamus and to either the paramedian midbrain or bilateral dorsolateral
diffusely to the cerebral cortex. Areas of the thalamus that pontine tegmentum causes coma.3 Lesions of other parts of
receive information from arousal centers in the brainstem in the brainstem outside of this ascending arousal system,
turn send widespread projections to both cerebral hemi- although often debilitating, do not cause global impairments
spheres. Thus isolated brainstem lesions, bilateral thalamic of consciousness.3,4 Multiple brainstem nuclei that form dis-
damage, or diffuse bilateral cerebral injury can cause global tinct cholinergic and monoaminergic projection systems are
impairments of consciousness and coma. An important corol- now known to constitute the ascending arousal system.4,5
lary to this general rule is that unilateral lesions should not Although these nuclei are not all located within the parame-
cause global reductions in consciousness unless they are large dian midbrain reticular formation, their output fibers in
enough to produce significant mass effect on the contralateral general course through this structure on their way to targets
hemisphere or there is a preexisting contralateral lesion. in the diencephalon and cerebral cortex.
84 © Copyright 2013 Elsevier Inc. All rights reserved.
Section II—Clinical and Laboratory Assessment 85

Multiple midline brainstem areas throughout the midbrain,


Cholinergic Projection Systems pons, and medulla that comprise the raphe nuclei provide
The main source of arousal promoting inputs to the thalamus serotonergic input to the brain.5 Of these, the rostral raphe
is from the pedunculopontine and laterodorsal tegmental nuclei in the midbrain and pons project diffusely throughout
nuclei in the pons.5-8 These areas send cholinergic projections the cortex, thalamus, and basal ganglia. Serotonin effects on
through the midbrain reticular formation mainly to the intra- these areas are complex and can be either excitatory or inhibi-
laminar thalamic nuclei and the thalamic reticular nucleus, tory. These pathways also are thought to play a major role in
although they also project to the thalamic relay nuclei. The psychiatric diseases such as depression, anxiety, and obsessive-
intralaminar nuclei are excited by their ascending cholinergic compulsive disorder.20,21
input and send widespread diffuse excitatory projections to A widespread histaminergic pathway arises from the tuber-
the cortex. The thalamic reticular nucleus, by contrast, sends omamillary nucleus in the posterior hypothalamus.5 Hista-
local inhibitory projections to thalamocortical relay nuclei, minergic neurons are important in sleep and arousal
and its principal neurons are inhibited by acetylcholine. Thus regulation, and activity of tuberomamillary nucleus neurons
activity in the pedunculopontine and laterodorsal tegmental is greatest during waking and rapid eye movement (REM)
nuclei—which is greatest during awakening and decreases sleep.4,7 Histamine receptor blockers that can cross the blood-
during sleep—both activate diffuse excitatory projections brain barrier, such as diphenhydramine, can exert a powerful
from the intralaminar nuclei and disinhibit excitatory thala- sleep-promoting effect.4
mocortical relay neurons by inhibiting the reticular nucleus.
A second source of cholinergic inputs arises from the basal
forebrain. These neurons receive inputs from brainstem Interactions Between Arousal-Promoting
monoaminergic projection systems (discussed following) and Brain Centers
neighboring excitatory brainstem nuclei and send widespread The anatomy of the pathways described previously and
projections throughout the cerebral cortex.5,9 Interestingly, their aggregate importance in arousal have been recognized
single axons from the basal forebrain have patchy, spatially for decades. However, the specific ways in which these path-
defined projection patterns to defined subsets of cortical ways interact to promote conscious behavior, attention, or
neurons.10 Thus regulated activity of different parts of the memory and regulate the transitions between multiple behav-
basal forebrain can have spatially restricted effects on different ioral states is only beginning to be elucidated. Other inputs,
cortical areas. including the galanin and gamma aminobutyric acid–secret-
ing (GABAergic) inputs from the ventrolateral preoptic
(VLPO) area of the hypothalamus,7,8 orexin-containing
Monoaminergic Projection Systems neurons from the lateral hypothalamus, and descending corti-
Multiple monoaminergic projection systems also provide tel- cal inputs provide modulatory control over the ARAS.8,22,23
encephalic input from the brainstem and diencephalon that Nevertheless, the concept of an ascending brainstem arousal
influence alertness and conscious behavior. Activity of these system whose activity promotes alertness and conscious
monoaminergic projections has complex effects on cortical behavior remains invaluable clinically when evaluating
activity and often decreases background activity but increases patients with disorders of impaired consciousness.
the responsiveness of a cortical neuron to its best stimulus,
thereby increasing signal to noise ratio.11-15 These systems
therefore have critical functions in regulating attention,
Coma Etiology
memory, and other higher order cognitive processes in Causes of altered mental status and coma are protean, and
addition to providing a more general arousal-promoting can be divided into primary brain disorders and systemic
stimulus. derangements that secondarily affect brain function (Table
Dopaminergic neurons are found in the midbrain in both 10.1). Primary brain disorders can be caused by structural
the substantia nigra and in the ventral tegmental area.5 Projec- abnormalities (e.g., cerebral infarction, tumors, subdural
tions from the substantia nigra terminate in the basal ganglia hematomas, intraparenchymal hemorrhages, and abscesses
and are crucial for motor control and modulation. The ventral among others) that either directly distort the circuitry of the
tegmental projections course through the midbrain reticular ascending arousal system or globally increase intracranial
formation and provide a mesolimbic projection to the nucleus pressure (ICP), or by diffuse nonstructural disturbances such
accumbens, basal forebrain, cingulate cortex, hippocampus, as seizures. Often both structural and nonstructural abnor-
and amygdala and a mesocortical projection to the prefrontal malities may exist together in the same patient, for example,
cortex.5 Mesolimbic dopaminergic pathways are involved in a patient with a cerebral abscess who develops seizures. Sys-
reward-mediated behavior,16,17 and overactivity of these pro- temic disturbances cause encephalopathy through diffuse
jections is thought to be central in the pathogenesis of thought bilateral cerebral dysfunction and can include metabolic
disorders such as schizophrenia.18 Mesocortical projections derangements, exposure to toxins, systemic infections, or
likely are involved in attention and working memory.19 diffuse encephalitis caused by primary central nervous system
Noradrenergic projections to the cerebral hemispheres arise (CNS) infections, autoimmune processes, or paraneoplastic
mainly from the locus ceruleus, located in the upper pons near syndromes.
the fourth ventricle. Locus ceruleus activity increases with The most common causes of coma are traumatic brain
awakening, and this area likely is important in behavioral state injury (TBI), hypoxic-ischemic encephalopathy (HIE), drug
switching (i.e., from sleep to wakefulness).7,8 These noradren- overdose, ischemic and hemorrhagic strokes, CNS infections,
ergic systems, like serotonergic systems (see following text), and brain herniation from space-occupying lesions. A detailed
also are pharmaceutical targets in mood disorders.20,21 discussion of these conditions is beyond the scope of this
86 Section II—Clinical and Laboratory Assessment

Table 10.1  A Partial List of the Etiologies


Differential Diagnosis (Mimics) of
of Coma and Altered Mental Status Coma and the Vegetative State
PRIMARY BRAIN DISORDERS
Patients may become unresponsive from conditions that
mimic coma or a vegetative state. Care must be taken not to
Structural Lesions
consider these patients as comatose because they have no
1. Traumatic brain injury impairments of conscious behavior and often can communi-
a. Diffuse axonal injury cate with clinical staff and other individuals if an appropriate
b. Contusions
c. Subdural hematomas communication system is devised.24 In the locked-in syn-
d. Epidural hematomas drome, destruction of the ventral pons leaves the patient
2. Cerebrovascular disorders quadriplegic and mute.4 Patients often are aware of their sur-
a. Ischemic strokes roundings and may communicate only through vertical eye
b. Spontaneous intracerebral hemorrhage
c. Subarachnoid hemorrhage
movements and blinking, which are spared. With more rostral
d. Hypoxic-ischemic encephalopathy pontine lesions, vertical eye movements and blinking are lost.
e. Cerebral venous sinus thrombosis In this state that can be likened to receiving a neuromuscular
3. Malignant disease blocking agent without a sedative, the patient has no means
a. Brain tumors of communication. Severe Guillain-Barré syndrome, botu-
4. Infectious diseases
a. Brain abscesses lism, and critical-illness neuropathy may similarly result in
5. Demyelinating disease complete de-efferentation. Catatonia is a manifestation of
a. Acute disseminated encephalomyelitis severe psychiatric illness in which patients open their eyes, do
b. Central pontine myelinolysis not speak or follow commands, and may exhibit waxy flexibil-
6. Hydrocephalus
ity.4,25 The remainder of the neurologic exam and the electro-
Nonstructural Disorders encephalogram (EEG) are normal. Akinetic mutism that
1. Infectious diseases results from bilateral medial frontal lobe injury is a profound
a. Bacterial meningoencephalitis form of abulia (lack of motivation) in which patients are
b. Viral encephalitis unable to speak or to move, but open their eyes, occasionally
2. Malignant disease
a. Carcinomatous or lymphomatous meningitis
track visual stimuli, and sometimes respond when prompted.26,27
3. Generalized seizures, status epilepticus
4. Basilar migraines
SYSTEMIC DISORDERS Neurologic Examination of the
Toxic Encephalopathies Comatose Patient
1. Medication overdose The initial neurologic examination of a patient with altered
a. Opioids, benzodiazepines, barbiturates, tricyclics, etc. consciousness allows the lesion to be localized: this helps to
2. Illicit drug exposure
a. Opioids, alcohols, amphetamines, etc. narrow the list of etiologic possibilities. For example, integrity
3. Environmental toxin exposure of brainstem function and absence of focal signs suggests a
a. Carbon monoxide toxic or metabolic disorder. In contrast, asymmetric findings
b. Heavy metals and brainstem dysfunction are more consistent with a struc-
c. Pesticides
tural etiology. The exam also is used to exclude conditions that
Metabolic Encephalopathies may mimic coma. Serial examinations of the comatose patient
1. Hypoglycemia, hyperglycemia over time are essential and provide information about treat-
2. Hyponatremia, hypernatremia ment efficacy, progression of the primary process, and prog-
3. Hypercalcemia nosis. The clinical examination of the comatose patient is
4. Hepatic encephalopathy
5. Uremia
discussed first, followed by some of the standardized scales
6. Vitamin deficiencies (thiamine, niacin) that have been developed to evaluate coma (Fig. 10.1).
7. Hypothermia, severe hyperthermia The coma examination is focused on four elements:
8. Hypothyroidism, hyperthyroidism (1) determination of the patient’s level of arousal (wake­
9. Urea cycle disorders fulness), (2) eye examination, (3) motor responses and pres-
Infections ence of abnormal reflexes, and (4) observation of breathing
1. Urinary tract infections patterns.
2. Pneumonia
3. Sepsis
Level of Consciousness
Adapted from Stevens RD, Bhardwaj A. Approach to the comatose patient.
Crit Care Med 2006;34(1):31–41. Patients who exhibit spontaneous eyes opening, verbalization
attempts, moaning, tossing, reaching, leg crossing, yawning,
coughing, or swallowing have a higher level of consciousness
chapter; however, a few warrant special mention because rapid than those who do not. The examiner should next assess
diagnosis and urgent treatment are essential to limit brain the patient’s response to a series of stimuli that escalate in
injury. These include seizures, infections, acute hydrocephalus, intensity. The patient’s name should be called loudly. If there
herniation, ischemic and hemorrhagic strokes, subarachnoid is no response, the examiner may stimulate the patient by
hemorrhage (SAH), cerebral venous sinus thrombosis, hyper- gently shaking him or her. If this produces no response, the
tensive encephalopathy, and TBI.4 examiner should use a noxious stimulus, such as pressure
Section II—Clinical and Laboratory Assessment 87

Initial resuscitation Full Outline of UnResponsiveness [FOUR] score, Reaction


Airway Level Scale 85 [RLS85], and Innsbruck Coma Score are dis-
Breathing cussed later in this chapter.
Circulation
Cervical spine stabilization
The Eye Examination
In coma, the neuro-ophthalmologic examination should focus
Laboratory evaluation
Serum glucose, electrolytes, blood
on: (1) the pupils, (2) resting eye position and eye movements,
gas, osmolality, liver and thyroid (3) retinal appearance, and (4) the corneal reflex.
function tests, ammonia, complete
blood count, toxicology screen
Pupillary Examination
Pupillary examination is perhaps the most important part of
Coma examination the coma examination, because it can help localize and dif-
ferentiate between structural lesions and diffuse metabolic
encephalopathies that cause bilateral cerebral hemispheric
Initial/empiric treatment dysfunction. Pupillary constriction is mediated by cholinergic
Hyperventilation, mannitol 0.75-1.25 g/kg if evidence of parasympathetic efferents that arise from the Edinger-
intercranial hypertension, herniation Westphal nucleus in the midbrain and travel within the ocu-
Thiamine (100 mg IV) then glucose (50 cc of 50% solution)
Naloxone (0.4-2 mg IV q 3 minutes) lomotor nerve. Pupillary dilation, in contrast, is mediated by
Flumazenil (0.2 mg/min to maximum dose of 1 mg IV) if sympathetic efferents originating in the hypothalamus that
benzodiazepine overdose suspected travel through the brainstem and cervical spinal cord and then
Gastric lavage with activated charcoal if other synapse onto postganglionic neurons in the superior cervical
drug intoxication suspected
Antibiotic administration if meningoencephalitis suspected ganglion. These noradrenergic neurons then course along the
Induced hypothermia if comatose after VT/VF carotid artery into the cavernous sinus and through the supe-
Cardiac arrest per ILCOR guidelines rior orbital fissure before reaching the iris.
Pupillary size, shape, and reactivity to light should be
assessed. In general, abnormalities of the pupillary light reflex
Head CT if etiology unknown suggest a structural abnormality. However, certain drugs also
or structural lesion suspected may affect the pupillary light reflex. These agents can cause
pupillary abnormalities that are mistakenly attributed to
structural brain lesions (Table 10.2). Metabolic causes of
Detailed history coma typically do not affect the pupils.
and physical Normal pupils are round, have equal diameters, and briskly
examination constrict when illuminated. When unequal pupils (anisoco-
ria) are observed, it is important to establish whether it is the
larger or the smaller pupil that is abnormal. This is accom-
Lumbar puncture, EEG, plished by examining the eyes both in the light and in the dark.
MRI as indicated
When the lights are extinguished, an abnormally small pupil
Fig. 10.1  Algorithm for initial approach to the comatose patient. will fail to dilate fully and the degree of anisocoria will increase.
CT, Computed tomography; EEG, electroencephalogram; MRI, magnetic In contrast, when the abnormal pupil is the larger one, the
resonance imaging; ILCOR, International Liaison Committee on degree of anisocoria will be maximal under full illumination
Resuscitation; IV, intravenous; VT/VF, Ventricular Tachycardia/Ventricular when the larger pupil fails to constrict fully.
Fibrillation. In the NCCU, the most important causes of a unilaterally
dilated pupil are compressive lesions of the oculomotor nerve
to the supraorbital ridge, nailbeds, or sternum, or nasal tickle complex (e.g., uncal herniation or a posterior communicating
with a cotton wisp. Responses such as grimacing, eye opening, artery aneurysm). A complete third nerve palsy results in ipsi-
grunting, or verbalization should be documented. Motor lateral mydriasis, inferolateral deviation of the eye, and ipsilat-
responses provide information not only about sensation and eral ptosis. As a rule a third nerve palsy with pupil involvement
limb strength but also level of consciousness. The examiner means a surgical lesion (i.e., compressive), whereas when the
should note whether stimuli produce “purposeful,” nonste- pupil is spared the cause is medical (e.g., diabetes, meningo-
reotyped limb movements, for example, reaching toward the vascular syphilis). Patients with myasthenia gravis may present
site of stimulation (“localization”). This implies a degree of with ptosis and ophthalmoparesis secondary to neuromuscu-
intact cortical function. Stereotyped limb movements gener- lar weakness that is often unilateral. Therefore, this condition
ally are mediated by brain and spinal reflexes and do not also should be considered in patients who present with what
require cortical input. Examples include extension and inter- appears to be a pupil-sparing third nerve palsy.
nal rotation of the limbs (decerebrate posturing), upper The most important cause of a unilateral small pupil is
extremity flexion (decorticate posturing), and flexion at the Horner syndrome, which is caused by damage to sympathetic
ankle, knee, and hip (“triple-flexion”). Several scales designed efferents to the eye and consists of miosis and mild ipsilateral
to quantitate level of consciousness are available to help ptosis. Horner syndrome can be seen from multiple areas of
reduce inter- and intraobserver variability and to facilitate brain damage, including (1) hypothalamic injury, (2) brain-
accurate follow-up. These scales (Glasgow Coma Scale [GCS], stem damage to descending inputs that synapse onto cervical
88 Section II—Clinical and Laboratory Assessment

Table 10.2  Drugs That Affect Pupillary Responses


Drug Class Examples Effect on Pupils Comments
Muscarinic Atropine, scopolamine, Dilate pupils by blocking action Ipratropium and tiotropium can cause
antagonists ipratropium, tiotropium, of acetylcholine inadvertent pupillary dilation if
tropicamide accidentally gets into eye during
respiratory treatment. Tropicamide
eyedrops commonly used to dilate pupils
for ophthalmologic evaluation.
Beta agonists Epinephrine, isoproterenol, Dilate pupils by activating Albuterol, salmeterol, and formoterol can
salmeterol, formoterol, beta-adrenergic receptors cause inadvertent pupillary dilation if
albuterol accidentally gets into eye during
respiratory treatment.
Cholinesterase Physostigmine, edrophonium, Constrict pupils by increasing Rivastigmine and donepezil used for
inhibitors pyridostigmine, rivastigmine, acetylcholine concentration treatment of dementia. Pyridostigmine
donepezil, organophosphates at synaptic cleft used for treatment of myasthenia gravis.
Muscarinic Pilocarpine Constrict pupils by activating Pilocarpine eyedrops are used to
agonists muscarinic acetylcholine distinguish pharmacologic pupil dilation
receptors from oculomotor nerve injury.
Serotonin-2A Lysergic acid diethylamide Dilate pupils through unclear Commonly used recreational hallucinogen.
(5HT-2A) (LSD) mechanism
receptor
agonists
Monoamine Cocaine, hydroxyamphetamine Dilate pupils by blocking Hydroxyamphetamine eyedrops can help
reuptake reuptake of norepinephrine distinguish if constricted pupil is from
inhibitors at synaptic cleft preganglionic or postganglionic injury.
Norepinephrine Hydroxyamphetamine Dilate pupils by increasing See above.
release norepinephrine concentration
potentiators at synaptic cleft
Opiate receptor Morphine, fentanyl, Constrict pupils Commonly used analgesics in hospitalized
agonists hydromorphone and critically ill patients

sympathetic preganglionic neurons, (3) cervical spine injury, conjugate deviation to the contralateral side. Downward devi-
(4) injury to sympathetic paravertebral ganglia (e.g., with an ation (or lack of upward gaze) of the eyes is caused by dys-
apical lung mass), or (5) damage to sympathetic postgangli- function of the dorsal midbrain and may be seen with
onic fibers because they course along the internal carotid hydrocephalus, tumors, strokes, and pineal region abnormali-
artery (e.g., carotid artery dissection). Depending on the ties. Dysconjugate gaze frequently is seen in sedated patients
lesion location, ipsilateral facial anhidrosis and other focal and usually represents unmasking of a latent eso- or exopho-
neurologic signs also may be present. The presence of Horner ria. Roving, or slow to-and-fro eye movements, implies
syndrome unequivocally places the lesion ipsilateral to the functional integrity of the brainstem.4 Ocular bobbing—fast
pupillary abnormality. Bilaterally fixed and dilated pupils are conjugate downward gaze followed by a slow upward correc-
seen in the terminal stages of brain death but also with anti- tion to midposition—implies extensive pontine injury.28
cholinergic medications, such as atropine. Hyperadrenergic Ocular dipping—slow conjugate downward gaze followed by
states (e.g., pain, anxiety, cocaine intoxication) produce bilat- fast upward gaze—also localizes to the pons.29 With a skew
erally large and reactive pupils. Reactive pinpoint (<1 mm) deviation, a vertical misalignment of the eyes that is not
pupils are observed with opiate and barbiturate intoxication, caused by an isolated cranial nerve palsy, the lesion usually is
and after extensive pontine injury. Patients with long-standing in the midbrain on the side of the higher eye, or in the ponto-
diabetes mellitus may have small pupils secondary to medullary junction on side of the lower eye.30,31 In comatose
hyperglycemia-induced damage of the sympathetic fibers that patients there is a high incidence of nonconvulsive seizures,
mediate pupillary dilation. and jerking movements of the eyes may be the only evidence
of seizure activity.
If spontaneous eye movements are absent, then an oculo-
Resting Eye Position and Eye Movements cephalic response (“doll’s eyes”) should be sought by turning
Eye position and spontaneous movements including horizon- the head horizontally and vertically. This maneuver should not
tal or vertical misalignment, spontaneous roving, or rhythmic be performed on trauma patients with known or suspected
and repetitive vertical movements should be looked for and cervical spine instability. Normally the eyes move opposite to
documented. The frontal lobe cortex (frontal eye fields) medi- the direction of head turning. Testing the oculocephalic
ates conjugate deviation of the eyes toward the contralateral response may uncover a vertical gaze paresis, a skew deviation,
side. Lateral deviation of both eyes therefore indicates a or a sixth nerve palsy that was not otherwise obvious.
destructive lesion in the ipsilateral frontal lobe or an excitatory If an oculocephalic response cannot be elicited, then an
focus (seizure) in the contralateral hemisphere. A destructive oculovestibular (“cold-caloric”) response is sought. First, the
unilateral pontine lesion, and rarely, a thalamic lesion, causes tympanic membrane should be visualized to ensure that is
Section II—Clinical and Laboratory Assessment 89

intact and unobstructed. The head of the bed should be set at externally rotated. Next, the patient is stimulated and the
30 degrees to align the patient’s horizontal semicircular canals examiner should observe for asymmetry in the patient’s face
parallel to the floor. Then, using an angiocatheter or a but- (grimace) and limb motor responses. A less vigorous response
terfly catheter without the needle, 30 to 60 cc of ice-cold water on one side of the body indicates a contralateral structural
are instilled into the external auditory canal against the tym- lesion that involves the motor pathways above the level of the
panic membrane. This inhibits the ipislateral vestibular system caudal medulla. In general, weakness associated with a pyra-
and normally causes the eyes first to move slowly toward the midal (upper motor neuron) lesion is found in the extensor
ipsilateral ear and then to jerk quickly toward the contralateral muscle groups of the arms and the flexor muscle groups of
ear. The initial slow response is mediated by the unopposed the legs. Hence a patient with a hemiparesis may lie with the
contralateral vestibular system in the brainstem, and the arm flexed and the leg extended. Tone, on the other hand, is
frontal eye fields mediate the subsequent corrective nystag- increased in the muscle groups that remain strong. Testing
mus. With bilateral cortical dysfunction and an intact brain- handgrip strength is not reliable because this function often
stem, slow tonic deviation of the eyes toward the ipislateral is affected late in the development of spastic hemiparesis. In
ear is observed and is not followed by contralateral nystagmus. addition a handgrip may represent an abnormal reflex associ-
In early metabolic coma, the oculocephalic and oculovestibu- ated with frontal lobe disease. Another important reflex is the
lar responses are preserved. Absent response indicates diffuse plantar response. An up-going toe is abnormal and is called a
brainstem dysfunction and is seen in primary brainstem Babinski reflex (i.e., there is no such thing as a negative Babin-
injury, late transtentorial herniation, barbiturate intoxication, ski). This reflex can be difficult to elicit and is best obtained
and brain death. by stroking the lateral border of the sole and then across the
ball of the foot. The toes must not be touched. It is the first
movement of the big toe that is important. For example, an
Retinal Examination individual with very sensitive feet may first flex the big toe and
A funduscopic examination should be considered to look for then extend all the toes and withdraw the leg—this is a normal
signs of intracranial hypertension. However, successful fun- response. An up-going toe is associated with an upper motor
duscopy often requires that the pupil be dilated; if doing this neuron lesion. The presence of an abnormal plantar reflex can
pharmacologically may confuse overall patient assessment, be cross-checked with the abdominal reflex obtained by gently
retinal examination may be deferred. Papilledema is swelling stroking the abdominal wall and looking for abdominal wall
of the optic nerve head from increased ICP. It is almost always muscle contraction. Absence of an abdominal reflex implies
bilateral and may be accompanied by retinal hemorrhages, an upper motor neuron lesion above T9. The reflex can be
exudates, and cotton wool spots, and ultimately by enlarge- difficult to assess in a patient who is obese or has had multiple
ment of the optic cup. Papilledema develops over hours to abdominal surgeries.
days. Its absence therefore does not imply normal ICP, espe- Paraparesis and quadraparesis raise the possibility of spinal
cially in the acute setting. Spontaneous pulsatility of the cord injury, especially in the setting of trauma. In these
retinal veins implies normal ICP. Terson syndrome is vitreous, patients, in whom spinal cord injury or compression is a con-
subhyaloid, or retinal hemorrhage associated with SAH. Pap- sideration, how they respond to sensory stimuli needs to be
illedema itself can be associated with visual loss,32 and in factored into what motor responses occur. Subtle findings
general when seen, even in a patient with normal conscious- such as sweat patterns may help indicate a sensory level in a
ness, should prompt immediate investigation as to its etiology comatose patient with a spinal cord abnormality. By looking
so treatment can be started in a timely manner. at plantar and abdominal reflexes, the level of injury can be
differentiated further. Priapism (reflex erection) results from
loss of sympathetic tone with injury to the spinal cord. The
The Corneal Reflex bulbocavernosus reflex is contraction of the anal sphincter
The cornea of each eye is gently touched with a drop of when the penile shaft is pinched or a Foley catheter is pulled.
saline or a cotton wisp while observing for eyelid closure Its absence is associated with a spinal cord injury. The Ameri-
to test the corneal reflex. Failure of unilateral eyelid closure can Spinal Injury Association (ASIA) Muscle Grading Scale
suggests facial nerve dysfunction on that side. Failure of can be used to classify patients with spinal cord injury (Table
bilateral eyelid closure with stimulation of one cornea, but 10.3). This is different from the ASIA Impairment Scale that
not the other, implies trigeminal nerve dysfunction on the defines the severity of the injury (Table 10.4).
stimulated side. Failure of bilateral eyelid closure on stimula-
tion of either cornea usually implies pontine dysfunction.
Breathing Patterns
Several different breathing patterns that depend on the type
Motor Responses and Abnormal Reflexes of injury and location of the pathology may be observed in
The symmetry of motor responses and reflexes, and the pres- coma. However, in clinical practice breathing patterns often
ence of abnormal movement often permits discrimination are obscured by the use of sedatives, paralytics, and mechani-
between structural and systemic etiologies of altered mental cal ventilation. Apneustic respirations are characterized by a
status. First, the patient should be observed for any abnormal prolonged end-inspiratory pause. This pattern may be seen
or spontaneous movements. Asterixis implies a metabolic dis- after focal injury to the dorsal lower half of the pons (e.g.,
turbance such as uremia or hepatic encephalopathy. Twitching stroke), but also may be observed with meningitis, hypoxia,
or jerking of the face or limbs, even if subtle, raises the suspi- and hypoglycemia. Sustained hyperventilation can be seen in
cion for seizures. Asymmetry of resting limb position may be metabolic encephalopathies such as hepatic coma, sepsis, and
a sign of weakness. For example, a paretic leg may lie diabetic ketoacidosis, but also can be seen as a consequence of
90 Section II—Clinical and Laboratory Assessment

fall along a spectrum, with coma at one end and normal con-
Table 10.3  American Spinal Injury sciousness at the other. Patients who are in coma do not
Association Muscle Grading for Spinal respond to external stimuli in a “purposeful” manner but may
Cord Injury demonstrate reflexive behavior. Their eyes are closed and
Grade Clinical Description sleep-wake cycles are absent. Coma is usually prolonged and
lasts for at least hours to days, but rarely is permanent. Instead
0 Total paralysis
coma progresses to death or to a higher level of consciousness
1 Palpable or visible contraction including the vegetative or minimally conscious state (see
2 Active movement, full range of motion, gravity Chapters 8 and 13). Deterioration of normal consciousness is
eliminated often designated by terms such as “confusion” or “delirium,”
3 Active movement, full range of motion, against “stupor,” and “coma.” These labels are imprecise and have been
gravity defined inconsistently from study to study, and even occasion-
4 Active movement, full range of motion, against ally within a given study. Several attempts have been made to
gravity and provides some resistance codify criteria for coma, vegetative state, minimally conscious
5 Active movement, full range of motion, against
state, or delirium.4 These attempts, although laudable, have
gravity and provides normal resistance not yet led to universal adoption of standard terminology. For
practical purposes for the intensive care unit (ICU) physician,
5* Muscle able to exert, in examiner’s judgment,
sufficient resistance to be considered normal if it is advisable to simply describe the clinical exam and use a
identifiable inhibiting factors were not present validated coma scale.
Several quantitative coma scales have been developed to
measure the degree of coma and extent of neurologic injury
in patients. Standardized scales provide an objective measure
Table 10.4  American Spinal Injury of coma that facilitates communication between physicians
Association Impairment Scale and ancillary medical staff, a means of comparing patients
A: Complete: no motor or sensory function is preserved in the with varying coma etiologies, and a simple mechanism to
sacral segments S4-S5. track clinical changes over time. Scales describing sedation
B: Incomplete: sensory but not motor function is preserved and agitation (e.g., Ramsay Sedation Scale),35 Richmond Agi-
below the neurologic level and includes the sacral tation Sedation Scale,36 Vancouver Interaction and Calmness
segments S4-S5.
C: Incomplete: motor function is preserved below the Scale,37 Riker Sedation-Agitation Scale,38 and the American
neurologic level, and more than half of key muscles below Association of Critical-Care Nurses’ Sedation Assessment
the neurologic level have a muscle grade less than 3. Scale for Critically Ill Patients39 also are used in critical
D: Incomplete: motor function is preserved below the care and are reviewed in Chapter 11. (See also www.mc.
neurologic level, and at least half of key muscles below the
neurologic level have a muscle grade of 3 or more.
vanderbilt.edu/icudelirium/docs/CAM_ICU_training.pdf for
E: Normal: motor and sensory function are normal. training in delirium assessment.)

focal injury to the upper pons—usually either from secondary Glasgow Coma Scale
neurogenic pulmonary edema caused by intrinsic brainstem The GCS (Table 10.5) is the most widely used standardized
injury33 or because of a tumor that creates a local cerebrospi- scale to assess the degree of coma and neurologic injury in
nal fluid (CSF) acidosis, which stimulates brain chemorecep- patients and has been incorporated into several models that
tors to trigger hyperventilation.34 Cluster breathing consists of predict outcome in a variety of neurologic insults. It was ini-
several rapid, shallow breaths followed by a prolonged pause, tially developed for triage in patients with head trauma40,41 but
and localizes to the upper medulla. Ataxic respirations, or it has since been shown to correlate with survival and neuro-
Biot’s breathing, is a chaotic pattern in which the length and logic outcome in TBI,42-46 nontraumatic coma,47 intracerebral
depth of the inspiratory and expiratory phases are irregularly hemorrhage,48,49 SAH,50 dementia,51 and meningitis.52-54 To
irregular. It may occur after injury to the respiratory centers calculate the GCS a patient is given a numeric score in three
in the lower medulla. Apnea may be seen in a variety of neu- categories (eye response, motor response, and verbal response)
rologic and non-neurologic disorders and by itself is of little and then these numbers are added together to get the full score
localizing value. Kussmaul respirations are rapid, deep breaths (see Table 10.5). The score can range from 3 to 15; a GCS of 13
that usually signal metabolic acidosis, but also may be observed or higher signifies mild brain injury, 9 to 12 signifies moderate
with pontomesencephalic lesions. Cheyne-Stokes respiration brain injury and 8 or less signifies severe brain injury and is
refers to alternating spells of apnea and crescendo-decrescendo frequently used to define coma.4 The GCS is simple to calcu-
hyperpnea. It has limited value in lesion localization and is late and provides a rapid estimate of the degree of neurologic
seen with diffuse cerebral injury, hypoxia, hypocapnea, and injury. It has been widely adopted by emergency medical per-
congestive heart failure. Agonal gasps reflect bilateral lower sonnel to evaluate patients both in the field and in the emer-
medullary injury and occur in the terminal stages of brain gency department (ED) to triage and guide initial therapy. The
injury. GCS has been incorporated into other validated scoring
systems that assess the severity of critical illness such as the
Acute Physiology and Chronic Healthy Evaluation (APACHE
Standardized Coma Rating Scales II) score55 and the Simplified Acute Physiology Score (SAPS II)
Coma scales are important because it can be difficult to oth- score56 or of SAH (e.g., the World Federation of Neurosurgical
erwise define levels of consciousness. States of consciousness Societies score; see following text) and independently predicts
Section II—Clinical and Laboratory Assessment 91

The GCS is also problematic in young children because the


Table 10.5  Glasgow Coma Scale verbal component is not appropriate and they will not be able
Motor Response to appropriately follow motor commands.68,69 Several coma
scores have been developed specifically for children in an
Follows commands 6
attempt to compensate for differences in verbal and motor
Localizes pain 5 capabilities.70 The Pediatric GCS modifies the verbal and
Withdraws to pain 4 motor scores such that the highest verbal score is “alert,
Flexion 3
babbles, coos words or sentences—normal for age” and the
highest motor score is “normal spontaneous” movement.71
Extension 2 Another score developed to assess level of consciousness in
None 1 infants with TBI has low interrater reliability and is not in
VERBAL RESPONSE widespread use.72
Oriented 5
Confused speech 4 Reaction Level Scale 85
Inappropriate words 3 Used almost exclusively in Sweden, the RLS8573 is a linear
Incomprehensible 2 score that is based on a patient’s level of consciousness from
1 (unconscious and unresponsive) to 8 (complete conscious-
None 1
ness). Rather than having subscores in different categories that
EYE OPENING are then added together, the RLS85 assigns a single number to
Spontaneous 4 the patient’s level of consciousness. This removes the potential
To command 3
ambiguity of having multiple ways to arrive at the same score
through different permutations. It has been studied and vali-
To pain 2 dated for a variety of neurologic conditions including isch-
None 1 emic and hemorrhagic stroke, TBI, and drug intoxication.74-77
Adapted from Teasdale G, Jennett B. Assessment of coma and impaired
Multiple studies have compared the RLS85 with the GCS and
consciousness: a practical scale. Lancet 1974;2:81–4. have found good correspondence between the two scoring
systems.74,76-78 In addition, one study used the RLS85 to calcu-
late APACHE II scores rather than the GCS and found good
agreement between GCS- and RLS85-based APACHE II
mortality in the general critical care patient.57,58 In addition, scores.77 However, the RLS85 has not been widely adopted and
the GCS is used to classify patients with acute brain injury and so there are few studies to validate interrater reliability.76,77
select them for various clinical trials.43-46 Websites such as In addition, there is some inherent ambiguity in the score
those based on the Corticosteroid Randomisation after Sig- because an RLS85 score of either 2 or 3 can signify “drowsy
nificant Head Injury (CRASH) study (www.crash.lshtm.ac.uk/ with response to strong stimulation.”
Risk%20calculator/index.html) or the International Mission
for Prognosis and Analysis of Clinical Trials in TBI (IMPACT)
project (www.tbi-impact.org/) use the GCS to help provide Innsbruck Coma Score
online prognostic predictor models after brain injury. This score79 is based on eight different categories: (1) eye
Despite its widespread use and proven utility, the GCS has opening, (2) reaction to acoustic stimuli, (3) reaction to pain,
potential limitations. For example, deafness or blindness may (4) body posture, (5) pupil size, (6) pupil response to light,
affect the GCS. In intubated patients, the verbal score cannot (7) position and movements of the eyeballs, and (8) oral
be calculated and usually is removed from the final calcula- automatisms. This score has not gained popularity outside of
tion. The GCS may be difficult to interpret when applied to the hospital where it was initially developed, and interrater
patients with swollen eyelids or eyes that cannot be evaluated reliability is unclear. In addition, the evaluation of “oral
accurately, locked-in patients, aphasic patients, or intoxicated automatisms” may be ambiguous in a comatose patient with
patients who have slurred speech and fluctuating compliance. occasional reflexive movements.80
In these circumstances the GCS motor score often is used
alone and seems to carry adequate predictor information.59,60
In addition, a given score in the midrange (6-12) may be Full Outline of Unresponsiveness Score
assigned to patients with different degrees of impaired con- The FOUR score was developed in an attempt to rectify the
sciousness through different combinations of scores in each difficulties that may occur when assessing nonverbal (aphasic
of the three categories. Early studies suggested that there was or intubated) patients and the inability to specifically test
excellent reliability between different raters who used the brainstem reflexes using the GCS (Fig. 10.2). The score has
GCS to assess the same patient.40,61 However, more recent four categories: eyes, motor, brainstem, and respiration. Each
studies suggest that interrater reliability may be lower than category is given a score from 0 to 4. The four categories and
expected.62-65 There is some evidence that the score is less consistent rating scales for each category are reinforced by the
accurate when used by untrained or less experienced medical score acronym (FOUR), making the scoring system potentially
staff65, 66 emphasizing the need for proper training for this or easier to remember than the GCS, which uses a different score
any scale or classification system that is used. Structured inter- for each category. Because of the structure of the FOUR score,
views are described to enhance the reliability of the GCS locked-in patients (no motor response but can blink on
and Glasgow Outcome Scale (GOS).67 command) and patients in a persistent vegetative state can be
92 Section II—Clinical and Laboratory Assessment

Fig. 10.2  The FOUR score. Instructions for the assessment of the individual categories of the FOUR (Full Outline of UnResponsiveness) score. A, For
eye response (E), grade the best possible response after at least three trials in an attempt to elicit the best level of alertness. A score of E4 indicates
at least three voluntary excursions. If eyelids are closed, the examiner should open them and examine tracking of a finger or object. Tracking with
the opening of one eyelid will suffice in cases of eyelid edema or facial trauma. If tracking is absent horizontally, examine vertical tracking. Alternatively,
two blinks on command should be documented. This will recognize a locked-in syndrome (patient is fully aware). A score of E3 indicates the absence
of voluntary tracking with open eyes. A score of E2 indicates eyelids opening to a loud voice. A score of E1 indicates eyelids open to pain stimulus.
A score of E0 indicates no eyelid opening to pain. B, For motor response (M), grade the best possible response of the arms. A score of M4 indicates
that the patient demonstrated at least one of three hand positions (thumbs-up, fist, or peace sign) with either hand. A score of M3 (localization)
indicates that the patient touched the examiner’s hand after a painful stimulus compressing the temporomandibular joint or supraorbital nerve. A
score of M2 indicates any flexion movement of the upper limbs. A score of M1 indicates extensor response to pain. A score of M0 indicates no motor
response to pain, or myoclonus status epilepticus. C, For brainstem reflexes (B), grade the best possible response. Examine pupillary and corneal
reflexes. Preferably, corneal reflexes are tested by instilling two or three drops sterile saline onto the cornea from a distance of 4 to 6 inches (this
minimizes corneal trauma from repeated examinations). Sterile cotton swabs can also be used. The cough reflex to tracheal suctioning is tested only
when both of these reflexes are absent. A score of B4 indicates pupil and corneal reflexes are present. A score of B3 indicates one pupil wide and
fixed. A score of B2 indicates either pupil or cornea reflexes are absent. A score of B1 indicates both pupil and cornea reflexes are absent. A score
of B0 indicates pupil, cornea, and cough reflex (using tracheal suctioning) are absent. D, For respiration (R), determine spontaneous breathing pattern
in a nonintubated patient and grade simply as regular (R4), or irregular (R2), Cheyne-Stokes (R3) breathing. In mechanically ventilated patients, assess
the pressure waveform of spontaneous respiratory pattern or the patient triggering of the ventilator (R1). The ventilator monitor displaying respiratory
patterns can be used to identify the patient-generated breaths on the ventilator. No adjustments are made to the ventilator while the patient is
graded, but grading is done preferably with PaCO2 within normal limits. A standard apnea (oxygen-diffusion) test may be needed when a patient
breathes at ventilator rate (R0). (From Wijdicks EF, Bamlet WR, Maramattom BV, et al. Validation of a new coma scale: the FOUR score. Ann Neurol 2005;58:
585–93.)
Section II—Clinical and Laboratory Assessment 93

unambiguously identified. The motor category allows “no _training.htm), but with proper training interrater reliability
motor response” or “myoclonic status epilepticus” both to be can be high.88,89 A potential limitation with the NIHSS is that
given a score of 0. This highlights the extremely poor long- it tends to give disproportionate weighting to symptoms
term prognosis of patients with neurologic injury who develop attributable to anterior circulation and cortical dysfunction
myoclonic seizures. In addition, the motor component allows (e.g., focal weakness, aphasia, neglect). Thus patients with
coarse testing of language comprehension and praxis because cerebellar or brainstem infarcts may be markedly debilitated
the best motor score involves the patient making a hand but have low to moderate NIHSS scores. Despite this, the
gesture such as a fist or peace sign. Finally, a combined score NIHSS has proven invaluable to assess the clinical course
of 0 (no eye opening, absent motor responses, no brainstem of hospitalized patients, to guide treatment decisions, and
reflexes, no breathing over the set ventilator rate) is consistent to allow comparison in research studies. A full NIHSS
with brain death. can be viewed at the National Stroke Association website
The FOUR score was initially validated in a prospective (www.stroke.org). Other scales used for ischemic stroke
single center study of 120 patients at the Mayo Clinic.81 These include the Scandinavian Neurological Stroke Scale,90 the
patients were either directly admitted to an NCCU or were Unified Stroke Scale,91 the European Stroke Scale,92 and the
seen by a consultant neurologist in a medical or surgical ICU Canadian Neurological Scale.93
for “unresponsiveness.” Patients had a variety of diagnoses Several scales are used to grade intracerebral hematomas.94
including stroke, TBI, craniotomy for brain tumor, SAH, The original is the most widely validated score and provides
seizure or status epilepticus, and primary CNS infection. This an estimate of 30-day mortality after intracerebral hematoma.
study found that interrater reliability was similar to the GCS. Its relationship to long-term functional outcome is less clear.
Subsequent studies validated this scale for use in both the ED It is the sum of individual points scored as follows: GCS 3 to
and medical intensive care unit setting.82-84 The FOUR score’s 4 (2 points), 5 to 12 (1), 13 to 15 (0); age equal to or more
simplicity and its ability to detect locked-in patients, uncal than 80 years yes (1), no (0); infratentorial origin yes (1), no
herniation and brain death make it an attractive alternative to (0); intracerebral hematoma volume equal or greater than 30
the GCS. Whether it will gain widespread acceptance remains cm3 (1), less than 30 cm3 (0); and intraventricular hemorrhage
to be seen. yes (1), no (0). Patients with a score of 0 should survive; those
with a score of 5 likely will die.95
AVPU and ACDU Score
These two scales were created to provide medical personnel Subarachnoid Hemorrhage
with a simple bedside tool to assess the severity of coma that Several assessment scales have been developed to assess clini-
does not rely on detailed physical examination. In the AVPU cal severity and provide prognostic information in SAH
scale the patient is given a score based on whether he or she patients. Of these, the Hunt and Hess scale and the World
is alert and oriented (A), responding to voice (V), respond- Federation of Neurological Societies (WFNS) scale are two of
ing to pain (P), or unresponsive (U). The similar ACDU the most widely used instruments.
asks whether the patient is alert and oriented (A), confused The Hunt and Hess scale was initially proposed as an assess-
(C), drowsy (D), or unresponsive (U). These scales have a ment tool to help neurosurgeons decide when to operate on
similar accuracy to the GCS, and their simplicity makes patients with aneurysmal SAH.96, 97 Patients receive a score
them attractive candidates for rapid assessment in emer- from I to V, with higher scores signifying increasing SAH
gency settings.85 severity (Table 10.6). Grades IV and V are considered poor-
grade SAH patients. If systemic disease is present the patient’s
score should be increased by one level. The Hunt and Hess
Clinical Scales Used in Stroke scale is simple to administer and widely recognized; however,
Many clinical scales have been developed to evaluate many of the terms used to define grades are vague (e.g.,
patients with ischemic stroke, intraparenchymal hemorrhage,
and subarachnoid hemorrhage. This section outlines some
of the most widely used stroke assessment scales. (The reader
also is referred to: www.strokecenter.org/professionals/stroke-
diagnosis/stroke-assessment-scales-overview, which details Table 10.6  Hunt and Hess Scale Used to
a large number of clinical stroke scales and outcome Classify Severity of Nontraumatic
measures.) Subarachnoid Hemorrhage
Grade* Clinical Description
Ischemic Stroke and I Asymptomatic, mild headache, slight nuchal rigidity
Intraparenchymal Hemorrhage II Moderate to severe headache, nuchal rigidity, no
neurologic deficit other than cranial nerve palsy
The most widely used neurologic score in stroke is the National
Institutes of Health Stroke Scale (NIHSS).86 It is based on III Drowsiness or confusion, mild focal neurologic
deficit
three previously used scales: the Toronto Stroke Scale, the
Oxbury Initial Severity Scale, and the Cincinnati Stroke IV Stupor, moderate to severe hemiparesis
Scale.87 The NIHSS is a 15-item test and has scores that range V Coma, decerebrate posturing
between 0 and 42, with a higher number indicating more
*A modified version adds “grade 0”: unruptured aneurysm, and “grade Ia”:
neurologic impairment. The NIHSS is complex and does no acute meningeal reaction but a fixed neurologic deficit.
require certification (www.ninds.nih.gov/doctors/stroke_scale
94 Section II—Clinical and Laboratory Assessment

archive/scores/iss.html). Other trauma scores include the


Table 10.7  World Federation of Trauma Injury Severity Score (TRISS)104 or the Revised
Neurosurgical Societies Subarachnoid Trauma Score (RTS).105 The RTS is a physiologic scoring
Hemorrhage Grading Scale system, with high interrater reliability and demonstrated
Motor Deficit (Also accuracy to predict death. It consists of GCS, systolic blood
Grade GCS Includes Speech Deficit) pressure (BP), and respiratory rate that is scored when the
patient is admitted (see www.trauma.org).
I 15 −
Several scores have been developed to follow a patient
II 14-13 − while he or she is in the ICU. The Sequential Organ Failure
III 14-13 + Assessment (SOFA)106,107 was initially designed to quantify
IV 12-7 ± the degree of sepsis-related organ dysfunction in critically
ill patients. It has since been used to track the severity of
V 6-3 ±
illness in nonseptic, critically ill patients. The score assigns
From Teasdale GM, Drake CG, Hunt W, et al. A universal subarachnoid 1 to 4 points to six different organ systems (respiratory,
hemorrhage scale: report of a committee of the World Federation of nervous, cardiovascular, liver, coagulation, and renal) based
Neurosurgical Societies. J Neurol Neurosurg Psychiatry 1988;51(11):1457.
GCS, Glasgow Coma Scale.
on the severity of organ dysfunction as measured by labora-
tory values (for respiratory, liver, coagulation, and renal),
vital signs and pressor requirements (for cardiovascular)
and the GCS (for nervous system) (see www.mdcalc.com/
sequential-organ-failure-assessment-sofa-score). High scores
drowsy, stupor) and interrater reliability can therefore be low, indicate more severe illness. The score has been used to
in particular of grade III patients. Despite this, the Hunt and sequentially track the severity of illness in patients during
Hess scale score and association with clinical outcome are well the course of their ICU stay. The SOFA score has also been
described.98,99,100 used to predict mortality, although it was not developed for
The WFNS scale was proposed in 1988 as an alternative to this purpose.108 Its use also has been validated in TBI,109 in
the Hunt and Hess scale to improve interrater reliability by which it may have superior discriminative ability and stron-
avoiding the inherent ambiguity in terms such as drowsy, ger association with outcome compared with other scoring
stupor, and deep coma.101 Like the Hunt and Hess scale, the systems of extracerebral organ dysfunction such as the
WFNS scale is graded from 0 to 5, with higher numbers rep- multiple organ dysfunction score110 or the multiple-system
resenting more severe SAH (Table 10.7). Patients are assigned organ-failure score.111
a score based on their GCS and the presence of focal neuro- The clinical assessment scores described here have not been
logic signs. The WFNS score is correlated with clinical validated for children younger than age 15 and should not be
outcome.98, 99 used in the pediatric population because of the different
normal physiologic parameters between children and adults.
There are, however, several pediatric specific scales that can be
Clinical Scores That Reflect Overall used to predict disease severity. For example, the Pediatric
Index of Mortality (PIM 2)112 assesses disease severity in criti-
Disease Severity cally ill children similar to the APACHE II and SAPS II scores
Several important scales have been developed to calculate used in adults. The PIM 2 uses the following pediatric inten-
overall disease severity, guide treatment decisions, and predict sive care unit (PICU) admission variables to predict outcome:
mortality and outcome in critically ill patients. The APACHE systolic blood pressure (BP), pupillary reaction (fixed or reac-
II score is a well-validated score that is widely used in both tive), ratio of arterial partial pressure of oxygen to fraction of
clinical practice and research literature to predict mortality inspired oxygen (PaO2/FiO2), base excess, elective admission
and outcome in adult patients.55 It is applied to patients (yes/no [Y/N]), mechanical ventilation (Y/N), recovery from
admitted to ICUs within the first 24 hours of admission. The surgery (Y/N), cardiac bypass (Y/N), and high risk/low risk
score is produced using physiologic measurements, laboratory diagnosis. For children with traumatic injury, the Pediatric
values, and the GCS, with higher numbers indicating more Trauma Score (PTS)113 has been used to assess the severity of
severe disease. APACHE II score calculation is used to deter- injury. The PTS includes six variables: weight, systolic BP,
mine eligibility for certain treatments in the ICU. Importantly, mental status, airway maintenance, skeletal injury, and open
the score is by definition an admission score and is not recal- wounds.
culated sequentially. There are later versions in use (APACHE In addition to the clinical assessment scales discussed, a
III, IV) and several online calculators (e.g., www.mdcalc.com/ variety of quantitative rating scales calculate the severity of
apache-ii-score-for-icu-mortality). The SAPS II is an admis- illness in a variety of conditions (e.g., liver failure, pulmonary
sion disease severity score similar to the APACHE II that also embolism, systemic inflammatory response syndrome, tran-
strongly predicts mortality56 and has been found useful in sient ischemic attack) that are beyond the scope of this chapter.
SAH patient assessment.102 An admission score specific to The reader is referred to www.mdcalc.com for a comprehen-
trauma is the Injury Severity Score (ISS).103 The ISS divides sive list of these different assessment instruments. Scores also
the body into six sections: head and neck, face, chest, abdomen, exist to predict development of ICU complications (e.g., pres-
extremity, and external. Each section is assigned an Abbrevi- sure sores)114, 115 (see www.bradenscale.com). Finally there are
ated Injury Score from 1 to 6 (1 being minor and 6 being an scores that help quantify the intensity of care, for example, the
unsurvivable injury). The highest scores in three categories are Therapeutic Intervention Scoring System.116 Of particular rel-
taken and squared and then summed (see www.trauma.org/ evance to the NCCU is the Therapeutic Intensity Level (TIL).117
Section II—Clinical and Laboratory Assessment 95

The TIL was developed to measure the effort required to hemorrhages, brain herniation, cerebral edema, and hydro-
control ICP in different clinical situations. The original score cephalus. Magnetic resonance imaging (MRI) is indicated in
does not include some therapies now in use (e.g., decompres- patients whose coma remains unexplained after CT. MRI
sive craniectomy, hypothermia, or hypertonic saline), and the has a higher sensitivity than CT for acute ischemic stroke,
maximum score originally was assigned for barbiturate use, intracerebral hemorrhage, inflammatory conditions, brain
which now is uncommon. It has, however, been modified for abscesses, brain tumors, cerebral edema, cerebral venous sinus
modern TBI care and is used in current phase III clinical trials thrombosis, and diffuse axonal injury. MRI also provides
(www.em.emory.edu/protect/toolbox.cfm). The TIL provides much better visualization of brainstem structures than CT.
information that provides insight into compliance of the (The role of CT, MRI, and other imaging studies in the NCCU
intracranial compartment; that is, a patient with an ICP of 20 is discussed in Chapters 26 through 29.) MRI is generally less
and a high TIL is far sicker than a patient with the same ICP widely available than CT, is more time consuming, and
and low TIL.118 The Pediatric Intensity Level of Therapy requires nonferromagnetic equipment, making it less feasible
(PILOT)119 aimed to correct the deficiencies of the TIL score. for many critically ill patients. The risks of transporting
It assigns points for hypertonic therapy and it does not assign patients to the CT or MRI scanner, and the time it takes to
maximal points for barbiturate use. complete the studies must be weighed against the benefit of
the information they may yield. The development of portable
CT scanning equipment now allows bedside head CT scans to
Laboratory Investigations to Help be obtained in critically ill patients, and many new ICUs now
incorporate advanced imaging such as MRI and positron
Evaluate Coma emission tomography (PET) scanning in nearby locations.
Metabolic and toxic encephalopathies account for a significant
proportion of altered mental status. Commonly encountered
metabolic disturbances include severe hyper- and hyponatre-
Prognosis of the Comatose Patient
mia, hypercalcemia, elevated blood urea nitrogen (BUN), Coma, by definition, is self-limited. Survivors may recover to
hyperammonemia, hypo- and hyperglycemia, hypercarbia, a persistent vegetative state, to complete neurologic recovery,
hypoxemia, and severe hyper- and hypothyroidism. A toxicol- or to an intermediate state of neurologic disability. The
ogy screen detects exposure to common drugs and toxins Glasgow Outcome Scale (GOS) is an established metric of
(Table 10.8). Plasma osmolality should be measured so that recovery120 (Table 10.9). Although it fails to capture the full
the plasma osmolal gap can be determined. The osmolal gap breadth of possible neurologic outcomes, it is widely used by
is the difference between the calculated osmolarity (2[Na] + clinical investigators for traumatic and nontraumatic coma.
[BUN]/2.8 + [glucose]/18) and the measured osmolality. The How to determine the prognosis of comatose patients poses
osmolal gap is elevated with intoxication from alcohols, such a significant clinical and sometime ethical challenge. Studies
as methanol, and ethylene glycol. A complete blood count that have addressed the prognosis of coma are plagued by
(white blood cell count) and differential should be obtained numerous problems that limit their clinical applicability. First,
to help assess for an infectious cause of encephalopathy. coma has been defined inconsistently across studies. Second,
virtually all studies involved patients in whom care was inten-
tionally limited, leading to a “self-fulfilling prophecy” bias.
Neuroimaging Studies and Third, most studies were performed before major advances in
Coma Evaluation
Noncontrast cranial compued tomography (CT) is indicated
in all new cases of unexplained coma and is the test of first
choice. CT rapidly identifies intra- and extra-axial cerebral Table 10.9  The Glasgow Outcome Scale
1 Death
2 Persistent Patient exhibits no obvious cortical
vegetative state function
Table 10.8  Agents Commonly Tested for on 3 Severe disability (Conscious but disabled.) Patient
Routine Toxicology Screens depends on others for daily
support due to mental or physical
Cocaine disability or both.
Amphetamines 4 Moderate (Disabled but independent.) Patient
Benzodiazepines disability is independent as far as daily life is
Barbiturates concerned. The disabilities found
Ethanol include varying degrees of
Tetrahydrocannabinol (active ingredient in marijuana) dysphagia, hemparesis, or ataxia,
Phencyclidine (i.e., PCP) as well as intellectual and memory
Opiates* deficits and personality changes.
Acetaminophen
Salicylates 5 Good recovery Resumption of normal activities even
though there may be minor
*Routine opiate panels generally only test for morphine and its derivatives neurologic or psychological deficits
(morphine, codeine, heroin). Testing for other synthetic opioids (e.g.,
hydrocodone, hydromorphone, methadone, fentanyl, oxycodone, Adapted from Jennett B, Bond M. Assessment of outcome after severe brain
buprenorphine) requires extended opioid testing. damage. Lancet 1975;1:480–4.
96 Section II—Clinical and Laboratory Assessment

intensive care (e.g., induced hypothermia) that have been The Vegetative and Minimally
shown to improve outcome in certain patient populations.
Last, studies have not yet systematically addressed the impact
Conscious State
of clinical confounders such as metabolic disturbances, seda- Coma survivors who fail to recover consciousness tradition-
tive and paralytic mediations, and shock. Recent efforts by the ally have been diagnosed as being in “vegetative” or “mini-
IMPACT investigators and others have attempted to address mally conscious” states. Patients with vegetative states regain
prognosis after TBI,43-46,121-123 and tools to help calculate prog- some arousal, but without apparent true awareness. If there is
nosis are available online (e.g., www.tbi-impact.org). no further improvement after 4 weeks, the patient is stated to
The prognosis of coma is based in part on its etiology. For be in a persistent vegetative state.144-146 Vegetative patients
example, coma after a high-velocity penetrating head injury may demonstrate sleep-wake cycles, may open their eyes and
usually has a worse prognosis than a closed TBI, whereas a track objects, and may be alert and swallow with feeding.
patient in coma with an extradural hematoma is far more Patients in minimally conscious states have greater evidence
likely to do well than one with a subdural hematoma. In of awareness and interaction including verbal or gestural
general nontraumatic coma has a worse outcome than trau- responses, reaching for objects, and emotional responses.144
matic coma, and among patients with nontraumatic coma, However, the anatomic and physiologic differences between
nonstructural etiologies are associated with a better prognosis vegetative and minimally conscious states are poorly under-
than structural etiologies. However, to “predict” outcome stood and remain difficult to define clinically. This results in
requires a careful consideration also of clinical signs and ancil- both ethical and legal challenges to managing patients in
lary tests including electrophysiologic, neuroimaging, and these conditions.
biochemical studies. Specialized rating scales that assess cognitive function in
Levy and colleagues124 performed a landmark prospective minimally conscious and vegetative patients have been devel-
study of outcome in 500 patients with nontraumatic coma, oped to help aid outcome prediction and monitor the effec-
primarily due to cardiac arrest, and found that prognosis tiveness of treatments in long-term rehabilitation centers.
depended in large part on the duration of coma, neuro- The JFK Coma Recovery Scale (CRS) was initially described
ophthalmologic signs, and motor function. The Levy criteria in 1991147 and is comprised of hierarchically arranged
have since become the benchmark to determine prognosis after items associated with brainstem and subcortical processes.
severe, nontraumatic brain injury, although newer advances The initial CRS was revised148 to aid in the differentiation of
in critical care, especially therapeutic hypothermia,125,126 have minimally conscious and vegetative patients. The CRS-R com-
altered the accuracy of these prognostic criteria. Multiple prises six subscales that address auditory, visual, motor, oro-
studies have examined the utility of additional clinical signs motor, communication, and arousal functions. Patients are
and ancillary tests for prognosis in cardiac arrest survivors. scored based on the presence or absence of different behav-
These data are summarized in a practice parameter issued by ioral responses to a variety of stimuli that range from cogni-
the Quality Standards Subcommittee of the American Academy tively complex behaviors (e.g., object recognition, functional
of Neurology.127 Clinical signs that most accurately portend object use, intelligible verbalization) to reflexive behaviors
poor prognosis in these patients include (1) myoclonic status (e.g., startle responses, posturing, oral reflexive movements)
epilepticus within the first 24 hours after primary circulatory to no response. Validity analyses support use of the scale as an
arrest, (2) absence of pupillary responses within days 1 to 3 index of neurobehavioral function and have shown that the
after cardiopulmonary resuscitation (CPR), (3) absent corneal CRS-R is capable of discriminating patients in minimally con-
reflexes within days 1 to 3 after CPR, and (4) absent or extensor scious state from those in vegetative state, which is of critical
motor responses after day 3. Although certain electroencepha- importance to establish prognosis and formulate treatment
lographic patterns, such as burst suppression and generalized intervention.149
epileptiform discharges are associated with poor prognosis, Although clinical assessment of the level of consciousness
EEG lacks sufficient prognostic accuracy and is not recom- has traditionally relied on behavioral responses, functional
mended as a tool to predict outcome.128, 129 Somatosenory imaging methods have begun to be used to directly interrogate
evoked potentials (SSEPs) appear to be more useful for brain function.150 In general, cerebral blood flow and metabo-
prognostication130-133 because SSEPs are less susceptible than lism reflects the level of arousal, with locked-in or minimally
EEG to the confounding effects of drugs and metabolic conscious patients showing relatively normal function, vegeta-
derangement. Bilateral absence of the N20 component of the tive patients showing reduced function, and brain dead
SSEP (see Chapter 24) with median nerve stimulation after patients showing no function, though certain networks of
day 1 is associated with poor prognosis. Numerous serum and brain regions are clearly more closely tied to the level of con-
CSF biomarkers have been studied for their prognostic value. sciousness than others. Mental imagery tasks are known to
To date, the only marker with sufficient prognostic accuracy produce regional brain activation approximating actual task
to be recommended is neuron-specific enolase (NSE). Serum performance, and a functional MRI study carried out in a
NSE levels of greater than 33 mcg/L on days 1 through 3 after patient thought to be in a persistent vegetative state 5 months
cardiac arrest predict poor outcome127, 134; however, this test is after severe TBI demonstrated task-specific activation patterns
not yet widely available and there is evidence that the false- during motor imagery similar to those observed in control
positive rate may be unacceptably high in patients treated with subjects, indicating that the patient was able to understand the
therapeutic hypothermia.135 Ongoing studies address the prog- task instruction and modulate brain activity in response to
nostic role of cranial CT, MRI, and magnetic resonance spec- exogenous cues despite the absence of convincing behavioral
troscopy after coma both from traumatic and nontraumatic responses.151 A follow-up study152 observed that 5 out of 54
causes.136-143 These are discussed further in Chapters 26 vegetative or minimally conscious patients were able to will-
and 28. fully modulate brain activity and produce activation patterns
Section II—Clinical and Laboratory Assessment 97

similar to those seen in healthy control subjects on functional 11. Bassant MH, Ennouri K, Lamour Y. Effects of iontophoretically applied
monoamines on somatosensory cortical neurons of unanesthetized rats.
MRI (fMRI), and in one case two types of mental imagery
Neuroscience 1990;39:431–9.
were used to verify “yes” or “no” answers to several questions, 12. Kolta A, Reader TA. Modulatory effects of catecholamines on neurons of the
demonstrating the feasibility of using brain responses to rat visual cortex: single cell iontophoretic studies. Can J Physiol Pharmacol
imagery as a means of communication. Although fMRI is not 1989;67:615–23.
a practical means for communication, other modalities such 13. McCormick DA. Cholinergic and noradrenergic modulation of
thalamocortical processing. Trends Neurosci 1989;12(6):215–21.
as transcranial optical spectroscopy and EEG could be used to 14. McCormick DA, Bal T. Sleep and arousal: thalamocortical mechanisms.
transduce brain responses at the bedside or to drive brain Annu Rev Neurosci 1997;20:185–215.
computer interfaces in patients who are otherwise locked-in. 15. Sato H, Fox K, Daw NW. Effect of electrical stimulation of locus coeruleus
Another fMRI approach to assess brain function is to on the activity of neurons in the cat visual cortex. J Neurophysiol 1989;62:
946–58.
examine the integrity of correlated activity within distributed
16. Bromberg-Martin ES, Matsumoto M, Hikosaka O. Dopamine in
networks that occurs independently of any voluntary task. motivational control: rewarding, aversive, and alerting. Neuron 2010;68:
Activity in one such network, termed the “default mode 815–34.
network,”153 is closely tied to a subject’s level of attentiveness 17. Schultz W. Predictive reward signal of dopamine neurons. J Neurophysiol
and shows graded alterations in patients with disorders of 1998;80(1):1–27.
18. Sawa A, Snyder SH. Schizophrenia: diverse approaches to a complex disease.
consciousness, with locked-in patients showing normal con- Science 2002;296(5568):692–5.
nectivity, and minimally conscious and unconscious patients 19. Marié RM, Defer GL. Working memory and dopamine: clinical and
showing reduced connectivity.154 These and related studies experimental clues. Curr Opin Neurol 2003;S29–35.
emerging in the literature suggest that functional brain 20. Haenisch B, Bönisch H. Depression and antidepressants: insights from
knockout of dopamine, serotonin, or noradrenaline re-uptake transporters.
imaging may ultimately supplement clinical examination in
Pharmacol Ther 2011;129(3):352–68.
assessing the integrity of conscious awareness, though signifi- 21. Ravindran LN, Stein MB. The pharmacologic treatment of anxiety disorders:
cant technical, practical, and ethical challenges remain in the a review of progress. J Clin Psychiatry 2010;71(7):839–54.
development and validation of their use. 22. Adamantidis AR, Zhang F, Aravanis AM, et al. Neural substrates of
awakening probed with optogenetic control of hypocretin neurons. Nature
2007;450(7168):720–4.
Conclusion 23. Willie JT, Chemelli RM, Sinton CM, et al. To eat or to sleep? Orexin in the
regulation of feeding and wakefulness. Annu Rev Neurosci 2001;24:429–58.
Altered mental status and coma occur commonly in the ICU. 24. Schjolberg A, Sunnerhagen KS. Unlocking the locked-in: a need for team
Physicians should develop an efficient diagnostic approach approach for survivors of locked-in syndrome. Acta Neurol Scand 2011.
Epub ahead of print.
that facilitates rapid treatment to minimize additional brain
25. Gelenberg AJ. The catatonic syndrome. Lancet 1976;1:1339–41.
injury. The clinical exam, laboratory tests, and neuroimaging 26. Cairns H. Head injuries in motor-cyclists: the importance of the crash
studies are powerful tools that help to determine whether a helmet. Br Med J 1941;2(4213):465–71.
structural or systemic abnormality exists, thus narrowing the 27. Kumral E, Bayulkem G, Evyapan D, et al. Spectrum of anterior cerebral
differential diagnosis. Standardized clinical scales allow the artery territory infarction: clinical and MRI findings. Eur J Neurol 2002;9(6):
615–24.
measurement of disease severity, help to track clinical prog- 28. Fisher CM. Ocular bobbing. Arch Neurol 1964;11:543–6.
ress, aid in communication between caregivers, and can give 29. Rosenberg ML. Spontaneous vertical eye movements in coma. Ann Neurol
valuable prognostic information. Currently there are few 1986;20(5):635–7.
widely available means of reliably determining prognosis in 30. Brandt TH, Dieterich M. Different types of skew deviation. J Neurol
Neurosurg Psychiatry 1991;54(6):549–50.
comatose patients. Genetic and metabolic markers and
31. Keane JR. Ocular skew deviation: analysis of 100 cases. Arch Neurol 1975;
advanced neuroimaging and neurophysiologic methods may 32(3):185–90.
ultimately prove helpful. 32. Schirmer CM, Hedges TR. Mechanisms of visual loss in papilledema.
Neurosurg Focus 2007;23(5):E5.
33. Simon RP. Neurogenic pulmonary edema. Neurol Clin 1993;11(2):309–23.
References 34. Tarulli AW, Lim C, Bui JD, et al. Central neurogenic hyperventilation: a case
report and discussion of pathophysiology. Arch Neurol 2005;62(10):
1. Von Economo C. Sleep as a problem of localization. J Nerv Ment Dis 1930; 1632–4.
71:249–59. 35. Ramsay MAE, Savege TM, Simpson BRJ, et al. Controlled sedation with
2. Morruzi G, Magoun HW. Brain stem reticular formation and activation of alphaxalone-alphadolone. BMJ 1974;2:656–9.
the EEG. J Neuropsychiatry Clin Neurosci 1949;7:251–67. 36. Sessler CN, Gosnell MS, Grap MJ, et al. The Richmond agitation-sedation
3. Parvizi J, Damasio AR. Neuroanatomical correlates of brainstem coma. Brain scale: validity and reliability in adult intensive care unit patients. Am J Respir
2003;126:1524–36. Crit Care Med 2002;166:1338–44.
4. Posner JB, Saper CB, Schiff ND, et al. Plum and Posner’s diagnosis of stupor 37. De Lemos J, Tweeddale M, Chittock D. Measuring quality of sedation in
and coma. 4th ed. New York: Oxford University Press; 2007. adult mechanically ventilated critically ill patients: the Vancouver interaction
5. Blumenfeld H. Neuroanatomy through clinical cases. Sunderland, MA: and calmness scale. J Clin Epidemiol 2000;53:908–19.
Sinauer Associates; 2010. 38. Fraser GL, Riker R. Monitoring sedation, agitation, analgesia, and delirium
6. Hallanger AE, Levey AI, Lee HJ, et al. The origins of cholinergic and other in critically ill adult patients. Crit Care Clin 2001;17:1–21.
subcortical afferents to the thalamus in the rat. J Comp Neurol 1987; 262: 39. De Jong MMJ, Burns SB, Campbell ML, et al. Development of the American
105–24. Association of Critical-Care Nurses’ Sedation Assessment Scale for Critically
7. Saper CB, Scammell TE, Lu J. Hypothalamic regulation of sleep and Ill Patients Am J Crit Care 2005;14:531–44.
circadian rhythms. Nature 2005;437(7063):1257–63. 40. Teasdale G, Jennett B. Assessment of coma and impaired consciousness:
8. Saper CB, Fuller PM, Pedersen NP, et al. Sleep state switching. Neuron 2010; a practical scale. Lancet 1974;2:81–4.
68(6):1023–42. 41. Teasdale G, Jennett B. Assessment and prognosis of coma after head injury.
9. Saper CB. Organization of cerebral cortical afferent systems in the rat. II. Acta Neurochir 1976;34:45–55.
Magnocellular basal nucleus. J Comp Neurol 1984;222:313–42. 42. Gabbe BJ, Cameron PA, Finch CF. The status of the Glasgow coma scale.
10. Saper CB. Diffuse cortical projection systems: anatomical organization and Emerg Med (Fremantle) 2003;15(4):353–60. Review.
role in cortical function. In: Plum F, editor. Handbook of physiology: the 43. Marmarou A, Lu J, Butcher I, et al. Prognostic value of the Glasgow coma
nervous system. V. Bethesda, MD: American Physiological Society; 1987. scale and pupil reactivity in traumatic brain injury assessed pre-hospital and
pp. 169–210. on enrollment: an IMPACT analysis. J Neurotrauma 2007;24(2):270–80.
98 Section II—Clinical and Laboratory Assessment

44. Murray GD, Butcher I, McHugh GS, et al. Multivariable prognostic analysis 48. Tuhrim S, Dambrosia JM, Price TR, et al. Prediction of intracerebral
in traumatic brain injury: results from the IMPACT study. J Neurotrauma hemorrhage survival. Ann Neurol 1988;24(2):258–63.
2007;24(2):329–37. 49. Tuhrim S, Dambrosia JM, Price TR, et al. Intracerebral hemorrhage: external
45. Steyerberg EW, Mushkudiani N, Perel P, et al. Predicting outcome after validation and extension of a model for prediction of 30-day survival. Ann
traumatic brain injury: development and international validation of Neurol 1991;29(6):658-63.
prognostic scores based on admission characteristics. PLoS Med 2008;5(8): 50. Hirai S, Ono J, Yamaura A. Evaluation of consciousness by the Glasgow
e165. Coma Scale in aneurysmal subarachnoid hemorrhage: appraisal using
46. Lingsma HF, Roozenbeek B, Steyerberg EW, et al. Early prognosis in logistic regression analysis. Cerebrovasc Dis 1997;99:S42–43.
traumatic brain injury: from prophecies to predictions. Lancet Neurol
2010;9(5):543–54. A complete list of references for this chapter can be found online at
47. Bates D. The prognosis of medical coma. J Neurol Neurosurg Psychiatry www.expertconsult.com.
2001;71(Suppl):i20–23.
Section II—Clinical and Laboratory Assessment 98.e1

References 34. Tarulli AW, Lim C, Bui JD, et al. Central neurogenic hyperventilation: a case
report and discussion of pathophysiology. Arch Neurol 2005;62(10):1632–4.
1. Von Economo C. Sleep as a problem of localization. J Nerv Ment Dis 35. Ramsay MAE, Savege TM, Simpson BRJ, et al. Controlled sedation with
1930;71:249–59. alphaxalone-alphadolone. BMJ 1974;2:656–9.
2. Morruzi G, Magoun HW. Brain stem reticular formation and activation of 36. Sessler CN, Gosnell MS, Grap MJ, et al. The Richmond agitation-sedation
the EEG. J Neuropsychiatry Clin Neurosci 1949;7:251–67. scale: validity and reliability in adult intensive care unit patients. Am J
3. Parvizi J, Damasio AR. Neuroanatomical correlates of brainstem coma. Respir Crit Care Med 2002;166:1338–44.
Brain 2003;126:1524–36. 37. De Lemos J, Tweeddale M, Chittock D. Measuring quality of sedation in
4. Posner JB, Saper CB, Schiff ND, et al. Plum and Posner’s diagnosis of adult mechanically ventilated critically ill patients: the Vancouver
stupor and coma. 4th ed. New York: Oxford University Press; 2007. interaction and calmness scale. J Clin Epidemiol 2000;53:908–19.
5. Blumenfeld H. Neuroanatomy through clinical cases. Sunderland, MA: 38. Fraser GL, Riker R. Monitoring sedation, agitation, analgesia, and delirium
Sinauer Associates; 2010. in critically ill adult patients. Crit Care Clin 2001;17:1–21.
6. Hallanger AE, Levey AI, Lee HJ, et al. The origins of cholinergic and other 39. De Jong MMJ, Burns SB, Campbell ML, et al. Development of the
subcortical afferents to the thalamus in the rat. J Comp Neurol 1987; American Association of Critical-Care Nurses’ Sedation Assessment Scale
262:105–24. for Critically Ill Patients. Am J Crit Care 2005;14:531–44.
7. Saper CB, Scammell TE, Lu J. Hypothalamic regulation of sleep and 40. Teasdale G, Jennett B. Assessment of coma and impaired consciousness:
circadian rhythms. Nature 2005;437(7063):1257–63. a practical scale. Lancet 1974;2:81–4.
8. Saper CB, Fuller PM, Pedersen NP, et al. Sleep state switching. Neuron 41. Teasdale G, Jennett B. Assessment and prognosis of coma after head injury.
2010;68(6):1023–42. Acta Neurochir 1976;34:45–55.
9. Saper CB. Organization of cerebral cortical afferent systems in the rat. II. 42. Gabbe BJ, Cameron PA, Finch CF. The status of the Glasgow Coma Scale.
Magnocellular basal nucleus. J Comp Neurol 1984;222:313–42. Emerg Med (Fremantle) 2003;15(4):353–60. Review.
10. Saper CB. Diffuse cortical projection systems: anatomical organization and 43. Marmarou A, Lu J, Butcher I, et al. Prognostic value of the Glasgow Coma
role in cortical function. In: Plum F, editor. Handbook of physiology: the Scale and pupil reactivity in traumatic brain injury assessed pre-hospital
nervous system. V. Bethesda, MD: American Physiological Society; 1987. and on enrollment: an IMPACT analysis. J Neurotrauma 2007;24(2):270–80.
pp. 169–210. 44. Murray GD, Butcher I, McHugh GS, et al. Multivariable prognostic analysis
11. Bassant MH, Ennouri K, Lamour Y. Effects of iontophoretically applied in traumatic brain injury: results from the IMPACT study. J Neurotrauma
monoamines on somatosensory cortical neurons of unanesthetized rats. 2007;24(2):329–37.
Neuroscience 1990;39:431–9. 45. Steyerberg EW, Mushkudiani N, Perel P, et al. Predicting outcome after
12. Kolta A, Reader TA. Modulatory effects of catecholamines on neurons of traumatic brain injury: development and international validation of
the rat visual cortex: single cell iontophoretic studies. Can J Physiol prognostic scores based on admission characteristics. PLoS Med 2008;5(8):
Pharmacol 1989;67:615–23. e165.
13. McCormick DA. Cholinergic and noradrenergic modulation of 46. Lingsma HF, Roozenbeek B, Steyerberg EW, et al. Early prognosis in
thalamocortical processing. Trends Neurosci 1989;12(6):215–21. traumatic brain injury: from prophecies to predictions. Lancet Neurol
14. McCormick DA, Bal T. Sleep and arousal: thalamocortical mechanisms. 2010;9(5):543–54.
Annu Rev Neurosci 1997;20:185–215. 47. Bates D. The prognosis of medical coma. J Neurol Neurosurg Psychiatry
15. Sato H, Fox K, Daw NW. Effect of electrical stimulation of locus coeruleus 2001;71(Suppl):i20–23.
on the activity of neurons in the cat visual cortex. J Neurophysiol 1989;62: 48. Tuhrim S, Dambrosia JM, Price TR, et al. Prediction of intracerebral
946–58. hemorrhage survival. Ann Neurol 1988;24(2):258–63.
16. Bromberg-Martin ES, Matsumoto M, Hikosaka O. Dopamine in 49. Tuhrim S, Dambrosia JM, Price TR, et al. Intracerebral hemorrhage:
motivational control: rewarding, aversive, and alerting. Neuron 2010;68: external validation and extension of a model for prediction of 30-day
815–34. survival. Ann Neurol 1991;29(6):658-63.
17. Schultz W. Predictive reward signal of dopamine neurons. J Neurophysiol 50. Hirai S, Ono J, Yamaura A. Evaluation of consciousness by the Glasgow
1998;80(1):1–27. Coma Scale in aneurysmal subarachnoid hemorrhage: appraisal using
18. Sawa A, Snyder SH. Schizophrenia: diverse approaches to a complex disease. logistic regression analysis. Cerebrovasc Dis 1997;99:S42–43.
Science 2002;296(5568):692–5. 51. Benesch CG, McDaniel KD, Cox C, et al. End-stage Alzheimer’s disease:
19. Marié RM, Defer GL. Working memory and dopamine: clinical and Glasgow Coma Scale and the neurologic examination. Arch Neurol 1993;
experimental clues. Curr Opin Neurol 2003;S29–35. 50:1309–15.
20. Haenisch B, Bönisch H. Depression and antidepressants: insights from 52. Chou CH, Lin GM, Ku CH, et al. Comparison of the APACHE II, GCS and
knockout of dopamine, serotonin, or noradrenaline re-uptake transporters. MRC scores in predicting outcomes in patients with tuberculous
Pharmacol Ther 2011;129(3):352–68. meningitis. Int J Tuberc Lung Dis 2010;14:86–92.
21. Ravindran LN, Stein MB. The pharmacologic treatment of anxiety 53. Ishihara M, Kamei S, Taira N, et al. Hospital-based study of the prognostic
disorders: a review of progress. J Clin Psychiatry 2010;71(7):839–54. factors in adult patients with acute community-acquired bacterial
22. Adamantidis AR, Zhang F, Aravanis AM, et al. Neural substrates of meningitis in Tokyo. Japan. Intern Med 2009;48(5):295–300.
awakening probed with optogenetic control of hypocretin neurons. Nature 54. Schut ES, Lucas MJ, Brouwer MC, et al. Cerebral infarction in adults with
2007;450(7168):720–4. bacterial meningitis. Neurocrit Care 2011;Oct. Epub ahead of print.
23. Willie JT, Chemelli RM, Sinton CM, et al. To eat or to sleep? Orexin in the 55. Knaus WA, Draper EA, Wagner DP, et al. APACHE II: a severity of disease
regulation of feeding and wakefulness. Annu Rev Neurosci 2001;24:429–58. classification system. Crit Care Med 1985;13:818–29.
24. Schjolberg A, Sunnerhagen KS. Unlocking the locked-in: a need for team 56. Le Gall JR, Lemeshow S, Saulnier F. A new simplified acute physiology score
approach for survivors of locked-in syndrome. Acta Neurol Scand 2011. (SAPS II) based on a European/North American multicenter study. JAMA
Epub ahead of print. 1993;270:2957–63.
25. Gelenberg AJ. The catatonic syndrome. Lancet 1976;1:1339–41. 57. Niskanen M, Kari A, Nikki P, et al. Acute physiology and chronic health
26. Cairns H. Head injuries in motor-cyclists: the importance of the crash evaluation (APACHE II) and Glasgow Coma Scores as predictors of
helmet. Br Med J 1941;2(4213):465–71. outcome from intensive care after cardiac arrest. Crit Care Med 1991;
27. Kumral E, Bayulkem G, Evyapan D, et al. Spectrum of anterior cerebral 19(12):1465–73.
artery territory infarction: clinical and MRI findings. Eur J Neurol 58. Bastos PG, Sun X, Wagner DP, et al. Glasgow Coma Scale score in the
2002;9(6):615–24. evaluation of outcome in the intensive care unit: findings from the Acute
28. Fisher CM. Ocular bobbing. Arch Neurol 1964;11:543–6. Physiology and Chronic Health Evaluation III study. Crit Care Med
29. Rosenberg ML. Spontaneous vertical eye movements in coma. Ann Neurol 1993;21(10):1459–65.
1986;20(5):635–7. 59. Buechler CM, Blostein PA, Koestner A, et al. Variation among trauma
30. Brandt TH, Dieterich M. Different types of skew deviation. J Neurol centers’ calculation of Glasgow Coma Scale score: results of a national
Neurosurg Psychiatry 1991;54(6):549–50. survey. J Trauma 1998;45:429–32.
31. Keane JR. Ocular skew deviation. Analysis of 100 cases. Arch Neurol 60. Healey C, Osler TM, Rogers FB, et al. Improving the Glasgow Coma Scale
1975;32(3):185–90. score: motor score alone is a better predictor. J Trauma 2003;54(4):671–8.
32. Schirmer CM, Hedges TR. Mechanisms of visual loss in papilledema. 61. Teasdale G, Knill-Jones R, van der Sande J. Observer variability in assessing
Neurosurg Focus 2007;23(5):E5. impaired consciousness and coma. J Neurol Neurosurg Psychiatry
33. Simon RP. Neurogenic pulmonary edema. Neurol Clin 1993;11(2):309–23. 1978;41:603–10.
98.e2 Section II—Clinical and Laboratory Assessment

62. Gill MR, Reiley DG, Green SM. Interrater reliability of Glasgow Coma Scale 93. Cote R, Battista RN, Wolfson C, et al. The Canadian Neurological Scale:
scores in the emergency department. Ann Emerg Med 2004;43:215–23. validation and reliability assessment. Neurology 1989;39:638–43.
63. Gill M, Martens K, Lynch EL, et al. Interrater reliability of 3 simplified 94. Hwang BY, Appelboom G, Kellner CP, et al. Clinical grading scales in
neurologic scales applied to adults presenting to the emergency department intracerebral hemorrhage. Neurocrit Care 2010;13(1):141–51.
with altered levels of consciousness. Ann Emerg Med 2007;49(4):403–7. 95. Hemphill JC 3rd, Bonovich DC, Besmertis L, et al. The ICH score: a simple,
64. Holdgate A, Ching N, Angonese L. Variability in agreement between reliable grading scale for intracerebral hemorrhage. Stroke 2001;32(4):
physicians and nurses when measuring the Glasgow Coma Scale in the 891–7.
emergency department limits its clinical usefulness. Emerg Med Australas 96. Hunt WE, Hess RM. Surgical risk as related to time of intervention in the
2006;18(4):379–84. repair of intracranial aneurysms. J Neurosurg 1968;28:14–20.
65. Rowley G, Fielding K. Reliability and accuracy of the Glasgow Coma Scale 97. Hunt WE, Kosnik EJ. Timing and perioperative care in intracranial
with experienced and inexperienced users. Lancet 1991;337:535–8. aneurysm surgery. Clin Neurosurg 1974;21:9–89.
66. Heim C, Schoettker P, Gilliard N, et al. Knowledge of Glasgow Coma Scale 98. Chiang VL, Claus EB, Awad IA. Toward a more rational prediction of
by air-rescue physicians. Scand J Trauma Resusc Emerg Med 2009; 17:39. outcome in patients with high-grade subarachnoid hemorrhage.
67. Wilson JT, Pettigrew LE, Teasdale GM. Structured interviews for the Neurosurgery 2000;46:28–35.
Glasgow Outcome Scale and the extended Glasgow Outcome Scale: 99. Oshiro EM, Walter KA, Piantadosi S, et al. A new subarachnoid hemorrhage
guidelines for their use. J Neurotrauma 1998;15:573–85. grading system based on the Glasgow Coma Scale: a comparison with the
68. Raimondi AJ, Hirschauer J. Head injury in the infant and toddler: coma Hunt and Hess and World Federation of Neurological Surgeons scales in a
coring and outcome scale. Childs Brain 1984;12:1018–20. clinical series. Neurosurgery 1997;41(1):140–7.
69. Simpson DA, Cockington RA, Hanieh A, et al. Head injuries in infants and 100. Salary M, Quigley MR, Wilberger JE Jr. Relation among aneurysm size,
young children: the value of the paediatric Coma Scale. Review of literature amount of subarachnoid blood and clinical outcome. J Neurosurg 2007;
and report on a study. Childs Nerv Syst 1991;7:183–90. 107(1):13–7.
70. Kirkham FJ, Newton CR, Whitehouse W. Pediatric Coma Scales. Dev Med 101. Teasdale GM, Drake CG, Hunt W, et al. A universal subarachnoid
Child Neurol. 2008;50(4):267–74. hemorrhage scale: report of a committee of the World Federation of
71. James HE, Anas NG, Perkin RM, et al. The Glasgow Coma Scale. In: James Neurosurgical Societies. J Neurol Neurosurg Psychiatry 1988;51(11):1457.
HE, Anas NG, Perkin RM, editors. Brain insults in infants and children. 102. Claassen J, Vu A, Kreiter KT, et al. Effect of acute physiologic derangements
Orlando, FL: Grune and Stratton; 1985. pp. 179–82. on outcome after subarachnoid hemorrhage. Crit Care Med 2004;32(3):
72. Morray JP, Tyler DC, Jones TK, et al. Coma Scale for use in brain-injured 832–8.
children. Crit Care Med 1984;12:1018–20. 103. Baker SP, O’Neill B, Haddon W Jr, et al. The Injury Severity Score: a
73. Starmark JE, Stålhammar D, Holmgren E. The Reaction Level Scale method for describing patients with multiple injuries and evaluating
(RLS85): manual and guidelines. Acta Neurochir 1988a;91:12–20. emergency care. J Trauma 1974;14(3):187–96.
74. Johnstone AJ, Lohlun JC, Miller JD, et al. A comparison of the Glasgow 104. Boyd CR, Tolson MA, Copes WS: Evaluating trauma care: the TRISS
Coma Scale and the Swedish Reaction Level Scale. Brain Inj 1993;7:501–6. method. J Trauma 1987;27:370–8.
75. Stålhammar D, Starmark JE, Holmgren E, et al. Assessment of 105. Champion HR, Sacco WH, Copes WS, et al. A revision of the trauma score.
responsiveness in acute cerebral disorders: a multicenter study on the J Trauma 1989;29:623–9.
reaction level scale (RLS 85). Acta Neurochir 1988;90:73–80. 106. Vincent JL, Moreno R, Takala J, et al. The SOFA (Sepsis-Related Organ
76. Tesseris J, Pantazidis N, Routsi C, et al. A comparative study of the Reaction Failure assessment) score to describe organ dysfunction/failure. Working
Level Scale (RLS85) with Glasgow Coma Scale (GCS) and Edinburgh-2 Group on sepsis-related problems of the European Society of Intensive
Coma Scale (modified) (E2CS(M)). Acta Neurochir 1991;10:65–76. Care Medicine. Intensive Care Med 1996;22(7):707–10.
77. Walther SM, Jonasson U, Gill H. Comparison of the Glasgow Coma Scale 107. Vincent JL, de Mendonca A, Cantraine F, et al. Use of the SOFA score to
and the reaction level scale for assessment of cerebral responsiveness in the assess the incidence of organ dysfunction/failure in intensive care units:
critically ill. Intensive Care Med 2003;29:933–8. results of a multicenter, prospective study. Working group on sepsis-related
78. Starmark JE, Stålhammar D, Holmgren E, et al. A comparison of the problems of the European Society of Intensive Care Medicine. Crit Care
Glasgow Coma Scale and the Reaction Level Scale (RLS85). J Neurosurg Med 1998;26(11):1793–800.
1988;69(5);699–706. 108. Vincent J, Ferreira F, Moreno R. Scoring systems for assessing organ
79. Benzer A, Mitterschiffthaler G, Marosi M, et al. Prediction of non-survival dysfunction and survival. Crit Care Clin 2000;16:353–66.
after trauma: Innsbruck Coma Scale. Lancet 1991;338(8773):977–8. 109. Zygun D, Berthiaume L, Laupland K, et al. SOFA is superior to MOD score
80. Diringer MN, Edwards DF. Does modification of the Innsbruck and the for the determination of non-neurologic organ dysfunction in patients with
Glasgow Coma Scales improve their ability to predict functional outcome? severe traumatic brain injury: a cohort study. Crit Care 2006;10(4):R115.
Arch Neurol 1997;54(5):606–11. 110. Marshall JC, Cook DJ, Christou NV, et al. Multiple Organ Dysfunction
81. Wijdicks EF, Bamlet WR, Maramattom BV, et al. Validation of a new Coma Score: a reliable descriptor of a complex clinical outcome. Crit Care Med
Scale: the FOUR score. Ann Neurol 2005;58:585–93. 1995;23(10):1638–52. Review.
82. Iyer VN, Mandrekar JN, Danielson RD, et al. Validity of the FOUR score 111. Ciesla DJ, Moore EE, Johnson JL, et al. The role of the lung in postinjury
Coma Scale in the medical intensive care unit. Mayo Clin Proc 2009;84: multiple organ failure. Surgery 2005;138(4):749–57, discussion 757–8.
694–701. 112. Slater A, Shann F, Pearson G. Paediatric Index of Mortality (PIM) study
83. Stead LG, Wijdicks EF, Bhagra A, et al. Validation of a new Coma Scale, the group: PIM2: a revised version of the Paediatric Index of Mortality.
FOUR score, in the emergency department. Neurocrit Care 2009;10:50–4. Intensive Care Med 2003;29:278–85.
84. Wolf CA, Wijdicks EF, Bamlet WR, et al. Further validation of the FOUR 113. Tepas JJ,3rd, Ramenofsky ML, Mollitt DL, et al. The pediatric trauma score
score Coma Scale by intensive care nurses. Mayo Clin Proc 2007;82:435–8. as a predictor of injury severity: an objective assessment. J Trauma 1988;
85. McNarry AF, Goldhill DR. Simple bedside assessment of level of 28:425–9.
consciousness: comparison of two simple assessment scales with the 114. Bergstrom N, Braden BJ, Laguzza A, et al. The Braden scale for predicting
Glasgow Coma Scale. Anesthesia 2004;59:34–7. pressure sore risk. Nurs Res 1987;36(4):205–10.
86. Brott T, Adams HP Jr, Olinger CP, et al. Measurements of acute cerebral 115. National Pressure Ulcer Advisory Panel and European Pressure Ulcer
infarction: a clinical examination scale. Stroke 1989;20(7):864–70. Advisory Panel. Prevention and treatment of pressure ulcers. Clinical
87. Lyden PD, Lau GT. A critical appraisal of stroke evaluation and rating practice guideline. Washington DC: NPUAP; 2009.
scales. Stroke 1991;22(11):1345–52. 116. Miranda DR, de Rijk A, Schaufeli W. Simplified therapeutic intervention
88. Lyden P, Brott T, Tilley B, et al. Improved reliability of the NIH stroke scale scoring system: the TISS-28 items—results from a multicenter study. Crit
using video training. Stroke 1994;25:2220–5. Care Med 1996;24:64–73.
89. Lyden P, Raman R, Liu L, et al. NIHSS training and certification using a 117. Maset AL, Marmarou A, Ward JD, et al. Pressure-volume index in head
new digital video disk is reliable. Stroke 2005;36(11):2446–9. injury. J Neurosurg 1987;67(6):832–40.
90. Scandinavian Stroke Study Group. Multicenter trial of hemodilution in 118. Weiner GM, Lacey MR, Mackenzie L, et al. Decompressive craniectomy for
ischemic stroke: background and study protocol. Stroke 1985;16:885–90. elevated intracranial pressure and its effect on the cumulative ischemic
91. Orgogozo JM, Asplundh K, Boysen G. A unified form for neurological burden and therapeutic intensity levels after severe traumatic brain injury.
scoring of hemispheric stroke with motor impairment. Stroke 1992;23; Neurosurgery 2010;66:1111–9.
1678–9. 119. Shore PM, Hand LL, Roy L, et al. Reliability and validity of the pediatric
92. Hantsen L, De Weerdt W, DeKeyser J, et al. The European Stroke Scale. intensity level of therapy (PILOT) scale: a measure of the use of
Stroke 1994;25:2215–19. intracranial pressure-directed therapies. Crit Care Med 2006;34(7):1981–7.
Section II—Clinical and Laboratory Assessment 98.e3

120. Jennett B, Bond M. Assessment of outcome after severe brain damage. 138. Perlbarg V, Puybasset L, Tollard E, et al. Relation between brain lesion
Lancet 1975;1:480–4. location and clinical outcome in patients with severe traumatic brain
121. Husson EC, Ribbers GM, Willemse-van Son AH, et al. Prognosis of injury: a diffusion tensor imaging study using voxel-based approaches.
six-month functioning after moderate to severe traumatic brain injury: a Hum Brain Mapp 2009;30(12):3924–33.
systematic review of prospective cohort studies. J Rehabil Med 2010;42: 139. Sidaros A, Engberg AW, Sidaros K, et al. Diffusion tensor imaging during
425–36. recovery from severe traumatic brain injury and relation to clinical
122. Lu J, Murray GD, Steyerberg EW, et al. Effects of Glasgow Outcome Scale outcome: a longitudinal study. Brain 2008;131:559–72.
misclassification on traumatic brain injury clinical trials. J Neurotrauma 140. Tollard E, Galanaud D, Perlbarg V, et al. Experience of diffusion tensor
2008;25(6):641–51. imaging and 1H spectroscopy for outcome prediction in severe traumatic
123. Menon DK, Zahed C. Prediction of outcome in severe traumatic brain brain injury: preliminary results. Crit Care Med 2009;37(4):1448–55.
injury. Curr Opin Crit Care 2009;15:437–41. 141. Tshibanda L, Vanhaudenhuyse A, Galanaud D, et al. Magnetic resonance
124. Levy DE, Caronna JJ, Singer BH, et al. Predicting outcome from hypoxic- spectroscopy and diffusion tensor imaging in coma survivors: promises and
ischemic coma. JAMA 1985;253:1420–6. pitfalls. Prog Brain Res 2009;177:215–29.
125. Bernard SA, Gray TW, Buist MD, et al. Treatment of comatose survivors of 142. Wu O, Sorensen AG, Benner T, et al. Comatose patients with cardiac arrest:
out-of-hospital cardiac arrest with induced hypothermia. N Engl J Med predicting clinical outcome with diffusion-weighted MR imaging.
2002;346:557–63. Radiology 2009;252:173–81.
126. Hypothermia after cardiac arrest (HACA) study group. Mild therapeutic 143. Wu O, Batista LM, Lima FO, et al. Predicting clinical outcome in comatose
hypothermia to improve the neurologic outcome after cardiac arrest. cardiac arrest patients using early noncontrast computed tomography.
N Engl J Med 2002;346:549–56. Stroke 2011;42:985–92.
127. Wijdicks EF, Hijdra A, Young GB, et al. Practice parameter: prediction 144. Giacino JT, Ashwal S, Childs N, et al. The minimally conscious state:
of outcome in comatose survivors after cardiopulmonary resuscitation definition and diagnostic criteria. Neurology 2002;58(3):349–53.
(an evidence-based review). Neurology 2006;67:203–10. 145. Giacino JT. The vegetative and minimally conscious states: consensus-based
128. Rossetti AO, Oddo , Liaudet L, et al. Predictors of awakening from criteria for establishing diagnosis and prognosis. NeuroRehabilitation 2004;
postanoxic status epilepticus after therapeutic hypothermia. Neurology 19:293–8.
2009;72:744–9. 146. Working Party of the Royal College of Physicians. The vegetative state:
129. Young GB. The EEG in coma. J Clin Neurophysiol 2000;17:473–85. guidance on diagnosis and management. Clin Med 2003;3(3):249–54.
130. Chen R, Bolton CF, Young B. Prediction of outcome in patients with 147. Giacino JT, Kezmarsky MA, DeLuca J, et al. Monitoring rate of recovery to
postanoxic coma: a clinical and electrophysiological study. Crit Care Med predict outcome in minimally responsive patients. Arch Phys Med Rehabil
1996;24:672–8. 1991;72:897–901.
131. Gendo A, Kramer L, Häfner M, et al. Time-dependency of sensory evoked 148. Giacino JT, Kalmar K, Whyte J. The JFK coma recovery scale-revised:
potentials in comatose cardiac arrest survivors. Intensive Care Med 2001; measurement characteristics and diagnostic utility. Arch Phys Med Rehabil
27(8):1305–11. 2004;85(12):2020–9.
132. Young GB, Doig G, Ragazzoni A. Anoxic-ischemic encephalopathy: clinical 149. Schnakers C, Majerus S, Giacino J, et al. A French validation study of the
and electrophysiological associations with outcome. Neurocrit Care Coma Recovery Scale-Revised (CRS-R). Brain Injury 2008;22(10):786–92.
2005;2:159–64. 150. Laureys S, Owen AM, Schiff ND. Brain function in coma, vegetative state,
133. Young GB. Clinical practice: neurologic prognosis after cardiac arrest. and related disorders. Lancet Neurol 2004;3(9):537–46.
N Engl J Med 2009;361(6):605–11. 151. Owen AM, Coleman MR, Boly M, et al. Detecting awareness in the
134. Zandbergen EG, Hijdra A, Koelman JH, et al. Prediction of poor outcome vegetative state. Science 2006;313(5792):1402.
within the first 3 days of postanoxic coma. Neurology 2006;66(1):1133. 152. Monti MM, Vanhaudenhuyse A, Coleman MR, et al. Willful modulation of
135. Fugate JE, Wijdicks EF, Mandrekar J, et al. Predictors of neurologic brain activity in disorders of consciousness. N Engl J Med 2010;362(7):
outcome in hypothermia after cardiac arrest. Ann Neurol 2010;68(6): 579–89.
907–14. 153. Raichle ME, MacLeod AM, Snyder AZ, et al. A default mode of brain
136. Galanaud D, Puybasset L. Cardiac arrest: has the time of MRI come? Crit function. Proc Nat Acad Sci U S A 2001;98(2):676–82.
Care 2010;14(2):135–6. 154. Vanhaudenhuyse A, Noirhomme Q, Tshibanda LJ, et al. Default network
137. Newcombe V, Chatfield D, Outtrim J, et al. Mapping traumatic axonal connectivity reflects the level of consciousness in non-communicative
injury using diffusion tensor imaging: correlations with functional brain-damaged patients. Brain 2010;133:161–71.
outcome. PLoS One 2011;6(5):e19214.
Chapter
11  
II

Pain, Sedation, and Delirium


in Critical Illness
Kyla P. Terhune, E. Wesley Ely, and Pratik P. Pandharipande

primary disease, invasive monitoring, endotracheal intuba-


Introduction tion, phlebotomy, and procedures performed while in the
Patient comfort in an intensive care unit (ICU), from both a ICU. Inadequately treated discomfort and pain may cause an
mental and physical health perspective, is of utmost impor- increased stress response manifested by tachycardia, increased
tance for patient safety and recovery. Proper management of oxygen consumption, hypercoagulability, immunosuppres-
analgesia and sedation is especially important in patients sion, hypermetabolism, increased endogenous catecholamine
admitted to the neurocritical care unit (NCCU), because these activity, and, in patients with acute brain injury, an increase
patients have significant comorbidities and often management in ICP.2,14,15 Insufficient pain relief also can lead to sleep defi-
goals (e.g., sedation vs. need to evaluate consciousness or neu- ciency, disorientation, and anxiety.2,14 Oxygen consumption
rologic function) are opposing. In addition, health care pro- can be decreased by about 15% from baseline when adequate
viders have to be cognizant of the anxiety experienced by analgesia is provided, thus reversing some of these unwanted
family members and understand how to alleviate it. Family effects.14 In addition, a blood pressure increase that can con-
anxiety in the ICU is beyond the scope of this chapter and is tribute to intracranial bleeding after a craniotomy can be
discussed elsewhere.1 This chapter provides an overview of attenuated by effective pain control rather than antihyperten-
analgesia and sedation in the critically ill and how to monitor sives in some patients.
its proper use. The reader also is referred to recent reviews and Critically ill patients often experience anxiety that can be
guidelines on analgesia, sedation, and delirium in the ICU.2-12 alleviated by judicious sedation. Apart from anxiolysis, seda-
Most research has been on patients in medical or surgical tives also can produce amnesia, an effect that should be sought
ICUs with far less specific information available for patients only when neuromuscular blocking agents are adminis-
in the NCCU. The following topics are discussed: tered.2,15 There are also potentially unwanted mental health
effects from how sedation is used in the ICU. Unpleasant
1. Indications for the use of analgesia and sedation in the memories of an ICU stay have been associated with post-
ICU traumatic stress disorder (PTSD) symptoms in survivors.16,17
2. Potential complications of analgesic and sedative medica- However, some studies suggest that the occurrence of PTSD
tions, with a focus on the role of these medications in may be more specifically related to whether the memories
delirium of critical illness retained are delusional, and that factual memories, even if
3. Strategies to optimize the delivery of analgesia and seda- unpleasant, are less likely to cause PTSD.18,19 In addition there
tion are some emerging concerns that some drugs themselves may
4. Selection of analgesia and sedation for the critically ill produce cognitive dysfunction post ICU.
5. Management of delirium The use of analgesia and sedation in the ICU can be
a double-edged sword. Both are inherently necessary but
both can have unwanted consequences. The administration
Indications for Analgesia of analgesia and sedation therefore should account for
patient heterogeneity, including severity and prognosis, to
and Sedation develop a tailored, optimized approach for each individual.2
Comfort and safety are the two main indications for analgesia There are many approaches to the same patient, and these
and sedation in the ICU, and both analgesia and sedation may often are influenced by unexpected factors such as resources,
be administered for prolonged periods, particularly when a technology, and culture.2,20-23 Given the potential deleterious
patient is mechanically ventilated.2 In addition, in the NCCU effects of mismanagement, the Society of Critical Care
analgesia and sedation are used to treat intracranial pressure Medicine (SCCM)2 guidelines provide recommendations
(ICP) and can help improve brain oxygen.13 for appropriate use of sedative and analgesic medications
Pain or discomfort is treated by analgesics. The ICU patient and outline specific monitoring to be used when these
often has multiple sources of discomfort that arise from medications are administered to ICU patients. Adherence
© Copyright 2013 Elsevier Inc. All rights reserved. 99
100 Section II—Clinical and Laboratory Assessment

to these guidelines can help ensure that the medications electromyography, and possibly even muscle biopsy increase
are used properly and side effects are minimized. the sensitivity to identify this entity.36,38 Additional potential
risk factors include steroid use, age, hyperglycemia, and the
presence of systemic inflammatory response syndrome
Complications of (SIRS).34,39 None are definitive, but the association with pro-
longed immobilization makes it reasonable that reduction of
Sedative Administration sedation may improve the ability to mobilize critically ill
Drug pharmacokinetics and pharmacodynamics can be patients early, thus minimizing the occurrence or the severity
unpredictable in ICU patients because of their disease, drug of CIPM.
interactions, organ dysfunction, irregular absorption, and
variable protein binding.15 Each of these parameters can
contribute to complications, including need for mechanical Neuro-Specific Complications
ventilation, infection, delirium, and chronic illness among Analgesics and sedatives often are administered in continuous
survivors.24 Structured multidisciplinary approaches, includ- drips and have variable accumulation in critically ill patients;
ing protocols and algorithms can help reduce these adverse this can predispose patients to experience a withdrawal syn-
effects associated with sedation and analgesia in the ICU. drome on discontinuation.2,15,40,41 In addition, many psycho-
Sedation and analgesia algorithms should include identifica- logical conditions have been studied in relation to prolonged
tion of goals and specific targets, use of valid and reliable tools analgesia and sedation, particularly depression and PTSD,
to assess analgesia, agitation, and sedation, and incorporation with symptoms that appear directly related to the number of
of logical medication selection.25 days receiving such medications.42 There is also some sugges-
tion that not being able to recall specifics of their ICU stay
may have negative effects on patients’ long-term cognitive
Respiratory Depression abilities compared with those who have specific recall.43 ICU
Overuse of opiates and sedatives in the conscious patient can survivors also may recall pain, anxiety, fear, and inability to
cause respiratory depression.15 It therefore is important to sleep during their ICU stay when questioned later.44 These are
individualize doses of opiates and sedatives to achieve levels important aspects for the intensivist to keep in mind because
of analgesia and anxiolysis that are appropriate without they are effects that are unlikely to be manifested in the ICU
needing to establish an artificial airway. but have significant impact on the patient as an individual.

Prolonged Mechanical Ventilation Delirium


Deep sedation, especially when administered through con- Delirium is defined by the Diagnostic and Statistical Manual
tinuous infusions without daily interruption, can be associ- of Mental Disorders (DSM IV)45 as a disturbance of con-
ated with prolongation of mechanical ventilation and ICU sciousness with inattention, accompanied by a change in
length of stay.26 The inability to perform adequate daily cognition or perceptual disturbance that develops over a
patient assessments and neurologic examinations then may short period (hours to days) and fluctuates over time. Delir-
result in increased need to evaluate the neurologic status ium occurs in between 20% and 80% of mechanically venti-
with imaging or electrophysiologic studies.27 This contributes lated medical, trauma, and surgical ICU patients.46-48
to increased costs of care and potential complications associ- However, the majority of the cases are not recognized49-51;
ated with transport of a critically ill patient.26,27 On the other this creates a delay in diagnosis, or the symptoms are attrib-
hand, inadequate sedation and analgesia in an intubated uted to another cause.
patient can contribute to coughing or compromise ventila- One approach to classify delirium is to group patients
tion (i.e., patient optimization arises from conflicting thera- according to their level of alertness (hyperactive, hypoactive,
peutic priorities). or mixed).52 Peterson et al. found the distribution of the delir-
ium subtypes among ICU patients to be 1.6% hyperactive,
43.5% hypoactive, and 54.1% a mixed type.53 Hyperactive
Critical Illness Polyneuromyopathy delirium is assigned to patients with characteristics of agi­
Patients who survive critical illness often have long-lasting tation, restlessness, attempts to remove catheters or tubes,
physical, neuropsychiatric, and quality of life impairments.28,29 violent behaviors, and emotional lability and has an overall
Prolonged bed rest and stay in the ICU can be associated with better prognosis compared with hypoactive delirium, which is
persistent functional debilitation and reduced quality of life, manifested by flat affect, withdrawal, apathy, lethargy, and
even after ICU discharge.30-33 This functional debilitation decreased responsiveness.52,54-57 Given its subtle appearance in
often manifests as a polyneuropathy or an associated myopa- a population that may be subject to polypharmacy, it is not
thy. Although the two conditions are physiologically distinct surprising that hypoactive delirium remains unrecognized in
from each another, they frequently coexist and are recognized 66% to 74% of patients in a variety of settings.51,58-60 Use of a
as critical illness polyneuropathy and myopathy (CIPNM), validated monitoring system can help guard against the failure
critical illness neuromuscular abnormalities (CINMA), or to recognize this type of delirium.
critical illness polyneuromyopathy (CIPM).34-36 Recognition of delirium in ICU patients is important
CIPM manifests itself physically as a generalized, sym­ because its development is associated with longer time on
metrical muscle weakness that spares facial muscles. Although mechanical ventilation, ICU length of stay, higher costs,46,61,62
some suggest that physical examination and history may be increased likelihood of discharge to a nursing home,45,63,64 an
sufficient for the diagnosis,37 nerve conduction studies, increased in-hospital mortality,46,61,65 and long-term cognitive
Section II—Clinical and Laboratory Assessment 101

impairment in survivors.66 Indeed, patients who develop delir- Feature 1: Acute onset of mental status
ium during their ICU stay have a two- to threefold increase changes or a fluctuating course
risk of death after adjustment for covariates such as age,
presence of sepsis or acute respiratory distress syndrome AND
(ARDS) and Acute Physiology and Chronic Health Evaluation
Feature 2: Inattention
(APACHE) II scores.46,61,65 In addition, a delay in initiating
treatment for delirium and a longer duration of delirium are AND
strong predictors of increased mortality in the ICU.67-69
The delirium research field continues to expand, and mul-
tiple potential mechanisms of delirium have been proposed
Feature 3: Disorganized Feature 4: Altered level
including imbalances in the synthesis, release, and inactivation thinking
OR
of consciousness
of neurotransmitters55,57; inflammatory abnormalities induced
by endotoxins and cytokines70-73; impaired oxidative metabo-
lism74; a relative cholinergic deficiency secondary to this = DELIRIUM
impaired oxidative metabolism75; and variations in levels of Fig. 11.1  The confusion assessment method for the intensive
neurotransmitter precursor amino acids.76,77 care unit (CAM-ICU). This delirium instrument can be used with a
Who is at risk for developing delirium in the ICU popula- sedation scale as a two-step approach to assess consciousness and
tion? Baseline characteristics including genetic factors, demen- diagnose delirium. Patients are considered to have delirium if they
tia, chronic illness, advanced age, and depression are associated have Richmond Agitation-Sedation Scale (RASS) scores of –3 and
with an increased risk of developing delirium.78-81 Although greater (responsive to verbal stimulus) and are CAM-ICU positive by
many of these risk factors are not modifiable, there are iatro- having features 1 and 2, and either 3 or 4 positive. (Adapted with
permission from Dr. E.W. Ely, www.icudelirium.org.)
genic factors to which clinicians can pay close attention to help
reduce the risk of delirium. These factors include hypoxia,
metabolic and electrolyte imbalances, substance withdrawal
syndromes, infection, dehydration, hyperthermia, vascular separate diagnostic instrument to diagnose pediatric delirium
disorders, and sleep deprivation. In addition neurologic in critically ill children, the Pediatric Confusion Assessment
insults such as seizures, head trauma, and intracranial space– Method for Intensive Care Unit (pCAM-ICU), has been vali-
occupying lesions have been suggested to have a role in dated.87 Importantly, unlike bedside pain assessment tools that
delirium.78-80 have been validated in critically ill patients with neurologic
Benzodiazepines and opiates are implicated in the develop- disorders, delirium and sedation tools have had less rigorous
ment of delirium in non-ICU and in ICU patients (including research in NCCU patients.10
the postsurgical population).48,82-84 This often puts the clini-
cian in a paradoxical situation because these medications may
be necessary and are established as standard practice in clinical Sedation Scales
practice guidelines by the SCCM2 to provide analgesia and The diagnosis of delirium first requires the assessment of
anxiolysis to patients who undergo procedures or are on arousal level, followed by an assessment for delirium. Exam-
mechanical ventilation. Hence the need for sedation and anal- ples of sedation scales include the Ramsay Sedation (RS)
gesia must be balanced against its associated complications. scale,88 the Riker Sedation-Agitation Scale (SAS),89 and the
Although benzodiazepines have been shown to be risk Richmond Agitation-Sedation Scale (RASS).90,91 Although any
factors for delirium, the data that implicate fentanyl and of these can be used to assess agitation, this chapter focuses
morphine are less convincing.48,82,83,85 For example, Morrison on the RASS (Table 11.2) because it has excellent interrater
et al.85 in a prospective cohort study showed that inadequate reliability and can be used to detect changes over consecutive
analgesia may predispose patients to delirium, with patients days.92 The RASS90,91 is a 10-point scale that ranges from +4 to
who receive lower doses of parenteral morphine after hip frac- –5. A score of 0 signifies a calm and alert patient. Positive
ture to be at higher risk for developing delirium. This may also RASS scores denote levels of aggressive behavior, and negative
depend in part on other medications (e.g., some anesthe­ RASS scores denote less responsiveness, and differentiate
siologists often comment that premedication with scopol- between response to verbal (–1 to –3) and physical stimuli (–4
amine makes patients who arouse in pain more likely to suffer and –5). Patients with a RASS score of –3 and higher (lighter
delirium). levels of sedation) can further be assessed for delirium.

Delirium Scales Confusion Assessment Method


Screening tools and guidelines allow the clinician to approach for Intensive Care Unit
the patient at risk for delirium in a systematic fashion. Two The CAM-ICU is a scale that assesses delirium, that can be
scales (Fig. 11.1 and Table 11.1), the Confusion Assessment done in approximately 60 to 90 seconds.93 It has high specific-
Method for the ICU (CAM-ICU)47 and the Intensive Care ity and interrater reliability but may not always help detect
Delirium Screening Checklist (ICDSC)86 have been validated early delirium.94,95 In addition, the CAM-ICU allows for de-
to diagnose delirium in mechanically ventilated patients. lirium assessment in the face of sedative medication. This then
Delirium monitoring was included in the 2002 SCCM practice may be a potential limitation in the NCCU, where it is im­
guidelines for the sustained use of sedatives and analgesics in portant to determine if there is neurologic deterioration as
the critically ill adult.2 The management guidelines for pain, well. The CAM-ICU assesses four features: acute change or
agitation, and sedation were under revision in 2012. A fluctuation in mental status (feature 1), inattention (feature
102 Section II—Clinical and Laboratory Assessment

Table 11.1  The Intensive Care Delirium Table 11.2  Richmond Agitation-Sedation
Screening Checklist (ICDSC) Scale (RASS)
Patient Evaluation Score Term Description
Altered level of +4 Combative Overtly combative, violent,
consciousness (A-E)* immediate danger to staff
Inattention Difficulty in following a +3 Very agitated Pulls or removes tube(s) or
conversation or instructions. Easily catheter(s); aggressive
distracted by external stimuli.
+2 Agitated Frequent nonpurposeful
Difficulty in shifting focuses. Any
movement, fights ventilator
of these scores 1 point.
+1 Restless Anxious but movements not
Disorientation Any obvious mistake in time, place,
aggressive or vigorous
or person scores 1 point.
0 Alert and calm
Hallucinations- The unequivocal clinical
delusion-psychosis manifestation of hallucination or −1 Drowsy* Not fully alert, but has sustained
of behavior probably due to awakening (eye opening/eye
hallucination or delusion. Gross contact) to voice (≥10 seconds)
impairment in reality testing. Any
of these scores 1 point. −2 Light Briefly awakens with eye
sedation* contact to voice (<10 seconds)
Psychomotor Hyperactivity requiring the use of
agitation or additional sedative drugs or −3 Moderate Movement or eye opening to
retardation restraints in order to control sedation† voice (but no eye contact)
potential danger to oneself or −4 Deep No response to voice, but
others. Hypoactivity or clinically sedation† movement or eye opening to
noticeable psychomotor slowing. physical stimulation
Inappropriate speech Inappropriate, disorganized, or −5 Unarousable No response to voice or physical
or mood incoherent speech. Inappropriate stimulation
display of emotion related to
events or situation. Any of these From Sessler CN, Gosnell MS, Grap MJ, et al. The Richmond Agitation-
scores 1 point. Sedation Scale: validity and reliability in adult intensive care unit patients.
Am J Respir Crit Care Med 2002;166:1338–44; Ely EW, Truman B, Shintani A,
Sleep-wake cycle Sleeping less than 4 hours or et al. Monitoring sedation status over time in ICU patients: reliability and
disturbance waking frequently at night (do validity of the Richmond Agitation-Sedation Scale (RASS). JAMA 2003;289:
not consider wakefulness initiated 2983–91.
by medical staff or loud *Verbal stimulation.
environment). Sleeping during †
Physical stimulation.
most of the day. Any of these
scores 1 point.
Symptom fluctuation Fluctuation of the manifestation of
any item or symptom over 24 Intensive Care Delirium Screening Checklist
hours scores 1 point.
The ICDSC has eight diagnostic features (see Table 11.1).86 To
Total score (0-8) Score of 4 or more is consistent be diagnosed with delirium, a patient should have four or
with diagnosis of delirium
more features from the checklist during the evaluation
Adapted from Bergeron N, Dubois MJ, Dumont M, et al. intensive care period.86 Patients who demonstrate some features from the
delirium screening checklist: evaluation of a new screening tool. Intensive
Care Med 2001;27:859–64.
ICDSC but do not meet all the criteria for delirium are con-
*Level of consciousness: sidered to have subsyndromal delirium.96 Work by Ouimet
A: No response, score: None. et al.96 has shown that subsyndromal delirium is part of the
B: Response to intense and repeated stimulation (loud voice and pain), score: spectrum of acute brain dysfunction between normal and
None. full-feature delirium and is associated with worse outcomes
C: Response to mild or moderate stimulation, score: 1.
D: Normal wakefulness, score: 0. than in patients with normal cognition, though better than in
E: Exaggerated response to normal stimulation, score: 1. those with delirium.

2), disorganized thinking (feature 3), or an altered level Strategies to Optimize the Delivery
of consciousness (feature 4) (see Fig. 11.1). The diagnosis
of delirium using the combination of the RASS and the
of Analgesia and Sedation
CAM-ICU requires: Systematic ICU management protocols including rating scales
are important for analgesia, sedation, and delirium, and their
1. RASS score of –3 or higher and
use likely improves outcome including in ventilated ICU
2. Feature 1 of CAM-ICU (acute change or fluctuation in
patients.25,97,98 Studies that evaluate the efficacy of sedation
mental status) and
protocols and target-based sedation all show benefits such as
3. Feature 2 of CAM-ICU (inattention) and
shorter time on mechanical ventilation and less time in the
4. One of the following:
ICU and hospitals.99,100 However, subjective sedation scales
 Feature 3 (disorganized thinking) that depend on patient behavior and clinical examination may
 Feature 4 (altered level of consciousness) be insensitive during deep sedation.101 This is evident from a
Section II—Clinical and Laboratory Assessment 103

study that showed that almost 40% of patients admitted to a 100


medical ICU had episodes of burst suppression on their elec-
troencephalogram (EEG), despite being managed by the use
80
of targeted sedation with sedation scales. In addition, burst
suppression was associated with an increased in-hospital and

Patients alive (%)


SAT + SBT (n = 167)
6-month mortality after adjusting for severity of illness and 60
presence of sepsis.102 The bispectral analysis of the EEG with
a Bispectral Index (BIS) monitor can be measured easily in the
ICU setting. A frontal temporal sensor is attached to the 40 Usual care + SBT (n = 168)
patient; this derives a signal from several EEG components
that can be visualized but also numerically graded on a scale 20
from 100 (completely awake) to 0 (deep sedation).103 The
visualization of potential burst suppression allows the clini-
cian to recognize the presence of a drug-induced coma,104,105 0
and the graded scale allows the clinician to otherwise titrate 0 60 120 180 240 300 360
sedative medications to their desired effect. This is particularly Days
helpful in the case of the paralyzed patient, in whom the depth
of sedation and amnesia is difficult to determine. However, Fig. 11.2  Mortality benefit in the Awakening Breathing Controlled
trial. This “wake up and breathe” intervention resulted in 14% absolute
even in the nonparalyzed patient, the BIS monitor may provide
reduction in the risk of death within 1 year (58% in control reduced to
further information to better titrate sedation. 44% in treatment group). SAT, Spontaneous awakening trial; SBT,
The BIS and the SAS or RASS have been compared in spontaneous breathing trial. (From Girard TD, Kress JP, Fuchs BD, et al. Efficacy
mechanically ventilated patients and appear comparable (i.e., and safety of a paired sedation and ventilator weaning protocol for mechanically
BIS can differentiate inadequate from adequate sedation).106-108 ventilated patients in intensive care [Awakening and Breathing Controlled trial]: a
Similarly Riker et al.109 studied 39 ICU patients sedated after randomised controlled trial. Lancet 2008;371:126–34.)
cardiac surgery and observed significant agreement between
BIS and SAS. In addition, the authors described significant
changes in the BIS values as patients became more arousable. wake-up is beneficial in the NCCU or the ideal method to
In trauma patients the BIS wave form appears to coincide with detect neurologic deterioration in a sedated patient is still to
propofol on/off during daily spontaneous awakening trials be fully elucidated.
(SATs).106 However, the role of the BIS to guide the titration
of sedation in ICU remains unclear, and in one study110 use of
a BIS monitor to support clinical sedation management deci- Selection of Analgesia and
sions in ventilated patients did not reduce the amount of
sedation used, the length of mechanical ventilation (MV), or
Sedation for Critically Ill Patients
the length of ICU stay. In addition, the interpretation of BIS The selection of an opioid traditionally has depended on the
findings in traumatic brain injury (TBI) patients can be sub- likely duration of analgesic infusion and the pharmacology of
jective.106 Further studies on the applicability of neurologic the specific opioid.2 There is little evidence that one sedative
monitoring beyond sedation scales are under way. However, agent is better than another for control of ICP or cerebral
in the interim, sedation protocols and scales need to be imple- perfusion pressure in adults with severe brain injury.114 Mor-
mented to prevent unnecessary administration of sedatives phine and hydromorphone because of their longer duration
and analgesics to critically ill patients. of action are used typically for cases of prolonged infusions.2
One method used in ICUs to assess sedation is the “daily Hydromorphone has several advantages over morphine
wake-up.”111 The role of daily interruptions of continuous including a lack of histamine release and lack of a clinically
sedative infusions in critically ill patients has been evaluated active metabolite. This may result in less vasodilation and
in several studies. Patients managed with sedation cessation hypotension and less prolonged sedation in patients with
had a significant reduction in the duration of MV, shorter ICU renal insufficiency. Fentanyl’s rapid onset and short duration
and hospital length of stay, and fewer neuroradiologic proce- of action make it better suited for shorter periods of infusion.
dures and tracheostomies. Importantly there was no increase Its ease of titration and lack of histamine release also make it
in long-term mental health sequelae associated with the daily better suited to the hemodynamically unstable patient.2
“wake-ups.”27,112 Girard et al. in a multicenter randomized Remifentanil is one of the newest µ agonists with a rapid
clinical trial, the Awakening Breathing Controlled trial, exam- onset and offset of action. Its lack of accumulation makes it a
ined the efficacy and safety of a linked sedation and ventilator useful drug for continuous infusion in the ICU, especially the
weaning protocol for mechanically ventilated ICU patients.113 NCCU. Dahaba et al. conducted a randomized double blind
Patients were managed by a “targeted-sedation” strategy and study on ventilated patients receiving remifentanil or mor-
control group patients received daily spontaneous breathing phine. Those in the remifentanil group had more optimal
trials (SBTs). In the treatment group patients had a mandatory sedation and a shorter duration of mechanical ventilation.115
SAT as step A (total cessation of sedation long enough to wake Muellejans et al. compared remifentanil with fentanyl in ICU
to verbal stimulus or tolerate for 4 hours), followed by the SBT patients; the efficacy of sedation was similar but more propo-
as step B. The “wake-up and breathe” intervention was associ- fol was required in the fentanyl group. The time to extubation
ated with a 3-day reduction of MV, 1 day less in coma, 4 days was similar. However, the percentage of patients who experi-
less in the ICU and hospital, and a 14% absolute reduction in enced pain after extubation was greater in the remifentanil
the risk of death within 1 year (Fig. 11.2). Whether the daily group, suggesting the need for proactive pain management
104 Section II—Clinical and Laboratory Assessment

when weaning remifentanil.40 This is important in patients propofol, independent of the length of sedation, and weaning
with neurologic compromise because increased pain and the times were shorter with propofol, but this was only significant
potential for withdrawal may contribute to hypertension and in patients sedated for less than 36 hours.126 Together these
tachycardia that may be detrimental to the patient. Other various data suggest that propofol is associated with better
studies suggest that fentanyl and remifentanil provide similar outcomes when compared with benzodiazepines and appears
amounts of analgesia.116 to be particularly useful in neurologic patients in whom eva­
The safety and efficacy of analgesia-based sedation with luation of the neurologic status is important. In addition
remifentanil have been compared with conventional sedation when compared with morphine, use of propofol may reduce
with hypnotic-based regimens for patients with brain injury the need for other interventions to control ICP in patients
who require prolonged sedation for MV. Neurologic assess- with TBI.114
ment times and time to extubation were shorter for patients Dexmedetomidine, an alpha-2 agonist, provides an alterna-
who received remifentanil than those who received propofol tive to gamma-aminobutyric acid (GABA)–agonist sedative
or midazolam supplemented with morphine or fentanyl.117 medications. There are several potential advantages to its use:
Breen et al., in another randomized controlled trial that com- “arousable sedation,” suppression of delirium, and preserved
pared remifentanil-based sedation with a midazolam-based ventilatory drive.127,128 In 2007, Pandharipande et al. compared
regimen, observed that the duration of mechanical ventilation dexmedetomidine to lorazepam in mechanically ventilated
and duration of weaning were shorter in patients who received ICU patients in an RCT, and found that sedation with dexme-
remifentanil. There also was a trend toward shortened ICU detomidine resulted in more days alive without delirium or
stay.118 coma, a lower prevalence of coma, greater achievement of
There are several comparative trials between hypnotic sed- target sedation, and an important trend toward reduction in
ative regimens. Barr et al. used a pharmacologic model to mortality (27% in lorazepam and 17% in dexmedetomidine
compare lorazepam and midazolam infusions for ICU seda- group).129 Studies in ICU patients comparing dexmedetomi-
tion. Lorazepam was found to have twice the sedative potency dine to midazolam130,131 have shown superior outcomes with
and four times the amnestic potency of midazolam. The dexmedetomidine, including time on mechanical ventilation
emergence times for light and deep sedation were longer for and lower probability of delirium.130 Propofol and dexme-
lorazepam than midazolam.119 In a prospective randomized detomidine use is associated with similar outcomes in criti-
controlled study in trauma patients in which infusions of cally ill patients.131 After coronary artery bypass surgery,
lorazepam, midazolam, and propofol were compared, McCol- sedation with dexmedetomidine has similar times to weaning
lam et al. observed that oversedation occurred most fre- and extubation as propofol but there is a significant reduction
quently with lorazepam, and the greatest number of dosage in narcotic, β-blocker, antiemetic, nonsteroidal anti-
adjustments was required by the lorazepam group. Cost was inflammatory drug, epinephrine, and diuretic use in patients
greatest, and undersedation occurred most often with propo- who receive dexmedetomidine.132 Observational data suggest
fol. The results suggest that midazolam is the most titratable patients with neurologic disorders in the ICU may require
drug with the least amount of oversedation or undersedation higher doses of dexmedetomidine to achieve desired sedation
and that lorazepam was the most cost-effective agent for levels.133 It does not seem that higher doses are associated with
sedation.120 more adverse side effects.134
Propofol has been compared to individual benzodiazepines Sedation paradigms have changed in the ICU; there is less
in several studies, including in randomized controlled trials benzodiazepine exposure and unnecessary deep sedation.
(RCTs). Carson et al.121 conducted a randomized trial that However, with use has also come identification of disorders
compared intermittent lorazepam boluses to propofol infu- such as propofol infusion syndrome,135 propylene glycol toxic-
sion with daily interruption of sedatives in both groups. ity from lorazepam (see Chapter 22), and lipid accumulation
Patients in the propofol group had fewer mechanical ventila- with very prolonged emergence with high doses. In addition,
tion days, with a trend toward greater number of ventilator- risks can be associated with off-label use of attractive sedatives
free survival days.121 In an economic evaluation of propofol (e.g., unexpected deaths were observed when etomidate was
and lorazepam, overall propofol was less costly per patient tried off label) and there have been some propofol deaths
than lorazepam despite the considerably lower pharmacy unit related to infusion syndrome. Postmarketing discoveries of
cost of lorazepam. The lower costs likely were associated with new risks associated with off-label use of dexmedetomidine
the greater number of ventilator-free days in the propofol and remifentanil may still arise before it can be concluded that
group.122 Compared with midazolam infusion, patients sedated the change in sedation paradigm is an independent factor
by propofol infusion have faster and more reliable wake-up associated with improved patient outcomes especially in the
times,123 have equal efficacy of achieving sedation goals,124 NCCU.
spend a larger percentage of time at their target sedation goal,
and have more rapid extubation, although this does not always
mean earlier ICU discharge.125 However, propofol appears to
Management of Delirium
require more frequent dose adjustments, has a lower nurse- Delirium is a symptom of an underlying disorder and it is
rated quality of sedation, and may cause more cardiovascular common in the ICU; its prevalence may be as high as 80%.
depression and less amnesia than midazolam, with higher Development of delirium is associated with increased morbid-
costs.124 Average time to sedation and change in oxygen ity and excess mortality.12,69,136 Ely et al. have suggested a prag-
consumption is similar in patients who receive propofol or matic approach to this: screening, prevention, and restoration
midazolam.123 Walder et al. performed a systematic review of of brain function,137 or a bundle known as the ABCDE (for
trials that compared sedation with propofol or midazolam in awakening and breathing coordination, delirium monitoring,
2001. The duration of adequate sedation was greater with and exercise/early mobility) approach to ventilated patients.9
Section II—Clinical and Laboratory Assessment 105

It is important to recognize and proactively treat reversible of delirium. However, proper diagnosis is critical because ben-
causes—hypoxia, hypercarbia, hypoglycemia, sleep depriva- zodiazepines remain the drugs of choice for alcohol with-
tion metabolic derangements and shock among others—that drawal and seizures. Use of benzodiazepines for delirium may
might lead to delirium.138,139 High baseline inflammatory bio- lead to oversedation, exacerbation of delirium, and respiratory
markers such as procalcitonin and C-reactive protein (CRP) suppression that could lead to hypoxia and hypercarbia and
also may predict the likelihood of delirium in ventilated thus start a vicious cycle. Elderly patients are particularly
patients.140 Intracranial hemorrhage and in particular intrace- susceptible to the effects of benzodiazepines. Haloperidol, a
rebral hemorrhage may increase the risk of delirium in NCCU butyrophenone “typical” antipsychotic, is a good choice in the
patients.12 absence of risk factors such as prolonged QT syndrome
Just as the diagnosis and likely causes of delirium are and is still the most widely used neuroleptic agent for delir-
multifaceted, so too is prevention. The most important, and ium.149 A variety of administration routes have been used—
certainly least expensive, interventions are noninvasive inter- intermittent intravenous IV, continuous IV infusion, oral or
ventions that can be used by every member of the health enteral or intramuscular (IM) haloperidol.150 It does not sup-
care team. For example, Inouye et al. trained volunteers and press respiratory drive and works as a dopamine receptor
nurses to focus on several key goals including provision of antagonist by blocking the D2 receptor. This treats positive
regular stimulating activities, a nonpharmacologic sleep pro- symptomatology (hallucinations, unstructured thoughts pat-
tocol, early mobilization, appropriate and early removal of terns, etc.) and produces a variable sedative effect. The SCCM
catheters and restraints, optimization of sensory input using guidelines recommend haloperidol to treat delirium, although
the patient’s own seeing and hearing aids, and attention to the class of evidence (level C data) supporting it is not par-
hydration to prevent delirium in 852 hospitalized patients ticularly strong.2 There are few data from non-ICU studies
greater than 70 years old. There was a 40% reduction in de- that support the use of haloperidol for delirium. For example,
lirium, but the interventions did not appear to show benefit Kalisvaart et al. in an RCT observed a reduction in the severity
when results were again assessed at 6 months.141 However, of delirium but not the incidence of delirium in elderly
nonpharmacologic approaches are unlikely to be successful patients who had hip surgery.151 Milbrandt et al.152 also showed
once delirium is established.142 Other studies have shown a that mechanically ventilated patients who received haloperi-
decrease in the duration and severity of delirium but with dol had reduced mortality after adjusting for covariates,
little effect on overall incidence,143,144 have shown benefit though patients were not evaluated for delirium.
only to subgroups of patients,145 or have not shown any There are newer “atypical” antipsychotic agents (e.g., ris-
benefit at all.146 Although these studies were not specific to peridone, ziprasidone, quetiapine, and olanzapine) that affect
the ICU setting, they provide some insight into how to alter neurotransmitters other than dopamine and may provide
the course of delirium. Recently, Wang et al. have suggested benefit in delirium.153-157 Several studies have addressed their
that low-dose intravenous (IV) prophylactic haloperidol may use. Skrobik et al.156 compared olanzapine to haloperidol in
prevent delirium in elderly patients admitted to the ICU critically ill patients, and found that both agents had a
after noncardiac surgery.147 similar effect on the severity of delirium, though olanzapine
Because sedatives and analgesics play a role in the genesis had fewer side effects. In cardiac surgery patients, Prakanrat-
of delirium, there may be benefit to daily interruption of seda- tana et al.158 showed that a single dose of respiridone reduced
tives and analgesics, and awakening and breathing trials in a delirum. One case study of a patient with the CYP2D6 geno-
protocol-driven fashion to decrease psychoactive medication type showed that a switch from risperidone to quetiapine
use.26,27,100,113,148 Although these strategies have been used in the cleared persistent delirium and extrapyramidal symptoms in
ICU, the studies have either not evaluated for delirium or a timely fashion (i.e., genetic predisposition may also affect
when delirium was evaluated,113 did not show any differences responses to treatment with different medications).159
in delirium rates or duration. However, in a subgroup of septic Further studies are needed and based on pilot studies are
patients, the wake-up-and-breathe strategy did reduce delir- feasible.160
ium duration. Similarly, the Maximizing Efficacy of Targeted Antipsychotics in general have several adverse effects,
Sedation and Reducing Neurological Dysfunction (MENDS) including hypotension, dystonias, extrapyramidal effects,
trial showed that sedation with dexmedetomidine resulted in malignant hyperthermia, and anticholinergic effects. However,
more days alive without delirium or coma and a lower preva- the most worrisome and life-threatening adverse effect is the
lence of coma.129 torsades de pointes. Antipsychotics therefore should not be
In delirious patients, particularly those with hyperactive given to patients with prolonged QT intervals, and periodic
delirium, it may still be necessary to use medication to main- QT interval measurements should be obtained in those on
tain a healthy sleep-wake cycle. This creates a paradox because such medications. In addition, a recent meta-analysis of out-
it is often difficult to discern whether the medications are the patients who received antipsychotic medications for delirium
root of the delirium. These medications also have the potential in the setting of dementia, shows that prolonged antipsychotic
to have psychoactive effects of their own, thus potentially pro- treatment is associated with increased mortality and stroke
longing delirium. The best approach is to use medications among elderly patients.161,162
sparingly and in the smallest dose possible to achieve the
desired effect. This seems self-explanatory, but it is often dis-
regarded with the use of “as needed” medications, which are
Conclusion
not regulated or regularly reassessed. Accumulating evidence shows that effective use of sedation
There is no specific U.S. Food and Drug Administration and analgesia protocols used in a target-based manner can
(FDA)–approved medication for the treatment of delirium in benefit patients admitted to the ICU. However, the most effec-
the ICU. Benzodiazepines are best avoided in the management tive protocols to use in the NCCU still are being elucidated.
106 Section II—Clinical and Laboratory Assessment

In addition, analgesia and sedation can be a double-edged 25. Sessler CN, Pedram S. Protocolized and target-based sedation and analgesia
in the ICU. Anesthesiol Clin 2011;29(4):625–50.
sword because of inherent risks and because the medications
26. Kollef MH, Levy NT, Ahrens TS, et al. The use of continuous IV sedation is
used may limit the ability to accurately assess consciousness associated with prolongation of mechanical ventilation. Chest 1998;114:
or neurologic function. Therefore patients who receive anal- 541–8.
gesia and sedation in the ICU need to be carefully monitored 27. Kress JP, Pohlman AS, O’Connor MF, et al. Daily interruption of sedative
through validated scales such as the CAM-ICU, the ICDSC, infusions in critically ill patients undergoing mechanical ventilation. N Engl
J Med 2000;342:1471–7.
the RS scale, the RASS, and the Riker SAS, among others, to 28. Jackson JC, Mitchell N, Hopkins RO. Cognitive functioning, mental health,
ensure an optimal approach targeted to each individual. and quality of life in ICU survivors: an overview. Crit Care Clin 2009;25(3):
615–28, x.
29. Desai SV, Law TJ, Needham DM. Long-term complications of critical care.
References Crit Care Med 2011;39(2):371–9.
30. Dowdy DW, Eid MP, Sedrakyan A, et al. Quality of life in adult survivors of
1. Sauls JL, Warise LF. Interventions for anxiety in the critically ill: a guide for critical illness: a systematic review of the literature. Intensive Care Med 2005;
nurses and families. Nurs Clin North Am 2010;45(4):555–67, vi. 31:611–20.
2. Jacobi J, Fraser GL, Coursin DB, et al. Clinical practice guidelines for the 31. Needham DM, Dowdy DW, Mendez-Tellez P, et al. Studying outcomes of
sustained use of sedatives and analgesics in the critically ill adult. Crit Care intensive care unit survivors: measuring exposures and outcomes. Intensive
Med 2002;30:119–41. Care Med 2005;31(9):1153–60.
3. Erstad BL, Puntillo K, Gilbert HC, et al. Pain management principles in the 32. Herridge MS, Cheung AM, Tansey CM, et al. One-year outcomes in survivors
critically ill. Chest 2009;135(4):1075–86. of the acute respiratory distress syndrome. N Engl J Med 2003;348:683–93.
4. Chamorro C, Borrallo JM, Romera MA, et al. Anesthesia and analgesia 33. Herridge MS, Batt J, Hopkins RO. The pathophysiology of long-term
protocol during therapeutic hypothermia after cardiac arrest: a systematic neuromuscular and cognitive outcomes following critical illness.
review. Anesth Analg 2010;110(5):1328–35. Crit Care Clin 2008;24:79–99, x.
5. Martin J, Heymann A, Bäsell K, et al. Evidence and consensus-based German 34. de Letter MCJ, Schmitz PIM, Visser LH, et al. Risk factors for the
guidelines for the management of analgesia, sedation and delirium in development of polyneuropathy and myopathy in critically ill patients.
intensive care: short version. Ger Med Sci 2010;8:Doc 02. Crit Care Med 2001;29:2281–6.
6. Honiden S, Siegel MD. Analytic reviews: managing the agitated patient in 35. Stevens RD, Dowdy DW, Michaels RK, et al. Neuromuscular dysfunction
the ICU: sedation, analgesia, and neuromuscular blockade. J Intensive Care acquired in critical illness: a systematic review. Intensive Care Med 2007;33:
Med 2010;25(4):187–204. 1876–91.
7. Wu CL, Raja SN. Treatment of acute postoperative pain. Lancet 2011; 36. Bednarik J, Lukas Z, Vondracek P. Critical illness polyneuromyopathy: the
377(9784):2215–25. electrophysiological components of a complex entity. Intensive Care Med
8. Patel SB, Kress JP. Sedation and analgesia in the mechanically ventilated 2003;29:1505–14.
patient. Am J Respir Crit Care Med 2012;185(5):486–97. Epub 2011 37. Morris C, Trinder JT. Electrophysiology adds little to clinical signs in critical
Oct 20. illness polyneuropathy and myopathy. Crit Care Med 2002;30:2612.
9. Morandi A, Brummel NE, Ely EW. Sedation, delirium and mechanical 38. Berek K, Margreiter J, Willeit J, et al. Polyneuropathies in critically ill
ventilation: the ‘ABCDE’ approach. Curr Opin Crit Care 2011;17(1):43–9. patients: a prospective evaluation. Intensive Care Med 1996;22:849–55.
10. Teitelbaum JS, Ayoub O, Skrobik Y. A critical appraisal of sedation, analgesia 39. Weber-Carstens S, Deja M, Koch S, et al. Risk factors in critical illness
and delirium in neurocritical care. Can J Neurol Sci 2011;38(6):815–25. myopathy during the early course of critical illness: a prospective
11. Power I. An update on analgesics. Br J Anaesth 2011;107(1):19–24. Epub observational study. Crit Care 2010;14(3):R119. Epub 2010, Jun 18.
2011, May 30. 40. Muellejans B, Lopez A, Cross MH, et al. Remifentanil versus fentanyl for
12. Frontera JA. Delirium and sedation in the ICU. Neurocrit Care analgesia based sedation to provide patient comfort in the intensive care
2011;14(3):463–74. unit: a randomized, double-blind controlled trial [ISRCTN43755713].
13. Pascual JL, Georgoff P, Maloney-Wilensky E, et al. Reduced brain tissue Crit Care 2004;8:R1–R11.
oxygen in traumatic brain injury: are most commonly used interventions 41. Vinik HR, Kissin I. Rapid development of tolerance to analgesia during
successful? J Trauma 2011;70(3):535–46. remifentanil infusion in humans. Anesth Analg 1998;86:1307–11.
14. Kress JP, Pohlman AS, Hall JB. Sedation and analgesia in the intensive care 42. Nelson BJ, Weinert CR, Bury CL, et al. Intensive care unit drug use and
unit. Am J Respir Crit Care Med 2002;166:1024–8. subsequent quality of life in acute lung injury patients. Crit Care Med 2000;
15. Gehlbach BK, Kress JP. Sedation in the intensive care unit. Curr Opin Crit 28:3626–30.
Care 2002;8:290–8. 43. Larson MJ, Weaver LK, Hopkins RO. Cognitive sequelae in acute respiratory
16. Weinert CR, Sprenkle M. Post-ICU consequences of patient wakefulness distress syndrome patients with and without recall of the intensive care unit.
and sedative exposure during mechanical ventilation. Intensive Care Med J Int Neuropsychol Soc 2007;13:595–605.
2008;34(1):82–90. Epub 2007 Aug 17. 44. Ethier C, Burry L, Martinez-Motta C, et al. Recall of intensive care unit stay
17. Rotondi AJ, Chelluri L, Sirio C, et al. Patients’ recollections of stressful in patients managed with a sedation protocol or a sedation protocol with
experiences while receiving prolonged mechanical ventilation in an intensive daily sedative interruption: a pilot study. J Crit Care 2011;26(2):127–32.
care unit. Crit Care Med 2002;30:746–52. 45. American Psychiatric Association. Diagnostic and statistical manual of
18. Jones C, Griffiths RD, Humprhis G. Disturbed memory and amnesia related mental disorders. Fourth edition, text revision. Washington, DC:
to intensive care. Memory 2000;8:79–94. American Psychiatric Association; 2000.
19. Jones C, Griffiths RD, Humphris G, et al. Memory, delusions, and the 46. Ouimet S, Kavanagh BP, Gottfried SB, et al. Incidence, risk factors and
development of acute posttraumatic stress disorder-related symptoms after consequences of ICU delirium. Intensive Care Med 2007;33:66–73.
intensive care. Crit Care Med 2001;29:573–80. 47. Ely EW, Inouye SK, Bernard GR, et al. Delirium in mechanically ventilated
20. Weinert CR, Chlan L, Gross C. Sedating critically ill patients: factors patients: validity and reliability of the confusion assessment method for the
affecting nurses’ delivery of sedative therapy. Am J Crit Care 2001;10:156–65. intensive care unit (CAM-ICU). JAMA 2001;286:2703–10.
21. Weinert CR, Calvin AD. Epidemiology of sedation and sedation adequacy 48. Pandharipande P, Cotton B, Shintani A, et al. Prevalence and risk factors for
for mechanically ventilated patients in a medical and surgical intensive care development of delirium in surgical and trauma intensive care unit patients.
unit. Crit Care Med 2007;35:393–401. J Trauma 2008;65:34–41.
22. Payen JF, Chanques G, Mantz J, et al. Current practices in sedation and 49. Ely EW, Siegel MD, Inouye SK. Delirium in the intensive care unit: an
analgesia for mechanically ventilated critically ill patients: a prospective under-recognized syndrome of organ dysfunction. Semin Respir Crit Care
multicenter patient-based study. Anesthesiology 2007;106:687–95. Med 2001;22:115–26.
23. Mehta S, Burry L, Fischer S. Canadian survey of the use of sedatives, 50. Francis J, Martin D, Kapoor WN. A prospective study of delirium in
analgesics, and neuromuscular blocking agents in critically ill patients. Crit hospitalized elderly. JAMA 1990;263:1097–101.
Care Med 2006;34:374–80.
24. Nseir S, Makris D, Mathieu D, et al. Intensive care unit-acquired infection as A complete list of references for this chapter can be found online at
a side effect of sedation. Crit Care 2010;14(2):R30. Epub 2010, Mar 15. www.expertconsult.com.
Section II—Clinical and Laboratory Assessment 106.e1

References 30. Dowdy DW, Eid MP, Sedrakyan A, et al. Quality of life in adult survivors of
critical illness: a systematic review of the literature. Intensive Care Med
1. Sauls JL, Warise LF. Interventions for anxiety in the critically ill: a guide for 2005;31:611–20.
nurses and families. Nurs Clin North Am 2010;45(4):555–67, vi. 31. Needham DM, Dowdy DW, Mendez-Tellez P, et al. Studying outcomes of
2. Jacobi J, Fraser GL, Coursin DB, et al. Clinical practice guidelines for the intensive care unit survivors: measuring exposures and outcomes. Intensive
sustained use of sedatives and analgesics in the critically ill adult. Crit Care Care Med 2005;31(9):1153–60.
Med 2002;30:119–41. 32. Herridge MS, Cheung AM, Tansey CM, et al. One-year outcomes in
3. Erstad BL, Puntillo K, Gilbert HC, et al. Pain management principles in the survivors of the acute respiratory distress syndrome. N Engl J Med 2003;
critically ill. Chest 2009;135(4):1075–86. 348:683–93.
4. Chamorro C, Borrallo JM, Romera MA, et al. Anesthesia and analgesia 33. Herridge MS, Batt J, Hopkins RO. The pathophysiology of long-term
protocol during therapeutic hypothermia after cardiac arrest: a systematic neuromuscular and cognitive outcomes following critical illness.
review. Anesth Analg 2010;110(5):1328–35. Crit Care Clin 2008;24:79–99, x.
5. Martin J, Heymann A, Bäsell K, et al. Evidence and consensus-based 34. de Letter MCJ, Schmitz PIM, Visser LH, et al. Risk factors for the
German guidelines for the management of analgesia, sedation and delirium development of polyneuropathy and myopathy in critically ill patients.
in intensive care: short version. Ger Med Sci 2010;8:Doc 02. Crit Care Med 2001;29:2281–6.
6. Honiden S, Siegel MD. Analytic reviews: managing the agitated patient in 35. Stevens RD, Dowdy DW, Michaels RK, et al. Neuromuscular dysfunction
the ICU: sedation, analgesia, and neuromuscular blockade. J Intensive Care acquired in critical illness: a systematic review. Intensive Care Med 2007;
Med 2010;25(4):187–204. 33:1876–91.
7. Wu CL, Raja SN. Treatment of acute postoperative pain. Lancet 2011; 36. Bednarik J, Lukas Z, Vondracek P. Critical illness polyneuromyopathy: the
377(9784):2215–25. electrophysiological components of a complex entity. Intensive Care Med
8. Patel SB, Kress JP. Sedation and analgesia in the mechanically ventilated 2003;29:1505–14.
patient. Am J Respir Crit Care Med 2012;185(5):486–97. Epub 2011 Oct 20. 37. Morris C, Trinder JT. Electrophysiology adds little to clinical signs in
9. Morandi A, Brummel NE, Ely EW. Sedation, delirium and mechanical critical illness polyneuropathy and myopathy. Crit Care Med 2002;30:2612.
ventilation: the ‘ABCDE’ approach. Curr Opin Crit Care 2011;17(1):43–9. 38. Berek K, Margreiter J, Willeit J, et al. Polyneuropathies in critically ill
10. Teitelbaum JS, Ayoub O, Skrobik Y. A critical appraisal of sedation, patients: a prospective evaluation. Intensive Care Med 1996;22:849–55.
analgesia and delirium in neurocritical care. Can J Neurol Sci 2011;38(6): 39. Weber-Carstens S, Deja M, Koch S, et al. Risk factors in critical illness
815–25. myopathy during the early course of critical illness: a prospective
11. Power I. An update on analgesics. Br J Anaesth 2011;107(1):19–24. Epub observational study. Crit Care 2010;14(3):R119. Epub 2010, Jun 18.
2011, May 30. 40. Muellejans B, Lopez A, Cross MH, et al. Remifentanil versus fentanyl for
12. Frontera JA. Delirium and sedation in the ICU. Neurocrit Care 2011;14(3): analgesia based sedation to provide patient comfort in the intensive care
463–74. unit: a randomized, double-blind controlled trial [ISRCTN43755713].
13. Pascual JL, Georgoff P, Maloney-Wilensky E, et al. Reduced brain tissue Crit Care 2004;8:R1–11.
oxygen in traumatic brain injury: are most commonly used interventions 41. Vinik HR, Kissin I. Rapid development of tolerance to analgesia during
successful? J Trauma 2011;70(3):535–46. remifentanil infusion in humans. Anesth Analg 1998;86:1307–11.
14. Kress JP, Pohlman AS, Hall JB. Sedation and analgesia in the intensive care 42. Nelson BJ, Weinert CR, Bury CL, et al. Intensive care unit drug use and
unit. Am J Respir Crit Care Med 2002;166:1024–8. subsequent quality of life in acute lung injury patients. Crit Care Med 2000;
15. Gehlbach BK, Kress JP. Sedation in the intensive care unit. Curr Opin Crit 28:3626–30.
Care 2002;8:290–8. 43. Larson MJ, Weaver LK, Hopkins RO. Cognitive sequelae in acute respiratory
16. Weinert CR, Sprenkle M. Post-ICU consequences of patient wakefulness distress syndrome patients with and without recall of the intensive care
and sedative exposure during mechanical ventilation. Intensive Care Med unit. J Int Neuropsychol Soc 2007;13:595–605.
2008;34(1):82–90. Epub 2007 Aug 17. 44. Ethier C, Burry L, Martinez-Motta C, et al. Recall of intensive care unit stay
17. Rotondi AJ, Chelluri L, Sirio C, et al. Patients’ recollections of stressful in patients managed with a sedation protocol or a sedation protocol with
experiences while receiving prolonged mechanical ventilation in an daily sedative interruption: a pilot study. J Crit Care 2011;26(2):127–32.
intensive care unit. Crit Care Med 2002;30:746–52. 45. American Psychiatric Association. Diagnostic and statistical manual of
18. Jones C, Griffiths RD, Humprhis G. Disturbed memory and amnesia mental disorders. Fourth edition, text revision. Washington, DC: American
related to intensive care. Memory 2000;8:79–94. Psychiatric Association; 2000.
19. Jones C, Griffiths RD, Humphris G, et al. Memory, delusions, and the 46. Ouimet S, Kavanagh BP, Gottfried SB, et al. Incidence, risk factors and
development of acute posttraumatic stress disorder-related symptoms after consequences of ICU delirium. Intensive Care Med 2007;33:66–73.
intensive care. Crit Care Med 2001;29:573–80. 47. Ely EW, Inouye SK, Bernard GR, et al. Delirium in mechanically ventilated
20. Weinert CR, Chlan L, Gross C. Sedating critically ill patients: factors patients: validity and reliability of the confusion assessment method for the
affecting nurses’ delivery of sedative therapy. Am J Crit Care 2001;10:156–65. intensive care unit (CAM-ICU). JAMA 2001;286:2703–10.
21. Weinert CR, Calvin AD. Epidemiology of sedation and sedation adequacy 48. Pandharipande P, Cotton B, Shintani A, et al. Prevalence and risk factors for
for mechanically ventilated patients in a medical and surgical intensive care development of delirium in surgical and trauma intensive care unit
unit. Crit Care Med 2007;35:393–401. patients. J Trauma 2008;65:34–41.
22. Payen JF, Chanques G, Mantz J, et al. Current practices in sedation and 49. Ely EW, Siegel MD, Inouye SK. Delirium in the intensive care unit: an
analgesia for mechanically ventilated critically ill patients: a prospective under-recognized syndrome of organ dysfunction. Semin Respir Crit Care
multicenter patient-based study. Anesthesiology 2007;106:687–95. Med 2001;22:115–26.
23. Mehta S, Burry L, Fischer S. Canadian survey of the use of sedatives, 50. Francis J, Martin D, Kapoor WN. A prospective study of delirium in
analgesics, and neuromuscular blocking agents in critically ill patients. Crit hospitalized elderly. JAMA 1990;263:1097–101.
Care Med 2006;34:374–80. 51. Inouye SK. The dilemma of delirium: clinical and research controversies
24. Nseir S, Makris D, Mathieu D, et al. Intensive care unit-acquired infection regarding diagnosis and evaluation of delirium in hospitalized elderly
as a side effect of sedation. Crit Care 2010;14(2):R30. Epub 2010, Mar 15. medical patients. Am J Med 1994;97:278–88.
25. Sessler CN, Pedram S. Protocolized and target-based sedation and analgesia 52. Milisen K, Foreman MD, Godderis J, et al. Delirium in the hospitalized
in the ICU. Anesthesiol Clin 2011;29(4):625–50. elderly: nursing assessment and management. Nurs Clin North Am 1998;
26. Kollef MH, Levy NT, Ahrens TS, et al. The use of continuous IV sedation is 33:417–36.
associated with prolongation of mechanical ventilation. Chest 1998;114: 53. Peterson JF, Pun BT, Dittus RS, et al. Delirium and its motoric subtypes: a
541–8. study of 614 critically ill patients. J Am Geriatr Soc 2006;54:479–84.
27. Kress JP, Pohlman AS, O’Connor MF, et al. Daily interruption of sedative 54. O’Keeffe ST, Lavan JN. Clinical significance of delirium subtypes in older
infusions in critically ill patients undergoing mechanical ventilation. N Engl people. Age Ageing 1999;28:115–9.
J Med 2000;342:1471–7. 55. Meagher DJ, Trzepacz PT. Motoric subtypes of delirium. Semin Clin
28. Jackson JC, Mitchell N, Hopkins RO. Cognitive functioning, mental health, Neuropsychiatry 2000;5:75–85.
and quality of life in ICU survivors: an overview. Crit Care Clin 2009;25(3): 56. Meagher DJ, Hanlon DO, Mahony EO, et al. Relationship between
615–28, x. symptoms and motoric subtype of delirium. J Neuropsychiatry Clin
29. Desai SV, Law TJ, Needham DM. Long-term complications of critical care. Neurosci 2000;12:51–6.
Crit Care Med 2011;39(2):371–9. 57. Justic M. Does “ICU psychosis” really exist? Crit Care Nurse 2000;20:28–37.
106.e2 Section II—Clinical and Laboratory Assessment

58. Marcantonio ER, Goldman L, Mangione CM, et al. A clinical prediction 87. Smith HA, Boyd J, Fuchs DC, et al. Diagnosing delirium in critically ill
rule for delirium after elective noncardiac surgery. JAMA 1994;271: children: validity and reliability of the Pediatric Confusion Assessment
134–9. Method for the Intensive Care Unit. Crit Care Med 2011;39(1):150–7.
59. Sanders AB. Missed delirium in older emergency department patients: 88. Ramsay MA, Savege TM, Simpson BR, et al. Controlled sedation with
a quality-of-care problem. Ann Emerg Med 2002;39:338–41. alphaxalone-alphadolone. BMJ 1974;2:656–9.
60. Hustey FM, Meldon SW. The prevalence and documentation of impaired 89. Riker RR, Picard JT, Fraser GL. Prospective evaluation of the Sedation-
mental status in elderly emergency department patients. Ann Emerg Med Agitation Scale for adult critically ill patients. Crit Care Med 1999;27:
2002;39:248–53. 1325–9.
61. Lin SM, Liu CY, Wang CH, et al. The impact of delirium on the survival of 90. Ely EW, Truman B, Shintani A, et al. Monitoring sedation status over time
mechanically ventilated patients. Crit Care Med 2004;32:2254–9. in ICU patients: reliability and validity of the Richmond Agitation-Sedation
62. Milbrandt EB, Deppen S, Harrison PL, et al. Costs associated with delirium Scale (RASS). JAMA 2003;289:2983–91.
in mechanically ventilated patients. Crit Care Med 2004;32:955–62. 91. Sessler CN, Gosnell MS, Grap MJ, et al. The Richmond Agitation-Sedation
63. Inouye SK, Schlesinger MJ, Lydon TJ. Delirium: a symptom of how hospital Scale: validity and reliability in adult intensive care unit patients. Am J
care is failing older persons and a window to improve quality of hospital Respir Crit Care Med 2002;166:1338–44.
care. Am J Med 1999;106:565–73. 92. Ely EW, Truman B, Shintani A, et al. Monitoring sedation status over time
64. Levkoff SE, Evans DA, Liptzin B, et al. Delirium: the occurrence and in ICU patients: reliability and validity of the Richmond Agitation-Sedation
persistence of symptoms among elderly hospitalized patients. Arch Intern Scale (RASS). JAMA 2003;289(22):2983–91.
Med 1992;152:334–40. 93. Pun BT, Gordon SM, Peterson JF, et al. Large-scale implementation of
65. Ely EW, Shintani A, Truman B, et al. Delirium as a predictor of mortality in sedation and delirium monitoring in the intensive care unit: a report from
mechanically ventilated patients in the intensive care unit. JAMA two medical centers. Crit Care Med 2005;33:1199–205.
2004;291:1753–62. 94. Guenther U, Popp J, Koecher L, et al. Validity and reliability of the
66. Girard TD, Jackson JC, Pandharipande PP, et al. Delirium as a predictor of CAM-ICU Flowsheet to diagnose delirium in surgical ICU patients. J Crit
long-term cognitive impairment in survivors of critical illness. Crit Care Care 2010;25(1):144–51. Epub 2009, Oct 13.
Med 2010;38(7):1513–20. 95. van Eijk MM, van den Boogaard M, van Marum RJ, et al. Routine
67. Pisani MA, Kong SY, Kasl SV, et al. Days of delirium are associated with use of the confusion assessment method for the intensive care unit: a
1-year mortality in an older intensive care unit population. Am J Respir multicenter study. Am J Respir Crit Care Med 2011;184(3):340–4. Epub
Crit Care Med 2009;180(11):1092–7. Epub 2009, Sep 10. 2011, May 11.
68. Heymann A, Radtke F, Schiemann A, et al. Delayed treatment of delirium 96. Ouimet S, Riker R, Bergeon N, et al. Subsyndromal delirium in the ICU:
increases mortality rate in intensive care unit patients. J Int Med Res 2010; evidence for a disease spectrum. Intensive Care Med 2007;33:1007–13.
38(5):1584–95. 97. Payen JF, Bosson JL, Chanques G, et al. Pain assessment is associated with
69. Shehabi Y, Riker RR, Bokesch PM, et al. Delirium duration and mortality in decreased duration of mechanical ventilation in the intensive care unit:
lightly sedated, mechanically ventilated intensive care patients. Crit Care a post hoc analysis of the DOLOREA study. Anesthesiology 2009;111(6):
Med 2010;38(12):2311–8. 1308–16.
70. Fulkerson W, MacIntyre N, Stamler J, et al. Pathogenesis and treatment of 98. Skrobik Y, Ahern S, Leblanc M, et al. Protocolized intensive care unit
the adult respiratory distress syndrome. Arch Intern Med 1996;156:29–38. management of analgesia, sedation, and delirium improves analgesia and
71. Bellingan G. The pulmonary physician in critical care, 6: the pathogenesis subsyndromal delirium rates. Anesth Analg 2010;111(2):451–63. Epub
of ALI/ARDS. Thorax 2002;57:540–6. 2010, Apr 7.
72. Arvin B, Neville LF, Barone FC, et al. Brain injury and inflammation: 99. Brattebo G, Hofoss D, Flaatten H, et al. Effect of a scoring system and
a putative role of TNF alpha. Ann N Y Acad Sci 1995;765:62–71. protocol for sedation on duration of patients’ need for ventilator support in
73. Fink MP, Evans TW. Mechanisms of organ dysfunction in critical illness: a surgical intensive care unit. BMJ 2002;324:1386–9.
report from a Round Table Conference held in Brussels. Intensive Care Med 100. Brook AD, Ahrens TS, Schaiff R, et al. Effect of a nursing-implemented
2002;28:369–75. sedation protocol on the duration of mechanical ventilation. Crit Care Med
74. Lipowski ZJ. Delirium: acute confusional states, Rev. edition. New York: 1999;27:2609–15.
Oxford University Press; 1990. 101. De Deyne C, Struys M, Decruyenaere J, et al. Use of continuous bispectral
75. Blass JP, Gibson GE, Duffy TE, et al. Cholinergic dysfunction: a common EEG monitoring to assess depth of sedation in ICU patients. Intensive Care
denominator in metabolic encephalopathies. In: Pepeu G, Landinsky H, Med 1998;24:1294–8.
editors. Cholinergic Mechanisms. New York, NY: Plenum Press; 1981. 102. Watson PL, Shintani A, Tyson R, et al. Presence of EEG burst suppression
p. 921–8. in critically ill patients is associated with increased mortality. Crit Care Med
76. Van Der Mast RC. Pathophysiology of delirium. J Geriatr Psychiatry Neurol 2008;36(12):3171–7.
1998;11:138–45. 103. Sigl JC, Chamoun NG. An introduction to bispectral analysis for the
77. Van Der Mast RC, van den Broek WW, Fekkes D, et al. Is delirium after electroencephalogram. J Clin Monit 1994;10:392–404.
cardiac surgery related to plasma amino acids and physical condition? 104. Wolter S, Friedel C, Bohler K, et al. Presence of 14Hz spindle oscillations in
J Neuropsychiatry Clin Neurosci 2000;12:57–63. the human EEG during deep anesthesia. Clin Neurophysiol 2006;117:157–68.
78. Webb JM, Carlton EF, Geeham DM. Delirium in the intensive care unit: 105. Leistritz L, Jager H, Schelenz C, et al. New approaches for the detection and
are we helping the patient? Crit Care Nurs Quat 2000;22:47–60. analysis of electroencephalographic burst-suppression patterns in patients
79. Francis J. Drug-induced delirium: diagnosis and treatment. CNS Drugs under sedation. J Clin Monit Comput 1999;15:357–67.
1996;5:103–14. 106. Ogilvie MP, Pereira BM, Ryan ML, et al. Bispectral index to monitor
80. Inouye SK, Charpentier PA. Precipitating factors for delirium in propofol sedation in trauma patients. J Trauma 2011;71(5):1415–21.
hospitalized elderly persons: predictive model and interrelationship with 107. Karamchandani K, Rewari V, Trikha A, et al. Bispectral index correlates well
baseline vulnerability. JAMA 1996;275:852–7. with Richmond agitation sedation scale in mechanically ventilated critically
81. McNicoll L, Pisani MA, Zhang Y, et al. Delirium in the intensive care unit: ill patients. J Anesth 2010;24(3):394–8. Epub 2010, Mar 12.
occurrence and clinical course in older patients. J Am Geriatr Soc 108. Simmons LE, Riker R, Prato M, et al. Assessing sedation during intensive
2003;51:591–8. care unit mechanical ventilation with the bispectral index and the
82. Marcantonio ER, Juarez G, Goldman L, et al. The relationship of postoperative sedation-agitation scale. Crit Care Med 1999;27:1499–504.
delirium with psychoactive medications. JAMA 1994;272:1518–22. 109. Riker RR, Fraser GL, Simmons LE, et al. Validating the sedation-agitation
83. Dubois MJ, Bergeron N, Dumont M, et al. Delirium in an intensive care scale with the bispectral index and visual analog scale in adult ICU patients
unit: a study of risk factors. Intensive Care Med 2001;27:1297–304. after cardiac surgery. Intensive Care Med 2001;27:853–8.
84. Pandharipande P, Shintani A, Peterson J, et al. Lorazepam is an independent 110. Weatherburn C, Endacott R, Tynan P, et al. The impact of bispectral index
risk factor for transitioning to delirium in intensive care unit patients. monitoring on sedation administration in mechanically ventilated patients.
Anesthesiology 2006;104:21–6. Anaesth Intensive Care 2007;35:204–8.
85. Morrison RS, Magaziner J, Gilbert M, et al. Relationship between pain and 111. Augustes R, Ho KM. Meta-analysis of randomised controlled trials on daily
opioid analgesics on the development of delirium following hip fracture. sedation interruption for critically ill adult patients. Anaesth Intensive Care
J Gerontol A Biol Sci Med Sci 2003;58:76–81. 2011;39(3):401–9.
86. Bergeron N, Dubois MJ, Dumont M, et al. intensive care delirium screening 112. Kress JP, Gehlbach B, Lacy M, et al. The long-term psychological effects of
checklist: evaluation of a new screening tool. Intensive Care Med 2001;27: daily sedative interruption on critically ill patients. Am J Respir Crit Care
859–64. Med 2003;168:1457–61.
Section II—Clinical and Laboratory Assessment 106.e3

113. Girard TD, Kress JP, Fuchs BD, et al. Efficacy and safety of a paired sedation 137. Vasilevskis EE, Pandharipande PP, Girard TD, et al. A screening, prevention,
and ventilator weaning protocol for mechanically ventilated patients in and restoration model for saving the injured brain in intensive care unit
intensive care (Awakening and Breathing Controlled trial): a randomised survivors. Crit Care Med. 2010;38(10 Suppl):S683–91.
controlled trial. Lancet 2008;371:126–34. 138. Weinhouse GL, Schwab RJ, Watson PL, et al. Bench-to-bedside review:
114. Roberts DJ, Hall RI, Kramer AH, et al. Sedation for critically ill adults with delirium in ICU patients: importance of sleep deprivation. Crit Care
severe traumatic brain injury: a systematic review of randomized controlled 2009;13(6):234. Epub 2009, Dec 7.
trials. Crit Care Med 2011;39(12):2743–51. 139. Van Rompaey B, Elseviers MM, Schuurmans MJ, et al. Risk factors for
115. Dahaba AA, Grabner T, Rehak PH, et al. Remifentanil versus morphine delirium in intensive care patients: a prospective cohort study. Crit Care
analgesia and sedation for mechanically ventilated critically ill patients: 2009;13(3):R77. Epub 2009, May 20.
a randomized double blind study. Anesthesiology 2004;101:640–6. 140. McGrane S, Girard TD, Thompson JL, et al. Procalcitonin and C-reactive
116. Spies C, Macguill M, Heymann A, et al. A prospective, randomized, protein levels at admission as predictors of duration of acute brain
double-blind, multicenter study comparing remifentanil with fentanyl in dysfunction in critically ill patients. Crit Care 2011;15(2):R78. Epub 2011,
mechanically ventilated patients. Intensive Care Med 2011;37(3):469–76. Mar 2.
Epub 2010, Dec 17. 141. Inouye SK, Bogardus ST Jr, Charpentier PA, et al. A multicomponent
117. Karabinis A, Mandragos K, Stergiopoulos S, et al. Safety and efficacy of intervention to prevent delirium in hospitalized older patients. N Engl J
analgesia-based sedation with remifentanil versus standard hypnotic-based Med 1999;340:669–76.
regimens in intensive care unit patients with brain injuries: a randomised, 142. Skrobik Y. Delirium prevention and treatment. Anesthesiol Clin
controlled trial [ISRCTN50308308]. Crit Care 2004;8:R268–80. 2011;29(4):721–7.
118. Breen D, Karabinis A, Malbrain M, et al. Decreased duration of mechanical 143. Milisen K, Foreman MD, Abraham IL, et al. A nurse-led interdisciplinary
ventilation when comparing analgesia-based sedation using remifentanil intervention program for delirium in elderly hip-fracture patients. JAGS
with standard hypnotic-based sedation for up to 10 days in intensive care 2001;49:523–32.
unit patients: a randomised trial [ISRCTN47583497]. Crit Care 2005;9: 144. Lundstrom M, Edlund A, Karlsson S, et al. A multifactorial intervention
R200–10. program reduces the duration of delirium, length of hospitalization, and
119. Barr J, Zomorodi K, Bertaccini EJ, et al. A double-blind, randomized mortality in delirious patients. J Am Geriatr Soc 2005;53:622–8.
comparison of IV lorazepam versus midazolam for sedation of ICU 145. Marcantonio ER, Flacker JM, Wright RJ, et al. Reducing delirium after hip
patients via a pharmacologic model. Anesthesiology 2001;95:286–98. fracture: a randomized trial. J Am Geriatr Soc 2001;49:516–22.
120. McCollam JS, O’Neil MG, Norcross ED, et al. Continuous infusions of 146. Cole MG, McCusker J, Bellavance F, et al. Systematic detection and
lorazepam, midazolam, and propofol for sedation of the critically ill multidisciplinary care of delirium in older medical inpatients: a
surgery trauma patient: a prospective, randomized comparison. Crit Care randomized trial. CMAJ 2002;167:753–9.
Med 1999;27:2454–8. 147. Wang W, Li HL, Wang DX, et al. Haloperidol prophylaxis decreases delirium
121. Carson SS, Kress JP, Rodgers JE, et al. A randomized trial of intermittent incidence in elderly patients after noncardiac surgery: a randomized
lorazepam versus propofol with daily interruption in mechanically controlled trial. Crit Care Med 2011;Nov 3. Epub ahead of print.
ventilated patients. Crit Care Med 2006;34:1326–32. 148. Hooper MH, Girard TD. Sedation and weaning from mechanical
122. Barrientos-Vega R, Mar Sanchez-Soria M, Morales-Garcia C, et al. ventilation: linking spontaneous awakening trials and spontaneous
Prolonged sedation of critically ill patients with midazolam or propofol: breathing trials to improve patient outcomes. Anesthesiol Clin
impact on weaning and costs. Crit Care Med 1997;25:33–40. 2011;29(4):651–61.
123. Kress JP, O’Connor MF, Pohlman AS, et al. Sedation of critically ill patients 149. Ely EW, Stephens RK, Jackson JC, et al. Current opinions regarding the
during mechanical ventilation: a comparison of propofol and midazolam. importance, diagnosis, and management of delirium in the intensive care
Am J Respir Crit Care Med 1996;153:1012–8. unit: a survey of 912 healthcare professionals. Crit Care Med
124. Weinbroum AA, Halpern P, Rudick V, et al. Midazolam versus propofol for 2004;32:106–12.
long-term sedation in the ICU: a randomized prospective comparison. 150. Wang EH, Mabasa VH, Loh GW, et al. haloperidol dosing strategies in the
Intensive Care Med 1997;23:1258–63. treatment of delirium in the critically ill. Neurocrit Care 2011;Oct 26. Epub
125. Hall RI, Sandham D, Cardinal P, et al. Propofol vs midazolam for ICU ahead of print.
sedation: a Canadian multicenter randomized trial. Chest 2001;119:1151–9. 151. Kalisvaart KJ, de Jonghe JF, Bogaards MJ, et al. Haloperidol prophylaxis for
126. Walder B, Elia N, Henzi I, et al. A lack of evidence of superiority of elderly hip-surgery patients at risk for delirium: a randomized placebo-
propofol versus midazolam for sedation in mechanically ventilated critically controlled study. J Am Geriatr Soc 2005;53:1658–66.
ill patients: a qualitative and quantitative systematic review. Anesth Analg 152. Milbrandt EB, Kersten A, Kong L, et al. Haloperidol use is associated with
2001;92:975–83. lower hospital mortality in mechanically ventilated patients. Crit Care Med
127. Pichot C, Ghignone M, Quintin L. Dexmedetomidine and clonidine: from 2005;33:226–9.
second-to-first-line sedative agents in the critical care setting? J Intensive 153. Tune LE, Damlouji NF, Holland A, et al. Association of postoperative
Care Med 2011;Apr 27. Epub ahead of print. delirium with raised serum levels of anticholinergic drugs. Lancet
128. Mantz J, Josserand J, Hamada S. Dexmedetomidine: new insights. 1981;2:651–3.
Eur J Anaesthesiol 2011;28(1):3–6. 154. Tune LE, Egeli S. Acetylcholine and delirium. Dement Geristr Cogn Disord
129. Pandharipande PP, Pun BT, Herr DL, et al. Effect of sedation with 2001;10:342–4.
dexmedetomidine vs lorazepam on acute brain dysfunction in mechanically 155. Foreman M, Milisen K, Marcantonia EM. Prevention and treatment
ventilated patients: the MENDS randomized controlled trial. JAMA 2007; strategies for delirium. Primary Psychiatry 2004;11:52–8.
298:2644–53. 156. Skrobik YK, Bergeron N, Dumont M, et al. Olanzapine vs haloperidol:
130. Riker RR, Shehabi Y, Bokesch PM, et al. Dexmedetomidine vs midazolam treating delirium in a critical care setting. Intensive Care Med
for sedation of critically ill patients: a randomized trial. JAMA 2004;30:444–9.
2009;301(5):489−99. 157. Alao AO, Soderberg M, Pohl EL, et al. Aripiprazole in the treatment of
131. Jakob SM, Ruokonen E, Grounds RM, et al. Dexmedetomidine vs delirium. Int J Psychiatry Med 2005;35:429–33.
midazolam or propofol for sedation during prolonged mechanical 158. Prakanrattana U, Prapaitrakool S. Efficacy of risperidone for prevention of
ventilation: two randomized controlled trials. JAMA 2012;307(11):1151−60. postoperative delirium in cardiac surgery. Anaesth.Intensive Care
132. Herr DL, Sum-Ping ST, England M. ICU sedation after coronary artery 2007;35:714–9.
bypass graft surgery: dexmedetomidine-based versus propofol-based 159. Kato D, Kawanishi C, Kishida I, et al. Delirium resolving upon switching
sedation regimens. J Cardiothorac Vasc Anesth 2003;17:576–84. from risperidone to quetiapine: implication of the CYP2D6 genotype.
133. Grof TM, Bledsoe KA. Evaluating the use of dexmedetomidine in Psychosomatics 2005;46:374–5.
neurocritical care patients. Neurocrit Care 2010;12(3):356–61. 160. Girard TD, Pandharipande PP, Carson SS, et al. Feasibility, efficacy, and
134. Jones GM, Murphy CV, Gerlach AT, et al. High-dose dexmedetomidine for safety of antipsychotics for intensive care unit delirium: the MIND
sedation in the intensive care unit: an evaluation of clinical efficacy and randomized, placebo-controlled trial. Crit Care Med 2010;38(2):428–37.
safety. Ann Pharmacother 2011;45(6):740–7. Epub 2011, Jun 10. 161. Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of
135. Diedrich DA, Brown DR. Analytic reviews: propofol infusion syndrome in conventional vs. atypical antipsychotic medications. N Engl J Med
the ICU. J Intensive Care Med 2011;26(2):59–72. 2005;353:2335–41.
136. Salluh JI, Soares M, Teles JM, et al. Delirium epidemiology in critical care 162. Schneider LS, Dagerman KS, Insel P. Risk of death with atypical
(DECCA): an international study. Crit Care 2010;14(6):R210. Epub 2010, antipsychotic drug treatment for dementia: meta-analysis of randomized
Nov 23. placebo-controlled trials. JAMA 2005;294:1934–43.
Chapter
12  
II

Outcome Scales and


Neuropsychological Outcome
Rosette C. Biester

clinical trials network, and the Common Data Elements


Introduction Workgroups.2-10
The incidence of mortality and illness-related complications
has long been documented in the medical literature. More
recently, the measurement of outcomes has gained increasing
Functional Outcome Scales
attention because advanced medical procedures, medications, Functional outcome scales have been used at the time of
and therapies have led to better survival rates. One of the main discharge from critical care units but more often in post–
reasons to measure status at the time of injury or illness or critical care settings to supplement physical and medical
soon after is that long-term outcomes can be predicted from measurements of status, progress, and stability.11-17 These
these initial assessments. In addition, the efficacy of treatment scales include general measures of disability, more detailed
must be determined through objective measures rather than measures of motor skills and other physical abilities, and com-
simply using clinical judgment or behavioral observation. The prehensive assessments of physical, cognitive, and emotional
medical community has recognized the need to measure out- functioning.18-20 Outcome scales are typically used by raters to
comes in several areas, including medical and physical prog- determine levels of functioning through observation of behav-
ress and functional or rehabilitation recovery. However, ior or alternatively, caregiver report of patterns of behavior.
inadequate or incomplete assessment occurs all too frequently Factors that should be considered in deciding which scales to
and does not provide enough data to reasonably predict long- use are:
term outcome.
1. Level of patient arousal and alertness: Patients who are
The term outcomes no longer refers to simply one or two
either in coma, have fluctuating consciousness, or are in a
functional areas. To establish a complete picture of an indi-
minimally conscious state will not exhibit many behaviors
vidual’s status or level of functioning, several outcomes
that can be rated on these measures. Instead, a coma-
should be considered, and if possible, measured at different
emergent scale should be chosen as a more meaningful
time points. In this chapter functional outcomes, including
alternative to standard outcome measures. A consistent
quality of life measures, and neuropsychologic or neurocog-
level of arousal and alertness is typically needed for mean-
nitive outcomes, are reviewed and discussed. These measures
ingful assessment on the measures that are reviewed in the
are of particular importance to the management of patients
following text.
with acute neurologic abnormalities as patients with no phys-
2. Environmental factors (e.g., limited available observation
ical deficit can be profoundly disabled because of cognitive or
time): Observation periods may be limited due to the
neurobehavioral deficits. It is now well documented that crit-
fragile medical status of the patient. This may dictate the
ical illness or a stay in an intensive care unit (ICU) can be
specific scale chosen for outcome ratings, or the frequency
associated with long-term neurocognitive function.1 Addi-
with which ratings can be made.
tional outcomes, including mood and behavioral function-
3. Training of raters: Raters who have extensive training in
ing, also are important in determining a patient’s complete
observation of critical care patients are much better suited
status, but they are not reviewed in this chapter. This chapter
to using the more in-depth scales with finer gradations of
does not present an exhaustive list of measures, but hopefully
functioning. In contrast, raters with less training may be
will provide the reader with a representative sampling of
limited to the more popular, easier-to-administer scales.
measures that could be useful in critical care and post–critical
care settings. The reader is referred to recent reviews on these Two types of functional outcome scales are reviewed:
topics, recommendations from the traumatic brain injury (1) brief functional outcome measures appropriate for ratings
© Copyright 2013 Elsevier Inc. All rights reserved. 107
108 Section II—Clinical and Laboratory Assessment

of critical care clients and (2) comprehensive tests of func- Disability Rating Scale
tional outcome. DRS is a widely used measure consisting of eight items that
fall within four categories. Scores range from 0 (no disabil-
ity) to 29 (extreme vegetative state). It is brief but more
Brief Tests of Functional Outcome comprehensive than other short scales because it contains
The measures reviewed here are essentially global ratings com- items that measure all three categories listed by the World
pleted by observers. Typically observed patients are either Health Organization: Impairment, Disability, and Handi-
critically ill or emerging from critical states. These patients are cap.30 The first three items were incorporated from the
usually incapable of either completing ratings themselves, or Glasgow Coma Scale (GCS)31 with slight modifications: Eye
participating in cognitive testing. Opening, Communication Ability, and Motor Response.
These items are measures of impairment, and are very
important behaviors to observe and rate in critically ill or
Glasgow Outcome Scale and Extended emerging patients whose behavioral repertoire is severely
Glasgow Outcome Scale restricted. Measures of Feed­ing, Toileting, and Grooming
The Glasgow Outcome Scale (GOS) is one of the earliest reflect disability, and the category of Employability is a
outcome measures developed. It has been used frequently in measure of handicap. An early study indicated that it was
outcome studies after acute neurologic disorders such as more sensitive than the GCS in measuring clinical changes in
traumatic brain injury (TBI) for which it was originally severe TBI patients.32 Similarly, it was found to be more sen-
described, as well as stroke and subarachnoid hemorrhage sitive to detect improvement than the GOS for an inpatient
(SAH).18,21 It has five levels of outcome, and is the basis for rehabilitation sample.33
other scales that have followed, including the Extended The DRS is used commonly because it has been demon-
Glasgow Outcome Scale (E-GOS) and the Disability Rating strated to be reliable and valid,34,35 is relatively brief and easy
Scale (DRS).22 These two scales are now more widely used to score, and can either be self-administered (though not a
than the GOS, which is regarded as too broad in its catego- common mode of administration) or scored through inter-
ries and therefore not sensitive to changes in patient status. view with the client, family member, or other caretaker. One
In addition the GOS is not an adequate assessment measure limitation of this scale is that it does not usually detect subtle
after mild brain injury such as concussion, for which there changes that may occur in higher functioning individuals,
are many specific concussion assessment tools: Axon Sports especially mild TBI survivors.
Computerized Cognitive Assessment Tool (CCAT) or
CogState-Sport,23 Standardized Concussion Assessment Tool
2 (SCAT2),24 and Immediate Post-Concussion Assessment Modified Rankin Scale
and Cognitive Testing (ImPACT),25 among others. Research The original Rankin Scale was developed in 1957 by J. Rankin.36
in mild TBI and sports-related concussion suggests that no It generally is used to measure the degree of handicap in stroke
one measure is ideal for obtaining a comprehensive picture patients. This six-level version has been supplanted by the
of the client’s status. Instead, a combination of measures or modified Rankin Scale (mRS), which was published in 1988.37
specific neurocognitive tests assessing multiple domains is It has an additional grade ranging from 0 (no symptoms) to
more effective. 6 (dead). The grades in between all provide a degree of detail
The E-GOS added three additional disability and outcome that sets it apart from other less descriptive measures, and
categories to the original five, resulting in these eight catego- hence increases its utility even though it is unidimensional.
ries: Dead, Vegetative State, Lower Severe Disability, Upper For example, level three is: “Moderate disability. Requires
Severe Disability, Lower Moderate Disability, Upper Moder- some help, but able to walk unassisted.” A study using neu-
ate Disability, Lower Good Recovery, and Upper Good rologists, nurses, and physiotherapists as raters found reliabil-
Recovery. Global outcome, rather than specific ability-related ity to be satisfactory.38 To improve interobserver reliability,
outcomes, is the main outcome assessed by the E-GOS. A another study used a multimedia training process,39 another
well-designed study of 399 patients with intracerebral hem- feature that sets it apart from other equally brief scales. The
orrhage utilized the E-GOS together with the modified mRS (or dichotomized mRS) has been used extensively in
Rankin Scale described in this chapter to determine global critical care studies to measure medical status and outcomes
outcome at 90 days.26 Although not as extensively cited as at various time points after serious neurologic events includ-
the DRS, several studies have used the E-GOS to determine ing intracerebral hemorrhage and acute ischemic stroke.40-48
functional outcome, including investigations several years
after the neurologic event. One study explored functional
outcome 10 years after TBI.27 Poorer outcome on the E-GOS Barthel Index
was associated with significantly longer posttraumatic The Barthel Activities of Daily Living (ADL) Index is one of
amnesia; more impairment on cognitive measures of infor- the earliest measures that focuses specifically on activities of
mation processing speed, attention, memory, and executive daily living as well as mobility. It was originally introduced in
function; and higher anxiety levels. The E-GOS is used also 1965, and although the initial version is still widely used, a
to measure functional outcome after SAH.28,29 Rating reli- modified Barthel Index with a 100-point assessment of inde-
ability has been improved from the original GOS with the pendence in 10 activities of daily living is even more compre-
addition of a structured interview. The E-GOS is popular in hensive and allows for finer discriminations between ratings.49
neurosurgical-based outcome studies, but in rehabilitation The variables measured are feeding, bathing, grooming, dress-
and neuropsychology studies it has not had the popularity ing, bowel, bladder, toilet use, transfers (bed to chair
afforded the DRS. and back), mobility, and stairs. The usefulness of the scale has
Section II—Clinical and Laboratory Assessment 109

been demonstrated in several diagnostic groups, including (7) role limitations due to emotional problems and (8) mental
stroke,50,51 TBI,52,53 and even spinal cord injury.54 Obviously, health (psychological distress and psychological well-being).
a scale such as the Barthel Index gives a more specific and Each of these domains is important to assess for patients
complete picture of the individual than global measures of who have had neurointensive illnesses, injuries, or interven-
disability or handicap. Yet ratings of the 10 variables in this tions, because they are all basic areas of functioning that can
index can only be made when the individual progresses to a be negatively affected in seriously ill patients. The SF-36 is
more advanced stage of recovery. widely cited in the health and medical literature for many dif-
ferent diagnostic groups including severe TBI, spinal cord
injury, or even elective surgery.61-63 It has even been discussed
Ordinal Analysis and Sliding Dichotomy as a quality-of-life measure.64 The SF-36 has been very useful
In most phase III clinical trials ordinal outcome measures are in determining progress over time with repeat assessments65
collapsed into a binary scale. For example, in TBI trials the and in assessing functional outcome many years post injury.66,67
5-point GOS at 6 months often is used as the primary outcome The Short Form-12 (SF-12) is a much shorter version of
measure. For analysis it is dichotomized to unfavorable (Dead, the SF-36 that still measures the eight domains but contains
Vegetative, or Severe Disability) or favorable outcome (Mod- only one or two items from each of these. The SF-12 is gaining
erate Disability or Good Recovery). In stroke trials the mRS, increasing popularity and more recently has been used in place
an ordinal scale (six categories), or Barthel Index also is col- of its longer counterpart.19,68,69 Several studies have compared
lapsed into binary scales. This collapse of an ordinal scale has the SF-36 with the SF-12, and although some information
two potential problems: (1) loss of information55 and (2) pre- inevitably will be missing with any shortened form including
cedence is given to a specific transition in the scale. For the SF-12, there appears to be enough evidence suggesting
example, for the GOS, this is the difference between moderate that this shorter version can be a good substitute, especially
and severe disability. The failure of many phase III TBI trials when assessment time is limited or a number of other mea-
over the past several decades has raised questions about con- sures are also employed in the study. In comparing the SF-12
ventional statistical approaches.56 To address this use of ordinal with the SF-36 in a trauma population, one conclusion was
rather than dichotomous outcome, analysis is recommended. that the Physical Component Score in both was moderately
Simulation studies suggest that this can increase statistical responsive to change and was equivalent using either the short
power57 because all patients can contribute to the detection of or longer measure.70 Other studies comparing these two mea-
a treatment effect. The analysis is performed through a sliding sures in non-neurologic patients have also shown the useful-
dichotomy in which the point of dichotomy is customized to ness of the SF-12 as an alternative to the longer version.71,72
each patient’s baseline prognosis.58 Analysis of the Corticoste-
roid Randomization After Significant Head Injury (CRASH)
trial data demonstrates that ordinal analysis of the GOS can Sickness Impact Profile
improve statistical power to detect a treatment effect with This health outcome measure is considerably longer than the
equal sample size.59 The Optimising Analysis of Stroke Trials more widely used SF-36 but is more comprehensive and
(OAST) collaboration suggests a benefit of ordinal analysis in detailed in assessing many important components of everyday
stroke research.60 functioning. The 12 categories of the Sickness Impact Profile
(SIP) are (1) Sleep and Rest, (2) Emotional Behavior, (3) Body
Care and Movement, (4) Home Management, (5) Mobility,
Comprehensive Tests of Functional (6) Social Interaction, (7) Ambulation, (8) Alertness Behavior,
Outcome, Quality of Life, or Health Status (9) Communication, (10) Work, (11) Recreation and Pas-
Measures designed to assess health status after the acute stages times, and (12) Eating. It is relatively lengthy, and may actually
of critical care are typically more comprehensive in evaluating be the longest well-known outcomes measure at 136 items
areas of functioning. These measures can be administered as that can be self- or interviewer administered. Scoring is done
early as several days after discharge from intensive care, and by categories, broader dimensions, or the total SIP score.
indefinitely afterward. These questionnaires are completed In deciding whether to use the shorter SF-36 or the longer
either by the patient or an observer or caretaker familiar with SIP, the clinician or researcher must weigh the importance of
the person’s functional status. These measures are different assessment time limitation versus the need for a very thorough
from the brief tests covered in the previous section in which assessment of everyday functioning. For example, in a study
an observer must rate the patient’s functions rather than the of the cognitive, mood, and quality-of-life impairments in
patient participating actively in answering test items. acute respiratory distress syndrome (ARDS) survivors, the
authors concluded that because ARDS patients had not
been well characterized in terms of the broad range of deficits
Short Form-36 and Short Form-12 that persist, the most comprehensive quality-of-life measure
The Short Form-36 Health Survey (SF-36) consists of 36 items should be selected.14 Similarly, Frontera et al. who studied the
that measure health status, outcomes, and quality of life. It effect of delayed cerebral ischemia on quality of life 3 months
measures 8 health concepts and yields 2 summary measures, after SAH, used the entire SIP rather than a shorter measure.13
making it a relatively comprehensive instrument often selected Others have used both the SIP, which some describe as a
by clinicians and researchers alike to assess status and change. measure of functional status, and the SF-36, considered to be
The 8 health domains cover physical and emotional or psy- a quality of life measure, even though the SIP has been
chological status: (1) physical functioning, (2) role limitations described as a quality-of-life measure also.12 In another study
due to physical health problems, (3) bodily pain, (4) general of long-term health status, the specific categories of Work,
health, (5) vitality (energy or fatigue), (6) social functioning, Ambulation, Home Management, Recreation and Pastimes,
110 Section II—Clinical and Laboratory Assessment

and Alertness Behavior were most problematic at 12- to rated on a 7-point scale: 7 is complete independence, 6 is modi-
18-month follow-up in survivors of major trauma.11 It is pos- fied independence, 5 equals supervision or setup, 4 equals
sible, however, to extract only those subscales of the SIP that minimal contact assistance (client expends 75% or more of the
are needed for a particular clinical or research purpose. For effort), 3 is moderate assistance (client expends between 50%
example, in a study of stroke survivors assessed at 6 months and 75% of the effort), 2 is maximal assistance (client expends
post stroke, only 6 of the 12 categories were utilized.16 The between 25% and 50% of the effort), and 1 equals total assis-
original SIP has sometimes been excluded from outcomes tance (client expends <25% of the effort).79 The FIM is typi-
research because of its length; hence, a shorter alternative cally completed by a group of rehabilitation specialists with
was developed. The SIP 68 was found to correlate well with expertise in particular functional areas, often in weekly patient
the SIP.73 care conferences. Eighteen areas are assessed: Eating; Groom-
ing; Bathing; Dressing—Upper Body; Dressing—Lower Body;
Toileting; Bladder Management—Level of Assistance; Bowel
Mayo Portland Adaptability Inventory-4 Management—Level of Assistance; Transfers: Bed, Chair,
The Mayo Portland Adaptability Inventory-4 (MPAI-4) is the Wheelchair; Transfers: Toilet; Transfers: Tub or Shower; Loco-
newest version of the original Portland Adaptability Inventory motion: Walk/Wheelchair; Locomotion: Stairs; Comprehen-
first published in 1987.74,75 This fifth version of the scale rates sion; Expression; Social Interaction; Prob­lem Solving; and
the most common sequelae of acquired brain injury (ABI), Memory.
including physical, cognitive, emotional, behavioral, and social
problems.76 The areas of communication and employment
were further refined for this version. The MPAI-4 is a carefully
Neuropsychological Measures
constructed inventory that provides clinical evaluations of Neuropsychological functioning typically refers to a broad
ABI survivors during the postacute stage following injury. range of cognitive abilities. However, a comprehensive defini-
Patients who are suitable for evaluation on this scale range tion entails not only cognitive, but also motor, perceptual, and
from mild to severe brain injury. The MPAI-4 may be com- emotional or psychological functions. After a neurologic
pleted by persons with ABI, their significant others, and/or event, some or all of these domains may be negatively affected.
medical or rehabilitation professionals. In one study, 134 indi- Patients who have been in ICUs are typically assessed after
viduals with ABI were rated by the persons with ABI, family hospital discharge, rather than in the ICU, because their
and significant others, and rehabilitation staff.77 Satisfactory medical status is usually too compromised to proceed with
internal consistency for the MPAI-4 was found, regardless of cognitive testing until after discharge.68,80 Tests that measure
the source of ratings. these functions (abilities) vary in length and scope. They can
The MPAI-4 has three subscales: (1) Ability Index, (2) Ad- be classified as:
justment Index, and (3) Participation Index. It has 29 clinical
1. Neuropsychological screens
items, as well as six additional items covering preinjury and
2. Neuropsychological batteries
postinjury information about alcohol use, drug use, psychotic
3. Individual tests measuring specific cognitive domains
symptoms, law violations, other conditions causing physical
impairment, and other conditions causing cognitive impair- In selecting tests and deciding on how extensive testing should
ment. Scoring consists of a total score and each of the three be, the evaluator needs to be aware of guidelines in the field
index scores. Outcomes are described by five functional levels: and cognitive issues specific to the diagnostic group of the
Good Outcome, Mild Limitations, Mild to Moderate Limita- individual being tested. This chapter reviews only neuropsy-
tions, Moderate to Severe Limitations, and Severe Limitations. chological screens because they are more appropriate for use
The manual, forms, and other language versions (including in acute care patient assessment. Moreover, an entire chapter
French and German translations) can be easily downloaded on neuropsychological assessment alone would be needed to
from the Center for Outcome Measurement in Brain Injury do full justice to comprehensive batteries and to specific
(COMBI) website (http://www.tbims.org/combi), making it domain-specific measures. Recent reviews, however, are avail-
an attractive and comprehensive functional battery. able that discuss this.

Functional Independence Measure Neuropsychological Screens


The Functional Independence Measure (FIM) is used in inpa- Neuropsychological screens are objective measures adminis-
tient rehabilitation settings and even in residential brain injury tered directly to testees to determine cognitive status, and to
programs to assess disability rather than impairment.78 It is a assess whether changes in functioning have occurred after
comprehensive measure of essential daily functions, and is neurologic injury or illness. Unlike functional outcome scales
widely used across rehabilitation programs. The FIM is in which observers typically rate patient levels, neuropsycho-
reviewed in this chapter because it is frequently used with logical tests are administered in face-to-face sessions to testees
post–critical care patients who have progressed from the ICU who actively participate in answering specific test questions.
to an acute rehabilitation setting and whose recovery is Several well-known screens are reviewed here, with limitations
advanced enough to warrant this level of detailed assessment. listed for each measure.
This instrument measures the person’s performance along
several behavioral dimensions, from total dependence to inde-
pendence. The scale provides classification of individuals by Mini-Mental Status Examination
their ability to carry out an activity either with some level of The Mini-Mental Status Examination (MMSE) is one of the
assistance or supervision, or independently. Each dimension is earliest screens used to measure cognitive functioning.81,82 It
Section II—Clinical and Laboratory Assessment 111

is a brief but relatively comprehensive test of cognitive abili- emphasis on attention and executive functions than the
ties, and is directly administered to the patient. As such, the MMSE, and contains a more difficult memory task, requiring
patient must have a reasonable degree of orientation and lan- the recitation and recall of five instead of three words. In
guage ability to participate. Otherwise, a rating scale such as comparing this measure to the MMSE, the MOCA has been
one of those reviewed earlier in this chapter, must be used to found to be significantly more sensitive. In the original study
determine functional level. It is widely used to detect dementia highlighting the development of the MOCA as an alternative
and even delirium, although there is caution in the literature to the MMSE, the MOCA had a sensitivity of 100% in detect-
about using the MMSE alone to yield a diagnosis for either of ing mild Alzheimer’s disease, whereas the MMSE had a 78%
these conditions.83 sensitivity.86 The difference in detecting Mild Cognitive
The cognitive skills measured are (1) time and place orien- Impairment was more dramatic, with a sensitivity of 90% for
tation, (2) registration (i.e., immediate auditory recall), the MOCA but 18% for the MMSE. This is expected, because
(3) attention and calculation, (4) recall, and (5) language one of the main reasons for the development of the MOCA
(receptive and expressive). A total of 30 points is the maximum was to create a measure that could detect MCI much more
score, and cutoff levels of 23 and higher have been proposed effectively than the MMSE.
to differentiate dementia or cognitive impairment from Limitations: (1) The MOCA measures a fairly comprehen-
normal functioning. It has been the screen of choice among sive set of cognitive domains, especially for a brief screen, but
health care professionals in medical settings, especially in does not sample visual perception, an important function that
assessing hospitalized patients with known or suspected neu- can be disrupted with TBI, brain tumor, stroke, or other neu-
rologic disorders. It is among the most widely used tests of rologic illness or injury. (2) Although both auditory and visual
adult mental status. A comprehensive review regarding the attention skills are evaluated, only auditory memory is
psychometric properties of the MMSE was done.84 Subjects assessed. It would be a more complete test if visual memory
ranging from cognitively intact community dwellers to indi- also were measured, especially because other visual functions
viduals with dementia were included in the studies reviewed. have been included (visual attention, visual executive func-
Satisfactory reliability and construct validity were found, and tion, and visual motor construction).
the test was shown to be sensitive for moderate to severe cog-
nitive impairment but had lower sensitivity for mild degrees
of impairment. Content analyses revealed the test to be highly Cognistat
verbal, and not all items were equally sensitive to cognitive The Cognistat is the short term for the Neurobehavioral Cog-
impairment. MMSE scores were affected by age, education, nitive Status Examination, a screening test that is more exten-
and one’s cultural background, but not gender. Age and edu- sive in scope than the MMSE or MOCA, and is used by
cational norms were included in one study, with suggestions neuropsychologists as preliminary assessment of known or
for using different cutoff levels.85 suspected neurologic disorders. In fact, it is the number one
Limitations: (1) The MMSE covers several but not all neu- choice for cognitive screening among U.S. and Canadian neu-
ropsychological domains. Some areas that are missing include ropsychologists.88 It is often used as a starting point, so that
visual perception, visual attention, and several subcompo- additional neuropsychological measures can be selected to
nents of executive functioning, including verbal fluency and further assess specific cognitive functions. It assesses a repre-
mental flexibility. (2) The MMSE can easily miss neurologi- sentative sampling but not complete array of cognitive func-
cally based impairments in higher-functioning individuals. tions, including level of consciousness, orientation, language,
Therefore false-negatives may result. (3) Only one test of memory, constructional ability, calculations, and reasoning.89
executive functioning or motor ability is included, namely, the Language is tested through four subtests: Speech Sample, in
design construction. Verbal and other nonverbal executive which the test taker verbally describes a detailed picture with
abilities are often disrupted with neurologic damage, and people and actions; Comprehension; Repetition; and Naming.
assessment of these areas is advisable, even with a relatively Reasoning has two subtests: Similarities and Judgment. The
brief screen. (4) There is an overemphasis on language: Other Cognistat takes longer than the MMSE and MOCA screens,
areas are disrupted as much as or even more than language typically 10 to 20 minutes to administer. It has a unique feature
after a neurologic event, and a battery that measures other that actually helps to quickly determine normal functioning;
areas more extensively would have had more utility. (5) Poor Each test has a screen item that is more difficult than the test
sensitivity has been found for the detection of mild cognitive items that follow, which are called the metric section. If an
impairment (MCI),84 which is a transition stage between individual successfully answers the difficult screen item, the
normal aging and the associated cognitive decreases that may metric items under that domain are skipped. If only screen
occur, and the more serious cognitive difficulties as a result of items are administered, the test actually is fast and efficient,
Alzheimer’s disease. and may take only 5 minutes to complete. Another useful
feature is that the scores are plotted on the profile sheet, so
that average and impaired scores are clearly illustrated. It has
Montreal Cognitive Assessment been translated and is available in nine languages, including
An alternative to the MMSE is a more recently developed Spanish, Japanese, and Hebrew.
cognitive test called the Montreal Cognitive Assessment Limitations: (1) Standardization data in the manual are
(MOCA), which takes about 10 minutes to administer and has limited, because only 60 subjects are used, divided into two
a total of 30 points.86 The cutoff for the MOCA is typically 26 age groups of 30 each (ages 20 to 30 and 40 to 66). For a
out of 30 points, i.e., scores below 26 are considered impaired. screen so extensively used and translated, a larger normative
It is gaining popularity, and has been used as a screen for more group would have been expected. (2) As with other screens, a
extensive neuropsychological testing.87 The MOCA has more select but not comprehensive battery of cognitive domains is
112 Section II—Clinical and Laboratory Assessment

provided. (3) There is a somewhat uneven emphasis on The MOANS database has one limitation: The database
certain but not other cognitive areas, particularly too many consists of predominantly Caucasian adults with an education
tests of language and no tests of executive functions, an area level higher than the average (M = 13.1 years).92 This implies
often affected by neurologic insult. (4) The utility of a sen- that scores for non-Caucasians and for persons with less than
tence repetition test in a general neuropsychological screen is 8 years of education must be interpreted with caution, espe-
questionable; for example, aphasic patients would be better cially if they fall in the dementia range.
tested using a language battery, such as the Boston Diagnostic
Aphasia Examination.90
Conclusion
Outcome in critical care often has been assessed by mortality.
Dementia Rating Scale-2 However, the efficacy of an ICU treatment can no longer be
The original Dementia Rating Scale was developed in 1988, judged simply by survival. After patients are discharged from
and further refined into a second edition in 2001. The same a neurocritical care unit, functional outcome is important to
36 tasks and 32 stimulus cards have been retained for the determine success of treatment and rate of recovery. Func-
Dementia Rating Scale-2, together with the 5 subscales that tional outcome includes many measures such as (1) physical
measure specific cognitive abilities vulnerable to deterioration abilities, (2) quality of life, (3) activities of daily living, (4)
in aging dementia patients: Attention (8 items), Initiation/ return to work, (5) cognitive function, (6) emotional or psy-
Perseveration (11), Construction (6), Conceptualization (6), chological status, and (7) social integration to name a few.
and Memory (5).91 A total score and the 5 subscale scores are Many different methods assess functional outcome, some of
generated, which assists the test evaluator to decide whether which are described in this chapter. These may be assessed by
additional testing is necessary. For example, a university pro- direct interview by a rater, by telephone, completion of a
fessor who scores considerably above the cutoff for his age questionnaire (self-report) using paper and pencil or online
group but who has significant problems with Memory and computerized systems, by caregiver reports, or through
Conceptualization probably should be referred for more com- observation. What is clear is that no single ideal way to assess
prehensive testing. The upgraded aspects of the Dementia outcome and variables such as age, sex, education, ethnicity,
Rating Scale-2 are expanded normative data, greater ease of response bias, test learning, and even the test itself can affect
administration and scoring based on user-friendly test forms, outcome. Outcome measures therefore should be carefully
and additional validity information in the manual’s literature selected based on the disease process and expectations.
review. The norms for the Dementia Rating Scale-2 are based Neuropsychological testing is a necessary and very effective
on a sample of healthy, nondementing adults, ages 56 to 105 method to assess cognition and functioning following the
from the Mayo Clinic’s impressive and comprehensive Mayo’s initial use of outcome scales.
Older Americans Normative Studies (MOANS) database,92
which also has been used for other testing instruments specific
to elderly populations.93 The authors of the Dementia Rating References
Scale-2 caution the user to rely more on the subscale scores 1. Hopkins RO, Jackson JC. Long-term neurocognitive function after critical
than the total score to determine meaningful cognitive decline, illness. Chest 2006;130(3):869–78.
2. Wilde EA, Whiteneck GG, Bogner J, et al. Recommendations for the use of
because an individual may score above the total score cutoff common outcome measures in traumatic brain injury research. Arch Phys
for his or her age range but demonstrate significant cognitive Med Rehabil 2010;91(11):1650–60.e17.
impairments in two or more subscale areas, for example. With 3. Bagiella E, Novack TA, Ansel B, et al. Measuring outcome in traumatic
the original Dementia Rating Scale, there was a single cut-off brain injury treatment trials: recommendations from the traumatic
score of 123 out of a total of 144, and the common practice brain injury clinical trials network. J Head Trauma Rehabil 2010;25(5):
375–82.
was to rate an individual as having a probable dementia dis- 4. McCauley SR, Wilde EA, Anderson VA, et al. Recommendations for the use
order if he/she scored below this total. However, researchers of common outcome measures in pediatric traumatic brain injury research.
in geriatric assessment have questioned this practice. One J Neurotrauma 2011; Aug 24. Epub ahead of print.
study argued that different cutoff scores should be used for 5. Fields JA, Ferman TJ, Boeve BF, et al. Neuropsychological assessment of
patients with dementing illness. Nat Rev Neurol 2011; Nov 1. Epub ahead of
different age groups.94 The Mayo Clinic research group found print.
that 16% of adults older than 85 years of age in the MOANS 6. Lewis MW, Babbage DR, Leathem JM. Assessing executive performance
normative data scored below the cutoff of 123 even though during cognitive rehabilitation. Neuropsychol Rehabil 2011;21(2):145–63.
they were not considered a dementia sample.92 Epub 2011 Jan 10.
The Dementia Rating Scale-2 also can be used with geriatric 7. American Congress of Rehabilitation Medicine, Brain Injury-
Interdisciplinary Special Interest Group, Disorders of Consciousness Task
patients who have suspected neuropsychological impairments Force, et al. Assessment scales for disorders of consciousness: evidence-based
that are not necessarily dementia, including head injury, or are recommendations for clinical practice and research. Arch Phys Med Rehabil
community dwellers with cognitive problems who have not 2010;91(12):1795–813.
yet been diagnosed with dementia.95 Like the Cognistat, test 8. Podell K, Gifford K, Bougakov D, et al. Neuropsychological assessment in
traumatic brain injury. Psychiatr Clin North Am 2010;33(4):855–76.
items can be eliminated based on successful performance of 9. Bilder RM. Neuropsychology 3.0: evidence-based science and practice. J Int
previous items, making it a time-efficient test to administer, Neuropsychol Soc 2011;17(1):7–13. Epub 2010 Nov 19.
especially for the geriatric population who may have limited 10. Gottesman RF, Hillis AE. Predictors and assessment of cognitive dysfunction
tolerance for testing. For example, a client may have no dif- resulting from ischaemic stroke. Lancet Neurol 2010;9(9):895–905.
ficulty in Initiation/Perseveration, but problems with Atten- 11. Holtslag HR, Post MW, Lindeman E, et al. Long-term functional health
status of severely injured patients. Injury 2007;38(3):280–9.
tion and Memory, requiring all of these latter two subscale 12. Van der Schaaf M, Beelen A, Dongelmans DA, et al. Functional status after
items to be administered but only a limited number of items intensive care: a challenge for rehabilitation professionals to improve
for the former. outcome. J Rehabil Med 2009;41(5):360–6.
Section II—Clinical and Laboratory Assessment 113

13. Frontera JA, Fernandez A, Schmidt JM, et al. Defining vasospasm after 32. Rappaport M, Hall KM, Hopkins K, et al. Disability rating scale for severe
subarachnoid hemorrhage: what is the most clinically relevant definition? head trauma: coma to community. Arch Phys Med Rehabil 1982;63:118–23.
Stroke 2009;40(6):1963–8. 33. Hall K, Cope N, Rappaport M. Glasgow outcome scale and disability rating
14. Mikkelsen ME, Shull WH, Biester RC, et al. Cognitive, mood and quality of scale: comparative usefulness in following recovery in traumatic head injury.
life impairments in a select population of ARDS survivors. Respirology 2009; Arch Phys Med Rehabil 1985;66:35–7.
14(1):76–82. 34. Gouvier WD, Blanton PD, LaPorte KK, et al. Reliability and validity of the
15. Van Baalen B, Odding E, van Woensel MP, et al. Reliability and sensitivity to disability rating scale and the levels of cognitive functioning scale in
change of measurement instruments used in a traumatic brain injury monitoring recovery from severe head injury. Arch Phys Med Rehabil
population. Clin Rehabil 2006;20(8):686–700. 1987;68(2):94–7.
16. de Haan R, Limburg M, Bossuyt P, et al. The clinical meaning of Rankin 35. Neese LE, Caroselli JS, Klaas R, et al. Neuropsychological assessment and the
“handicap” grades after stroke. Stroke 1995;26:2027–30. disability rating scale (DRS): a concurrent validity study. Brain Inj
17. Lipsett PA, Swoboda SM, Dickerson J, et al. Survival and functional outcome 2000;14(8):719–24.
after prolonged intensive care unit stay. Ann Surg 2000;231(2):262–8. 36. Rankin J. Cerebral vascular accidents in patients over the age of 60:
18. Boake C, High WM. Functional outcome from traumatic brain injury. Am J prognosis. Scott Med J 1957;2(5):200–15.
Phys Med Rehabil 1996;75(2):105–13. 37. Van Swieten J, Koudstaal P, Visser M, et al. Interobserver agreement for the
19. von Elm E, Osterwalder JJ, Graber C, et al. Severe traumatic brain injury in assessment of handicap in stroke patients. Stroke 1988;19:604–7.
Switzerland—feasibility and first results of a cohort study. Swiss Med Wkly 38. Shinohara Y, Minematsu K, Amano T, et al. Modified Rankin scale with
2008;138(23-24):327–34. expanded guidance scheme and interview questionnaire: interrater
20. Lippert-Gruner M, Maegele M, Haverkamp H, et al. Health-related quality agreement and reproducibility of assessment. Cerebrovasc Dis 2006;21:
of life during the first year after severe brain trauma with and without 271–8.
polytrauma. Brain Inj 2007;21(5):451–5. 39. Quinn TJ, Lees KR, Hardemark HG, et al. Initial experience of a digital
21. Broessner G, Helbok R, Lackner P, et al. Survival and long-term functional training resource for modified Rankin scale assessment in clinical trials.
outcome in 1155 consecutive neurocritical care patients. Crit Care Med Stroke 2007;38:2257–61.
2007;35(9):2025–30. 40. Gresham GE, Phillips TF, Labi ML. ADL status in stroke: relative merit s of
22. Sander A. The Extended Glasgow Outcome Scale. The Center for Outcome these standard indexes. Arch Phys Med Rehabil 1980;61(8):355–8.
Measurement in Brain Injury, 2002, http://www.tbims.org/combi/gose 41. Koch S, Romano JG, Forteza AM, et al. Rapid blood pressure reduction in
(accessed September 24, 2012). acute intracerebral hemorrhage: feasibility and safety. Neurocrit Care
23. Collie A, Maruff P, Makdissi M, et al. CogSport: reliability and correlation 2008;8:316–21.
with conventional cognitive tests used in postconcussion medical 42. Naval NS, Abdelhak TA, Zeballos P, et al. Prior statin use reduces mortality
evaluations. Clin J Sport Med 2003;13:28–32. in intracerebral hemorrhage. Neurocrit Care 2008;8:6–12.
24. McCrory P, Meeuwisse W, Johnston K, et al. Consensus statement on 43. Kwiatkowski TG, Libman RB, Frankel M, et al. Effects of tissue plasminogen
concussion in sport: the 3rd International Conference on Concussion in activator for acute ischemic stroke at one year. N Engl J Med
Sport, Zurich, November 2008. Br J Sports Med 2009;43(Suppl 1):i76–i90. 1999;40:1781–7.
25. Iverson GL, Lovell MR, Collins MW. Validity of ImPACT for measuring 44. Albers GW, Bates VE, Clark WM, et al. Intravenous tissue-type plasminogen
processing speed following sports-related concussion. J Clin Exp activator for treatment of acute stroke. JAMA 2000;283(9):1145–50.
Neuropsychol 2005;27(6):683–9. 45. Stead LG, Gilmore RM, Bellolio MF, et al. Hyperglycemia as an independent
26. Mayer SA, Brun NC, Begtrup K, et al. Recombinant activated factor VII for predictor of worse outcome in non-diabetic patients presenting with acute
acute intracerebral hemorrhage. N Engl J Med 2005;352:777–85. ischemic stroke. Neurocrit Care 2009;10(2):181–6.
27. Ponsford J, Draper K, Schonberger M. Functional outcome 10 years after 46. Smith WS, Tsao JW, Billings ME, et al. Prognostic significance of
traumatic brain injury: its relationship with demographic, injury severity, angiographically confirmed large vessel intracranial occlusion in patients
and cognitive and emotional status. J Int Neuropsychol Soc presenting with acute brain ischemia. Neurocrit Care 2006;4(1):14–7.
2008;14(2):233–42. 47. Murthy JM, Chowdary GV, Murthy TV, et al. Decompressive craniectomy
28. Kirkness CJ, Burr RL, Mitchell PH. Intracranial and blood pressure with clot evacuation in large hemispheric hypertensive intracerebral
variability and long-term outcome after aneurysmal sub-arachnoid hemorrhage. Neurocrit Care 2005;2(3):258–62.
hemorrhage. Am J Crit Care 2009;8(3):241–51. 48. Schmidt JM, Rincon F, Fernandez A, et al. Cerebral infarction associated
29. Poon WS, Ng SC, Wong GK, et al. Chronic hydrocephalus that requires with acute subarachnoid hemorrhage. Neurocrit Care 2007;7(1):10–7.
shunting in aneurysmal subarachnoid haemorrhage: its impact on clinical 49. Shah S, Vanclay F, Cooper B. Improving the sensitivity of the Barthel index
outcome. Acta Neurochir Suppl 2008;102:129–30. for stroke rehabilitation. J Clin Epidemiol 1989;42(8):703–9.
30. World Health Organization (WHO). International Classification of 50. Loewen SC, Anderson BA. Predictors of stroke outcome using objective
Impairments, Disabilities, and Handicaps: a manual of classification relating measurement scales. Stroke 1990;21:78–81.
to the consequences of disease. Geneva, 1980.
31. Teasdale G, Jeanette B. Assessment of coma and impaired consciousness: A complete list of references for this chapter can be found online at
practical scale. Lancet 1974;2:81–4. www.expertconsult.com.
Section II—Clinical and Laboratory Assessment 113.e1

References 28. Kirkness CJ, Burr RL, Mitchell PH. Intracranial and blood pressure
variability and long-term outcome after aneurysmal sub-arachnoid
1. Hopkins RO, Jackson JC. Long-term neurocognitive function after critical hemorrhage. Am J Crit Care 2009;8(3):241–51.
illness. Chest 2006;130(3):869–78. 29. Poon WS, Ng SC, Wong GK, et al. Chronic hydrocephalus that requires
2. Wilde EA, Whiteneck GG, Bogner J, et al. Recommendations for the use of shunting in aneurysmal subarachnoid haemorrhage: its impact on clinical
common outcome measures in traumatic brain injury research. Arch Phys outcome. Acta Neurochir Suppl 2008;102:129–30.
Med Rehabil 2010;91(11):1650–60.e17. 30. World Health Organization (WHO). International Classification of
3. Bagiella E, Novack TA, Ansel B, et al. Measuring outcome in traumatic brain Impairments, Disabilities, and Handicaps: a manual of classification relating
injury treatment trials: recommendations from the traumatic brain injury to the consequences of disease. Geneva, 1980.
clinical trials network. J Head Trauma Rehabil 2010;25(5):375–82. 31. Teasdale G, Jeanette B. Assessment of coma and impaired consciousness:
4. McCauley SR, Wilde EA, Anderson VA, et al. Recommendations for the use practical scale. Lancet 1974;2:81–4.
of common outcome measures in pediatric traumatic brain injury research. 32. Rappaport M, Hall KM, Hopkins K, et al. Disability rating scale for severe
J Neurotrauma 2011; Aug 24. Epub ahead of print. head trauma: coma to community. Arch Phys Med Rehabil 1982;63:118–23.
5. Fields JA, Ferman TJ, Boeve BF, et al. Neuropsychological assessment of 33. Hall K, Cope N, Rappaport M. Glasgow outcome scale and disability rating
patients with dementing illness. Nat Rev Neurol 2011; Nov 1. Epub ahead of scale: comparative usefulness in following recovery in traumatic head injury.
print. Arch Phys Med Rehabil 1985;66:35–7.
6. Lewis MW, Babbage DR, Leathem JM. Assessing executive performance 34. Gouvier WD, Blanton PD, LaPorte KK, et al. Reliability and validity of the
during cognitive rehabilitation. Neuropsychol Rehabil 2011;21(2):145–63. disability rating scale and the levels of cognitive functioning scale in
Epub 2011 Jan 10. monitoring recovery from severe head injury. Arch Phys Med Rehabil
7. American Congress of Rehabilitation Medicine, Brain Injury- 1987;68(2):94–7.
Interdisciplinary Special Interest Group, Disorders of Consciousness Task 35. Neese LE, Caroselli JS, Klaas R, et al. Neuropsychological assessment and the
Force, et al. Assessment scales for disorders of consciousness: evidence-based disability rating scale (DRS): a concurrent validity study. Brain Inj
recommendations for clinical practice and research. Arch Phys Med Rehabil 2000;14(8):719–24.
2010;91(12):1795–813. 36. Rankin J. Cerebral vascular accidents in patients over the age of 60:
8. Podell K, Gifford K, Bougakov D, et al. Neuropsychological assessment in prognosis. Scott Med J 1957;2(5):200–15.
traumatic brain injury. Psychiatr Clin North Am 2010;33(4):855–76. 37. Van Swieten J, Koudstaal P, Visser M, et al. Interobserver agreement for the
9. Bilder RM. Neuropsychology 3.0: evidence-based science and practice. J Int assessment of handicap in stroke patients. Stroke 1988;19:604–7.
Neuropsychol Soc 2011;17(1):7–13. Epub 2010 Nov 19. 38. Shinohara Y, Minematsu K, Amano T, et al. Modified Rankin scale with
10. Gottesman RF, Hillis AE. Predictors and assessment of cognitive dysfunction expanded guidance scheme and interview questionnaire: interrater
resulting from ischaemic stroke. Lancet Neurol 2010;9(9):895–905. agreement and reproducibility of assessment. Cerebrovasc Dis
11. Holtslag HR, Post MW, Lindeman E, et al. Long-term functional health 2006;21:271–8.
status of severely injured patients. Injury 2007;38(3):280–9. 39. Quinn TJ, Lees KR, Hardemark HG, et al. Initial experience of a digital
12. Van der Schaaf M, Beelen A, Dongelmans DA, et al. Functional status after training resource for modified Rankin scale assessment in clinical trials.
intensive care: a challenge for rehabilitation professionals to improve Stroke 2007;38:2257–61.
outcome. J Rehabil Med 2009;41(5):360–6. 40. Gresham GE, Phillips TF, Labi ML. ADL status in stroke: relative merit s of
13. Frontera JA, Fernandez A, Schmidt JM, et al. Defining vasospasm after these standard indexes. Arch Phys Med Rehabil 1980;61(8):355–8.
subarachnoid hemorrhage: what is the most clinically relevant definition? 41. Koch S, Romano JG, Forteza AM, et al. Rapid blood pressure reduction in
Stroke 2009;40(6):1963–8. acute intracerebral hemorrhage: feasibility and safety. Neurocrit Care
14. Mikkelsen ME, Shull WH, Biester RC, et al. Cognitive, mood and quality of 2008;8:316–21.
life impairments in a select population of ARDS survivors. Respirology 42. Naval NS, Abdelhak TA, Zeballos P, et al. Prior statin use reduces mortality
2009;14(1):76–82. in intracerebral hemorrhage. Neurocrit Care 2008;8:6–12.
15. Van Baalen B, Odding E, van Woensel MP, et al. Reliability and sensitivity to 43. Kwiatkowski TG, Libman RB, Frankel M, et al. Effects of tissue plasminogen
change of measurement instruments used in a traumatic brain injury activator for acute ischemic stroke at one year. N Engl J Med
population. Clin Rehabil 2006;20(8):686–700. 1999;40:1781–7.
16. de Haan R, Limburg M, Bossuyt P, et al. The clinical meaning of Rankin 44. Albers GW, Bates VE, Clark WM, et al. Intravenous tissue-type plasminogen
“handicap” grades after stroke. Stroke 1995;26:2027–30. activator for treatment of acute stroke. JAMA 2000;283(9):1145–50.
17. Lipsett PA, Swoboda SM, Dickerson J, et al. Survival and functional outcome 45. Stead LG, Gilmore RM, Bellolio MF, et al. Hyperglycemia as an independent
after prolonged intensive care unit stay. Ann Surg 2000;231(2):262–8. predictor of worse outcome in non-diabetic patients presenting with acute
18. Boake C, High WM. Functional outcome from traumatic brain injury. Am J ischemic stroke. Neurocrit Care 2009;10(2):181–6.
Phys Med Rehabil 1996;75(2):105–13. 46. Smith WS, Tsao JW, Billings ME, et al. Prognostic significance of
19. von Elm E, Osterwalder JJ, Graber C, et al. Severe traumatic brain injury in angiographically confirmed large vessel intracranial occlusion in patients
Switzerland—feasibility and first results of a cohort study. Swiss Med Wkly presenting with acute brain ischemia. Neurocrit Care 2006;4(1):14–7.
2008;138(23-24):327–34. 47. Murthy JM, Chowdary GV, Murthy TV, et al. Decompressive craniectomy
20. Lippert-Gruner M, Maegele M, Haverkamp H, et al. Health-related quality with clot evacuation in large hemispheric hypertensive intracerebral
of life during the first year after severe brain trauma with and without hemorrhage. Neurocrit Care 2005;2(3):258–62.
polytrauma. Brain Inj 2007;21(5):451–5. 48. Schmidt JM, Rincon F, Fernandez A, et al. Cerebral infarction associated
21. Broessner G, Helbok R, Lackner P, et al. Survival and long-term functional with acute subarachnoid hemorrhage. Neurocrit Care 2007;7(1):10–7.
outcome in 1155 consecutive neurocritical care patients. Crit Care Med 49. Shah S, Vanclay F, Cooper B. Improving the sensitivity of the Barthel index
2007;35(9):2025–30. for stroke rehabilitation. J Clin Epidemiol 1989;42(8):703–9.
22. Sander A. The Extended Glasgow Outcome Scale. The Center for Outcome 50. Loewen SC, Anderson BA. Predictors of stroke outcome using objective
Measurement in Brain Injury, 2002, http://www.tbims.org/combi/gose measurement scales. Stroke 1990;21:78–81.
(accessed September 24, 2012). 51. Granger CV, Dewis LS, Peters NC, et al. Stroke rehabilitation: analysis of
23. Collie A, Maruff P, Makdissi M, et al. CogSport: reliability and correlation repeated Barthel index measures. Arch Phys Med Rehabil 1979;60(1):14–7.
with conventional cognitive tests used in postconcussion medical 52. Morgalla MH, Will BE, Roser F, et al. Do long-term results justify
evaluations. Clin J Sport Med 2003;13:28–32. decompressive craniectomy after severe traumatic brain injury? J Neurosurg
24. McCrory P, Meeuwisse W, Johnston K, et al. Consensus statement on 2008;109:685–90.
concussion in sport: the 3rd International Conference on Concussion in 53. Powell J, Heslin J, Greenwood R. Community based rehabilitation after
Sport, Zurich, November 2008. Br J Sports Med 2009;43(Suppl 1):i76–i90. severe traumatic brain injury: a randomized controlled trial. J Neurol
25. Iverson GL, Lovell MR, Collins MW. Validity of ImPACT for measuring Neurosurg Psychiatry 2002;72:193–202.
processing speed following sports-related concussion. J Clin Exp 54. Yarkony GM, Roth, EJ, Heinemann AW, et al. Benefits of rehabilitation for
Neuropsychol 2005;27(6):683–9. traumatic spinal cord injury. Arch Neurol 1987;44(1):93–6.
26. Mayer SA, Brun NC, Begtrup K, et al. Recombinant activated factor VII for 55. Altman DG, Royston P: The cost of dichotomising continuous variables.
acute intracerebral hemorrhage. N Engl J Med 2005;352:777–85. BMJ 2006;332:1080.
27. Ponsford J, Draper K, Schonberger M. Functional outcome 10 years after 56. Maas AI, Steyerberg EW, Marmarou A, et al. IMPACT recommendations for
traumatic brain injury: its relationship with demographic, injury severity, and improving the design and analysis of clinical trials in moderate to severe
cognitive and emotional status. J Int Neuropsychol Soc 2008;14(2):233–42. traumatic brain injury. Neurotherapeutics 2010;7:127–34.
113.e2 Section II—Clinical and Laboratory Assessment

57. McHugh GS, Butcher I, Steyerberg EW, et al. A simulation study evaluating 76. Malec J. The Mayo Portland Adaptability Inventory. The Center for Outcome
approaches to the analysis of ordinal outcome data in randomized controlled Measurement in Brain Injury, 2005, http://www.tbims.org/combi/mpai
trials in traumatic brain injury: results from the IMPACT project. Clin Trials (accessed September 24, 2012).
2010;7:44–57. 77. Malec J. Comparability of Mayo-Portland adaptability inventory ratings by
58. Murray GD, Barer D, Choi S, et al. Design and analysis of phase III trials staff, significant others and people with acquired brain injury. Brain Inj
with ordered outcome scales: the concept of the sliding dichotomy. 2004;18(6):563–75.
J Neurotrauma 2005;22:511–17. 78. Bezner JR, Hunter DL. Wellness perception in persons with traumatic brain
59. Roozenbeek B, Lingsma HF, Perel P, et al. IMPACT (International Mission injury and its relation to functional independence. Arch Phys Med Rehabil
on Prognosis and Clinical Trial Design in Traumatic Brain Injury) study 2001;82(6):787–92.
group; CRASH (corticosteroid randomisation after significant head injury) 79. Wright J. The functional assessment measure. The Center for Outcome
trial collaborators. The added value of ordinal analysis in clinical trials: an Measurement in Brain Injury, 2000, http://www.tbims.org/combi/FAM
example in traumatic brain injury. Crit Care 2011;15(3):R127. (accessed September 24, 2012).
60. Optimising Analysis of Stroke Trials (OAST) Collaboration, Bath PM, Gray 80. Jackson JC, Obremskey W, Bauer R, et al. Long-term cognitive, emotional,
LJ, et al. Can we improve the statistical analysis of stroke trials? Statistical and functional outcomes in trauma intensive care unit survivors without
reanalysis of functional outcomes in stroke trials. Stroke 2007;38:1911–15. intracranial hemorrhage. J Trauma Inj Infect Crit Care 2007;62(1):80–8.
61. Findler M, Cantor J, Haddad L, et al. The reliability and validity of the SF-36 81. Folstein MF, Folstein SE, McHugh PR. Mini-mental state: a practical method
health survey questionnaire for use with individuals with traumatic brain for grading the state of patients for the clinician. J Psychiatr Res
injury. Brain Inj 2001;15(8):715–23. 1975;12:189–98.
62. MacKenzie EJ, McCarthy ML, Ditunno JF, et al. Using the SF-36 for 82. Cockrell JR, Folstein MF. Mini mental state examination (MMSE).
characterizing outcome after multiple trauma involving head injury. Psychopharmacology 1988;24(4):689–92.
J Trauma 2002;52(3):527–34. 83. Anthony JC, LeResche L, Niaz U, et al. Limits of the ‘mini-mental state’ as a
63. Timofeev I, Kirkpatrick PJ, Corteen E, et al. Decompressive craniectomy in screening test for dementia and delirium among hospital patients. Psychol
traumatic brain injury: outcome following protocol-driven therapy. Acta Med 1982;12(2):397–408.
Neurochir Suppl 2006;96:11–6. 84. Tombaugh TN, McIntyre NJ. The mini-mental state examination: a
64. Neugebauer E, Bouillon B, Bullinger M, et al. Quality of life after multiple comprehensive review. J Am Geriatr Soc 1992;40(9):922–35.
trauma: summary and recommendations of the consensus conference. Rest 85. Crum RM, Anthony JC, Bassett SS, et al. Population-based norms for the
Neurol Neurosci 2002;20: 161–7. mini-mental state examination by age and educational level. JAMA
65. McNaughton HK, Weatherall M, McPherson KM. Functional measures 1993;18:2386–91.
across neurologic disease states: analysis of factors in common. Arch Phys 86. Nasreddine ZS, Phillips NA, Bedirian V, et al. The Montreal cognitive
Med Rehabil 2005;86(11):2184–8. assessment, MOCA: a brief screening tool for mild cognitive impairment.
66. Nestvold K, Stavem K. Determinants of health-related quality of life 22 years J Am Geriatr Soc 2005;53(4):695–9.
after hospitalization for traumatic brain injury. Brain Inj 2009;23(1):15–21. 87. Ruff RL, Ruff SS, Wang X-F. Headaches among operation Iraqi freedom/
67. Andelic N, Hammergren N, Bautz-Holter E, et al. Functional outcome and operation enduring freedom veterans with mild traumatic brain injury
health-related quality of life 10 years after moderate-to-severe traumatic associated with exposures to explosions. J Rehabil Res Dev
brain injury. Acta Neurol Scand 2008;120(1):16–23. 2008;45(7):941–52.
68. Jackson JC, Hart RP, Gordon SM, et al. Six-month neuropsychological 88. Rabin LA, Barr WB, Burton LA. Assessment practices of clinical
outcome of medical intensive care unit patients. Crit Care Med neuropsychologists in the United States and Canada: a survey of INS, NAN
2003:31(4):1226–34. and APA division 40 members. Arch Clin Neuropsychol 2005;20:33–65.
69. Cote I, Gregoire JP, Moisan J, et al. Quality of life in hypertension: the SF-12 89. Mueller J, Kiernan R, Langston JW. Cognistat manual. Fairfax, CA:
compared to the SF-36. Can J Clin Pharmacol 2004;11(2):232–8. Cognistat, Inc.; 2009.
70. Kiely JM, Grasel KJ, Guse CE, et al. Correlation of SF-12 and SF-36 in a 90. Goodglass H, Kaplan E, Barresi B. Boston diagnostic aphasia examination.
trauma population. J Surg Res 2006;132(2):214–18. 3rd ed. Lutz, FL: Psychological Assessment Resources; 2000.
71. Riddle DL, Lee KT, Stratford PW. Use of SF-36 and SF-12 health status 91. Jurica PJ, Leitten CL, Mattis S. DRS-2: Dementia rating scale-2 professional
measures: a quantitative comparison for groups versus individual patients. manual. Lutz, FL: Psychological Assessment Resources, Inc; 2001.
Med Care 2001;39(8):867–78. 92. Lucas JA, Ivnik RJ, Smith GE, et al. Normative date for the Mattis demential
72. Wee CC, Davis RB, Hamel MB. Comparing the SF-12 and SF-36 health rating scale. J Clin Exper Neuropsychol 1998;20(40):536–47.
status questionnaires in patients with and without obesity. Health Qual Life 93. Ivnik RJ, Malec JF, Smith GE, et al. Mayo’s older Americans normative
Out 2008;30(6):6–11. studies: WAIS-R, WMS-R, AND AVLT norms for ages 56 through 97. Clin
73. Nanda U, McLendon PM, Andresen EM, et al. The SIP68: an abbreviated Neuropsychol 1992;6(Suppl.):1–104.
sickness impact profile for disability outcomes research. Qual Life Res 94. Bank AL, Yochim BP, MacNeill SE, et al. Expanded normative data for the
2003;12(5):583–95. Mattis dementia rating scale for use with urban, elderly medical patients.
74. Lezak M. Relationships between personality disorders, social disturbances, Clin Neuropsychol 2000;14:149–56.
and physical disability following traumatic brain injury. J Head Trauma 95. Schmidt KS, Mattis PJ, Adams J, et al. Alternate-form reliability of the
Rehabil 1987;2(1):57–69. dementia rating scale-2. Arch Clin Neuropsychol 2005;20(4):435–41.
75. Lezak M, O’Brien K. Longitudinal study of emotional, social, and physical
changes after traumatic brain injury. J Learn Disabil 1988;21(8):456–63.
II
Chapter
13  

Brain Death
Farzana Tariq and Peter M. Black

in the 40 years since the report, the fundamental criteria have


Introduction not changed markedly—lack of brainstem reflexes, lack of
Brain death is one of the most important diagnoses to be cortical activity as shown by electroencephalography (EEG)
made in the neurocritical care unit (NCCU). The increasing and clinical examination, and irreversibility.
ability to maintain the heart and other vital organs in the face The initial development of this concept was marked by two
of devastating brain injury has made it mandatory to recog- major refinements, both of considerable importance for
nize circumstances in which further intervention is futile intensive care worldwide. The first was acceptance of the
because of widespread irreversible brain destruction. Imple- concept of brain death. In most Western cultures, acceptance
menting criteria for brain death is important for: of this concept is complete; in Japan and other Eastern tradi-
tions, this concept is increasingly being accepted. In the United
1. Family and patient decision making
States, an important landmark was the drafting of a model act
2. Effective use of expensive technology and resources
in 1980 known as the Uniform Determination of Death Act,5
3. Potential organ donation to allow other patients a new life
which states:
Bioethicists, physicians, philosophers, religious leaders, and
An individual who has sustained either irreversible cessa-
those in the legal profession, among others, will continue to
tion of circulatory and respiratory functions or irreversible
discuss (and disagree) about how to best define human death.
cessation of all function of the entire brain including the
These theories can range from the Harvard Criteria of Brain
brainstem, is dead. The determination of brain death must
Death that defines death as the cessation of all brain activity,
be made in accordance with accepted medical standards.
through the Irreversibility Standard that defines death when
the capacity for consciousness is lost forever, to the Cognitive The adoption of this statute by many states helped establish
Standard that defines death as the loss of almost all core the concept that brain death was death. Uniform acceptance
mental properties (e.g., memory, self-consciousness, moral of the concept in case law in other states confirmed the idea
agency, and the capacity for reason).1 This chapter emphasizes that brain death equates to human death. The concept of brain
the practical determination of brain death in the NCCU in death became standard. Although there are two explicit defini-
adults and relevance to organ donation, and discusses the tions of death in this statute, cardiorespiratory function can be
philosophical concepts of brain death. End-of-life care, brain said to be important only to perfuse the brain, so death of the
death criteria, and the concept and understanding of brain brain becomes the major criterion for death in both definitions.
death vary in different countries.2 In this chapter, the focus is Since the adoption of the Uniform Determination of Death
on how brain death is diagnosed in the United States. Act, all court rulings in which brain death has been challenged
in the United States have upheld the medical practice of death
determination using neurologic criteria according to state law.
Historical Background Two major issues that arise in court rulings are consequences
Death was described for many centuries as cessation of circu- of documentation of the time of brain death and family-
lation and respiration. This definition was no longer valid physician discord on withdrawal of intensive care support.5
when modern medical devices, increasingly sophisticated The second area of increasing refinement has been the cri-
ICUs, and greater understanding of physiology in the face of teria used to make the determination of death. With time,
severe brain injury made it possible to maintain artificial res- increasing emphasis has been placed on blood flow determi-
piration and circulation indefinitely despite massive brain nation to establish this diagnosis, although the fundamental
destruction.3 This brought a need for more precise criteria to criteria of absent brainstem and cortical function and irrevers-
define brain death. ibility remain the core criteria.6
An early landmark was the publication of criteria for irre-
versible coma in 1968 by an ad hoc committee at Harvard
Medical School.4 This group described a condition known as
Brain Death Medical Criteria
irreversible coma that they equated with cerebral death or The American Academy of Neurology has provided evidence-
brain death. They created a set of practical steps that could be based practice parameters to determine brain death in
used to make this diagnosis. Although the field has changed adults.7,8 However, there still is variance in how this is done in
114 © Copyright 2013 Elsevier Inc. All rights reserved.
Section II—Clinical and Laboratory Assessment 115

different hospitals, states, and countries that can create poten- Determination of Brain Death
tial pitfalls in the process.9 Parameters that commonly vary
include prerequisites to declare brain death, qualification Clinical Examination
of physicians, acceptable body temperature when the exami- A careful well-documented clinical examination is central to
nation is performed, number of required examinations, need the diagnosis of brain death. The time and detailed set of tests
for a second physician to confirm the diagnosis, and docu- performed should be noted in the record; the apnea test needs
mentation. Despite these variances, the basic medical criteria to be done only once, usually at the end of the time period
for brain death include: used for the determination (6 hours in most cases). Prerequi-
sites to the examination include the following: (1) establish
 Coma or unresponsiveness (with a known cause)
the cause and irreversible nature of the coma, (2) exclude the
 Absence of purposeful motor responses in all extremities

use of any central nervous system depressant drugs or neuro-
Absence of brainstem reflexes including apnea

muscular blockade, (3) ensure normal temperature (>36° C)
Demonstrated irreversibility
and systolic blood pressure (>100 mm Hg, and (4) ensure
The details of the clinical testing are described later. It is laboratory values are close to normal.
important to know the cause of the patient’s coma and to
determine that the condition is irreversible. This means
excluding conditions that may mimic brain death, such as Coma
barbiturate intoxication, valproic acid intoxication, severe Brain dead patients are in a state of deep coma. The cause of
alcohol intoxication, sedative overdose, organophosphate poi- coma should be determined and should be irreversible. If the
soning, baclofen overdose, high cervical cord injury, or hypo- cause is not known, then longer observation is required.12
thermia. In rare instances, snake bites, Miller Fisher syndrome Based on the Glasgow Coma Scale (see Chapter 11), the
(cranial nerve palsy, ataxia, areflexia), fulminant Guillain- patient should have a score of 3; based on the FOUR score,13
Barré syndrome that includes both central and peripheral the patient should have a score of E(0)M(0)B(0)R(0).
nervous system involvement, and brainstem encephalitis also
may mimic brain death. It also means observing the patient
for long enough that it is clear that the condition is not going Motor Responses
to resolve—a minimum of 6 hours in most current proto- There should be no purposeful motor response to painful
cols.10,11 Finally, some protocols require confirmation of brain stimuli such as pressure on the supraorbital ridge or nail bed.
death by a neurologist or neurosurgeon. While there can be Pinching a patient on the chest is not an adequate way to elicit
potential mimics that neurointensivists should be aware of, these responses. Reflex motor responses that do not preclude
there are no published reports in adults of recovery of neuro- the diagnosis of brain death include the following: facial myo-
logic function after a brain death diagnosis using the criteria kymia (spontaneous twitching of the cheek); undulating toe
described in the 1995 American Academy of Neurology prac- movements, reflecting L5 and S1 activity; the Lazarus sign
tice parameter.8 In children, brain death may be more compli- (movements of upper limb from spinal cord integrated reflex
cated to diagnose. Guidelines add a component of continued activity); abdominal reflexes; flexion reflexes; and periodic leg
unresponsiveness for up to 48 hours. Table 13.1 presents these movements, especially if they are generated during apnea
criteria in summary form. testing, respiratory acidosis, or brisk neck flexion. It is impor-
tant to understand the origin of these movements because
they may pose concern to nurses, physicians, and family
members about whether there is whole brain destruction.
Table 13.1  Criteria for the Clinical These spinal cord reflexes occur more often in young adults
Determination of Brain Death than in older adults.14-18
Coma
Absence of reversible medical conditions: Brainstem Reflexes
Hypothermia <32°C
Drug overdose (barbiturate, benzodiazepines, narcotics)
The following tests are performed to evaluate brainstem
Electrolyte anomalies (metabolic acidosis) function.
Pupillary response: The pupils may be round, oval, or irregu-
Absent pupillary light reflex
larly shaped. They are usually midsize (4 to 6 mm) but may
Absent corneal reflex be dilated. Drugs and preexisting anatomic ocular abnormali-
Absent caloric response ties may be a confounding factor.19 The pupillary light reflex
Absent gag reflex (testing the second and third cranial nerves) must be absent.
Pilocarpine supersensitivity due to denervation following
Absent cough in response to tracheal suctioning
instillation of 0.06% of the drug also may be seen. In some
Absent sucking and rooting reflex instances spontaneous asynchronous pupillary constriction
Absent respiratory drive at PaCO2 that is 60 mm Hg or 20 mm and dilation may occur, which should not be a confounding
Hg above normal baseline values factor for brain death diagnosis.20,21
Interval between two evaluations is age dependent: Ocular movements: Both oculocephalic (doll’s eye response)
Term to 2 months old, 48 hr and vestibulo-ocular (caloric test) reflexes must be absent in
>2 months to 1 year old, 24 hr brain death patients. The oculocephalic reflex is elicited by
>1 year to <18 years old, 12 hr
>18 years old interval is optional, usually 6 hr
rapidly and vigorously turning the head to 90 degrees laterally
on both sides. A normal response is deviation of the eyes to
116 Section II—Clinical and Laboratory Assessment

the opposite side of the head turning; in brain death, there is


no deviation of eyes. This test should not be done if there is Time of Observation
a suspected fracture or instability of the spine. The vestibulo- The clinical examination should be repeated after 6 hours
ocular reflex is elicited by raising the head to 30 degrees and before labeling a person brain dead. The need for repeat exam-
irrigating both tympanic membranes with 50 mL of iced ination and the timing of repeat examination may vary among
saline or water. In brain death patients, there is no eye devia- institutions. In some described criteria for brain death, the
tion. The patient should be observed for up to 1 minute fol- examination should be done by two physicians.32-35 In practice,
lowing irrigation, with a 5-minute wait between testing for however, the observation time to a second neurologic exami-
each ear. Contraindications to the test include rupture of the nation is often much greater. For example, in a recent review
tympanic membrane. of 1229 adult and 82 pediatric patients pronounced brain
Drugs including sedatives, aminoglycoside antibiotics, tri- dead in New York hospitals serviced by the New York Organ
cyclic antidepressants, and some antiseizure agents can dimin- Donor Network, the mean brain death declaration interval
ish the oculocephalic and vestibulo-ocular reflexes. Facial between two examinations was 19.2 hours, that is three times
trauma involving auditory canal and petrous bone also can longer than the state guideline.36 Brain death examinations
inhibit these reflexes. were less frequent on weekends and the interval longer in
Corneal and facial motor responses: Both corneal and facial smaller hospitals. A longer interval was associated with a
reflexes are absent in brain death patients. The corneal reflex decrease in organ donation. The authors found that none of
is elicited by touching the cornea of the patient with a cotton the patients declared brain dead regained brainstem function
swab; the normal response is a blink, which is absent in brain upon repeat examination and so suggested that a single brain
death. The facial motor response is elicited by applying pres- death examination for patients older than 1 year may suffice
sure to the temporomandibular joint, supraorbital ridge, or and so increase organ donation and reduce intensive care unit
nail bed. A normal response is facial grimacing, which is not (ICU) costs.36 Longer brain death duration (i.e., time between
seen in patients with brain death. Interpretation of these declaration of brain death and organ harvesting), however,
maneuvers can be difficult in patients with facial trauma. may not adversely affect organ survival on transplant.37 Newer
Pharyngeal and tracheal reflexes: Both of these reflexes are practice parameters support a single examination rather than
absent in brain death patients. The pharyngeal reflex is elicited dual examination; the impact is unclear, although early studies
by touching the posterior pharyngeal wall with a tongue blade suggest equivalence.38
that normally results in a gag. The cough reflex is elicited by
using bronchial suctioning. These reflexes may be difficult to
evaluate in orally intubated patients but may be feasible with
Confirmatory Tests
deep suctioning in the orally intubated patient.22-24 Confirmatory tests are not mandatory unless clinical tests are
Apnea testing: The medulla controls the central breathing inconclusive. While there are several reports where the value
center via chemoreceptors. These receptors sense changes in of ancillary radiologic imaging is described in decisions about
PaCO2. During apnea testing, target PaCO2 levels are up to brain death,39 a comprehensive clinical evaluation performed
60 mm Hg. To do this test, the patient is removed from the by skilled examiners is considered to have perfect diagnostic
ventilator; 100% oxygen may be administered via the endo- accuracy.40 Possible confirmatory tests are listed in Table 13.2
tracheal tube. In case of chronic hypercarbia (e.g., a patient and are presented briefly here as tests of cerebral blood flow
with chronic obstructive pulmonary disease), higher target (CBF) and electrodiagnostic tests.
values of PaCO2 are required. Cardiac dysrhythmias and
hypotension are complications associated with apnea testing
and can be reduced by taking precautionary measures, such as Tests of Cerebral Blood Flow
preoxygenation and adequate maintenance of the baseline sys- Cerebral angiography. Cerebral angiography is the “gold stan-
tolic blood pressure up to 120 mm Hg with pressors. Both dard” to demonstrate absent CBF. It is the most reliable test
hypocarbia and hypercarbia diminish viability of the organs to confirm brain death, and can save time and reduce an
for organ donation; one suggestion is to administer CO2 exog- unnecessary stay in the ICU. However, it is cumbersome and
enously after preoxygenation to shorten the interval for hyper- requires moving a critically ill patient into an angiography
carbia.25,26 Many patients who undergo apnea testing are on suite for several hours. In brain death patients, cerebral circu-
vasopressors, and an apnea test is not possible in between 5% lation is absent at the level of the carotid bifurcation, although
and 10% of patients because of hemodynamic instability or flow may be demonstrated in the external carotid circulation.
inadequate lung function.27 In hemodynamically stable Extravasation of contrast may result from autolysis.41-44
patients, it is rare that apnea testing is aborted (<5%). Patients Transcranial Doppler ultrasonography (TCD). TCD is an
who fail completion of apnea testing tend to be younger, have important noninvasive confirmatory indirect blood flow test
significantly greater alveolar arterial (A-a) gradients, and are that can be done daily in the NCCU. When criteria recom-
more acidotic.28 Confirmatory tests then are needed if apnea mended by the Task Force Group on Cerebral Death of the
testing is not possible or is aborted. Neurosonology Research Group of the World Federation of
Unstable blood pressure and heart rate: Unstable blood pres- Neurology are used, TCD is a reliable method to establish
sure and heart rate may provide an early indication of hypo- brain death.45 Brain death patients may show the following
thalamic disturbance and impending death. Patients who findings: (1) brief systolic flow and no diastolic flow, (2) no
subsequently meet the criteria for brain death have a higher flow at all in a patient who previously had documented flow,
heart rate and less variability as a result of loss of vegetative and (3) brief systolic forward flow with systolic spikes and
impulses from the brainstem. This may be a useful finding in diastolic reverse flow. In patients with open skull fractures,
deeply comatose patients.29-31 external cerebrospinal fluid drainage, or large decompressive
Section II—Clinical and Laboratory Assessment 117

Table 13.2  Confirmatory Tests That Can Be Electrodiagnostic Testing


Used to Help Confirm the Clinical Diagnosis EEG. An EEG is usually performed for 30 minutes before
of Brain Death concluding brain death because of electrocerebral silence. It is
Confirmatory Test Results
recommended that a minimum of eight scalp electrodes be
used with a distance between electrodes of at least 10 cm.
Cerebral angiography No circulation at level of However, the EEG is not entirely reliable as a confirmatory test
carotid bifurcation and circle
of Willis to document brain death in that minimal electrical activity
Patent external circulation may be seen in some patients who otherwise fulfill brain death
Electroencephalogram Electrocerebral silence for
criteria, and the EEG may appear flat in cases of comatose
30 minutes patients with intact brainstem functioning. According to the
American Encephalographic Society guidelines, electrocere-
Transcranial Doppler Brief systolic flow and reverse
ultrasonography diastolic flow bral silence is confirmed when there is absence of cortical
Brief systolic flow and no activity greater than 2 µV for 30 minutes. EEG results may
diastolic flow be contaminated by electromyographic artifacts from scalp
No flow in patients with motor units.61,62
previously documented flow
Somatosensory and brainstem auditory evoked potentials.
Somatosensory and No response The somatosensory evoked potential (SSEP) is a bedside test
auditory evoked potential done with a portable instrument via stimulation of the median
Technetium 99 scan No radionuclide uptake by nerve. When SSEP is used for brain death determination, it
brain parenchyma presupposes that there is absent pathology in the intervening
Xenon computed Average global flow of less structures between the peripheral nerve and the cortex (e.g.,
tomography than 5 mL/100 mL/min cervical spine injury). Patients with brainstem death have no
Brain tissue oxygenation Persistent zero value for more response. These tests are useful in patients in whom mislead-
than 30 min ing factors such as depressant drugs, hypothermia, and meta-
Blood pressure and heart Absent variability bolic disturbances are present. These tests are less sensitive
rate variability and not recommended for children younger than 6 months
of age.63-65

craniotomies, oscillating flow can be seen, and TCD should be


carried out by an experienced person to avoid misinterpreta-
Controversies in Brain Death
tion of collateral flow signals.46-50 There remain several important philosophical and practical
Radionuclide studies. Cerebral planar scintigraphy or cere- issues in brain death despite its increasing acceptance world-
bral scintotomography with Tc-99 hexamethylpropylenea- wide (see also Chapter 8). Some suggest that the definition of
mine oxime is a safe, sensitive, and cost-effective test that may brain death should be broadened further, and others suggest
be extremely helpful in patients with a skull defect or scalp it already is too broad. The central issue for both groups is the
trauma in whom angiography may be difficult to interpret concept of irreversible coma and particularly loss of respira-
or when there are other confounding factors that may make tory drive. A patient who is unresponsive and who has no
diagnosis equivocal.51 Approximately 25 mCi is given intra­ corneal reflexes but who has spontaneous respiration is not
venously and images are obtained with the help of a mobile brain dead by present criteria. Although recent changes may
scintillation camera.52,53 allow such patients to have support stopped, this is different
Xenon-enhanced computed tomography (Xe-CT). Xe-CT from declaring them dead by brain criteria. A similar issue
is an important tool to measure CBF arrest as a confirmatory arises in anencephalic infants, in whom there is no cortex but
test for brain death. An average global flow of less than some of the brainstem is functioning including spontaneous
5 mL/100 mL/min confirms brain death. This also is a clini- breathing. Even though the child can never gain conscious-
cally useful tool to determine prognosis of the patient.54 Pres- ness, he or she is not brain dead.10-12
ently, the U.S. Food and Drug Administration limits the use Some authors have suggested that irreversible prolonged
of xenon to research protocols only in the United States. loss of consciousness is adequate to establish brain death.
Brain tissue oxygenation and other tests. Monitoring of brain Proponents of this position suggest that:
tissue oxygenation in the comatose patient may help establish
the diagnosis of brain death. A sustained (>30 min) partial 1. Some clinically dead patients maintain residual vegetative
pressure of brain tissue oxygen of zero is consistent with brain functions that are mediated by the brain or brainstem (e.g.,
death. This measurement may be helpful in pharmacologically hypothalamic function when they do not manifest diabe-
depressed patients, including children, to suggest when formal tes insipidus).
brain death assessment should be performed.55-57 However, 2. Some patients may retain slight cerebral electrical activity
brain oxygen levels have yet to be incorporated into defini- on EEG despite having no brainstem reflexes.
tions of brain death. Newer diagnostic tests, such as bispectral 3. Some patients have stereotyped movements in the form of
index, magnetic resonance imaging, computed tomography complex spinal reflexes that are difficult to tell from pur-
angiography (CTA), and computed tomography perfusion poseful movements.
(CTP), have been suggested as useful,58-60 but there is insuffi- 4. Confirmatory tests are necessarily positive predictors or
cient evidence to determine whether newer ancillary tests can may have ambiguity in their interpretation to reliably
confirm the cessation of brain function.8 establish death.
118 Section II—Clinical and Laboratory Assessment

Proponents of this view postulate that brain death criteria liable for injuries that arise from the determination of
should be based on the diagnosis of permanent loss of con- death.81 Interdisciplinary teamwork that includes nurses and
sciousness rather than loss of total brain function.66-73 Their other clinicians in the ICU, palliative care, and the local
new formulation has three components: definition, criteria, organ procurement organization, among others, is necessary
and diagnosis: for organ donation in non–heart-beating donation and do-
nation after cardiac death. Debate or controversy about
Definition: Irreversible loss of consciousness, which is the
death criteria can be healthy in a purely philosophical sense
most integrating function of the organism
but in health care can be harmful, for example, creating dis-
Criteria: Loss of cortical-subcortical connections to gener-
tress for donor families and health care staff, and so institu-
ate both components of consciousness (capacity and
tional standards are necessary to ensure separation of death
cognition)
criteria and organ donation.82
Diagnostic tests: No waking response to stimuli (capacity),
no cognitive or affective functions (content)74
A second idea is to turn time back to 1968 and reform the Family Discussions
concept of “irreversible coma” to “irreversible apneic coma” The family should be told clearly that the patient is dead by
and so abandon the idea of whole brain death but accept brain criteria when these rules are followed; subsequent
instead lack of brainstem function as a sufficient criterion. removal of the ventilator is not “termination of life support”
This was the principle behind British guidelines. These ideas but rather recognizing that death has occurred. In general,
have also generated discussion about organ procurement and dead patients are not ventilated. How families respond to
the dead donor rule that currently states that organ donors brain death or organ donation is beyond the scope of this
must be dead before donation. Donors when declared brain chapter, and the reader is referred to other narratives.83
dead are dead by neurologic criteria. Consequently there is
debate whether patients who have suffered severe and irrevers-
ible brain damage can become organ donors, even though
Brain Death and Organ Donation
they are not yet dead and organ removal then becomes the One of the most important aspects to establishing clear crite-
proximal cause of death.75 For the moment, none of these ria for brain death is organ donation, although the number of
points of view have enough proponents to make a decisive organ donors varies greatly across nations. In many U.S. states,
change in the criteria already presented. it is mandatory to raise the possibility of organ donation with
Since the ad hoc Harvard report was issued, the medical the family of a patient who may be imminently brain dead.
and legal definitions of death have evolved and concepts Transplant coordinators help with the subsequent steps, and
such as loss of integration of the whole organism, loss of this along with an institutional multidisciplinary organ dona-
autonomy, and loss of personhood have generated discus- tion approach or even countrywide efforts can help increase
sion. Functional neuroimaging studies also have provided organ procurement.84-86 Current belief is that it is better for
new insights into patients with severe brain injury and the treating intensivist, neurosurgeon, or neurologist not to be
coma.76 In addition, some scholars advocate return to the involved directly in the solicitation of organs. Their role is
traditional circulatory and respiratory criteria to diagnose simply to establish that brain death is imminent. Uniform
death since brain dead patients may manifest a large variety criteria that define imminent brain death and hence potential
of biologic function when mechanically ventilated.77 Recent- organ donation are not defined in detail but in general will
ly the Institute of Medicine, the Joint Commission on the include a mechanically ventilated deeply comatose patient,
Accreditation of Healthcare Organizations, and the United with irreversible catastrophic brain injury with a clear etiology
Network for Organ Sharing have proposed organ donation of known origin, a Glasgow Coma Score of 3, and the progres-
after cessation of circulation and respiration as a way to in- sive absence of at least three out of six brainstem reflexes or a
crease organ procurement. In ICU care, this concept of FOUR score of zero.87
“timed death” and subsequent organ donation (i.e., con- There remains a chronic shortage of organ donors. Family
trolled organ donation after cardiac death) has generated consent represents one limiting factor for successful dona-
controversy about the limits of treatment that aid organ tion; less than two thirds of families agree to organ donation.
transplant but that may hasten death, and the period of Associated factors in the United States include family
cardiac arrest needed to declare death and commence organ members of minority populations, older potential donors,
procurement.78 A critical factor to understand the meaning medical brain deaths, and a longer duration between brain
of death using circulatory-respiratory tests is the distinction death declaration and discussion about organ donation.88 In
between the “permanent” (will not reverse) and “irrevers- other countries, family refusal for organ donation or low
ible” (cannot reverse) cessation of circulation, not heart- number of organ donations may include denial and rejection
beat.79,80 Legal statutes specify irreversible cessation of of brain death, religious beliefs or fear about organ trade,89
circulatory and respiratory functions. However, the accepted and small hospital size.90 General public awareness is
medical standard is permanent cessation because physicians required to support organ and tissue transplantation.74 In
recognize that irreversible cessation inevitably and swiftly addition, structured education and instruction on definitions
follows once circulation no longer restores itself spontane- of brain and cardiac death and organ donation policies are
ously and cannot be restored medically (i.e., permanent is a necessary in medical and nursing schools and even in sub-
surrogate for irreversibility). The revised Uniform Anatomi- sequent training.91-93
cal Gift Act of 2006 provides protection to those involved in The care of the brain dead individual between declaration
organ procurement. However, the courts have not provided of death and organ procurement and of donation after cardiac
an opinion on whether health care providers may be held death is beyond the scope of this chapter. There are however
Section II—Clinical and Laboratory Assessment 119

many factors that become important to organ procurement 13. Wijdicks EF, Bamlet WR, Maramattom BV, et al. Validation of a new coma
scale: the FOUR score. Ann Neurol 2005;58(4):585–93.
and subsequent recipient survival.94-99 The transplant team
14. Jain S, DeGeorgia M. Brain death associated reflexes and automatism.
rather then the neurointensivist is better equipped to address Neurocrit Care 2005;3(2):122–6.
this period of care, but the neurointensivist should be aware 15. Han SG, Kim GM, Lee KH, et al. Reflex movements in patients with brain
of issues such as ventilatory status, inflammation, hemody- death: a prospective study in tertiary medical center. J Korean Med
namic instability, endocrine and metabolic disturbances, and Sci.2006;21(3):588–90.
16. Saposnik G, Maurino J, Saizar R. Facial myokymia in brain death. Eur J
altered internal homeostasis that may jeopardize organs that Neurol 2001;8(3):227–30.
could be donated. Finally, there are rare circumstances that 17. Saposnik G, Maurino J, Saizar R, et al. Undulating toe movements in brain
lead to maternal brain death during pregnancy. Management death. Eur J Neurol 2004;11(11):723–7.
in this circumstance requires a multidisciplinary approach 18. Van den bent MJ, Ronday M, Oosterlee A. Unexpected movements in a brain
dead patient. Ned Tijdschr Geneeskd 1993;137(51):2653–5.
that follows current guidelines and recommendations for the
19. Ishiguro T, Tamagawa S, Ogawa H. Changes of pupil size in brain death
fetus and for organ preservation of the potential donor.100 patient. Seishin Shinkeigaku Zasshi 1992;94(9):864–73.
20. Ishikawa S. Pupil recent development of investigation. Rinsho Shinkeigaku
1990;30(12):1329–33.
Withdrawal of Life Support 21. Shlugman D, Parulekar M, Elston JS, et al. Abnormal pupil activity in a brain
stem–dead patient. Br J Anaesth 2001;86(5):717–20.
Although a separate issue, withdrawal of life-sustaining mea- 22. Wang MY, Wallace P, Gruen JP. Brain death documentation: analysis and
sures in the individual with catastrophic neurologic injury is issues. Neurosurgery 2002;15(3):731–6.
in part interwoven with the concept of brain death because 23. Thomke F, Weilmann LS. Current concepts in diagnosing brain death in
this decision incorporates “irreversibility” and “permanence.” Germany. Med Klin (Munich) 2000;95(2):85–9.
24. Nathanson M, Bergman PS, Anderson PJ. Significance of oculocephalic
In addition, some of these patients may be suitable for organ
and caloric response in the unconscious patient. Neurology 1957;7(12):
donation after cardiac death protocols. When care is with- 829–32.
drawn in ventilated and comatose patients, up to half will die 25. Sharpe MD, Young GB, Harris C. The apnea test for brain death
within 60 minutes, usually from cardiac arrest. Factors such determination: an alternative approach. Neurocrit Care 2004;1(3):363–6.
as absent corneal and cough reflexes, extensor or absent motor 26. Wu XL, Fang Q, Li L, et al. Complication associated with the apnea test
in the determination of the brain death. Chin Med J (Engl) 2008;121(13):
response, and poor oxygenation are associated with earlier 1169–72.
death.101 This information can be used to guide family and 27. Wijdicks EF, Rabinstein AA, Manno EM, et al. Pronouncing brain death:
health care provider expectations. In these patients, the pres- contemporary practice and safety of the apnea test. Neurology 2008;71(16):
ence and severity of pain cannot be assessed and so palliative 1240–4.
28. Yee AH, Mandrekar J, Rabinstein AA, et al. Predictors of apnea test failure
analgesia and sedation are necessary. Palliative sedation can be
during brain death determination. Neurocrit Care 2010;12(3):352–5.
complex and requires consideration about whether the medi- 29. Conci F, Di Rienzo M, Castiglioni P. Blood pressure and heart variability and
cation contributes to covert euthanasia or acceleration of baroreflex sensitivity before and after brain death. J Neurosurg Psychiatry
death. 2001;71(5):621–31.
30. Freitas J, Puig J, Rocha AP, et al. Heart rate variability in brain death. Clin
Auton Res 1996;6(3):141–6.
References 31. Knuttgen D, Zur Nieden K, Muller-Georges MR, et al. Computer aided
analysis of heart rate variability in brain death. Int J Clin Monit Comput
1. Manninen BA. Defining human death: an intersection of bioethics and 1997;14(1):37–42.
metaphysics. Rev Neurosci 2009;20(3-4):283–92. 32. Ashwal S, Scheneider S. Brain death in the newborn. Pediatrics 1989;84(3):
2. Ball CG, Navsaria P, Kirkpatrick AW, et al. The impact of country and 429–37.
culture on end-of-life care for injured patients: results from an international 33. Villani A, Onofri A, Bianchi R, et al. Determination of brain death in
survey. J Trauma 2010;69(6):1323–34. intensive pediatric therapy. Pediatr Med Chir 1998;20(1):19–23.
3. Practice parameters for determining brain death in adults: summary 34. Parker BL, Frewen TC, Levine SD, et al. Declaring pediatric brain death:
statement. Report of the quality Standards Subcommittee of the American current practice in a Canadian pediatric critical care unit. CMAJ 1995;
Academy of Neurology. In: Practice handbook: American Academy of 153(7):909–16.
Neurology. St Paul, Minn: American Academy of Neurology; 1994. 35. Religious, cultural, legal, and ethical considerations. Crit Care Med 1993;
4. A definition of irreversible coma: report of the Ad Hoc Committee of the 21(12):1951–65.
Harvard Medical School to Examine the Definition of the brain death. 36. Schimizu N, Shemie S, Miyasaka E, et al. Preliminary report: use of clinical
JAMA 1968;205:337–40. criteria for the determination of pediatric brain death and confirmation by
5. Burkle CM, Schipper AM, Wijdicks EF. Brain death and the courts. radionuclide cerebral blood flow. Masi 2000;49(10):1126–32.
Neurology 2011;76(9):837–41. 37. Lustbader D, O’Hara D, Wijdicks EF, et al. Second brain death examination
6. Guidelines for the determination of death: report of the medical may negatively affect organ donation. Neurology 2011;76(2):119–24.
consultation on the diagnosis of death to the President’s Commission for 38. Nijboer WN, Moers C, Leuvenink HG, et al. How important is the duration
the Study of Ethical Problems in Medicine and Biomedical and Behavioral of the brain death period for the outcome in kidney transplantation? Transpl
Research. JAMA 1981;246:2184–6. Int 2011;24(1):14–20.
7. The Quality Standards Subcommittee of the American Academy of 39. Varelas PN, Rehman M, Abdelhak T, et al. Single brain death examination
Neurology. Practice parameters for determining brain death in adults is equivalent to dual brain death examinations. Neurocrit Care 2011;15(3):
(summary statement). Neurology 1995;45:1012–14. 547–53.
8. Wijdicks EF, Varelas PN, Gronseth GS, et al, American Academy of 40. Roberts DJ, MacCulloch KA, Versnick EJ, et al. Should ancillary brain blood
Neurology. Evidence-based guideline update: determining brain death in flow analyses play a larger role in the neurological determination of death?
adults: report of the Quality Standards Subcommittee of the American Can J Anaesth 2010;57(10):927–35.
Academy of Neurology. Neurology 2010;74(23):1911–18. 41. Wijdicks EF. The case against confirmatory tests for determining brain death
9. Busl KM, Greer DM. Pitfalls in the diagnosis of brain death. Neurocrit Care in adults. Neurology 2010;75(1):77–83.
2009;11(2):276–87. 42. Young GB, Lee D. A critique of ancillary tests for brain death. Neurocrit Care
10. John J, Gane BD, Plakkal N, et al. Snake bite mimicking brain death. Case J 2004;1(4):499–508.
2008;1(1):16. 43. Young GB, Shemie SD, Doig J, et al. Brief review: the role of ancillary
11. Miller RK. Fisher syndrome, a brainstem encephalitis, mimics brain death. tests in the neurological determination of death. Can Anaesth 2006;53(6):
Clin Pediatr (Phila) 1993;32(11):685–7. 540–3.
12. Singounas EG. Glasgow coma scale and brain death: a proposal. Acta 44. Wijdicks EFM. Determining brain death in adults. Neurology 1995;45:
Neurochir (Wein) 1995;133(1-2):60. 1003–11.
120 Section II—Clinical and Laboratory Assessment

45. Alvarez LA, Lipto RB, Hirschfeld A, et al. Brain death determination by 49. Kuo JR, Chen CF, Chico CC, et al. Time dependant validity in the diagnosis
angiography in the setting of a skull defect. Arch Neurol 1988;45(2):225–7. of brain death using transcranial Doppler sonography. J Neurolsurg
46. Sharma D, Souter MJ, Moore AE, et al. Clinical experience with transcranial Psychiatry 2006;77(5):646–9.
Doppler ultrasonography as a confirmatory test for brain death: a 50. Calleja S,Tembl JI, Sequra T. Sociedad Espanola Neurosonologia.
retrospective analysis. Neurocrit Care 2011;14(3):370–6. Recommendations of the use of transcranial Doppler to determine the
47. Poularas J, Karakitsos D, Kouraklis G, et al. Comparison between existence of cerebral circulatory arrest as diagnostic support of brain death.
transcranial color Doppler ultrasonography and angiography in the Neurolgia 2007;22(7):441–7.
confirmation of brain death. Transplant Proc 2006;38(5):1213–17.
48. Valentin A, Karnik R, Winkler WB, et al. Transcranial Doppler for early A complete list of references for this chapter can be found online at
identification of potential organ transplant donors. Wien Klin Wochenschr www.expertconsult.com.
1997;109(21):836–9.
Section II—Clinical and Laboratory Assessment 120.e1

References 31. Knuttgen D, Zur Nieden K, Muller-Georges MR, et al. Computer aided
analysis of heart rate variability in brain death. Int J Clin Monit Comput
1. Manninen BA. Defining human death: an intersection of bioethics and 1997;14(1):37–42.
metaphysics. Rev Neurosci 2009;20(3-4):283–92. 32. Ashwal S, Scheneider S. Brain death in the newborn. Pediatrics
2. Ball CG, Navsaria P, Kirkpatrick AW, et al. The impact of country and 1989;84(3):429–37.
culture on end-of-life care for injured patients: results from an 33. Villani A, Onofri A, Bianchi R, et al. Determination of brain death in
international survey. J Trauma 2010;69(6):1323–34. intensive pediatric therapy. Pediatr Med Chir 1998;20(1):19–23.
3. Practice parameters for determining brain death in adults: summary 34. Parker BL, Frewen TC, Levine SD, et al. Declaring pediatric brain death:
statement. Report of the quality Standards Subcommittee of the American current practice in a Canadian pediatric critical care unit. CMAJ
Academy of Neurology. In: Practice handbook: American Academy of 1995;153(7):909–16.
Neurology. St Paul, Minn: American Academy of Neurology; 1994. 35. Religious, cultural, legal, and ethical considerations. Crit Care Med
4. A definition of irreversible coma: report of the Ad Hoc Committee of the 1993;21(12):1951–65.
Harvard Medical School to Examine the Definition of the brain death. 36. Schimizu N, Shemie S, Miyasaka E, et al. Preliminary report: use of clinical
JAMA 1968;205:337–40. criteria for the determination of pediatric brain death and confirmation by
5. Burkle CM, Schipper AM, Wijdicks EF. Brain death and the courts. radionuclide cerebral blood flow. Masi 2000;49(10):1126–32.
Neurology 2011;76(9):837–41. 37. Lustbader D, O’Hara D, Wijdicks EF, et al. Second brain death examination
6. Guidelines for the determination of death: report of the medical may negatively affect organ donation. Neurology 2011;76(2):119–24.
consultation on the diagnosis of death to the President’s Commission for 38. Nijboer WN, Moers C, Leuvenink HG, et al. How important is the duration
the Study of Ethical Problems in Medicine and Biomedical and Behavioral of the brain death period for the outcome in kidney transplantation?
Research. JAMA 1981;246:2184–6. Transpl Int 2011;24(1):14–20.
7. The Quality Standards Subcommittee of the American Academy of 39. Varelas PN, Rehman M, Abdelhak T, et al. Single brain death examination
Neurology. Practice parameters for determining brain death in adults is equivalent to dual brain death examinations. Neurocrit Care
(summary statement). Neurology 1995;45:1012–4. 2011;15(3):547–53.
8. Wijdicks EF, Varelas PN, Gronseth GS, et al. American Academy of 40. Roberts DJ, MacCulloch KA, Versnick EJ, et al. Should ancillary brain blood
Neurology. Evidence-based guideline update: determining brain death in flow analyses play a larger role in the neurological determination of death?
adults: report of the Quality Standards Subcommittee of the American Can J Anaesth 2010;57(10):927–35.
Academy of Neurology. Neurology 2010;74(23):1911–18. 41. Wijdicks EF. The case against confirmatory tests for determining brain
9. Busl KM, Greer DM. Pitfalls in the diagnosis of brain death. Neurocrit Care death in adults. Neurology 2010;75(1):77–83.
2009;11(2):276–87. 42. Young GB, Lee D. A critique of ancillary tests for brain death. Neurocrit
10. John J, Gane BD, Plakkal N, et al. Snake bite mimicking brain death. Case J Care 2004;1(4):499–508.
2008;1(1):16. 43. Young GB, Shemie SD, Doig J, et al. Brief review: the role of ancillary tests
11. Miller RK. Fisher syndrome, a brainstem encephalitis, mimics brain death. in the neurological determination of death. Can Anaesth 2006;53(6):540–3.
Clin Pediatr (Phila) 1993;32(11):685–7. 44. Wijdicks EFM. Determining brain death in adults. Neurology 1995;45:
12. Singounas EG. Glasgow coma scale and brain death: a proposal. Acta 1003–11.
Neurochir (Wein) 1995;133(1-2):60. 45. Alvarez LA, Lipto RB, Hirschfeld A, et al. Brain death determination by
13. Wijdicks EF, Bamlet WR, Maramattom BV, et al. Validation of a new coma angiography in the setting of a skull defect. Arch Neurol 1988;45(2):225–7.
scale: the FOUR score. Ann Neurol 2005;58(4):585–93. 46. Sharma D, Souter MJ, Moore AE, et al. Clinical experience with transcranial
14. Jain S, DeGeorgia M. Brain death associated reflexes and automatism. Doppler ultrasonography as a confirmatory test for brain death: a
Neurocrit Care 2005;3(2):122–6. retrospective analysis. Neurocrit Care 2011;14(3):370–6.
15. Han SG, Kim GM, Lee KH, et al. Reflex movements in patients with brain 47. Poularas J, Karakitsos D, Kouraklis G, et al. Comparison between
death: a prospective study in tertiary medical center. J Korean Med Sci transcranial color Doppler ultrasonography and angiography in the
2006;21(3):588–90. confirmation of brain death. Transplant Proc 2006;38(5):1213–17.
16. Saposnik G, Maurino J, Saizar R. Facial myokymia in brain death. Eur J 48. Valentin A, Karnik R, Winkler WB, et al. Transcranial Doppler for early
Neurol 2001;8(3):227–30. identification of potential organ transplant donors. Wien Klin Wochenschr
17. Saposnik G, Maurino J, Saizar R, et al. Undulating toe movements in brain 1997;109(21):836–9.
death. Eur J Neurol 2004;11(11):723–7. 49. Kuo JR, Chen CF, Chico CC, et al. Time dependant validity in the diagnosis
18. Van den bent MJ, Ronday M, Oosterlee A. Unexpected movements in a of brain death using transcranial Doppler sonography. J Neurolsurg
brain dead patient. Ned Tijdschr Geneeskd 1993;137(51):2653–5. Psychiatry 2006;77(5):646–9.
19. Ishiguro T, Tamagawa S, Ogawa H. Changes of pupil size in brain death 50. Calleja S,Tembl JI, Sequra T. Sociedad Espanola Neurosonologia.
patient. Seishin Shinkeigaku Zasshi 1992;94(9):864–73. Recommendations of the use of transcranial Doppler to determine the
20. Ishikawa S. Pupil recent development of investigation. Rinsho Shinkeigaku existence of cerebral circulatory arrest as diagnostic support of brain death.
1990;30(12):1329–33. Neurolgia 2007;22(7):441–7.
21. Shlugman D, Parulekar M, Elston JS, et al. Abnormal pupil activity in a 51. Burger R. Value of transcranial Doppler ultrasonography compared with
brain stem–dead patient. Br J Anaesth 2001;86(5):717–20. scintigraphic techniques and EEG in brain death. Zentrailbl Neurochir
22. Wang MY, Wallace P, Gruen JP. Brain death documentation: analysis and 2000;61(10):7–13.
issues. Neurosurgery 2002;15(3):731–6. 52. Bertagna F, Barozzi O, Puta E, et al. Residual brain viability, evaluated by
23. Thomke F, Weilmann LS. Current concepts in diagnosing brain death in (99m)Tc-ECD SPECT, in patients with suspected brain death and with
Germany. Med Klin (Munich) 2000;95(2):85–9. confounding clinical factors. Nucl Med Commun 2009;30(10):815–21.
24. Nathanson M, Bergman PS, Anderson PJ. Significance of oculocephalic 53. Conrad GR, Sinha P. Scintography as a confirmatory test of brain death.
and caloric response in the unconscious patient. Neurology 1957;7(12): Semin Nucl Med 2003;33(4):312–23.
829–32. 54. Kurtek RW, Lia KK, Tauxe WN, et al. Tc 99 hexamethylpropylene amino
25. Sharpe MD, Young GB, Harris C. The apnea test for brain death oxime scintography in the diagnosis of brain death and its implications
determination: an alternative approach. Neurocrit Care 2004;1(3):363–6. in the harvesting of organs used for transplant. Clin Nucl Med 2000;
26. Wu XL, Fang Q, Li L, et al. Complication associated with the apnea test 25(1):7–10.
in the determination of the brain death. Chin Med J (Engl) 2008; 55. Pistoia F, Johnson DW, Darby JM, et al. The role of xenon CT
121(13):1169–72. measurements of cerebral blood flow in the clinical determination of brain
27. Wijdicks EF, Rabinstein AA, Manno EM, et al. Pronouncing brain death: death. AJNR Am J Neurodial 1991;12(1):97–103.
contemporary practice and safety of the apnea test. Neurology 56. Figaji AA, Kent SJ. Brain tissue oxygenation in children diagnosed with
2008;71(16):1240–4. brain death. Neurocrit Care 2010;12(1):56–61.
28. Yee AH, Mandrekar J, Rabinstein AA, et al. Predictors of apnea test failure 57. Smith ML, Counelis GJ, Maloney-Welinsky E, et al. Brain tissue oxygen
during brain death determination. Neurocrit Care 2010;12(3):352–5. tension in clinical brain death: a case series. Neurol Res 2007;29(7):755–9.
29. Conci F, Di Rienzo M, Castiglioni P. Blood pressure and heart variability 58. Palmer S, Badar MK. Brain tissue oxygenation in brain death. Neurocrit
and baroreflex sensitivity before and after brain death. J Neurosurg Care 2005;2(1):17–22.
Psychiatry 2001;71(5):621–31. 59. Bohatyrewicz R, Sawicki M, Walecka A, et al. Computed tomographic
30. Freitas J, Puig J, Rocha AP, et al. Heart rate variability in brain death. Clin angiography and perfusion in the diagnosis of brain death. Transplant Proc
Auton Res 1996;6(3):141–6. 2010;42(10):3941–6.
120.e2 Section II—Clinical and Laboratory Assessment

60. Escudero D, Otero J, Marqués L, et al. Diagnosing brain death by CT 82. Verheijde JL, Rady MY, McGregor J. Presumed consent for organ
perfusion and multislice CT angiography. Neurocrit Care 2009;11(2): preservation in uncontrolled donation after cardiac death in the United
261–71. States: a public policy with serious consequences. Philos Ethics Humanit
61. Berenguer CM, Davis FE, Howington JU. Brain death confirmation: Med 2009;4:15.
comparison of computed tomographic angiography with nuclear medicine 83. Dubois JM. The ethics of creating and responding to doubts about death
perfusion scan. J Trauma 2010;68(3):553–9. criteria. J Med Philos 2010;35(3):365–80.
62. Rimmele T, Malhicheikh A, Boselli E, et al. The electroencephalogram is 84. Manuel A, Solberg S, MacDonald S. Organ donation experiences of family
not an adequate test to confirm the diagnosis of brain death. Can J Anaesth members. Nephrol Nurs J 2010;37(3):229–37.
2008;55(3):188–90. 85. Kong AP, Barrios C, Salim A, et al. A multidisciplinary organ donor council
63. Sethi NK, Sethi PK, Torgounick J. EMG artifact in brain death and performance improvement initiative can improve donation outcomes.
electroencephalogram, is it a cry of medullary death? Clin Neurol Am Surg 2010;76(10):1059–62.
Neurosurg 2008;110(7):729–31. 86. Bleakley G. Implementing minimum notification criteria for organ donation
64. Guerit JM. Evoked potential, a safe brain death confirmatory tool? Eur J in an acute hospital’s critical care units. Nurs Crit Care 2010;15(4):185–91.
Med 1992;1(4):233–43. 87. Matesanz R, Marazuela R, Dominguez-Gil B, et al. The 40 donors per
65. Wagner W. Scalp, ear lobe and nasopharyngeal recording of the median million population plan: an action plan for improvement of organ
nerve somatosensory evoked P14 potential in coma and brain death patient donation and transplantation in Spain. Transplant Proc 2009;41(8):3453–6.
and amplitude analysis. Brain 1996;119(Pt 5):1507–21. 88. de Groot YJ, Jansen NE, Bakker J, et al. Imminent brain death: point of
66. Jarddim M. Brainstem auditory evoked potential as a model to assist departure for potential heart-beating organ donor recognition. Intensive
diagnosis of brain death. Pro Fono 2008;20(2):123–8. Care Med 2010;36(9):1488–94.
67. Bacigalupo F. The debate about death: an imperishable discussion. Biol Res 89. Brown CV, Foulkrod KH, Dworaczyk S, et al. Barriers to obtaining family
2007;40(4):523–34. consent for potential organ donors. J Trauma 2010;68(2):447–51.
68. Nojszewska M. Determination of brain death. Neurol Neurochir Pol 90. Ghorbani F, Khoddami-Vishteh HR, et al. Causes of family refusal for
2002;36(1):91–104. organ donation. Transplant Proc 2011;43(2):405–6.
69. Boobes Y, Al Daker N. What it means to die in Islam and modern medicine. 91. Formanek M, Schöffski O. Difficulties with the organ donation process in
Saudi J Kidney Dis Transpl 1996;7(2):121–7. small hospitals in Germany. Transplant Proc 2010;42(5):1445–8.
70. Bryne PA, O’Reilly S, Quary PM. Brain death: an opposing viewpoint. 92. Anker AE, Feeley TH, Friedman E, et al. Teaching organ and tissue
JAMA 1979;242(18):1985–90. donation in medical and nursing education: a needs assessment. Prog
71. Trueba J. Clinical death, a diagnosis and a testimony. An Sist Sanit Nava Transplant 2009;19(4):343–8.
2007;30(Suppl 3):57–70. 93. Lima CX, Lima MV, Cerqueira RG, et al. Organ donation: cross-sectional
72. Aker J, Pearson KS. AANA Journal Course. Update for nurse anesthetists survey of knowledge and personal views of Brazilian medical students and
brain death: terminology, clinical criteria, and diagnostic testing. AANA J physicians. Transplant Proc 2010;42(5):1466–71.
1997;65(2):129–35. 94. Rykhoff ME, Coupland C, Dionne J, et al. A clinical group’s attempt to raise
73. Truog RD, Fackler JC. Rethinking brain death. Crit Care Med awareness of organ and tissue donation. Prog Transplant 2010;20(1):33–9.
1992(12):1705–13. 95. Murugan R, Venkataraman R, Wahed AS, et al, HIDonOR Study
74. Walker AE, Diamond EL, Moseley J. The neuropathological findings in Investigators. Increased plasma interleukin-6 in donors is associated with
irreversible coma: a critique of the respirator. J Neuropathol Exp Neurol lower recipient hospital-free survival after cadaveric organ transplantation.
1975;34(4):295–323. Crit Care Med 2008;36(6):1810–16.
75. Machado-Curbelo C. A new formulation of death, definition, criteria and 96. Dictus C, Vienenkoetter B, Esmaeilzadeh M, et al. Critical care management
diagnostic tests. Rev Neurol 1998;26(154):1040–7. of potential organ donors: our current standard. Clin Transplant
76. Collins M. Reevaluating the dead donor rule. J Med Philos 2010;35(2): 2009;23(Suppl 21):2–9.
154–79. 97. Mascia L, Pasero D, Slutsky AS, et al. Effect of a lung protective strategy
77. Cavanna AE, Cavanna SL, Servo S, et al. The neural correlates of impaired for organ donors on eligibility and availability of lungs for transplantation:
consciousness in coma and unresponsive states. Discov Med a randomized controlled trial. JAMA 2010;304(23):2620–7.
2010;9(48):431–8. 98. Dubbeld J, van Hoek B, Ringers J. Use of a liver from donor after cardiac
78. Miller FG, Truog RD. Decapitation and the definition of death. J Med death: is it appropriate for the sick or the stable? Curr Opin Organ
Ethics 2010;36(10):632–4. Transplant 2011;16(2):239–42.
79. Souter M, Van Norman G. Ethical controversies at end of life after 99. Frontera JA. How I manage the adult potential organ donor: donation after
traumatic brain injury: defining death and organ donation. Crit Care Med cardiac death (part 2). Neurocrit Care 2010;12(1):111–16.
2010;38(9 Suppl):S502–9. 100. Esmaeilzadeh M, Dictus C, Kayvanpour E, et al. One life ends, another
80. Thomas AG. Continuing the definition of death debate: the report of the begins: management of a brain-dead pregnant mother—a systematic
President’s Council on Bioethics on Controversies in the Determination of review. BMC Med 2010;8:74.
Death. Bioethics 2012;26(2):101–7. 101. Yee AH, Rabinstein AA, Thapa P, et al. Factors influencing time to death
81. Veatch RM. Transplanting hearts after death measured by cardiac criteria: after withdrawal of life support in neurocritical patients. Neurology
the challenge to the dead donor rule. J Med Philos 2010;35(3):313–29. 2010;74(17):1380–5.
Chapter
14  
II

Glucose and Nutrition


Sarice L. Bassin and Thomas P. Bleck

2010, the American Diabetes Association (ADA) proposed a


Introduction threshold of 140 mg/dL (7.8 mmol/L; ADA Standards of
Hyperglycemia is common in acutely ill patients. In the past, Medical Care in Diabetes–2010 http://care.diabetesjournals.
stress hyperglycemia was considered to be a beneficial adaptive org/content/33/Supplement_1/S11.full). Hyperglycemia may
response. However, prospective observational and clinical be considered as mild (110-180 mg/dL), moderate (180-
trial data show a consistent and clear association between 400 mg/dL) or severe (>400 mg/dL). Table 14.1 lists the
hyperglycemia and increased morbidity and mortality in all diagnostic criteria for diabetic ketoacidosis or hyperos-
critically ill populations and in particular those with acute molar hyperglycemic state. In the hospitalized patient stress
brain injury, stroke, and subarachnoid hemorrhage. Glycemic and medication (e.g., steroids) are factors associated with
control therefore has become an important aspect of critical elevation of blood glucose. In addition, there can be relative
care. However, the optimal level of glycemic control in critical insulin resistance after administration of catecholamines or
illness remains in debate since Van den Berghe et al. published glucocorticoids.
their study conclusions in 2001 showing that strict glucose
control was associated with reduced mortality.1,2 In these ran-
domized trials intensive insulin therapy (IIT) was used to Assessment of Glucose in the Intensive
target a blood glucose of 80 to 110 mg/dL. Studies since then Care Unit
have not been able to duplicate these results (Normoglycemia Blood to measure glucose can be obtained from a variety of
in Intensive Care Evaluation-Survival Using Glucose Algo- sites (e.g., a fingerstick device, venipuncture, or through an
rithm Regulation [NICE-SUGAR]),3 and so a more liberal intra-arterial or venous catheter). Fingersticks may be inac-
glucose target of 140 to 180 mg/dL is favored. In addition, an curate when there is edema, hypoperfusion, or anemia,
association between hypoglycemia that can be caused by IIT, whereas capillary blood glucose monitoring may not be accu-
and high glycemic variability and poor outcome also is appar- rate in shock.16 In addition, care to prevent contamination
ent, further emphasizing the importance of glucose control.4-7 by intravenous solutions is needed when using samples from
The safety and efficacy of IIT may be influenced by patient- arterial or venous catheters because pseudohyperglycemia
related and intensive care unit–related variables. In particular, may be present if a blood sample is drawn from a line where
nutrition is closely associated with glycemic control.8,9 Fur- the patient is receiving dextrose 5% in water (D5W) or total
thermore, guidelines recommend use of early nutritional parenteral nutrition (TPN). Laboratory analysis of plasma
support, preferably enteral nutrition. However, optimal energy is the best means to analyze blood glucose levels. However,
requirements and the timetable of nutritional support still are this technique may be too slow for care of critically ill patients.
not fully resolved, in part because energy expenditure needs Many hospitals now have laboratories operated by licensed
may not be met with enteral nutrition only, and there are medical technologists in, or close to, the ICU that allow
different recommendations for parental nutrition in Europe rapid assessment of blood gases, electrolytes, hemoglobin,
and North America.10-15 This chapter addresses the impor- prothrombin time (PT), and glucose through a blood gas
tance of glucose control: how to monitor glucose levels in ICU analyzer or “stat” platform (e.g., i-STAT [Abbott Point of
patients, practical aspects of glucose control in ICUs, nutrition Care, Princeton, NJ], Radiometer ABL 825, or RapidLab
therapy, and methods to calculate nutritional requirements in 865).17 These devices generally are very accurate and can be
critical care. used interchangeably with core laboratory devices,18 but turn-
around time can still be a limiting factor.
Glucose
Plasma glucose usually is between 60 and 100 mg/dL before Point-of-Care Testing
meals and up to 150 mg/dL after a meal. Hypoglycemia is Glucose testing using point-of-care (POC) devices has become
defined as a blood glucose less than 50 mg/dL. Several factors increasingly common in hospital and ICU settings as well as
such as malnutrition, cirrhosis, renal failure, and alcoholic in the operating room and in the outpatient environment.19
ketoacidosis or drugs such as pentamidine, haloperidol, In addition, there are many different self-monitoring blood
bactrim, or salicylates increase the risk of developing hypogly- glucose (SMBG) devices available for patients to use, for
cemia. Traditionally, acute hyperglycemia was defined as a example, Accu-Chek Active and Accu-Chek Performa (Roche
random glucose concentration greater than 200 mg/dL. In Diagnostics, Indianapolis, IN), OneTouch Ultra2 (LifeScan
© Copyright 2013 Elsevier Inc. All rights reserved. 121
122 Section II—Clinical and Laboratory Assessment

Table 14.1  Diagnostic Criteria for Diabetic Ketoacidosis and Hyperosmolar Hyperglycemic State
Criteria Diabetic Ketoacidosis Hyperosmolar Hyperglycemic State
Mild Moderate Severe
Plasma glucose (mg/dL) >250 >250 >250 >600
Arterial pH 7.25-7.30 7.00-7.24 <7.00 >7.30
Serum bicarbonate (mEq/L) 15-18 10 to <15 <10 >15
Urine ketones Positive Positive Positive Small
Serum ketones Positive Positive Positive Small
Effective serum osmolality* Variable Variable Variable >320
Anion gap †
>10 >12 >12 Variable
Level of consciousness Alert Alert/drowsy Stupor/coma Stupor/coma

Adapted from Kitabachi AE, Umpierrez GE, Murphy MB, et al. Hyperglycemic crisis in adult patients with diabetes: a consensus statement from the American
Diabetes Association. Diabetes Care 2006;29:2739–48.
*
Effective serum osmolality = 2[measured Na+] + glucose/18.

Anion gap = Na+ − (Cl− + HCO3−)

Inc., Milpitas, CA), MediSense Optium or Xceed (Abbott Dia-


betes Care Inc., Alameda, CA), and others. These devices all GEM 3000
are subject to strict regulatory standards (i.e., International The GEM 3000 (Instrumentation Laboratories, Bedford, MA)
Organization for Standardization. In vitro diagnostic test has thick film sensors or electrodes in strips that can measure
systems: requirements for blood glucose monitoring systems glucose, lactate, and blood gases. The sensors contain reagents
for self-testing in managing diabetes mellitus. [ISO 15197:2003 and calibrators packed into small cartridges that are placed in
http://www.iso.org/iso/home/store/catalogue_tc/catalogue_ the body of the analyzer. The cartridges can test multiple
detail.htm?csnumber=26309; accessed Oct. 19, 2012.) samples and the sensors are reusable. Samples with increased
Although POC devices are used at bedside, they are licensed osmolalities, high protein, or high lipid volumes adversely
under laboratory medical directors and governed by several affect the function. In addition, gallamine triethiodide,
regulatory bodies such as Health Care Financing Administra- ethanol, sodium fluoride, potassium oxalate, acetaminophen,
tion (HCFA), The Joint Commission, Clinical Laboratory isoniazide, or thiocyanate may affect glucose or lactate levels.
Improvements Amendments of 1988 (CLIA 88), the College
of American Pathologists (CAP), and individual state depart-
ments of health, hospital policies, and procedure manuals for
each device. This means use of the devices must follow regula- Minimally Invasive Devices
tion, and there needs to be proficiency testing for accuracy and Continuous monitoring sensors (e.g., CDI 500 Parameter
testing of unknown samples provided by CAP or commercial Monitoring System, Terumo Cardiovascular Systems, Ann
sources for comparison to reference range values to calibrate Arbor, MI) that are inserted into the bloodstream are available
and verify the device. Staff who use the devices also are but their benefit still needs to be elucidated. These monitors
expected to receive annual competency testing. POC monitors based on optical technology are primarily used to measure
are reasonably accurate, particularly when blood from a cath- blood gases and most commonly have been used in cardiac
eter is used. However, POC glucometers can be inaccurate surgery. However, electrochemical sensors that can monitor
(20%) when patients have low blood glucose levels17 and can glucose are available.
differ from laboratory values20; not all achieve ISO 15197
accuracy criteria.21,22 This can limit their use when tight
glucose control is targeted.23 Consequently it may be better to
examine blood glucose levels using central lab or blood gas Microdialysis
analyzers rather than POC devices24 and when POC devices In 2009 the Eirus system (Dipylon, Solna, Sweden) was CE-
are used careful clinical evaluation of each system for glucose marked for continuous glucose and lactate monitoring in
measurement is critical. blood, and in 2011, the Eirus TLC catheter received its CE
mark. This technology is based on microdialysis so no blood
is removed from the patient, and it does not require a separate
i-STAT Analyzer line because the device includes a central venous catheter. It
This device can measure blood gases, electrolytes, glucose, and allows second-by-second monitoring of glucose and lactate.
certain coagulation parameters through single-use sensors The glucose value is presented on a monitor as a numeric
constructed using thin-film technology. Because the sensor value and a trend curve is provided. Studies have verified its
film is thin, it can be used immediately and unlike thick film technical feasibility, accuracy, and performance in the ICU.25,26
sensors does not require a calibration period. Waste, however, However, as of 2012 the device has not yet become commer-
is a potential disadvantage of use. cially available.
Section II—Clinical and Laboratory Assessment 123

to have a poor outcome at 10 months after SAH.37 Others


Why Is Glycemic Control Important in the have found that mean glucose concentrations greater than
Neurocritical Care Unit? 140  mg/dL are associated with the development of symp-
In critical illness, release of stress hormones (e.g., epinephrine tomatic vasospasm after SAH.38,44 In traumatic brain injury
and cortisol), medications such as exogenous glucocorticoids (TBI) hyperglycemia also is associated with decreased sur-
or catecholamines, and release of inflammatory mediators vival, higher intracranial hypertension, and increased length
(e.g., in sepsis or trauma) all inhibit insulin release and action. of hospital stay.45,46
This results in stress hyperglycemia, defined as an increase in Whether hyperglycemia in the setting of acute neurologic
blood glucose in the presence of acute illness,27 through injury is simply an epiphenomenon or actually serves a pro-
enhanced gluconeogenesis, inhibition of glycogen synthesis, tective purpose remains controversial. The stress response
and impaired insulin-mediated glucose uptake by tissues. In associated with neurologic injury can lead to hyperglycemia
addition, intravenous dextrose (e.g., in parenteral nutrition or through liberation of catecholamines that induce gluconeo-
in antibiotic solutions), can exacerbate or contribute to hyper- genesis and insulin resistance. This may be an adaptive and
glycemia. Overall, hyperglycemia may be present in 40% of protective response because glucose is the primary source of
critically ill patients; 80% of ICU patients with hyperglycemia energy in the brain. However, some have postulated that in the
have no history of diabetes before admission.28 setting of anaerobic metabolism, hyperglycemia will result in
Hyperglycemia during critical illness is associated with the accumulation of lactate and an intracellular acidosis, a
adverse outcome and an increased risk for infection, myocar- potential mediator of secondary injury. Conversely, if the
dial infarction, and neurologic and renal damage, particularly supply of glucose, the main energy substrate of the brain, is
in patients who are not diabetic before admission.29 The effect inadequate, there could be insufficient fuel for glycolysis in
of hyperglycemia also depends in part on patient pathology already ischemic tissue. Termed the glucose paradox of cerebral
(i.e., ICU admission diagnosis).30 However, the impact of ischemia, this conundrum makes determination of an exact
hyperglycemia on patients with preexisting diabetes is less target for plasma glucose concentrations in patients with
clear even though diabetic patients are at greater risk for com- severe neurologic injury difficult.47 The conundrum is further
plications.31 In fact, in diabetic patients with elevated hemo- complicated by accumulating evidence that in some circum-
globin A1c (HbA1c) levels (>7%), glucose levels that are stances after acute brain injury lactate is used as a fuel in the
desirable in other patients appear to be “unsafe” in diabetic brain and may even be protective.48,49
patients with chronic hyperglycemia.32 Although the likeli-
hood of hyperglycemia is greater in sicker patients, it appears
that hyperglycemia itself is an independent factor associated
with outcome particularly in stroke, both hemorrhagic and Insulin Therapy in Critical Care
ischemic, and acute coronary syndromes.33-40 In addition, sur- In 2001 and 2006 two single center randomized clinical
vival is inversely associated with the duration of hyperglyce- trials1,2 demonstrated the association between IIT and
mia.29 However, the exact threshold at which elevated blood improved outcomes in critically ill patients in surgical ICUs
glucose is deleterious remains uncertain. The relationship (SICU) and medical ICUs (MICU). In these trials patients
between hyperglycemia and increased risk of worse outcome were randomly assigned to IIT (target glucose 80-110 mg/dL
has driven research into glucose control in the ICU, the value [4.4-6.1 mmol/L]) or standard care (target glucose 180-
and structure of which remain debated, in part because tight 200 mg/dL [9.9-11 mmol/L]). In SICU patients (n = 1548),
glucose control is associated with an increased risk for hypo- Van den Berghe et al. observed that an IIT protocol that
glycemia that in turn is associated with adverse outcome and treated hyperglycemia by administration of intravenous infu-
mortality. Furthermore, glucose variability also is associated sion insulin adjusted to maintain blood glucose between 80
with an increased risk of poor outcome. For example, Cueni- and110 mg/dL was associated with reduced mortality during
Villoz et al.7 observed that increased blood glucose variability intensive care (8% for conventional therapy vs. 4.6% for IIT)
during therapeutic hypothermia was associated with in-hos- especially among patients who were in the ICU more than 5
pital mortality after cardiac arrest, independent of injury days. Moreover, IIT reduced in-hospital mortality (10.9% for
severity and mean blood glucose levels. conventional care vs. 7.2% for IIT), bloodstream infection,
Consistent evidence from experimental and clinical studies renal failure, critical-illness polyneuropathy, and transfu-
demonstrates that hyperglycemia in the setting of acute neu- sions.1 The greatest reduction in mortality was observed for
rologic injury is associated with worse outcomes; this concept deaths associated with multiple-organ failure with a proven
is widely accepted in the management of critical neurologic septic focus. A follow-up MICU study2 (n = 1200 patients)
illness in adults as well as children.41,42 Hyperglycemia at showed IIT was associated with prevention of newly acquired
admission is present in almost half of acute stroke patients, kidney injury, accelerated weaning from mechanical ventila-
and is an independent risk factor for mortality at 30 days, 1 tion, and quicker discharge from the ICU and the hospital. In
year, and 6 years after stroke.33 Baird et  al. found that serum patients who received IIT, mortality was greater among those
glucose greater than 130  mg/dL during the first 72 hours who were in the ICU less than 3 days but significantly decreased
after a stroke was associated with significant increase in infarct in those patients with MICU stays longer than 3 days.2 The
size as measured by magnetic resonance imaging (MRI) 3 to mechanism by which IIT lowered mortality remains poorly
6 days after the ictus.34 Likewise, increased blood glucose is understood and probably involves both glycemic and non­
a risk factor for death and disability after aneurysmal sub- glycemic pathways. The potential nonglycemic benefits of
arachnoid hemorrhage (SAH).35,36,43 For example, McGirt insulin include myocardial protection, partial correction of
et  al. observed that patients with blood glucose greater than abnormal serum lipid profiles, and the prevention of excessive
200  mg/dL for 2 or more days were seven times more likely inflammation.50
124 Section II—Clinical and Laboratory Assessment

Table 14.2  Guidelines for Glycemic Control in the Intensive Care Unit
Updated Since
Patient Treatment Target Glucose Definition of NICE-SUGAR
Year Organization Population Threshold mg/dL Hypoglycemia Trial
2009 American Association of ICU patients 180 140-180 <70 Yes
Clinical Endocrinologists
and American Diabetes
Association
2009 Surviving Sepsis Campaign ICU patients 180 150 Not stated Yes
2009 Institute for Healthcare ICU patients 180 <180 <40 Yes
Improvement
2008 American Heart Association Acute 180 90-140 Not stated No
coronary
syndromes
2007 European Society of ICU patients Not stated Strict (refers to Not stated No
Cardiology and European with 80-110 mg/dL)
Association for the Study cardiac
of Diabetes disorders

Modified from Kavanagh BP, McCowen KC. Clinical practice: glycemic control in the ICU. N Engl J Med 2010;363(26):2540–6.
ICU, Intensive care unit.

However, other studies have not been able to duplicate the whom had isolated brain injury, Van den Berge et al. also
results of Van den Berghe et al.1,2 and in a meta-analysis that observed that IIT reduced both the incidence of critical illness
included 29 randomized trials with 8432 patients Wiener polyneuropathy and time on the ventilator.54 In the patients
et al.3 found that tight glucose control (IIT) increased the risk with isolated brain injury, 1-year functional outcomes were
of critical hypoglycemia in critically ill adult patients but did significantly improved in those patients in the IIT group,
not reduce hospital mortality. In turn, hypoglycemia increases despite the admission Glasgow Coma Scale (GCS) score being
the risk of mortality.5 Although IIT was popular in the years higher in the conventional insulin therapy group. For MICU
immediately before and after 2000, recommendations (Table patients who needed intensive care for at least 7 days, IIT
14.2) now target a blood glucose concentration between 140 reduced the incidence of critical illness myopathy and neu-
and 180 mg/dL (8-10 mmol/L) according to NICE-SUGAR, ropathy and was protective against prolonged mechanical ven-
published in 2009.51 In this randomized clinical trial 6104 tilation.55 Subsequent studies suggest glycemic control can
patients were randomized to IIT (target blood glucose help reduce critical illness polyneuropathy or myopathy,
81-108 mg/dL (4.5-6 mmol/L), or conventional glucose although this may depend on the diagnostic criteria used.56
control (target <180 mg/dL [<10 mmol/L]). Ninety-day mor- Several studies have focused on glucose control specifically
tality was significantly less in the conventional glucose control in patients with primary neurologic diseases (e.g., TBI and
group. In addition, severe hypoglycemia (<40 mg/dL) was rare stroke); overall it does not appear that IIT improves outcome,
when using conventional glucose control (0.5% vs. 6.8% in whereas its use is associated with more frequent episodes of
the IIT group). Other investigators also have observed that hypoglycemia that in some patients aggravates outcome.57-64
hypoglycemia is rare using the current American Association The UK Glucose Insulin in Stroke Trial (GIST) randomized
of Clinical Endocrinologists and American Diabetes Associa- patients with acute ischemic stroke to either conventional
tion (AACE/ADA) consensus recommendations that use a insulin therapy or IIT (72 and 126 mg/dL); 90-day mortality
blood glucose target of 140 to 180 mg/dL in the ICU.52 was similar in the two groups.57 Bilotta et al. randomized 78
However, there are differences, in particular glycemic control SAH patients to conventional or IIT. The patients randomized
superimposed on different nutritional strategies, between the to the IIT group had a significant decrease in infection but no
trials in Leuven1,2 and NICE-SUGAR that may limit direct difference in mortality or neurologic outcome.58 The same
comparison. Indeed it is conceivable that the safety and effi- researchers randomized 97 patients with severe TBI to either
cacy of IIT may be influenced by patient-related and ICU conventional treatment for glucose greater than 220 mg/dL or
setting–related variables. IIT for (80-120 mg/dL). The patients treated with IIT had
more episodes of hypoglycemia but rates of infection, poor
outcome, and mortality were similar in the two groups.59
Insulin Therapy in Neurocritical Care Two randomized phase II trials of IIT for acute ischemic
The potential mechanisms by which IIT is theorized to benefit stroke were completed in the United States. The Treatment of
the critically ill—attenuation of the cortisol stress response, Hyperglycemia in Ischemic Stroke trial aimed for glucose con-
improved immune cell function, reduced endothelial dysfunc- centrations less than 130 mg/dL in the tight control group,
tion, correction of abnormal lipids, prevention of toxicity to and less than 200 mg/L in the loose control group. In the
mitochondria, and reduced hyperglycemic axonal damage53— Glucose Regulation in Acute Stroke trial, the goals were 80 to
may be relevant in patients suffering from acute neurologic 120 mg/dL and 80 to 180 mg/dL, respectively. In both, the
illness. In a preplanned subanalysis of SICU patients, 63 of investigators were able to achieve their targets for tight glucose
Section II—Clinical and Laboratory Assessment 125

control without significant hypoglycemia or other major


adverse events.65,66 Beyond a Glucose Threshold
Other data in primary neurologic injury raise concerns There are several areas of research beyond just the threshold
about the effects of IIT on cerebral metabolism. Microdialysis at which to treat elevated blood glucose that are relevant to
studies in patients with severe TBI or SAH suggest that tight glycemic control: (1) treatment algorithms and patient safety
systemic glucose control is associated with reduced cerebral mechanisms (see also Chapter 6),75 (2) use of computerized
extracellular glucose and increased risk of brain energy decision support systems to deliver insulin or development
crisis.67-71 In addition, Vespa et al. reported that low extracel- of continuous monitors with automated closed-loop systems,
lular cerebral glucose after TBI was associated with poor (3) glucose variability, (4) whether target levels should differ
outcomes, even in the absence of increased microdialysate according to the indication for ICU admission or the stage of
lactate.68 Similarly in patients with severe SAH, Oddo et al.70 acute illness (i.e., a population-specific glycemic control strat-
observed that increased insulin dose independently predicted egy), and (5) the relationship between glucose, glycemic
brain energy crisis after adjustment for intracranial pressure control, and nutrition. In addition, attention to ICU processes
(ICP) and cerebral perfusion pressure (CPP) and that this in is important. For example, the risk of hypoglycemia associated
turn was associated with increased mortality. In addition, the with IIT is associated with nursing schedules and fatigue,76
relationship between brain interstitial and systemic glucose whereas when nurses override clinical decision support
concentrations is complex and may change when the systemic systems for IIT they tend to use lower-than-recommended
glucose concentration is greater than 180 mg/dL.72 Other doses to avoid hypoglycemia.77 Finally bioinformatics systems
microdialysis studies suggest administration of insulin could can be used to facilitate integration of glucose control and
impede the glucose supply of the brain73 and that brain energy nutrition.78
metabolic crisis (elevated lactate-to-pyruvate ratio) may occur Glucose variability, that is, changes in the measured level of
with acute reduction in serum glucose even when those levels blood glucose, may be of particular importance because it
are normal.74 Together these various data suggest that IIT in appears to be an independent or even casual factor associated
patients with already marginal serum glucose levels may be with adverse outcome.7,79-81 This in part may explain the
detrimental, and the optimal goal for a glucose level in the limited effect of IIT because IIT seems to increase glucose
setting of acute neurologic injury remains to be elucidated. variability.80 On the other hand IIT also is associated with
The available evidence suggests a less restrictive target for hypoglycemia, the severity of which is independently associ-
systemic glucose control (8-10 mmol/L; 144-180 mg/dL) may ated with increased mortality.80,82 However, there are no
be reasonable. Furthermore the data emphasize the impor- prospective studies that examine whether reduced glucose
tance of monitoring blood glucose and the effects of therapy variability is associated with better outcome, and so insulin
and that an approach that is perhaps more individualized to therapy is for the moment targeted to a glucose threshold
avoid both hypoglycemia and hyperglycemia is warranted rather than reduced glucose variability.
until specific glycemic control guidelines for patients with
severe brain injury in the neurocritical car unit (NCCU) are
better defined.43 An example of a continuous insulin protocol
Nutrition
is provided in Table 14.3. Since the 1990s, research has verified the importance of nutri-
tion in the critically ill. In addition to maintaining weight,
providing energy, and ensuring a positive nitrogen balance,
nutrition, particularly successful enteral nutrition, can help
Table 14.3  An Example of a Continuous augment immune function and is associated with fewer noso-
Insulin Infusion Therapy Protocol comial infections,83,84 reduced mortality,85 and improved neu-
rologic outcomes.86 However, it appears that the mean time to
 Regular insulin in 0.9% sodium chloride (1 U/mL) start enteral nutrition (EN) in critically ill patients is 3 days,87
administered through large-bore peripheral or central and average caloric intake is only 50% of American College of
venous catheter.
 The insulin infusion is adjusted based on every-hour
Chest Physicians (ACCP) goals.88 Despite national guidelines
Accu-Cheks to maintain target blood glucose. that support early and aggressive nutritional support,89,90
 Accu-Chek measurements performed every 2 hours when patients in the ICU are often underfed.
blood glucose is controlled for 8 hours or more and then Supplying adequate nutrition to those patients with severe
switch to subcutaneous insulin therapy. neurologic injury is particularly problematic. Many of those
Blood Glucose Initial Insulin Infusion admitted to the hospital are already malnourished, including
Level (mg/dL) (U/hr) one fifth of patients with acute stroke.91 Acute neurologic
>351 14 (plus 0.1 U/kg IV bolus) injury is associated with profound hypermetabolic and
hypercatabolic states. Patients therefore can lose nitrogen in
280-350 10
quantities sufficient to reduce weight by 15% per week.89
240-279 8 Nutritional status often deteriorates further because of poor
200-239 6 intake associated with reduced level of consciousness, dyspha-
170-199 4 gia, and weakness that limits the ability to self-feed. Therefore
optimizing nutritional status is an essential but often over-
140-169 3
looked aspect of neurocritical care. In addition, nutrition is
111-139 2 closely associated with glycemic control such that balanced
IV, Intravenous.
enteral nutrition may reduce the risk of hypoglycemia associ-
ated with IIT.8 For example, a meta-analysis suggests that IIT
126 Section II—Clinical and Laboratory Assessment

is associated with reduced mortality only when a high propor- residual volumes—retention, diarrhea, and aspiration—that
tion of calories is provided parenterally.9 Consistent with this may limit enteral access for nutrition. Moreover, EN may be
parenteral dextrose was an important source of energy in the interrupted by frequent displacement of access tubes, discon-
first Leuven study, which showed that IIT was associated with tinued feeds during incompatible medicine administration
reduced mortality.1 Furthermore, the rates of hypoglycemia1 and procedures, and inadequate ordering by staff. For
were less than those in NICE-SUGAR51 in which intravenous example, McClave et  al. observed that only 14% of patients
dextrose rarely was used and nutritional support was less reached 90% of goal feeding within 72 hours of the start
aggressive. Hence the benefit of IIT may depend on nutritional of EN.101
support or adequate calorie supply to avoid the hypoglycemia Successful tube feeding requires adequate gastric emptying,
of IIT. Consistent with this, microdialysis studies show that which is often impaired by the primary neurologic condition,
brain glucose is favorably influenced by enteral nutrition use of opiates, and limited mobility. A high residual volume is
without an adverse effect on other markers such as glutamate the most frequent cause to discontinue enteral tube feeding,87,102
or the lactate-to-pyruvate ratio.92 but the threshold volume for which tube feeds should be
stopped varies between practices. ACCP guidelines recom-
mend that a gastric residual greater than 150 mL should
Initiation and Optimization of Feeding prompt a decrease in the infusion rate, consideration of PN,
Studies that address the appropriate timing, amount, and or use of small bowel feeding.83 However, McClave et al.
method of feeding are difficult to generalize because of small studied the incidence of aspiration over a wide range of resid-
sample sizes, variable definitions of “early” and “late” feeding, ual volumes from 200 to 400 mL and found no relation
and inconsistent adherence to guidelines. Overall, the observa- between the residual volume and aspiration.102 The authors
tions support a recommendation of “early” enteral feeding.93-96 suggest that feeds should not be stopped when residual
Marik and Zaloga93 performed a meta-analysis of 15 random- volumes are less than 400 to 500 mL unless there are other
ized controlled trials that included 753 critically ill patients. signs of intolerance. A strict bowel regimen that includes pro-
Early feeding, defined as nutrition that commenced within 36 motility agents, stool softeners, and liberalization of activity
hours of admission, was associated with a 50% decrease in orders may help improve tolerance to enteral feeding.
infectious complications and a 2.2-day reduction in length of Both jejunal and gastric tubes are used routinely to provide
stay (LOS). The reduction in LOS was most pronounced in EN. Although the gastric route is associated with a higher
those patients with trauma, head injury, and burns. Rubinson incidence of high gastric residual volumes, it is safe and well
et al. followed 138 patients after MICU admission and found tolerated in most patients.103 Montejo studied the efficacy and
that failing to provide more than or equal to 25% of the rec- complication rate of gastric and jejunal feeding in 101 ran-
ommended calories was associated with a higher risk of domized patients. There were no differences in the incidence
bloodstream infection.94 Likewise, Villet et al. reported that of hospital-acquired pneumonia, length of stay, multiple-
in 48 critically ill patients, a negative energy balance was organ failure score, or mortality between groups, despite the
associated with an increased number of infectious and gastric feeding group having a significantly higher incidence
noninfectious complications.95 of high gastric residuals.87 Early data suggesting that TBI
Studies in neurocritical care also demonstrate an associa- patients benefit from jejunal over gastric feeding have been
tion between lack of nutrition and adverse outcomes. For criticized because the gastric group received no nutrition until
example, Hartl et al.97 evaluated the effect of early nutrition in bowel sounds returned, whereas the jejunal group received
nearly 800 patients with severe TBI. Patients who were not fed nutrition within 36 hours of admission.104 Furthermore,
within 5 days of injury had twice the incidence of death than gastric tubes are easy to place, and generally eliminate the need
those fed early. Every 10 kcal/kg decrease in caloric intake was for multiple radiographs to document correct placement.
associated with a 30% to 40% increase in mortality, even after Jejunal tubes have been linked to a higher caloric intake,105
controlling for hypotension, age, pupillary status, initial GCS, perhaps associated with decreased cessation of EN secondary
and computed tomography scan findings. Taylor et al. in a to high gastric residual volumes.
prospective study found that TBI patients randomized to a PN is advised when the enteral route is either unusable or
tube-feeding regimen that supplied full energy requirements unable to provide adequate calories. When PN is used, criti-
from the day of admission had nearly 25% fewer infections cally ill patients should be given a mixture that consists of
and decreased markers of inflammation than patients who amino acids (between 0.8 and 1.5 g/kg/day), carbohydrates
were started on tube feeds that were gradually increased to (about 60% of the nonprotein energy) and fat (about 40% of
goal.98 In patients with acute stroke, protein-energy malnutri- the nonprotein energy), electrolytes, and micronutrients.12
tion is associated with increased infection and worse outcome, Close monitoring is required when PN is used for (1) toler-
independent of age or baseline nutritional status.99 ance of electrolytes and macronutrients, (2) glycemia control,
and (3) specific micronutrients.106 PN may be used alone or
in combination with EN; therefore, even if the patient requires
Route of Acute Nutritional Delivery supplemental PN to meet calorie and nitrogen requirements,
There is limited information to recommend whether EN or the enteral route should be used unless it is contraindicated.
parenteral nutrition (PN) is superior for patients in the When PN and EN are used together, late (8 days) may be
NCCU. Many ICU practitioners were trained by the motto, preferable to early (48 hours) initiation of PN.107 A potential
“If the gut works, use it.” Lack of EN increases susceptibility advantage of PN is that nutritional goals may be reached more
to infection by impairing the delicate systems of mucosal quickly with PN, especially in trauma patients who may
immunity, gut cytokines, and endothelial markers of inflam- require 130% to 150% of predicted energy expenditure for
mation.100 However, EN may be associated with high gastric many weeks after injury.108 However, some studies suggest PN
Section II—Clinical and Laboratory Assessment 127

is associated with increased risk of infection, particularly hos-


pital-acquired candidemia,109 and increased cost. By contrast Table 14.4  ACCP Recommendations for
other studies suggest the risk of infection is not increased. For Nutrition in ICU Patients
example, Woodcock et al. evaluated 500 patients who received Total calories  25 total kilocalories per kilogram usual
adjuvant nutritional support and found no evidence that PN body weight per day
was associated with a higher incidence of infection.110 Simi-  1 mL of water per kilocalorie

larly, in a small study that evaluated PN and EN in 45 TBI Glucose  30%-70% of total calories per day
patients, Hadley et al. observed that infection was similar in
Fats  15%-30% of total calories per day
the two groups.111  7% of total calories administered as
Vasopressor use is considered a contraindication to EN. The omega-6 PUFA triglycerides
American Society of Parenteral and Enteral Nutrition (ASPEN)  Monitoring of triglyceride clearance is

2002 guidelines state that EN should be deferred until the recommended.


 Serum triglyceride levels above
patient is hemodynamically stable.10 However, vasopressors in 500 mg/dL should initiate consideration
the NCCU do not necessarily indicate hemodynamic instabil- for a decrease in total calories and/or
ity because they often are used to manage cerebral vasospasm omega-6 PUFA triglycerides.
or maintain cerebral perfusion pressure. Furthermore, the Protein  15%-20% of total calories per day
data that support the recommendation to limit EN during  Initiate therapy at 1.2 to 1.5 g/kg/day;
vasopressor use are based largely on expert opinions and con- consider increase dosing in patients
tradictory literature. On the other hand Doshi and Divgiovine with trauma or burns.
 BUN level exceeding 100 mg/dL should
found that early enteral feeding was associated with a 20% initiate consideration for a decrease in
decrease in ICU mortality and a 25% decrease in hospital dosing.
mortality among 1174 patients who required mechanical ven-  Nitrogen balance should be assessed

tilation and vasopressors.112 Similarly Berger et al. found that every 5-7 days and protein dosing
no patient with circulatory failure who received EN and adjusted accordingly.
required norepinephrine or dopamine after cardiac bypass Micronutrients  Potassium, magnesium, zinc, and
experienced any serious gastrointestinal complication.113 phosphate should be provided in doses
adequate to maintain normal serum
These data suggest that although EN should be used with levels.
caution in patients who are hypotensive, it is well tolerated
and likely of benefit to patients on vasopressors. ACCP, American College of Chest Physicians; BUN, blood urea nitrogen; ICU,
intensive care unit; PUFA, polyunsaturated fatty acid.

How to Determine Energy Needs


There are several methods to calculate calorie needs, but the Finland) can be used to perform indirect calorimetry with
best method to determine nutritional needs in hospitalized most commonly used ventilators. Indirect calorimetry is a
patients remains debated. There are multiple predictive equa- technique that measures oxygen consumption and carbon
tions, but these may over- or underestimate caloric needs.114 dioxide production to calculate resting energy expenditure
Indirect calorimetry may be a more useful measure of energy and respiratory quotient (RQ) and to measure the actual
needs in hospitalized patients. A metabolic cart, which mea- energy expenditure (measured energy expenditure [MEE]) of
sures the oxygen consumed and the carbon dioxide produced a patient. Because more than 95% of the energy generated
by the patient and then calculates the energy expenditure, is requires oxygen, there is a direct proportion between oxygen
felt to be particularly beneficial for patients who are obese, in consumption and the metabolic rate. One liter of oxygen con-
a pharmacologically induced coma, or being treated for neu- sumed generates 3.8 kcal (16.32 kJ): 1 L of carbon dioxide
rotrauma.115 General nutrient requirements for ICU patients produced generates 1.1 kcal (4.60 kJ). Ideally the patient
recommended by the ACCP are listed in Table 14.4.90 These should be assessed in a thermoneutral environment, at rest for
guidelines should be adapted to the specific disease state and more than 30 minutes, and without food for 2 hours for
baseline nutritional status of each individual patient. For precise measurements. A stress factor to account for injury is
example, a high-protein, hypocaloric regimen may be prefer- not necessary because the MEE accounts for the effects of the
able in the obese patient, although specific guidelines for obese disease. However, because the measurement is at rest, it is
patients are limited.116,117 This means understanding each necessary to multiply by an “activity” factor of 1 to 1.3,
patient’s premorbid nutritional state, basal metabolic rate, depending on whether the patient is intubated, at bed rest,
activity level, and stress factors that contribute to greater paralyzed, or out of bed.
energy needs.
Formulas to Determine Caloric Requirements
Indirect Calorimetry Instead of indirect calorimetry, which is not widely avail-
Assessment of nutritional status is multidimensional and able, there are a number of predictive equations that can
requires consideration of several factors, including nutrition be used to derive nutritional needs based on the degree of
status, nutritional intake, and energy expenditure. The refer- injury and patient weight among other variables.90 These
ence standard to measure energy expenditure and caloric various equations and nutritional status indices, however,
requirements in the ICU is indirect calorimetry.118 For example, may be less accurate than indirect calorimetry.119
the Deltatrac II MBM-200 Metabolic Monitor (Datex-Eng- The Maastricht Index (MI)120 and the Nutritional Risk
strom Division, Instrumentation Corp., Datex-Engstrom, Index (NRI),121 can be used to assess the nutritional status of
128 Section II—Clinical and Laboratory Assessment

a patient to provide a sense of the individual’s premorbid of the BMR. On the other hand excessive muscle activity such
condition. The MI is calculated as follows: as from excessive work of breathing or a combative disori-
ented state increases the energy expenditure.
 MI = [20.68 − (0.24 × albumin, g/L) − (19.21 ×
serum transthyretin, g/L) − (1.86 × lymphocytes, 106/L)
− (0.04 × ideal weight)] Stress-Induced Energy Needs
Nutritional status is graded as either malnourished or not The stress factor (SF) defines the hypermetabolism induced
malnourished with the MI; patients with a MI greater than by illness and is a multiple of the BMR (normalized to 1). The
0 are considered malnourished. Use of the MI is limited value takes into consideration the type of injury and the
to patients younger than 70 years of age.The NRI was devel- average metabolic response with this degree of injury. For
oped to assess malnutrition of surgical patients. There also example, an elective operation increases needs about 10% (SF
are geriatric-specific NRIs. = 1.1), major infection about 25% (SF = 1.25), and multiple
blunt trauma about 50% (SF = 1.5). Burn injury leads to the
 NRI = (1.519 × serum albumin, g/dL) +
greatest increase in metabolic demands.
(41.7 × present weight [kg]/ideal body weight [kg])
A value of NRI greater than100 indicates that the subject is Anthropometric Measurements
not malnourished, 97.5 to 100 indicates the subject is mildly Anthropometric measurements such as patients’ height and
malnourished, 83.5 to 97.5 indicates that the subject is mod- weight to calculate body mass index (BMI), triceps skinfold
erately malnourished, and NRI less than 83.5 indicates that thickness (TSF), and midarm circumference (MAC) to calcu-
the subject is severely malnourished. late muscle area among others may be used to help calculate
Basal metabolic rate (BMR) is a simple and accurate system energy and nutritional needs.
that can be used to estimate caloric needs is according to the
following formula:  BMI = weight (kg)/height m2
 Arm muscle area (mm2) = (MAC [mm] −
 Energy requirements = BMR ×
[TSF —3.14])2/4π
activity level (usually 1.25) × stress factor
BMR describes the energy required to maintain cell integrity A BMI less than 18.5 is underweight; a BMI between 25 and
in the resting state and at thermoneutrality. A significant 29.9 is overweight; a BMI greater than or equal to 30 is obese,
portion of energy expenditure goes into thermogenesis, the and a BMI greater than or equal to 40 is morbidly obese. These
maintenance of normal body temperature through heat pro- values may need to be factored in when calculating calorie and
duction. Thermoneutrality usually is close to 80° F [28°  C] protein needs.
when the heat loss and the need for increased heat produc-
tion to maintain the body temperature are minimal.  The Biochemical Measurements
BMR for man has been defined by direct calorimetry. Body While in the ICU, biochemical measurements including serum
size is a key factor, and so BMR is expressed in terms of albumin and prealbumin, transferrin, retinol binding protein,
square meters of body surface or body weight. Other vari- magnesium, phosphorus, total lymphocyte count (TLC), and
ables include age and sex. The average adult BMR value is a 24-hour urine collection to measure urine urea nitrogen
1500 to 1800  kcal per day (850 to 950  kcal/m2/day or 20 to (UUN) and creatinine can help guide nutritional therapy. The
24  kcal/kg/day). creatinine height index is calculated from 24-hour urinary
The Harris-Benedict equation can be used to calculate creatinine excretion.123 The nitrogen balance is derived from
resting energy expenditure (REE).122 the daily volume of infusate minus the daily output of urinary
nitrogen:
 For men, REE = 66.5 + (13.75 × weight [kg]) +
(5.003 × height [cm]) − (6.775 × age [yr])  Nitrogen balance (g/day) = protein intake (grams)/6.25 −
 Or, REE = 65 + (6.2 × weight [lb]) +
(UUN + 4g obligatory loss)
(12.7 × height [in]) − (6.8 × age [yr])  Or, nitrogen balance = nitrogen intake − nitrogen output
 For women, REE = 655.1 + (9.563 × weight [kg]) +
(1.850 × height [cm]) − (4.676 × age [yr])
UUN is measured in grams of nitrogen excreted in the
 Or, REE = 655 + (4.3 × weight [lb]) +
urine over a 24-hour period. The following values are con-
(4.7 × height [in]) − (4.7 × age [yr])
sidered normal (although this may vary between laborato-
A variety of multiplication factors (activity, stress, tempera- ries): serum albumin, 3.5 to 5  g/dL; prealbumin, 18 to
ture that range from 1.2 to >2) are introduced to calculate 43mg/dL; total lymphocyte count (TLC) greater than 1500/
adequate calorie intake. mm3; and creatinine height index greater than 90%.
Patients admitted to the ICU should be weighed on admis-
 Total caloric requirements = REE × by the sum of the stress
sion and then at least twice weekly. Ideal body weights (IBWs)
and activity factors, for example, total calorie require-
then can be used to calculate caloric goals:
ments/day = 1.25 × REE. For each 1° C more than 37° C
add 10% extra allowance.  IBW males = 106 lb for 5'0" + 6 lb/inch >5'
The activity level reflects energy used by muscles for average (or IBW = 50 kg + 2.3 kg for each inch >5 feet)
daily activity. In the ICU, patient activity is usually limited and  IBW females = 100 lb for 5'0" + 5 lb/inch >5'
is usually considered to be no greater than an additional 25% (or IBW = 45.5 kg + 2.3 kg for each inch over 5 feet)
Section II—Clinical and Laboratory Assessment 129

18. Leino A, Kurvinen K. Interchangeability of blood gas, electrolyte and


Conclusion metabolite results measured with point-of-care, blood gas and core
laboratory analyzers. Clin Chem Lab Med 2011;49(7):1187–91. Epub 2011,
Initial caloric goals are 25 to 35 kcal/kg of IBW. An exception Apr 20.
may be patients with spinal cord injury who are paralyzed and 19. Penfornis A, Personeni E, Borot S. Evolution of devices in diabetes
who may require fewer initial calories that are inversely related management. Diabetes Technol Ther 2011;13(Suppl 1):S93–102.
to the level of injury (i.e., the higher the lesion, the lower the 20. Shearer A, Boehmer M, Closs M, et al. Comparison of glucose point-of-care
values with laboratory values in critically ill patients. Am J Crit Care
energy expenditure). The protein requirement is 1.5 to 1.8 g/ 2009;18(3):224–30.
kg/day using IBW for all patients. However, protein require- 21. Roth-Kleiner M, Stadelmann Diaw C, Urfer J, et al. Evaluation of different
ments are altered by conditions such as acute pancreatitis and POCT devices for glucose measurement in a clinical neonatal setting.
hepatic encephalopathy in which protein is restricted to 0.6 g/ Eur J Pediatr 2010;169(11):1387–95. Epub 2010, Jun 24.
22. Meynaar IA, van Spreuwel M, Tangkau PL, et al. Accuracy of AccuChek
kg/day for 3 days and then increased to less than or equal to
glucose measurement in intensive care patients. Crit Care Med 2009;37(10):
1.2 g/kg/day or reduced if the mental status deteriorates. In 2691–6.
addition, protein needs may need to be modified for renal 23. Egi M, Finfer S, Bellomo R. Glycemic control in the ICU. Chest
failure (creatinine [Cr] >2.5). For example, the initial protein 2011;140(1):212–20.
intake may need to be reduced to 1 to 1.3 g/kg/day and the 24. Ichai C, Preiser JC, Société Française d’Anesthésie-Réanimation, et al.
International recommendations for glucose control in adult non diabetic
daily blood urea nitrogen (BUN) followed. If the patient is critically ill patients. Crit Care 2010;14(5):R166. Epub 2010, Sep 14.
oliguric, protein intake may need to be started at less than or 25. Rooyackers O, Blixt C, Mattsson P, Wernerman J. Continuous glucose
equal to 0.6 g/kg/day and the daily BUN followed. Specific monitoring by intravenous microdialysis. Acta Anaesthesiologica
enteral nutritional supplements are available for these various Scandinavica 2010;54(7):841–7.
26. Möller F, Liska J, Franco-Cereceda A. Evaluation of a continuous blood
conditions (e.g., Magnacal, Peptinex, NutriHep).
glucose monitoring system using central venous microdialysis. Crit Care
2011;15(Suppl 1):P405. Published online 2011 March 11. doi: 10.1186/
References cc9825. PMCID: PMC3068334.
27. Dungan KM, Braithwaite SS, Preiser JC. Stress hyperglycaemia. Lancet
1. Van den Berghe G, Wouters P, Weekers F, et al. Intensive insulin therapy in 2009;373:1798–807.
critically ill patients. N Engl J Med 2001;345:1359–67. 28. Farrokhi F, Smiley D, Umpierrez GE. Glycemic control in non-diabetic
2. Van den Berghe G, Wilmer A, Hermans G, et al. Intensive insulin therapy in critically ill patients. Best Pract Res Clin Endocrinol Metab
medical intensive care patients. N Engl J Med 2006;354:449–61. 2011;25(5):813–24.
3. Wiener RS, Wiener DC, Larson RJ. Benefits and risks of tight glucose control 29. Egi M, Bellomo R, Stachowski E, et al. Blood glucose concentration and
in critically ill adults: a meta-analysis. JAMA 2008;300(8):933–44. outcome of critical illness: the impact of diabetes. Crit Care Med 2008;36:
4. Jacka MJ, Torok-Both CJ, Bagshaw SM. Blood glucose control among 2249–55.
critically ill patients with brain injury. Can J Neurol Sci 2009;36(4): 30. Falciglia M, Freyberg RW, Almenoff PL, et al. Hyperglycemia-related
436–42. mortality in critically ill patients varies with admission diagnosis.
5. Krinsley JS, Schultz MJ, Spronk PE, et al. Mild hypoglycemia is Crit Care Med 2009;37(12):3001–9.
independently associated with increased mortality in the critically ill. 31. Siegelaar SE, Hoekstra JB, DeVries JH. Special considerations for the diabetic
Crit Care 2011;15(4):R173. patient in the ICU: targets for treatment and risks of hypoglycaemia.
6. Duning T, van den Heuvel I, Dickmann A, et al. Hypoglycemia aggravates Best Pract Res Clin Endocrinol Metab 2011;25(5):825–34.
critical illness-induced neurocognitive dysfunction. Diabetes Care 2010; 32. Egi M, Bellomo R, Stachowski E, et al. The interaction of chronic and acute
33(3):639–44. Epub 2009, Dec 23. glycemia with mortality in critically ill patients with diabetes. Crit Care Med
7. Cueni-Villoz N, Devigili A, Delodder F, et al. Increased blood glucose 2011;39(1):105–11.
variability during therapeutic hypothermia and outcome after cardiac arrest. 33. Williams LS, Rotich J, Qi R, et al. Effects of admission hyperglycemia on
Crit Care Med 2011;39(10):2225–31. mortality and costs in acute ischemic stroke. Neurology 2002;59:67–71.
8. Kauffmann RM, Hayes RM, Jenkins JM, et al. Provision of balanced 34. Baird TA, Parsons MW, Phanh T, et al. Persistent post stroke hyperglycemia
nutrition protects against hypoglycemia in the critically ill surgical patient. is independently associated with infarct expansion and worse clinical
JPEN J Parenter Enteral Nutr 2011;35(6):686–94. Epub 2011, Jul 12. outcomes. Stroke 2003;34:2208–14.
9. Marik PE, Preiser JC. Toward understanding tight glycemic control in the 35. Juvela S, Siironen J, Kuhmonen J. Hyperglycemia, excess weight, and history
ICU: a systematic review and metaanalysis. Chest 2010;137:544–51. of hypertension as risk factors for poor outcome and cerebral infarction
10. ASPEN Board of Directors and the Clinical Guidelines Task Force. after aneurysmal subarachnoid hemorrhage. J Neurosurg
Guidelines for the use of parenteral and enteral nutrition in adult and 2005;102(6):998–1003.
pediatric patients. J Parenter Enteral Nutr 2002;26S:1SA–138SA. 36. Frontera JA, Fernandez A, Claassen J, et al. Hyperglycemia after SAH:
11. Kreymann KG, Berger MM, Deutz NE, et al. ESPEN guidelines on enteral predictors, associated complications, and impact on outcome. Stroke
nutrition: intensive care. Clin Nutr 2006;25:210–23. 2006;37(1):199–203.
12. Kreymann G, Adolph M, Druml W, et al. Intensive medicine: guidelines on 37. McGirt MJ, Woodworth GF, Ali M, et al. Persistent perioperative
parenteral nutrition. Ger Med Sci 2009;7:doc14. hyperglycemia as an independent predictor of poor outcome after
13. Berger MM, Chiolero RL. Hypocaloric feeding: pros and cons. Curr Opin aneurysmal subarachnoid hemorrhage. J Neurosurg 2007;107(6):1080–5.
Crit Care 2007;13:180–6. 38. Badjatia N, Topcuoglu MA, Buonanno FS, et al. Relationship between
14. Heyland DK, Dhaliwal R, Drover JW, et al. Canadian clinical practice hyperglycemia and symptomatic vasospasm alter subarachnoid hemorrhage.
guidelines for nutrition support in mechanically ventilated, critically ill adult Crit Care Med 2005;33:1603–9.
patients. JPEN J Parenter Enteral Nutr 2003;27:355–73. 39. Worthley MI, Shrive FM, Anderson TJ, et al. Prognostic implication of
15. McClave SA, Martindale RG, Vanek VW, et al. Guidelines for the provision hyperglycemia in myocardial infarction and primary angioplasty. Am J Med
and assessment of nutrition support therapy in the adult critically ill patient: 2007;120(7):643.e1–e7.
Society of Critical Care Medicine (SCCM) and American Society for 40. Fogelholm R, Murros K, Rissanen A, et al. Admission blood glucose and
Parenteral and Enteral Nutrition (ASPEN). JPEN J Parenter Enteral Nutr short term survival in primary intracerebral haemorrhage: a population
2009;33(3):277–316. based study. J Neurol Neurosurg Psychiatry 2005;76:349–53.
16. Juneja D, Pandey R, Singh O. Comparison between arterial and capillary 41. Salim A, Hadjizacharia P, Dubose J, et al. Persistent hyperglycemia in severe
blood glucose monitoring in patients with shock. Eur J Intern Med traumatic brain injury: an independent predictor of outcome. Am Surg
2011;22(3):241–4. Epub 2011, Feb 16. 2009;75(1):25–9.
17. Corstjens AM, Ligtenberg JJ, van der Horst IC, et al. Accuracy and feasibility 42. Smith RL, Lin JC, Adelson PD, et al. Relationship between hyperglycemia
of point-of-care and continuous blood glucose analysis in critically ill ICU and outcome in children with severe traumatic brain injury. Pediatr Crit
patients. Crit Care 2006;10(5):R135. Care Med 2012;13(1):85–91.
130 Section II—Clinical and Laboratory Assessment

43. Schmutzhard E, Rabinstein AA, Participants in the International 47. McCormick MT, Muir KW, Gray CS, et al. Management of hyperglycemia in
Multi-Disciplinary Consensus Conference on the Critical Care Management acute stroke: how, when, and for whom? Stroke 2008;39:2177–85.
of Subarachnoid Hemorrhage. Spontaneous subarachnoid hemorrhage and 48. Oddo M, Levine JM, Frangos S, et al. Brain lactate metabolism in humans
glucose management. Neurocrit Care 2011;15(2):281–6. with subarachnoid hemorrhage. Stroke 2012. Epub ahead of print.
44. Dumont T, Rughani A, Silver J, et al. Diabetes mellitus increases risk of 49. Wyss MT, Jolivet R, Buck A, et al.. In vivo evidence for lactate as a neuronal
vasospasm following aneurysmal subarachnoid hemorrhage independent of energy source. J Neurosci 2011;31(20):7477–85.
glycemic control. Neurocrit Care 2009;11(2):183–9. Epub 2009, May 27. 50. Vanhorebeek I, Langouche L, Van den Berghe G. Glycemic and nonglycemic
45. Vogelzang M, Nijboer JMM, van der Horst ICC, et al. Hyperglycemia has a effects of insulin: how do they contribute to a better outcome of critical
stronger relation with outcome in trauma patients than in other critically ill illness? Curr Opin Crit Care 2005;11:304–11.
patients. J Trauma 2006;60:873–9.
46. Jeremitsky E, Omert LA, Dunham M, et al. The impact of hyperglycemia on A complete list of references for this chapter can be found online at
patients with severe brain injury. J Trauma 2005;58:47–50. www.expertconsult.com.
Section II—Clinical and Laboratory Assessment 130.e1

References 27. Dungan KM, Braithwaite SS, Preiser JC. Stress hyperglycaemia. Lancet
2009;373:1798–807.
1. Van den Berghe G, Wouters P, Weekers F, et al. Intensive insulin therapy in 28. Farrokhi F, Smiley D, Umpierrez GE. Glycemic control in non-diabetic
critically ill patients. N Engl J Med 2001;345:1359–67. critically ill patients. Best Pract Res Clin Endocrinol Metab
2. Van den Berghe G, Wilmer A, Hermans G, et al. Intensive insulin therapy 2011;25(5):813–24.
in medical intensive care patients. N Engl J Med 2006;354:449–61. 29. Egi M, Bellomo R, Stachowski E, et al. Blood glucose concentration and
3. Wiener RS, Wiener DC, Larson RJ. Benefits and risks of tight glucose outcome of critical illness: the impact of diabetes. Crit Care Med
control in critically ill adults: a meta-analysis. JAMA 2008;300(8):933–44. 2008;36:2249–55.
4. Jacka MJ, Torok-Both CJ, Bagshaw SM. Blood glucose control among 30. Falciglia M, Freyberg RW, Almenoff PL, et al. Hyperglycemia-related
critically ill patients with brain injury. Can J Neurol Sci 2009;36(4): mortality in critically ill patients varies with admission diagnosis.
436–42. Crit Care Med 2009;37(12):3001–9.
5. Krinsley JS, Schultz MJ, Spronk PE, et al. Mild hypoglycemia is 31. Siegelaar SE, Hoekstra JB, DeVries JH. Special considerations for the
independently associated with increased mortality in the critically ill. Crit diabetic patient in the ICU: targets for treatment and risks of
Care 2011;15(4):R173. hypoglycaemia. Best Pract Res Clin Endocrinol Metab 2011;25(5):
6. Duning T, van den Heuvel I, Dickmann A, et al. Hypoglycemia aggravates 825–34.
critical illness-induced neurocognitive dysfunction. Diabetes Care 2010; 32. Egi M, Bellomo R, Stachowski E, et al. The interaction of chronic and acute
33(3):639–44. Epub 2009, Dec 23. glycemia with mortality in critically ill patients with diabetes. Crit Care
7. Cueni-Villoz N, Devigili A, Delodder F, et al. Increased blood glucose Med 2011;39(1):105–11.
variability during therapeutic hypothermia and outcome after cardiac 33. Williams LS, Rotich J, Qi R, et al. Effects of admission hyperglycemia on
arrest. Crit Care Med 2011;39(10):2225–31. mortality and costs in acute ischemic stroke. Neurology 2002;59:67–71.
8. Kauffmann RM, Hayes RM, Jenkins JM, et al. Provision of balanced 34. Baird TA, Parsons MW, Phanh T, et al. Persistent post stroke hyperglycemia
nutrition protects against hypoglycemia in the critically ill surgical patient. is independently associated with infarct expansion and worse clinical
JPEN J Parenter Enteral Nutr 2011;35(6):686–94. Epub 2011, Jul 12. outcomes. Stroke 2003;34:2208–14.
9. Marik PE, Preiser JC. Toward understanding tight glycemic control in the 35. Juvela S, Siironen J, Kuhmonen J. Hyperglycemia, excess weight, and history
ICU: a systematic review and metaanalysis. Chest 2010;137:544–51. of hypertension as risk factors for poor outcome and cerebral infarction
10. ASPEN Board of Directors and the Clinical Guidelines Task Force. after aneurysmal subarachnoid hemorrhage. J Neurosurg
Guidelines for the use of parenteral and enteral nutrition in adult and 2005;102(6):998–1003.
pediatric patients. J Parenter Enteral Nutr 2002;26S:1SA–138SA. 36. Frontera JA, Fernandez A, Claassen J, et al. Hyperglycemia after SAH:
11. Kreymann KG, Berger MM, Deutz NE, et al. ESPEN guidelines on enteral predictors, associated complications, and impact on outcome. Stroke
nutrition: intensive care. Clin Nutr 2006;25:210–23. 2006;37(1):199–203.
12. Kreymann G, Adolph M, Druml W, et al. Intensive medicine: guidelines on 37. McGirt MJ, Woodworth GF, Ali M, et al. Persistent perioperative
parenteral nutrition. Ger Med Sci 2009;7:doc14. hyperglycemia as an independent predictor of poor outcome after
13. Berger MM, Chiolero RL. Hypocaloric feeding: pros and cons. Curr Opin aneurysmal subarachnoid hemorrhage. J Neurosurg 2007;107(6):1080–5.
Crit Care 2007;13:180–6. 38. Badjatia N, Topcuoglu MA, Buonanno FS, et al. Relationship between
14. Heyland DK, Dhaliwal R, Drover JW, et al. Canadian clinical practice hyperglycemia and symptomatic vasospasm alter subarachnoid
guidelines for nutrition support in mechanically ventilated, critically ill hemorrhage. Crit Care Med 2005;33:1603–9.
adult patients. JPEN J Parenter Enteral Nutr 2003;27:355–73. 39. Worthley MI, Shrive FM, Anderson TJ, et al. Prognostic implication of
15. McClave SA, Martindale RG, Vanek VW, et al. Guidelines for the provision hyperglycemia in myocardial infarction and primary angioplasty. Am J Med
and assessment of nutrition support therapy in the adult critically ill 2007;120(7):643.e1–e7.
patient: Society of Critical Care Medicine (SCCM) and American Society 40. Fogelholm R, Murros K, Rissanen A, et al. Admission blood glucose and
for Parenteral and Enteral Nutrition (ASPEN). JPEN J Parenter Enteral short term survival in primary intracerebral haemorrhage: a population
Nutr 2009;33(3):277–316. based study. J Neurol Neurosurg Psychiatry 2005;76:349–53.
16. Juneja D, Pandey R, Singh O. Comparison between arterial and capillary 41. Salim A, Hadjizacharia P, Dubose J, et al. Persistent hyperglycemia in severe
blood glucose monitoring in patients with shock. Eur J Intern Med traumatic brain injury: an independent predictor of outcome. Am Surg
2011;22(3):241–4. Epub 2011, Feb 16. 2009;75(1):25–9.
17. Corstjens AM, Ligtenberg JJ, van der Horst IC, et al. Accuracy and 42. Smith RL, Lin JC, Adelson PD, et al. Relationship between hyperglycemia
feasibility of point-of-care and continuous blood glucose analysis in and outcome in children with severe traumatic brain injury. Pediatr Crit
critically ill ICU patients. Crit Care 2006;10(5):R135. Care Med 2012;13(1):85–91.
18. Leino A, Kurvinen K. Interchangeability of blood gas, electrolyte and 43. Schmutzhard E, Rabinstein AA. Participants in the International
metabolite results measured with point-of-care, blood gas and core Multi-Disciplinary Consensus Conference on the Critical Care
laboratory analyzers. Clin Chem Lab Med 2011;49(7):1187–91. Epub 2011, Management of Subarachnoid Hemorrhage. Spontaneous
Apr 20. subarachnoid hemorrhage and glucose management. Neurocrit Care
19. Penfornis A, Personeni E, Borot S. Evolution of devices in diabetes 2011;15(2):281–6.
management. Diabetes Technol Ther 2011;13(Suppl 1):S93–102. 44. Dumont T, Rughani A, Silver J, et al. Diabetes mellitus increases risk of
20. Shearer A, Boehmer M, Closs M, et al. Comparison of glucose point-of- vasospasm following aneurysmal subarachnoid hemorrhage independent of
care values with laboratory values in critically ill patients. Am J Crit Care glycemic control. Neurocrit Care 2009;11(2):183–9. Epub 2009, May 27.
2009;18(3):224–30. 45. Vogelzang M, Nijboer JMM, van der Horst ICC, et al. Hyperglycemia has a
21. Roth-Kleiner M, Stadelmann Diaw C, Urfer J, et al. Evaluation of different stronger relation with outcome in trauma patients than in other critically
POCT devices for glucose measurement in a clinical neonatal setting. ill patients. J Trauma 2006;60:873–9.
Eur J Pediatr 2010;169(11):1387–95. Epub 2010, Jun 24. 46. Jeremitsky E, Omert LA, Dunham M, et al. The impact of hyperglycemia
22. Meynaar IA, van Spreuwel M, Tangkau PL, et al. Accuracy of AccuChek on patients with severe brain injury. J Trauma 2005;58:47–50.
glucose measurement in intensive care patients. Crit Care Med 2009; 47. McCormick MT, Muir KW, Gray CS, et al. Management of hyperglycemia
37(10):2691–6. in acute stroke: how, when, and for whom? Stroke 2008;39:2177–85.
23. Egi M, Finfer S, Bellomo R. Glycemic control in the ICU. Chest 2011; 48. Oddo M, Levine JM, Frangos S, et al. Brain lactate metabolism in humans
140(1):212–20. with subarachnoid hemorrhage. Stroke 2012; Epub ahead of print.
24. Ichai C, Preiser JC, Société Française d’Anesthésie-Réanimatio, et al. 49. Wyss MT, Jolivet R, Buck A, et al. In vivo evidence for lactate as a neuronal
International recommendations for glucose control in adult non diabetic energy source. J Neurosci 2011;31(20):7477–85.
critically ill patients. Crit Care 2010;14(5):R166. Epub 2010, Sep 14. 50. Vanhorebeek I, Langouche L, Van den Berghe G. Glycemic and nonglycemic
25. Rooyackers O, Blixt C, Mattsson P, Wernerman J. Continuous glucose effects of insulin: how do they contribute to a better outcome of critical
monitoring by intravenous microdialysis. Acta Anaesthesiologica illness? Curr Opin Crit Care 2005;11:304–11.
Scandinavica 2010;54(7):841–7. 51. NICE-SUGAR Study Investigators, Finfer S, Chittock DR, et al. Intensive
26. Möller F, Liska J, Franco-Cereceda A. Evaluation of a continuous blood versus conventional glucose control in critically ill patients. N Engl J Med
glucose monitoring system using central venous microdialysis. Crit Care 2009;360(13):1283–97. Epub 2009, Mar 24.
2011;15(Suppl 1):P405. Published online 2011 March 11. doi: 10.1186/ 52. Shetty S, Inzucchi SE, Goldberg PA, et al. Adapting to the New Consensus
cc9825. PMCID: PMC3068334. Guidelines for Managing Hyperglycemia During Critical Illness: the
130.e2 Section II—Clinical and Laboratory Assessment

Updated Yale Insulin Infusion Protocol. Endocr Pract 2011;2:1–19. Epub 76. Louie K, Cheema R, Dodek P, et al. Intensive nursing work schedules and
ahead of print. the risk of hypoglycaemia in critically ill patients who are receiving
53. Van den Berghe G. How does blood glucose control with insulin save lives intravenous insulin. Qual Saf Health 2010;19(6):e42. Epub 2010,
in intensive care? J Clin Invest 2004;114:1187–95. Aug 4.
54. Ven den Berghe G, Schoonheydt K, Becx P, et al. Insulin therapy protects 77. Campion TR Jr, May AK, Waitman LR, et al. Characteristics and effects of
the central and peripheral nervous system of intensive care patients. nurse dosing over-rides on computer-based intensive insulin therapy
Neurology 2005;64:1348–53. protocol performance. J Am Med Inform Assoc 2011;18(3):251–8. Epub
55. Hermans G, Wilmer A, Meersseman W, et al. Impact of intensive insulin 2011, Mar 14.
therapy on neuromuscular complications and ventilator dependency in 78. Berger MM, Que YA. Bioinformatics assistance of metabolic and nutrition
the medical intensive care unit. Am J Respir Crit Care Med 2007;175: management in the ICU. Curr Opin Clin Nutr Metab Care 2011;14(2):
480–98. 202–8.
56. Wieske L, Harmsen RE, Schultz MJ, et al. Is critical illness neuromyopathy 79. Krinsley JS. Glycemic variability: a strong independent predictor of
and duration of mechanical ventilation decreased by strict glucose control? mortality in critically ill patients. Crit Care Med 2008;36:3008–13.
Neurocrit Care 2011;14(3):475–81. 80. Meyfroidt G, Keenan DM, Wang X, et al. Dynamic characteristics of blood
57. Gray CS, Hildreth AJ, Sandercock PA, et al. Glucose-potassium-insulin glucose time series during the course of critical illness: effects of intensive
infusions in the management of post-stroke hyperglycemia: the UK Glucose insulin therapy and relative association with mortality. Crit Care Med
Insulin in Stroke Trial (GIST-UK). Lancet Neurol 2007;6:397–406. 2010;38:1021–9.
58. Bilotta F, Spinelli A, Giovannini F, et al. The effect of intensive insulin 81. Matsushima K, Peng M, Velasco C, et al. Glucose variability negatively
therapy ion infection rate, vasospasm, neurologic outcome, and mortality impacts long-term functional outcome in patients with traumatic brain
in the neurointensive care unit after intracranial aneurysm clipping in injury. J Crit Care 2011. Epub ahead of print.
patients with acute subarachnoid hemorrhage: a randomized prospective 82. Egi M, Bellomo R, Stachowski E, et al. Hypoglycemia and outcome in
pilot trial. J Neurosurg Anesthesiol 2007;19:156–60. critically ill patients. Mayo Clin Proc 2010;85:217–24.
59. Bilotta F, Caramia R, Cernak I, et al. Intensive insulin therapy after severe 83. Rubinson L, Diette GB, Song X, et al. Low caloric intake is associated with
traumatic brain injury: a randomized clinical trial. Neurocrit Care nosocomial blood stream infections in patients in the medical intensive
2008;9(2):159–66. care unit. Crit Care Med 2004;(32):350–7.
60. Green DM, O’Phelan KH, Bassin SL, et al. Intensive versus conventional 84. Heyland DK, Stephens KE, Day AG, et al. The success of enteral nutrition
insulin therapy in critically ill neurologic patients. Neurocrit Care and ICU-acquired infections: a multicenter observational study. Clin Nutr
2010;13(3):299–306. 2011;30(2):148–55. Epub 2010, Oct 25.
61. Graffagnino C, Gurram AR, Kolls B, et al. Intensive insulin therapy in the 85. Serón-Arbeloa C, Puzo-Foncillas J, Garcés-Gimenez T, et al. A retrospective
neurocritical care setting is associated with poor clinical outcomes. study about the influence of early nutritional support on mortality and
Neurocrit Care 2010;13(3):307–12. nosocomial infection in the critical care setting. Clin Nutr 2011;30(3):
62. Tiemessen CA, Hoedemaekers CW, van Iersel FM, et al. Intensive insulin 346–50. Epub 2010, Dec 4.
therapy increases the risk of hypoglycemia in neurocritical care patients. 86. Hartl R, Gerber LM, Ni Q, et al. Effect of early nutrition on deaths due to
J Neurosurg Anesthesiol 2011;23(3):206–14. severe traumatic brain injury. J Neurosurg 2008;109:50–6.
63. Shan L, Hao PP, Chen YG. Efficacy and safety of intensive insulin therapy 87. Montejo JC. Enteral nutrition-related gastrointestinal complications in
for critically ill neurologic patients: a meta-analysis. J Trauma critically ill patients: a multicenter study. Crit Care Med 1999;27:
2011;71(5):1460–4. 1447–53.
64. Coester A, Neumann CR, Schmidt MI. Intensive insulin therapy in severe 88. Krishnan JA, Parce PB, Martinz A, et al. Caloric intake in medical ICU
traumatic brain injury: a randomized trial. J Trauma 2010;68(4):904–11. patients: consistency of care with guidelines and relationship to clinical
65. Bruno A, Kent TA, Coull BM, et al. Treatment of hyperglycemia in ischemic outcomes. Chest 2003;124:297–305.
stroke (THIS): a randomized pilot trial. Stroke 2008;39:384–9. 89. Brain Trauma Foundation, American Association of Neurological Surgeons,
66. Johnston KC, Hall CL, Kissela BM, et al. Glucose Regulation in Acute Congress of Neurological Surgeons. Guidelines for the management of
Stroke Patients (GRASP) trial: a randomized pilot trial. Stroke severe traumatic brain injury. Nutrition J Neurotrauma 2007;24(Suppl 1):
2009;40(12):3804–9. S77–82.
67. Graetz D, Nagel A, Schlenk F, et al. Role of hyperglycemia and insulin 90. Cerra F, Benitez MR, Blackburn, et al. Applied nutrition in ICU patients:
treatment for cerebral metabolism in aneurysmal SAH. Meeting abstract. a consensus statement of the American College of Chest Physicians. Chest
59th Annual Meeting of the German Society of Neurosurgery, Wurzberg, 1997;111:769–78.
Germany, 2008. 91. Dennis M. Nutrition after stroke. Br Med Bull 2000;56:466–75.
68. Vespa PM, McArthur D, O’Phelan K, et al. Persistently low extracellular 92. Kinoshita K, Moriya T, Utagawa A, et al. Change in brain glucose after
glucose correlates with poor outcome 6 months after human traumatic enteral nutrition in subarachnoid hemorrhage. J Surg Res 2010;162(2):
brain injury despite a lack of increased lactate: a microdialysis study. 221–4. Epub 2009, Sep 22.
J Cerebral Blood Flow Metab 2003;23:865–77. 93. Marik PE, Zaloga GP. Early enteral nutrition in acutely ill patients:
69. Vespa P, Boonyaputthikul R, McArthur DL, et al. Intensive insulin therapy a systematic review. Crit Care Med 2001;29:2264–70.
reduces microdialysis glucose values without altering glucose utilization or 94. Rubinson L, Diette GB, Song X, et al. Low caloric intake is associated with
improving the lactate/pyruvate ratio after traumatic brain injury. Crit Care nosocomial bloodstream infections in patients in the medical intensive care
Med 2006;34:850–6. unit. Crit Care Med 2004;32:350–7.
70. Oddo M, Schmidt JM, Carrera C, et al. Impact of tight glycemic control on 95. Villet S, Chiolero RL, Bollmann MD, et al. Negative impact of hypocaloric
cerebral glucose metabolism after severe brain injury: a microdialysis study. feeding and energy balance on clinical outcome in ICU patients. Clin Nutr
Crit Care Med 2008;36:3233–8. 2005;24:502–9.
71. Schlenk F, Vajkoczy P, Sarrafzadeh A. Inpatient hyperglycemia following 96. Nguyen NQ, Besanko LK, Burgstad C, et al. Delayed enteral feeding impairs
aneurysmal subarachnoid hemorrhage: relation to cerebral metabolism and intestinal carbohydrate absorption in critically ill patients. Crit Care Med
outcome. Neurocrit Care 2009;11(1):56–63. Epub 2009, May 6. 2012;40(1):50–4.
72. Liogier LE, Macyszyn L, Kosty JA, et al. Brain and blood glucose after 97. Hartl R, Gerber LM, Ni Q, et al. Effect of early nutrition on deaths due to
subarachnoid hemorrhage: a change in relationship that depends on blood severe traumatic brain injury. J Neurosurg 2008;109:50–6.
glucose levels. New Orleans: AANS/CNS Joint Section on Cerebrovascular 98. Taylor S, Fettes S, Jewkes C, et al. Prospective, randomized, controlled trial
Disease Annual Meeting; 2012. to determine the effect of early enhanced enteral nutrition on clinical
73. Zetterling M, Hillered L, Enblad P, et al. Relation between brain interstitial outcome in mechanically ventilated patients suffering head in-jury.
and systemic glucose concentrations after subarachnoid hemorrhage. Crit Care Med 1999;27:2525–31.
J Neurosurg 2011;115(1):66–74. Epub 2011, Apr 8. 99. Davalos A, Ricart W, Gonzalex-Huix F. Effect of malnutrition after acute
74. Helbok R, Schmidt JM, Kurtz P, et al. Systemic glucose and brain energy stroke on clinical outcome. Stroke 1996;27:1028–32.
metabolism after subarachnoid hemorrhage. Neurocrit Care 100. Kudsk KA. Effect of route and type of nutrition on intestine-derived
2010;12(3):317–23. inflammatory responses. Am J Surg 2003;185:16–21.
75. Davidson PC, Steed RD, Bode BW. Glucommander: a computer-directed 101. McClave SA, Sexton LK, Spain DA, et al. Enteral tube feeding in the
intravenous insulin system shown to be safe, simple, and effective in intensive care unit: factors impeding adequate delivery. Crit Care Med
120,618 h of operation. Diabetes Care 2005;28:2418–23. 1999;27:1252–6.
Section II—Clinical and Laboratory Assessment 130.e3

102. McClave SA, Lukan JK, Stefater JA, et al. Poor validity of residual volumes 113. Berger MM, Revelly JP, Cayeux MC, et al. Enteral nutrition in critically ill
as a marker for risk of aspiration in critically ill patients. Crit Care Med patients with severe hemodynamic failure after cardiopulmonary bypass.
2005;33:324–30. Clin Nutr 2005;24:124–32.
103. Rice TW, Swope T, Bozeman S, et al. Variation in enteral nutrition delivery 114. Boullata J, Williams J, Cottrell F, et al. Accurate determination of energy
in mechanically ventilated patients. Nutrition 2005;21:786–92. needs in hospitalized patients. J Am Diet Assoc 2007;107:393–401.
104. Grahm T, Zadroxny D, Harrington T. The benefits of early jejunal 115. American Association for Respiratory Care. AARC clinical practice
hyperalimentation in the head-injured patients. Neurosurgery 1989;25: guidelines. Metabolic measurement using indirect calorimetry during
729–35. mechanical ventilation. Respir Care 1994;39(12):1170–5.
105. Montecalvo MA, Steger KA, Farber HW, et al. Nutritional outcome and 116. McClave SA, Kushner R, Van Way CW 3rd, et al. Nutrition therapy of
pneumonia in critical care patients randomized to gastric versus jejunal the severely obese, critically ill patient: summation of conclusions and
tube feedings. The Critical Care Research Team. Crit Care Med recommendations. JPEN J Parenter Enteral Nutr 2011;35(5 Suppl):88S–96S.
1992;20(10):1377–87. 117. Martindale RG, DeLegge M, McClave S, et al. Nutrition delivery for obese
106. Peterson S, Chen Y. Systemic approach to parenteral nutrition in the ICU. ICU patients: delivery issues, lack of guidelines, and missed opportunities.
Curr Drug Saf 2010;5(1):33–40. JPEN J Parenter Enteral Nutr 2011;35(5 Suppl):80S–7S.
107. Casaer MP, Mesotten D, Hermans G, et al. Early versus late parenteral 118. Flancbaum L, Choban PS, Sambucco S, et al. Comparison of indirect
nutrition in critically ill adults. N Engl J Med 2011;365(6):506–17. Epub calorimetry, the Fick method, and prediction equations in estimating the
2011, Jun 29. energy requirements of critically ill patients. Am J Clin Nutr 1999;69:461–6.
108. Borzotta AP, Pennings J, Papasadero B, et al. Enteral versus 119. Frankenfield DC, Rowe WA, Smith JS, et al. Validation of several established
parenteral nutrition after severe closed head injury. J Trauma equations for resting metabolic rate in obese and nonobese people. J Am
1994;37:459–68. Diet Assoc 2003;103:1152–9.
109. Snydman DR. Shifting patterns in the epidemiology of nosocomial Candida 120. de Jong PCM, Wesdorp RIC, Volovics A, et al. The value of objective
infections. Chest 2003;123:500S–3S. measurements to select patients who are malnourished. Clin Nutr
110. Woodcock, NP, Zeigler D, Palmer D, et al. Enteral versus parenteral 1985;4:61–6.
nutrition: a pragmatic study. Nutrition 2001;17:1–12. 121. Hall JC. The use of internal validity in the construct of an index of
111. Hadley M, Grahn T, Harrington T, et al. Nutritional support and undernutrition. J Parenter Enteral Nutr 1990;14:582–7.
neurotrauma: a critical review of early nutrition in forty-five acute head 122. Harris J, Benedict F. A biometric study of basal metabolism in man.
injury patients. Neurosurgery 1986;19:367–73. Washington, DC: Carnegie Institute of Washington; 1919.
112. Doshi P, Divgiovine B. Effects of early enteral feeding on the outcome of 123. Bistrian BR, Blackburn GL, Sherman M, et al. Therapeutic index of
critically ill mechanically ventilated medical patients on vasopressors. Chest nutritional depletion in hospitalized patients. Surg Gynecol Obstet
2006;130 (Supp):101S. 1975;141:512–16.
Chapter
15  
II

Hematology and Coagulation


Monisha A. Kumar

have an average Hgb 10% lower than men of the same age.1
Introduction Because of the wide range of normal Hgb and hematocrit
Bleeding and thrombosis are common complications in the (Hct) values, it often is difficult to diagnose mild anemia.
intensive care unit (ICU); their impact on neurologically ill The RBC count is the number of RBCs per unit volume. It
patients is of particular concern because these complications generally ranges between 4.2 and 5.9 million cells per micro-
may result in permanent neurologic disability. Sequelae from liter.1,2 RBCs are the most common cell type present in blood.
unrecognized bleeding can lead to cerebral ischemia, organ They are smaller than white blood cells but larger than plate-
hypoperfusion, multiorgan failure, and death. Thrombotic lets. A normal platelet count is between 150,000 and
complications, such as deep vein thrombosis (DVT), are 400,000/µL. Thrombocytopenia is defined as a platelet count
common in neurologically injured patients given the associ- less than 150,000/µL.2
ated limb weakness and complicated by the altered mental The Hct is the ratio of RBCs to blood volume. With modern
state. Multimodality monitoring in the neurologic ICU may laboratory equipment, the Hct is calculated and not measured
preempt and thus mitigate the consequences of these common directly. The mean concentration of Hgb within the red cell
conditions. population (mean corpuscular hemoglobin concentration
The first part of this chapter briefly reviews the physiology [MCHC] is the quotient of the Hgb divided by the Hct. The
of erythropoises and the consequences of anemia and throm- red cell distribution width (RDW), another parameter of red
bocytopenia, explores the complications of blood transfusion, cell size, measures the variability in red cell size of circulating
and discusses alternatives to blood product administration in erythrocytes.3 For example, the etiology of anemia with a low
neurologically critically ill patients. The clinical utility and MCV and a high RDW is most likely iron deficiency; even
application of new monitoring techniques available to evalu- though the anemia is microcytic, the variability of RBC size is
ate anemia and thrombocytopenia in the ICU are discussed. high. In contrast, a macrocytic anemia would yield both a high
The latter portion of this chapter is devoted to the diagnosis, MCV and RDW. Together with the reticulocyte count, the
monitoring, and management of venous thromboembolism. MCV and RDW can narrow the differential diagnosis of
anemia.
The reticulocyte count (RC) measures the number of
Hematology immature RBCs in circulation and is a marker of RBC produc-
Erythropoiesis and Blood Counts tion. This count may be inaccurate in the presence of nucle-
ated RBCs or nuclear debris in the peripheral blood. When
Erythropoiesis is catalyzed by the hormone erythropoietin the RC is reported as a percentage, it should be adjusted for
and leads to production of mature erythrocytes. Reticulocytes the total number of RBCs present. This correction can be
are immature erythrocytes that are released into the circula- made by multiplying the reticulocyte count by the Hct and
tion. Within 1 day, reticulocytes transform into mature red divided by age and gender specific normative Hct values.
blood cells that circulate for 100 to 120 days. Ultimately, senes- Alternatively, the percent of reticulocytes can be multipied by
cent red blood cells (RBCs) are removed from circulation by the red cell count to determine the absolute RC.2 The normal
macrophages in the spleen and other reticuloendothelial absolute reticulocyte count (ARC) is between 25,000 and
tissues. On average, the number of erythrocytes formed is 75,000/µL.2 In the presence of anemia, an ARC less than
equivalent to the number destroyed. 75,000/µL indicates a hypoproliferative process, whereas an
The complete blood count (CBC) provides accurate infor- ARC greater than 100,000/µL indicates hemolysis or an appro-
mation about hemoglobin (Hgb) concentration and cell priate erythropoietin response. RCs between 75,000 and
counts as well as calculated indices of erythrocyte size and 100,000/µL must be interpreted for the degree of anemia
Hgb content.1 The Hgb concentration varies by both age and present.2
gender (Table 15-1). In the average adult male, the Hgb con- Review of the peripheral blood smear remains an informa-
centration varies between 13 and 17.5 g/dL; in women it tive diagnostic tool. Not only can it confirm findings of an
ranges from 12 to 16 g/dL. A high white blood cell (WBC) automated CBC, but it also may suggest a marrow disease or
count, lipemia, or a precipitating monoclonal protein can hemolytic process. Microcytic, hypochromic cells may suggest
result in a spuriously high Hgb. High altitude will result in an iron deficiency or thalassemia, whereas macrocytosis with
increased Hgb concentration; the rise in Hgb is proportional ovalocytes may suggest a megaloblastic anemia. The periph-
to the magnitude of elevation. Women of childbearing age eral smear may demonstrate echinocytes, seen in uremia;
© Copyright 2013 Elsevier Inc. All rights reserved. 131
132 Section II—Clinical and Laboratory Assessment

Table 15.1  Reference Hematologic Values in Children and Adults*


Hemoglobin Hematocrit Red Blood Cell
(g/dL) (%) Count (1012/L) MCV (fL) MCH (pg) MCHC (g/dL)
Age Mean −2 SD Mean −2 SD Mean −2 SD Mean −2 SD Mean −2 SD Mean −2 SD
Birth (cord blood) 16.5 13.5 51 42 4.7 3.9 108 98 34 31 33 30
1-3 days (capillary) 18.5 14.5 56 45 5.3 4.0 108 95 34 31 33 29
1 week 17.5 13.5 54 42 5.1 3.9 107 88 34 28 33 28
2 weeks 16.5 12.5 51 39 4.9 3.6 105 86 34 28 33 28
1 month 14.0 10.0 43 31 4.2 3.0 104 85 34 28 33 29
2 months 11.5 9.0 35 28 3.8 2.7 96 77 30 26 33 29
3-6 months 11.5 9.5 35 29 3.8 3.1 91 74 30 25 33 30
0.5-2 years 12.0 10.5 36 33 4.5 3.7 78 70 27 23 33 30
2-6 years 12.5 11.5 37 34 4.6 3.9 81 75 27 24 34 31
6-12 years 13.5 11.5 40 35 4.6 4.0 86 77 29 25 34 31
12-18 YEARS
Female 14.0 12.0 41 36 4.6 4.1 90 78 30 25 34 31
Male 14.5 13.0 43 37 4.9 4.5 88 78 30 25 34 31
18-49 YEARS
Female 14.0 12.0 41 36 4.6 4.0 90 80 30 26 34 31
Male 15.5 13.5 47 41 5.2 4.5 90 80 30 26 34 31

Data from Dallman PR: Blood-forming tissues. In: Rudolph A, editor. Pediatrics. 16th ed. New York: Appleton-Century-Crofts; 1977. p. 1111.
MCH, Mean corpuscular hemoglobin; MCHC, mean corpuscular hemoglobin concentration; MCV, mean corpuscular volume.
*These data have been compiled from several sources. Emphasis is given to studies employing electronic counters and to the selection of populations that are likely
to exclude individuals with iron deficiency. The mean −2 SD can be expected to include 95% of the observations in a normal population.

acanthocytes, observed in liver disease; or schistocytes, found mechanical ventilation and may be particularly detrimental
in disseminated intravascular coagulation (DIC). Special for patients in the neurocritical care unit (NCCU) with
stains of the peripheral smear may assist in the diagnosis of reduced cerebrovascular reserve.
anemia, particularly when there is rouleaux formation and Anemia is associated with increased ICU length of stay and
automated counters may be inaccurate. increased mortality in critically ill patients.5 In surgical ICU
Early after acute hemorrhage, red cell mass and plasma patients with cardiac disease, anemia is associated with
volume contract at the same rate; therefore, there may not be increased risk of death.8 Anemia also is associated with the
any observed reduction in Hgb concentration or Hct. The development of left ventricular hypertrophy in patients with
platelet count often increases after acute blood loss and there end-stage renal disease, and its correction is associated with
often is an increase in reticulocyte count. Because young fewer cardiovascular events.9 Preoperative anemia is associ-
erythrocytes are larger than mature ones, the MCV is usually ated with worse surgical outcomes10 and preprocedural
slightly elevated. Bleeding is usually accompanied by an anemia among patients undergoing percutaneous coronary
increase in indirect bilirubin, which reflects the catabolism of intervention is associated with increased adverse in-hospital
heme from extravasated RBCs. outcomes.11

Anemia Causes of Anemia in the Intensive


Care Unit
Anemia in the Intensive Care Unit The causes of anemia in the ICU are varied and may occur
Anemia is a common problem among critically ill patients. concurrently. These include sepsis, trauma, burns, infections,
According to the World Health Organization, anemia is clin- hemolysis, immune-associated iron deficiency, decreased
ically defined as an Hgb concentration less than or equal to endogenous erythropoietin production, renal failure, liver
12 g/dL for women and 13 g/dL for men.4 Nearly two thirds disease, malignancy, collagen vascular disease, nutritional
of patients are anemic on admission to the ICU5 and deficiency, gastrointestinal bleeding, drug-related causes, and
between 70% and 95% of patients become anemic by day phlebotomy. Phlebotomy is the most significant modifiable
3.6,7 Anemia is potentially deleterious because lower Hgb factor associated with anemia in the ICU and is a predictor of
levels decrease oxygen carrying capacity of the blood and blood transfusion12; it accounts for 40 to 70 mL per day of
may reduce tissue oxygenation. This is detrimental for blood loss early in the ICU period.13,14 Smoller observed that
patients with critical illness who often have increased meta- the amount of blood drawn per day varied from 12 mL per
bolic demand for oxygen from infection, sepsis, fever, and day on the ward, to 33 mL a day in the ICU in patients without
Section II—Clinical and Laboratory Assessment 133

an arterial line, to 74 mL a day in ICU patients with an arterial IRF (total reticulocyte count low, IRF high).20 It also is useful
line.15 For every 100 mL of blood removed by phlebotomy, to identify resumption of marrow activity after bone marrow
Hgb and Hct levels fall by 0.7 g/dL and 1.9%, respectively.12 transplantation or chemotherapy.
ICU patients with evidence of inflammation and multior- The CHr and the MCVr are reticulocyte indices that reflect
gan failure develop anemia despite a lack of active bleeding. the adequacy of iron stores available for erythropoiesis. The
This “anemia of critical illness” results from underproduction CHr is important because it can help identify iron-deficiency
of erythrocytes and is similar to anemia of chronic disease, anemia in the face of the acute phase response when total iron
which is thought to be due to inflammation. Inflammatory stores, as evidenced by an elevated ferritin and transferrin, are
cytokines, such as tumor necrosis factor-α (TNFα), adequate. This may be seen in anemia of chronic disease. The
interferon-α (IFNα), and interleukin-1 (IL-1), suppress eryth- MCVr has not been well studied, but has been used to evaluate
ropoiesis and are central in the pathogenesis of anemia in possible EPO abuse in sports.22 The main limitations of these
these patients.16 These inflammatory mediators inhibit new indices include limited availability of the machines and
hypoxia-induced (as opposed to anemia-induced) erythro- variable sensitivity of the analyses.
poietin (EPO) production by up to 89%.17 FRBCs can be found in a variety of diseases from microan-
Proinflammatory cytokines not only induce bone marrow giopathies to cardiovascular disorders (valve prostheses and
suppression of erythropoiesis but also aggravate bleeding and endocarditis). Microangiopathies need immediate diagnosis
disrupt iron metabolism. These cytokines exacerbate intesti- and treatment, and expedient identification and quantifica-
nal bleeding through increased permeability of the intestinal tion is paramount. The sensitivity of the new analyzers to
wall. They alter iron metabolism by stimulating phagocytosis diagnose microangiopathy is very high (>90%), but the speci-
of RBCs. More than 90% of ICU patients have low levels of ficity is low (20%-50%).23,24 Therefore in certain populations,
serum iron (Fe), total iron binding capacity (TIBC), and Fe- the FRBC count may be a good screening test for microangi-
to-TIBC ratio, but have normal or elevated serum ferritin.18,19 opathies, but further study is warranted.
Despite low levels of circulating iron, EPO levels remain only In addition to new laboratory indices of red cell size and
modestly increased and are not markedly elevated. This function, new monitoring devices permit measurements of
blunted EPO response results from the inhibition of EPO total Hgb concentration. In contrast to the standard cyano-
transcription by inflammatory mediators. Rogiers et al. found methemoglobin method of Hgb determination, co-oximeters
that at the same serum level of Hgb, ambulatory anemic use spectrophotometry to determine Hgb concentration. The
patients had an eight times higher level of EPO than did septic methodology of Hgb measurement is similar to that of oxy-
ICU patients.18 hemoglobin measurement by conventional pulse oximetry,
The etiology of anemia in the ICU is multifactorial and can except that instead of two wavelengths of light, multiple wave-
be difficult to determine. The diagnosis of anemia in the ICU lengths are transmitted through the finger to measure the light
is complicated because serial monitoring of hematologic pro- absorbance characteristics of Hgb.25 Point-of-care co-oximeters,
files (i.e., phlebotomy) can exacerbate the condition. such as the HemoCue (Angelholm, Sweden), allow for capil-
lary measurement of Hgb concentration at the bedside,
whereas noninvasive co-oximeters, such as the Radical-7
Monitoring Anemia in the Intensive (Masimo Corp, Irvine, CA) device, provide continuous Hgb
Care Unit measurements. The accuracy of these devices compare well
Modern laboratory testing and the availability of automated against standard laboratory assays, although the Radical-7
cell counters provide a wealth of diagnostic information for may give lower readings than the HemoCue during surgery.26,27
interpretation. Hgb measurements with these devices have the additional
The extended differential count (EDC) quantifies other cell benefit of providing rapid results that may facilitate monitor-
types in the peripheral blood including nucleated RBCs ing in the operating room, ICU, and emergency department.
(NRBCs). Although NRBCs typically are only seen in neo-
nates, their presence has been identified in adults, notably in
patients with septicemia, massive hemorrhage, and severe Anemia in the Neurocritical Care Unit
hypoxia.20,21 When identified in adults, the presence and Anemia is common in the NCCU and, depending on the defi-
burden of NRBC may correlate with mortality and poor nition, occurs in nearly 40% of patients.28 It has been identi-
outcome. For example, Stachon observed a mortality rate of fied as an independent factor associated with death or severe
44% in surgical patients with NRBCs compared with 4.2% for disability in several disorders including subarachnoid hemor-
patients without.21 rhage (SAH), particularly in patients who develop delayed
Other parameters of cellular analysis available through cerebral ischemia, traumatic brain injury (TBI), or intracere-
automated analyzers include the immature reticulocyte frac- bral hemorrhage (ICH).28-34 However, there remains variation
tion (IRF), mean reticulocyte hemoglobin content (CHr), in how anemia is managed.
mean reticulocyte volume (MCVr), fragmented RBC (FRBC) In the uninjured brain, vasodilation of the cerebral arteries
count, and the immature platelet fraction (IPF). IRF is an early compensates for anemia-related reduction in oxygen carrying
and sensitive index of erythropoiesis. When characterized capacity; therefore, frank brain hypoxia does not typically
with total reticulocyte count, it can discriminate between occur at Hgb concentrations greater than 6 g/dL.35 The ability
anemias with increased erythropoiesis (increased total reticu- of cerebral vessels to vary their caliber to maintain constant
locyte count and IRF) such as hemolytic anemias; decreased perfusion is termed cerebral autoregulation. With impairment
marrow function (decreased total reticulocyte count and IRF); of cerebral autoregulation, such as may occur after SAH or
and conditions such as acute infections and myelodysplastic TBI, tissue hypoxia may occur at higher Hgb concentrations.
syndrome with dissociation of total reticulocyte count and Consistent with this, fewer ischemic events are observed in
134 Section II—Clinical and Laboratory Assessment

SAH patients with higher Hgb concentrations.33 However, the associated with greater mortality than admission APACHE II,
Hgb threshold below which cerebral metabolic dysfunction Simplified Acute Physiology Score (SAPS), or multiple organ
occurs is only beginning to be elucidated. dysfunction syndrome (MODS) scores.49
A fundamental tenet of neurocritical care is that ischemia ICU-related thrombocytopenia is primarily caused by
is an important cause of brain injury. Anemia and compro- peripheral loss, destruction, and margination of platelets
mised oxygen delivery may exacerbate ischemia; therefore rather than bone marrow hypoplasia or nutritional deficiency.
anemia may represent a therapeutic target in the NCCU. Sepsis accounts for 48% of cases. Other causes include liver
However, the ramifications of anemia may vary by the type of disease, hypersplenism, consumption, platelet destruction,
brain injury. The consequences of anemia after SAH, particu- DIC, medications, immune phenomena, and intravascular
larly among patients with severe vasospasm or delayed cere- devices.55 The cause of a low platelet count in the ICU can be
bral ischemia (DCI), may be dire. In contrast, the significance difficult to determine and in more than 25% of critically ill
of cerebral ischemia in other neorocritically ill subpopulations patients the cause is multifactorial. Pseudothrombocytopenia
is less clear; therefore, anemia may be of less concern. occurs when platelets clump in association with an ethylene-
diamine tetra-acetic acid (EDTA)–dependent platelet aggluti-
nin.52 An accurate platelet count can be determined by
Optimal Hemoglobin Threshold in the collecting the sample in a citrated collection tube or by using
Neurocritical Care Unit Population a heparinized blood sample. Postresuscitative hemodilution
Several physiology studies in neurocritically ill patients have can result in thrombocytopenia (i.e., after blood transfusion
sought to define an Hgb threshold by measuring markers of or with crystalloid or colloid replacement therapy for blood
cerebral metabolism.35-37 Brain tissue oxygen tension (PbtO2) loss). The post-transfusion reduction in platelet count is asso-
monitoring and lactate-to-pyruvate ratio (LPR) measure- ciated with both splenic sequestration of platelets and a reduc-
ments from cerebral microdialysis demonstrated increased tion in viable platelets in stored blood.
brain hypoxia and cell energy dysfunction when Hgb levels Platelets play a complex role in sepsis and multiorgan dys-
were less than 9 g/dL in SAH patients.37 However, the physi- function. Platelets of septic patients may display increased
ologic data in TBI are conflicting.37-39 Although TBI patients adhesion and aggregation.56,57 Bacteria and bacterial products
frequently have pathologic evidence of cerebral infarction, may affect platelet function. Lipopolysaccharide increases
premortem evidence of ischemia is debatable.40-42 Early evi- platelet aggregation in some animal models, but has the oppo-
dence demonstrated compromised cerebral blood flow sug- site effect on human platelets in vitro.48 Lipoteichoic acid, a
gestive of ischemia; however, it was later demonstrated that cellular membrane component of gram-positive bacteria,
the altered blood flow correlated with decreased cerebral inhibits platelet aggregation in human platelets.48 Escherichia
metabolism.40,43,44 Regardless, anemia appears to be a predictor coli endotoxin may indirectly reduce platelet responsiveness.48
of poor outcome, although transfusion of PRBCs does not
always appear to mitigate its effect.45
The treatment of anemia in the NCCU is unclear in part Drug-Induced Thrombocytopenia
because of the heterogeneous pathophysiology of diseases Drug-induced thrombocytopenia (DIT) is a common
treated therein. Moreover, transfusion trials in general ICUs nonimmune-mediated disorder, associated with bone marrow
included very few or excluded neurologically critically ill suppression and accelerated platelet destruction and defined
patients and therefore results cannot be easily extrapolated as a platelet count between 50 and 150,000 × 106/L with any
from them.46,47 Transfusion studies in patients with active degree of spontaneous bleeding.58 It is observed in 19% to
cardiac ischemia have not demonstrated consistent benefit 25% of ICU patients59,60 and associated with medications such
from a liberal transfusion approach.5,8 It is unclear whether as cytotoxic chemotherapeutic agents,61,62 thiazide diuretics,
this is analogous to patients with cerebral ischemia. ethanol, tolbutamide, and bleomycin. DIT typically develops
2 to 3 days after a previously administered medication, and 7
days after a virgin drug. Discontinuation of the medication
Thrombocytopenia normalizes the platelet count by 5 to 8 days.63 A major bleed-
ing event can occur in 4% of patients,63 and these patients may
Thrombocytopenia in the Critically Ill benefit from intravenous immunoglobulin (IVIg), plasma-
The importance of platelets and their role in thrombus forma- pheresis, or platelet transfusion64 but not corticosteroids.65
tion is clear, but their significance in sepsis is being eluci-
dated.48 Thrombocytopenia occurs in 13% to 58% of ICU
patients49-53; 23% have at least one platelet count less than Drug-Induced Immune Thrombocytopenia
100,000/mm3 and 10% had counts less than 50,000 mm3.50 Drug-induced immune thrombocytopenia (DITP) is a disor-
Sepsis is considered the major independent risk factor for der characterized by the development of drug-dependent
thrombocytopenia in the ICU. Other factors associated with antibodies that bind to glycoproteins on the surface of plate-
thrombocytopenia are episodes of bleeding or transfusion, lets and accelerate their destruction in the presence of the
longer ICU stays, and Acute Physiology and Chronic Health specific drug. This type of disorder may be more severe than
Evaluation II (APACHE II) scores greater than 15.52 The devel- DIT. Hundreds of drugs have been associated with this type
opment of thrombocytopenia during the ICU stay is associ- of thrombocytopenia; the most common are heparin, quinine
ated with decreased ICU survival and may have a greater (cinchona alkaloids), penicillin, sulfonamides, nonsteroidal
implication for prognosis than admission thrombocytope- anti-inflammatory drugs (NSAIDs), anticonvulsants, anti-
nia.51,53,54 For example, Cawley et al. observed that a drop in rheumatics, oral hypoglycemics, gold salts, diuretics, rifampi-
platelet count to less than 50% of admission levels was cin, ranitidine, and the glycoprotein (GP) IIb/IIIa inhibitors
Section II—Clinical and Laboratory Assessment 135

(e.g., abciximab, tirofiban, or eptifibatide).58 The precise Clinical complications of HIT include both venous and
mechanisms by which various drugs incite the immune arterial thromboses.55 Arterial thrombosis is more common in
response are beyond the scope of this chapter. Some patients those with known cardiac disease, whereas DVT is seen in
can make both drug-independent and drug-dependent anti- postoperative patients. Cerebral sinus thrombosis, venous
bodies during the course of medication exposure. The drug- limb gangrene, and hemorrhagic adrenal infarction also are
independent antibodies (autoantibodies) usually are transient reported.67 Concomitant prothrombotic risk factors include
but if they persist, can result in a chronic autoimmune throm- diabetes mellitus, malignancy, systemic lupus erythematosus
bocytopenic purpura. When agents such as GPIIb/IIIa inhibi- (SLE), antiphospholipid antibody syndrome, indwelling cath-
tors are used, platelet counts should be frequently monitored. eters, and trauma.55 The incidence of stroke is 3.1% among
If the platelet count is less than 50,000/µL, the drug should be patients with HIT and is more common in females, in patients
discontinued. If the platelet count is less than 10,000/µL and with more severe thrombocytopenia, and early in the course
there is severe bleeding or if an invasive procedure is required, of HIT. Myocardial infarction can occur in 3% to 5% of
then platelet transfusion should be considered.66 Readminis- patients and amputation in as many as 20% of patients with
tration of GPIIb/IIIa at a later date is not recommended HIT and limb thrombosis. In SAH, HIT may occur in up to
because the rechallenge might prove more severe than the 15% of patients, and its risk is increased in Fisher grade 3
initial event. patients, females, and perhaps endovascular treatments.70 The
incidence of hypodensity on computed tomography (CT)
scans and worse outcome is increased.
Heparin-Induced Thrombocytopenia PF4 antibodies are transient and are undetectable a median
Heparin-induced thrombocytopenia (HIT) is an anticoagulant- of 50 to 85 days after an episode of HIT.71 In some patients
induced prothrombotic immune disorder caused by heparin- antibodies remain detectable at low levels for several months.
dependent platelet-activating IgG antibodies that recognize If heparin is readministered to a patient with high levels of
complexes of platelet factor 4 (PF4) bound to heparin.67,68 HIT antibodies, development of thrombocytopenia can be rapid.
should be suspected when the platelet count falls to less than This is unlikely more than 100 days from exposure. The “4
50% of the baseline level or an absolute number of less than T’s”—Thrombocytopenia, Timing, Thrombosis, and oTher
150,000/µL between days 5 and 14 of heparin exposure. plausible explanations for the clinical scenario—is an eight-
Several factors are associated with the development of HIT: point clinical scoring system that evaluates the likelihood of
(1) the dose or duration of heparin use; (2) the type of heparin developing HIT (Table 15.3).72 It has a high negative predic-
administered; (3) the patient population; and (4) patient tive value and can help rule out HIT. If HIT is suspected all
gender69 (Table 15.2). A greater risk of HIT is observed in heparin sources must be stopped, preferably before the dem-
patients exposed to a high dose of heparin or a long duration onstration of HIT antibodies. The direct thrombin inhibitor
of treatment, with unfractionated heparin rather than low family of anticoagulants should replace heparin because treat-
molecular weight heparin (LMWH), with surgical rather than ment with LMWH does not fully eradicate the risk of HIT.
medical patients, and in females.
PF4 is a tetrameric protein, involved with hemostasis,
immunoregulation, and angiogenesis. Heparin binding Thrombotic Microangiopathies: Thrombotic
induces a conformational change in PF4. HIT differs from Thrombocytopenic Purpura
most other drug-induced immune thrombocytopenias in that Thrombotic thrombocytopenic purpura (TTP) is a rare but
the induced antibodies engage the Fc receptor (not Fab region) potentially fatal disorder that mostly affects adult women. It
and thereby activate platelets (i.e., they promote thrombosis is characterized by red cell fragmentation (microangiopathy),
and a hypercoagulable state, which is characteristic of HIT). hemolytic anemia, and thrombosis (consumptive thrombocy-
PF4 also neutralizes the anticoagulant effect of heparin, which topenia) that lead to ischemic organ damage.73 The predilec-
exacerbates the prothrombotic milieu. tion for thrombosis is a consequence of abnormal numbers of

Table 15.2  Risk Factors for Immune Heparin-Induced Thrombocytopenia


Risk Factor Odds Ratio Comment
Duration of heparin use ∼20-100 Odds ratio (OR) estimated as follows: risk for HIT postcardiac surgery (with
(>1 week vs. <1 day) UFH prophylaxis for >1 week) ∼2% versus risk for “delayed-onset HIT”
(without prophylaxis) ∼0.02%-0.1%
Type of Heparin ∼10-15 Difference in risk for HIT between heparin types is established for postsurgical
UFH > LMWH > thromboprophylaxis (UFH vs. LMWH) and is more pronounced in women.
fondaparinux ORs shown are for UFH versus LMWH (woman, postsurgical prophylaxis) [18]
Type of patient (surgery ∼3-4 Highest reported frequencies of HIT are in postsurgical thromboprophylaxis
> medical > pregnancy) (OR shown is for surgery vs. medical) [18]
Gender (female > male) ∼1.5-2.0 Difference in risk for HIT between women and men has only been established
for UFH thromboprophylaxis

Used with permission from Warkentin TE. Heparin-induced thrombocytopenia. Hematol Oncol Clin North Am 2007;21(4):592.
HIT, Heparin-induced thrombocytopenia; LMWH, low molecular weight heparin; UFH, unfractionated heparin.
136 Section II—Clinical and Laboratory Assessment

Table 15.3  Estimating the Pretest Probability of Heparin-Induced Thrombocytopenia:


The “4 T’s” Scoring System
Points (0, 1, or 2 for each of 4 categories: maximum possible score = 8)
Date : 2 1 0
Thrombocytopenia* >50% platelet decrease to 30%-50% platelet count decrease <30% platelet
Score = _____________ nadir ≥20 × 109/L (or >50% directly resulting from decrease or
surgery) or nadir 10 − 19 × 109/L nadir <10 × 109/L
Timing† of platelet count Days 5-10 onset† or ≤1 day Consistent with days 5-10 decrease, Platelet count
decrease, thrombosis, or (with recent heparin but not clear (e.g. , missing decrease ≥4 days
other sequelae (first day of exposure within past 5-30 platelet counts), or ≤1 day (heparin without recent
heparin course = day 0) days) exposure within past 31-100 days), heparin exposure
Score = ________ or platelet decrease after day 10
Thrombosis (including adrenal Proven new thrombosis, or Progressive or recurrent thrombosis None
infarction) or other sequelae skin necrois (at injection or erythematous skin lesions
(e.g., skin lesions) site), or post-IV heparin (at injection sites), or suspected
Score = ____________ bolus anaphylactoid reaction thrombosis (not proven)
OTher cause for No explantion for platelet Possible other cause is evident Definite other
thrombocytopenia count decrease is evident cause is present
Score = ____________
Total score = _______________ Pretest probability score: 6-8 = high, 4-5 = intermediate, 0-3 = low

IV, Intravenous.
*Changes to score can occur, based on new information (e.g., further decrease in platelets, new thrombosis, other causes for platlet decrease).

First day of immunizing heparin exposure considered day 0; the day the platelet count begins to decrease is considered the day of onset of thrombocytopenia (it
generally takes 1 to 3 more days until an arbitrary threshold that defines thrombocytopenia is passed. Usually, heparin administered at or near surgery is the most
immunizing situation).

von Willebrand multimers (µL-vWF) in the circulation due Disseminated Intravascular


to altered vWF homeostasis. vWF is normally released in
response to prothrombotic stimuli, and persistence of these
Coagulation
multimers leads to platelet activation and clumping with sub- Disseminated intravascular coagulation (DIC) is a syndrome
sequent microvascular thrombosis. The protease, ADAMTS13, of thrombosis and systemic hemorrhage caused by an abnor-
a disintegrin and metalloproteinase with a thrombospondin mally activated extrinsic clotting cascade or tissue factor
type 1 motif, member 13 normally cleaves the µL-vWF into system. Tissue factor (TF) is a tissue membrane glycoprotein,
smaller peptides.73 When the protease activity is decreased or which binds to factor VII. Normally, the TF/factor VIIa
absent there is accumulation of the large multimers. The activ- complex activates factors X and XI that initiate clotting. In the
ity of ADAMTS13 can be decreased by many different factors face of systemic inflammation, circulating cytokines, IL-1,
but most cases of acquired adult TTP result from autoanti- IL-6, and TNFα cause the constitutive release of tissue factor
bodies to ADAMTS13. Congenital TTP results from complete and down-regulate thrombomodulin, an endothelial cell
absence of this protease. surface receptor that activates anticoagulants and inhibits
TTP often is seen with infection, pregnancy, or estrogen- factors Va and VIIa.74 The down-regulation of thrombomodu-
containing compounds. General clinical features include fever, lin converts the antithrombotic blood vessel endothelial
anemia, thrombocytopenia, renal failure, and neurologic surface to a prothrombotic one. The early thrombotic phase
symptoms from multiorgan damage due to microvascular causes widespread fibrin deposition, which is followed by con-
thrombi. Patients present with complaints of weakness, sumption of platelets and clotting factors that culminate in
malaise, nausea, vomiting, abdominal pain, hallucinations, fibrinolysis and hemorrhage.
and seizures. Diagnostic laboratory tests for TTP include The diagnosis of DIC should always be suspected if the
hemoglobin, platelet count, peripheral smear (to document patient has acrocyanosis, purpura, dusky cyanosis of the nose,
fragmentation), lactate dehydrogenase (LDH), bilirubin, and ears, and genitalia, or is oozing blood from venipuncture or
reticulocyte count. ADAMTS13 activity level and antibody arterial catheter sites. Neurologic complications of DIC
tests may be helpful. The differential diagnosis for TTP include coma, encephalopathy, SAH, small and large vessel
includes DIC, the HELLP syndrome (hemolytic anemia, ele- stroke, embolic infarction from nonbacterial thrombotic
vated liver enzymes, low platelets), hemolytic uremic syn- emboli, and intracerebral hemorrhage.73 DIC is a major cause
drome (HUS), malignant hypertension, preeclampsia, and of stroke in medical ICUs, is a frequent complication in ter-
some infections. Treatment is aimed at prompt removal of minally ill patients, and may aggravate neurologic deficit after
circulating antibodies (by plasma exchange), replacement of TBI.74 Several scoring systems help diagnose DIC. One such
ADAMTS13 (by administration of FFP, cryoprecipitate) and system developed by the International Society of Thrombosis
immunosuppression (with steroids, cyclosporine, vincristine, and Haemostasis combines platelet count, the ratio of fibrin
cyclophosphamide, or rituximab).73 monomers to fibrin degradation products, a prolonged
Section II—Clinical and Laboratory Assessment 137

prothrombin time (PT), and fibrinogen level to determine a is an optical detection device that measures closure time, or
DIC score.75 The sensitivity and specificity of this scoring time to the cessation of the blood flow through a channel
system is greater than 90%. Other scoring systems includes a coated with collagen and a platelet activator, either adenosine
D-dimer test and can help predict mortality.76 diphosphate (ADP) or epinephrine. Eight hundred microliters
of citrated whole blood is required for the analysis. Although
PFA-100 results are abnormal in both congenital platelet
Monitors of Platelet Function abnormalities88 and moderate-severe von Willebrand’s disease
Circulating platelets vary in size, metabolism, and activity. (vWD), it is not sensitive enough to serve as an adequate
Newly released platelets contain RNA and owing to the simi- screening test to exclude all congenital abnormalities or mild
larity to reticulocytes, are called reticulated platelets. The vWD. A normal ADP closure time has been found to be a
largest are more reactive and secrete a greater quantity of good negative predictor of bleeding89; however, an elevated
thrombogenic factors.20 Automated counters can quantify closure time has a poor positive predictive value. Other POCT
reticulated platelets and determine platelet size variability is intended primarily to monitor antiplatelet medications such
and mean platelet volume. Fluorescent dyes and cytometers as aspirin, clopidogrel, or glycoprotein IIb/IIIa inhibitors.
have allowed quantification of reticulated platelets. Increased Correlation with platelet aggregometry has been poor; one
presence of reticulated platelets may be seen in peripheral study found the sensitivity of the PFA-100 and the Verify-Now
platelet destruction and acute blood loss, whereas a de- (Accumetrics, San Diego, CA) to be 62% and 39%, respec-
creased number may be seen in marrow hypoplasia or cyto- tively. Another study of 100 patients treated with 75 to 100 mg
toxic chemotherapy. The increase in immature platelet of aspirin for CAD demonstrated variable levels of aspirin
fraction (IPF) may precede the rise in platelet count by resistance (12%-22%) when measured by four tests of platelet
days,77 and may serve to limit prophylactic platelet trans­ aggregability.90 Only 2% of patients were aspirin resistant by
fusion after chemotherapy.78 Moreover, an increase in reticu- all tests, and correlation between tests was poor.
lated platelets may be associated with an increased risk of
thrombosis in thrombocytosis and myeloproliferative
disorders.85 Transfusion in the Critically Ill
New automated counters can provide the mean platelet
volume (MPV) and platelet distribution width (PDW), which Packed Red Blood Cell Transfusion
are analogous to the erythrocyte indices. There is a nonlinear Blood transfusion in the ICU is common due in large part to
inverse relationship between MPV and platelet concentra- the increased severity of illness, the burden of chronic diseases,
tion; MPV decreases with higher platelet counts.80 Therefore improved ICU management, and blood-intensive surgeries.91
the MPV should be assessed relative to platelet number. MPV Annually, approximately 15 millions units of blood are
is also a known marker of platelet activation. MPV elevation donated, and 13 million are transfused.92,93 Up to 85% of
is associated with increased risk of myocardial infarction patients remaining in the ICU more than 1 week are trans-
(MI) in patients with coronary artery disease (CAD) and fused, with an average total transfusion of 9.5 units.6 Overall
increased mortality after acute MI.81,82 Studies have demon- packed red blood cell (PRBC) transfusion rate among ICU
strated MPV elevations in acute ischemic stroke82-85; more- patients is between 37% and 50%.7,13,14
over, some studies suggest that increased MPV is related to Since 2000, concern about the safety of PRBC transfusion,
poor functional outcome, although this remains controver- including immunosuppressive and microcirculatory compli-
sial.86,87 Although these parameters provide novel informa- cations, has led to a reappraisal of transfusion practices. In
tion, there are many limitations to the use of platelet indices. 1999, the Transfusion Requirements in Critical Care (TRICC)
The anticoagulant used for CBC determination incites non- trial, a randomized study, examined whether a restrictive
uniform structural changes in platelets. As a consequence, the (Hgb threshold of 7 g/dL) and liberal (Hgb threshold of 10 g/
MPV may increase or decrease depending on the method of dL) transfusion strategy in the ICU was associated with mor-
measurement. There is no mathematical correction for this tality.5 The primary endpoint of 30-day mortality was similar
phenomenon, and therefore platelet indices are still consid- between two groups and in subgroups, including in patients
ered experimental. with cardiac disease, severe infections or septic shock, or
trauma. However, in patients with an APACHE II score less
than 20, or those younger than 55 years of age, a restrictive
Platelet Function Assays approach was associated with less mortality. The authors rec-
Platelet aggregation with a platelet aggregometer is the gold ommended that critically ill patients with isovolemic anemia
standard to test platelet function. However, this method is receive PRBC transfusion when the Hgb concentration is less
labor intensive and requires technologic expertise; therefore, than 7 g/dL and that Hgb levels be maintained between 7 and
it is unsuitable for bedside monitoring. Since 2000, devices 9 g/dL. Many observational studies are consistent with this
that can be used in the laboratory or at the point of care observation.43 The most appropriate transfusion strategies
(POC) have become available; however, the results do not remain unclear for patients in the NCCU; although guide-
always correlate with the gold standard, aggregometry, and lines regarding transfusion practice have been published for
more studies need to define the role of these new assays. general critical care,94 neurocritical care,95 TBI,96 and SAH.46
A variety of POC testing (POCT) devices are available for
platelet function. These devices have been developed primar-
ily to measure the effect of antiplatelet medications and not Efficacy of Transfusion
necessarily to diagnose platelet function defects. The Platelet RBC transfusions are intended to augment oxygen delivery
Function Analyzer 100 (PFA-100, Siemans Corporation, NY) and improve tissue oxygenation. It is presumed that increased
138 Section II—Clinical and Laboratory Assessment

Hgb concentration increases oxygen carrying capacity and completed transfusion.115 The most common clinical features
provides more oxygen to delivery-dependent tissue. However, include bilateral pulmonary edema, hypoxemia, fever, dyspnea,
whether blood transfusion successfully achieves improved and hypotension in the presence of normal cardiac func-
tissue oxygenation remains unclear. In the NCCU population tion.115 Plasma-rich blood components (FFP and platelets)
several studies have examined the effect of transfusion on and high-volume transfusion may predispose to TRALI,
brain oxygen. In summary, transfusion increases brain oxygen although it has been associated with all blood product com-
in about 75% of patients.96-98 However, even when transfusion ponents including IVIg and cryoprecipitate.115,116
increases brain oxygen it may not always correct other markers Tolerance induction after transfusion is associated with a
of cellular distress (e.g., the LPR).35 decrease in natural killer cell function, defective antigen pre-
Sequelae of blood storage, termed the storage lesion, sentation, and a reduction in helper/suppressor T-lymphocyte
may be responsible for the observed lack of efficacy of RBC ratio. Consequently transfusion is linked to increased predis-
transfusion (RBCT). First, during storage, RBCs undergo a position to hospital-acquired and postoperative infections in
predictable change from biconcave disks to deformed spher- variety of disorders and ICU patients, multiorgan dysfunc-
oechinocytes, resulting in loss of deformability and decreased tion, acute respiratory distress syndrome (ARDS), systemic
ability to navigate the microcirculation (vessels of 3-8 µm).99 inflammatory response system (SIRS), and even to cancer
Second, increased osmotic fragility may lead to decreased recurrence.117-129 A recent meta-analysis of transfusion in the
RBC survival.100,101 Third, adenosine triphosphate (ATP) is ICU130 suggests that the pooled odds ratio for developing an
depleted.102 Fourth, the p50, a measure of Hgb affinity for infectious complication was 1.8 (95% CI, 1.5-2.2).
oxygen, is lower in stored blood, which results in less oxygen The deleterious effects of transfusion are thought to
unloading to tissues.103 Fifth, after 7 to 14 days, stored blood occur in large part from inflammatory cytokines (e.g.,
is depleted of 2,3-diphosphoglycerate (2,3-DPG)104; this shifts IL-1, IL-6, IL-8), inflammatory mediators (e.g., bactericidal
the oxygen dissociation curve to the left and reduces the permeability-increasing protein [BPI] and TNFα)131; neutro-
amount of oxygen available for tissue consumption. Sixth, phil activation132 and the accumulation of histamine, lipids,
prolonged storage results in adhesion of RBCs to endothelial cytokines, cellular membranes, and HLA class antigens.
cells, which can compromise microvascular circulation.105,106 Some of these compounds are derived from WBCs,
Finally, storage for more than 42 days may cause vasoconstric- which are present in the cellular components of standard-
tion upon transfusion due to free Hgb and lysophosphatidyl preparation blood products. Consequently although it seems
choline species released from the cellular membrane of senes- reasonable to expect that leukoreduction of stored blood
cent RBCs.107 would mitigate the immunomodulatory effects of transfused
RBCs, its clinical impact and cost effectiveness have not
been fully elucidated.133-138 There also may be non–leukocyte
Potential Side Effects of Transfusion mediated mechanisms of immunosuppression139 (i.e., RBC
A variety of deleterious side effects are associated with lysis leads to release of arginase, an enzyme that degrades
PRBC transfusion. First, although rare, risks are associated arginine, an amino acid that normally stimulates lymphatic
with crossmatching and the transmission of viral and prion function).140
diseases. The risk of transmission of human immunodefi-
ciency virus (HIV), hepatitis C virus, and hepatitis B virus
are 1 in 1,900,000, 1 in 600,000, and 1 in 220,000 respec- Strategies to Prevent Anemia
tively.108 Cytomegalovirus (CMV) is present in 4% of trans- Anemia can be detrimental to ICU patients, but so is its cor-
fusions from healthy donors due to the reactivation of latent rection with blood transfusion. One way to reduce the inci-
CMV in leukocytes.109,110 Other transfusion-transmitted dence of anemia in the ICU is to decrease the amount of
infections are thought to be due to contaminated leuko- phlebotomized blood. Strategies to decrease phlebotomy
cytes.111 Contaminated leukocytes111 may also contribute to include the use of pediatric collection tubes in adults,
infection or febrile nonhemolytic transfusion reactions recycling of discard volumes, POC technology, and new
(FNHTR) and hemolytic transfusion reactions.112 In addi- paradigms in laboratory-ordering practices. The use of
tion, leukocyte activity on RBC membranes during storage pediatric-size laboratory tubes can reduce blood loss by
and degradation during storage that releases oxygen free almost 50%.141 Discard volumes required for “clearing the
radicals and proteases, may incite inflammation in the trans- line” amount to almost a third of daily blood loss in the ICU.19
fusion recipient.113,114 Recycling of discard volumes through the use of blood con-
Transfusion may have detrimental effects on the immune servatory devices can minimize iatrogenic blood loss. POC
system. Transfusion-related immunomodulation may lead to technologies employ rapid bedside analyzers of whole blood
alloimmunization,91 a heightened immune response as in that provide accurate analyses of multiple analytes with
transfusion reactions, or tolerance induction, a suppression of minimal blood sampling. One milliliter of whole blood is suf-
the immune response that predisposes to hospital-acquired ficient for bedside analysis of pH measurements, arterial
infections. Alloimmunization and consequent induction of blood gases, selected electrolytes, glucose, and hemato-
human leukocyte antigen (HLA) antibodies and T cell activa- crit.142,143 Use of another POC device, namely thromboelas-
tion, results in several clinical syndromes, including transfu- tography, has been shown to reduce not only administration
sion reactions, transfusion-associated graft-versus-host of blood components but also bleeding.144,145 Education
disease, transfusion-related acute lung injury (TRALI), and of physicians and nurses to promote judicious laboratory
perhaps various autoimmune diseases.91 TRALI is defined as ordering and testing is paramount. Documentation of phle-
a new episode of respiratory distress not explained by an alter- botomy volume on ICU flow sheets has been shown to effec-
nate etiology that occurs during, or within 6 hours of, a tively decrease iatrogenic blood loss.146 Discontinuation of
Section II—Clinical and Laboratory Assessment 139

laboratory panels and standing laboratory orders may further the fibrinolytic system, which is mediated by plasmin.
reduce unnecessary blood draws. Batching of blood draws can Plasmin is formed from endothelial-derived plasminogen
also minimize the wastage associated with each phlebotomy activators.
attempt. Four bleeding tests have traditionally been used to evaluate
the bleeding patient: the PT, activated partial thromboplastin
time (aPTT), bleeding time, and platelet count.161 The PT
Alternatives to Transfusion measures the integrity of the extrinsic and common pathways,
Because RBCT can worsen outcome, alternative strategies including factors VII, X, and V; prothrombin; and fibrinogen;
have been tried, including stimulation of red cell production whereas the aPTT measures the integrity of the intrinsic
by recombinant human EPO (rhuEPO), Hgb-based oxygen and common pathways, including high molecular weight kal-
carriers, and perfluorocarbons. A detailed discussion of Hgb- likrein (HMWK); prekallikrein; factors XII, XI, IX, VIII, X,
based oxygen carriers and perfluorocarbons is beyond the and V; prothrombin; and fibrinogen.161 Bleeding time is a
scope of this chapter but is available elsewhere.147-149 The use screening test of platelet-endothelial interactions. It measures
of EPO is of particular interest to the NCCU patient popu- the time to cessation of bleeding after a standard incision in
lation because both EPO and its receptor (EPOR) are present the volar aspect of the forearm. It is prolonged in thrombocy-
in neural and vascular tissue. Furthermore, several lines of topenia, vWD, platelet abnormalities, and primary vascular
evidence suggest that EPO may be neuroprotective.150-152 disorders; it is usually not affected by coagulation factor defi-
Whether rhuEPO is useful in the ICU remains unclear, ciencies. This test, however, lacks reproducibility and has poor
because some studies suggest that although transfusion sensitivity and specificity.
needs may be reduced153-156 there is a significant increase in Normal results of these screening tests essentially exclude
the incidence of thrombotic vascular events.157 In addition, clinically significant bleeding. Notable exceptions include
a variety of side effects including seizures, hypertension, factor XIII deficiency and vWD, which may have normal
headache, tachycardia, nausea, and clotted vascular access results to these screening tests. If the bleeding time is pro-
are described.158 Enthusiasm for the use of rhuEPO therefore longed and thrombocytopenia is not present, evaluation for
has waned and many studies of erythropoiesis-stimulating medications (e.g., aspirin) or diseases (e.g., end-stage renal
agents (ESAs) have been halted. In 2008, the U.S. Food and disease [ESRD]) that may affect platelet function is warranted.
Drug Administration (FDA) published a black box warning Prolongation of the PT or partial thromboplastin time (PTT)
on all ESAs for increased risk of mortality, cardiovascular usually indicates a factor deficiency or presence of an inhibi-
and thrombotic events, and tumor progression or recur- tor. A mixing study can discriminate between these etioloiges
rence. Because of its neuroprotective effects, EPO studies because a deficiency should correct with addition of normal
in ischemic stroke, SAH, and TBI continue (clinicaltrials. plasma, whereas the presence of an inhibitor would not correct
gov/sho/NCT00313716 and www.controlled-trials.com/mrct/ the prolonged time.
trial/661629/bellomo). There are promising results from pre-
liminary human randomized controlled trials (RCTs) in
SAH.159 However, mortality was actually greater with EPO Venous Thromboembolism
in a recent multicenter RCT of acute ischemic stroke
patients.160 Venous Thromboembolism in the Intensive
Care Unit
Venous thromboembolism (VTE) is composed of DVT and
Coagulation pulmonary embolism (PE) and occurs in 117 to 145 per
100,000 individuals each year.162 In the United States, the fre-
Hemostasis quency of DVT approaches 2 million people per year,163 with
The coagulation system is normally quiescent. Endothelial fatal PE in 50,000 to 200,000 patients.164,165 The 1-year mortal-
cells that line the intimal surface of blood vessels promote ity rate is as high as 21% for DVT and 39% for PE,161 and PE
smooth blood flow by inhibiting platelet function and fibrin accounts for 5% to 10% of potentially preventable hospital
accumulation.161 Upon vascular injury, the antithrombotic deaths.166 Risk factors for VTE include: (1) age greater than 40
properties of the endothelial monolayer are lost, and pro- years; (2) prolonged immobility; (3) prior VTE; (4) malig-
thrombotic substances contained within the subendothelium nancy; (5) myeloproliferative disorders; (6) cardiac dysfunc-
are exposed, inciting platelet adhesion. This is mediated by tion; (7) stroke; (8) respiratory failure; (9) pregnancy; (10)
vWF, which binds platelets at its receptor in the platelet obesity; (11) varicose veins; (12) estrogen use; (13) inflamma-
membrane and attaches to the damaged vessel wall. Adherent tory bowel disease; (14) nephritic syndrome; (15) femoral vein
platelets release the contents of the storage granules, includ- catheterization; (16) sepsis; (17) major surgery; (18) trauma;
ing ADP and thromboxane A2, thus activating circulating and (19) inherited or acquired thrombophilias. Among hos-
platelets to bind the damaged vessel wall. These activated pitalized patients the risk is much greater in ICU than other
platelets expose binding sites for fibrinogen and lead to fibrin hospitalized patients167,168; in particular, patients who remain
deposition, the final step in the formation of the hemostatic in the ICU more than 72 hours and who have end-stage renal
plug. Fibrin is converted from plasma fibrinogen by throm- failure, personal or family history of VTE, platelet transfusion,
bin. Thrombin is formed from prothrombin, its zymogen and vasopressor therapy are at increased risk.165 Despite
precursor, and activated factors X and V (the common routine thromboprophylaxis, 10% of ICU patients develop
pathway). Factor X is most often activated by the tissue factor lower extremity DVT, and VTE remains one of the most
(extrinsic) pathway, but may be activated by the contact acti- common unsuspected autopsy findings in critically ill
vation (intrinsic) pathway. Fibrin is ultimately digested by patients.169
140 Section II—Clinical and Laboratory Assessment

intracranial bleeding was greater than the benefits of heparin-


Venous Thromboembolism ization, so they concluded that overall outcomes were best
in Neurosurgical Patients with mechanical prophylaxis alone. In a single center study the
Neurosurgical patients have several unique risks for VTE combination of low-dose unfractionated heparin and twice-
including: (1) treatment-related immobilization, (2) disease- weekly surveillance Doppler ultrasound in neurosurgical
related or postoperative paralysis, (3) prolonged surgical pro- patients was associated with an incidence of PE of 0.09% and
cedures (>4 hours), (4) delayed initiation of activity, (5) of DVT of 2.6%, a rate of VTE lower than that in the other
corticosteroid use, and (6) pharmacologic dehydration. In ICUs within the same institution.197 In a randomized trial of
particular patients with TBI, malignant tumors, advanced age 307 patients undergoing elective neurosurgical procedures,
and those who undergo craniotomy (in contrast to spinal 17% of patients with compression stocking therapy and
surgery) are at greater risk.170,171 Kaolin-activated thrombo- enoxaparin developed DVT, whereas 32% who received only
elastography has identified a transient hypercoagulable state compression devices had DVTs.175 The incidence of major
during and after neurosurgical operations; indices of hyper- bleeding was similar. The ninth American College of Chest
coagulability were more pronounced with longer operative Physicians (ACCP) conference on antithrombotic and throm-
times and cranial rather than spinal procedures.172 Brain tissue bolytic therapy recommends mechanical prophylaxis, prefer-
is rich in tissue factor (factor VII), which may incite a pro- ably with intermittent pneumatic compression (IPC) or
thrombotic state. After surgical procedures there is increased pharmacolagic prophylaxis be used over no prophylaxis for
inhibition of tissue plasminogen activator (tPA), a potent clot- patients undergoing craniotomy.198 The risk of a DVT in
dissolving enzyme that converts plasminogen to plasmin.173 patients who undergo elective spinal surgery is low. The 9th
Diminished tPA results in a reduced capacity to clear thrombi. edition ACCP guidelines recommend mechanical prophylaxis,
Local hypoxemia in venous channels that may occur with preferably with IPC, over no prophylaxis (Grade 2c), unfrac-
lengthy neurosurgical procedures or with prolonged inactivity tionated heparin (Grade 2c), or LMWH (Grade 2c) unless
can promote clot formation in as little as 2 hours.174 they have additional risk factors such as advanced age, known
Among neurosurgical patients after craniotomy, the malignancy, presence of a neurologic deficit, previous VTE, or
reported rates of VTE vary widely from zero to 60%.175-182 an anterior surgical approach.199 There also is debate on what
Although the prevalence of PE is between 1.5% and 8.4% to do after TBI or spinal cord injury (SCI). The 9th edition
among neurosurgical patients, the risk of mortality from PEs ACCP guidelines suggest that in the absence of a contraindica-
can be as high as 50% in this population.183,184 The greatest tion, all TBI patients receive LDUH (Grade 2c), LMWH
risk for DVT is in patients with brain tumors (28%-43%), (Grade 2c) or mechanical prophylaxis, preferably with IPC
followed by those undergoing craniotomy (25%), and those (Grade 2c).198 If LMWH is contraindicated, then IPC is rec-
with TBI (20%).185,186 ommended. Inferior vena cava (IVC) filters are not recom-
The overall risk of developing DVT in patients with brain mended as primary prophylaxis in trauma patients. In patients
tumors is 3% to 60%.185-187 The incidence of DVT is generally with acute SCI, which carries the highest risk of VTE, the
considered highest with malignant gliomas, although some ACCP guidelines recommend VTE prophylaxis with the use
meningiomas, especially those that adhere to the sinuses, can of IPC and either low-dose unfractionated heparin (LDUH)
be associated with higher DVT rates.188-191 The pathogenesis or LWMH, once primary hemostasis has been achieved.198
of VTE in brain tumor patients is thought to be associated
with the high concentration of TF, the cell surface receptor of
factor VII/VIIa, which plays a pivotal role in the initiation of Venous Thromboembolism Prophylaxis
the coagulation cascade. High-grade tumors may express in Stroke Patients
higher concentrations of TF, procoagulants, and fibrinolytic VTE is a common complication of stroke, similar to that for
inhibitors than low-grade tumors.192 Elevated levels of surgical patients, and is associated with increased mortality
D-dimer, homocysteine, lipoprotein (a) (lp[a]), vascular and morbidity.185,198 A meta-analysis of more than 23,000 isch-
endothelial growth factor (VEGF), tPA and plasminogen acti- emic stroke patients suggested that low-dose LMWH therapy
vator inhibitor (PAI) are found in patients with malignant rather than unfractionated heparin (UFH) or placebo had the
glioma and likely contribute to the observed hypercoagulable best risk-benefit profile, decreasing the risk of both DVT and
state.193,194 Chemotherapeutic agents used to treat malignancy, PE without an increased risk of spontaneous intracerebral
including nitrosurea and cisplatin, may also increase the risk hemorrhage (sICH).200 The PREVAIL study, a randomized
of DVT in these patients.195 trial of 1762 ischemic stroke patients, found that 40 mg of
once-daily enoxaparin reduced the risk of VTE by 43% com-
pared with UFH (5000 units subcutaneously twice daily)
Thromboprophylaxis in including inpatients with a National Institutes of Health
Neurosurgical Patients Stroke Scale (NIHSS) score greater than or less than 14.201 The
There is a general reluctance to use pharmacologic prophy- rate of symptomatic ICH was similar but there was a small
laxis for VTE in neurosurgical patients because the conse- increase in the rate of extracranial bleeding in the enoxaparin
quences of bleeding may be devastating. Several studies have group (1% versus 0%).
attempted to determine whether the risk of pharmacoprophy- There is less evidence about optimal VTE prophylaxis in
laxis outweighs the risk of VTE. Danish et al. sought to deter- patients with hemorrhagic strokes, although the risk of
mine the best method of thromboprophylaxis in neurosurgical DVT may be up to four times greater in ICH than in patients
patients by use of a decision analysis model196 that incorpo- with ischemic stroke. The increased risk is likely related to
rated the incidence, prevalence, and likelihood ratios of VTE reduced rates of pharmacoprophylaxis and a higher degree of
and ICH. The addition of heparin lowered the incidence of neurologic impairment. Abrupt cessation and reversal of oral
VTE but increased the incidence of ICH. The impact of anticoagulation also may increase this risk, although one study
Section II—Clinical and Laboratory Assessment 141

found that patients with oral anticoagulation–related hemor- adjusted life-year.217 The authors concluded that optimizing
rhages were not at increased risk of thromboembolic events.202 thromboprophylaxis was a better value in terms of cost and
Review of data from the National Hospital Discharge Survey health gains when compared with routine ultrasound
demonstrates that of 14 million patients with ischemic stroke screening.
and 1.6 million patients with ICH the incidence of PE
and DVT were 0.51% and 0.74% and 0.68% and 1.1%,
respectively.177
Cerebral Venous Thrombosis
The incidence of VTE after ICH may be reduced with the Thrombosis of the cerebral veins and sinuses is a rare but
addition of intermittent pneumatic compression to elastic serious disorder characterized by headache, seizures, focal
stockings203 and may be further reduced with administration neurologic signs, papilledema, and occasionally coma and
of low-dose UFH administered three times a day when started death.218 A prothrombotic factor is found in nearly two thirds
2 days after ICH.204 Pharmacoprophylaxis with either UFH or of cases.219 Cavernous sinus thrombosis is usually caused by
LMWH may safely be initiated 2 days after ictus.204,205 The infection and is characterized by periorbital edema, proptosis,
American Heart Association and American Stroke Association chemosis, papilledema, and ocular paresis.218 Occlusion of the
(AHA-ASA) guidelines for ICH management recommend cerebral veins leads to brain infarction in a nonarterial distri-
intermittent pneumatic compression in addition to elastic bution, petechial hemorrhage, and localized edema, whereas
stockings for prevention of VTE (class I, level B evidence) and occlusion of the venous sinuses can cause decreased venous
consideration of low-dose subcutaneous LMWH or UFH after outflow and increased ICP. Etiologies include antecedent head
1 to 4 days from onset in patients with immobility and docu- trauma, hypercoagulable states, ear and sinus infections (espe-
mented cessation of bleeding.206,207 cially otitis media or mastoiditis with hypoplasia of the con-
tralateral transverse sinus), collagen vascular and hematologic
diseases, oral contraceptive use, pregnancy and the puerpe-
Diagnosis and Surveillance rium, neurosurgical procedures, and dehydration. Lumbar
for Venous Thromboembolism puncture also may lead to sinus thrombosis by traction placed
The presence of clinical symptoms is not a reliable marker on cortical veins associated with downward shift of the brain
of DVT, and hospital-acquired VTE is usually clinically after spinal fluid removal.220
silent.199 Traditional clinical features such as a tender, warm, The frequency of puerperal cerebral venous thrombosis
and swollen calf; a positive Homans’ sign; and venous dila- (CVT) is only slightly less than the incidence of puerperal
tion are unreliable.196 The vast majority of patients with PE arterial strokes. Incidence estimates for CVT in women during
exhibit no stigmata of DVT.168 The highest risk of pulmo- pregnancy and the puerperal period range from 1 per 2500 to
nary embolism is associated with thromboses of the proxi- 1 per 10,000 deliveries in the United States.221,222 CVT does not
mal veins above the popliteal fossa. In less than 1% of preclude future pregnancy; however, formal consultation with
patients, calf vein thrombi cause PE. DVTs of the proximal a hematologist or maternal-fetal medicine specialist is reason-
leg contribute to 90% of pulmonary emboli, whereas the able.219 Pregnant patients with CVT are preferentially treated
remainder is thought to arise from the pelvis or the upper with LMWH rather than UFH. For pregnant women with a
extremities. history of CVT, prophylactic anticoagulation during future
The gold standard diagnostic test to identify a DVT remains pregnancies may be beneficial.219
contrast venography; however, it is being rapidly replaced with In the acute setting, diagnosis should include routine blood
Duplex ultrasonography (DUS) given the invasive nature, studies (CBC, chemistry panel, PT, aPTT), erythrocyte sedi-
technical demands, costs, and potential risks of venogra- mentation rate, and screening for oral contraceptive use and
phy.208,209 For proximal DVT, ultrasonography has diagnostic inflammatory or infectious diseases.219 Testing for prothrom-
sensitivities of 89% to 96% and specificities of 94% to botic conditions including protein C, protein S, antithrombin
99%.209,210 The sensitivity and specificity of DUS is lower for deficiency, antiphospholipid syndrome, prothrombin gene
symptomatic calf DVT, 73% and 93%, respectively.211 It should mutation, and factor V Leiden can be beneficial for the man-
be noted that for asymptomatic patients DUS has a sensitivity agement of patients and is generally indicated 2 to 4 weeks
of only 50%; therefore DUS should only be performed when after anticoagulation has been discontinued. A normal
there is a clinical suspicion of DVT. Radiologic imaging is D-dimer level, by sensitive immunoassay or enzyme-linked
necessary for the diagnosis of PE in patients with at least an immunosorbent assay (ELISA), may be considered to identify
intermediate pretest probability of PE. patients with a low probability of CVT; a normal level should
D-dimer is protein produced when cross-linked fibrin is not preclude diagnosis in a patient with a high clinical suspi-
degraded by plasmin, the principal enzyme involved in fibri- cion.219,223,224 Magnetic resonance imaging with venography is
nolysis. D-dimer has been studied extensively.211 Although a highly sensitive diagnostic tool. Both T1- and T2-weighted
D-dimer levels coupled with a low pretest probability of DVT images demonstrate hyperintense signal from the thrombosed
may reliably exclude DVT in some populations,212 its utility in sinuses. Thrombi are isointense on T1-weighted images early
hospitalized or critically ill patients is limited.213,214 In some in the course of the disease (<5 days). Angiography can be
patients with confirmed DVT, the D-dimer level may be particularly helpful when there is cortical vein thrombosis in
normal.215 the absence of sinus thrombosis. The mainstay of treatment
Routine ultrasound surveillance of lower extremities has is anticoagulation regardless of presence of ICH; duration of
been proposed to decrease the PE rate, especially in neurologi- treatment varies according to etiology (i.e., 3-6 months for
cally injured patients often with altered mental states and limb transient risk factors compared with lifelong treatment
immobility.197,216 A decision and cost effectiveness analysis for those with severe thrombophilias).225,226 Treatment for
demonstrated that adherence to pharmacoprophylaxis resulted refractory CVT includes endovascular thrombectomy,
in fewer DVTs and was eight times less costly per quality thrombolysis, or thromborrhexis.
142 Section II—Clinical and Laboratory Assessment

increased risk of VTE.237 Fibrinogen levels of greater than


Thrombophilias 500 mg/dL are associated with approximately a fourfold
Testing for hypercoagulable states in unselected populations increased risk of VTE.235 The remaining factors have not been
is low yield, although a thrombophilic defect can be demon- shown to be prothrombotic.
strated in more than 50% of the patients who present with a
VTE.227 Thrombophilias increase the risk of first-time VTE,
but it is unclear whether they increase the risk of recurrent Plasminogen Activator Inhibitor 1 and
VTE . Approximately 1% to 4% of ischemic strokes are caused Lipoprotein (a)
by thrombophilias.228 Strokes may be caused by alterations in genetic regulation of
fibrinolysis by plasminogen activator inhibitor (PAI)-1 and
lp(a).227,232 PAI-1 inhibits tissue plasminogen activator activity
Protein C, Protein S, Antithrombin and suppresses fibrinolysis. Lp(a) is a modified low-density
Deficiency lipoprotein that exhibits structural homology to plasminogen,
Activated proteins C and S regulate the coagulation cascade a plasma protein important in dissolving blood clots.
by inactivating factor Va. Protein C deficiency is relatively rare, Lp(a) competes with plasminogen, which interferes with
with a prevalence of approximately 0.2% in the general popu- fibrinolysis.
lation and 2.5% to 6% in patients with first-time VTE.228 Ret-
rospective analyses of family studies indicate that the risk of
VTE in patients with protein C deficiency is approximately 10 Antiphospholipid Syndrome
times that of nondeficient patients. Recurrent thromboses in The antiphospholipid syndrome (APS) is defined by the com-
these patients have been estimated between 4.8% and 17% per bination of thrombosis and/or pregnancy-related morbidity
year.229 The prevalence of protein S deficiency, an autosomal and medium to high titiers of persistent circulating antiphos-
dominant defect, is about 0.03% to 1.6% in the general popu- pholipid antibodies (aPL): lupus anticoagulant (IA), anticar-
lation.230 Among patients with demonstrated VTE, the preva- diolipin antibodies (aCL), and/or anti-b2 glycoprotein I
lence is 1% to 2%. One third of patients with protein S antibodies (anti-b2 GPI).238 aPL antibodies are a heteroge-
deficiency will have a VTE event by the age of 60 years. Anti- neous family of antibodies to membrane phospholipids that
thrombin deficiency occurs 0.5% to 7.5% with a first-time are primarily associated with venous thrombosis and fetal loss,
VTE.230 The inheritance pattern is also autosomal dominant although stroke is the most common arterial thrombotic
and almost all patients are heterozygous. It should be sus- manifestation. The combination of aPL antibodies and
pected when unusually high doses of heparin are required to tobacco or oral contraceptive use significantly increases the
increase bleeding time. risk of thrombosis.239

Factor V Leiden and Prothrombin Evaluation and Treatment


Gene Mutation (G20210A) of Thrombophilias
Factor V Leiden and prothrombin gene mutations are “gain of The inherited thrombophilias rarely contribute to strokes in
function” lesions that result in enhanced thrombosis and pro- adults; therefore, screening for these states is not routinely
coagulation. Inactivation of factor Va occurs by cleavage at recommended.226 For those patients with known inherited
three sites. Failure of cleavage by activated protein C at the thrombophilia who present with stroke, full evaluation for
first site, position 506, is known as factor V Leiden (FVL). alternative etiologies (atherosclerosis, nonvalvular atrial fibril-
Factor Va is inactivated 10 times more slowly than normal in lation) should be performed. However, evaluation for DVT
FVL carriers.231 FVL is responsible for at least 90% of acti- should be considered because presence of DVT may alter
vated protein C–resistant conditions. It is the most prevalent treatment (e.g., anticoagulation). Antiplatelet therapy may be
thrombophilia and is found in 5% to 12% of the general reasonable treatment in stroke patients with low-titer aPL
population.231 antibodies; for patients with APS, treatment with anticoagula-
The second most prevalent thrombophilia in the general tion to a goal target international normalized ratio (INR) of
population is the prothrombin gene mutation. It is found in 2 to 3 or even >3 may be warranted.240
1% of the population and is implicated as a cause in 4% to
8% of patients with first-time VTE. The risk of first thrombo-
sis is 0.46% for a person carrying this gene.232 A prothrombin Hyperhomocysteinemia
level higher than 115% (90th percentile) results in a 2.1-fold Homocystinuria is a rare condition associated with elevated
increased VTE risk.233,234 Data on recurrent events in these levels of homocysteine (>100 µmol/L), marfanoid features,
patients are not as compelling as they are for deficiencies of mental retardation, and premature arterial and venous
proteins C and S and antithrombin III. thromboses. It is caused by mutations in either cystathionine-
β-synthase or methyl tetrahydrofolate reductase. Contrary to
homocystinuria, mild hyperhomocysteinemia is common
Elevated Levels of Coagulation Factors and occurs in about 5% to 10% of the general population.233
Elevated levels of coagulation factors VIII, IX, XI and fibrino- Its clinical relevance and contribution to stroke risk are
gen increase the risk for VTE. Of these, factor VIII is the most unknown. Folate supplementation may be considered in
prothrombotic and in a dose-dependent manner.235-237 Every patients with hyperhomocysteinemia; however, there is no
10% increase in factor VIII results in a 10% increased risk for compelling evidence to suggest that reducing homocysteine
VTE. Elevated factor IX levels are associated with a twofold levels abrogates stroke risk.
Section II—Clinical and Laboratory Assessment 143

the same instruments used to measure the aPTT.257-261 There-


Anticoagulants fore the therapeutic range for the aPTT is laboratory spe-
Parenteral anticoagulants may be broadly divided into cific.261,262 Although many heparin nomograms exist, none is
two groups: indirect and direct anticoagulants. Indirect applicable to all aPTT reagents. For patients who require
anticoagulants rely on plasma cofactors for thrombin inacti- high levels of heparin administration (>35,000 units/day
vation, whereas direct anticoagulants do not. The indirect UFH), it may be reasonable to monitor anti-Xa levels and
anticoagulants include UFH, LMWHs, fondaparinux, and adjust the dose accordingly because this practice lessens the
danaparoid. The direct agents target thrombin and include amount of heparin administered without compromising clin-
recombinant hirudins, bivalirudin and argatroban. ical outcomes.251
The anticoagulant effects of heparin can be reversed with
IV protamine sulfate, a protein that complexes with heparin
Heparin to form a stable salt: 1 mg of IV protamine will neutralize 100
Heparin is a mucopolysaccharide of heterogeneous molecu- units of IV heparin.244 The half-life of heparin is 60 to 90
lar size; it can vary in weight from 3,000 to 30,000 kDa and minutes; therefore, when reversing with protamine, only
averages approximately 45 saccharide units.241-243 Its major heparin administered over the last few hours needs to be con-
anticoagulant effect is mediated by its interaction with anti- sidered when calculating the dose. Caution is needed when
thrombin (AT), but it also inhibits factors Xa, IXa, XIa, and administering protamine because it can cause anaphylaxis.244
XIIa. Heparin binds to AT and converts it from a slow to a The main side effects of heparin therapy are HIT (see earlier)
fast inactivator of thrombin. Heparin binds to AT through a and osteoporosis.
glucosamine unit, which contains a unique pentasaccharide
sequence. One third of heparin chains are of sufficient length
to bridge the heparin/AT to thrombin, forming a ternary AT/ Low Molecular Weight Heparin
heparin/thrombin complex, which potentiates inhibition. LMWHs are derived from UFH and are less active against
Heparin then dissociates from thrombin and is available for thrombin relative to factor Xa.263,264 The anti–Xa-to-anti–IIa
reuse. Smaller chains that contain the high affinity pentasac- ratio of LWMHs is 2 to 4 : 1 (based on molecular size distribu-
charide can inhibit factor Xa, although thrombin is 10-fold tion), whereas the anti–Xa-to-anti–IIa ratio of UFH is 1 : 1.244
more sensitive to inhibition than factor Xa.244 Heparin also LMWHs are one third the molecular weight of UFH, which
has anticoagulant effects independent of the pentasaccharide roughly corresponds to 15 saccharide units. They have a more
sequence. It binds to heparin cofactor II and catalyzes the predictable pharmacokinetic profile than UFH due to a
inactivation of factor IIa; however, this effect is chain length reduced binding affinity for cells and proteins; this also extends
dependent, requiring heparin chains of at least 24 saccharide their plasma half-life.265 The lower incidence of HIT also is
units.245 related to the decrease in platelet binding. The various LMWHs
Heparin is administered intravenously or subcutaneously. have different anticoagulant profiles and are thus not inter-
Subcutaneous administration requires higher dosing given the changeable. Routine laboratory monitoring generally is not
reduced bioavailability. Intravenous (IV) bolus administra- recommended for most patients treated with LMWH. The
tion can provide an immediate effect. Heparin binds plasma ACCP recommends anti-Xa monitoring in some special pop-
proteins, endothelial cells, and macrophages246,247; this phe- ulations, such as in pregnant women who require treatment
nomenon results in an unpredictable anticoagulant response with LMWH.244 Others suggest monitoring in obese patients
and may explain some of the apparent heparin resistance. and those with renal insufficiency. The ACCP recommends
Heparin resistance may also be due to AT deficiency,248 weight-based dosing in the treatment or prophylaxis of obese
increased heparin clearance,249,250 elevated factor VIII,251,252 patients and preferential use of UFH in patients with severe
and elevated fibrinogen.252 renal dysfunction.126-128
The risk of bleeding associated with heparin increases at Although protamine sulfate incompletely neutralizes the
higher doses and with concomitant administration of fibri- anticoagulant effect of LMWHs, it should be administered at
nolytic agents253,254 or GPIIb/IIIa inhibitors.255 Recent surgery, a dose of 1 mg per 100 anti-Xa units of LMWH in the acutely
trauma, invasive procedures, and defects of hemostasis are bleeding patient if it was administered during the preceding 8
also risk factors for bleeding.244 Monitoring of heparin and hours.244 A second dose of protamine sulfate at 0.5 mg per 100
dose adjustment has become standard practice given the units of anti-Xa should be given if bleeding continues. Side
variable anticoagulant response. When given in therapeutic effects of LMWH include HIT and osteoporosis. The fre-
doses, the anticoagulant effect is monitored by the aPTT. The quency of HIT is threefold lower with LMWH; however,
activated clotting time (ACT) is used to monitor high LMWHs can form complexes with PF4 that are capable of
heparin doses administered to patients, such as during endo- binding HIT antibodies and therefore LMWHs should not be
vascular neurosurgical procedures or percutaneous coronary used in the treatment of HIT.
interventions.
Unlike the evidence supporting a target range for the INR
for warfarin monitoring, the data to adjust the dose of Fondaparinux
heparin to target a therapeutic range are weak. An aPTT ratio Fondaparinux is a synthetic analog of the AT-binding penta-
of 1.5 to 2.5 times control was suggested by a retrospective saccharide found in both UFH and LMWH that has higher
study in the 1970s that demonstrated lower risk of VTE; this anti-Xa activity than LMWH and a longer half-life (~17
practice has been widely adopted despite a lack of strong evi- hours).266,267 It does not increase the rate of thrombin inhibi-
dence in its support.256 However, neither has this practice tion by AT. Due to its predictable pharmacokinetics and neg-
been rigorously studied by randomized control trials, nor are ligible nonspecific binding, fondaparinux can be administered
144 Section II—Clinical and Laboratory Assessment

subcutaneously once daily in fixed doses without laboratory and the ACT, but the results are variable and unreli-
monitoring. It is not intended for use in patients with renal able.244,272 New anti-factor IIa assays hold promise to better
insufficiency. Routine monitoring is not recommended, but if monitor the effects of this anticoagulant without a veritable
needed the anticoagulant activity can be measured with antidote.272
anti-Xa assays. The anticoagulant effects of fondaparinux are
not reversible with protamine sulfate; recombinant factor VIIa
may be effective.268 There have been no reports of HIT with Argatroban
fondaparinux, and it has been used to safely treat patients with Argatroban is a reversible thrombin inhibitor with a 45-
this disorder.269 minute plasma half-life. It is hepatically metabolized via the
cytochrome p450 system and caution must be used in the
face of liver dysfunction.244 It is indicated for the treatment
Danaparoid of HIT and as an alternative to heparin for PCI in patients
Danaparoid is a mixture of glycosaminoglycans (heparan with HIT. All of the direct thrombin inhibitors increase the
sulfate, dermatan sulfate, and chondroitin sulfate); its antico- INR, but this is especially true of argatroban. This compli-
agulant properties are derived from inhibition of factor Xa in cates the transition from argatroban to vitamin K antago-
an AT-dependent manner.244 Its use is limited to treatment of nists. Some target a goal INR greater than 4 when vitamin K
HIT. An advantage of danaparoid is that it does not prolong antagonists (VKAs) are given in conjunction with direct
INR, which is beneficial when transitioning patients to vitamin thrombin inhibitors; however, monitoring a chromogenic
K antagonists. However, the 25-hour half-life is a significant assay of factor X levels may be a safer practice.244
disadvantage because there is no antidote for it.
Oral Direct Thrombin Inhibitors
Direct Anticoagulants The need for a fixed-dose oral anticoagulant with a broader
In contrast to the indirect anticoagulants, the direct thrombin therapeutic window and lower inter- and intrapatient vari-
inhibitors bind to thrombin and block its enzymatic activity. ability that does not require serial laboratory testing and dose
The aPTT is used to monitor the anticoagulant effects of adjustment led to the novel anticoagulants dabigatran, rivar-
direct thrombin inhibitors; however, this test is not always oxaban, apixaban, and edoxaban. These agents inhibit throm-
accurate. The dose response is not linear and the aPTT pla- bin (dabigatran) or factor Xa (rivaroxaban, apixaban, and
teaus at high doses of these drugs. The ecarin clotting time edoxaban) directly rather than through a cofactor. Although
may be more accurate, but lack of availability and standardiza- they reversibly bind their substrate, their anticoagulant effect
tion generally precludes its routine use. is irreversible because there is no veritable antidote. These
agents provide rapid onset to therapeutic effect in contrast to
warfarin. They all demonstrate low variability in pharmaco-
Hirudin dynamic characteristics, thus obviating the need for routine
Hirudin, derived from the salivary gland of the medicinal coagulation monitoring.
leech, is available in recombinant form as lepirudin or desi- Apixaban has demonstrated efficacy and safety for the pre-
rudin. They both irreversibly inactivate thrombin. They have vention of venous thromboembolism after elective hip or knee
an identical pharmacokinetic profile and mechanism of replacement273,274 and to prevent stroke in patients with non-
action; the amino-terminal domain interacts with active site valvular atrial fibrillation (AF).275 The FDA has approved riva-
of thrombin, and the carboxy-terminus binds to the roxaban for the prevention of stroke secondary to AF.
substrate-binding site. The plasma half-life of the hirudins is Dabigatran has been compared with warfarin to prevent
60 minutes when administered IV and 120 minutes when stroke and systemic embolization in more than 18,000 patients
given subcutaneously.270 Hirudins are renally excreted and so with AF.276 At a dose of 110 mg twice daily, dabigatran proved
are contraindicated in patients with renal failure.271 When noninferiority to warfarin and demonstrated lower rates of
used to treat HIT, the anticoagulant effect of lepirudin is symptomatic ICH. A higher dose of 150 mg twice daily was
monitored by the aPTT with the goal ratio between 1.5 and associated with lower rates of stroke but similar rates of symp-
2.5. When administered for thromboprophylaxis, routine tomatic hemorrhage compared with warfarin. Although these
monitoring is unnecessary.244 oral anticoagulants have proven effective at reducing second-
Antibodies against hirudin develop in 40% of patients ary stroke due to AF, concerns remain about the lack of revers-
treated with lepirudin.244 These antibodies generally do not ibility of its anticoagulant effect.
have much clinical impact; however, analphylaxis may occur
if patients with antibodies are reexposed to hirudin.
Vitamin K Antagonists
Warfarin is a vitamin K antagonist (VKA) that produces an
Bivalirudin anticoagulant effect by obstructing the cyclic interconversion
Bivalirudin is a hirudin analog that reversibly binds to the of vitamin K and its 2,3 epoxide, thereby modulating
active site of thrombin. Bivalirudin has a plasma half-life γ-carboxylation of glutamate residues on the N-terminal
of 25 minutes after IV injection. Bivalirudin is licensed as regions of vitamin K–dependent proteins. Not only do VKAs
an alternative to heparin for patients undergoing percuta- have anticoagulant properties due to inhibition of vitamin
neous coronary interventions (PCIs) in patients with HIT. K–dependent coagulation factors II, VII, IX, and X, but they
In contrast to hirudin, bivalirudin is not immunogenic. also may have procoagulant properties by inhibiting carbox-
The anticoagulant effect may be monitored with the aPTT ylation of the regulatory anticoagulant proteins C, S, and Z.277
Section II—Clinical and Laboratory Assessment 145

VKAs are used to treat venous thromboembolism, atrial fibril- generation through cleavage of a peptide substrate.143 Inac-
lation, and valvular heart disease.198 They are complex agents curacies may be predicted by the principle underlying the
with a narrow therapeutic index and many food and drug technology. For example, POCT monitors that measure capil-
interactions. VKAs require routine monitoring and frequent lary blood flow may give lower INR values for samples with a
dose adjustments to maintain their safety and efficacy. high Hct.
Advances in warfarin management have been made through
dedicated anticoagulation clinics and patient self-testing and
management. Despite these advances, in 2007 more than Activated Partial Thromboplastin Time
60,000 emergency department visits were related to warfarin The aPTT may be used to screen preoperatively for abnor-
therapy, the majority of which were for acute hemorrhage.278 malities in hemostasis (especially when used with the PT) or
Health care resources dedicated to warfarin management sub- to guide heparin dosing. A number of studies have shown that
stantially exceed the cost of the drug itself. POCT devices can accurately measure the aPTT and thus
guide management of patients on UFH during a variety of
procedures.279-284 However, the correlation between POCT
Point of Care Testing aPTT and central laboratory aPTT remains controversial.285-287
POCT is diagnostic testing performed with rapid generation Moreover, POC aPTT may be more susceptible to effects of
of results at or near the site of the patient in order to optimize other medications such as aprotinin or epsilon aminocaproic
patient care. These assays often do not require laboratory acid.288 The demand for POCT aPTT has been low and the
support, are easy to perform, and efficiently deliver results. reliability of the results remains suspect. Only the Hemochron
Some of these tests use capillary blood and others use venous device remains available.289
blood or citrated samples. Careful attention to manufacturers’
recommendations is necessary because erroneous values may
be obtained if the inappropriate sample type is used. This Activated Clotting Time
discussion focuses on those POCTs with a self-contained The ACT is exclusively used to monitor anticoagulation with
reagent system. Much of this literature has been driven by use high doses of UFH (e.g., during cardiac bypass surgery or
of POCT in cardiac surgery, because these patients have plate- interventional vascular procedures) because high concentra-
let dysfunction and high bleeding risk and require antiplatelet tions of UFH result in an unclottable aPTT. A baseline ACT
and anticoagulant medications. should be obtained on all patients before UFH administration.
Some POCTs do not contain a self-contained reagent system The ACT may be affected by the choice of activator, hypother-
and may require samples to be pipetted or centrifuged. Quality mia, hemodilution, or medications. Currently available POCT
assurance may be limited because there are no formal external devices for the aPTT include the Hepcon Haemostasis Man-
quality assessment measures for many of these devices, agement System (HMS), the Hemochron Response device, the
although some commercially available quality control samples Actalyke MAX-ACT, the Hemochron Junior II and the i-Stat
with established reference ranges are available. Internal quality analyzer.
controls also must be performed routinely to ensure reliability
of results. Maintenance of current standard operating proce-
dures specifying sample collection, handling and storing prac- Estimations and Modifications
tices, selection of appropriate reagents, and generation of of the Thrombin Time
adequate records and reporting are mandatory for optimal Historically, the thrombin time was used to monitor patients
utility. The quality of the results also depends on personnel on UFH; however, there is a poor correlation between heparin
and frequency of practice. The combination of internal and concentration and thrombin time. Furthermore, at high con-
external quality assurance is imperative to provide both centrations of heparin, the thrombin time becomes unclot-
precise and accurate results. table and therefore is unsuitable to monitor patients treated
Practical limitations of POCT devices include (1) difficulty with high UFH doses. The high dose thrombin time (HiTT,
in obtaining sufficient sample volume; (2) difficulty in access- Hemochron) is more specific to heparin effects than the
ing test strip or application reservoir; (3) insufficient time to ACT.290 In contrast to the ACT, the HiTT is not affected by
deliver blood sample; (4) inaccuracies based on presence of hypothermia or hemodilution. However, there is poor correla-
heparin or other medications/drugs; and (5) ineligibility for tion between the HiTT and the ACT, which has been previ-
patients with blood abnormalities such as presence of lupus ously validated in the cardiac surgery population. The Hepcon
anticoagulant or extremes of Hct. HMS establishes whole blood heparin concentration using an
automated heparin-protamine titration method. There are
some concerns that HMS may overestimate the heparin dose
Prothrombin Time/International required to maintain adequate anticoagulation as compared
Normalized Ratio with the ACT method.291
Historically, blood product replacement after surgery has been
based on clinical judgment. The POCT PT/INR can guide
blood product administration expediently in patients who Thromboelastography
undergo major surgery. Different devices are based on differ- Thromboelastography (TEG) and rotational thromboelas-
ent principles to generate the INR value. Some use light reflec- tometry (ROTEM) are POCTs that measure clot formation,
tance properties as metal particles move through a magnetic clot strength, and clot dissolution. It has been suggested that
field, whereas others monitor movement of blood in capillary this technology better reflects in vivo hemostasis and thus may
tubes, changes in electrochemical impedence, or thrombin provide more information than standard coagulation assays,
146 Section II—Clinical and Laboratory Assessment

especially in the perioperative setting. TEG analyzes hemostasis of anemia, through serial laboratory testing, exacerbates the
using whole blood, which better accounts for the effect of cel- condition. Anemic patients may be at higher risk of cerebral
lular elements in the coagulation process than the traditional ischemia. Neurosurgical patients are among the most frequent
“cascade” model. The cascade model of coagulation is helpful of surgical populations to develop DVT and fatal PE given
to understand hemostasis in vitro but does not fully explain postoperative immobility, hemiparesis, and altered mental
it in vivo. TEG has been shown to significantly reduce the use state. Combined modalities of thromboprophylaxis and inten-
of blood component therapy144,145 and overall blood loss.292 sive surveillance may prove the best method to identify and
TEG yields several parameters of clotting.293 The reaction treat VTE. Understanding the pathogenesis of these various
time, R, represents the time until to initial fibrin formation conditions, suspicion that they may exist, and multimodality
after mechanical stress and reflects the ability to generate monitoring in the NCCU may identify these disorders earlier,
thrombin. Prolongation of this time reflects anticoagulant thereby minimizing the devastating impact of these common
activity, whereas a shortened R time suggests hypercoagulabil- ailments.
ity. The alpha, or angle parameter, represents the rate of fibrin
generation and cross linkage. The faster the rate of fibrin
generation, the larger is the measured angle. The K value also References
measures clot kinetics; specifically it assesses the time it takes 1. Bunn HF. Approach to the anemias. 24th ed. Philadelphia: Elsevier; 2012.
to reach amplitude of 20 mm. The maximum amplitude 2. Marks PW, Glader B. Approach to anemia in the adult and child. 5th ed.
(MA) represents the tensile strength of the clot; the stronger Philadelphia: Churchill Livingstone; 2009.
3. Ginder G. Microcytic and hypochromic anemias. In: Goldman L, Schafer A,
the clot, the larger the MA. Platelets contribute most to clot editors. Goldman’s Cecil Textbook of Medicine. 24th ed. Philadelphia:
strength and therefore contribute most significantly (80%) to Elsevier; 2012. p. 1039–44.
the MA, whereas fibrinogen, which binds platelets, contrib- 4. Nutritional anaemias. Report of a WHO scientific group. World Health
utes about 20%. The G value is an exponential expression of Organ Tech Rep Ser 1968;405:5–37.
the MA; it is more sensitive to changes in platelet function 5. Hebert PC, Wells G, Tweeddale M, et al. Does transfusion practice affect
mortality in critically ill patients? Transfusion Requirements in Critical Care
than the MA at higher values. The coagulation index (CI) (TRICC) investigators and the Canadian Critical Care Trials Group. Am J
represents the linear relationship between the R value, the K Respir Crit Care Med 1997;155(5):1618–23.
value, the angle, and MA. A CI greater than 3 represents 6. Corwin HL. Blood transfusion in the critically ill patient. Dis Mon
hypercoagulability. The estimated percent lysis (EPL) and 1999;45(10):409–26.
7. Groeger JS, Guntupalli KK, Strosberg M, et al. Descriptive analysis of critical
LY30 (lysis after 30 minutes) are measures of clot dissolution. care units in the United States: patient characteristics and intensive care unit
The EPL is the percent change in the MA over time and the utilization. Crit Care Med 1993;21(2):279–91.
LY30 is the amplitude 30 minutes after the MA divided by the 8. Carson JL, Duff A, Poses RM, et al. Effect of anaemia and cardiovascular
MA. An EPL greater than 7.5% and an LY30 greater than 15% disease on surgical mortality and morbidity. Lancet 1996;348(9034):
represent fibrinolysis; if associated with a CI less than 1 is it 1055–60.
9. Spiess BD, Ley C, Body SC, et al. Hematocrit value on intensive care
primary fibrinolysis with a tendency toward bleeding, and if unit entry influences the frequency of Q-wave myocardial infarction after
CI is greater than 3 it is secondary fibrinolysis, not character- coronary artery bypass grafting. The Institutions of the Multicenter Study of
ized by bleeding. Perioperative Ischemia (McSPI) research group. J Thorac Cardiovasc Surg
The analysis was originally performed on whole blood 1998;116(3):460–7.
10. Kuriyan M, Carson JL. Anemia and clinical outcomes. Anesthesiol Clin
without an activator. Subsequent modifications have been North America 2005;23(2):315–25, vii.
developed that allow faster result generation and test stan- 11. McKechnie RS, Smith D, Montoye C, et al. Prognostic implication of anemia
dardization. The choice of activator depends on the clinical on in-hospital outcomes after percutaneous coronary intervention.
setting. Rapid-thromboelastography (r-TEG), uses TF as the Circulation 2004;110(3):271–7.
activator; it allows for faster curve generation but compro- 12. Chant C, Wilson G, Friedrich JO. Anemia, transfusion, and phlebotomy
practices in critically ill patients with prolonged ICU length of stay: a cohort
mises some of the results (e.g., the R value). Without activa- study. Crit Care 2006;10(5):R140.
tion TEG takes 30 to 60 minutes to provide all information. 13. Corwin HL, Gettinger A, Pearl RG, et al. The CRIT Study: anemia and blood
Quality assurance needs to be performed daily. transfusion in the critically ill—current clinical practice in the United States.
Since 2000, a variety of POC testing (POCT) devices have Crit Care Med 2004;32(1):39–52.
14. Vincent JL, Baron JF, Reinhart K, et al. Anemia and blood transfusion in
become available to examine platelet function and measure critically ill patients. JAMA 2002;288(12):1499–507.
the effect of antiplatelet medications (see previous section on 15. Smoller BR, Kruskall MS. Phlebotomy for diagnostic laboratory tests in
Platelet Function Assays). adults: pattern of use and effect on transfusion requirements. N Engl J Med
1986;314(19):1233–5.
16. Shander A. Anemia in the critically ill. Crit Care Clin 2004;20(2):159–78.
D-Dimer Assays 17. Faquin WC, Schneider TJ, Goldberg MA. Effect of inflammatory cytokines
on hypoxia-induced erythropoietin production. Blood 1992;79(8):1987–94.
Combined with a pretest probability score, D-dimer assays can 18. Rogiers P, Zhang H, Leeman M, et al. Erythropoietin response is blunted in
be used to reliably exclude a diagnosis of VTE. Some D-dimer critically ill patients. Intensive Care Med 1997;23(2):159–62.
assays use capillary blood whereas others use heparinized 19. Rodriguez RM, Corwin HL, Gettinger A, et al. Nutritional deficiencies and
blunted erythropoietin response as causes of the anemia of critical illness.
blood or citrated plasma. The sensitivity of POC D-dimer tests J Crit Care 2001;16(1):36–41.
appears to be less sensitive than the best available clinical labo- 20. Buttarello M, Plebani M. Automated blood cell counts: state of the art. Am J
ratory tests.211,294-296 Clin Pathol 2008;130(1):104–16.
21. Stachon A, Kempf R, Holland-Letz T, et al. Daily monitoring of nucleated
red blood cells in the blood of surgical intensive care patients. Clin Chim
Conclusion Acta 2006;366:329–35.
22. Parisotto R, Wu M, Ashenden MJ, et al. Detection of recombinant human
Anemia, bleeding, and thrombosis occur frequently in the erythropoietin abuse in athletes utilizing markers of altered erythropoiesis.
NCCU, and their treatment can be complicated. The diagnosis Haematologica. 2001;86:128–37.
Section II—Clinical and Laboratory Assessment 147

23. Zini G, DiMario A, Garzia M. Clinical usefulness of red cell fragments 37. Oddo M, Milby A, Chen I, et al. Hemoglobin concentration and cerebral
identified by the Sysmex XE-2100 hematological analyser [lecture]. metabolism in patients with aneurysmal subarachnoid hemorrhage. Stroke
Proceeding of the Sysmex European Haematology Symposium. Lisbon, 2009;40:1275–81.
Portugal; June 13-14, 2007. 38. Leal-Noval SR, Munoz-Gomez M, Murillo-Cabezas F. Optimal hemoglobin
24. Lesesve JF, Salignac S, Alla F, et al. Comparative evaluation of schistocyte concentration in patients with subarachnoid hemorrhage, acute ischemic
counting by an automated method and by microscopic determination. Am J stroke and traumatic brain injury. Curr Opin Crit Care 2008;14(2):
Clin Pathol 2004;121:739–45. 156–62.
25. Berkow L, Rotolo S, Mirski E. Continuous noninvasive hemoglobin 39. Sahuquillo J, Poca MA, Garnacho A, et al. Early ischaemia after severe head
monitoring during complex spine surgery. Anesth Analg 2011;113(6): injury. Preliminary results in patients with diffuse brain injuries. Acta
1396–402. Neurochir (Wien) 1993;122(3-4):204–14.
26. Frasca D, Dahyot-Fizelier C, Catherine K, et al. Accuracy of a continuous 40. Vespa P, Bergsneider M, Hattori N, et al. Metabolic crisis without brain
noninvasive hemoglobin monitor in intensive care unit patients. Crit Care ischemia is common after traumatic brain injury: a combined microdialysis
Med 2011;39(10):2277–82. and positron emission tomography study. J Cereb Blood Flow Metab
27. Lamhaut L, Apriotesei R, Combes X, et al. Comparison of the accuracy of 2005;25(6):763–74.
noninvasive hemoglobin monitoring by spectrophotometry (SpHb) and 41. Menon DK. Brain ischaemia after traumatic brain injury: lessons from
HemoCue with automated laboratory hemoglobin measurement. 15O2 positron emission tomography. Curr Opin Crit Care 2006;12(2):
Anesthesiology 2011;115(3):548–54. 85–9.
28. Wartenberg KE, Schmidt JM, Claassen J, et al. Impact of medical 42. Graham DI, Adams JH, Doyle D. Ischaemic brain damage in fatal
complications on outcome after subarachnoid hemorrhage. Crit Care Med non-missile head injuries. J Neurol Sci 1978;39(2-3):213–34.
2006;34(3):617–23; quiz 24. 43. Vespa PM. The implications of cerebral ischemia and metabolic dysfunction
29. Kramer AH, Gurka MJ, Nathan B, et al. Complications associated with for treatment strategies in neurointensive care. Curr Opin Crit Care
anemia and blood transfusion in patients with aneurysmal subarachnoid 2006;12(2):119–23.
hemorrhage. Crit Care Med 2008;36(7):2070–5. 44. McLaughlin MR, Marion DW. Cerebral blood flow and vasoresponsivity
30. Van Beek JG, Mushkudiani NA, Steyerberg EW, et al. Prognostic value of within and around cerebral contusions. J Neurosurg 1996;85(5):871–6.
admission laboratory parameters in traumatic brain injury: results from the 45. Carlson AP, Schermer CR, Lu SW. Retrospective evaluation of anemia and
IMPACT study. J Neurotrauma 2007;24(2):315–28. transfusion in traumatic brain injury. J Trauma 2006;61(3):567–71.
31. Salim A, Hadjizacharia P, DuBose J, et al. Role of anemia in traumatic brain 46. Diringer MN, Bleck TP, Claude Hemphill J, 3rd, et al. Critical care
injury. J Am Coll Surg 2008;207(3):398–406. management of patients following aneurysmal subarachnoid hemorrhage:
32. Steyerberg EW, Mushkudiani N, Perel P, et al. Predicting outcome after recommendations from the Neurocritical Care Society’s Multidisciplinary
traumatic brain injury: development and international validation of Consensus Conference. Neurocrit Care 2011;15(2):211–40.
prognostic scores based on admission characteristics. PLoS Med 47. Le Roux PD. Anemia and transfusion after subarachnoid hemorrhage.
2008;5(8):e165; discussion e. Neurocrit Care 2011;15(2):342–53.
33. Kramer AH, Zygun DA, Bleck TP, et al. Relationship between hemoglobin 48. Vincent JL, Yagushi A, Pradier O. Platelet function in sepsis. Crit Care Med
concentrations and outcomes across subgroups of patients with aneurysmal 2002;30(5 Suppl):S313–17.
subarachnoid hemorrhage. Neurocrit Care 2009;10(2):157–65. 49. Cawley MJ, Wittbrodt ET, Boyce EG, et al. Potential risk factors associated
34. Kimberly WT, Wu O, Arsava EM, et al. Lower hemoglobin correlates with with thrombocytopenia in a surgical intensive care unit. Pharmacotherapy
larger stroke volumes in acute ischemic stroke. Cerebrovasc Dis Extra 1999;19(1):108–13.
2011;1(1):44–53. 50. Baughman RP, Lower EE, Flessa HC, et al. Thrombocytopenia in the
35. Zygun DA, Nortje J, Hutchinson PJ, et al. The effect of red blood cell intensive care unit. Chest 1993;104(4):1243–7.
transfusion on cerebral oxygenation and metabolism after severe traumatic
brain injury. Crit Care Med 2009;37(3):1074–8.
36. Kurtz P, Schmidt JM, Claassen J, et al. Anemia is associated with metabolic A complete list of references for this chapter can be found online at
distress and brain tissue hypoxia after subarachnoid hemorrhage. Neurocrit www.expertconsult.com.
Care 2010;13(1):10–6.
Section II—Clinical and Laboratory Assessment 147.e1

References 28. Wartenberg KE, Schmidt JM, Claassen J, et al. Impact of medical
complications on outcome after subarachnoid hemorrhage. Crit Care Med
1. Bunn HF. Approach to the anemias. 24th ed. Philadelphia: Elsevier; 2012. 2006;34(3):617–23; quiz 24.
2. Marks PW, Glader B. Approach to anemia in the adult and child. 5th ed. 29. Kramer AH, Gurka MJ, Nathan B, et al. Complications associated with
Philadelphia: Churchill Livingstone; 2009. anemia and blood transfusion in patients with aneurysmal subarachnoid
3. Ginder G. Microcytic and hypochromic anemias. In: Goldman L, Schafer A, hemorrhage. Crit Care Med 2008;36(7):2070–5.
editors. Goldman’s Cecil Textbook of Medicine. 24th ed. Philadelphia: 30. Van Beek JG, Mushkudiani NA, Steyerberg EW, et al. Prognostic value of
Elsevier; 2012. p. 1039–44. admission laboratory parameters in traumatic brain injury: results from the
4. Nutritional anaemias. Report of a WHO scientific group. World Health IMPACT study. J Neurotrauma 2007;24(2):315–28.
Organ Tech Rep Ser 1968;405:5–37. 31. Salim A, Hadjizacharia P, DuBose J, et al. Role of anemia in traumatic brain
5. Hebert PC, Wells G, Tweeddale M, et al. Does transfusion practice affect injury. J Am Coll Surg 2008;207(3):398–406.
mortality in critically ill patients? Transfusion Requirements in Critical 32. Steyerberg EW, Mushkudiani N, Perel P, et al. Predicting outcome after
Care (TRICC) investigators and the Canadian Critical Care Trials Group. traumatic brain injury: development and international validation of
Am J Respir Crit Care Med 1997;155(5):1618–23. prognostic scores based on admission characteristics. PLoS Med
6. Corwin HL. Blood transfusion in the critically ill patient. Dis Mon 2008;5(8):e165; discussion e.
1999;45(10):409–26. 33. Kramer AH, Zygun DA, Bleck TP, et al. Relationship between hemoglobin
7. Groeger JS, Guntupalli KK, Strosberg M, et al. Descriptive analysis of concentrations and outcomes across subgroups of patients with aneurysmal
critical care units in the United States: patient characteristics and intensive subarachnoid hemorrhage. Neurocrit Care 2009;10(2):157–65.
care unit utilization. Crit Care Med 1993;21(2):279–91. 34. Kimberly WT, Wu O, Arsava EM, et al. Lower hemoglobin correlates with
8. Carson JL, Duff A, Poses RM, et al. Effect of anaemia and cardiovascular larger stroke volumes in acute ischemic stroke. Cerebrovasc Dis Extra
disease on surgical mortality and morbidity. Lancet 1996;348(9034): 2011;1(1):44–53.
1055–60. 35. Zygun DA, Nortje J, Hutchinson PJ, et al. The effect of red blood cell
9. Spiess BD, Ley C, Body SC, et al. Hematocrit value on intensive care unit transfusion on cerebral oxygenation and metabolism after severe traumatic
entry influences the frequency of Q-wave myocardial infarction after brain injury. Crit Care Med 2009;37(3):1074–8.
coronary artery bypass grafting. The Institutions of the Multicenter Study 36. Kurtz P, Schmidt JM, Claassen J, et al. Anemia is associated with metabolic
of Perioperative Ischemia (McSPI) research group. J Thorac Cardiovasc distress and brain tissue hypoxia after subarachnoid hemorrhage. Neurocrit
Surg 1998;116(3):460–7. Care 2010;13(1):10–6.
10. Kuriyan M, Carson JL. Anemia and clinical outcomes. Anesthesiol Clin 37. Oddo M, Milby A, Chen I, et al. Hemoglobin concentration and cerebral
North America 2005;23(2):315–25, vii. metabolism in patients with aneurysmal subarachnoid hemorrhage. Stroke
11. McKechnie RS, Smith D, Montoye C, et al. Prognostic implication of 2009;40:1275–81.
anemia on in-hospital outcomes after percutaneous coronary intervention. 38. Leal-Noval SR, Munoz-Gomez M, Murillo-Cabezas F. Optimal hemoglobin
Circulation 2004;110(3):271–7. concentration in patients with subarachnoid hemorrhage, acute ischemic
12. Chant C, Wilson G, Friedrich JO. Anemia, transfusion, and phlebotomy stroke and traumatic brain injury. Curr Opin Crit Care 2008;14(2):156–62.
practices in critically ill patients with prolonged ICU length of stay: a 39. Sahuquillo J, Poca MA, Garnacho A, et al. Early ischaemia after severe head
cohort study. Crit Care 2006;10(5):R140. injury. Preliminary results in patients with diffuse brain injuries. Acta
13. Corwin HL, Gettinger A, Pearl RG, et al. The CRIT Study: anemia and Neurochir (Wien) 1993;122(3-4):204–14.
blood transfusion in the critically ill—current clinical practice in the 40. Vespa P, Bergsneider M, Hattori N, et al. Metabolic crisis without brain
United States. Crit Care Med 2004;32(1):39–52. ischemia is common after traumatic brain injury: a combined microdialysis
14. Vincent JL, Baron JF, Reinhart K, et al. Anemia and blood transfusion in and positron emission tomography study. J Cereb Blood Flow Metab
critically ill patients. JAMA 2002;288(12):1499–507. 2005;25(6):763–74.
15. Smoller BR, Kruskall MS. Phlebotomy for diagnostic laboratory tests in 41. Menon DK. Brain ischaemia after traumatic brain injury: lessons from
adults: pattern of use and effect on transfusion requirements. N Engl J Med 15O2 positron emission tomography. Curr Opin Crit Care 2006;12(2):
1986;314(19):1233–5. 85–9.
16. Shander A. Anemia in the critically ill. Crit Care Clin 2004;20(2):159–78. 42. Graham DI, Adams JH, Doyle D. Ischaemic brain damage in fatal
17. Faquin WC, Schneider TJ, Goldberg MA. Effect of inflammatory cytokines non-missile head injuries. J Neurol Sci 1978;39(2-3):213–34.
on hypoxia-induced erythropoietin production. Blood 1992;79(8):1987–94. 43. Vespa PM. The implications of cerebral ischemia and metabolic
18. Rogiers P, Zhang H, Leeman M, et al. Erythropoietin response is blunted in dysfunction for treatment strategies in neurointensive care. Curr Opin Crit
critically ill patients. Intensive Care Med 1997;23(2):159–62. Care 2006;12(2):119–23.
19. Rodriguez RM, Corwin HL, Gettinger A, et al. Nutritional deficiencies and 44. McLaughlin MR, Marion DW. Cerebral blood flow and vasoresponsivity
blunted erythropoietin response as causes of the anemia of critical illness. within and around cerebral contusions. J Neurosurg 1996;85(5):871–6.
J Crit Care 2001;16(1):36–41. 45. Carlson AP, Schermer CR, Lu SW. Retrospective evaluation of anemia and
20. Buttarello M, Plebani M. Automated blood cell counts: state of the art. Am transfusion in traumatic brain injury. J Trauma 2006;61(3):567–71.
J Clin Pathol 2008;130(1):104–16. 46. Diringer MN, Bleck TP, Claude Hemphill J, 3rd, et al. Critical care
21. Stachon A, Kempf R, Holland-Letz T, et al. Daily monitoring of nucleated management of patients following aneurysmal subarachnoid hemorrhage:
red blood cells in the blood of surgical intensive care patients. Clin Chim recommendations from the Neurocritical Care Society’s Multidisciplinary
Acta 2006;366:329–35. Consensus Conference. Neurocrit Care 2011;15(2):211–40.
22. Parisotto R, Wu M, Ashenden MJ, et al. Detection of recombinant human 47. Le Roux PD. Anemia and transfusion after subarachnoid hemorrhage.
erythropoietin abuse in athletes utilizing markers of altered erythropoiesis. Neurocrit Care 2011;15(2):342–53.
Haematologica. 2001;86:128–37. 48. Vincent JL, Yagushi A, Pradier O. Platelet function in sepsis. Crit Care Med
23. Zini G, DiMario A, Garzia M. Clinical usefulness of red cell fragments 2002;30(5 Suppl):S313–17.
identified by the Sysmex XE-2100 hematological analyser [lecture]. 49. Cawley MJ, Wittbrodt ET, Boyce EG, et al. Potential risk factors associated
Proceeding of the Sysmex European Haematology Symposium. Lisbon, with thrombocytopenia in a surgical intensive care unit. Pharmacotherapy
Portugal; June 13-14, 2007. 1999;19(1):108–13.
24. Lesesve JF, Salignac S, Alla F, et al. Comparative evaluation of schistocyte 50. Baughman RP, Lower EE, Flessa HC, et al. Thrombocytopenia in the
counting by an automated method and by microscopic determination. Am intensive care unit. Chest 1993;104(4):1243–7.
J Clin Pathol 2004;121:739–45. 51. Stephan F, Hollande J, Richard O, et al. Thrombocytopenia in a surgical
25. Berkow L, Rotolo S, Mirski E. Continuous noninvasive hemoglobin ICU. Chest 1999;115(5):1363–70.
monitoring during complex spine surgery. Anesth Analg 52. Hanes SD, Quarles DA, Boucher BA. Incidence and risk factors of
2011;113(6):1396–402. thrombocytopenia in critically ill trauma patients. Ann Pharmacother
26. Frasca D, Dahyot-Fizelier C, Catherine K, et al. Accuracy of a continuous 1997;31(3):285–9.
noninvasive hemoglobin monitor in intensive care unit patients. Crit Care 53. Moreau D, Timsit JF, Vesin A, et al. Platelet count decline: an early
Med 2011;39(10):2277–82. prognostic marker in critically ill patients with prolonged ICU stays. Chest
27. Lamhaut L, Apriotesei R, Combes X, et al. Comparison of the accuracy of 2007;131(6):1735–41.
noninvasive hemoglobin monitoring by spectrophotometry (SpHb) and 54. Nijsten MW, ten Duis HJ, Zijlstra JG, et al. Blunted rise in platelet count in
HemoCue with automated laboratory hemoglobin measurement. critically ill patients is associated with worse outcome. Crit Care Med
Anesthesiology 2011;115(3):548–54. 2000;28(12):3843–6.
147.e2 Section II—Clinical and Laboratory Assessment

55. Napolitano LM, Warkentin TE, Almahameed A, et al. Heparin-induced 83. Ntaios G, Gurer O, Faouzi M, et al. Hypertension is an independent
thrombocytopenia in the critical care setting: diagnosis and management. predictor of mean platelet volume in patients with acute ischaemic stroke.
Crit Care Med 2006;34(12):2898–911. Intern Med J 2011;41(9):691–5.
56. Gawaz M, Dickfeld T, Bogner C, et al. Platelet function in septic 84. Ha SI, Choi DH, Ki YJ, et al. Stroke prediction using mean platelet volume
multiple organ dysfunction syndrome. Intensive Care Med 1997;23(4): in patients with atrial fibrillation. Platelets 2011;22(6):408–14.
379–85. 85. Ilhan D, Ozbabalik D, Gulcan E, et al. Evaluation of platelet activation,
57. Mavrommatis AC, Theodoridis T, Orfanidou A, et al. Coagulation system coagulation, and fibrinolytic activation in patients with symptomatic
and platelets are fully activated in uncomplicated sepsis. Crit Care Med lacunar stroke. Neurologist 2010;16(3):188–91.
2000;28(2):451–7. 86. D’Erasmo E, Aliberti G, Celi FS, et al. Platelet count, mean platelet volume
58. Visentin GP, Liu CY. Drug-induced thrombocytopenia. Hematol Oncol Clin and their relation to prognosis in cerebral infarction. J Intern Med
North Am 2007;21(4):685–96, vi. 1990;227(1):11–4.
59. Bonfiglio MF, Traeger SM, Kier KL, et al. Thrombocytopenia in intensive 87. O’Malley T, Langhorne P, Elton RA, et al. Platelet size in stroke patients.
care patients: a comprehensive analysis of risk factors in 314 patients. Ann Stroke 1995;26(6):995–9.
Pharmacother 1995;29(9):835–42. 88. Hayward CP, Harrison P, Cattaneo M, et al. Platelet function analyzer
60. Shalansky SJ, Verma AK, Levine M, et al. Risk markers for (PFA)-100 closure time in the evaluation of platelet disorders and platelet
thrombocytopenia in critically ill patients: a prospective analysis. function. J Thromb Haemost 2006;4(2):312–19.
Pharmacotherapy 2002;22(7):803–13. 89. Slaughter TF, Sreeram G, Sharma AD, et al. Reversible shear-mediated
61. Carey PJ. Drug-induced myelosuppression: diagnosis and management. platelet dysfunction during cardiac surgery as assessed by the PFA-100
Drug Saf 2003;26(10):691–706. platelet function analyzer. Blood Coagul Fibrinolysis 2001;12(2):85–93.
62. Curtis BR, Kaliszewski J, Marques MB, et al. Immune-mediated 90. Harrison P, Segal H, Blasbery K, et al. Screening for aspirin responsiveness
thrombocytopenia resulting from sensitivity to oxaliplatin. Am J Hematol after transient ischemic attack and stroke: comparison of 2 point-of-care
2006;81(3):193–8. platelet function tests with optical aggregometry. Stroke 2005;36(5):
63. Pedersen-Bjergaard U, Andersen M, Hansen PB. Drug-specific 1001–5.
characteristics of thrombocytopenia caused by non-cytotoxic drugs. Eur J 91. Raghavan M, Marik PE. Anemia, allogenic blood transfusion, and
Clin Pharmacol 1998;54(9-10):701–6. immunomodulation in the critically ill. Chest 2005;127(1):295–307.
64. Nguyen TC, Stegmayr B, Busund R, et al. Plasma therapies in thrombotic 92. Becker C. Bled dry: fiscal woes threaten supply data from National Blood
syndromes. Int J Artif Organs 2005;28(5):459–65. Data Resource Center. Mod Healthc 2003;33(2):12–3.
65. Pedersen-Bjergaard U, Andersen M, Hansen PB. Drug-induced 93. Sullivan MT, Wallace EL. Blood collection and transfusion in the United
thrombocytopenia: clinical data on 309 cases and the effect of States in 1999. Transfusion 2005;45(2):141–8.
corticosteroid therapy. Eur J Clin Pharmacol 1997;52(3):183–9. 94. Napolitano LM, Kurek S, Luchette FA, et al. Clinical practice guideline: red
66. Llevadot J, Coulter SA, Giugliano RP. A practical approach to the diagnosis blood cell transfusion in adult trauma and critical care. Crit Care Med
and management of thrombocytopenia associated with glycoprotein IIb/ 2009;37(12):3124–57.
IIIa receptor inhibitors. J Thromb Thrombolysis 2000;9(2):175–80. 95. Kramer AH, Zygun DA. Anemia and red blood cell transfusion in
67. Arepally GM, Ortel TL. Clinical practice. Heparin-induced neurocritical care. Crit Care 2009;13:1–22.
thrombocytopenia. N Engl J Med 2006;355(8):809–17. 96. Utter GH, Shahlaie K, Zwienenberg-Lee M, et al. Anemia in the setting of
68. Warkentin TE. Heparin-induced thrombocytopenia: pathogenesis and traumatic brain injury: the arguments for and against liberal transfusion.
management. Br J Haematol 2003;121(4):535–55. J Neurotrauma 2011;28:155–65.
69. Warkentin TE. Heparin-induced thrombocytopenia. Hematol Oncol Clin 97. Smith ML, Counelis GJ, Maloney-Wilensky E, et al. Brain tissue oxygen
North Am 2007;21(4):589–607, v. tension in clinical brain death: a case series. Neurol Res 2007;29:755–9.
70. Hoh BL, Aghi M, Pryor JC, et al. Neurosurgery 2005;57:243–48. 98. Dhar R, Zazulia AR, Videen TO, et al. Red blood cell transfusion increases
71. Warkentin TE, Kelton JG. Temporal aspects of heparin-induced cerebral oxygen delivery in anemic patients with subarachnoid hemorrhage.
thrombocytopenia. N Engl J Med 2001;344(17):1286–92. Stroke 2009;40:3039–44.
72. Lo GK, Juhl D, Warkentin TE, et al. Evaluation of pretest clinical score 99. Card RT, Mohandas N, Perkins HA, et al. Deformability of stored red blood
(4 T’s) for the diagnosis of heparin-induced thrombocytopenia in two cells. Relationship to degree of packing. Transfusion 1982;22(2):96–101.
clinical settings. J Thromb Haemost 2006;4:759–65. 100. Beutler E, Kuhl W, West C. The osmotic fragility of erythrocytes after
73. Zheng XL, Sadler JE. Pathogenesis of thrombotic microangiopathies. Ann prolonged liquid storage and after reinfusion. Blood 1982;59(6):1141–7.
Rev Pathol Mech Dis 2008;3:249–77. 101. Card RT, Mohandas N, Mollison PL. Relationship of post-transfusion
74. Schwartzman RJ, Hill JB. Neurologic complications of disseminated viability to deformability of stored red cells. Br J Haematol
intravascular coagulation. Neurology 1982;32:791–7. 1983;53(2):237–40.
75. Voves C, Waillemin WA, Zeerleder S. International Society on Thrombosis 102. Tinmouth A, Fergusson D, Yee IC, et al. Clinical consequences of red cell
and Haemostasis score for overt disseminated intravascular coagulation storage in the critically ill. Transfusion 2006;46(11):2014–27.
predicts organ dysfunction and fatality in sepsis patients. Blood Coagul 103. Napolitano LM, Corwin HL. Efficacy of red blood cell transfusion in the
Fibrinolysis 2006;17:445–51. critically ill. Crit Care Clin 2004;20(2):255–68.
76. Angstwurm MW, Dempfle CE, Spannagl M. New disseminated 104. Apstein CS, Dennis RC, Briggs L, et al. Effect of erythrocyte storage and
intravascular coagulation score: a useful tool to predict mortality in oxyhemoglobin affinity changes on cardiac function. Am J Physiol
comparison with Acute Physiology and Chronic Health Evaluation II 1985;248(4 Pt 2):H508–15.
and Logistic Organ Dysfunction scores. Crit Care Med 2006;34(2):314–20; 105. Hovav T, Yedgar S, Manny N, et al. Alteration of red cell aggregability and
quiz 28. shape during blood storage. Transfusion 1999;39(3):277–81.
77. Stohlawetz P, Stiegler G, Knobl P, et al. The rise of reticulated platelets after 106. Berezina TL, Zaets SB, Morgan C, et al. Influence of storage on red blood
intensive chemotherapy for AML reduces the need for platelet transfusion. cell rheological properties. J Surg Res 2002;102(1):6–12.
Ann Hematol 1999;78:271–3. 107. Knight JA, Voorhees RP, Martin L, et al. Lipid peroxidation in stored red
78. Chaoui D, Chakroun T, Robert F, et al. Reticulated platelets: a reliable cells. Transfusion 1992;32(4):354–7.
measure to reduce prophylactic platelet transfusions after intensive 108. Goodnough LT. Risks of blood transfusion. Crit Care Med 2003;31(12 Supp
chemotherapy. Transfusion 2005;45:766–72. l):S678–86.
79. Rinder HM, Schuster JE, Rinder CS, et al. Correlation of thrombosis with 109. Soderberg-Naucler C, Fish KN, et al. Reactivation of latent human
increased platelet turnover in thrombocytosis. Blood 1998;91:1288–94. cytomegalovirus by allogeneic stimulation of blood cells from healthy
80. Bessman JD, Williams LJ, Gilmer PR. Mean platelet volume: the inverse donors. Cell 1997;91(1):119–26.
relation of platelet size and count in normal subjects, and an artifact of 110. Bowden RA, Sayers M, Flournoy N, et al. Cytomegalovirus immune
other particles. Am J Clin Pathol 1981;76:289–93. globulin and seronegative blood products to prevent primary
81. Endler G, Klimesch A, Sunder-Plassmann H, et al. Mean platelet volume is cytomegalovirus infection after marrow transplantation. N Engl J Med
an independent risk factor for myocardial infarction but not for coronary 1986;314(16):1006–10.
artery disease. Br J Haematol 2002;117:399–404. 111. Chu RW. Leukocytes in blood transfusion: adverse effects and their
82. Arevalo-Lorido JC, Carretero-Gomez J, Alvarez-Oliva A, et al. Mean platelet prevention. Hong Kong Med J 1999;5(3):280–4.
volume in acute phase of ischemic stroke, as predictor of mortality and 112. Phelan HA, Gonzalez RP, Patel HD, et al. Prestorage leukoreduction
functional outcome after 1 year. J Stroke Cerebrovasc Dis 2011 Oct 15 ameliorates the effects of aging on banked blood. J Trauma
(Epub ahead of print). 2010;69(2):330–7.
Section II—Clinical and Laboratory Assessment 147.e3

113. Silliman CC, Moore EE, Kelher MR, et al. Identification of lipids that 140. Prins HA, Houdijk AP, Nijveldt RJ, et al. Arginase release from red blood
accumulate during the routine storage of prestorage leukoreduced red cells: possible link in transfusion induced immune suppression? Shock
blood cells and cause acute lung injury. Transfusion 2011;51(12):2549–54. 2001;16(2):113–15.
114. Silliman CC, Clay KL, Thurman GW, et al. Partial characterization of lipids 141. Smoller BR, Kruskall MS, Horowitz GL. Reducing adult phlebotomy blood
that develop during the routine storage of blood and prime the neutrophil loss with the use of pediatric-sized blood collection tubes. Am J Clin Pathol
NADPH oxidase. J Lab Clin Med 1994;124(5):684–94. 1989;91(6):701–3.
115. Shander A, Popovsky MA. Understanding the consequences of transfusion- 142. Khor B, Van Cott EM. Laboratory evaluation of hypercoagulability. Clin
related acute lung injury. Chest 2005;128(5 Suppl 2):598S–604S. Lab Med 2009;29(2):339–66.
116. Reese Jr EP, McCullough JJ, Craddock PR. An adverse pulmonary reaction 143. Perry DJ, Fitzmaurice DA, Kitchen S, et al. Point-of-care testing in
to cryoprecipitate in a hemophiliac. Transfusion 1975;15(6):583–8. haemostasis. Br J Haematol 2010;150(5):501–14.
117. Offner PJ, Moore EE, Biffl WL, et al. Increased rate of infection associated 144. Ak K, Isbir CS, Tetik S, et al. Thromboelastography-based transfusion
with transfusion of old blood after severe injury. Arch Surg algorithm reduces blood product use after elective CABG: a prospective
2002;137(6):711–16; discussion 6–7. randomized study. J Card Surg 2009;24(4):404–10.
118. Edna TH, Bjerkeset T. Association between blood transfusion and infection 145. Shore-Lesserson L, Manspeizer HE, DePerio M, et al.
in injured patients. J Trauma 1992;33(5):659–61. Thromboelastography-guided transfusion algorithm reduces transfusions in
119. Moore FA, Moore EE, Sauaia A. Blood transfusion: an independent risk complex cardiac surgery. Anesth Analg 1999;88(2):312–19.
factor for postinjury multiple organ failure. Arch Surg 1997;132(6):620–4; 146. Foulke GE, Harlow DJ. Effective measures for reducing blood loss from
discussion 4–5. diagnostic laboratory tests in intensive care unit patients. Crit Care Med
120. Agarwal N, Murphy JG, Cayten CG, et al. Blood transfusion increases the 1989;17(11):1143–5.
risk of infection after trauma. Arch Surg 1993;128(2):171–6; discussion 6–7. 147. Natanson C, Kern SJ, Lurie P, et al. Cell-free hemoglobin-based blood
121. Claridge JA, Sawyer RG, Schulman AM, et al. Blood transfusions correlate substitutes and risk of myocardial infarction and death: a meta-analysis.
with infections in trauma patients in a dose-dependent manner. Am Surg JAMA 2008;299(19):2304–12.
2002;68(7):566–72. 148. Moore EE, Moore FA, Fabian TC, et al. Human polymerized hemoglobin
122. Malone DL, Dunne J, Tracy JK, et al. Blood transfusion, independent of for the treatment of hemorrhagic shock when blood is unavailable: the
shock severity, is associated with worse outcome in trauma. J Trauma USA multicenter trial. J Am Coll Surg 2009;208(1):1–13.
2003;54(5):898–905; discussion 905-7. 149. Harrington JP, Wollocko H. Pre-clinical studies using OxyVita hemoglobin,
123. Dunne JR, Malone DL, Tracy JK, et al. Allogenic blood transfusion in the a zero-linked polymeric hemoglobin: a review. J Artif Organs
first 24 hours after trauma is associated with increased systemic 2010;13(4):183–8.
inflammatory response syndrome (SIRS) and death. Surg Infect (Larchmt) 150. Mammis A, McIntosh TK, Maniker AH. Erythropoietin as a
2004;5(4):395–404. neuroprotective agent in traumatic brain injury. Surg Neurol
124. Wobbes T, Bemelmans BL, Kuypers JH, et al. Risk of postoperative septic 2009;71(5):527–31; discussion 31.
complications after abdominal surgical treatment in relation to 151. Assaraf MI, Diaz Z, Liberman A, et al. Brain erythropoietin receptor
perioperative blood transfusion. Surg Gynecol Obstet 1990;171(1):59–62. expression in Alzheimer disease and mild cognitive impairment. J
125. Chelemer SB, Prato BS, Cox Jr PM, et al. Association of bacterial infection Neuropathol Exp Neurol 2007;66(5):389–98.
and red blood cell transfusion after coronary artery bypass surgery. Ann 152. Ehrenreich H, Kästner A, Weissenborn K, et al. Circulating damage marker
Thorac Surg 2002;73(1):138–42. profiles support a neuroprotective effect of erythropoietin in ischemic
126. Leal-Noval SR, Jara-Lopez I, Garcia-Garmendia JL, et al. Influence of stroke patients. Mol Med 2011;17(11–12):1306–10.
erythrocyte concentrate storage time on postsurgical morbidity in cardiac 153. Corwin HL, Gettinger A, Rodriguez RM, et al. Efficacy of recombinant
surgery patients. Anesthesiology 2003;98(4):815–22. human erythropoietin in the critically ill patient: a randomized, double-
127. Leal-Noval SR, Rincon-Ferrari MD, Garcia-Curiel A, et al. Transfusion of blind, placebo-controlled trial. Crit Care Med 1999;27(11):2346–50.
blood components and postoperative infection in patients undergoing 154. Corwin HL, Gettinger A, Pearl RG, et al. Efficacy of recombinant human
cardiac surgery. Chest 2001;119(5):1461–8. erythropoietin in critically ill patients: a randomized controlled trial. JAMA
128. Shorr AF, Jackson WL, Kelly KM, et al. Transfusion practice and blood 2002;288(22):2827–35.
stream infections in critically ill patients. Chest 2005;127(5):1722–8. 155. Gabriel A, Kozek S, Chiari A, et al. High-dose recombinant human
129. Taylor RW, Manganaro L, O’Brien J, et al. Impact of allogenic packed red erythropoietin stimulates reticulocyte production in patients with multiple
blood cell transfusion on nosocomial infection rates in the critically ill organ dysfunction syndrome. J Trauma 1998;44(2):361–7.
patient. Crit Care Med 2002;30(10):2249–54. 156. van Iperen CE, Gaillard CA, Kraaijenhagen RJ, et al. Response of
130. Marik PE, Corwin HL. Efficacy of red blood cell transfusion in the critically erythropoiesis and iron metabolism to recombinant human erythropoietin
ill: a systematic review of the literature. Crit Care Med 2008;36(9):2667–74. in intensive care unit patients. Crit Care Med 2000;28(8):2773–8.
131. Fransen E, Maessen J, Dentener M, et al. Impact of blood transfusions on 157. Corwin HL, Gettinger A, Fabian TC, et al. Efficacy and safety of epoetin
inflammatory mediator release in patients undergoing cardiac surgery. alfa in critically ill patients. N Engl J Med 2007;357(10):965–76.
Chest 1999;116(5):1233–9. 158. Epoetin alfa [package insert]. Thousand Oaks, CA. 2008; Amgen Inc.
132. Zallen G, Moore EE, Ciesla DJ, et al. Stored red blood cells selectively 159. Tseng MY, Hutchinson PJ, Richards HK, et al. Acute systemic erythropoietin
activate human neutrophils to release IL-8 and secretory PLA2. Shock therapy to reduce delayed ischemic deficits following aneurysmal
2000;13(1):29–33. subarachnoid hemorrhage: a phase II randomized, double-blind,
133. Stec N, Shirey RS, Smith B, et al. The efficacy of performing red cell elution placebo-controlled trial. Clinical article. J Neurosurg 2009;111(1):171–80.
studies in the pretransfusion testing of patients with positive direct 160. Ehrenreich H, Weissenborn K, Prange H, et al. Recombinant human
antiglobulin tests. Transfusion 1986;26(3):225–6. erythropoietin in the treatment of acute ischemic stroke. Stroke
134. Heddle NM, Klama LN, Griffith L, et al. A prospective study to identify the 2009;40(12):e647–56.
risk factors associated with acute reactions to platelet and red cell 161. Schafer AI. Approach to the patient with bleeding and thrombosis. In:
transfusions. Transfusion 1993;33(10):794–7. Goldman I, Schafer A, editors. Goldman’s Cecil Textbook of Medicine. 24th
135. Hebert PC, Fergusson D, Blajchman MA, et al. Clinical outcomes following ed. Philadelphia: Elsevier; 2012. p. 1121–4.
institution of the Canadian universal leukoreduction program for red blood 162. Lim W, Crowther MA, Ginsberg JS. Venous thromboembolism. In:
cell transfusions. JAMA 2003;289(15):1941–9. Hoffman R, Furie B, Benz EJ, et al, editors. Hematology. 5th ed.
136. Dzik WH, Anderson JK, O’Neill EM, et al. A prospective, randomized Philadelphia: Churchill Livingstone; 2009. p. 2043–54.
clinical trial of universal WBC reduction. Transfusion 2002;42(9): 163. Haines ST. Venous thromboembolism: pathophysiology and clinical
1114–22. presentation. Am J Health Syst Pharm 2003;60(22 Suppl 7):S3–5.
137. Watkins TR, Rubenfeld GD, Martin TR, et al. Effects of leukoreduced blood 164. Becker DM. Venous thromboembolism: epidemiology, diagnosis,
on acute lung injury after trauma: a randomized controlled trial. Crit Care prevention. J Gen Intern Med 1986;1(6):402–11.
Med 2008;36(5):1493–9. 165. Anderson Jr FA, Wheeler HB, Goldberg RJ, et al. A population-based
138. Nathens AB, Nester TA, Rubenfeld GD, et al. The effects of leukoreduced perspective of the hospital incidence and case-fatality rates of deep vein
blood transfusion on infection risk following injury: a randomized thrombosis and pulmonary embolism. The Worcester DVT Study. Arch
controlled trial. Shock 2006;26(4):342–7. Intern Med 1991;151(5):933–8.
139. Hogman CF, Meryman HT. Storage parameters affecting red blood cell 166. Cohen AT, Tapson VF, Bergmann JF, et al. Venous thromboembolism risk
survival and function after transfusion. Transfus Med Rev and prophylaxis in the acute hospital care setting (ENDORSE study): a
1999;13(4):275–96. multinational cross-sectional study. Lancet 2008;371(9610):387–94.
147.e4 Section II—Clinical and Laboratory Assessment

167. Schonhofer B, Kohler D. Prevalence of deep-vein thrombosis of the leg in 193. Sciacca FL, Ciusani E, Silvani A, et al. Genetic and plasma markers of
patients with acute exacerbation of chronic obstructive pulmonary disease. venous thromboembolism in patients with high grade glioma. Clin Cancer
Respiration 1998;65(3):173–7. Res 2004;10(4):1312–17.
168. Harris LM, Curl GR, Booth FV, et al. Screening for asymptomatic deep 194. Chan AT, Atiemo A, Diran LK, et al. Venous thromboembolism occurs
vein thrombosis in surgical intensive care patients. J Vasc Surg 1997;26(5): frequently in patients undergoing brain tumor surgery despite prophylaxis.
764–9. J Thromb Thrombolysis 1999;8(2):139–42.
169. Twigg SJ, McCrirrick A, Sanderson PM. A comparison of post mortem 195. Muchmore JH, Dunlap JN, Culicchia F, Kerstein MD. Deep vein
findings with post hoc estimated clinical diagnoses of patients who die in a thrombophlebitis and pulmonary embolism in patients with malignant
United Kingdom intensive care unit. Intensive Care Med 2001;27(4):706–10. gliomas. South Med J 1989;82(11):1352–6.
170. Bostrom S, Holmgren E, Jonsson O, et al. Post-operative thromboembolism 196. Danish SF, Burnett MG, Ong JG, et al. Prophylaxis for deep venous
in neurosurgery: a study on the prophylactic effect of calf muscle thrombosis in craniotomy patients: a decision analysis. Neurosurgery
stimulation plus dextran compared to low-dose heparin. Acta Neurochir 2005;56(6):1286–92; discussion 92–4.
(Wien) 1986;80(3-4):83–9. 197. Misra M, Roitberg B, Ebersole K, et al. Prevention of pulmonary embolism
171. Quevedo JF, Buckner JC, Schmidt JL, et al. Thromboembolism in patients by combined modalities of thromboprophylaxis and intensive surveillance
with high-grade glioma. Mayo Clin Proc 1994;69(4):329–32. protocol. Neurosurgery 2004;54(5):1099–102; discussion 102–3.
172. Abrahams JM, Torchia MB, McGarvey M, et al. Perioperative assessment of 198. Guyatt GH, Akl EA, Crowther M, et al. Executive summary: antithrombotic
coagulability in neurosurgical patients using thromboelastography. Surg therapy and prevention of thrombosis, 9th ed: American College of chest
Neurol 2002;58(1):5–11; discussion 11-2. physicians evidence based clinical practice guidelines. Chest
173. Juhan-Vague I, Conte-Devolx B, Aillaud MF, et al. Effects of DDAVP and 2012;141:7S–47S.
venous occlusion on the release of tissue-type plasminogen activator and 199. Geerts WH, Bergqvist D, Pineo GF, et al. Prevention of venous
von Willebrand factor in patients with panhypopituitarism. Thromb Res thromboembolism. American College of Chest Physicians
1984;33(6):653–9. Evidence-Based Clinical Practice Guidelines (8th ed). Chest 2008;
174. Hamer JD, Malone PC, Silver IA. The PO2 in venous valve pockets: its 133(6 Suppl):381S–453S.
possible bearing on thrombogenesis. Br J Surg 1981;68(3):166–70. 200. Kamphuisen PW, Agnelli G. What is the optimal pharmacological
175. Agnelli G, Piovella F, Buoncristiani P, et al. Enoxaparin plus compression prophylaxis for the prevention of deep-vein thrombosis and pulmonary
stockings compared with compression stockings alone in the prevention of embolism in patients with acute ischemic stroke? Thromb Res
venous thromboembolism after elective neurosurgery. N Engl J Med 2007;119(3):265–74.
1998;339(2):80–5. 201. Sherman DG, Albers GW, Bladin C, et al. The efficacy and safety of
176. Auguste KI, Quinones-Hinojosa A, Berger MS. Efficacy of mechanical enoxaparin versus unfractionated heparin for the prevention of venous
prophylaxis for venous thromboembolism in patients with brain tumors. thromboembolism after acute ischaemic stroke (PREVAIL study): an
Neurosurg Focus 2004;17(4):E3. open-label randomised comparison. Lancet 2007;369(9570):1347–55.
177. Raslan AM, Fields JD, Bhardwaj A. Prophylaxis for venous thrombo- 202. Goldstein JN, Fazen LE, Wendell L, et al. Risk of thromboembolism
embolism in neurocritical care: a critical appraisal. Neurocrit Care 2010; following acute intracerebral hemorrhage. Neurocrit Care 2009;10(1):
12(2):297–309. 28–34.
178. Cerrato D, Ariano C, Fiacchino F. Deep vein thrombosis and low-dose 203. Lacut K, Bressollette L, Le Gal G, et al. Prevention of venous thrombosis in
heparin prophylaxis in neurosurgical patients. J Neurosurg 1978;49(3): patients with acute intracerebral hemorrhage. Neurology 2005;65(6):
378–81. 865–9.
179. Powers SK, Edwards MS. Prophylaxis of thromboembolism in the 204. Boeer A, Voth E, Henze T, et al. Early heparin therapy in patients with
neurosurgical patient: a review. Neurosurgery 1982;10(4):509–13. spontaneous intracerebral haemorrhage. J Neurol Neurosurg Psychiatry
180. Smith SF, Simpson JM, Sekhon LH. Prophylaxis for deep venous 1991;54(5):466–7.
thrombosis in neurosurgical oncology: review of 2779 admissions over a 205. Tetri S, Hakala J, Juvela S, et al. Safety of low-dose subcutaneous
9-year period. Neurosurg Focus 2004;17(4):E4. enoxaparin for the prevention of venous thromboembolism after primary
181. Swann KW, Black PM. Deep vein thrombosis and pulmonary emboli in intracerebral haemorrhage. Thromb Res 2008;123(2):206–12.
neurosurgical patients: a review. J Neurosurg 1984;61(6):1055–62. 206. Broderick J, Connolly S, Feldmann E, et al. Guidelines for the management
182. Valladares JB, Hankinson J. Incidence of lower extremity deep vein of spontaneous intracerebral hemorrhage in adults, 2007 update: a
thrombosis in neurosurgical patients. Neurosurgery 1980;6(2):138–41. guideline from the American Heart Association/American Stroke
183. Hamilton MG, Hull RD, Pineo GF. Prophylaxis of venous thromboembolism Association Stroke Council, High Blood Pressure Research Council, and the
in brain tumor patients. J Neurooncol 1994;22(2):111–26. Quality of Care and Outcomes in Research Interdisciplinary Working
184. Altschuler E, Moosa H, Selker RG, et al. The risk and efficacy of Group. Stroke 2007;38(6):2001–23.
anticoagulant therapy in the treatment of thromboembolic complications 207. Morgenstern LB, Hemphill 3rd JC, Anderson C, et al. Guidelines for the
in patients with primary malignant brain tumors. Neurosurgery management of spontaneous intracerebral hemorrhage: a guideline for
1990;27(1):74–6; discussion 7. healthcare professionals from the American Heart Association/American
185. Hamilton MG, Hull RD, Pineo GF. Venous thromboembolism in Stroke Association. Stroke 2010;41(9):2108–29.
neurosurgery and neurology patients: a review. Neurosurgery 1994;34(2): 208. Palareti G, Cosmi B, Legnani C. Diagnosis of deep vein thrombosis. Semin
280–96; discussion 96. Thromb Hemost 2006;32(7):659–72.
186. Sawaya R, Zuccarello M, Elkalliny M, et al. Postoperative venous 209. Bates SM, Ginsberg JS. Clinical practice. Treatment of deep-vein
thromboembolism and brain tumor. Part I. Clinical profile. J Neurooncol thrombosis. N Engl J Med 2004;351(3):268–77.
1992;14(2):119–25. 210. Segal JB, Eng J, Tamariz LJ, et al. Review of the evidence on diagnosis of
187. Marras LC, Geerts WH, Perry JR. The risk of venous thromboembolism is deep venous thrombosis and pulmonary embolism. Ann Fam Med
increased throughout the course of malignant glioma: an evidence-based 2007;5(1):63–73.
review. Cancer 2000;89(3):640–6. 211. Heim SW, Schectman JM, Siadaty MS, et al. D-dimer testing for deep
188. Sawaya R, Hammoud M, Schoppa D, et al. Neurosurgical outcomes in a venous thrombosis: a metaanalysis. Clin Chem 2004;50(7):1136–47.
modern series of 400 craniotomies for treatment of parenchymal tumors. 212. Wells PS, Anderson DR, Rodger M, et al. Evaluation of D-dimer in the
Neurosurgery 1998;42(5):1044–55; discussion 55–6. diagnosis of suspected deep-vein thrombosis. N Engl J Med
189. Sawaya R, Decourteen-Meyers G, Copeland B. Massive preoperative 2003;349(13):1227–35.
pulmonary embolism and suprasellar brain tumor: case report and review 213. Crowther MA, Cook DJ, Griffith LE, et al. Neither baseline tests of
of the literature. Neurosurgery 1984;15(4):566–71. molecular hypercoagulability nor D-dimer levels predict deep venous
190. Sawaya R, Cummins CJ, Kornblith PL. Brain tumors and plasmin thrombosis in critically ill medical-surgical patients. Intensive Care Med
inhibitors. Neurosurgery 1984;15(6):795–800. 2005;31(1):48–55.
191. Turpie AG, Hirsh J, Gent M, et al. Prevention of deep vein thrombosis in 214. Engelberger RP, Aujesky D, Calanca L, et al. Comparison of the diagnostic
potential neurosurgical patients: a randomized trial comparing graduated performance of the original and modified Wells score in inpatients and
compression stockings alone or graduated compression stockings plus outpatients with suspected deep vein thrombosis. Thromb Res
intermittent pneumatic compression with control. Arch Intern Med 2011;127(6):535–9.
1989;149(3):679–81. 215. Soto MJ, Grau E, Gadelha T, et al. Clinical significance of a negative
192. Hamada K, Kuratsu J, Saitoh Y, et al. Expression of tissue factor correlates D-dimer level in patients with confirmed venous thromboembolism:
with grade of malignancy in human glioma. Cancer 1996;77(9):1877–83. findings from the RIETE Registry. J Thromb Haemost 2011;9(2):407–10.
Section II—Clinical and Laboratory Assessment 147.e5

216. Flinn WR, Sandager GP, Silva MB, Jr., et al. Prospective surveillance for 243. Harenberg J. Pharmacology of low molecular weight heparins. Semin
perioperative venous thrombosis: experience in 2643 patients. Arch Surg Thromb Hemost 1990;(16 Suppl):12–8.
1996;131(5):472–80. 244. Hirsh J, Bauer KA, Donati MB, et al. Parenteral anticoagulants: American
217. Sud S, Mittmann N, Cook DJ, et al. Screening and prevention of venous College of Chest Physicians Evidence-Based Clinical Practice Guidelines
thromboembolism in critically ill patients: a decision analysis and (8th ed.). Chest 2008;133(6 Suppl):141S–59S.
economic evaluation. Am J Respir Crit Care Med 2011;184(11):1289–98. 245. Tollefsen DM, Majerus DW, Blank MK. Heparin cofactor II. Purification
218. Stam J. Thrombosis of the cerebral veins and sinuses. N Engl J Med and properties of a heparin-dependent inhibitor of thrombin in human
2005;352(17):1791–8. plasma. J Biol Chem 1982;257(5):2162–9.
219. Saposnik G, Barinagarrementeria F, Brown RD Jr., et al. Diagnosis and 246. Young E, Prins M, Levine MN, et al. Heparin binding to plasma proteins,
management of cerebral venous thrombosis: a statement for healthcare an important mechanism for heparin resistance. Thromb Haemost
professionals from the American Heart Association/American Stroke 1992;67(6):639–43.
Association. Stroke 2011;42(4):1158–92. 247. Whitfield LR, Lele AS, Levy G. Effect of pregnancy on the relationship
220. Wilder-Smith E, Kothbauer-Margreiter I, Lammle B, et al. Dural puncture between concentration and anticoagulant action of heparin. Clin
and activated protein C resistance: risk factors for cerebral venous sinus Pharmacol Ther 1983;34(1):23–8.
thrombosis. J Neurol Neurosurg Psychiatry 1997;63(3):351–6. 248. Olson JD, Arkin CF, Brandt JT, et al. College of American Pathologists
221. Wilterdink JL, Easton JD. Cerebral ischemia in pregnancy. Adv Neurol conference XXXI on laboratory monitoring of anticoagulant therapy:
2002;90:51–62. laboratory monitoring of unfractionated heparin therapy. Arch Pathol Lab
222. Lanska DJ, Kryscio RJ. Peripartum stroke and intracranial venous Med 1998;122(9):782–98.
thrombosis in the National Hospital Discharge survey. Obstet Gynecol 249. Marks PW. Treatment of venous thromboembolism. In: Colman RW,
1997;89(3):413–18. Marder VJ, Clowes AW, et al, editors. Thrombosis and Hemostasis: Basic
223. Cucchiara B, Messe S, Taylor R, et al. Utility of D-dimer in the diagnosis of Principles and Clinical Practice. Philadelphia: Lippincott Williams &
cerebral venous sinus thrombosis. J Thromb Haemost 2005;3(2):387–9. Wilkins; 2006. p. 1307–16.
224. Kosinski CM, Mull M, Schwarz M, et al. Do normal D-dimer levels reliably 250. Hirsh J, van Aken WG, Gallus AS, et al. Heparin kinetics in venous
exclude cerebral sinus thrombosis? Stroke 2004;35(12):2820–5. thrombosis and pulmonary embolism. Circulation 1976;53(4):691–5.
225. Stam J, De Bruijn SF, DeVeber G. Anticoagulation for cerebral sinus 251. Levine MN, Hirsh J, Gent M, et al. A randomized trial comparing activated
thrombosis. Cochrane Database Syst Rev 2002;(4):CD002005. thromboplastin time with heparin assay in patients with acute venous
226. Furie KL, Kasner SE, Adams RJ, et al. Guidelines for the prevention of thromboembolism requiring large daily doses of heparin. Arch Intern Med
stroke in patients with stroke or transient ischemic attack: a guideline for 1994;154(1):49–56.
healthcare professionals from the American Heart Association/American 252. Edson JR, Krivit W, White JG. Kaolin partial thromboplastin time: high
Stroke Association. Stroke 2011;42(1):227–76. levels of procoagulants producing short clotting times or masking
227. Cohn DM, Roshani S, Middeldorp S. Thrombophilia and venous deficiencies of other procoagulants or low concentrations of anticoagulants.
thromboembolism: implications for testing. Semin Thromb Hemost J Lab Clin Med 1967;70(3):463–70.
2007;33(6):573–81. 253. Antman EM. Hirudin in acute myocardial infarction: safety report from the
228. Fields MC, Levine SR. Thrombophilias and stroke: diagnosis, treatment, Thrombolysis and Thrombin Inhibition in Myocardial Infarction (TIMI)
and prognosis. J Thromb Thrombolysis 2005;20(2):113–26. 9A Trial. Circulation 1994;90(4):1624–30.
229. Van Cott EM, Soderberg BL, Laposata M. Activated protein C resistance, 254. GUSTO Investigators. Randomized trial of intravenous heparin versus
the factor V Leiden mutation, and a laboratory testing algorithm. Arch recombinant hirudin for acute coronary syndromes: the Global Use of
Pathol Lab Med 2002;126(5):577–82. Strategies to Open Occluded Coronary Arteries (GUSTO) IIa investigators.
230. van den Belt AG, Sanson BJ, Simioni P, et al. Recurrence of venous Circulation 1994;90:1631–7.
thromboembolism in patients with familial thrombophilia. Arch Intern 255. EPIC Investigators. Use of a monoclonal antibody directed against the
Med 1997;157(19):2227–32. platelet glycoprotein IIb/IIIa receptor in high-risk coronary angioplasty: the
231. Griffin JH, Heeb MJ, Kojima Y, et al. Activated protein C resistance: EPIC investigation. N Engl J Med 1994;330:956–61.
molecular mechanisms. Thromb Haemost 1995;74(1):444–8. 256. Basu D, Gallus A, Hirsh J, Cade J. A prospective study of the value of
232. Rahemtullah A, Van Cott EM. Hypercoagulation testing in ischemic stroke. monitoring heparin treatment with the activated partial thromboplastin
Arch Pathol Lab Med 2007;131(6):890–901. time. N Engl J Med 1972;287(7):324–7.
233. Poort SR, Rosendaal FR, Reitsma PH, Bertina RM. A common genetic 257. Rosborough TK. Comparing different lots of activated partial
variation in the 3’-untranslated region of the prothrombin gene is thromboplastin time reagent: analysis of two methods. Am J Clin Pathol
associated with elevated plasma prothrombin levels and an increase in 1998;110(2):173–7.
venous thrombosis. Blood 1996;88(10):3698–703. 258. Baker BA, Adelman MD, Smith PA, et al. Inability of the activated partial
234. Folsom AR, Cushman M, Tsai MY, et al. A prospective study of venous thromboplastin time to predict heparin levels: time to reassess guidelines
thromboembolism in relation to factor V Leiden and related factors. Blood for heparin assays. Arch Intern Med 1997;157(21):2475–9.
2002;99(8):2720–5. 259. Bates SM, Grand’Maison A, Johnston M, et al. A latex D-dimer reliably
235. Koster T, Blann AD, Briet E, et al. Role of clotting factor VIII in effect of excludes venous thromboembolism. Arch Intern Med 2001;161(3):447–53.
von Willebrand factor on occurrence of deep-vein thrombosis. Lancet 260. Bates SM, Weitz JI, Johnston M, et al. Use of a fixed activated partial
1995;345(8943):152–5. thromboplastin time ratio to establish a therapeutic range for
236. van Hylckama Vlieg A, van der Linden IK, Bertina RM, et al. High levels of unfractionated heparin. Arch Intern Med 2001;161(3):385–91.
factor IX increase the risk of venous thrombosis. Blood 261. Brandt JT, Triplett DA. Laboratory monitoring of heparin. Effect of
2000;95(12):3678–82. reagents and instruments on the activated partial thromboplastin time. Am
237. Meijers JC, Tekelenburg WL, Bouma BN, et al. High levels of coagulation J Clin Pathol 1981;76(4 Suppl):530–7.
factor XI as a risk factor for venous thrombosis. N Engl J Med 262. van den Besselaar AM, Meeuwisse-Braun J, Bertina RM. Monitoring
2000;342(10):696–701. heparin therapy: relationships between the activated partial thromboplastin
238. Brey RL, Muscal E, Chapman J. Antiphospholipid antibodies and the brain: time and heparin assays based on ex-vivo heparin samples. Thromb
a consensus report. Lupus 2011;20:153–7. Haemost 1990;63(1):16–23.
239. Urbanus RT, Siegerink B, Roest M, et al. Antiphospholipid antibodies and 263. Holmer E, Kurachi K, Soderstrom G. The molecular-weight dependence of
risk of myocardial infarction and ischaemic stroke in young women in the the rate-enhancing effect of heparin on the inhibition of thrombin, factor
RATIO study: a case-control study. Lancet Neurol 2009;8:998–1005. Xa, factor IXa, factor XIa, factor XIIa and kallikrein by antithrombin.
240. Ruiz-Irastorza G, Cuadrado MJ, Ruiz-Arruza I, et al. Evidence-based Biochem J 1981;193(2):395–400.
recommendations for the prevention and long-term management of 264. Holmer E, Soderberg K, Bergqvist D, et al. Heparin and its low molecular
thrombosis in antiphospholipid antibody-positive patients: report of a task weight derivatives: anticoagulant and antithrombotic properties.
force at the 13th International Congress on Antiphospholipid Antibodies. Haemostasis 1986;16(Suppl 2):1–7.
Lupus 2011;20:206–18. 265. Hirsh J, Weitz JI. New antithrombotic agents. Lancet 1999;353(9162):
241. Andersson LO, Barrowcliffe TW, Holmer E, et al. Anticoagulant properties 1431–6.
of heparin fractionated by affinity chromatography on matrix-bound 266. Choay J, Petitou M, Lormeau JC, et al. Structure-activity relationship in
antithrombin III and by gel filtration. Thromb Res 1976;9(6):575–83. heparin: a synthetic pentasaccharide with high affinity for antithrombin III
242. Johnson EA, Mulloy B. The molecular-weight range of mucosal-heparin and eliciting high anti-factor Xa activity. Biochem Biophys Res Commun
preparations. Carbohydr Res 1976;51(1):119–27. 1983;116(2):492–9.
147.e6 Section II—Clinical and Laboratory Assessment

267. Choay J. Biologic studies on chemically synthesized pentasaccharide and 283. Eiswirth G, Walch S, Bommer J. New bedside test for monitoring
tetrasaccharide fragments. Semin Thromb Hemost 1985;11(2):81–5. anticoagulation during hemodialysis. Artif Organs 1998;22(4):346–8.
268. Bijsterveld NR, Moons AH, Boekholdt SM, et al. Ability of recombinant 284. Reiner JS, Coyne KS, Lundergan CF, et al. Bedside monitoring of heparin
factor VIIa to reverse the anticoagulant effect of the pentasaccharide therapy: comparison of activated clotting time to activated partial
fondaparinux in healthy volunteers. Circulation 2002;106(20):2550–4. thromboplastin time. Cathet Cardiovasc Diagn 1994;32(1):49–52.
269. Kuo KH, Kovacs MJ. Fondaparinux: a potential new therapy for HIT. 285. Smythe MA, Koerber JM, Westley SJ, et al. Use of the activated partial
Hematology 2005;10(4):271–5. thromboplastin time for heparin monitoring. Am J Clin Pathol
270. Wallis RB. Hirudins: from leeches to man. Semin Thromb Hemost 2001;115(1):148–55.
1996;22(2):185–96. 286. Ferring M, Reber G, de Moerloose P, et al. Point of care and central
271. Lefevre G, Duval M, Gauron S, et al. Effect of renal impairment on the laboratory determinations of the aPTT are not interchangeable in surgical
pharmacokinetics and pharmacodynamics of desirudin. Clin Pharmacol intensive care patients. Can J Anaesth 2001;48(11):1155–60.
Ther 1997;62(1):50–9. 287. Reiss RA, Haas CE, Griffis DL, et al. Point-of-care versus laboratory
272. Salemi A, Agrawal YP, Fontes MA. An assay to monitor bivalirudin levels on monitoring of patients receiving different anticoagulant therapies.
cardiopulmonary bypass. Ann Thorac Surg 2011;92(1):332–4. Pharmacotherapy 2002;22(6):677–85.
273. Connolly SJ, Eikelboom J, Joyner C, et al. Apixaban in patients with atrial 288. Choi TS, Greilich PE, Shi C, et al. Point-of-care testing for prothrombin
fibrillation. N Engl J Med 2011;364:806–17. time, but not activated partial thromboplastin time, correlates with
274. Connolly SJ, Ezekowitz MD, Yusuf S, et al. Dabigatran versus warfarin in laboratory methods in patients receiving aprotinin or epsilon-aminocaproic
patients with atrial fibrillation. N Engl J Med 2009;361(12):1139–51. acid while undergoing cardiac surgery. Am J Clin Pathol 2002;117(1):74–8.
275. Ansell J, Hirsh J, Hylek E, et al. Pharmacology and management of the 289. Jaryno S, Bennett K, Loder C, et al. Validation of a new whole blood
vitamin K antagonists. American College of Chest Physicians Evidence- coagulation monitoring system. J Extra Corpor Technol 2002;34(4):
Based Clinical Practice Guidelines (8th ed.). Chest 2008;133:160S–98S. 271–5.
276. Shehab N, Sperling LS, Kegler SR, et al. National estimates of emergency 290. Wang JS, Lin CY, Karp RB. Comparison of high-dose thrombin time with
department visits for hemorrhage-related adverse events from clopidogrel activated clotting time for monitoring of anticoagulant effects of heparin in
plus aspirin and from warfarin. Arch Intern Med 2010;170:1926–33. cardiac surgical patients. Anesth Analg 1994;79(1):9–13.
277. Lassen MR, Gallus A, Raskob GE, et al. Apixaban versus enoxaparin for 291. Despotis GJ, Levine V, Joist JH, et al. Antithrombin III during cardiac
thromboprophylaxis after hip replacement. N Engl J Med 2010;363: surgery: effect on response of activated clotting time to heparin and
2487–98. relationship to markers of hemostatic activation. Anesth Analg
278. Lassen MR, Raskob GE, Gallus A, et al. Apixaban versus enoxaparin for 1997;85(3):498–506.
thromboprophylaxis after knee replacement (ADVANCE-2): a randomised 292. Royston D, von Kier S. Reduced haemostatic factor transfusion using
double-blind trial. Lancet 2010;375:807–15. heparinase-modified thrombelastography during cardiopulmonary bypass.
279. Chavez JJ, Weatherall JS, Strevels SM, et al. Evaluation of a point-of-care Br J Anaesth 2001;86(4):575–8.
coagulation analyzer on patients undergoing cardiopulmonary bypass 293. Gonzalez E, Pieracci FM, Moore EE, et al. Coagulation abnormalities in the
surgery. J Clin Anesth 2004;16(1):7–10. trauma patient: the role of point of care thromboelastography. Semin
280. Merlani PG, Chenaud C, Cottini S, et al. Point of care management of Thromb Hemost 2010;36:723–37.
heparin administration after heart surgery: a randomized, controlled trial. 294. Baker J, Quick H, Hullsiek KH, et al. Interleukin-6 and d-dimer levels are
Intensive Care Med 2006;32(9):1357–64. associated with vascular dysfunction in patients with untreated HIV
281. Zabel KM, Granger CB, Becker RC, et al. Use of bedside activated partial infection. HIV Med 2010;11(9):608–9.
thromboplastin time monitor to adjust heparin dosing after thrombolysis 295. Dempfle CE, Suvajac N, Elmas E, et al. Performance evaluation of a new
for acute myocardial infarction: results of GUSTO-I. Global Utilization of rapid quantitative assay system for measurement of D-dimer in plasma and
Streptokinase and TPA for Occluded Coronary Arteries. Am Heart J whole blood: PATHFAST D-dimer. Thromb Res 2007;120(4):591–6.
1998;136(5):868–76. 296. Dempfle CE, Korte W, Schwab M, et al. Sensitivity and specificity of a
282. Schroeder AP, Knudsen LL, Husted SE, et al. Bedside coagulometry during quantitative point of care D-dimer assay using heparinized whole blood, in
intravenous heparin therapy after coronary angioplasty. J Thromb patients with clinically suspected deep vein thrombosis. Thromb Haemost
Thrombolysis 2001;12(2):157–63. 2006;96(1):79–83.
II
Chapter
16  

Monitoring Inflammation
Alejandro M. Spiotta, Alan Siu, and J. Javier Provencio

neurotrauma, SAH, and various chronic CNS diseases. Inflam-


Introduction matory mediators are indeed elevated in the cerebrospinal
The basic tenet of neurocritical care is prevention of secondary fluid (CSF) and serum of patients with acute and chronic
injury. Secondary injuries following the initial ictus include brain injury, and are associated with poor outcome.19-25 Various
global increases in intracranial pressure (ICP), alterations in animal models also point to potential protective mechanisms
cerebral perfusion, hydrocephalus, and seizures, among others. with the inhibition of proinflammatory mediators. As the role
In addition, systemic insults such as infections, hypotension, of neuroinflammation in secondary brain injury becomes
thromboemboli, hyperglycemia, hyperthermia, malnutrition, more evident, the need to monitor its activity in the NCCU
and other complications can affect patients in the neurocritical becomes important.
care unit (NCCU) adversely. Accumulating evidence suggests The knowledge that may be gained by monitoring neuro-
that inflammation can exacerbate secondary injury in brain inflammation and its clinical utility can be organized into
disease and in particular subarachnoid hemorrhage, vaso- three main areas:
spasm, traumatic brain injury (TBI), and stroke; it can even
1. Identifying risk factors to predict secondary injury
be epileptogenic.1-6 Monitoring neuroinflammation therefore
2. Providing endpoints to monitor responses to specific ther-
may allow better understanding of the impact of therapies
apies aimed at modulating the inflammatory state
aimed at preventing or minimizing secondary injury. Whether
3. Gaining insights into the pathophysiology of the disease
prevention of inflammation will improve patient outcome fol-
process
lowing brain injury is still to be defined because although
pharmacologic therapies to attenuate inflammation can ame- Monitoring of neuroinflammation thus may provide a more
liorate pathology, microglial activation that is central to the sensitive and earlier detection of secondary injury and in turn
inflammatory response also plays a role in recovery and regen- widen the therapeutic window. Furthermore, studying the
eration (i.e., inflammation may have a dual role). response of the various measured parameters to conventional
and experimental medical and interventional therapies should
augment our knowledge of the disease course and potentially
Monitoring Neuroinflammation lead to new therapeutic strategies.
As the understanding of the pathophysiologic process follow-
ing acute brain injury including subarachnoid hemorrhage
(SAH), stroke, and TBI has increased, awareness about the
Basic Science of Inflammation
deleterious effects of inflammatory activation as both a trigger The brain was once considered to be an “immune-privileged”
and exacerbating factor of secondary injury has created new organ system, inaccessible and independent from systemic
opportunities in monitoring neuroinflammation. For example, inflammation. Although the CNS has unique features, this
findings from clinical studies suggest that neutrophils play a view has largely fallen out of favor because there is accumulat-
critical role in ischemia-reperfusion injury, because tissue ing evidence of crosstalk between the CNS and the immune
damage is associated with extent of neutrophil infiltration.7-9 system outside of the CNS. For example, CNS cells have been
The critical role that systemic as well as local inflammatory shown to modulate peripheral immune function in response
activators play in determining the extent of central nervous to peripheral disease and injury through the systemic release
system (CNS) injury following an ictus is highlighted by of various cytokines (i.e., interleukin-1 [IL-1], IL-6, tumor
the emergence and widespread use of anti-inflammatory necrosis factor-α [TNFα]) and vagal efferents.26,27
pharmaceuticals, such as statins, in neurocritical care.10-14 Cerebral edema has many potential causes but can be con-
3-Hydroxy-3-methylglutaryl coenzyme A (HMG CoA) reduc- sidered an inflammatory process. Indeed IL-1 neutralizing
tase inhibitors, commonly known as statins, have lipid- antibodies in animal models can attenuate brain edema.28
lowering properties, and animal models of acute brain injury Because the skull is functionally a closed box, significant
have demonstrated their anti-inflammatory properties.15-18 development of edema occupies the excess space in the
There is mounting evidence for the role of neuroinflammation cranium, after which pressure increases and so can damage
as a contributor to secondary brain tissue injury in stroke, neurons and glia. Cerebral edema occurs at different times in
148 © Copyright 2013 Elsevier Inc. All rights reserved.
Section II—Clinical and Laboratory Assessment 149

different cerebral injuries. In TBI and SAH, cerebral edema chronic neurodegeneration such as multiple sclerosis and
develops quickly and can cause increased ICP in many patients. Alzheimer’s disease have also implicated a role for inflamma-
Conversely, in ischemic stroke, cerebral edema develops slowly tion,44,45 because cytokines such as TNFα and interferon-γ
over the first 72 hours after the initial ictus. The reason for the (IFNγ) are elevated in the CSF. In addition, in vitro studies
differences in these time courses is not well defined; however, point to inflammation’s damaging effects on neurons, whereas
the course of cerebral edema could be predicted by monitor- cytokine inhibition in animal models slows the disease
ing inflammation. process.46 Interestingly, immune responses also are necessary
Inflammatory processes contribute to CNS injury and neu- for neuroregeneration, suggesting there is an innate balance
rodegeneration.20,27 In addition there is accumulating evidence between adaptive and deleterious immune activation or
that inflammation and in particular leukocyte-endothelial cell immunomodulation.47
interactions, play a critical role in the pathogenesis of vaso- The entry of leukocytes into the CNS parenchyma depends
spasm after SAH2,29 and that inflammatory processes contrib- on the expression of specific molecules by the endothelium
ute to formation of cerebral aneurysms.30-32 Various types of (intracellular adhesion molecules [ICAMs], integrins, selec-
acute and chronic brain injuries result in gliosis, a reaction tins, and chemokines) along the capillary and postcapillary
characterized by the activation, proliferation, and hypertrophy venules in the brain. The production of chemokines and the
of mononuclear phagocytic-type cells,33-35 which occurs in a activation of the endothelium are initiated by the activated
well-defined manner to promote a systematic restoration of parenchymal microglia, which produce the cytokines and
the brain parenchyma in the setting of neuronal injury. This other inflammatory mediators to further stimulate the inflam-
process is mediated by the resident astrocytes and microglia, matory cascade.36
which produce various chemical mediators to recruit periph- The activated endothelium is marked by an up-regulation
erally derived immune cells to further enhance the inflamma- of cell-adhesion molecules (i.e., selectins and integrins) that
tory response. The inhibition of these chemical mediators act to increase interactions to anchor peripheral leukocytes
(i.e., TNFα, IL-1) and the key components that modulate to promote transmigration. In this multistep paradigm of
leukocyte trafficking across the blood-brain barrier (BBB) leukocyte-endothelial interactions to cross the BBB, the first
have shown great potential to improve outcome in neuroin- step is tethering, which is characterized by a transient contact
flammatory diseases such as multiple sclerosis and hold the between the glycosylated ligands on leukocytes and selectins
same potential in acute brain injury (natalizumab therapy). on the endothelium. The next step is rolling, which is medi-
ated by the shear forces of flowing blood to promote locomo-
tion of the leukocyte and increase exposure to activating
Biology of the Brain’s factors (i.e., cytokines, chemokines) immobilized on the
endothelial luminal surface. Further activation of the leuko-
Immune System cytes is characterized by conformational changes of integrins
Inflammatory processes within the CNS are different from the into a higher affinity state for stronger interactions with endo-
periphery because the CNS parenchyma lacks resident den- thelial cell-adhesion molecules (ICAM-1, vascular cell adhe-
dritic cells, with inflammation instead being initiated by the sion molecule 1 [VCAM-1]), resulting in leukocyte arrest onto
resident microglia.36 The exact role of microglia remains the endothelial surface. The arrested leukocytes then can
under debate because they release a number of factors that extend protrusions enriched in chemokine receptors, and
modulate both secondary injury and recovery after injury, extravasate across the BBB by either a paracellular or transcel-
including pro- and anti-inflammatory cytokines, chemokines, lular route. Various matrix metalloproteinases (MMPs) are
nitric oxide, prostaglandins, growth factors, and superoxide secreted to facilitate the transmigration process.
species.37 The most unique quality, however, is the presence of
a BBB that regulates the subsequent immune amplification by
the peripheral inflammatory cells. The BBB, along with the The Febrile State: Monitoring Brain
neuroepithelial cells, modulate the inflammatory mediators
and ultimately the peripheral leukocytes that participate in a
and Body Temperature
neuroinflammatory state.36,38-41 These key mediators, namely Fever is the oldest and most easily measured marker of an
the cytokines and adhesion molecules, are the focus of current underlying inflammatory condition, with careful records of
drug development.42 elevated temperature and its relationship to disease course
CNS injury can result in acute or chronic neurodegenera- dating back to antiquity.48 Raised core body temperature is
tion. In the acute setting, conditions such as TBI and cerebral associated with worse outcome following a variety of acute
ischemia lead to parenchymal injury that is characterized by neurologic insults.49-54 The exact mechanism by which hyper-
substantial nerve cell loss from excitotoxicity (i.e., glutamate thermia exacerbates CNS injury is not yet fully elucidated.
release), oxidative stress, and regional electrolyte disturbances Animal infarct models demonstrate that hyperthermia can
among other pathophysiologic processes. Proinflammatory enhance the depth and extent of injury by one of two mecha-
cytokines are elevated in human CSF and serum after TBI. In nisms: increased cytokine release and inflammation or
addition, increased microglial activation can be detected in increased energy expenditure and a widened metabolic gap.55-
59
vivo using the positron emission tomography (PET) ligand However, hyperthermia in a brain-injured patient has many
[11C](R)PK11195 even years after an acute injury.43 Animal disparate causes. First, pharmaceutical and infectious etiolo-
models of acute injury have further implicated these proin- gies are common.60-62 Second, there may be direct hypotha-
flammatory cytokines as contributors to the extent of brain lamic injury. Third, factors such as heat shock protein (HSP)
injury because their inhibition can reduce the extent of cell synthesis, release of proinflammatory cytokines such as IL-1,
death. Extensive investigations into various conditions of IL-6, TNFα, and interferon, or increased glutamate63-65 each,
150 Section II—Clinical and Laboratory Assessment

or in combination, may act as the stimulus to trigger fever rupture. This likely represents the response to ischemia. There
after TBI (for review see reference 62). Factors that may aggra- was a trend observed for higher CRP values on the preceding
vate neurologic outcome in the setting of hyperthermia days but these did not achieve significance. Serum CRP also
include increased glucocorticoid production, increased meta- was greater in poor-grade patients, which makes it difficult to
bolic expenditure, altered oxygen consumption, glutamate or conclude a “causal” relationship between a heightened acute
nitric oxide release, increased myeloid cell and cytokine activ- phase response and vasospasm, or that CRP simply reflects the
ity, or increased vascular permeability.58,66-70 Given the complex severity of the initial brain injury.
interplay between factors for fever following SAH, it is not In ischemic stroke, CRP and leukocytosis also are indepen-
surprising that fever has been found to be an independent risk dent factors associated with outcome including cognitive
factor for the development of vasospasm following SAH in outcome among survivors.88-90 In addition, several lines of
some studies,53,54 whereas others have not found this relation- evidence demonstrate a relationship between progression of
ship.71 Fever at admission also is a risk factor for vasospasm infarction and worse neurologic outcome and elevated serum
after TBI72 and is associated with poor clinical grade after cytokines IL-1, IL-6, and TNFα.89,91-95 What is unclear is
aneurysm rupture.73 whether there is a causal relationship.
It is now possible to routinely monitor brain temperature
(BT; see Chapter 37). Although brain and body temperature
may correlate after SAH and TBI, average BT is usually greater Cerebrospinal Fluid
than the average rectal temperature by about 1° C. The differ-
ence between brain and body temperature becomes greater as
Inflammatory Markers
a patient becomes febrile.74-77 The effect of BT on brain physi- Cytokines and downstream activators of the inflammatory
ology is not well understood. Some studies have demonstrated response can be measured in the CSF. This is better than brain
that fever may be associated with increased ICP,75 while others biopsy but there are limitations to CSF sampling including:
suggest that brain hyperthermia may not affect brain oxygen78 (1) sampling error when relative concentrations of proteins
or may even increase it.79 In part these different results may and molecules must diffuse into the CSF from a heteroge-
be associated with changes in cerebral blood volume or cere- neously injured brain (i.e., the measured values likely reflect
bral blood flow. On the other hand, microdialysis studies a weighted average from surrounding tissue and its proximity
suggest that fever control can attenuate markers of cerebral to the subarachnoid space); (2) CSF levels of individual cyto-
energy dysfunction in the brain.80 However, exactly what the kines may be related to their diffusion properties in fluids and
relationship between BT and energy dysfunction is, is not distance from the source; (3) it is restricted to intermittent
clear. The clinical utility of BT monitoring, and in particular (e.g., daily) sampling; and (4) different half-lives of these
the relationship of BT with cerebral metabolism requires inflammatory markers both in the CSF and collecting vials. In
more study. addition, it should be remembered that most cytokines act
over short distances to effect local cells. It may therefore be
desirable in some patients to obtain tissue and fluid closest to
Serum Inflammatory Markers the injury, to allow for more specific sampling of brain
Systemic indices of inflammation revolve around markers of inflammation.
acute phase reactants such as serum white blood cell (WBC) Several clinical studies suggest that CSF and blood are
count and C reactive protein (CRP). Elevation in WBCs can functionally separate compartments when cytokine concen-
occur in the setting of infection, inflammation, and stress or trations are measured. For example, Shiozaki et  al.96 com-
tissue necrosis. The limitation of these parameters is their pared CSF and serum concentrations of several cytokines
nonspecific nature in CNS injury. This can limit an interpreta- from patients suffering from multiorgan trauma and TBI to
tion about CNS inflammation because elevation of these those with isolated TBI. The concentrations of the proinflam-
parameters may reflect other organ injury or secondary infec- matory cytokines TNFα and IL-1 were greater in CSF than
tion. Despite these limitations, several converging lines of serum in those with isolated brain injury. In patients with
evidence suggest that monitoring acute phase reactants may multiorgan trauma, serum levels were greater than in CSF,
help develop models to predict secondary injury following whereas CSF concentrations of both proinflammatory and
brain injury.81 anti-inflammatory cytokines were similar to those with iso-
SAH is a disease in which inflammation monitoring may be lated CNS injury. There was an association between Injury
the most advantageous and in particular to evaluate vaso- Severity Score, an index of systemic injury burden, and
spasm.2,82-84 For example, leukocytosis has been found to be an tumor necrosis factor-α receptor 1 (TNFr1). Patients with
independent risk factor for delayed cerebral ischemia (DCI) elevated ICP had higher concentrations of CSF IL-1, IL-10,
following aneurysmal SAH, with each increase of 1000 in the IL-Ira, and TNFr1 than those with normal ICP. These results
serum WBC associated with a 9% increase in the likelihood suggest that CSF cytokine monitoring will more accurately
of developing DCI.73,85 Furthermore, a peak serum WBC reflect CNS pathology than serum cytokines.
greater than 15,000 is associated with a 3.3-fold greater inci- Hydrocephalus that requires external ventricular drainage
dence of DCI,73 whereas a CSF neutrophil content of greater is common following SAH; hence CSF samples often are avail-
than 62% on the third day after SAH is an independent factor able. SAH has been found to cause an increase in cytokines
associated with later vasospasm.86 IL-1, IL-6, transforming growth factor-β (TGFβ) and TNFα
The relationship between CRP, vasospasm and the develop- released from leukocytes in CSF21,97-99 and so monitoring the
ment of DCI is still to be fully elucidated.85,87 For example, inflammatory response and in particular IL-6 maybe a useful
Rothoerl et al.87 observed that CRP values were higher in tool to help guide care of SAH patients.84 However, in
patients who developed DCI 5 to 8 days after aneurysm SAH patients who develop systemic inflammatory response
Section II—Clinical and Laboratory Assessment 151

syndrome (SIRS) ventricular CSF cytokine levels do not the findings. It appears that IL-1b and vascular endothelial
appear to correlate with serum levels of IL-1 and TNFα.97 In growth factor (VEGF) levels are greater in brain ECF than in
addition, although SIRS is associated with poor outcome, it CSF, whereas IL-6, TNF, IL-8 and IL-1ra are greater in CSF
may not always be associated with vasospasm.100,101 In general than in microdialysate.3 Third, there are complex interactions
SIRS is more common in patients in poor clinical grade, and of the immune system with the BBB and blood-CSF barriers
some but not all studies suggest it is associated with how an that influence the findings. Finally, it is unclear whether it is
aneurysm is occluded.100,101 the peak cytokine level, the area under the curve, or the spe-
IL-1 and IL-6 levels consistently have been observed to be cific cytokine production relative to other mediators that is
greater in poor-grade than good-grade patients although an most important to the biologic effect.
initial inflammatory response is observed in patients of all In clinical practice there are four basic approaches to
clinical grades.102 There also appears to be a relationship with examine the role of inflammatory mediators: (1) blood sam-
DCI and inflammatory markers particularly in the CSF.102,103 pling (both arterial and jugular venous), (2) CSF sampling,
For example, Mathiesen et al.21 analyzed CSF daily following (3) microdialysis, and (4) direct tissue sampling from ex vivo
SAH and found IL-1 to increase during delayed ischemic neu- or postmortem tissue. In addition, PET studies may be used
rologic deficit (DIND). Elevated IL-1 and TNFα levels also to examine microglial activation. Blood sampling is the easiest
correlated with poor outcome.21 Kwon and Jeon found that technique because it is readily available, can be easily repeated,
admission IL-6 concentration correlated with delayed isch- requires no specialized equipment, and cytokines can be
emic deficits.98 SAH also appears to induce the production of screened for with multiplex assay techniques. However, cyto-
nitric oxide metabolites, as inferred by the association with kines in arterial blood samples may indicate both intracranial
increased levels of nitric oxide breakdown products, nitrite and extracranial pathology. Paired arterial and jugular venous
and nitrate (NOx), in CSF.104 However, nitric oxide metabo- sampling can be used to help examine the relative contribu-
lites in CSF appear to be different from serum concentra- tions of the brain or the systemic inflammatory response.
tions.104,105 In patients who develop vasospasm and DCI, CSF However, detection of a gradient is not always feasible because
concentrations of NOx are greater between 2 and 8 days after the absolute concentrations of some cytokines are close to the
aneurysm rupture.106 limit of sensitivity of the assay techniques.
In TBI, CSF cytokine concentrations are elevated.65,107-109 External ventricular drainage when used provides a useful
The highest concentrations usually are measured during the method to evaluate CSF inflammation because CSF produc-
first 24 hours, after which there is a decline.110 However, there tion of cytokines is thought to represent brain production.
is a difference between CSF and serum concentrations of However, the concentration of a cytokine may depend on how
various cytokines.96,111,112 The difference between plasma and the CSF is drained (i.e., intermittently or continuously)117 and
CSF levels of cytokines does not appear to be associated with CSF production. In addition, it is unclear whether CSF drain-
BBB breakdown. This suggests that the cytokines are released age is a method to eliminate various cytokines (i.e., is “thera-
by microglia, astrocytes, and neurons112 rather than simply peutic”). This then raises the question of the biologic relevance
reflecting “spill-over” from the blood. There are conflicting of any immune mediator detected in the CSF.
data on what cytokine levels in the CSF mean. Some studies Microdialysis continuously samples the brain interstitial
show that IL-1 concentration in CSF is associated with unfa- space (see Chapter 36). This may be the most biologically
vorable outcome96,108 following TBI. Other studies, however, relevant compartment because inflammatory mediators are
suggest that peak CSF IL-6 levels are associated with improved secreted here and act on their surface membrane target recep-
outcome,113 IL-6 deficiency is associated with poor outcome,114 tors. It can be difficult to assay cytokines and inflammatory
and up-regulation of IL-10 production may play a neuro­ mediators using microdialysis because these proteins have a
protective role by attenuating TNFα release.111,115 greater molecular weight than the metabolic intermediaries
(glucose, lactate, pyruvate) commonly examined with this
technique. Consequently cytokines diffuse across the micro-
Methods to Sample Cytokines dialysis membrane at a slower rate, and a smaller proportion
and Monitor Neuroinflammation crosses into the catheter (i.e., the relative recovery is low).
Several techniques can be used to address this, including use
in Clinical Practice of catheters with larger molecular weight cutoff membranes
Immune system cells and brain cells produce cytokines that (e.g., 100 kDa), addition of colloids (such as dextrans or
act as intercellular signaling molecules. Depending on their albumin) to the perfusate, and use of sensitive multiplex tech-
concentration, cytokines can have toxic or trophic effects and niques to assay the cytokines (e.g., the Luminex xMAP plat-
can be pro- or anti-inflammatory.116 In addition several cyto- form). In the laboratory setting cytokine binders (e.g., heparin)
kines can have synergistic effects, and therefore a full under- have been added to the perfusion fluid. This has not been used
standing of the interactions between both deleterious and clinically because it is conceivable that heparin is lost from the
protective cytokines rather than just the absolute concentra- perfusate into the ECF.118
tion of a particular cytokine may be needed to derive mean-
ingful interpretation of clinical data. Several other factors
make clinical interpretation of human cytokine data complex. Future Investigation
First, each cytokine is produced during a defined time period
and so results may depend on when the samples are obtained. Further Understanding of the Dual Effects
Second, cytokines are produced in different amounts within of Cytokines Following Acute Brain Injury
different compartments—blood, brain, extracellular fluid Acute brain injury is followed by a cytokine-triggered influx
(ECF), and CSF—and hence what is sampled will influence of lymphocytes and myeloid cells into the injured region.
152 Section II—Clinical and Laboratory Assessment

These infiltrating cells are further stimulated in an autocrine Consistent with this, animal studies suggest that the inhibition
and paracrine fashion to produce an inflammatory environ- of proinflammatory mediators can provide neuroprotection.
ment, which may contribute to further tissue injury. However, Monitoring of inflammatory markers in the CNS that may be
some signals are adaptive and confer neuroprotection as well measured in the serum, CSF, or brain interstitial fluid using
as promote tissue repair.119 For example, IL-6 is both a neuro- microdialysis therefore will likely play an increasing role in the
tropic and inflammatory factor.120 The complex interplay of NCCU.
the various factors and downstream effectors involved in an
inflammatory response in the setting of acute brain injury is
only beginning to be elucidated.
References
The major challenge when monitoring cytokines as markers 1. Mashaly HA, Provencio JJ. Inflammation as a link between brain injury and
heart damage: the model of subarachnoid hemorrhage. Cleve Clin J Med
of inflammation is that many cytokines can exert different 2008;75(Suppl 2):S26–30.
effects that depend on the local environment. This also pres- 2. Pradilla G, Chaichana KL, Hoang S, et al. Inflammation and cerebral
ents an obstacle when interpreting animal and clinical studies vasospasm after subarachnoid hemorrhage. Neurosurg Clin N Am 2010;
that relate inflammatory mediators and extent of brain injury 21(2):365–79.
or outcome. It is likely a balance between the various impli- 3. Helmy A, De Simoni MG, Guilfoyle MR, et al. Cytokines and innate
inflammation in the pathogenesis of human traumatic brain injury. Prog
cated cytokines and their intracellular signaling cascades that Neurobiol 2011;95(3):352–72. Epub 2011 Sep 16.
determines whether the inflammatory response is adaptive or 4. Brough D, Tyrrell PJ, Allan SM. Regulation of interleukin-1 in acute brain
maladaptive. Thus, monitoring cytokines in isolation may not injury. Trends Pharmacol Sci 2011;32(10):617–22. Epub 2011 Jul 23.
be useful unless its role is incorporated into the greater context 5. Ravizza T, Balosso S, Vezzani A. Inflammation and prevention of
epileptogenesis. Neurosci Lett 2011;497(3):223–30. Epub 2011 Feb 26.
of the inflammatory environment and individualized to each 6. Denes A, Thornton P, Rothwell NJ, et al. Inflammation and brain injury:
particular patient. acute cerebral ischaemia, peripheral and central inflammation. Brain Behav
Immun 2010;24:708–23.
Further Investigation on the Interplay 7. Ott L, McClain CJ, Gillespie M, et al. Cytokines and metabolic dysfunction
after severe head injury. J Neurotrauma 1994;11:447–72.
Between Systemic and CNS 8. Ross SA, Halliday MI, Campbell GC, et al. The presence of tumour necrosis
Inflammatory Pathways factor in CSF and plasma after severe head injury. Br J Neurosurg 1994;8:
419–25.
The relationship between cerebral inflammatory response and 9. Shohami E, Novikov M, Bass R, et al. Closed head injury triggers early
the systemic, extracerebral response needs to be better under- production of TNF alpha and IL-6 by brain tissue. J Cereb Blood Flow
Metab 1994;14:615–19.
stood. They are distinct but not independent pathways, and it 10. Chou SH, Smith EE, Badjatia N, et al. A randomized, double-blind,
is likely that systemic factors influence those occurring in the placebo-controlled pilot study of simvastatin in aneurysmal subarachnoid
CNS and that the neuroinflammatory pathway modulates the hemorrhage. Stroke 2008;39:2891–3.
systemic cascade. 11. Lynch JR, Wang H, McGirt MJ, et al. Simvastatin reduces vasospasm after
aneurysmal subarachnoid hemorrhage: results of a pilot randomized clinical
trial. Stroke 2005;36:2024–6.
12. McGirt MJ, Blessing R, Alexander MJ, et al. Risk of cerebral vasospasm after
Continuous, Bedside Monitoring of subarachnoid hemorrhage reduced by statin therapy: a multivariate analysis
Neuroinflammation of an institutional experience. J Neurosurg 2006;105:671–4.
13. Sillberg VA, Wells GA, Perry JJ. Do statins improve outcomes and reduce the
incidence of vasospasm after aneurysmal subarachnoid hemorrhage: a
It is feasible to measure intrathecal interleukins at bedside meta-analysis. Stroke 2008;39:2622–6.
using point-of-care testing with an immunoassay chip-test. 14. Tseng MY, Czosnyka M, Richards H, et al. Effects of acute treatment with
These bedside results are similar to those obtained with labo- pravastatin on cerebral vasospasm, autoregulation, and delayed ischemic
ratory testing.81 Microdialysis techniques to sample inflamma- deficits after aneurysmal subarachnoid hemorrhage: a phase II randomized
tory mediators in the tissue most at risk for secondary injury, placebo-controlled trial. Stroke 2005;36:1627–32.
15. Chen SF, Hung TH, Chen CC, et al. Lovastatin improves histological and
may further advance the ability to monitor neuroinflamma- functional outcomes and reduces inflammation after experimental traumatic
tion.110 For example, the balance between members of the IL-1 brain injury. Life Sci 2007;81:288–98.
cytokine family, in particular between IL-1b and its endoge- 16. Lu D, Goussev A, Chen J, et al. Atorvastatin reduces neurological deficit and
nous inhibitor IL-1ra, may be more important than absolute increases synaptogenesis, angiogenesis, and neuronal survival in rats
subjected to traumatic brain injury. J Neurotrauma 2004;21:21–32.
levels of IL-1b. In TBI patients, the microdialysate IL-1ra:IL-1b 17. Lu D, Qu C, Goussev A, et al. Statins increase neurogenesis in the dentate
ratio is greater in patients with favorable rather than unfavor- gyrus, reduce delayed neuronal death in the hippocampal CA3 region, and
able outcomes.121 The critical factor is whether following improve spatial learning in rats after traumatic brain injury. J Neurotrauma
trends in neuroinflammation heralds the onset of secondary 2007;24:1132–46.
injury in a timely and predictable manner to alter manage- 18. Turkoglu OF, Eroglu H, Okutan O, et al. Atorvastatin efficiency after
traumatic brain injury in rats. Surg Neurol 2009;72:146–52.
ment, or whether it is a late sign at a time when intervention 19. Feuerstein G, Wang X, Barone FC. Cytokines in brain ischemia—the role of
is not likely to affect the outcome. TNF alpha. Cell Mol Neurobiol 1998;18:695–701.
20. Lucas SM, Rothwell NJ, Gibson RM. The role of inflammation in CNS injury
and disease. Br J Pharmacol 2006;147(Suppl 1):S232–40.
Conclusion 21. Mathiesen T, Edner G, Ulfarsson E, et al. Cerebrospinal fluid interleukin-1
receptor antagonist and tumor necrosis factor-alpha following subarachnoid
Inflammatory processes, initiated by the resident microglia hemorrhage. J Neurosurg 1997;87:215–20.
within the CNS, occur after a variety of acute neurological 22. Rodriguez-Yanez M, Castillo J. Role of inflammatory markers in brain
insults. These processes may have a dual role, that is, inflam- ischemia. Curr Opin Neurol 2008;21:353–7.
matory responses can play a role in recovery whereas accumu- 23. Stefini R, Catenacci E, Piva S, et al. Chemokine detection in the cerebral
tissue of patients with posttraumatic brain contusions. J Neurosurg 2008;
lating evidence indicates that inflammation can aggravate 108:958–62.
secondary injury in conditions such as TBI, stroke, or SAH.
Section II—Clinical and Laboratory Assessment 153

24. Waje-Andreassen U, Krakenes J, Ulvestad E, et al. An early marker for 39. Man S, Ubogu EE, Ransohoff RM. Inflammatory cell migration into the
outcome in acute ischemic stroke. Acta Neurol Scand 2005;111:360–5. central nervous system: a few new twists on an old tale. Brain Pathol 2007;
25. Welsh P, Lowe GD, Chalmers J, et al. Associations of proinflammatory 17:243–50.
cytokines with the risk of recurrent stroke. Stroke 2008;39:2226–30. 40. Pachter JS, de Vries HE, Fabry Z. The blood-brain barrier and its role in
26. Borovikova LV, Ivanova S, Zhang M, et al. Vagus nerve stimulation attenuates immune privilege in the central nervous system. J Neuropathol Exp Neurol
the systemic inflammatory response to endotoxin. Nature 2000;405:458–62. 2003;62:593–604.
27. Rothwell NJ, Luheshi GN. Interleukin 1 in the brain: biology, pathology and 41. Ransohoff RM, Kivisakk P, Kidd G. Three or more routes for leukocyte
therapeutic target. Trends Neurosci 2000;23:618–25. migration into the central nervous system. Nat Rev Immunol 2003;3:569–81.
28. Clausen F, Hanell A, Israelsson C, et al. Neutralization of interleukin-1beta 42. Simard JM, Geng Z, Woo SK, et al. Glibenclamide reduces inflammation,
reduces cerebral edema and tissue loss and improves late cognitive outcome vasogenic edema, and caspase-3 activation after subarachnoid hemorrhage.
following traumatic brain injury in mice. Eur J Neurosci 2011;34:110–23. J Cereb Blood Flow Metab 2009;29:317–30.
29. Chaichana KL, Pradilla G, Huang J, et al. Role of inflammation (leukocyte- 43. Ramlackhansingh AF, Brooks DJ, Greenwood RJ, et al. Inflammation after
endothelial cell interactions) in vasospasm after subarachnoid hemorrhage. trauma: microglial activation and traumatic brain injury. Ann Neurol
World Neurosurg 2010;73(1):22–41. Epub 2009 Aug 6. 2011;70(3):374–83. Epub 2011 Jun 27.
30. Kataoka H, Aoki T. Molecular basis for the development of intracranial 44. De Santi L, Polimeni G, Cuzzocrea S, et al. Neuroinflammation and
aneurysm. Expert Rev Neurother 2010;10(2):173–17. neuroprotection: an update on (future) neurotrophin-related strategies in
31. Tulamo R, Frösen J, Hernesniemi J, et al. Inflammatory changes in the multiple sclerosis treatment. Curr Med Chem 2011;18(12):1775–84.
aneurysm wall: a review. J Neurointerv Surg 2010;2(2):120–30. Epub 2010 45. Amor S, Puentes F, Baker D, van der Valk P. Inflammation in
Mar 12. neurodegenerative diseases. Immunology 2010;129(2):154–69.
32. Aoki T, Nishimura M. Targeting chronic inflammation in cerebral 46. Laskowitz DT, Vitek MP. Apolipoprotein E and neurological disease:
aneurysms: focusing on NF-kappaB as a putative target of medical therapy. therapeutic potential and pharmacogenomic interactions.
Expert Opin Ther Targets 2010;14(3):265–73. Pharmacogenomics 2007;8:959–69.
33. Norton WT, Aquino DA, Hozumi I, et al. Quantitative aspects of reactive 47. Iannotti CA, Clark M, Horn KP, et al. A combination immunomodulatory
gliosis: a review. Neurochem Res 1992;17:877–85. treatment promotes neuroprotection and locomotor recovery after contusion
34. O’Callaghan JP. Quantitative features of reactive gliosis following toxicant- SCI. Exp Neurol 2011;230(1):3–15. Epub 2010 Mar 23.
induced damage of the CNS. Ann N Y Acad Sci 1993;679:195–210. 48. Thompson HJ. Fever: a concept analysis. J Adv Nurs 2005;51:484–92.
35. Perry VH, Gordon S. Macrophages and the nervous system. Int Rev Cytol 49. Busto R, Dietrich WD, Globus MY, et al. Small differences in intraischemic
1991;125:203–44. brain temperature critically determine the extent of ischemic neuronal
36. Town T, Nikolic V, Tan J. The microglial “activation” continuum: from innate injury. J Cereb Blood Flow Metab 1987;7:729–38.
to adaptive responses. J Neuroinflammation 2005;2:24. 50. Diringer MN, Reaven NL, Funk SE, et al. Elevated body temperature
37. Loane DJ, Byrnes KR. Role of microglia in neurotrauma. Neurotherapeutics independently contributes to increased length of stay in neurologic intensive
2010;7(4):366–77. care unit patients. Crit Care Med 2004;32:1489–95.
38. Engelhardt B, Ransohoff RM. The ins and outs of T-lymphocyte trafficking
to the CNS: anatomical sites and molecular mechanisms. Trends Immunol A complete list of references for this chapter can be found online at
2005;26:485–95. www.expertconsult.com.
Section II—Clinical and Laboratory Assessment 153.e1

References 28. Clausen F, Hanell A, Israelsson C, et al. Neutralization of interleukin-1beta


reduces cerebral edema and tissue loss and improves late cognitive outcome
1. Mashaly HA, Provencio JJ. Inflammation as a link between brain injury and following traumatic brain injury in mice. Eur J Neurosci 2011;34:110–23.
heart damage: the model of subarachnoid hemorrhage. Cleve Clin J Med 29. Chaichana KL, Pradilla G, Huang J, et al. Role of inflammation (leukocyte-
2008;75(Suppl 2):S26–30. endothelial cell interactions) in vasospasm after subarachnoid hemorrhage.
2. Pradilla G, Chaichana KL, Hoang S, et al. Inflammation and cerebral World Neurosurg 2010;73(1):22–41. Epub 2009 Aug 6.
vasospasm after subarachnoid hemorrhage. Neurosurg Clin N Am 2010; 30. Kataoka H, Aoki T. Molecular basis for the development of intracranial
21(2):365–79. aneurysm. Expert Rev Neurother 2010;10(2):173–17.
3. Helmy A, De Simoni MG, Guilfoyle MR, et al. Cytokines and innate 31. Tulamo R, Frösen J, Hernesniemi J, et al. Inflammatory changes in the
inflammation in the pathogenesis of human traumatic brain injury. Prog aneurysm wall: a review. J Neurointerv Surg 2010;2(2):120–30. Epub 2010
Neurobiol 2011;95(3):352–72. Epub 2011 Sep 16. Mar 12.
4. Brough D, Tyrrell PJ, Allan SM. Regulation of interleukin-1 in acute 32. Aoki T, Nishimura M. Targeting chronic inflammation in cerebral
brain injury. Trends Pharmacol Sci 2011;32(10):617–22. Epub 2011 aneurysms: focusing on NF-kappaB as a putative target of medical therapy.
Jul 23. Expert Opin Ther Targets 2010;14(3):265–73.
5. Ravizza T, Balosso S, Vezzani A. Inflammation and prevention of 33. Norton WT, Aquino DA, Hozumi I, et al. Quantitative aspects of reactive
epileptogenesis. Neurosci Lett 2011;497(3):223–30. Epub 2011 Feb 26. gliosis: a review. Neurochem Res 1992;17:877–85.
6. Denes A, Thornton P, Rothwell NJ, et al. Inflammation and brain injury: 34. O’Callaghan JP. Quantitative features of reactive gliosis following
acute cerebral ischaemia, peripheral and central inflammation. Brain Behav toxicant-induced damage of the CNS. Ann N Y Acad Sci 1993;679:195–210.
Immun 2010;24:708–23. 35. Perry VH, Gordon S. Macrophages and the nervous system. Int Rev Cytol
7. Ott L, McClain CJ, Gillespie M, et al. Cytokines and metabolic dysfunction 1991;125:203–44.
after severe head injury. J Neurotrauma 1994;11:447–72. 36. Town T, Nikolic V, Tan J. The microglial “activation” continuum: from
8. Ross SA, Halliday MI, Campbell GC, et al. The presence of tumour necrosis innate to adaptive responses. J Neuroinflammation 2005;2:24.
factor in CSF and plasma after severe head injury. Br J Neurosurg 1994;8: 37. Loane DJ, Byrnes KR. Role of microglia in neurotrauma. Neurotherapeutics
419–25. 2010;7(4):366–77.
9. Shohami E, Novikov M, Bass R, et al. Closed head injury triggers early 38. Engelhardt B, Ransohoff RM. The ins and outs of T-lymphocyte trafficking
production of TNF alpha and IL-6 by brain tissue. J Cereb Blood Flow to the CNS: anatomical sites and molecular mechanisms. Trends Immunol
Metab 1994;14:615–19. 2005;26:485–95.
10. Chou SH, Smith EE, Badjatia N, et al. A randomized, double-blind, 39. Man S, Ubogu EE, Ransohoff RM. Inflammatory cell migration into the
placebo-controlled pilot study of simvastatin in aneurysmal subarachnoid central nervous system: a few new twists on an old tale. Brain Pathol
hemorrhage. Stroke 2008;39:2891–3. 2007;17:243–50.
11. Lynch JR, Wang H, McGirt MJ, et al. Simvastatin reduces vasospasm after 40. Pachter JS, de Vries HE, Fabry Z. The blood-brain barrier and its role in
aneurysmal subarachnoid hemorrhage: results of a pilot randomized immune privilege in the central nervous system. J Neuropathol Exp Neurol
clinical trial. Stroke 2005;36:2024–6. 2003;62:593–604.
12. McGirt MJ, Blessing R, Alexander MJ, et al. Risk of cerebral vasospasm after 41. Ransohoff RM, Kivisakk P, Kidd G. Three or more routes for leukocyte
subarachnoid hemorrhage reduced by statin therapy: a multivariate analysis migration into the central nervous system. Nat Rev Immunol 2003;3:
of an institutional experience. J Neurosurg 2006;105:671–4. 569–81.
13. Sillberg VA, Wells GA, Perry JJ. Do statins improve outcomes and reduce 42. Simard JM, Geng Z, Woo SK, et al. Glibenclamide reduces inflammation,
the incidence of vasospasm after aneurysmal subarachnoid hemorrhage: a vasogenic edema, and caspase-3 activation after subarachnoid hemorrhage.
meta-analysis. Stroke 2008;39:2622–6. J Cereb Blood Flow Metab 2009;29:317–30.
14. Tseng MY, Czosnyka M, Richards H, et al. Effects of acute treatment with 43. Ramlackhansingh AF, Brooks DJ, Greenwood RJ, et al. Inflammation after
pravastatin on cerebral vasospasm, autoregulation, and delayed ischemic trauma: microglial activation and traumatic brain injury. Ann Neurol
deficits after aneurysmal subarachnoid hemorrhage: a phase II randomized 2011;70(3):374–83. Epub 2011 Jun 27.
placebo-controlled trial. Stroke 2005;36:1627–32. 44. De Santi L, Polimeni G, Cuzzocrea S, et al. Neuroinflammation and
15. Chen SF, Hung TH, Chen CC, et al. Lovastatin improves histological and neuroprotection: an update on (future) neurotrophin-related strategies in
functional outcomes and reduces inflammation after experimental multiple sclerosis treatment. Curr Med Chem 2011;18(12):1775–84.
traumatic brain injury. Life Sci 2007;81:288–98. 45. Amor S, Puentes F, Baker D, van der Valk P. Inflammation in
16. Lu D, Goussev A, Chen J, et al. Atorvastatin reduces neurological deficit neurodegenerative diseases. Immunology 2010;129(2):154–69.
and increases synaptogenesis, angiogenesis, and neuronal survival in rats 46. Laskowitz DT, Vitek MP. Apolipoprotein e and neurological disease:
subjected to traumatic brain injury. J Neurotrauma 2004;21:21–32. therapeutic potential and pharmacogenomic interactions.
17. Lu D, Qu C, Goussev A, et al. Statins increase neurogenesis in the dentate Pharmacogenomics 2007;8:959–69.
gyrus, reduce delayed neuronal death in the hippocampal CA3 region, and 47. Iannotti CA, Clark M, Horn KP, et al. A combination immunomodulatory
improve spatial learning in rats after traumatic brain injury. J Neurotrauma treatment promotes neuroprotection and locomotor recovery after
2007;24:1132–46. contusion SCI. Exp Neurol 2011;230(1):3–15. Epub 2010 Mar 23.
18. Turkoglu OF, Eroglu H, Okutan O, et al. Atorvastatin efficiency after 48. Thompson HJ. Fever: a concept analysis. J Adv Nurs 2005;51:484–92.
traumatic brain injury in rats. Surg Neurol 2009;72:146–52. 49. Busto R, Dietrich WD, Globus MY, et al. Small differences in intraischemic
19. Feuerstein G, Wang X, Barone FC. Cytokines in brain ischemia—the role of brain temperature critically determine the extent of ischemic neuronal
TNF alpha. Cell Mol Neurobiol 1998;18:695–701. injury. J Cereb Blood Flow Metab 1987;7:729–38.
20. Lucas SM, Rothwell NJ, Gibson RM. The role of inflammation in CNS 50. Diringer MN, Reaven NL, Funk SE, et al. Elevated body temperature
injury and disease. Br J Pharmacol 2006;147 Suppl 1:S232–40. independently contributes to increased length of stay in neurologic
21. Mathiesen T, Edner G, Ulfarsson E, et al. Cerebrospinal fluid interleukin-1 intensive care unit patients. Crit Care Med 2004;32:1489–95.
receptor antagonist and tumor necrosis factor-alpha following 51. Hajat C, Hajat S, Sharma P. Effects of poststroke pyrexia on stroke
subarachnoid hemorrhage. J Neurosurg 1997;87:215–20. outcome: a meta-analysis of studies in patients. Stroke 2000;31:410–14.
22. Rodriguez-Yanez M, Castillo J. Role of inflammatory markers in brain 52. Naidech AM, Bendok BR, Bernstein RA, et al. Fever burden and functional
ischemia. Curr Opin Neurol 2008;21:353–7. recovery after subarachnoid hemorrhage. Neurosurgery 2008;63:212–17;
23. Stefini R, Catenacci E, Piva S, et al. Chemokine detection in the cerebral discussion 217–18.
tissue of patients with posttraumatic brain contusions. J Neurosurg 53. Oliveira-Filho J, Ezzeddine MA, Segal AZ, et al. Fever in subarachnoid
2008;108:958–62. hemorrhage: relationship to vasospasm and outcome. Neurology 2001;56:
24. Waje-Andreassen U, Krakenes J, Ulvestad E, et al. An early marker for 1299–304.
outcome in acute ischemic stroke. Acta Neurol Scand 2005;111:360–5. 54. Weir B, Disney L, Grace M, et al. Daily trends in white blood cell count and
25. Welsh P, Lowe GD, Chalmers J, et al. Associations of proinflammatory temperature after subarachnoid hemorrhage from aneurysm. Neurosurgery
cytokines with the risk of recurrent stroke. Stroke 2008;39:2226–30. 1989;25:161–5.
26. Borovikova LV, Ivanova S, Zhang M, et al. Vagus nerve stimulation 55. Chen H, Chopp M, Welch KM. Effect of mild hyperthermia on the
attenuates the systemic inflammatory response to endotoxin. Nature 2000; ischemic infarct volume after middle cerebral artery occlusion in the rat.
405:458–62. Neurology 1991;41:1133–5.
27. Rothwell NJ, Luheshi GN. Interleukin 1 in the brain: biology, pathology 56. Dietrich WD. The importance of brain temperature in cerebral injury.
and therapeutic target. Trends Neurosci 2000;23:618–25. J Neurotrauma 1992;9 Suppl 2:S475–85.
153.e2 Section II—Clinical and Laboratory Assessment

57. Morimoto T, Ginsberg MD, Dietrich WD, et al. Hyperthermia enhances 84. Muroi C, Mink S, Seule M, et al. Monitoring of the inflammatory response
spectrin breakdown in transient focal cerebral ischemia. Brain Res 1997; after aneurysmal subarachnoid haemorrhage in the clinical setting: review
746:43–51. of literature and report of preliminary clinical experience. Acta Neurochir
58. Whalen MJ, Carlos TM, Clark RS, et al. The relationship between brain Suppl 2011;110(Pt 1):191–6.
temperature and neutrophil accumulation after traumatic brain injury in 85. Kasius KM, Frijns CJ, Algra A, et al. Association of platelet and
rats. Acta Neurochir Suppl 1997;70:260–1. leukocyte counts with delayed cerebral ischemia in aneurysmal
59. Matthews DS, Bullock RE, Matthews JN, et al. Temperature response to subarachnoid hemorrhage. Cerebrovasc Dis 2010;29(6):576–83. Epub
severe head injury and the effect on body energy expenditure and cerebral 2010 Apr 8.
oxygen consumption. Arch Dis Child 1995;72:507–15. 86. Provencio JJ, Fu X, Siu A, et al. CSF neutrophils are implicated in the
60. Stocchetti N, Rossi S, Zanier ER, et al. Pyrexia in head-injured patients development of vasospasm in subarachnoid hemorrhage. Neurocrit Care
admitted to intensive care. Intensive Care Med 2002;28:1555–62. 2010;12(2):244–51.
61. Stubbe HD, Greiner C, Van Aken H, et al. Cerebral vascular and metabolic 87. Rothoerl RD, Axmann C, Pina AL, et al. Possible role of the C-reactive
response to sustained systemic inflammation in ovine traumatic brain protein and white blood cell count in the pathogenesis of cerebral
injury. J Cereb Blood Flow Metab 2004;24:1400–8. vasospasm following aneurysmal subarachnoid hemorrhage. J Neurosurg
62. Thompson HJ, Tkacs NC, Saatman KE, et al. Hyperthermia following Anesthesiol 2006;18:68–72.
traumatic brain injury: a critical evaluation. Neurobiol Dis 2003;12:163–73. 88. Muir KW, Weir CJ, Alwan W, et al. C-reactive protein and outcome after
63. Allan SM, Rothwell NJ. Cytokines and acute neurodegeneration. Nat Rev ischemic stroke. Stroke 1999;30:981–5.
Neurosci 2001;2:734–44. 89. Smith CJ, Emsley HC, Gavin CM, et al. Peak plasma interleukin-6 and
64. Huang WT, Tsai SM, Lin MT. Involvement of brain glutamate release in other peripheral markers of inflammation in the first week of ischaemic
pyrogenic fever. Neuropharmacology 2001;41:811–18. stroke correlate with brain infarct volume, stroke severity and long-term
65. Morganti-Kossmann MC, Rancan M, Otto VI, et al. Role of cerebral outcome. BMC Neurol 2004;4:2.
inflammation after traumatic brain injury: a revisited concept. Shock 90. Rothenburg LS, Herrmann N, Swardfager W, et al. The relationship
2001;16:165–77. between inflammatory markers and post stroke cognitive impairment.
66. Adachi H, Fujisawa H, Maekawa T, et al. Changes in the extracellular J Geriatr Psychiatry Neurol 2010;23(3):199–205. Epub 2010 Jul 2.
glutamate concentrations in the rat cortex following localized by 91. Blanco M, Castellanos M, Rodriguez-Yanez M, et al. High blood pressure
hyperthermia. Int J Hyperthermia 1995;11:587–99. and inflammation are associated with poor prognosis in lacunar
67. Chatzipanteli K, Alonso OF, Kraydieh S, et al. Importance of posttraumatic infarctions. Cerebrovasc Dis 2006;22:123–9.
hypothermia and hyperthermia on the inflammatory response after fluid 92. Castellanos M, Castillo J, Garcia MM, et al. Inflammation-mediated damage
percussion brain injury: biochemical and immunocytochemical studies. in progressing lacunar infarctions: a potential therapeutic target. Stroke
J Cereb Blood Flow Metab 2000;20:531–42. 2002;33:982–7.
68. Rovlias A, Kotsou S. The influence of hyperglycemia on neurological 93. Leira R, Rodriguez-Yanez M, Castellanos M, et al. Hyperthermia is a
outcome in patients with severe head injury. Neurosurgery 2000;46:335–42; surrogate marker of inflammation-mediated cause of brain damage in
discussion 342–3. acute ischaemic stroke. J Intern Med 2006;260:343–9.
69. Saper CB, Breder CD. The neurologic basis of fever. N Engl J Med 94. Oto J, Suzue A, Inui D, et al. Plasma proinflammatory and anti-
1994;330:1880–6. inflammatory cytokine and catecholamine concentrations as predictors
70. Young B. Nutritional and metabolic management of the head-injured of neurological outcome in acute stroke patients. J Anesth 2008;22:
patient. Neurotrauma 1996:345–63. 207–12.
71. McGirt MJ, Mavropoulos JC, McGirt LY, et al. Leukocytosis as an 95. Vila N, Castillo J, Davalos A, et al. Proinflammatory cytokines and early
independent risk factor for cerebral vasospasm following aneurysmal neurological worsening in ischemic stroke. Stroke 2000;31:2325–9.
subarachnoid hemorrhage. J Neurosurg 2003;98:1222–6. 96. Shiozaki T, Hayakata T, Tasaki O, et al. Cerebrospinal fluid concentrations
72. Shahlaie K, Keachie K, Hutchins IM, et al. Risk factors for posttraumatic of anti-inflammatory mediators in early-phase severe traumatic brain
vasospasm. J Neurosurg 2011;115(3):602–11. Epub 2011 Jun 10. injury. Shock 2005;23:406–10.
73. Hanafy KA, Morgan Stuart R, Fernandez L, et al. Cerebral inflammatory 97. Gruber A, Rossler K, Graninger W, et al. Ventricular cerebrospinal fluid and
response and predictors of admission clinical grade after aneurysmal serum concentrations of STNFR-I, IL-1RA, and IL-6 after aneurysmal
subarachnoid hemorrhage. J Clin Neurosci 2010;17(1):22–5. Epub 2009 subarachnoid hemorrhage. J Neurosurg Anesthesiol 2000;12:297–306.
Dec 8. 98. Kwon KY, Jeon BC. Cytokine levels in cerebrospinal fluid and delayed
74. Gupta AK, Al-Rawi PG, Hutchinson PJ, et al. Effect of hypothermia on ischemic deficits in patients with aneurysmal subarachnoid hemorrhage.
brain tissue oxygenation in patients with severe head injury. Br J Anaesth J Korean Med Sci 2001;16:774–80.
2002;88:188–92. 99. Takizawa T, Tada T, Kitazawa K, et al. Inflammatory cytokine cascade
75. Rossi S, Zanier ER, Mauri I, et al. Brain temperature, body core released by leukocytes in cerebrospinal fluid after subarachnoid
temperature, and intracranial pressure in acute cerebral damage. J Neurol hemorrhage. Neurol Res 2001;23:724–30.
Neurosurg Psychiatry 2001;71(4):448–54. 100. Tam AK, Ilodigwe D, Mocco J, et al. Impact of systemic inflammatory
76. Rumana CS, Gopinath SP, Uzura M, et al. Brain temperature exceeds response syndrome on vasospasm, cerebral infarction, and outcome after
systemic temperature in head-injured patients. Crit Care Med 1998;26: subarachnoid hemorrhage: exploratory analysis of CONSCIOUS-1
562–7. database. Neurocrit Care 2010;13(2):182–9.
77. Soukup J, Zauner A, Doppenberg EM, et al. The importance of brain 101. Dhar R, Diringer MN. The burden of the systemic inflammatory response
temperature in patients after severe head injury: relationship to intracranial predicts vasospasm and outcome after subarachnoid hemorrhage. Neurocrit
pressure, cerebral perfusion pressure, cerebral blood flow, and outcome. Care 2008;8(3):404–12.
J Neurotrauma 2002;19:559–71. 102. Sarrafzadeh A, Schlenk F, Gericke C, et al. Relevance of cerebral
78. Spiotta AM, Stiefel MF, Heuer GG, et al. Brain hyperthermia after interleukin-6 after aneurysmal subarachnoid hemorrhage. Neurocrit Care
traumatic brain injury does not reduce brain oxygen. Neurosurgery 2010;13(3):339–46.
2008;62:864–72; discussion 872. 103. Zanier ER, Brandi G, Peri G, et al. Cerebrospinal fluid pentraxin 3 early
79. Stocchetti N, Protti A, Lattuada M, et al. Impact of pyrexia on after subarachnoid hemorrhage is associated with vasospasm. Intensive
neurochemistry and cerebral oxygenation after acute brain injury. J Neurol Care Med 2011;37(2):302–9. Epub 2010 Nov 12.
Neurosurg Psychiatry 2005;76(8):1135–9. 104. Suzuki Y, Osuka K, Noda A, et al. Nitric oxide metabolites in the cisternal
80. Oddo M, Frangos S, Milby A, et al. Induced normothermia attenuates cerebral spinal fluid of patients with subarachnoid hemorrhage.
cerebral metabolic distress in patients with aneurysmal subarachnoid Neurosurgery 1997;41(4):807–11.
hemorrhage and refractory fever. Stroke 2009;40(5):1913–6. 105. Rejdak K, Petzold A, Sharpe MA, et al. Serum and urine nitrate and nitrite
81. Dengler J, Schefold JC, Graetz D, et al. Point-of-care testing for are not reliable indicators of intrathecal nitric oxide production in acute
interleukin-6 in cerebro spinal fluid (CSF) after subarachnoid brain injury. J Neurol Sci 2003;208:1–7.
haemorrhage. Med Sci Monit 2008;14(12):BR265–8. 106. Woszczyk A, Deinsberger W, Boker DK. Nitric oxide metabolites in cisternal
82. Maiuri F, Gallicchio B, Donati P, et al. The blood leukocyte count and its CSF correlate with cerebral vasospasm in patients with a subarachnoid
prognostic significance in subarachnoid hemorrhage. J Neurosurg Sci haemorrhage. Acta Neurochir (Wien) 2003;145(4):257–63.
1987;31:45–8. 107. Bell MJ, Kochanek PM, Doughty LA, et al. Interleukin-6 and interleukin-10
83. Rovlias A, Kotsou S. The blood leukocyte count and its prognostic in cerebrospinal fluid after severe traumatic brain injury in children.
significance in severe head injury. Surg Neurol 2001;55:190–6. J Neurotrauma 1997;14:451–7.
Section II—Clinical and Laboratory Assessment 153.e3

108. Hayakata T, Shiozaki T, Tasaki O, et al. Changes in CSF S-100B and 114. Ley EJ, Clond MA, Singer MB, et al. IL6 deficiency affects function after
cytokine concentrations in early-phase severe traumatic brain injury. Shock traumatic brain injury. J Surg Res 2011;170(2):253–6. Epub 2011 Mar 29.
2004;22:102–7. 115. de Waal Malefyt R, Abrams J, Bennett B, et al. Interleukin 10(IL-10) inhibits
109. Kossmann T, Stahel PF, Lenzlinger PM, et al. Interleukin-8 released into the cytokine synthesis by human monocytes: an autoregulatory role of il-10
cerebrospinal fluid after brain injury is associated with blood-brain barrier produced by monocytes. J Exp Med 1991;174:1209–20.
dysfunction and nerve growth factor production. J Cereb Blood Flow 116. McCombe PA, Read SJ. Immune and inflammatory responses to stroke:
Metab 1997;17:280–9. good or bad? Int J Stroke 2008;3:254–65.
110. Perez-Barcena J, Ibáñez J, Brell M, et al. Lack of correlation among 117. Suzuki S, Tanaka K, Suzuki N. Ambivalent aspects of interleukin-6 in
intracerebral cytokines, intracranial pressure, and brain tissue oxygenation cerebral ischemia: Inflammatory versus neurotrophic aspects. J Cereb Blood
in patients with traumatic brain injury and diffuse lesions. Crit Care Med Flow Metab 2009;29:464–79.
2011;39(3):533–40. 118. Viviani B, Bartesaghi S, Corsini E, et al. Cytokines role in neurodegenerative
111. Csuka E, Morganti-Kossmann MC, Lenzlinger PM, et al. IL-10 levels in events. Toxicol Lett 2004;149:85–9.
cerebrospinal fluid and serum of patients with severe traumatic brain 119. Shore PM, Thomas NJ, Clark RS, et al. Continuous versus intermittent
injury: relationship to IL-6, TNF-alpha, TGF-beta1 and blood-brain barrier cerebrospinal fluid drainage after severe traumatic brain injury in children:
function. J Neuroimmunol 1999;101:211–21. effect on biochemical markers. J Neurotrauma 2004;21:1113–22.
112. Maier B, Schwerdtfeger K, Mautes A, et al. Differential release of 120. Duo J, Stenken JA. In vitro and in vivo affinity microdialysis sampling of
interleukines 6, 8, and 10 in cerebrospinal fluid and plasma after traumatic cytokines using heparin-immobilized microspheres. Anal Bioanal Chem
brain injury. Shock 2001;15:421–6. 2011;399:783–93.
113. Singhal A, Baker AJ, Hare GM, et al. Association between cerebrospinal 121. Hutchinson PJ, O’Connell MT, Rothwell NJ, et al. Inflammation in human
fluid interleukin-6 concentrations and outcome after severe human brain injury: intracerebral concentrations of IL-1alpha, IL-1beta, and their
traumatic brain injury. J Neurotrauma 2002;19:929–37. endogenous inhibitor IL1-ra. J of Neurotrauma 2007;24:1545–57.
II
Chapter
17  

Infection
Barnett R. Nathan and John J. Stern

Introduction Clinical Presentation of Bacterial Central


Infections in the intensive care setting are common, both as Nervous System Space-Occupying Lesions
the reason for the intensive care unit (ICU) admission and Parenchymal brain abscess may present with many symp-
as a nosocomial complication of critical illness. Nosocomial toms. Although the classic triad of fever, headache, and focal
infections, now referred to as hospital acquired infections neurologic signs is helpful, this occurs in less than half of
(HAIs), are thought to complicate between one quarter and patients, and the presentation may be more of a mass lesion
one half of all ICU admissions. In the Extended Prevalence than an infection. Other manifestations, including symptoms
of Infection in Intensive Care (EPIC II) study, a 1-day, pro- and signs of the original infection (otitis or sinusitis), may
spective, point prevalence study, 51% of 13,796 adult (older be present and more impressive. Although the average course
than 18 years) patients in 1265 participating ICUs from 75 of brain abscesses from the time of symptom presentation to
countries were considered infected; 9084 (71%) were receiv- hospital admission can be as short as 5 days,3 the course may
ing antibiotics.1 The ICU mortality rate of the infected be more indolent. Subdural empyema also may present with
patients was twofold greater than that of noninfected the triad of sinusitis, fever, and neurologic deficit. However,
patients (25% vs. 11%). Although central nervous system signs of cortical irritation such as seizures (50%), raised
(CNS) infections are far less common as admission diagno- intracranial pressure (headache, vomiting, and papilledema
ses or as complications of critical care, these infections, [50%]), or focal deficits (75%) are a frequent presentation.
including the hospital-acquired complications of neurosurgi- Spinal epidural abscess is a neurosurgical emergency, and the
cal intervention and the community-acquired infections of typical presenting features include back pain, malaise, and
meningitis, encephalitis, and brain abscess are the focus of fever. Neurologic deficits associated with spinal epidural
this chapter. Each of these infections is discussed separately abscesses often are associated with venous thrombophlebitis
to provide insights on what tests are needed to make the and so may evolve rapidly or be greater than the lesion size
diagnosis. Infections such as catheter-associated bloodstream suggests.
infections (BSIs) and ventilator-associated pneumonia (VAP)
are reviewed in Chapter 6. Finally, the role of infection sur-
veillance, prevention, and control is reviewed. Epidemiology
The frequency of brain abscesses in the United States ranges
from 0.3 to 1.3 cases per 100,000 persons per year, with a
Bacterial Infections predominance occurring in men between the ages of 30 to 40.
Twenty five percent of all brain abscesses occur in children
Bacterial Central Nervous System between the ages of 4 to 7, resulting from either cyanotic heart
Space-Occupying Lesions disease or extension from an otic source. The organism causing
Examples of space-occupying lesions of the CNS include the infection depends in part on the source of the infection
parenchymal abscesses, epidural abscesses, and empyemas. and the patient’s age. Common organisms in adults include
The most common sources of brain abscesses are from hema- anaerobic organisms, in particular Bacteroides and Peptostrep-
tologic spread (most common lung), direct extension from a tococcus in some series, whereas in others, aerobic organisms
parameningeal source (including the sinuses, middle ear, or especially Staphylococcus, Streptococcus (most common),
after dental procedures, head trauma, or surgery), suppurative Enterobacteriaceae, and Haemophilus are more common. In
lung disease, or congenital heart disease (e.g., patent foramen infants Proteus and Citrobacter are the most frequent cause,
ovale or patent ductus arteriosus, i.e., right to left shunting). whereas in children (3-5 years old) Haemophilus, Streptococcus
Rare causes of brain abscess have been reported after tongue pneumoniae, and Bacteroides fragilis (otogenic spread) are
piercing or endovascular occlusion of aneurysms using Gug- common. Staphylococcus, S. pneumoniae, and Haemophilus
lielmi detachable coils.2 Infective endocarditis occasionally influenzae are common from sinus spread and Streptococcus
presents first with a brain abscess. or Staphylococcus aureus from cyanotic heart disease. Since
154 © Copyright 2013 Elsevier Inc. All rights reserved.
Section II—Clinical and Laboratory Assessment 155

1981 the incidence of human immunodeficiency virus (HIV)– Microbiologic Analysis


related brain abscesses due to toxoplasmosis has become the Blood cultures should always be obtained on admission to
most common cause of a protozoal space-occupying brain identify a potential hematogenous source for the CNS or epi-
lesion and must be considered in any patient presenting with dural infection. CNS-obtained material must be sent to a labo-
a ring-enhancing brain lesion. These lesions may be single or ratory for Gram stain, routine aerobic and anaerobic culture,
multiple and should lead to expeditious HIV serologic testing. and in select patients for mycobacterial and fungal cultures.
Aspergillus is the most common fungal organism to cause a In the immunocompromised patient, a silver stain should be
brain abscess. requested to identify Aspergillus species along with a request
Subdural empyemas (SDEs)—a collection of purulence for modified acid-fast bacillus (AFB) staining to potentially
located between the arachnoid and dura—account for 15% to identify a Nocardia species. In an HIV-infected patient patho-
20% of all space-occupying infections and result from exten- logic specimens should be sent to a lab to look for Toxoplasma
sion of a suppurative process in a paranasal sinus or otorhi- gondii tachyzoites. In the event that routine cultures fail to
nologic infection. Less commonly, subdural empyemas result grow after 4 to 5 days, the clinician must consider whether he
from head trauma or after a neurosurgical procedure. Tewari or she is dealing with a routine anaerobic process that failed
et al., found the majority of SDEs are identified in infancy to grow or a more unusual pathogen such as a Nocardia
through the third decade of life (60%).4 species, fungal or mycobacterial infection, or T. gondii in an
Spinal epidural abscesses usually occur as the result of HIV-infected patient. In these patients, consultation with an
hematogenous seeding of the epidural space from a distant infectious diseases expert and the microbiology laboratory is
suppurative focus such as infective endocarditis, from an advised.
infected central line catheter, or from the injection of intrave-
nous drugs. S. aureus, and less commonly aerobic and anaero-
bic streptococci, and gram-negative rods cause the vast Follow-up
majority of epidural infections. The most effective approach to evaluate a patient’s response
to therapy is to follow the patient’s temperature and WBC
trend and the patient’s mental status and to obtain CNS
Diagnosis of Central Nervous System imaging on a regular basis to assess structural improvement.
Space-Occupying Lesions
Fever with or without a peripheral white count and symptoms
of new CNS deficit suggest an intracranial infection. Other Management of Space-Occupying
signs and symptoms such as headache, vomiting, and papill- Infections
edema are not specific to these diagnoses. Cranial imaging with Treatment of brain abscess is both surgical and medical.
computed tomography (CT) or magnetic resonance imaging Abscesses greater than 2.5 cm in diameter or those associ-
(MRI) are sensitive tests; MRI is able to provide more infor­ ated with mass effect should be excised or aspirated. Aspira-
mation and better resolution.5 On MRI diffusion-weighted tion may be performed with image guidance and through a
imaging (DWI) an intraparenchymal abscess demonstrates twist drill or burr hole. Brain imaging should be used to
high signal on DWI and low apparent diffusion coefficient follow the treatment response, with repeat scans every 1 to 2
(ADC) in the abscess cavity.6,7 In subdural empyema, there weeks.
may be a disproportionate amount of underlying cortical and In patients with ventriculitis, meningitis, or hydrocephalus
white matter edema and enhancement relative to the size of that requires cerebrospinal fluid (CSF) drainage, or those with
the fluid collection. In patients with spinal epidural abscess inaccessible abscesses or early abscess formation (cerebritis),
there may evidence of diskitis and vertebral body infection. medical treatment alone can be attempted preferably after
However, even imaging does not have the sensitivity required microbiologic analysis on blood or CSF. Broad-spectrum
to definitively diagnose all infections. Lumbar puncture should antibiotics are started and refined when culture results are
not be performed in those with brain abscesses given the risk available. Empiric drug regimens for immunocompetent
of herniation. patients should include coverage for methicillin-resistant
S. aureus (MRSA), anaerobes, and gram-negative bacilli
Laboratory and Microbiologic Analysis (including Pseudomonas coverage in the setting of trauma or
post neurosurgery) with a regimen such as vancomycin, met-
Laboratory Studies ronidazole, and cefotaxime, typically for 6 to 8 weeks. The
The most useful blood test is the complete blood count with treatment response can be followed initially with weekly brain
a manual differential. Although an elevated white blood cell imaging. This can be spaced out to every 2 weeks during the
(WBC) count is always helpful, patients presenting with a remainder of the 6- to 8-week antibiotic course, with follow-up
significant suppurative process may have a normal peripheral scans every 2 to 4 months for the following year to assess for
WBC count. In these patients bandemia may be the only recurrences.
abnormal finding to guide the clinician toward making the The treatment of subdural empyema is surgical either
diagnosis of an infectious process. It is imperative that the through a craniotomy or burr holes. Early treatment (within
treating team request a manual differential on presentation. 72 hours of symptom onset or sooner) is preferable.8 Broad-
Furthermore, a markedly elevated platelet count such as one spectrum empiric antibiotics (similar to that described for
greater than 500,000 can also help suggest the presence of a brain abscess) should be started early. Once the cause of the
suppurative CNS process. An elevated erythrocyte sedimenta- infection is defined from culture analysis, antibiotics can be
tion rate (ESR) and C-reactive protein (CRP) may be useful tailored to the organism and continued for 3 to 4 weeks
but are nonspecific. after drainage. Spinal epidural abscess is a neurosurgical
156 Section II—Clinical and Laboratory Assessment

emergency. Without treatment, severe and permanent spinal Diagnosis


cord damage can occur in just a few hours in part because of The diagnosis of bacterial meningitis is made based on the
venous thrombosis rather than compression alone. Treatment clinical presentation and CSF examination. At the time of a
includes surgical decompression and drainage, a search for lumbar puncture many patients have an increased opening
the infectious source (if possible), and antibiotics (empiric pressure (>15 cm H2O). CSF total WBC count is in the 1000
as described earlier, then specific antibiotics based on the to 10,000 range, and almost all patients with bacterial menin-
pathogens). gitis have a predominance of neutrophils. CSF glucose is
usually decreased, although the CSF-to-serum ratio is more
accurate. The CSF protein is almost always elevated (>100 g/
Outcome dL). C-reactive protein (CRP), either in the serum or the CSF,
The mortality of patients with brain abscess is between or procalcitonin can help differentiate bacterial meningitis
20% and 30%9 and greater in those with a depressed level from viral meningitis. The specific diagnosis is made on CSF
of consciousness or in coma at admission and distant meta- Gram stain and culture.
static focus of infection. Factors such as age, multiple abscesses, The Gram stain, developed by Hans Christian Gram in
and steroid use do not appear to alter outcomes. Mortality Berlin in the late 19th century, colors bacteria by using a
in subdural empyema is between 10% and 40%. Younger crystal violet stain to colorize what are referred to as gram-
age and early treatment is associated with better outcome.4,8 positive organisms blue, namely streptococci and staphylo-
For example, Renaudin and Frazee observed that patients cocci. In a subsequent step the specimen is then decolorized
treated in less than 72 hours had less than 10% mortality, with alcohol and counter-stained with the red dye, safranin.
whereas 70% of patients died or were disabled when treated Gram-negative bacteria such as coliforms are unable to hold
more than 72 hours after symptom onset.8 Outcome also their crystal violet stain but take up the safranin stain, render-
is associated with neurologic status at presentation and ing these bacteria red. These procedures are now done exclu-
whether sinus thrombophlebitis occurs and its extent. sively in a microbiology laboratory. This procedure can guide
Outcome after spinal epidural abscess is associated with antibiotic therapy until culture and sensitivity results are
how quickly the diagnosis is made and surgical decompression available.
is achieved and the severity of the spinal cord symptoms A head CT scan is typically not required and not needed to
at the time of decompression. The more symptoms at the diagnosis bacterial meningitis. However, it can be useful to
time of decompression, the worse the outcome, although exclude mass lesions that are a contradiction to lumbar punc-
compared with similar neurologic presentations in traumatic ture (LP). The literature supporting the necessity of doing a
spinal cord injury, those with epidural abscess tend to do CT scan of the brain prior to LP is minimal. Between 3% and
better.10 4% of patients with meningitis develop herniation syn-
dromes12,13; about 1% of these patients herniate within several
Bacterial Meningitis hours of the LP (1%). However, a normal CT scan does not
eliminate the risk for herniation nor does an abnormal CT
Epidemiology scan predict herniation after LP.14 In 2004 Tunkel et al.15 wrote
S. pneumoniae (or pneumococcus) and Neisseria meningitidis the “Practice Guidelines for the Management of Bacterial
remain the most common etiology for community- Meningitis.” A head CT is recommended before LP in patients
acquired bacterial meningitis. With the widespread use of with suspected bacterial meningitis who (a) have a history of
H. influenzae–conjugated vaccine in children, the incidence of CNS disease, (b) are immunocompromised, (c) present with
H. influenzae meningitis has decreased.11 S. pneumoniae (or a seizure, or have (d) papilledema, (e) an abnormal level of
pneumococcus) is now the most common cause of meningi- consciousness, or (f) a focal neurologic deficit. Furthermore,
tis in the United States, although its incidence too is decreas- in the setting of suspected bacterial meningitis the decision to
ing with the use of the multivalent pneumococcal vaccine. begin antimicrobial therapy should be made expeditiously
Group B streptococcus is the most common cause of bacterial and independent of when and if a head CT is performed
meningitis in newborns but rare after the neonatal period, because earlier treatment is associated with better outcome.
N. meningitidis in children, teens, and young adults and
pneumococcus is the most common cause of bacterial men-
ingitis in adults. Management
Treatment of bacterial meningitis should not be delayed while
waiting for a head CT scan. Empiric treatment of the patient
Clinical Features should be provided based on the patient’s relative risk for
The typical presenting signs and symptoms of bacterial specific organisms. Most immunocompetent adults are at
meningitis are headache, fever, stiff neck, and altered mental highest risk for S. pneumoniae. Those greater than 50 years old
status that each occur in 80% of patients. Kernig’s sign and are also at risk for Listeria monocytogenes. Teens and young
Brudzinski’s sign are found in 50% of patients. In most adults are also susceptible to N. meningitidis (meningococ-
cases of bacterial meningitis there also is associated inflam- cus). Empiric antibiotics should be administered based on
mation of the underlying brain tissue (cerebritis). In addi- these likely causes. Beta-lactam–resistant pneumococcus is
tion, secondary venous sinus thrombosis, vasculitis, or becoming more prevalent in the community (up to 15% in
cranial nerve inflammation may occur. Therefore focal neu- some metropolitan areas in the United States) and so vanco-
rologic deficits are seen in about 20% of patients and sei- mycin should be used as an empiric treatment in the appropri-
zures in about 30% of patients. Papilledema at presentation ate patient population. Once the Gram stain is obtained
is uncommon (<1%). and organisms identified by morphology and staining
Section II—Clinical and Laboratory Assessment 157

characteristics the antibiotics can be modified to be more Clinical Features


specific. The antibiotics can be changed once again if neces- The common clinical manifestations of HSE are a change in
sary once the organism has been grown and identified in personality, seizures, and decreased level of consciousness.
culture and sensitivities have been obtained. Antibiotic treat- Fever is common, and focal or generalized seizures occur in
ment durations are based on the organism found. Both two thirds of patients. Focal neurologic findings such aphasia
meningococcus and H. influenzae can be treated successfully or neglect associated with a cortical abnormality or dysphagia
for 7 days. S. pneumoniae requires 2 weeks of treatment, and hemiparesis associated with cortical dysfunction may be
whereas Listeria and other gram-negative organisms require seen. Headache, nausea, vomiting, and papilledema also are
at least 3 weeks of treatment. In addition, an infectious disease common. The clinical presentation may be subtle and some-
consultant should guide antibiotic therapy. what slow moving.
The role of beta corticosteroids has been controversial
since the early 1990s. A randomized clinical trial of dexa-
methasone (10 mg of dexamethasone every 6 hours for 4 Diagnosis
days) that included 301 patients with bacterial meningitis has The diagnosis of HSE is made based on the clinical course,
been published.16 The first dose of steroid was given before imaging (CT and MRI), and laboratory evaluation, in particu-
antibiotic administration. Outcome was better in those who lar CSF analysis and CSF PCR. Electroencephalography and
received dexamethasone. This improvement in outcome was brain biopsy may be helpful in some patients, but brain biop-
almost exclusively in the patients who had S. pneumoniae sies are now infrequently performed in this setting. The CSF
meningitis; however, due to a small sample size, conclusions WBC count in patients with HSE is in the 10s to the 100s but
regarding the effectiveness of steroids in H. influenzae and values of 1000 to 2000 cell/mm3 may be observed. Most of the
meningococcal meningitis are unclear. Because at the time of WBCs are lymphocytes. However, 10% to 25% of patients can
presentation the offending organism is unknown, the dexa- have high numbers of neutrophils in their CSF early in the
methasone can be started before antibiotics. If an organism disease course. Red cells in the range of 10s to 1000s/mm3 are
other than S. pneumoniae is found to be the offending patho- seen in the CSF of about 50% of patients and xanthochromia
gen, clinical guidelines for treatment of bacterial meningitis is common. CSF protein may be mild to moderately elevated
recommend against continued routine use of corticoste- in approximately 50% of patients. Glucose is usually normal
roids.15 Good supportive intensive care medicine is important but may be slightly reduced. Opening pressure is elevated in
in these patients. In bacterial meningitis increased intracra- one third of patients. The CSF PCR for herpes simplex is now
nial pressure does occur, yet it is unclear whether monitoring the test of choice for diagnosis and has a sensitivity of 96%
of intracranial pressure or treatment with hypertonic agents and specificity of 99%.17 Very early in the disease (<24 hours)
such as mannitol or hypertonic saline is useful in this patient the test may be negative, and with treatment the sensitivity
population. may decrease.
On MRI imaging, temporal lobe abnormalities, often hem-
orrhagic, are frequently found. Hypodensities in the temporal
Herpes Simplex Encephalitis lobe or frontal lobes are found on CT in a severe case of herpes
encephalitis. On MRI there are hyperintensities on T2 and
Epidemiology gadolinium enhancement around the lesion in the lobe that is
Herpes simplex viruses (HSVs) are distributed worldwide, and infected. Electroencephalograms (EEGs) are sensitive (84%)
humans are the sole reservoir of this virus. Between 25% and but nonspecific (32% specificity) for HSE. The characteristic
60% of humans are seropositive for herpes simplex virus by EEG demonstrates spike and slow activity with periodic later-
adulthood. Herpes simplex encephalitis (HSE) is the most alized epileptiform discharge (PLED). Today brain biopsy
common cause of sporadic fatal encephalitis in the United rarely is required.
States. It is estimated to occur in 1/250,000 to 1/500,000 in the
United States or approximately 250 to 500 cases per year.
Treatment
Acyclovir (10 mg/kg every 8 hours for 21 days assuming
Pathophysiology normal renal function) is the drug of choice to treat HSE.18
It is unclear whether herpes simplex encephalitis represents a Higher doses and a longer treatment duration may help
primary or recurrent infection. The virus can lie dormant in prevent relapse.18 Outcome for HSE has improved signifi-
neurons and ganglia once the patient has been infected. cantly since the introduction of acyclovir, but mortality
However, it has been demonstrated that patients with HSE remains 19% at 6 months and 28% at 18 months.19 Predictors
and concomitant cutaneous herpes simplex may have two of poor outcome include age greater than 30 years and coma
different strains of HSV. No specific triggers for HSE have on presentation. In these patients mortality is nearly 70%.19
been identified. Immunosuppression does not predispose Significant neurologic sequelae are often observed in those
patients to HSE. However, patients who are immunosup- that survive.
pressed are at risk for a more aggressive disease with a worse
outcome.
HSE is always a cortical infection, and no cases of HSE have West Nile Viral Encephalitis
been reported to involve the brainstem. The temporal lobe is
the most commonly infected brain region, although other Characteristics of the West Nile Virus
lobes can develop encephalitis. The virus causes a hemor- The West Nile virus (WNV) is a single-stranded RNA virus
rhagic and necrotizing encephalitis. that belongs to the family Flaviviridae. This family of viruses
158 Section II—Clinical and Laboratory Assessment

includes the Japanese encephalitis virus, the St. Louis cortex, brainstem, basal ganglia, or spinal cord also may be
encephalitis virus, and the Kunjin virus. There is serologic seen on MRI.27,28
cross-reactivity with all of these viruses, and so those vac-
cinated against one may be seropositive against another.
This virus was thought to have originated in the West Nile Treatment and Prognosis
valley of Uganda. Genetic linkage of the WNV epidemic The care of patients who develop WNVE can be very resource
in the United States suggests that the viral serotype virus intensive, particularly in those who develop motor weakness
comes from the Middle East. Clinically, only the United and use up many ICU patient-days.29 There is currently no
States and Israeli WNV infections have caused severe human proven treatment for WNV infection, although there are small
disease. case series using ribavirin, interferon alpha-2b, hyperimmune
gammaglobulin,30,31 and a monoclonal antibody to the WNV.
The mortality in those patients who develop WNVE (usually
Epidemiology the elderly) is 10% to 18%29 and about half the survivors do
WNV infection is now considered epidemic in the United not return to their premorbid function. In those who develop
States.20 Between 1999 and 2008 there were more than 31,000 motor neuron damage up to two thirds have continued weak-
cases and 1150 deaths (≈3.5% mortality). Of these cases nearly ness. One year after discharge, 55% of patients report that they
12,000 (≈35%) were West Nile virus encephalitis (WNVE).21, 22 were not fully recovered; symptoms include fatigue, weakness,
The incidence of WNVE increases in patients greater than 50 gait difficulty, and memory problems.32
years old; there is a 10-times higher risk of meningitis or
encephalitis in those who are 50 to 59 years old and a 43-times
higher risk of encephalitis in patients older than 80 years of Infections After Neurosurgical
age. There also is an increased risk of encephalitis in patients Procedures
who are immunocompromised secondary to malignancy,
transplant, or HIV. Background
Infections after neurosurgical procedures, including those
associated with intracranial monitors or drains (e.g., external
Clinical Presentation ventricular drains or lumbar drains), or after cranial or spinal
There is a clinical spectrum of West Nile viral infections that surgical procedures are rare but when they occur contribute
ranges from mild, almost asymptomatic cases to the most to prolonged length of stay in the ICU or hospital and adversely
severe version, encephalitis. WNV infection without enceph- affect outcome.33
alitis is typically asymptomatic or a “flulike illness” with
fever, malaise, myalgias, and headache. In those patients who
develop WNVE, fever, fatigue, nausea, vomiting, headache, External Ventricular Drains
myalgias and altered mental status are common. Half of the The external ventricular drain or ventriculostomy was devel-
patients may develop objective weakness23 that appears to be oped in the early 1950s. During the 1960s Lundberg reported
associated with anterior horn cell injury in the spinal cord positive CSF cultures in 6% of patients but no clinical infec-
appearing similar to a polio presentation.24 These patients tions in patients with ventriculostomies.34 Most reports that
also may develop compromised respiratory function or frank describe infections associated with external ventricular drains
respiratory failure. A subset of WNVE patients can present (EVDs) are retrospective: the incidence is between zero and
with movement disorders associated with basal ganglia 22%. Lozier et al.35 in a literature review of ventriculostomy-
involvement.25 related infections, reported a combined infection rate between
8% and 9%. However, the manner in which ventriculostomy-
associated infections are described varies greatly and not every
Diagnosis paper defines what an “infection” is. There are several possible
The diagnosis of WNV is made by examination of immuno- scenarios based on the ranked certainty of infectious ventricu-
globulin M (IgM) antibodies in the serum or CSF. Because litis: (1) ventriculostomy-related infection, (2) suspected
IgM does not cross the blood-brain barrier, the finding of ventriculostomy-related infection, (3) ventriculitis, (4) ven-
IgM antibodies specific for WNV in the CSF indicates an triculostomy colonization, and (5) contamination (Table
acute infection with the virus. Vaccines to yellow fever and 17.1).35 According to this classification a positive CSF culture
Japanese encephalitis are cross-reactive with the antibodies from an EVD means colonization or contamination when
to WNV; however, this is IgG rather than IgM. The remain- other CSF parameters are normal. When the CSF WBC count
ing CSF analysis is nonspecific: up to 2000 white cells, pre- and protein also are abnormal, then there is infection in the
dominantly lymphocytes, a mild to moderately elevated face of a positive CSF culture. Infections associated with
protein, and normal glucose are found.23,26 Peripheral blood lumbar drain infection are about 3%, although fewer studies
analysis typically is nonspecific; the WBC count is normal have addressed this question.36
or slightly elevated, or lymphopenia may be observed.26 Risk factors for infection associated with ventriculostomies
Patients who develop encephalitis may have hyponatremia include intraventricular hemorrhage, subarachnoid hemor-
associated with the syndrome of inappropriate antidiuretic rhage, open depressed skull fracture, basilar cranial fracture
hormone (SIADH) secretion. Imaging studies are nonspe- with CSF leak, neurosurgical operation, ventriculostomy irri-
cific. Head CTs often are normal. However, one third of gation, systemic infection, and longer duration of catheteriza-
patients have enhancement of the meninges or periventricu- tion.35 However, the role of insertion duration remains unclear
lar areas on brain MRI. Abnormalities that involve the and it seems that if no infection has developed by 9 days, then
Section II—Clinical and Laboratory Assessment 159

Table 17.1  Classification Scheme of Ventriculostomy-Related Infections


Suspected
Ventriculostomy- Ventriculostomy- Ventriculostomy
Parameters Ventriculitis Related Infection Related Infection Colonization Contamination
Cultures/GS Variable One or more Absence Multiple positive Isolated culture
culture or GS cultures or GS or GS
Glucose Low Declining Declining Normal/expected Normal/expected
Protein High Increasing Increasing Normal/expected Normal/expected
CSF WBC Pleocytosis Increasing Increasing Normal/expected Normal/expected
pleocytosis
Clinical Fever, meningeal signs, Fever None Sometimes fever None
photophobia,
altered mental status

Adapted from Lozier AP, Sciacca RR, Romagnoli MF, et al. Ventriculostomy-related infections: a critical review of the literature. Neurosurgery 2002;51(1):170–81;
discussion 181–2.
CSF, Cerebrospinal fluid; GS, Gram stain; WBC, white blood cell.

the risk may decline. Most studies suggest that gram-negative demonstrated a significant reduction in ventriculitis with less
bacilli and gram-positive cocci are the most common species frequent sampling.
that infect ventricular drains.37,38
Prevention
Intraparenchymal Pressure Monitors The data supporting the role of antibiotic prophylaxis or
The infection rate associated with intracranial pressure moni- changing the catheter to help prevent EVD associated infec-
tors (“bolts”) is reported to be between zero and 4%, much tions are conflicting. In a randomized trial Poon et al. com-
lower than that associated with ventriculostomies.39-42 In part pared continuous prophylactic ampicillin/sulbactam plus
this may result from the limited, if any, manipulation of a bolt aztreonam to a single dose at the time of the procedure.51
once inserted, whereas ventriculostomies may be sampled and Continuous antibiotic prophylaxis was associated with fewer
drugs injected into them. infections but greater mortality if an infection occurred. Ret-
rospective data suggest that the risk of infection increases once
an EVD has been in place between days 5 and 935 but that
Craniotomy Infections routine changes may not help prevent this.38,52 For example
Infections including meningitis or ventriculitis complicate Wong et al. in a randomized trial of 103 patients observed a
about 4% of craniotomies.43,44 The bacteria associated with similar infection rate whether catheters were changed every
these infections are typically skin flora such as Staphylococcus, 5 days or not.53 Some institutions use prophylactic vancomy-
Streptococcus, and less frequently gram-negative bacilli. Risk cin or other antibiotics in this setting but data as to its efficacy
factors for a craniotomy infection include emergency surgery, are lacking.
longer duration of surgery, reoperation during the same
admission, bleeding complications, CSF leak, or the surgeon
who performs the craniotomy.43 Treatment
When there is evidence of infection, treatment should begin
immediately. If cultures are available, the antibiotics should be
Diagnosis directed at the appropriate organisms. Without available cul-
The diagnosis of true ventriculostomy-associated infection tures or Gram stains, antibiotic choice should be directed at
has the following characteristics: (1) progressive decline in gram-negative bacilli (including Pseudomonas) and both
CSF glucose, (2) increasing CSF protein, (3) progressive CSF Streptococcus and Staphylococcus (including S. aureus).
pleocytosis, (4) one or more positive CSF cultures or Gram
stain, and (5) fever.36 Similar findings are evident when there
is ventriculitis, but the patient is far sicker. A reduced CSF-to- Surveillance and Evaluating
serum glucose ratio can help in diagnosis. A ratio of 0.4 has a the Febrile Neurocritical
sensitivity of 77% and a specificity of 87%.45 Other methods
including CSF lactate, cytokine analysis, and cell index
Care Unit Patient
studies45-47 are useful but lack sensitivity and specificity.48 The Often a patient in the neurocritical care unit (NCCU) (e.g., a
routine analysis of CSF to help predict infection does not postoperative neurosurgical patient), receives both steroids
appear to be helpful and instead the frequent access to the and antipyretics, making an assessment of the patient’s poten-
system may increase the risk of contamination or infec- tial for an infection challenging. Of paramount importance
tion.35,48,49 Furthermore, in a prospective patient cohort in is a careful daily examination of the patient, paying close
which CSF was sampled every 3 days compared with a histori- attention to level of consciousness, lung sounds, wounds,
cal control group sampled more frequently, Williams et al.50 skin, peripheral and central line insertion sites, presence of
160 Section II—Clinical and Laboratory Assessment

diarrhea, and urine clarity. Given the frequency of intubation the hospital environment, producing toxic megacolon, and
and ventilator support in these patients, the risk of a VAP is occasional deaths are now being reported in increasing
relatively high, and careful evaluation of the integrity and numbers particularly in the elderly.57 C. difficile colitis presents
position of the endotracheal tube (ET) should be made daily, with a fever and diarrhea but with few other clinical signs and
along with continual vigilance regarding the color and consis- symptoms. The diarrhea often has a unique odor. The diag-
tency of any sputum aspirated by the nursing staff. Should a nosis of C. difficile requires identification of C. difficile toxin
patient develop a fever (>38o C), a chest x-ray is indicated. A or B in diarrheal stool. However, the accuracy of these diag-
Modern ICU care of the intubated patient now includes nostic tests is limited and the optimal diagnostic testing algo-
careful mouth care using chlorhexidine-containing mouth rithm is still being elucidated; for example, it is difficult to
products, inline suctioning, and elevation of the head of the predict and prevent development of severe or relapsing C.
bed to 30 degrees, all in an effort to minimize VAPs (see difficile infection in patients who initially present with mild
Chapter 6). In a patient destined for prolonged ventilator symptoms.58 The use of probiotics such as Lactobacillus plan-
support, data now support early tracheotomy placement. tarum 299/299v plus fiber (LAB), to prevent and or help with
In the presence of an unexplained fever, blood cultures treatment of C. difficile is conflicting.59
also are essential to evaluate for a potential central venous Urinary tract infections (UTIs) and in particular catheter-
catheter (CVC)–associated infection. Although CVCs often associated UTIs, remain a common source of HAIs and
are silver impregnated to help reduce the incidence of account for 20% to 50% of all HAIs in the ICU.60 Longer
catheter-related infections, these infections remain common duration of Foley catheterization (>5 days) and poor urinary
and a threat to an ICU patient. Blood cultures should always catheter management, specifically disconnection of the closed
be obtained in a febrile patient as soon as possible and pref- system, are significant factors associated with hospital-
erably during the febrile episode to increase the yield of cap- acquired UTIs.61 In a patient with an indwelling Foley catheter
turing the offending organism. Customary practice is to (FC), simple evaluation of the urine may be confusing. Fur-
draw aerobic and anaerobic cultures both peripherally and thermore, nearly all patients with an FC in place for more than
through the central line. This method increases the yield, 4 weeks have pyuria and bacteriuria. In the febrile patient with
and, when a blood culture is positive from the central line cloudy urine and a fever, it is incumbent on the team of physi-
source yet negative from the periphery, points toward a cians to rule out other potential sources of fever before attrib-
central line source and early removal of the offending line. uting the fever to the urine. Treatment of UTIs with antibiotics
Occasionally Candida bloodstream infections occur in an is more likely to clear bacteriuria and relieve symptoms but
ICU patient (6.9 per 1000 patients), in particular in those also selects for resistant uropathogens and commensal bacte-
whom have been on broad-spectrum antibiotics for pro- ria and can adversely affect the gut and vaginal microbiota.
longed periods, on steroids, or are receiving TPN.54 The Uropathogens are increasingly becoming resistant to currently
source is most often from a central venous catheter (CVC) available antibiotics and the case-fatality rate of hospital-
and responds to removal of the catheter followed by a 10-day acquired UTIs remains high.62 Consequently, alternative strat-
course of an appropriate antifungal agent such as fluconazole egies including avoidance of inappropriate FC use, removal of
or caspofungin. Antigen detection and PCR assays could a Foley, intermittent catheterization, and condom catheters
provide an alternative to microscopy, culture, and serology to are necessary to manage UTIs.63
detect fungal infections, but further study and validation still
are required.
Other potential sources for fever are deep venous thrombo-
Sepsis
sis with or without pulmonary emboli that can present with Sepsis is associated with increased morbidity, mortality, and
tachycardia, fever, hypotension, and a sepsis-like syndrome. costs of care64 and remains the leading cause of death in criti-
Careful examination of the patient and his or her extremities cally ill patients.65 The definition of sepsis is provided in Table
can lead to a timely duplex ultrasound of the legs or arms if 17.2. The initial stage of sepsis is referred to as systemic
indicated. Numerous medications used in the NCCU patient inflammatory response syndrome (SIRS) and is defined as a
can produce fever including phenytoin, carbamazepine, van- temperature greater than 38o C or less than 36o C, pulse more
comycin, beta-lactam antibiotics, and blood products. Looking than 90 beats per minute, respiratory rate greater than 20 per
carefully for a new rash may assist in determining that the minute, and a WBC more than 12,000 or less than 4000.
fever is indeed a medication-related problem. A careful review However, there are no specific laboratory studies at present to
of the medication list is mandatory to evaluate a febrile post- help guide practitioners in defining an early septic patient
operative or NCCU patient. Serum procalcitonin, and use although serum procalcitonin levels may provide some insight
of scores such as the Simplified Acute Physiology Score II into severe infections.66,67 Instead careful attention to the
(SAPS II), Acute Physiology and Chronic Health Evaluation patient is the bedrock of early detection and treatment of an
(APACHE), or Sequential Organ Failure Assessment (SOFA)* infected patient, including a postoperative neurosurgical
may differentiate between infectious and noninfectious fever patient in the NCCU. Table 17.3 lists common laboratory
in the ICU.55 findings in septic patients. In addition, PCR testing provides
Clostridium difficile–induced colitis is an ever-expanding increased sensitivity for bloodstream infections.68
complication of hospitalization and primarily associated with The Surviving Sepsis Campaign (SSC) was launched in
antibiotic use but occasionally is associated with chemothera- 2002 as a collaborative initiative between the European
peutic agents.56 Recently identified strains of hypervirulent Society of Intensive Care Medicine (ESICM), the Interna-
C. difficile (PCR ribotype 027 or BI/NAP1/027) have entered tional Sepsis Forum (ISF), and the Society of Critical
Care Medicine (SCCM) to address the poor outcome associ-
*Note: SOFA also refers to Sepsis-Related Organ Failure Assessment. ated with sepsis. The SSC developed evidence-based
Section II—Clinical and Laboratory Assessment 161

management guidelines that were published in 2004 and others—have been identified to help evaluate quality of
updated in 200869 with the goal of improving outcomes in sepsis care and improve delivery of evidence-based care in
sepsis and septic shock. Since the development of these the ICU.73 Finally, ICU patients may be followed for disease
guidelines, more than 20,000 patients have been entered into progression using a number of techniques, for example, the
the SSC database; analysis of this showed that participation SOFA system (Table 17.4) that originally was developed by
in the SSC was associated with continuous quality improve- the ESICM to describe organ failure in sepsis.74,75 The SOFA
ment in sepsis care and a 5.4% absolute survival benefit.70,71 is a six-organ dysfunction/failure score that measures multi-
Compliance with the overall sepsis bundle and its individual ple organ failure daily. Each organ is graded from 0 (normal)
elements is important. This can be facilitated by weekly feed- to 4 (the most abnormal), providing a daily score of 0 to 24
back sessions.72 In addition, a variety of potential measures points. Sequential assessment of organ dysfunction during
of quality of care for septic patients—vancomycin adminis- the first few days of ICU care is a good indicator of progno-
tration and its timing, steroid administration, blood culture sis. The mean and highest SOFA scores are associated with
collection, activated protein C administration, and broad- outcome. In addition an increase in SOFA score during the
spectrum antibiotic administration and its timing, among first 48 hours in the ICU predicts a mortality rate of at least
50%.74-77 SOFA use has been validated in the NCCU and
traumatic brain injury (TBI).78
Table 17.2  Consensus Definition of SIRS
and Sepsis by the American College
of Chest Physicians and the Society Infection Prevention
of Critical Care Medicine in the Intensive Care Unit
SIRS Two or more of the following criteria: HAIs are common (about 5% of all hospitalized patients), and
—Temperature <36° C or >38° C
—HR >90
in particular catheter-related bloodstream infection, VAP, sur-
—RR >20 or PaCO2 <32 gical site infection, and catheter-associated urinary tract infec-
—WBC >12,000 <4000, or >10% tions increase the cost of care and patient length of stay and
immature (band) forms adversely affect outcome.79-81 In patients with acute neurologic
Sepsis Documented infection together with 2 disorders (e.g., TBI or stroke) or those admitted to the ICU
or more SIRS criteria above the incidence of HAIs is greater—30%.82-84 For example, Wes-
Severe sepsis Sepsis associated with organ dysfunction tendorp et al. in a meta-analysis of 87 studies (8 of which were
restricted to patients admitted to the ICU) that included
Septic shock Sepsis with refractory hypotension or
hypoperfusion abnormalities in spite 137,817 patients after stroke observed that infection compli-
of adequate fluid resuscitation cated 30% of these patients.83 In ICUs today multiresistant
gram-negative bacteria are becoming more frequent as the
Modified from: Bone RC, Balk RA, Cerra FB, et al. Definitions for sepsis and
organ failure and guidelines for the use of innovative therapies in sepsis. The cause of HAIs.85 However, several lines of evidence demon-
ACCP/SCCM Consensus Conference Committee. American College of Chest strate that hand hygiene, isolation, antibiotic control mea-
Physicians/Society of Critical Care Medicine. Chest 1992;101(6):1644–55 sures, and compliance with protocols designed to prevent
HR, Heart rate; RR, respiratory rate; SIRS, Systemic inflammatory response
syndrome; WBC, white blood cell.
specific infections are associated with a reduction in HAIs
including central line–associated bloodstream infections and

Table 17.3  Common Laboratory Findings in Sepsis


Laboratory Test Findings Comments
White blood cell count Leukocytosis or leukopenia Endotoxemia may cause early leukopenia.
Platelet count Thrombocytosis or Early in the disease course a high platelet count may be
thrombocytopenia observed as an acute-phase response.
Low platelet counts found in DIC.
Coagulation Protein C deficiency Coagulation abnormalities may be identified before onset
Antithrombin deficiency of organ failure and without frank bleeding.
Elevated D-dimer level
Prolonged INR (PT) and PTT
Creatinine Increase from baseline Doubling indicates acute renal injury.
Lactic acid >4 mmol/L (36 mg/dL) Consistent with tissue hypoxia
Liver function and Elevated bilirubin, alkaline Hypoperfusion can contribute to acute hepatocellular injury.
enzymes phosphatase, AST, and ALT
Serum phosphate Hypophosphatemia Inversely correlated with proinflammatory cytokine levels
CRP Elevated Acute-phase response
Procalcitonin Elevated May help differentiate infectious from noninfectious SIRS

ALT, Alanine aminotransferase; AST, aspartate transaminase; CRP, C-reactive protein; DIC, disseminated intravascular coagulation; INR, international normalized ratio;
PT, prothrombin time; PTT, partial thromboplastin time; SIRS, systemic inflammatory response syndrome.
162 Section II—Clinical and Laboratory Assessment

Table 17.4  The SOFA (Sepsis-Related Organ Failure Assessment)* Score to Describe Organ
Dysfunction or Failure
Organ  0  1 2 3 4
Respiration †
>400 <400 <300 <200 <100
PaO2/FiO2 (mm Hg) SaO2/FiO2 221-301 142–220 67-141 <67
Coagulation >150 <150 <100 <50 <20
platelets 103/mm3
Liver <1.2 1.2-1.9 2-5.9 6-11.9 >12
bilirubin (mg/dL)
Cardiovascular‡ No hypotension MAP <70 Dopamine ≤5 or Dopamine >5 or Dopamine >15 or
hypotension dobutamine (any) norepinephrine norepinephrine
≤0.1 >0.1
CNS 15 13-14 10-12 6-9 <6
Glasgow Coma score
Renal <1.2 1.2-1.9 2-3.4 3.5-4.9 or <500 >5 or <200
creatinine (mg/dL)
or urine output
(mL/day)

Modified from Vincent JL, Moreno R, Takala J, et al. The SOFA (Sepsis-related Organ Failure Assessment) score to describe organ dysfunction/failure. On behalf of
the Working Group on Sepsis-Related Problems of the European Society of Intensive Care Medicine. Intensive Care Med 1996;22(7):707–10.
*The SOFA is a six-organ dysfunction/failure score measuring multiple organ failure daily. Each organ is graded from 0 (normal) to 4 (the most abnormal), providing
a daily score of 0 to 24 points.

PaO2/FiO2 ratio was used preferentially. If not available, the SaO2/FiO2 ratio was used.

Vasoactive medications administered for at least 1 hr (dopamine and norepinephrine µg/kg/min).
CNS, Central nervous system; FiO2, inspired oxygen fraction; MAP, mean arterial pressure; PaO2, arterial oxygen tension; SaO2, arterial oxygen saturation.

VAP86-92 (see also Chapter 6). The most important aspect of effort to reduce the spread of MRSA within hospitals.100
infection prevention in the ICU including of MRSA and Whether universal screening rather than targeted screening
vancomycin-resistant enterococcus (VRE) is compliance with and expanded use of barrier precautions is cost effective is still
handwashing.93,94 This can be monitored using World Health being elucidated.101-105 However, some form of screening and
Organization hand hygiene observation methods. For routine use of general quality measures is better than no screening to
care alcohol-based gels are easy to use and extremely effective. decrease the incidence of endemic MRSA in ICUs; same-day
However, for patients with C. difficile infections, alcohol gels PCR testing for high-risk patients may provide the greatest
have no activity against the C. difficile spore and careful hand- cost savings.106-108
washing with soap is required. Hands should be washed exten- There are no data or recommendations on the screening of
sively with soap and water for at least 15 seconds, but preferably health care workers for the presence of MRSA nasal carriage.
30 seconds. A strong environmental cleaning program also is In the event that a particular surgeon or surgical team experi-
needed to help reduce the risk of HAIs.95 For example, a previ- ences a higher than anticipated number of epidemiologically
ous room occupant with C. difficile is an independent signifi- related postoperative infections, the hospital infection preven-
cant risk factor for C. difficile acquisition in the ICU.96 Patient tion team should get involved to identify the problem.109 These
visitation does not appear to increase the risk for infection in evaluations often include culturing the team’s nares, axillae,
the ICU,97 whereas conversion to single rooms in the ICU can and groin for MRSA; carefully evaluating the perioperative
reduce the rate at which patients develop infections while in procedures in place including the timing and appropriateness
the ICU.98 of the preoperative antibiotic, and identifying any breach in
Appropriate isolation of infected patients also is required to sterile technique within the operative suite. In the event a
reduce the colonization of health care workers and the spread surgical team member is found to be carrying MRSA, a variety
of these organisms to other patients. All health care workers of antimicrobial agents are available to eliminate carriage.
must wear barriers, including gown and gloves, when they These same principles may be used to examine clustering of
enter the room of a patient colonized or infected with MRSA, infections in the ICU.110
VRE, or C. difficile. These items must be removed before
exiting the room and not reused unless washed in the hospital
laundry. Currently all patients who enter the hospital environ- Health Care Worker–Acquired
ment transferred from another health care institution includ- Infections
ing a rehabilitation facility are screened for nasal carriage for
MRSA and isolated if necessary. This may be important Human Immunodeficiency Virus
because 11% of patients who enter the ICU have a nares The prevalence of HIV in the United States is approximately
sample culture result positive for MRSA.99 This form of active 1 million infected individuals with 20% currently not aware
surveillance remains controversial but has become standard of their status and not in care. The overall rate of HIV trans-
across the United States and other industrialized nations in an mission through percutaneous inoculation (i.e., a needle or
Section II—Clinical and Laboratory Assessment 163

other instrument that pierces the skin) is 0.3%. In the event 9. Qureshi HU, Habib AA, Siddiqui AA, et al. Predictors of mortality in brain
abscess. J Pak Med Assoc 2002;52(3):111–6.
of a sharps or significant splash injury in the ICU, it is impera-
10. Koo DW, Townson, AF, Dvorak MF, et al. Spinal epidural abscess: a 5-year
tive that the health care worker seek immediate attention at case-controlled review of neurologic outcomes after rehabilitation. Arch
the hospital’s employee health department or emergency Phys Med Rehabil 2009;90(3):512–16.
department. Early postexposure prophylaxis of an HIV- 11. Schuchat A, Robinson K, Wenger JD, et al. Bacterial meningitis in the United
infected body fluid with highly active antiretroviral therapy is States in 1995. N Engl J Med 1997;337(14):970–6.
12. Rennick G, Shann F, de Campo J. Cerebral herniation during bacterial
both effective and well tolerated.111 Guidelines for prophylaxis meningitis in children [see comments]. BMJ 1993;306(6883):953–5.
after occupational exposure are available from the Centers for 13. Durand ML, Calderwood SB, Weber DJ, et al. Acute bacterial meningitis in
Disease Control and Prevention (CDC),91 the Department of adults: a review of 493 episodes. N Engl J Med 1993;328:21–8.
Health and Human Services (DHHS), and the World Health 14. Hasbun R, Abrahams J, Jekel J, et al. Computed tomography of the head
before lumbar puncture in adults with suspected meningitis. N Engl J Med
Organization (WHO).112-114
2001;345(24):1727–33.
15. Tunkel AR, Hartman BJ, Kaplan SL, et al. Practice guidelines for the
management of bacterial meningitis. Clin Infect Dis 2004;39(9):1267–84.
Hepatitis B 16. de Gans J, van de Beek D. Dexamethasone in adults with bacterial
Since wide spread mandatory use of the hepatitis B vaccine, meningitis. N Engl J Med 2002;347(20):1549–56.
17. Lakeman FD, Whitley RJ. Diagnosis of herpes simplex encephalitis:
hepatitis B is far less common as an HAI. However, hepatitis application of polymerase chain reaction to cerebrospinal fluid from
C remains a problem, with more than 4 million infected indi- brain-biopsied patients and correlation with disease. National Institute of
viduals in the United States alone. The risk for hepatitis C Allergy and Infectious Diseases Collaborative Antiviral Study Group [see
transmission following a needlestick with contaminated blood comments]. J Infect Dis 1995;171(4):857–63.
18. Tunkel AR, Glaser CA, Bloch KC, et al. The management of encephalitis:
is approximately 3%, but there are currently no effective pro-
clinical practice guidelines by the Infectious Diseases Society of America.
phylactic agents for these injuries or exposure. Care, avoiding Clin Infect Dis 2008;47(3):303–27.
fatigue, and patience in the operating room or ICU are the 19. Whitley RJ, Alford CA, Hirsch MS, et al. Vidarabine versus acyclovir therapy
most effective methods to avoid a sharps injury. in herpes simplex encephalitis. N Engl J Med 1986;314(3):144–9.
20. Provisional surveillance summary of the West Nile virus epidemic: United
States, January-November 2002. MMWR Morb Mortal Wkly Rep 2002;
Conclusion 51(50):1129–33.
21. Division of Vector Borne Infectious Diseases. CDC West Nile virus
Infection is a common reason for admission to the ICU in homepage. 2009 [cited; Available at www.cdc.gov/ncidod/dvbid/westnile/
general and in the NCCU HAIs such as catheter-related blood- index.htm.
22. Acute flaccid paralysis syndrome associated with West Nile virus
stream infections, UTIs, VAP, surgical site infections (includ-
infection: Mississippi and Louisiana, July-August 2002. MMWR Morb
ing those associated with a ventriculostomy), and C. difficile Mortal Wkly Rep 2002;51(37):825–8.
may complicate a third of all admissions. Several factors such 23. Jeha LE, Sila CA, Lederman RJ, et al. West Nile virus infection: a new acute
as poor nutrition and hyperglycemia may increase the risk of paralytic illness. Neurology 2003;61(1):55–9.
an HAI, which when it develops has significant effects on the 24. Leis AA, Stokic DS, Polk JL, et al. A poliomyelitis-like syndrome from West
Nile virus infection. N Engl J Med 2002;347(16):1279–80.
cost of care and may adversely affect patient outcome. Strate- 25. Robinson RL, Shahida S, Madan N, et al. Transient parkinsonism in West
gies such as hand hygiene, environmental cleaning, appropri- Nile virus encephalitis. Am J Med 2003;115(3):252–3.
ate hospital staffing, and education and use of evidence-based 26. Guarner J, Shieh WJ, Hunter S, et al. Clinicopathologic study and laboratory
guidelines among others can reduce NCCU infection rates. diagnosis of 23 cases with West Nile virus encephalomyelitis. Hum Pathol
2004;35(8):983–90.
Infection-control committees can assist in implementing
27. Petropoulou KA, Gordon SM, Prayson RA, et al. West Nile virus
these policies, the success of which requires careful monitor- meningoencephalitis: MR imaging findings. AJNR Am J Neuroradiol
ing of the individual patient and the ICU as a whole.115 2005;26(8):1986–95.
28. Ali M, Safriel Y, Sohi J, et al. West Nile virus infection: MR imaging findings
References in the nervous system. AJNR Am J Neuroradiol 2005;26(2):289–97.
29. Pepperell C, Rau N, Krajden S, et al. West Nile virus infection in 2002:
1. Vincent JL, Rello J, Marshall J, et al. International study of the prevalence morbidity and mortality among patients admitted to hospital in
and outcomes of infection in intensive care units. JAMA 2009;302(21): southcentral Ontario. CMAJ 2003;168(11):1399–405.
2323–9. 30. Makhoul B, Braun E, Herskovitz M, et al. Hyperimmune gammaglobulin for
2. Kirollos RW, Bosma JJ, Radhakrishnan J, et al. Endovascularly treated the treatment of West Nile virus encephalitis. Isr Med Assoc J
cerebral aneurysm using Guglielmi detachable coils acting as a nidus for 2009;11(3):151–3.
brain abscess formation secondary to Salmonella bacteremia: case report. 31. Lewis M, Amsden JR. Successful treatment of West Nile virus infection after
Neurosurgery 2002;51(1):234–7; discussion 237–8. approximately 3 weeks into the disease course. Pharmacotherapy
3. Cavusoglu H, Kaya RA, Turkmenoglu ON, et al. Brain abscess: analysis of 2007;27(3):455–8.
results in a series of 51 patients with a combined surgical and medical 32. Gottfried K, Quinn R, Jones T. Clinical description and follow-up
approach during an 11-year period. Neurosurg Focus 2008;24(6):E9. investigation of human West Nile virus cases. South Med J 2005;98(6):
4. Tewari MK, Sharma RR, Shiv VK, et al. Spectrum of intracranial subdural 603–6.
empyemas in a series of 45 patients: current surgical options and outcome. 33. Beer R, Pfausler B, Schmutzhard E. Infectious intracranial complications in
Neurol India 2004;52(3):346–9. the neuro-ICU patient population. Curr Opin Crit Care 2010;Mar 18. Epub
5. Falcone S, Post MJ. Encephalitis, cerebritis, and brain abscess: ahead of print.
pathophysiology and imaging findings. Neuroimaging Clin N Am 2000; 34. Lundberg N. Continuous recording and control of ventricular fluid pressure
10(2):333–53. in neurosurgical practice. Acta Psychiatr Scand Suppl 1960;36(149):1–193.
6. Ebisu T, Tanaka C, Umeda M, et al. Discrimination of brain abscess from 35. Lozier AP, Sciacca RR, Romagnoli MF, et al. Ventriculostomy-related
necrotic or cystic tumors by diffusion-weighted echo planar imaging. Magn infections: a critical review of the literature. Neurosurgery 2002;51(1):170–
Reson Imaging 1996;14(9):1113–6. 81; discussion 181–2.
7. Cartes-Zumelzu FW, Stavrou I, Castillo M, et al. Diffusion-weighted imaging 36. Coplin WM, Avellino Am, Kim DK, et al. Bacterial meningitis associated
in the assessment of brain abscesses therapy. AJNR Am J Neuroradiol 2004; with lumbar drains: a retrospective cohort study. J Neurol Neurosurg
25(8):1310–7. Psychiatry 1999;67(4):468–73.
8. Renaudin JW, Frazee J. Subdural empyema: importance of early diagnosis. 37. Sundbarg G, Nordstrom CH, Soderstrom S. Complications due to prolonged
Neurosurgery 1980;7(5):477–9. ventricular fluid pressure recording. Br J Neurosurg 1988;2(4):485–95.
164 Section II—Clinical and Laboratory Assessment

38. Arabi Y, Memish ZA, Balkhy HH, et al. Ventriculostomy-associated 46. Pfausler B, Beer R, Engelhardt K, et al. Cell index: a new parameter for the
infections: incidence and risk factors. Am J Infect Control early diagnosis of ventriculostomy (external ventricular drainage)-related
2005;33(3):137–43. ventriculitis in patients with intraventricular hemorrhage? Acta Neurochir
39. Flibotte JJ, Lee KE, Koroshetz WJ, et al. Continuous antibiotic prophylaxis (Wien) 2004;146(5):477–81.
and cerebral spinal fluid infection in patients with intracranial pressure 47. Lopez-Cortes LF, Marquez-Arbizu R, Jimenez-Jimenez LM, et al.
monitors. Neurocrit Care 2004;1(1):61–8. Cerebrospinal fluid tumor necrosis factor-alpha, interleukin-1beta,
40. Rebuck JA, Murry KR, Rhoney DH, et al. Infection related to intracranial interleukin-6, and interleukin-8 as diagnostic markers of cerebrospinal fluid
pressure monitors in adults: analysis of risk factors and antibiotic infection in neurosurgical patients [see comments]. Crit Care Med
prophylaxis. J Neurol Neurosurg Psychiatry 2000;69(3):381–4. 2000;28(1):215–9.
41. Martinez-Manas RM, Santamarta D, de Campos JM, et al. Camino 48. Schade RP, Schinkel J, Roelandse FW, et al. Lack of value of routine analysis
intracranial pressure monitor: prospective study of accuracy and of cerebrospinal fluid for prediction and diagnosis of external drainage-
complications. J Neurol Neurosurg Psychiatry 2000;69(1):82–6. related bacterial meningitis. J Neurosurg 2006;104(1):101–8.
42. Yablon JS, Lantner HJ, McCormack TM, et al. Clinical experience with a 49. Hader WJ, Steinbok P. The value of routine cultures of the cerebrospinal
fiberoptic intracranial pressure monitor. J Clin Monit 1993;9(3):171–5. fluid in patients with external ventricular drains. Neurosurgery
43. Blomstedt GC. Infections in neurosurgery: a retrospective study of 1143 2000;46(5):1149–53; discussion 1153–5.
patients and 1517 operations. Acta Neurochir (Wien) 1985;78(3-4):81–90. 50. Williams TA, Leslie GD, Dobb GJ, et al. Decrease in proven ventriculitis by
44. Korinek AM, Golmard JL, Elcheick A, et al. Risk factors for neurosurgical reducing the frequency of cerebrospinal fluid sampling from extraventricular
site infections after craniotomy: a critical reappraisal of antibiotic drains. J Neurosurg 2011;115(5):1040–6. Epub 2011, Jul 29.
prophylaxis on 4,578 patients. Br J Neurosurg 2005;19(2):155–62.
45. Leib SL, Boscacci R, Gratzl O, et al. Predictive value of cerebrospinal fluid A complete list of references for this chapter can be found online at
(CSF) lactate level versus CSF/blood glucose ratio for the diagnosis of www.expertconsult.com.
bacterial meningitis following neurosurgery. Clin Infect Dis
1999;29(1):69–74.
Section II—Clinical and Laboratory Assessment 164.e1

References 28. Ali M, Safriel Y, Sohi J, et al. West Nile virus infection: MR imaging findings
in the nervous system. AJNR Am J Neuroradiol 2005;26(2):289–97.
1. Vincent JL, Rello J, Marshall J, et al. International study of the prevalence 29. Pepperell C, Rau N, Krajden S, et al. West Nile virus infection in 2002:
and outcomes of infection in intensive care units. JAMA 2009;302(21): morbidity and mortality among patients admitted to hospital in
2323–9. southcentral Ontario. CMAJ 2003;168(11):1399–405.
2. Kirollos RW, Bosma JJ, Radhakrishnan J, et al. Endovascularly treated 30. Makhoul B, Braun E, Herskovitz M, et al. Hyperimmune gammaglobulin
cerebral aneurysm using Guglielmi detachable coils acting as a nidus for for the treatment of West Nile virus encephalitis. Isr Med Assoc J 2009;
brain abscess formation secondary to Salmonella bacteremia: case report. 11(3):151–3.
Neurosurgery 2002;51(1):234–7; discussion 237–8. 31. Lewis M, Amsden JR. Successful treatment of West Nile virus infection after
3. Cavusoglu H, Kaya RA, Turkmenoglu ON, et al. Brain abscess: analysis of approximately 3 weeks into the disease course. Pharmacotherapy
results in a series of 51 patients with a combined surgical and medical 2007;27(3):455–8.
approach during an 11-year period. Neurosurg Focus 2008;24(6):E9. 32. Gottfried K, Quinn R, Jones T. Clinical description and follow-up
4. Tewari MK, Sharma RR, Shiv VK, et al. Spectrum of intracranial subdural investigation of human West Nile virus cases. South Med J 2005;98(6):
empyemas in a series of 45 patients: current surgical options and outcome. 603–6.
Neurol India 2004;52(3):346–9. 33. Beer R, Pfausler B, Schmutzhard E. Infectious intracranial complications in
5. Falcone S, Post MJ. Encephalitis, cerebritis, and brain abscess: the neuro-ICU patient population. Curr Opin Crit Care 2010;Mar 18. Epub
pathophysiology and imaging findings. Neuroimaging Clin N Am 2000; ahead of print.
10(2):333–53. 34. Lundberg N. Continuous recording and control of ventricular fluid
6. Ebisu T, Tanaka C, Umeda M, et al. Discrimination of brain abscess from pressure in neurosurgical practice. Acta Psychiatr Scand Suppl 1960;
necrotic or cystic tumors by diffusion-weighted echo planar imaging. Magn 36(149):1–193.
Reson Imaging 1996;14(9):1113–6. 35. Lozier AP, Sciacca RR, Romagnoli MF, et al. Ventriculostomy-related
7. Cartes-Zumelzu FW, Stavrou I, Castillo M, et al. Diffusion-weighted infections: a critical review of the literature. Neurosurgery 2002;51(1):170–
imaging in the assessment of brain abscesses therapy. AJNR Am J 81; discussion 181–2.
Neuroradiol 2004;25(8):1310–7. 36. Coplin WM, Avellino Am, Kim DK, et al. Bacterial meningitis associated
8. Renaudin JW, Frazee J. Subdural empyema: importance of early diagnosis. with lumbar drains: a retrospective cohort study. J Neurol Neurosurg
Neurosurgery 1980;7(5):477–9. Psychiatry 1999;67(4):468–73.
9. Qureshi HU, Habib AA, Siddiqui AA, et al. Predictors of mortality in brain 37. Sundbarg G, Nordstrom CH, Soderstrom S. Complications due to
abscess. J Pak Med Assoc 2002;52(3):111–6. prolonged ventricular fluid pressure recording. Br J Neurosurg 1988;2(4):
10. Koo DW, Townson, AF, Dvorak MF, et al. Spinal epidural abscess: a 5-year 485–95.
case-controlled review of neurologic outcomes after rehabilitation. Arch 38. Arabi Y, Memish ZA, Balkhy HH, et al. Ventriculostomy-associated
Phys Med Rehabil 2009;90(3):512–16. infections: incidence and risk factors. Am J Infect Control 2005;33(3):
11. Schuchat A, Robinson K, Wenger JD, et al. Bacterial meningitis in the 137–43.
United States in 1995. N Engl J Med 1997;337(14):970–6. 39. Flibotte JJ, Lee KE, Koroshetz WJ, et al. Continuous antibiotic prophylaxis
12. Rennick G, Shann F, de Campo J. Cerebral herniation during bacterial and cerebral spinal fluid infection in patients with intracranial pressure
meningitis in children [see comments]. BMJ 1993;306(6883):953–5. monitors. Neurocrit Care 2004;1(1):61–8.
13. Durand ML, Calderwood SB, Weber DJ, et al. Acute bacterial meningitis in 40. Rebuck JA, Murry KR, Rhoney DH, et al. Infection related to intracranial
adults: a review of 493 episodes. N Engl J Med 1993;328:21–8. pressure monitors in adults: analysis of risk factors and antibiotic
14. Hasbun R, Abrahams J, Jekel J, et al. Computed tomography of the head prophylaxis. J Neurol Neurosurg Psychiatry 2000;69(3):381–4.
before lumbar puncture in adults with suspected meningitis. N Engl J Med 41. Martinez-Manas RM, Santamarta D, de Campos JM, et al. Camino
2001;345(24):1727–33. intracranial pressure monitor: prospective study of accuracy and
15. Tunkel AR, Hartman BJ, Kaplan SL, et al. Practice guidelines for the complications. J Neurol Neurosurg Psychiatry 2000;69(1):82–6.
management of bacterial meningitis. Clin Infect Dis 2004;39(9):1267–84. 42. Yablon JS, Lantner HJ, McCormack TM, et al. Clinical experience with a
16. de Gans J, van de Beek D. Dexamethasone in adults with bacterial fiberoptic intracranial pressure monitor. J Clin Monit 1993;9(3):171–5.
meningitis. N Engl J Med 2002;347(20):1549–56. 43. Blomstedt GC. Infections in neurosurgery: a retrospective study of 1143
17. Lakeman FD, Whitley RJ. Diagnosis of herpes simplex encephalitis: patients and 1517 operations. Acta Neurochir (Wien) 1985;78(3-4):81–90.
application of polymerase chain reaction to cerebrospinal fluid from 44. Korinek AM, Golmard JL, Elcheick A, et al. Risk factors for neurosurgical
brain-biopsied patients and correlation with disease. National Institute of site infections after craniotomy: a critical reappraisal of antibiotic
Allergy and Infectious Diseases Collaborative Antiviral Study Group [see prophylaxis on 4,578 patients. Br J Neurosurg 2005;19(2):155–62.
comments]. J Infect Dis 1995;171(4):857–63. 45. Leib SL, Boscacci R, Gratzl O, et al. Predictive value of cerebrospinal fluid
18. Tunkel AR, Glaser CA, Bloch KC, et al. The management of encephalitis: (CSF) lactate level versus CSF/blood glucose ratio for the diagnosis of
clinical practice guidelines by the Infectious Diseases Society of America. bacterial meningitis following neurosurgery. Clin Infect Dis 1999;29(1):
Clin Infect Dis 2008;47(3):303–27. 69–74.
19. Whitley RJ, Alford CA, Hirsch MS, et al. Vidarabine versus acyclovir 46. Pfausler B, Beer R, Engelhardt K, et al. Cell index: a new parameter for the
therapy in herpes simplex encephalitis. N Engl J Med 1986;314(3): early diagnosis of ventriculostomy (external ventricular drainage)-related
144–9. ventriculitis in patients with intraventricular hemorrhage? Acta Neurochir
20. Provisional surveillance summary of the West Nile virus epidemic: United (Wien) 2004;146(5):477–81.
States, January-November 2002. MMWR Morb Mortal Wkly Rep 2002; 47. Lopez-Cortes LF, Marquez-Arbizu R, Jimenez-Jimenez LM, et al.
51(50):1129–33. Cerebrospinal fluid tumor necrosis factor-alpha, interleukin-1beta,
21. Division of Vector Borne Infectious Diseases. CDC West Nile virus interleukin-6, and interleukin-8 as diagnostic markers of cerebrospinal fluid
homepage. 2009 [cited; Available at www.cdc.gov/ncidod/dvbid/westnile/ infection in neurosurgical patients [see comments]. Crit Care Med
index.htm. 2000;28(1):215–9.
22. Acute flaccid paralysis syndrome associated with West Nile virus 48. Schade RP, Schinkel J, Roelandse FW, et al. Lack of value of routine analysis
infection: Mississippi and Louisiana, July-August 2002. MMWR Morb of cerebrospinal fluid for prediction and diagnosis of external drainage-
Mortal Wkly Rep 2002;51(37):825–8. related bacterial meningitis. J Neurosurg 2006;104(1):101–8.
23. Jeha LE, Sila CA, Lederman RJ, et al. West Nile virus infection: a new acute 49. Hader WJ, Steinbok P. The value of routine cultures of the cerebrospinal
paralytic illness. Neurology 2003;61(1):55–9. fluid in patients with external ventricular drains. Neurosurgery
24. Leis AA, Stokic DS, Polk JL, et al. A poliomyelitis-like syndrome from West 2000;46(5):1149–53; discussion 1153–5.
Nile virus infection. N Engl J Med 2002;347(16):1279–80. 50. Williams TA, Leslie GD, Dobb GJ, et al. Decrease in proven ventriculitis by
25. Robinson RL, Shahida S, Madan N, et al. Transient parkinsonism in West reducing the frequency of cerebrospinal fluid sampling from
Nile virus encephalitis. Am J Med 2003;115(3):252–3. extraventricular drains. J Neurosurg 2011;115(5):1040–6. Epub 2011, Jul 29.
26. Guarner J, Shieh WJ, Hunter S, et al. Clinicopathologic study and 51. Poon WS, Ng S, Wai S. CSF antibiotic prophylaxis for neurosurgical
laboratory diagnosis of 23 cases with West Nile virus encephalomyelitis. patients with ventriculostomy: a randomised study. Acta Neurochir Suppl
Hum Pathol 2004;35(8):983–90. 1998;71:146–8.
27. Petropoulou KA, Gordon SM, Prayson RA, et al. West Nile virus 52. Holloway KL, Barnes T, Choi S, et al. Ventriculostomy infections: the effect
meningoencephalitis: MR imaging findings. AJNR Am J Neuroradiol of monitoring duration and catheter exchange in 584 patients. J Neurosurg
2005;26(8):1986–95. 1996;85(3):419–24.
164.e2 Section II—Clinical and Laboratory Assessment

53. Wong GK, Poon WS, Wai S, et al. Failure of regular external ventricular 77. Jones AE, Trzeciak S, Kline JA. The Sequential Organ Failure Assessment
drain exchange to reduce cerebrospinal fluid infection: result of a score for predicting outcome in patients with severe sepsis and evidence of
randomised controlled trial. J Neurol Neurosurg Psychiatry 2002;73(6): hypoperfusion at the time of emergency department presentation. Crit
759–61. Care Med 2009;37(5):1649–54.
54. Kett DH, Azoulay E, Echeverria PM, et al. Extended Prevalence of Infection 78. Zygun D, Berthiaume L, Laupland K, et al. SOFA is superior to MOD score
in ICU Study (EPIC II) Group of Investigators. Candida bloodstream for the determination of non-neurologic organ dysfunction in patients with
infections in intensive care units: analysis of the extended prevalence of severe traumatic brain injury: a cohort study. Crit Care 2006;10(4):R115.
infection in intensive care unit study. Crit Care Med 2011;39(4):665–70. 79. Roberts RR, Scott RD 2nd, Hota B, et al. Costs attributable to healthcare-
55. Jeon EJ, Jung JW, Choi JC, et al. The value of procalcitonin and the SAPS II acquired infection in hospitalized adults and a comparison of economic
and APACHE III scores in the differentiation of infectious and non- methods. Med Care 2010;48(11):1026–35.
infectious fever in the ICU: a prospective, cohort study. J Korean Med Sci 80. Hassan M, Tuckman HP, Patrick RH, et al. Cost of hospital-acquired
2010;25(11):1633–7. Epub 2010, Oct 26. infection. Hosp Top 2010;88(3):82–9.
56. Ananthakrishnan AN. Clostridium difficile infection: epidemiology, risk 81. Waters HR, Korn R Jr, Colantuoni E, et al. The business case for quality:
factors and management. Nat Rev Gastroenterol Hepatol 2011;8(1):17–26. economic analysis of the Michigan Keystone Patient Safety Program in
Epub 2010, Nov 30. ICUs. Am J Med Qual 2011;26(5):333–9.
57. Freeman J, Bauer MP, Baines SD, et al. The changing epidemiology of 82. Kourbeti IS, Vakis AF, Papadakis JA, et al. Infections in traumatic brain
Clostridium difficile infections. Clin Microbiol Rev 2010;23(3):529–49. injury patients. Clin Microbiol Infect 2011;Jun 30. Epub ahead of print.
58. Bobo LD, Dubberke ER, Kollef M. Clostridium difficile in the ICU: the 83. Westendorp WF, Nederkoorn PJ, Vermeij JD, et al. Post-stroke infection: a
struggle continues. Chest 2011;140(6):1643–53. systematic review and meta-analysis. BMC Neurol 2011;11:110.
59. Oudhuis GJ, Bergmans DC, Dormans T, et al. Probiotics versus antibiotic 84. Cairns S, Reilly J, Booth M. Prevalence of healthcare-associated infection in
decontamination of the digestive tract: infection and mortality. Intensive Scottish intensive care units. J Hosp Infect 2010;76(4):308–10. Epub 2010,
Care Med 2011;37(1):110–7. Epub 2010, Aug 19. Oct 23.
60. Chenoweth CE, Saint S. Urinary tract infections. Infect Dis Clin North Am 85. Doyle JS, Buising KL, Thursky KA, et al. Epidemiology of infections
2011;25(1):103–15. Epub 2010, Dec 18. acquired in intensive care units. Semin Respir Crit Care Med
61. Markovic-Denic L, Mijovic B, Jankovic S. Risk factors for hospital-acquired 2011;32(2):115–38. Epub 2011, Apr 19.
urinary tract infection: a case-control study. Int Urol Nephrol 86. Ramirez P, Bassi GL, Torres A. Measures to prevent nosocomial infections
2011;43(2):303–8. Epub 2010, Sep 17. during mechanical ventilation. Curr Opin Crit Care 2012;18(1):86–92.
62. Chang R, Greene MT, Chenoweth CE, et al. Epidemiology of hospital- 87. Bouadma L, Deslandes E, Lolom I, et al. Long-term impact of a
acquired urinary tract-related bloodstream infection at a university multifaceted prevention program on ventilator-associated pneumonia in a
hospital. Infect Control Hosp Epidemiol 2011;32(11):1127–9. Epub 2011, medical intensive care unit. Clin Infect Dis 2010;51(10):1115–22. Epub
Oct 6. 2010, Oct 11.
63. Knoll BM, Wright D, Ellingson L, et al. Reduction of inappropriate urinary 88. Sihler KC, Chenoweth C, Zalewski C, et al. Catheter-related vs. catheter-
catheter use at a Veterans Affairs hospital through a multifaceted quality associated blood stream infections in the intensive care unit: incidence,
improvement project. Clin Infect Dis 2011;52(11):1283–90. microbiology, and implications. Surg Infect (Larchmt) 2010;11(6):529–34.
64. Levinson AT, Casserly BP, Levy MM. Reducing mortality in severe sepsis 89. Hospenthal DR, Green AD, Crouch HK, et al. Infection prevention and
and septic shock. Semin Respir Crit Care Med 2011;32(2):195–205. Epub control in deployed military medical treatment facilities. J Trauma
2011, Apr 19. 2011;71(2 Suppl 2):S290–8.
65. Angus D, Linde-Zwirble W, Lidicker J, et al. Epidemiology of severe sepsis 90. Cherry-Bukowiec JR, Denchev K, Dickinson S, et al. Prevention of
in the United States: analysis of incidence, outcome, and associated costs of catheter-related blood stream infection: back to basics? Surg Infect
care. Crit Care Med 2001;29:1303–10 (Larchmt) 2011;12(1):27–32. Epub 2010, Dec 20.
66. Bloos F, Marshall JC, Dellinger RP, et al. Multinational, observational study 91. Centers for Disease Control and Prevention (CDC). Vital signs: central
of procalcitonin in ICU patients with pneumonia requiring mechanical line-associated blood stream infections—United States, 2001, 2008, and
ventilation: a multicenter observational study. Crit Care 2011;15(2):R88. 2009. MMWR Morb Mortal Wkly Rep 2011;60(8):243–8.
Epub 2011, Mar 7. 92. Longmate AG, Ellis KS, Boyle L, et al. Elimination of central-venous-
67. Giamarellos-Bourboulis EJ, Tsangaris I, Kanni T, et al. Procalcitonin as an catheter-related bloodstream infections from the intensive care unit. BMJ
early indicator of outcome in sepsis: a prospective observational study. Qual Saf 2011;20(2):174–80.
J Hosp Infect 2011;77(1):58–63. Epub 2010, Dec 4. 93. Lin MY, Hayden MK. Methicillin-resistant Staphylococcus aureus and
68. Lehmann LE, Hunfeld KP, Steinbrucker M, et al. Improved detection of vancomycin-resistant enterococcus: recognition and prevention in intensive
blood stream pathogens by real-time PCR in severe sepsis. Intensive Care care units. Crit Care Med 2010;38(8 Suppl):S335–44.
Med 2010;36(1):49–56. Epub 2009, Sep 15. 94. Monistrol O, Calbo E, Riera M, et al. Impact of a hand hygiene educational
69. Dellinger RP, Levy MM, Carlet JM, et al. Surviving Sepsis Campaign: programme on hospital-acquired infections in medical wards. Clin
international guidelines for management of severe sepsis and septic shock: Microbiol Infect 2011;Nov 22. Epub ahead of print.
2008. Crit Care Med 2008;36(1):296–327. 95. Kleypas Y, McCubbin D, Curnow ES. The role of environmental cleaning in
70. Marshall JC, Dellinger RP, Levy M. The Surviving Sepsis Campaign: a health care-associated infections. Crit Care Nurs Q 2011;34(1):11–7.
history and a perspective. Surg Infect (Larchmt) 2010;11(3):275–81. 96. Shaughnessy MK, Micielli RL, DePestel DD, et al. Evaluation of hospital
71. Levy MM, Dellinger RP, Townsend SR, et al. The Surviving Sepsis room assignment and acquisition of Clostridium difficile infection. Infect
Campaign: results of an international guideline-based performance Control Hosp Epidemiol 2011;32(3):201–6.
improvement program targeting severe sepsis. Intensive Care Med 2010; 97. Adams S, Herrera A 3rd, Miller L, Soto R. Visitation in the intensive care
36(2):222–31. Epub 2010, Jan 13. unit: impact on infection prevention and control. Crit Care Nurs Q 2011;
72. Schramm GE, Kashyap R, Mullon JJ, et al. Septic shock: a multidisciplinary 34(1):3–10.
response team and weekly feedback to clinicians improve the process of 98. Teltsch DY, Hanley J, Loo V, et al. Infection acquisition following intensive
care and mortality. Crit Care Med 2011;39(2):252–8. care unit room privatization. Arch Intern Med 2011;171(1):32–8.
73. Berenholtz SM, Pronovost PJ, Ngo K, et al. Developing quality measures 99. Nair N, Kourbatova E, Poole K, et al. Molecular epidemiology of
for sepsis care in the ICU. Jt Comm J Qual Patient Saf 2007;33(9): methicillin-resistant Staphylococcus aureus (MRSA) among patients
559–68. admitted to adult intensive care units: the STAR*ICU trial. Infect Control
74. Vincent JL, Moreno R, Takala J, et al. The SOFA (Sepsis-related Organ Hosp Epidemiol 2011;32(11):1057–63. Epub 2011, Sep 20.
Failure Assessment) score to describe organ dysfunction/failure. On behalf 100. Prabaker K, Weinstein RA. Trends in antimicrobial resistance in intensive
of the Working Group on Sepsis-Related Problems of the European Society care units in the United States. Curr Opin Crit Care 2011;17(5):472–9.
of Intensive Care Medicine. Intensive Care Med 1996;22(7):707–10. 101. Leonhardt KK, Yakusheva O, Phelan D, et al. Clinical effectiveness and cost
75. Vincent JL, de Mendonça A, Cantraine F, et al. Use of the SOFA score to benefit of universal versus targeted methicillin-resistant Staphylococcus
assess the incidence of organ dysfunction/failure in intensive care units: aureus screening upon admission in hospitals. Infect Control Hosp
results of a multicenter, prospective study. Working group on “sepsis-related Epidemiol 2011;32(8):797–803.
problems” of the European Society of Intensive Care Medicine. Crit Care 102. Kohlenberg A, Schwab F, Behnke M, et al. Screening and control of
Med 1998;26(11):1793–800. methicillin-resistant Staphylococcus aureus in 186 intensive care units:
76. Ferreira FL, Bota DP, Bross A, et al. Serial evaluation of the SOFA score to different situations and individual solutions. Crit Care 2011;15(6):R285.
predict outcome in critically ill patients. JAMA 2001;286(14):1754––8 Epub ahead of print.
Section II—Clinical and Laboratory Assessment 164.e3

103. Huskins WC, Huckabee CM, O’Grady NP, et al. Intervention to reduce in a medical-surgical intensive care unit. Intensive Care Med
transmission of resistant bacteria in intensive care. N Engl J Med 2011; 2010;36(11):1890–8. Epub 2010, Aug 6.
364(15):1407–18. 109. Cook E, Marchaim D, Kaye KS. Building a successful infection prevention
104. Kypraios T, O’Neill PD, Huang SS, et al. Assessing the role of undetected program: key components, processes, and economics. Infect Dis Clin North
colonization and isolation precautions in reducing methicillin-resistant Am 2011;25(1):1–19.
Staphylococcus aureus transmission in intensive care units. BMC Infect Dis 110. Patterson JE, Malani PN, Maragakis LL. Infection control in the intensive
2010;10:29. care unit: progress and challenges in systems and accountability. Crit Care
105. Martinez-Capolino C, Reyes K, Johnson L, et al. Impact of active Med 2010;38(8 Suppl):S265–8.
surveillance on meticillin-resistant Staphylococcus aureus transmission and 111. Raphael J, Landovitz MD, Currier JS. Postexposure prophylaxis for HIV
hospital resource utilisation. J Hosp Infect 2010;74(3):232–7. Epub 2010, infection. N Engl J Med 2009;361:1768–75.
Feb 12. 112. Updated U.S. Public Health Service guidelines for the management of
106. Olchanski N, Mathews C, Fusfeld L, et al. Assessment of the influence of occupational exposures to HIV and recommendations for postexposure
test characteristics on the clinical and cost impacts of methicillin-resistant prophylaxis. Atlanta: Centers for Disease Control and Prevention; 2005.
Staphylococcus aureus screening programs in US hospitals. Infect Control 113. Updated information regarding antiretroviral agents used as HIV
Hosp Epidemiol 2011;32(3):250–7. postexposure prophylaxis for occupational HIV exposures. MMWR Morb
107. Byrnes MC, Adegboyega T, Riggle A, et al. Nasal swabs collected routinely Mortal Wkly Rep 2007;56:1291–2.
to screen for colonization by methicillin-resistant Staphylococcus aureus in 114. Post-exposure prophylaxis to prevent HIV infection: joint WHO/ILO
intensive care units are a sensitive screening test for the organism in clinical guidelines on post-exposure prophylaxis (PEP) to prevent HIV infection.
cultures. Surg Infect (Larchmt) 2010;11(6):511–5. Geneva: World Health Organization; 2007.
108. Ananda-Rajah MR, McBryde ES, Buising KL, et al. The role of general 115. Barsanti MC, Woeltje KF. Infection prevention in the intensive care unit.
quality improvement measures in decreasing the burden of endemic MRSA Infect Dis Clin North Am 2009;23(3):703–25.
Chapter
18  
II

Biomarkers
Robert G. Siman

discordance between laboratory advances and clinical success


Introduction particularly in TBI. Included among the recommendations to
Acute brain injuries triggered by head trauma, intracranial address this is the application of emerging technologies (e.g.,
aneurysm rupture, cardiac arrest, and stroke among others are biomarkers, genomics, and advanced magnetic resonance
an enormous medical, social, and economic problem. Acute imaging [MRI]) to better understand pathophysiology, help
traumatic brain injury (TBI) is the most common cause of classify disease heterogeneity, and examine treatment effects.
death and disability in children and young adults.1 Cardiac Simple diagnostic and prognostic tests sensitive to the onset
arrest kills about 300,000 individuals annually in the United and magnitude of brain damage could have widespread and
States, and even successful resuscitation of out-of-hospital far-reaching applications in the NCCU, and in particular help
cases leads to neurologic dysfunction in the vast majority of identify patients at risk of irreversible brain injury and dys-
instances (www.ctsnet.org/doc/2988). Approximately 800,000 function. In TBI, computed tomography (CT) and MRI can
coronary artery bypass surgeries are performed yearly in the confirm the diagnosis of moderate to severe brain injury.
United States, and it is estimated that as many as 80% are However, CT scanning does not efficiently diagnose mild or
associated with postsurgical impairments in neurologic or repetitive mild TBI, which can lead to neurodegeneration and
cognitive function.2,3 More than 750,000 strokes occur in the neurologic dysfunction, and MRI has suboptimal sensitivity.5
U.S. annually, and treatment with clot-dissolving drugs is Similarly, the impact of mild stroke or coronary bypass surgery
indicated for only a small proportion of cases.4 About 30,000 remains difficult to quantify with routine neuroimaging
cases of aneursymal subarachnoid hemorrhage (SAH) occur methods.6 Prognostic tests that define patients at high risk for
per year, and severe aneurysmal SAH carries a high risk of death or disability and sensitive tests to detect secondary inju-
brain damage and neurologic dysfunction, especially when ries also could guide the direction and aggressiveness of patient
accompanied by cerebral vasospasm. care. There is a variable risk for ischemic brain damage after
Treatments conducted in the neurocritical care unit cardiac arrest, and currently there are no methods to identify
(NCCU) and emergency department are directed at the the subset of patients susceptible to irreversible brain injury
pathophysiologic processes that occur with acute brain inju- that would benefit most from neuroprotective treatment. The
ries. Progress is being made, in part through the use of neu- significance of this shortcoming is heightened with the success
romonitoring and neuroimaging to better understand these of induced hypothermia after cardiac arrest. Although this
pathophysiologic processes, identify deleterious risk factors, helps preserve short- and long-term brain function, the treat-
improve preservation of brain tissue, and reduce mortality, ment is complex, expensive to administer, efficacious for only
morbidity, and long-term brain dysfunction. However, it has a subset of resuscitated patients, and dependent on treatment
been difficult to assess in a rigorous fashion the relative merits variables that have yet to be optimized.
of the many treatment options available in the NCCU, and Following severe SAH or TBI, monitoring often generates
considerable strides still need to be made to identify patients data about the onset, severity, and location of secondary
at risk for irreversible brain damage, detect early and poten- pathologic events through measures of blood flow, energy
tially treatable stages before the onset of irreversible injury, metabolism, local oxygen partial pressure, and electroenceph-
rapidly identify patients who most benefit from specific treat- alographic activity. However, there are no direct indices or
ment measures to reduce subsequent neurodegeneration and surrogate measures for the onset and magnitude of irrevers-
brain dysfunction, and optimize treatment parameters for ible brain damage. There are many treatment interventions
reducing brain damage and improving outcome. Further- that still need to be evaluated fully or optimized in controlled
more, despite 25 years of extensive basic and clinical research clinical trials, for example, the relative merits of mannitol or
into intraneuronal signaling mechanisms for neuron death hyperosmotic saline; determining the optimal osmolarity or
and pharmacologic strategies for neuroprotection, there still target pressures when using hyperosmolar therapy; the role of
are no U.S. Food and Drug Administration (FDA)–approved induced normothermia; finding the optimal glucose or hemo-
neuroprotective therapies for any form of acute brain injury. globin; different ventilation strategies; and how best to prevent
Recent multidisciplinary workshops (e.g., Classification of venous thrombosis, among many others that have to be
TBI for Targeted Therapies, October 2007; Common Data answered in the NCCU. These questions can be difficult to
Elements workshop for Research and Psychological Health study in a controlled trial, in part because of the sample size
in TBI, March 2009, Comparative Effectiveness Research in needed, the complexity of patient care, and the insensitivity
TBI, June 2010) have identified important reasons for the and imprecision of commonly used functional outcome
© Copyright 2013 Elsevier Inc. All rights reserved. 165
166 Section II—Clinical and Laboratory Assessment

measures. Surrogate markers for irreversible acute brain Project and the growth of microarrays, proteomics, and
damage could play a major role in helping to answer these nanotechnology provides new methods to develop sensitive
questions in the NCCU. The clinical evaluation of potential high-throughput assays. Second, advances in bioinformatics
neuroprotectant drugs also often requires large and expensive and cross-disciplinary collaborations have enhanced the
trials that can present a barrier for pharma­ceutical and bio- ability to retrieve and analyze large quantities of data. Third,
technology companies with promising but unproven experi- there now is understanding that diseases develop from the
mental therapeutics. Surrogate measures for efficacy that dynamic dysregulation of several gene regulatory networks,
reduce the complexity and cost while increasing the discrimi- proteins, and metabolic alterations that reflect complex per-
native power of early-stage drug trials would facilitate clinical turbations (genetic and environmental) of the “biologic
research for novel neuroprotectants and help drug discovery system.” Best practice guidelines to standardize the quality and
and development.7 In addition, use of state-of-the-art bio- accessibility of biosamples and so realize the full potential and
marker platforms may help during the recovery phase of any accelerate genomic, proteomic, and metabolomic research in
acute brain injury to identify effective therapies to enhance acute brain injury have been published.10 It is beyond the
recovery in a targeted and individualized fashion.8 This scope of this chapter to review the many technical, financial,
concept may apply not only to strategies used in rehabilitation legal, and regulatory hurdles that have to be overcome to
after moderate or severe TBI but also to managing the cogni- develop clinically acceptable and validated biomarkers that are
tive or neurobehavioral sequelae even after mild injury with commercially produced and FDA proved.11
techniques such as evaluation of micro-ribonucleic acid To evaluate patients with potential acute brain injuries, the
(RNA) species in peripheral blood mononuclear cells.9 measurement of biochemical markers from a CSF source are
There are many potential uses for surrogate markers for useful in many settings, whereas serum measures would have
brain damage in preclinical studies. This is especially true for broader utility. Studies also suggest that use of microdialysis
markers that relate directly and mechanistically to the under- (i.e., from brain interstitial fluid) to recover some biomarkers
lying molecular pathways for neurodegeneration. Rather than may be feasible.12-14 An ideal CSF or serum marker should have
relying solely on neurologic tests that reflect in complex and several characteristics,15,16 including: (1) high degrees of sen-
often indirect ways the anatomy, pathology and physiology of sitivity and specificity for brain damage, (2) appearance only
brain damage, or histopathologic methods that restrict analy- after irreversible degeneration of brain tissue, (3) rapid transit
ses to the time of sacrifice, it may be possible to examine to the bloodstream in the case of serum markers, and (4) cor-
mechanisms of neurodegeneration and assess their temporal relations with the time course and extent of brain injury.
characteristics by serial sampling in living animals using sur- There should be low variability in the measurement of an ideal
rogate markers. These markers also could help identify optimal marker to guarantee a predictable relationship between its
dosing, evaluate specific neurodegenerative mechanisms, or concentration in CSF, serum, or urine and the degree of brain
define efficacy of neuroprotective therapeutics over time in injury. The ideal marker should provide information that is
living animals. Furthermore, a surrogate marker with the sen- unavailable from other clinical data alone, for example, rapid
sitivity to detect mild brain damage may facilitate mechanistic diagnosis of at-risk patients, and levels should be responsive
and experimental therapeutic studies of both single and repet- to treatment interventions that improve brain function,
itive mild insults, and of factors that regulate the threshold for thereby providing a pharmacodynamic measure of irreversible
brain damage. Finally, comparative analyses of a panel of brain damage. Finally, there should be a reliable and facile
potential surrogate markers for brain damage in human clini- assay for the ideal marker with the simplicity, speed, and low
cal samples and preclinical experimental models should cost needed to quickly inform and direct patient care and
provide new information about the clinical relevance of achieve widespread and routine application.
various animal model systems, none of which have been vali-
dated pharmacologically because of the lack of clinically
proven neuroprotective drug treatments. Current Status of
Surrogate Markers
Desirable Characteristics of a A surrogate measure for brain damage that meets the stringent
requirements of the ideal marker has not emerged yet.
Surrogate Marker for Brain Damage However, several biochemical markers have demonstrated
Biomarker (biologic marker) was introduced in 1989 as a promise in preclinical and clinical studies and so raise the
Medical Subject Heading (MeSH) term. In 2001 a National possibility that a simple cerebrospinal fluid, blood, or urine
Institutes of Health (NIH) working group standardized the test for acute brain damage is feasible. Examples of clinical
definition of a biomarker as “a characteristic that is objectively studies are summarized in Table 18.1. Conceptually, proteins
measured and evaluated as an indicator of normal biologic that are expressed primarily in the nervous system could
processes, pathogenic processes, or pharmacologic responses appear outside the central nervous system (CNS) preferen-
to a therapeutic intervention.” The working group also tially under pathologic neurodegenerative conditions that are
described different types of biomarkers including that mea- accompanied by disruption of the blood-brain and blood-
sured on a biosample (e.g., blood, urine, cerebrospinal fluid cerebrospinal fluid barriers. Consistent with this concept, it is
[CSF], or tissue), a recording obtained from a person (e.g., well-established that TBI, ischemia, stroke, or SAH among
blood pressure, electrocardiogram [ECG]), or an imaging others cause localized, reversible breakdown of the blood-
study (e.g., MRI or CT scan). This chapter focuses on biosam­ brain barrier that, although transient, can persist for several
ples. Several recent developments have stimulated the bio- hours or occur even days following the insult.17,18 In addition,
marker field: first, the completion of the Human Genome certain nervous system–enriched proteins are detectable in
Section II—Clinical and Laboratory Assessment 167

Table 18.1  Examples of Clinical Studies of Surrogate Markers for Acute Brain Damage
Biomarkers Source Key Finding Reference
TRAUMATIC BRAIN INJURY
S100β, NSE Serum Prognostic for poor neurologic outcome 22
Tau CSF Prognostic for long-term outcome 120
α-Spectrin CSF Related to CT findings, long-term outcome 75
Cleaved tau CSF Prognostic for elevated intracranial pressure and long-term outcome 58
SUBARACHNOID HEMORRHAGE
NFL CSF Prognostic for long-term outcome 70
S100β, GFAP Serum Associated with hemorrhage type, neurologic dysfunction, intracranial pressure 50
pNFH CSF, serum Associated with vasospasm 67
CARDIAC ARREST WITH RESUSCITATION
S100β Serum Prognostic for mortality 24
S100β, NSE Serum Prognostic for mortality (both) and memory dysfunction (S100β) 121
STROKE
S100β, GFAP CSF Predictive of infarct volume 25
S100β Serum Prognostic for long-term neurologic outcome and related to infarct volume 122
Tau CSF Prognostic for long-term neurologic outcome and related to infarct volume 123

CSF, Cerebrospinal fluid; CT, computed tomography; GFAP, glial fibrillary acidic protein; NFL, neurofilament light chain; NSE, neuron-specific enolase;
pNFH, hypophospho-neurofilament H.

human CSF and serum following brain injury or in associa- analysis models that include mass lesion, pupils, S100β
tion with chronic neurodegenerative disease. So far the devel- and another astrocyte marker, glial fibrillary acidic protein
opment of clinically useful surrogate markers has been most (GFAP), best predict death and unfavorable outcome.29
successful in the prion disorder Creutzfeldt-Jakob disease, for In SAH, admission serum level of S100β is associated with
which CSF measurement of the brain-enriched 14-3-3 protein short-term survival, injury severity as assessed by neuroimag-
family is accepted into clinical practice as a diagnostic marker ing, and poor outcome at 1 year, whereas therapeutic inter­
for the disorder.19,20 vention with external ventricular drainage is associated with
reduced serum S100β level.38
However, not all studies have found that use of S100β is
Glial Proteins reliable to discriminate severity of injury or prognosis.39-41
Several limitations also are associated with S100β as a bio-
S100β marker for acute brain injury. First, high serum concentra-
Several non-neuronal proteins whose expression either is tions of S100β are observed in association with chronic
brain enriched or induced in the brain in response to neuronal diseases unrelated to acute brain damage42,43; this can occur in
injury have been evaluated in preclinical and clinical studies humans even in the absence of brain injury and may be
as potential surrogate markers for acute brain damage. The released after injury by tissue other than the brain.44,45 Second,
most extensively studied is S100β, an astrocyte-enriched not all types of brain damage are associated with measurable
protein. S100β expression is increased with the astroglial increases in serum concentration of S100β46 Third, S100β
hypertrophic response to brain injury,21 and increases are elevations are not always predictive of long-term neurologic
detectable based on two-antibody sandwich immunoassay in dysfunction47 and have shown inconsistent prognostic value
CSF and serum of patients following TBI, SAH, cardiopulmo- in mild forms of acute brain damage.48 In children, these
nary bypass surgery, cardiac arrest with resuscitation, carbon inconsistencies may be associated with patient age.34 Fourth,
monoxide poisoning, and ischemic or hemorrhagic stroke.22-33 the data that demonstrate a positive relationship between
Many of these studies demonstrate a prognostic relationship serum and CSF levels of S100β and patient prognosis have
between serum or CSF level of S100β and neurologic outcome, been largely derived from retrospective analysis, that is, cutoff
mortality, or secondary cerebral insults (e.g., elevated intra- biomarker levels that relate to distinct outcomes all have been
cranial pressure),28 although there is no consensus on cutoff selected from observing the data post-hoc. Consequently,
values or time of sampling. Furthermore, there is a suggestion multiple, reproducible studies have yet to establish definitively
that S100β elevation may be an indicator of impending dete- the clinical utility of S100β as a diagnostic or prognostic
rioration, for example, an increase in intracranial pressure measure of acute brain injuries, or as a pharmacodynamic tool
(ICP)28 or of when to obtain a head CT scan in mild TBI.34-36 to measure treatment effectiveness. Finally, methodologic
In severe TBI, maximal serum levels of S100β are associated considerations add variance to the clinical measure of S100β
with poor long-term function assessed by the Glasgow Coma that will require both independent confirmation and inter-
Scale,37 and some studies suggest that after multivariable laboratory standardization to promote the prognostic utility
168 Section II—Clinical and Laboratory Assessment

of the biomarker.49 In summary, although certainly worthy of results, however, are mixed. In some studies the magnitude of
study, S100β is not yet established as either a specific and serum NSE elevation is associated with long-term brain func-
broadly applicable surrogate marker for acute brain damage, tional outcome following cardiopulmonary resuscitation.60,61
or a highly reproducible and consistent index of short-term However, the cutoff serum NSE level for maximal prognostic
and long-term prognosis that may be used in a defined sensitivity and specificity was selected in a post-hoc fashion.
clinical application in a prospective fashion across multiple Thus it remains to be determined whether serum NSE mea-
laboratories. sures have sufficient reliability and robustness for prospective
applications in routine clinical practice. In severe pediatric
TBI, serum NSE changes have a poor association with
Other Glial Proteins outcome in children less than 4 years old or in victims of
There have been fewer studies on other glial-specific proteins inflicted TBI.62 In severe adult TBI, both NSE and S100β
that show promise as candidate biomarkers for acute brain serum levels correlated with contusion volumes or episodes
injuries; these include the astroglial GFAP and the oligoden- of reduced cerebral perfusion pressure (CPP), but their
droglial myelin basic protein (MBP). In SAH, serum levels of respective release patterns over time differ for cases of primary
GFAP on hospital arrival are associated with scales of neuro- cortical contusion, diffuse axonal injury, and cerebral edema
logic dysfunction and CT Fisher grade scores, and increase in without focal mass lesions.28,63 Increases in NSE have been
patients with rebleeding or ischemic events.50,51 Serum GFAP observed in boxers with predominantly blows to the head
levels at admission are associated with abnormal head CT rather than the body64 and after an extended resting period
findings, mortality, and poor long-term outcome following that suggests repetitive head trauma results in sustained
severe TBI; 20- to 30-fold increases are observed in patients release of this brain-specific protein to the peripheral circula-
with poor outcome.29,52,53 In pediatric TBI, initial serum MBP tion.65 In SAH, serum NSE levels are not always elevated.50
and S100β levels correlate with poor outcome, although the Consequently, whereas NSE has received increased attention
relationship is stronger for children 4 years or younger.54 as a marker for brain damage associated with cardiac arrest
Serum and CSF GFAP levels also are elevated in the severe and resuscitation, its use in other acute brain injuries appears
pediatric TBI.55 Although this elevation is associated with thus far to be limited.
Pediatric Cerebral Performance Category scores at 6 months,
the GFAP increase does not discriminate admission injury
severity or differentiate therapeutic strategies. In children who Neurofilaments
receive extracorporeal membrane oxygenation, peak GFAP Neurofilaments are the intermediate filaments of neurons,
levels are higher in children with brain injury than those and because of their unique protein composition the constitu-
without.56 It remains uncertain, however, whether these glial ent neurofilament triplet polypeptides neurofilament heavy
markers can guide therapy. chain (NFH), neurofilament middle chain (NFM), and neu-
rofilament light chain (NFL) have been evaluated as markers
for acute brain damage in several studies. In SAH, CSF levels
Neuronal Proteins of a particular phosphoform of NFH are increased in patients
Several nervous system–enriched proteins expressed predomi- with poor 3-month outcome,66 whereas serum NFH phospho-
nantly or exclusively in neurons have been studied as potential form levels increase when there is cerebral vasospasm.67 A
surrogate markers for acute brain injuries, including tau, particular hypophosphorylated form of NFH is expressed in
neuron-specific enolase (NSE), neurofilament polypeptides, neurons predominantly within axons, and CSF alterations in
and α-spectrin (see Table 18.1). this phosphoform have been proposed as an index of axonal
damage.66 CSF NFL levels also have been shown to increase
following severe TBI or SAH68 and reportedly correlate with
Tau long-term outcome following both cardiac arrest and resusci-
The microtubule-associated protein tau is increased in CSF tation69 and SAH.70 However, other studies suggest that
and serum of severely head-injured patients primarily as tau although CSF NFL concentrations are elevated in SAH, espe-
proteolytic fragments of approximately 30 to 50 kDa of inde- cially those with acute ischemia, there is no association with
terminate sequence rather than the intact protein and may outcome suggesting that neurofilament as a biomarker may be
serve as indicators of outcome.57-59 Zemlan and colleagues better suited to TBI when there is axonal injury.71 Consistent
developed a monoclonal antibody preferentially reactive with with this, the level of neurofilament protein heavy chain in
these unidentified tau fragments and established a two- serum is not associated with brain ischemia on routine brain
antibody immunoassay to provide evidence for measurable imaging techniques after carotid endarectomy.72 Proteolytic
elevations in serum tau concentrations in association with breakdown products of neurofilament heavy chains can be
human TBI.57 However, serum-cleaved tau was not detected recovered using microdialysis, and these brain extracellular
in several head-injury cases, and without an unambiguous fluid markers might be of prognostic value.13,21
characterization of these tau fragments, it is difficult to relate
their appearance in body fluids to mechanisms that underlie
neurodegeneration. Spectrin
Another cytoskeletal protein that is expressed in neurons and
shows promise as a systemic marker for acute brain damage
Neuron-Specific Enolase is the actin-binding protein spectrin. The alpha subunit of the
NSE is another neuronal protein that has been studied exten- nonerythroid form of spectrin, referred to as αII, although
sively as a surrogate marker for acute brain damage. The expressed in a diverse number of cell types, is expressed at high
Section II—Clinical and Laboratory Assessment 169

specifically with preferred caspase recognition motifs. Cleav-


levels in neurons.73 Levels of αII-spectrin and spectrin break- age site-specific antibodies reactive with caspase-derived
down products (SBDP145 produced by calpain, and SBDP120 proteolytic fragments of lamin A, αII-spectrin, polyADP-
produced by caspase-3)74 increase in CSF following severe ribose polymerase, actin, and other caspase substrates have
TBI,75 SAH,76 and cardiac arrest with resuscitation,77 and the been useful tissue markers for caspase-mediated apoptosis.85-87
increases are associated with various measures of patient Thus far the only caspase substrates evaluated as potential
outcome. In addition is a correlation between patients with CSF and serum markers for acute brain injury have been
longer ICP elevations and CSF αII-SBDPs.78 The specific the caspase-1 products interleukin-1β and interleukin-18.88,89
nature of these breakdown products and their relevance to the However, specificity of these proinflammatory cytokines as
pathophysiologic processes that underlie acute neurodegen- markers for brain damage has been limited by their abundant
eration are described further in the following section. expression in hematopoietic cells that mediate diverse inflam-
In summary, there are a number of glial- and neuron- matory processes throughout the body. Recent clinical studies
enriched proteins and markers from brain endothelial cells however suggest serum interleukin-1β and to a lesser extent
that have shown promise as diagnostic or prognostic indica- tumor necrosis factor alpha (TNFα) may show promise as
tors of acute brain damage deserving of further study, but markers of poor outcome or intracranial hypertension after
unfortunately none has emerged yet as a validated and reliable TBI.90 The identification of additional caspase substrate pro-
prospective marker with routine clinical applicability.79,80 In teins that are expressed at high levels predominantly in the
addition, whether a combination of markers is more valuable brain, degraded during apoptotic neurodegeneration in asso-
than single markers is still to be validated.81,82 Moreover, the ciation with acute brain damage, and leaked into the CSF or
CSF and serum elevations in the most widely studied bio- blood in an injury-specific manner could lead to the identifi-
markers have yet to be related to underlying pathophysiologic cation of a mechanism-based surrogate marker for the apop-
brain processes or brain regions and neural pathways being totic component of acute brain injury.
impacted, and with the exception of a single NFH phospho- Activation of the calpain family of calcium-activated cyste-
form, to neuronal subcellular elements that may be preferen- ine proteases has been linked in numerous studies to necrotic
tially affected. neurodegeneration.91-94 Calpains are inactive at normal, physi-
ologic intraneuronal free calcium concentrations, but are acti-
vated by the intraneuronal calcium overload that accompanies
excitotoxicity; global and focal cerebral ischemia; traumatic
Proteolytic Mechanisms for Acute injury to the brain, spinal cord, and peripheral nerves; hypoxia-
Neurodegeneration That Could hypoglycemia; status epilepticus; oxidative and nitrative stress;
Be Exploited to Develop and mitochondrial respiratory failure. Calpains also are acti-
vated in some instances of apoptosis, and very brief episodes
Surrogate Markers of calpain activation can occur without neurodegeneration.
The biochemical signaling pathways that underlie neurode- However, strong and sustained calpain activation without an
generation offer the prospect to develop mechanism-based accompanying caspase activation is diagnostic for neuronal
markers for brain damage potentially measurable in accessible necrosis.95-97 One of the most abundant high-affinity calpain
body fluids. In particular, apoptotic and necrotic neurodegen- substrate proteins is the alpha subunit of spectrin, and cleav-
erative signaling are characterized by the activation of distinct age site–specific antibodies reactive with calpain-derived frag-
families of cysteine proteases, and the proteolytic “finger- ments of α-spectrin, but not with caspase-derived fragments
prints” of degraded protein substrates for these cell death– or the spectrin holoprotein, have been used widely as cell
activated proteases have been used widely as tissue markers and tissue markers for calpain activity. They have linked sus-
for experimental neurodegeneration. Most modes of apop- tained calpain activation with the necrotic neurodegeneration
totic neuronal death are triggered by the activation of cas- that accompanies TBI, global and focal cerebral ischemia,
pases, cysteine proteases with the unusual specificity for and hypoxia-hypoglycemia.87,95,98-101 Similar to the caspase-
cleavage of protein substrates on the carboxyl side of aspartate derived protein fragments, the immunodetection of calpain
residues. Caspase activation and neuronal apoptosis have been substrate protein fragments that are generated from brain-
demonstrated as contributing factors in numerous studies of enriched proteins during neuronal necrosis and enter the CSF
acute brain damage after TBI, global and focal cerebral isch- or bloodstream with acute brain damage could be valuable
emia, and hypoxia-hypoglycemia.83,84 Eleven human caspases mechanism-based surrogate markers for the necrotic compo-
have been identified, at least 8 of which are expressed in the nent of acute brain injuries.
brain and may be activated during apoptosis. These proteases
are synthesized as inactive zymogens and require proteolytic
processing and oligomeric assembly for their activation. Con-
sequently, cleavage site-specific antibodies have been prepared Novel Approaches to Identify
that react preferentially with activated, processed subunits of Potential Surrogate Markers
many of the caspases, and these have facilitated the biochemi-
cal quantitation and histochemical localization of the activa-
for Brain Damage
tion of caspases 3, 6, 8, and 9 that occur during acute brain Enormous strides have been made with proteomics methods
damage. Furthermore, the substrate specificity of caspases is to identify proteins in complex biologic samples, and these
determined primarily by short domains of four or five amino methods now are being applied to define CSF and serum
acids that end in an aspartate residue, making it possible to proteomes and analyses of changes in the levels of specific
design and prepare cleavage site-specific antibodies that react proteins in association with disease states. Protein separation
170 Section II—Clinical and Laboratory Assessment

and identification are performed by either two-dimensional well-established proteomics methods of two-dimensional
polyacrylamide gel electrophoresis (2D PAGE), liquid chro- electrophoresis and mass spectrometry. It identified 14 of the
matography (LC), mass spectrometry (MS), or combinations most abundant polypeptides released preferentially during
of 2D PAGE, LC, and MS.102,103 Although it may be theoreti- neuronal necrosis.97 Seven are known to be expressed pri-
cally possible to measure a protein within protein-rich serum marily in the brain, and are present in the adult CNS: 14-3-
whose levels change in relation to the time course and extent 3β and ζ isoforms, ubiquitin C-terminal hydrolase L1
of brain damage, no such marker protein has been identified (UCH-L1), collapsin response mediator proteins 2 and 4
yet by proteomics methods. The sensitivity of detection by 2D (CRMP-2 and CRMP-4), GAP-43 and creatine kinase B.106-110
PAGE or MS approaches may need to improve to detect the Because these 7 proteins are brain-enriched proteins released
expected minute (attomolar [10–18 molar]) quantity of brain- in response to neurodegeneration, they are candidate surro-
enriched proteins that may circulate in the blood post injury. gate markers for brain injury potentially measurable in acces-
The complexity of methods for MS and their considerable sible body fluids. A second set of candidate markers was
expense are further barriers to their adoption into routine identified by examining abundant cytoskeletal proteins that
clinical practice. Although proteomics approaches to identify are expressed predominantly or exclusively in neurons. This
and quantitate surrogate markers for acute brain damage have approach demonstrated that dying neurons release several
promise, it remains to be seen whether they offer sufficient intact structural proteins and numerous proteolytic frag-
simplicity, sensitivity, and specificity to identify and quantify ments, including NFH, the microtubule-associated proteins 2
surrogate markers for brain damage that will have broad clini- (MAP2) and tau, the α-subunit of the actin-binding protein
cal applicability. spectrin, calpain-derived fragments of α-spectrin and tau
Unlike proteomics methods, immunoassays may offer the and caspase derivatives of α-spectrin and the nuclear matrix
simplicity, sensitivity, and specificity that would be required protein lamin A.
of a clinically useful surrogate marker for brain damage. To address whether proteins and their proteolyic frag-
Rather than evaluating target antigens one by one as potential ments released from degenerating cultured neurons may be
bases for a surrogate measure for brain damage in accessible markers for brain damage, CSF and serum samples were
body fluids, as has been conducted so far, a biologically based evaluated from rats subjected to lateral fluid-percussion TBI
approach to identify numerous potential surrogate markers or transient global cerebral ischemia. It was found that two
could expedite the development and validation of a clinically sets of cytoskeletal proteolytic fragments and several intact
useful immunoassay. The Siman laboratory initiated a global, proteins increase markedly in CSF following experimental
systematic analysis of protein release by degenerating neurons TBI in the rat.97,104 Tau is detectable as five calpain deriva-
maintained in primary culture to establish a neurobiologic tives that co-migrate with tau fragments released during
approach to identify new potential surrogate markers.97,104,105 necrotic neurodegeneration in vitro. Additionally, α-spectrin
The value of this neurobiologic approach is supported by the fragments of 120 to 150 kDa and intact 14-3-3 protein iso-
finding that proteins released preferentially by degenerating forms β and ζ and NFH also increase in rat CSF following
cultured neurons also are increased markedly in CSF and TBI. Cleavage site-specific antibodies reactive with calpain-
serum following TBI or cerebral ischemia in rodent experi- derived α-spectrin fragments confirm the appearance of
mental models (see following section). This neurobiologic approximately 150 and 145 kDa calpain derivatives of spec-
approach can identify new markers and provide immunoas- trin. The additional set of α-spectrin derivatives of approxi-
says for them with the potential to aid in diagnosis, prognosis, mately 120 kDa that also increase after brain injury, albeit to
and therapeutic evaluation of acute brain damage. a lesser degree, are similar in size to an α-spectrin fragment
The identification of a large number of novel candidate formed by caspase proteolysis93,111 and likely are caspase
markers for acute brain injuries would foster development of derived. The biomarker results are consistent with histopa-
a panel of markers that in turn can be expected to have diag- thology studies that indicate TBI triggers brain cell death
nostic and prognostic sensitivity and specificity beyond that both by necrosis and apoptosis, with necrosis predominating
of individual markers. Indeed, clinical practices in oncology under most circumstances.100,112,113 These findings demon-
and cardiology have benefited considerably from the avail- strate that several proteins released from dying cultured
ability of multiple markers to classify tumors and detect and neurons increase in an accessible body fluid after experi-
characterize myocardial infarction. In addition, a neurobio- mental TBI, and suggest that different pathophysiologic
logic approach to identify surrogate markers for brain damage mechanisms that contribute to brain damage may be distin-
based on underlying proteolytic pathways for necrotic and guishable in living animals by mechanistically distinct bio-
apoptotic neurodegeneration would be valuable to define chemical markers for neurodegeneration.
further the biochemical mechanisms for brain damage both To determine whether the injury-induced increases in intact
in patients and preclinical experimental models. neuronal proteins and proteolytic fragments reflect general-
ized changes in CSF protein levels, by 2D PAGE and silver
staining the proteome for rat CSF proteins following moder-
Neuron-Enriched Proteins Released ate TBI was examined. The 2D patterns of the most abundant
During Neurodegeneration Are rat CSF polypeptides were similar with or without injury.96 It
Markers for Acute Brain Damage was concluded that the TBI-induced elevations in CSF levels
of the neuron-enriched proteins detectable by immunoassay
in Experimental Models are not accompanied by generalized changes in the levels of
As a first step to develop and validate a panel of biomarkers major CSF proteins. The results highlight the sensitivity of
for acute brain damage, the neurobiologic approach men- immunoassay methods to detect brain injury–induced altera-
tioned earlier examined the most prevalent polypeptides tions in neuronal polypeptides present in relatively low abun-
released from degenerating cultured cortical neurons using dance in an accessible body fluid.
Section II—Clinical and Laboratory Assessment 171

Surrogate markers could have important clinical uses to


Proteins Released from identify patients at risk for acute brain injury, stratify patients
Degenerating Neurons as based on an early indicator of the severity of acute brain
Pharmacodynamic Markers for damage, and provide relatively fast, simple, and inexpensive
assessment of candidate neuroprotectant treatments in early
Experimental Acute Brain Damage stage trials. To investigate whether proteins released from
A surrogate marker for acute brain damage that has preclinical degenerating neurons may be pharmacodynamic markers that
and clinical use should be sensitive to the injury magnitude. are responsive to neuroprotectant therapy, 14-3-3 isoform
“Mild” TBI in the rat resembles mild human TBI because levels were studied in CSF of rats subjected to moderate TBI
there is relatively sparse neurodegeneration and injury is and treated with magnesium that is a known neuroprotectant
without accompanying mortality, apnea, or seizures. Even in the rat.114-117 As shown on the right of Figure 18.2, magne-
mild TBI in the rat leads to increased CSF levels of 14-3-3β, sium treatment significantly attenuated the TBI-induced
14-3-3ζ, calpain-cleaved tau, NFH, UCH-L1 and calpain- increase in 14-3-3β by 30% and the increase in 14-3-3ζ by
cleaved α-spectrin (Fig. 18.1). At all time points examined, 37%. The reduced marker levels were observed at 6 hours post
CSF concentrations of the six markers are greater following injury. This early attenuation of biomarker increases occurs
moderate TBI than mild TBI. well before the full temporal development of injury-induced
To assess whether CSF levels of proteins released from histopathology, and supports the potential of diagnostic,
degenerating neurons could serve as markers for ischemic prognostic, and therapeutic applications of surrogate markers
brain injury and reflect its severity, CSF was examined 48 in the clinic.
hours after an episode of forebrain ischemia varying from 3
to 10 minutes.88,104 Levels of 14-3-3β, 14-3-3ζ, and α-spectrin
fragments are below the limit of detection in CSF of sham rats Novel CSF Neurodegeneration
subjected to surgical procedures only, but are readily measur- Markers Provide Evidence for Acute
able following ischemia and increase by up to 20-fold. Quan- Central Nervous System Damage
titative Western blot analysis demonstrates that CSF levels of
14-3-3β increase with increasing ischemia duration, and cor-
in Humans
relate with the severity of acute cerebral histopathology as Some patients who undergo cardiopulmonary bypass develop
assessed by Fluoro-Jade and silver impregnation labeling of lasting cognitive and neurologic problems post surgery.2,3
acutely degenerating cells (Fig. 18.2; see also reference 68). Proteins released from degenerating neurons obtained from
When cases of ischemia are analyzed separate from the insult patients’ lumbar CSF during thoracoabdominal surgeries to
duration, CSF levels of 14-3-3β and the calpain-cleaved repair aortic aneurysms were analyzed; some of the patients
17 kDa tau fragment correlate significantly with the numbers underwent cardiopulmonary bypass with complete circula-
of acutely degenerating neurons. This relationship is not ob- tion arrest. As shown in Figure 18.3, a subset of patients
served for all CSF markers because calpain-cleaved α-spectrin exhibit time-dependent increases in 14-3-3β, 14-3-3ζ, calpain-
levels have a limited relationship with injury severity. These cleaved α-spectrin, a hypophosphorylated form of NFH
results support the hypothesis that neuron-enriched proteins reactive with a phosphoform-specific monoclonal antibody
such as 14-3-3β change dynamically in CSF in relation to the (pNFH), and UCH-L1, manifesting in most instances between
degree of acute brain injury, and raise the possibility that brain 2 and 12 hours after circulation arrest. All seven cases of com-
injuries of mild severity that may be difficult to diagnose with plete circulation arrest exhibit time-dependent increases in
routine neuroimaging, may be detectable instead using sur- multiple novel markers for neurodegeneration.77 More recent
rogate markers for neurodegeneration. experiments have increased the biomarker panel to eight

24 6 6 24 24 Sham Ischemic
Sham Mild Mod Mild Mod

Intact
Intact 150 kDa α-spectrin
150 kDa α-spectrin 120 kDa
120 kDa

30 kDa 14-3-3 ζ 14-3-3 ζ

30 kDa 14-3-3 β 14-3-3 β


A B

Fig. 18.1  Proteins released by degenerating neurons increase in cerebrospinal fluid (CSF) of rats following traumatic brain injury (TBI) (A) or cerebral
ischemia (B). TBI of varying severity was produced in a lateral fluid-percussion rat model. At 6 or 24 hours following TBI or sham surgery, CSF was
collected and subjected to Western blot analysis for α-spectrin, 14-3-3β, and 14-3-3ζ. Compared to sham controls, TBI rapidly increased intact
α-spectrin, a ≈150 kDa calpain cleavage product, and to a lesser degree a ≈120 kDa caspase cleavage product, and the two 14-3-3 isoforms. CSF
levels of all these proteins varied in relation to injury severity. In B, a similar analysis was conducted in rats subjected to either sham surgery or 10
minutes of global forebrain ischemia with 48-hour reperfusion. In both models the calpain derivative of α-spectrin predominates but the caspase
derivative also is sometimes increased, that is, both necrotic and apoptotic mechanisms contribute to the acute brain injuries. (Modified from Siman R,
McIntosh TK, Soltesz KM, et al. Proteins released from degenerating neurons are surrogate markers for acute brain damage in the rat. Neurobiol Dis 2004;16:311–20.)
172 Section II—Clinical and Laboratory Assessment

21,000 14-3-3β 14-3-3β 14-3-3ζ


18,000 R2 = 0.8046

Protein level (U/µL)


15,000 * **
12,000
9000 400
600
6000

CSF protein level (U/µL)


3000
0 300
0 10,000 20,000 30,000 40,000 50,000 400
Fluoro-Jade Positive Cells
200

5000 Calpain-cleaved spectrin 200


R2 = 0.0184
Protein level (U/µL)

100
4000

3000

2000

Magnesium

Magnesium
Vehicle

Vehicle
1000

0
0 10,000 20,000 30,000 40,000 50,000
Fluoro-Jade Positive Cells
Fig. 18.2  14-3-3 protein levels in rat cerebrospinal fluid (CSF) reflect the magnitude of experimental brain injury and are pharmcodynamic
measures of neuroprotectant therapy. Left, CSF content of 14-3-3β but not cleaved α-spectrin correlates with global ischemic neurodegeneration
irrespective of the duration of the ischemia. The total numbers of acutely degenerating neurons at 48 hours post injury were determined from Fluoro-
Jade staining and compared with CSF 14-3-3β and calpain-cleaved α-spectrin content from rats given episodes of global forebrain ischemia ranging
from 3 to 10 minutes. Degenerating cell counts are from the major vulnerable regions: hippocampus, parietal cortex, striatum, and inferior colliculus.
Right, The increase in CSF 14-3-3β and 14-3-3ζ following experimental traumatic brain injury (TBI) in the rat are attenuated by a neuroprotectant
treatment. Levels of the two 14-3-3 variants are shown 6 hours after lateral fluid percussion TBI in the rat, either in the absence or presence of
treatment with magnesium chloride (n = 12/group), an established neuroprotectant in this TBI model. *, p < 0.05; **, p < 0.01. (Left, modified from
Siman R, Zhang C, Roberts VL, et al. Novel surrogate markers for acute brain damage: cerebrospinal fluid levels correlate with severity of ischemic neurodegeneration in
the rat. J Cereb Blood Flow Metab 2005;25:1433–44.)

Cleaved
14-3-3β 14-3-3ζ α-spectrin pNFH UCH-L1
Case # 0 24 0 24 0 24 0 24 0 24
Circ arrest
1
5
13
16
18
38

No arrest
3
6
8
9
11
12
14
31
37
45
49

Fig. 18.3  Increases in multiple cerebrospinal fluid (CSF) markers occur in patients who undergo aortic surgery using cardiopulmonary
bypass with deep hypothermic circulatory arrest (DHCA). CSF levels of 14-3-3β, 14-3-3ζ, cleaved α-spectrin, a hypophosphorylated form of
neurofilament H (pNFH), and UCH-L1 were quantified before cardiopulmonary bypass with DHCA and 24 hours after reestablishment of circulation,
and divided into four quadrants: low (blue), moderate (yellow), high (orange), and very high (red) levels. The aortic surgical cases are shown separated
into groups that either were subjected to DHCA or not. Note that all cases of DHCA are accompanied by time-dependent CSF increases in at least
four of the proteins. In addition, 3 of 12 cases that did not involve cardiopulmonary bypass with DHCA also show time-dependent CSF increases in
at least four of the proteins. All three of these cases had acute neurologic complications indicative of CNS injury. (From Siman R, Roberts VL, McNeil E,
et al. Biomarker evidence for mild central nervous system injury after surgically-induced circulation arrest. Brain Res 2008;1213:1–11.)
Section II—Clinical and Laboratory Assessment 173

members, as NSE and two additional phosphoforms of NFH following TBI. Peak CSF levels of these neuron-enriched pro-
(dephosphorylated NFH and hyperphosphorylated NFH) also teins are reached typically between 72 and 96 hours after
increase following clinically induced circulation arrest. Among severe TBI. The same panel of neuron-enriched proteins also
12 cases of aneurysm repair that did not require complete may be sensitive, specific, and clinically useful serum markers
circulation arrest, increased CSF biomarkers for neurodegen- for acute brain damage because severe TBI increases serum
eration were restricted to the subset that developed acute neu- levels of the three distinct neurofilament H phosphoforms.
rologic complications.77 The findings indicate that candidate The results identify eight neuron-enriched proteins that may
markers identified initially in cultured rat cortical neurons can serve as a panel of surrogate markers for the minimally inva-
be elevated in an accessible body fluid in humans, and raise sive clinical detection, management, and therapeutic evalua-
the possibility that CSF detection of a panel of neurodegen- tion of TBI and ischemic brain injuries, including mild forms
eration markers may have clinical use to identify and guide of damage.
treatment of ischemic CNS injury. A clinical utility in ischemic
brain damage is supported by a recent study of CSF neurode-
generation biomarkers in aneurysmal subarachnoid hemor-
Future Directions
rhage patients, in which elevations in the marker panel Despite recent research efforts that have increased the number
were associated with the incidence and severity of cerebral of promising candidate surrogate markers for acute brain
vasospasm, brain infarction, and long-term dysfunctional damage, there remain several significant hurdles to bring these
outcomes.118 biomarkers to widespread clinical practice. First will be the
The same neuron-enriched protein panel that serves as a establishment of validated biomarker detection methods along
CSF biomarker for ischemic central nervous system injury with prospectively defined cutoff levels that can provide valu-
following circulation arrest also may have clinical use in TBI. able diagnostic or prognostic information as part of routine
The Siman laboratory examined a panel of eight biomarker clinical practice. Precedents are available from other fields for
candidates in CSF samples derived from patients with severe the successful development and clinical application of bio-
TBI, and presurgical and normal pressure hydrocephalus con- marker immunoassays. However, it still is uncertain whether
trols. By quantitative Western blot (Fig. 18.4) and fluorescence surrogate markers for acute brain damage can be identified
enzyme immunoassays (data not shown), each of the eight for which CSF or serum levels change consistently and can be
proteins is near or below the detection limit in controls, but measured with sufficient speed to direct the course of patient
all eight are detected readily and elevated consistently in CSF care, and with sufficient reliability to apply across multiple
hospitals. Second, additional research is required to determine
TBI-3 if either the time course or magnitude of biomarker altera-
TB1-10
NPH-1

TB1-8

tions, or both, has value in the diagnosis, prognosis, and thera-


24 48 72 96 peutic treatment of brain injuries. There are only a handful of
replicated studies that demonstrate relationships between bio-
14-3-3β marker alterations and neurologic, behavioral, or neuroradio-
logic measures of short- and long-term brain dysfunction, and
biomarker alterations have yet to demonstrate clinical signifi-
14-3-3ζ cance when obtained in a prospective fashion, as would be
required of a clinical tool for the diagnostic, prognostic, and
UCH-L1 therapeutic evaluation of acute brain damage. Finally, whereas
immunoassay methods are in place to measure many candi-
NSE date biomarkers at the laboratory bench with high sensitivity
and specificity that may be completed within a period of
hours, clinical application of these biomarkers would benefit
Intact α-spectrin
from quantitation that can be completed within minutes and
with extremely high reliability to provide the rapid diagnostic
or prognostic information needed at the bedside.
Calpain fragment To date, studies of biomarkers for acute brain injury have
Caspase fragment
been conducted mostly with CSF or serum. Brain interstitial
fluid, measured through intracerebral microdialysis, is another
Total NFH potential source to measure clinically meaningful biomarker
changes. Studies suggest the feasibility of microdialysis12-14 to
Fig. 18.4  A panel of biomarkers for human traumatic brain injury assess protein biomarkers for acute brain damage. It is con-
(TBI) based on proteins released from degenerating neurons. ceivable that by using probes that allow macromolecules such
Western blot analysis is shown here of ventricular cerebrospinal fluid as moderately sized proteins and their proteolytic fragments
(CSF) samples taken from cases of severe TBI (Glasgow Coma Scale <9) to be collected, it may be possible to use brain interstitial
or normal pressure hydrocephalus (NPH).All six proteins released from
fluids to detect biomarkers that may help assess, on a near-
degenerating neurons increase in human CSF after TBI. As exemplified
by case TBI-3, neurodegeneration markers are elevated by 24 hours and
continuous basis, the onset, severity, and localization of sec-
reach peak levels typically between 72 and 96 hours post injury. NFH, ondary pathophysiologic processes and how management
Neurofilament heavy chain; NSE, neuron-specific enolase; UCH, ubiquitin affects these processes.
C-terminal hydrolase. (From Siman R, Toraskar N, Dang A, et al. A panel of Another potential application to be explored is the contin-
neuron-enriched proteins as markers for traumatic brain injury in humans. J ued development of biomarkers that distinguish different
Neurotrauma 2009; 26:1867–77.) mechanisms of brain injury, for example, apoptotic and
174 Section II—Clinical and Laboratory Assessment

necrotic neurodegeneration. The identification of specific Michael McGarvey, M. Sean Grady, and Tracy McIntosh, along with
proteolytic fragments of brain-enriched proteins generated Eileen Maloney, Justin Plaum, Victoria Roberts, Lindsay Dressler, Eliza-
by either calpains or caspases raises the possibility of develop- beth McNeil, Antony Dang, and Nikhil Toraskar. This research was sup-
ing an approach to assess the relative contributions of calpain- ported by R01 NS048234 (RS) and P50 NS08803 (TKM, MSG).
driven necrosis and caspase-mediated apoptosis in brain
damage. Such patient-specific information may help target
pharmacologic and other interventions to the appropriate References
patient most likely to benefit from particular treatments that 1. Goldstein M. Traumatic brain injury: a silent epidemic. Ann Neurol
differentially impact pathophysiologic processes. A panel of 1990;27:327.
brain-enriched protein biomarkers also may provide clinically 2. Roche GW, Kanchuger M, Mangano CM, et al. Adverse cerebral outcomes
after coronary bypass surgery. N Engl J Med 1996;335:1857–64.
useful information on the relative involvement of particular 3. Van Dijk D, Jansen WE, Hijman R, et al. Cognitive outcome after off-pump
brain regions or neuronal elements. For example, a panel of and on-pump coronary artery bypass graft surgery: a randomized trial.
biomarkers may be differentially expressed across the neur- JAMA 2002;287:1405–12.
axis, raising the possibility that differential changes in them 4. Williams GR, Jiang JG, Matchar DB, et al. Incidence and occurrence of total
may provide clinically useful information on the relative (first-ever and recurrent) stroke. Stroke 1999;30:2523–8.
5. Teasdale G, Teasdale E, Hadley D. Computed tomographic and magnetic
involvement of particular brain regions in evolving degenera- resonance imaging classification of head injury. J Neurotrauma
tive pathology.104 Petzold66 has proposed that the CSF and 1992;9(Suppl. 1):S249–57.
serum alterations in an NFH phosphoform that is concen- 6. Guadagno JV, Calautti C, Baron JC. Progress in imaging stroke: emerging
trated in axons reflects axonal damage. It is possible that with clinical applications. Br Med Bull 2003;65:145–57.
7. Zhang Z, Larner SF, Kobeissy F, et al. Systems biology and theranostic
knowledge about markers such as somatodendritic neuronal approach to drug discovery and development to treat traumatic brain injury.
proteins (e.g., MAP2, astroglial proteins [e.g., S100β], and Methods Mol Biol 2010;662:317–29.
oligodendroglial proteins [e.g., MBP]), it may be possible to 8. Wagner AK. TBI translational rehabilitation research in the 21st century:
distinguish pathophysiologic processes that affect particular exploring a Rehabilomics research model. Eur J Phys Rehabil Med 2010;
and subcellular elements and so may respond differently to 46(4):549–56.
9. Pasinetti GM, Fivecoat H, Ho L. Personalized medicine in traumatic brain
pharmacologic intervention. injury. Psychiatr Clin North Am 2010;33(4):905–13.
10. Manley GT, Diaz-Arrastia R, Brophy M, et al. Common data elements for
Conclusion traumatic brain injury: recommendations from the biospecimens and
biomarkers working group. Arch Phys Med Rehabil 2010;91(11):1667–72.
11. Oli MW, Hayes RL, Robinson G, et al. Traumatic brain injury biomarkers:
An important future application of biomarkers is the potential from pipeline to diagnostic assay development. Methods Mol Biol 2009;
to serve as pharmacodynamic measures of injury. Only a 566:293–302.
limited number of clinical studies have evaluated the impact 12. Afinowi R, Tisdall M, Keir G, et al. Improving the recovery of S100β protein
of treatment on the magnitude and time course of biomarker in cerebral microdialysis: implications for multimodal monitoring in
alterations that accompany acute brain injuries, and even neurocritical care. J Neurosci Methods 2009;181(1):95–9. Epub 2009 Mar 14.
13. Petzold A, Tisdall MM, Girbes AR, et al. In vivo monitoring of neuronal loss
fewer examples of biomarker levels that serve as pharmacody- in traumatic brain injury: a microdialysis study. Brain 2011;134(Pt 2):
namic measures that respond to intervention (e.g., Baker 464–83.
et al).119 Such pharmacodynamic markers could serve as end- 14. Marklund N, Blennow K, Zetterberg H, et al. Monitoring of brain interstitial
point measures in early stage clinical trials to assess the com- total tau and beta amyloid proteins by microdialysis in patients with
traumatic brain injury. J Neurosurg 2009;110(6):1227–37.
parative efficacies of various interventions, assess the beneficial 15. Qureshi AI. Evaluation of brain injury using a blood test: is there a role in
effects of experimental treatments, and guide current strate- clinical practice? Crit Care Med 2002;30:2778–9.
gies of management, for example, determining the optimal 16. Ingebrigtsen T, Romner B. Biochemical serum markers of traumatic brain
CPP or allow practitioners to identify not only whether injury. J Trauma 2002;52:798–808.
patients are at risk of damage, but also whether they are still 17. Betz A, Dietrich W. Blood-brain barrier dysfunction in cerebral ischemia.
In: Ginsberg MD, Bogousslavsky J, editors. Cerebrovascular disease. Oxford,
able to benefit from therapy. Pharmacodynamic markers that England: Blackwell; 1998. p. 358–70.
correlate with the severity of an evolving injury can be simply 18. Del Zoppo GJ, Hallenbeck JM. Advances in the vascular pathophysiology of
and rapidly measured and could reduce the complexity and ischemic stroke. Thromb Res 2000;98:73–81.
expense and increase the discriminative power of early stage 19. Wiltfang J, Otto M, Baxter HC, et al. Isoform pattern of 14-3-3 proteins in
the cerebrospinal fluid of patients with Creutzfeldt-Jakob disease.
intervention trials. This is an important potential application J Neurochem 1997;73:2485–90.
for surrogate markers given the large size and expense of con- 20. Green AJE. Cerebrospinal fluid brain-derived proteins in the diagnosis of
ducting clinical trials and the failure of many trials of experi- Alzheimer’s disease and Creutzfeldt-Jakob disease. Neuropathol Appl
mental agents in the past that relied on clinical outcome Neurobiol 2002;28:427–40.
measures. In addition, knowledge biomarkers may help relate 21. Donato R. S100: a multigenic family of calcium-modulated proteins of the
EF-hand type with intracellular and extracellular functional roles. Int J
the neurologic and neurobehavioral endpoint measures to the Biochem Cell Biol 2001;33:637–68.
underlying pathophysiologic processes and neuroanatomic 22. McKeating E, Andrews P, Mascia L. Relationship of neuron specific enolase
correlates for brain dysfunction. Basic science research has and S100 concentrations in systemic and jugular venous system to injury
increased our knowledge about the mechanisms involved in severity and outcome after traumatic brain injury. Acta Neurochir 1998;
71:117–9.
brain injury and led to the discovery of new biomarkers. 23. Blomquist S, Johnsson P, Luhrs C, et al. The appearance of S100 protein in
However, translating this research to patients’ benefits still serum during and immediately after cardiopulmonary bypass surgery:
remains a formidable challenge. a possible marker for cerebral injury. J Cardiotharac Vasc Anesth 1997;11:
699–703.
24. Rosen H. Increased serum levels of the S-100 protein are associated with
Acknowledgments hypoxic brain damage after cardiac arrest. Stroke 1998;29:473–7.
25. Aurell A, Rosengren, I, Karlsson B, et al. Determination of S-100 and glial
The research described in this chapter from the Siman laboratory was fibrillary acidic protein concentration in cerebrospinal fluid after brain
conducted with many colleagues: Drs. Chen Zhang, Robert Neumar, infarction. Stroke 1991;22:1254–8.
Section II—Clinical and Laboratory Assessment 175

26. Mussack T, Biberthaler P, Kanz K-G, et al. Serum S100β and interleukin-8 as 39. Geyer C, Ulrich A, Gräfe G, et al. Diagnostic value of S100β and neuron-
predictive markers for comparative neurologic outcome analysis of patients specific enolase in mild pediatric traumatic brain injury. J Neurosurg Pediatr
after cardiac arrest and severe traumatic brain injury. Crit Care Med 2002; 2009;4(4):339–44.
30:2669–74. 40. Bechtel K, Frasure S, Marshall C, et al. Relationship of serum S100β levels
27. Wunderlich MT, Ebert AD, Kratz T, et al. Early neurobehavioral outcome and intracranial injury in children with closed head trauma. Pediatrics
after stroke is related to release of neurobiochemical markers of brain 2009;124:e697–704.
damage. Stroke 1999;30:1190–5. 41. Morochovic R, Rácz O, Kitka M, et al. Serum S100β protein in early
28. Stein DM, Kufera JA, Lindell A, et al. Association of CSF biomarkers and management of patients after mild traumatic brain injury. Eur J Neurol
secondary insults following severe traumatic brain injury. Neurocrit Care 2009;16(10):1112–7.
2011;14(2):200–7. 42. Green A, Harvey R, Thompson E, et al. Increased S100 beta in the
29. Vos PE, Jacobs B, Andriessen TM, et al. GFAP and S100β are biomarkers of cerebrospinal fluid of patients with frontotemporal dementia. Neurosci Lett
traumatic brain injury: an observational cohort study. Neurology 2010; 1997;235:5–8.
75(20):1786–93. 43. Otto M, Wiltfang J, Schutz E, et al. Diagnosis of Creutzfeldt-Jakob disease by
30. Wojtczak-Soska K, Lelonek M. S-100β protein: an early prognostic marker measurement of S100 protein in serum: prospective case-control study. BMJ
after cardiac arrest. Cardiol J 2010;17(5):532–6. 1998;316:577–82.
31. Murillo-Cabezas F, Muñoz-Sánchez MA, Rincón-Ferrari MD, et al. The 44. Anderson RE, Hansson LO, Nilsson O, et al. High serum S100β levels for
prognostic value of the temporal course of S100beta protein in post-acute trauma patients without head injuries. Neurosurgery 2001;48:1255–58.
severe brain injury: a prospective and observational study. Brain Inj 2010; 45. Savola O, Pyhtinen J, Leino TK, et al. Effects of head and extracranial
24(4):609–19. injuries on serum protein S100B levels in trauma patients. J Trauma
32. Cakir Z, Aslan S, Umudum Z, et al. S-100β and neuron-specific enolase levels in 2004;56:1229–34.
carbon monoxide-related brain injury. Am J Emerg Med 2010;28(1):61–7. 46. Beems T, Simons KS, van Geel WJA, et al. Serum and CSF concentrations
33. James ML, Blessing R, Phillips-Bute BG, et al. S100β and brain natriuretic of brain-specific proteins in hydrocephalus. Acta Neurochir 2003;145:
peptide predict functional neurological outcome after intracerebral 37–43.
haemorrhage. Biomarkers 2009;14(6):388–94. 47. Piazza O, Storti MP, Cotena S, et al. S100β is not a reliable prognostic index
34. Filippidis AS, Papadopoulos DC, Kapsalaki EZ, et al. Role of the S100β in paediatric TBI. Pediatr Neurosurg 2007;43:258–64.
serum biomarker in the treatment of children suffering from mild traumatic 48. Bazarian JJ, Zemlan FP, Mookerjee S, et al. Serum S100β and cleaved tau are
brain injury. Neurosurg Focus 2010;29(5):E2. poor predictors of long-term outcome after mild traumatic brain injury.
35. Hallén M, Karlsson M, Carlhed R, et al. S-100β in serum and urine after Brain Inj 2006;20:759–65.
traumatic head injury in children. J Trauma 2010;69(2):284–9. 49. Muller K, Elverland A, Romner B, et al. Analysis of protein S-100β in serum:
36. Undén J, Romner B. Can low serum levels of S100β predict normal CT a methodological study. Clin Chem Lab Med 2006;44:1111–4.
findings after minor head injury in adults?: an evidence-based review and 50. Vos PE, van Gils M, Beems T, et al. Increased GFAP and S100β but not NSE
meta-analysis. J Head Trauma Rehabil 2010;25(4):228–40. serum levels after subarachnoid haemorrhage are associated with clinical
37. Nylen K, Ost M, Csajbok LZ, et al. Serum levels of S100B, S100A1B and severity. Eur J Neurol 2006;13:632–8.
S100BB are all related to outcome after severe traumatic brain injury. Acta
Neurochir 2008;150:221–7. A complete list of references for this chapter can be found online at
38. Stranjalis G, Korfias S, Psachoulia C, et al. The prognostic value of serum www.expertconsult.com.
S-100β protein in spontaneous subarachnoid haemorrhage. Acta Neurochir
2007;149:231–7.
Section II—Clinical and Laboratory Assessment 175.e1

References 27. Wunderlich MT, Ebert AD, Kratz T, et al. Early neurobehavioral outcome
after stroke is related to release of neurobiochemical markers of brain
1. Goldstein M. Traumatic brain injury: a silent epidemic. Ann Neurol damage. Stroke 1999;30:1190–5.
1990;27:327. 28. Stein DM, Kufera JA, Lindell A, et al. Association of CSF biomarkers and
2. Roche GW, Kanchuger M, Mangano CM, et al. Adverse cerebral outcomes secondary insults following severe traumatic brain injury. Neurocrit Care
after coronary bypass surgery. N Engl J Med 1996;335:1857–64. 2011;14(2):200–7.
3. Van Dijk D, Jansen WE, Hijman R, et al. Cognitive outcome after off-pump 29. Vos PE, Jacobs B, Andriessen TM, et al. GFAP and S100β are biomarkers of
and on-pump coronary artery bypass graft surgery: a randomized trial. traumatic brain injury: an observational cohort study. Neurology 2010;
JAMA 2002;287:1405–12. 75(20):1786–93.
4. Williams GR, Jiang JG, Matchar DB, et al. Incidence and occurrence of total 30. Wojtczak-Soska K, Lelonek M. S-100β protein: an early prognostic marker
(first-ever and recurrent) stroke. Stroke 1999;30:2523–8. after cardiac arrest. Cardiol J 2010;17(5):532–6.
5. Teasdale G, Teasdale E, Hadley D. Computed tomographic and magnetic 31. Murillo-Cabezas F, Muñoz-Sánchez MA, Rincón-Ferrari MD, et al. The
resonance imaging classification of head injury. J Neurotrauma 1992; prognostic value of the temporal course of S100beta protein in post-acute
9(Suppl. 1):S249–57. severe brain injury: a prospective and observational study. Brain Inj 2010;
6. Guadagno JV, Calautti C, Baron JC. Progress in imaging stroke: emerging 24(4):609–19.
clinical applications. Br Med Bull 2003;65:145–57. 32. Cakir Z, Aslan S, Umudum Z, et al. S-100beta and neuron-specific enolase
7. Zhang Z, Larner SF, Kobeissy F, et al. Systems biology and theranostic levels in carbon monoxide-related brain injury. Am J Emerg Med 2010;
approach to drug discovery and development to treat traumatic brain 28(1):61–7.
injury. Methods Mol Biol 2010;662:317–29. 33. James ML, Blessing R, Phillips-Bute BG, et al. S100β and brain natriuretic
8. Wagner AK. TBI translational rehabilitation research in the 21st century: peptide predict functional neurological outcome after intracerebral
exploring a Rehabilomics research model. Eur J Phys Rehabil Med 2010; haemorrhage. Biomarkers 2009;14(6):388–94.
46(4):549–56. 34. Filippidis AS, Papadopoulos DC, Kapsalaki EZ, et al. Role of the S100β
9. Pasinetti GM, Fivecoat H, Ho L. Personalized medicine in traumatic brain serum biomarker in the treatment of children suffering from mild
injury. Psychiatr Clin North Am 2010;33(4):905–13. traumatic brain injury. Neurosurg Focus 2010;29(5):E2.
10. Manley GT, Diaz-Arrastia R, Brophy M, et al. Common data elements 35. Hallén M, Karlsson M, Carlhed R, et al. S-100β in serum and urine after
for traumatic brain injury: recommendations from the biospecimens and traumatic head injury in children. J Trauma 2010;69(2):284–9.
biomarkers working group. Arch Phys Med Rehabil 2010;91(11):1667–72. 36. Undén J, Romner B. Can low serum levels of S100β predict normal CT
11. Oli MW, Hayes RL, Robinson G, et al. Traumatic brain injury biomarkers: findings after minor head injury in adults?: an evidence-based review and
from pipeline to diagnostic assay development. Methods Mol Biol meta-analysis. J Head Trauma Rehabil 2010;25(4):228–40.
2009;566:293–302. 37. Nylen K, Ost M, Csajbok LZ, et al. Serum levels of S100B, S100A1B and
12. Afinowi R, Tisdall M, Keir G, et al. Improving the recovery of S100β S100BB are all related to outcome after severe traumatic brain injury. Acta
protein in cerebral microdialysis: implications for multimodal monitoring Neurochir 2008;150:221–7.
in neurocritical care. J Neurosci Methods 2009;181(1):95–9. Epub 2009 38. Stranjalis G, Korfias S, Psachoulia C, et al. The prognostic value of serum
Mar 14. S-100β protein in spontaneous subarachnoid haemorrhage. Acta Neurochir
13. Petzold A, Tisdall MM, Girbes AR, et al. In vivo monitoring of neuronal 2007;149:231–7.
loss in traumatic brain injury: a microdialysis study. Brain 2011;134(Pt 2): 39. Geyer C, Ulrich A, Gräfe G, et al. Diagnostic value of S100β and
464–83. neuron-specific enolase in mild pediatric traumatic brain injury.
14. Marklund N, Blennow K, Zetterberg H, et al. Monitoring of brain J Neurosurg Pediatr 2009;4(4):339–44.
interstitial total tau and beta amyloid proteins by microdialysis in patients 40. Bechtel K, Frasure S, Marshall C, et al. Relationship of serum S100β levels
with traumatic brain injury. J Neurosurg 2009;110(6):1227–37. and intracranial injury in children with closed head trauma. Pediatrics
15. Qureshi AI. Evaluation of brain injury using a blood test: is there a role in 2009;124:e697–704.
clinical practice? Crit Care Med 2002;30:2778–9. 41. Morochovic R, Rácz O, Kitka M, et al. Serum S100β protein in early
16. Ingebrigtsen T, Romner B. Biochemical serum markers of traumatic brain management of patients after mild traumatic brain injury. Eur J Neurol
injury. J Trauma 2002;52:798–808. 2009;16(10):1112–7.
17. Betz A, Dietrich W. Blood-brain barrier dysfunction in cerebral ischemia. 42. Green A, Harvey R, Thompson E, et al. Increased S100 beta in the
In: Ginsberg MD, Bogousslavsky J, editors. Cerebrovascular disease. Oxford, cerebrospinal fluid of patients with frontotemporal dementia. Neurosci Lett
England: Blackwell; 1998. p. 358–70. 1997;235:5–8.
18. Del Zoppo GJ, Hallenbeck JM. Advances in the vascular pathophysiology of 43. Otto M, Wiltfang J, Schutz E, et al. Diagnosis of Creutzfeldt-Jakob disease
ischemic stroke. Thromb Res 2000;98:73–81. by measurement of S100 protein in serum: prospective case-control study.
19. Wiltfang J, Otto M, Baxter HC, et al. Isoform pattern of 14-3-3 proteins in BMJ 1998;316:577–82.
the cerebrospinal fluid of patients with Creutzfeldt-Jakob disease. 44. Anderson RE, Hansson LO, Nilsson O, et al. High serum S100β levels
J Neurochem 1997;73:2485–90. for trauma patients without head injuries. Neurosurgery 2001;48:
20. Green AJE. Cerebrospinal fluid brain-derived proteins in the diagnosis of 1255–58.
Alzheimer’s disease and Creutzfeldt-Jakob disease. Neuropathol Appl 45. Savola O, Pyhtinen J, Leino TK, et al. Effects of head and extracranial
Neurobiol 2002;28:427–40. injuries on serum protein S100β levels in trauma patients. J Trauma
21. Donato R. S100: a multigenic family of calcium-modulated proteins of the 2004;56:1229–34.
EF-hand type with intracellular and extracellular functional roles. Int J 46. Beems T, Simons KS, van Geel WJA, et al. Serum and CSF concentrations
Biochem Cell Biol 2001;33:637–68. of brain-specific proteins in hydrocephalus. Acta Neurochir 2003;145:
22. McKeating E, Andrews P, Mascia L. Relationship of neuron specific enolase 37–43.
and S100 concentrations in systemic and jugular venous system to injury 47. Piazza O, Storti MP, Cotena S, et al. S100β is not a reliable prognostic index
severity and outcome after traumatic brain injury. Acta Neurochir 1998; in paediatric TBI. Pediatr Neurosurg 2007;43:258–64.
71:117–9. 48. Bazarian JJ, Zemlan FP, Mookerjee S, et al. Serum S100β and cleaved tau
23. Blomquist S, Johnsson P, Luhrs C, et al. The appearance of S100 protein in are poor predictors of long-term outcome after mild traumatic brain
serum during and immediately after cardiopulmonary bypass surgery: injury. Brain Inj 2006;20:759–65.
a possible marker for cerebral injury. J Cardiotharac Vasc Anesth 1997;11: 49. Muller K, Elverland A, Romner B, et al. Analysis of protein S-100β in
699–703. serum: a methodological study. Clin Chem Lab Med 2006;44:1111–4.
24. Rosen H. Increased serum levels of the S-100 protein are associated with 50. Vos PE, van Gils M, Beems T, et al. Increased GFAP and S100beta but not
hypoxic brain damage after cardiac arrest. Stroke 1998;29:473–7. NSE serum levels after subarachnoid haemorrhage are associated with
25. Aurell A, Rosengren, I, Karlsson B, et al. Determination of S-100 and glial clinical severity. Eur J Neurol 2006;13:632–8.
fibrillary acidic protein concentration in cerebrospinal fluid after brain 51. Nylen K, Csajbok LZ, Ost M, et al. Serum glial fibrillary acidic protein is
infarction. Stroke 1991;22:1254–8. related to focal brain injury and outcome after aneurysmal subarachnoid
26. Mussack T, Biberthaler P, Kanz K-G, et al. Serum S100β and interleukin-8 hemorrhage. Stroke 2007;38:1489–94.
as predictive markers for comparative neurologic outcome analysis of 52. Vos PE, Lamers KJ, Hendriks JC, et al. Glial and neuronal proteins in serum
patients after cardiac arrest and severe traumatic brain injury. Crit Care predict outcome after severe traumatic brain injury. Neurology 2004;62:
Med 2002;30:2669–74. 303–10.
175.e2 Section II—Clinical and Laboratory Assessment

53. Honda M, Tsuruta R, Kaneko T, et al. Serum glial fibrillary acidic protein is 77. Siman R, Roberts VL, McNeil E, et al. Biomarker evidence for mild central
a highly specific biomarker for traumatic brain injury in humans compared nervous system injury after surgically-induced circulation arrest. Brain Res
with S-100β and neuron-specific enolase. J Trauma 2010;69(1):104–9. 2008;1213:1–11.
54. Berger RP, Beers SR, Richichi R, et al. Serum biomarker concentrations and 78. Brophy GM, Pineda JA, Papa L, et al. AlphaII-spectrin breakdown product
outcome after pediatric traumatic brain injury. J Neurotrauma cerebrospinal fluid exposure metrics suggest differences in cellular injury
2007;24:1793–801. mechanisms after severe traumatic brain injury. J Neurotrauma
55. Fraser DD, Close TE, Rose KL, et al. Severe traumatic brain injury in 2009;26(4):471–9.
children elevates glial fibrillary acidic protein in cerebrospinal fluid and 79. Ning M, Sarracino DA, Kho AT, et al. Proteomic temporal profile of human
serum. Pediatr Crit Care Med 2011;12(3):319–24. brain endothelium after oxidative stress. Stroke 2011;42(1):37–43. Epub
56. Bembea MM, Savage W, Strouse JJ, et al. Glial fibrillary acidic protein as a 2010 Dec 16.
brain injury biomarker in children undergoing extracorporeal membrane 80. Foerch C, Montaner J, Furie KL, et al. Invited article: searching for oracles?
oxygenation. Pediatr Crit Care Med 2010; Nov 4. Epub ahead of print. Blood biomarkers in acute stroke. Neurology 20094;73(5):393–9.
57. Zemlan FP, Rosenberg WS, Luebbe PA, et al. Quantification of axonal 81. Lo TY, Jones PA, Minns RA. Combining coma score and serum biomarker
damage in traumatic brain injury: affinity purification and characterization levels to predict unfavorable outcome following childhood brain trauma.
of cerebrospinal fluid tau levels. J Neurochem 1999;72:741–50. J Neurotrauma 2010;27(12):2139–45. Epub 2010 Nov 2.
58. Shaw GJ, Jauch EC, Zemlan FP. Serum cleaved tau protein levels and 82. Turck N, Vutskits L, Sanchez-Pena P, et al. A multiparameter panel method
clinical outcome in adult patients with closed head injury. Ann Emerg Med for outcome prediction following aneurysmal subarachnoid hemorrhage.
2002;39:254–57. Intensive Care Med 2010;36(1):107–15. Epub 2009 Sep 17.
59. Liliang PC, Liang CL, Weng HC, et al. Tau proteins in serum predict 83. Springer JE. Apoptotic cell death following traumatic injury to the central
outcome after severe traumatic brain injury. J Surg Res 2010;160(2):302–7. nervous system. J Biochem Mol Biol 2002;35:94–105.
Epub 2009 Jan 10. 84. Ferrer I. Apoptosis: future targets for neuroprotective strategies.
60. Zingler VC, Krumm B, Bertsch T, et al. Early prediction of neurological Cerebrovasc Dis 2006;21 (Suppl. 2):9–20.
outcome after cardiopulmonary resuscitation: a multimodal approach 85. Yang F, Sun X, Beech W, et al. Antibody to caspase-cleaved actin detects
combining neurobiochemical and electrophysiological investigations may apoptosis in differentiated neuroblastoma and plaque-associated
provide high prognostic certainty in patients after cardiac arrest. Eur neurons and microglia in Alzheimer’s disease. Am J Pathol 1998;152:
Neurol 2003;49:79–84. 379–89.
61. Reisinger J, Hollinger K, Lang W, et al. Prediction of neurological outcome 86. Siman R, Bozyczko-Coyne D, Meyer SL, et al. Immunolocalization of
after cardiopulmonary resuscitation by serial determination of serum caspase proteolysis in situ: evidence for widespread caspase-mediated
neuron-specific enolase. Eur Heart J 2007;28:52–8. apoptosis of neurons and glia in the postnatal rat brain. Neuroscience
62. Shore PM, Berger RP, Varma S, et al. Cerebrospinal fluid biomarkers versus 1999;92:1425–42.
Glasgow Coma Scale and Glasgow Outcome Scale in pediatric traumatic 87. Zhang C, Siman R, Xu YA, et al. Comparison of calpain and caspase
brain injury: the role of young age and inflicted injury. J Neurotrauma activities in adult rat brain after transient forebrain ischemia. Neurobiol Dis
2007;24:75–86. 2002;10:289–305.
63. Herrmann M, Jost S, Kutz S, et al. Temporal profile of release of 88. Yatsiv I, Morganti-Kossmann MC, Perez D, et al. Elevated intracranial IL-18
neurobiochemical markers of brain damage after traumatic brain injury is in humans and mice after traumatic brain injury and evidence of
associated with intracranial pathology as demonstrated in cranial neuroprotective effects of IL-18 binding protein after experimental closed
computerized tomography. J Neurotrauma 2000;17:113–22. head injury. J Cereb Blood Flow Metab 2002;22:971–8.
64. Graham MR, Myers T, Evans P, et al. Direct hits to the head during amateur 89. Hayakata T, Shiozaki T, Tasaki O, et al. Changes in CSF S100beta and
boxing is associated with a rise in serum biomarkers for brain injury. Int J cytokine concentrations in early-phase severe traumatic brain injury. Shock
Immunopathol Pharmacol 2011;24(1):119–25. 2004;22:102–7.
65. Zetterberg H, Tanriverdi F, Unluhizarci K, et al. Sustained release of 90. Stein DM, Lindell A, Murdock KR, et al. Relationship of serum and
neuron-specific enolase to serum in amateur boxers. Brain Inj 2009; cerebrospinal fluid biomarkers with intracranial hypertension and cerebral
23(9):723–6. hypoperfusion after severe traumatic brain injury. J Trauma 2011;70(5):
66. Petzold A, Keir G, Kay A, et al. Axonal damage and outcome in 1096–103.
subarachnoid haemorrhage. J Neurol Neurosurg Psychiatry 2006;77:753–9. 91. Roberts-Lewis JM, Siman R. Spectrin proteolysis in the hippocampus:
67. Lewis SB, Wolper RA, Miralia L, et al. Detection of phosphorylated NF-H a biochemical marker for neuronal injury and neuroprotection. Ann N Y
in the cerebrospinal fluid and blood of aneurysmal subarachnoid Acad Sci 1993;679:78–86.
hemorrhage patients. J Cereb Blood Flow Metab 2008;28:1261–71. 92. Siman R, Bozyczko-Coyne D, Savage MJ, et al. The calcium-activated
68. Van Geel WJ, Rosengren LE, Verbeek MM. An enzyme immunoassay to protease calpain I and ischemia-induced neurodegeneration. Adv Neurol
quantify neurofilament light chain in cerebrospinal fluid. J Immunol 1996;71:167–74.
Methods 2005;296:179–85. 93. Wang KKW. Calpain and caspase: can you tell the difference? Trends
69. Rosen H, Karlsson JE, Rosengren L. CSF levels of neurofilament is a Neurosci 2000;23:20–6.
valuable predictor of long-term outcome after cardiac arrest. J Neurol Sci 94. Saatman KE, Creed J, Raghupathi R. Calpain as a therapeutic target in
2004;221:19–24. traumatic brain injury. Neurotherapeutics 2010;7(1):31–42.
70. Nylen K, Csajbok LZ, Ost M, et al. CSF neurofilament correlates with 95. Siman R, Reaume AG, Savage MJ, et al. Presenilin-1 P264L knock-in
outcome after aneurysmal subarachnoid hemorrhage. Neurosci Lett 2006; mutation: differential effects on Abeta production, amyloid deposition, and
404:132–6. neuronal vulnerability. J Neurosci 2000;20:8717–26.
71. Zanier ER, Refai D, Zipfel GJ, et al. Neurofilament light chain levels in 96. Neumar RW, Xu YA, Gada H, et al. Cross-talk between calpain and caspase
ventricular cerebrospinal fluid after acute aneurysmal subarachnoid proteolytic systems during neuronal apoptosis. J Biol Chem 2003;278:
haemorrhage. J Neurol Neurosurg Psychiatry 2011;82(2):157–9. 14162–67.
72. Sellner J, Petzold A, Sadikovic S, et al. The value of the serum 97. Siman R, McIntosh TK, Soltesz KM, et al. Proteins released from
neurofilament protein heavy chain as a biomarker for peri-operative brain degenerating neurons are surrogate markers for acute brain damage in the
injury after carotid endarterectomy. Neurochem Res 2009;34(11):1969–74. rat. Neurobiol Dis 2004;16:311–20.
Epub 2009 May 9. 98. Saido TC, Yokota M, Nagao S, et al. Spatial resolution of fodrin proteolysis
73. Glenney Jr JR, Glenney P. Fodrin is the general spectrin-like protein found in postischemic brain. J Biol Chem 1993;268:25239–43.
in most cells whereas spectrin and the TW protein have a restricted 99. Roberts-Lewis JM, Savage MJ, Marcy VR, et al. Immunolocalization of
distribution. Cell 1983;34:503–12. calpain I-mediated spectrin degradation to vulnerable neurons in the
74. Mondello S, Robicsek SA, Gabrielli A, et al. αII-Spectrin breakdown ischemic gerbil brain. J Neurosci 1994;14:3934–44.
products (SBDPs): diagnosis and outcome in severe traumatic brain injury 100. Saatman KE, Bozyczko-Coyne D, Marcy V, et al. Prolonged calpain-
patients. J Neurotrauma 2010;27(7):1203–13. mediated spectrin breakdown occurs regionally following experimental
75. Pineda JA, Lewis SB, Valadka AB, et al. Clinical significance of alphaII- brain injury in the rat. J Neuropathol Exp Neurol 1996;55:850–60.
spectrin breakdown products in cerebrospinal fluid after severe traumatic 101. Yamashima T, Saido TC, Takita M, et al. Transient brain ischemia provokes
brain injury. J Neurotrauma 2007;24:354–66. Ca2+, PIP2 and calpain responses prior to delayed neuronal death in
76. Lewis SB, Velat GJ, Miralia L, et al. Alpha-II spectrin breakdown products monkeys. Eur J Neurosci 1996;8:1932–44.
in aneurysmal subarachnoid hemorrhage: a novel biomarker of proteolytic 102. Aebersold R, Mann M. Mass spectrometry-based proteomics. Nature
injury. J Neurosurg 2007;107:792–6. 2003;422:198–207.
Section II—Clinical and Laboratory Assessment 175.e3

103. McDonald WH, Yates 3rd JR. Shotgun proteomics and biomarker discovery. 114. Smith DH, Okiyama K, Gennarelli TA, et al. Magnesium and ketamine
Dis Markers 2002;18:99–105. attenuate cognitive dysfunction following experimental brain injury.
104. Siman R, Zhang C, Roberts VL, et al. Novel surrogate markers for acute Neurosci Lett 1993;157:211–4.
brain damage: cerebrospinal fluid levels correlate with severity of ischemic 115. Feldman Z, Gurevitch B, Artru AA, et al. Effect of magnesium given 1 hour
neurodegeneration in the rat. J Cereb Blood Flow Metab 2005;25:1433–44. after head trauma on brain edema and neurological outcome. J Neurosurg
105. Siman R, Toraskar N, Dang A, et al. A panel of neuron-enriched proteins as 1996;85:131–7.
markers for traumatic brain injury in humans. J Neurotrauma 116. Bareyre FM, Saatman KE, Raghupathi R, et al. Postinjury treatment with
2009;26(11):1867–77. magnesium chloride attenuates cortical damage after traumatic brain injury
106. De Graan PN, Oestreicher AB, Schrama LH, et al. Phosphoprotein B-50 in rats. J Neurotrauma 2000;17:1029–39.
(GAP-43): localization and function. Prog Brain Res 1986;69:37–50. 117. Browne KD, Leoni MJ, Iwata A, et al. Acute treatment with MgSO4
107. Hemmer W, Wallimann T. Functional aspects of creatine kinase in brain. attenuates long-term hippocampal tissue loss after brain trauma in the rat.
Dev Neurosci 1993;15:249–60. J Neurosci Res 2004;77:878–83.
108. Aitken A, Jones D, Soneji Y, et al. 14-3-3 Proteins: biological function and 118. Siman R, Giovannone N, Toraskar N, et al. Evidence that a panel of
domain structure. Biochem Soc Trans 2003;23:605–11. neurodegeneration biomarkers predicts vasospasm, infarction, and outcome
109. Nacher J, Rosell DR, McEwen BS. Widespread expression of rat collapsin in aneurysmal subarachnoid hemorrhage. PLoS One 2011;12:e28938.
response-mediated protein 4 in the telencephalon and other areas of the 119. Baker AJ, Rhind SG, Morrison LJ, et al. Resuscitation with hypertonic
adult rat central nervous system. J Comp Neurol 2000; 424:628–39. saline-dextran reduces serum biomarker levels and correlates with outcome
110. Dahmen N, Jungblut PR, Fischer V, et al. Detection of TOAD-64 in adult in severe traumatic brain injury patients. J Neurotrauma
rat brain as revealed by two-dimensional protein gel electrophoresis 2009;26(8):1227–40.
followed by MALDI mass spectrometry: possible modulatory effect of 120. Ost M, Nylen K, Csajbok L, et al. Initial CSF total tau correlates with 1 year
chronic haloperidol treatment. Prog Neuropsychopharmacol Biol Psychiatry outcome in patients with traumatic brain injury. Neurology
2002;26:693–8. 2006;67:1600–4.
111. Pike BR, Zhao X, Newcomb JK, et al. Temporal relationships between de 121. Grubb NR, Simpson C, Sherwood RA, et al. Prediction of cognitive
novo protein synthesis, calpain and caspase 3-like protease activation, and dysfunction after resuscitation from out-of-hospital cardiac arrest using
DNA fragmentation during apoptosis in septo-hippocampal cultures. serum neuron-specific enolase and protein S-100. Heart 2007;93:1268–73.
J Neurosci Res 1998;52:505–20. 122. Wunderlich MT, Wallesch CS, Goertler M. Release of neurobiochemical
112. Yakovlev AG, Knoblach SM, Fan L, et al. Activation of CPP32-like caspases markers of brain damage is related to the neurovascular status on
contributes to neuronal apoptosis and neurological dysfunction after admission and the site of arterial occlusion in acute ischemic stroke.
traumatic brain injury. J Neurosci 1997;17:7415–24. J Neurol Sci 2004;227:49–53.
113. Raghupathi R, Graham DI, McIntosh TK. Apoptosis after traumatic brain 123. Bitsch A, Horn C, Kemmling Y, et al. Serum tau protein level as a marker of
injury. J Neurotrauma 2000;17:927–38. axonal damage in acute ischemic stroke. Eur Neurol 2002;47:45–51.
II
Chapter
19  

Volume Status and


Cardiac Function
Jose L. Pascual, Jiri Horak, Vicente H. Gracias, and Patrick J. Neligan

expansion or vasopressors frequently are used to augment


Introduction cerebral blood flow (CBF). However, positive fluid balance or
The injured brain is exquisitely sensitive to global changes in induced hypertension also can have adverse effects on outcome
blood flow and oxygen delivery. Brain injury rarely occurs in or intracranial physiology.3-5 Hence, an understanding of a
the absence of systemic injury, either as a primary (trauma) patient’s volume status is fundamental to his or her care. Acute
or secondary process (subarachnoid hemorrhage). Hence, and chronic intravascular fluid deficits may manifest with
volume and circulatory abnormalities may induce or worsen varying degrees of clinical findings. Patients with chronic
brain injuries, and brain injuries may induce circulatory volume deficits may present with a decreased skin turgor,
abnormalities. In the setting of cerebral autoregulatory dys- weight loss, sunken eyes, hypothermia, oliguria, orthostatic
function, large swings in cardiac output or peripheral resis- hypotension, and chronic low-grade tachycardia. On the other
tance may have an adverse effect on brain tissue perfusion. hand, those with acute volume deficits may present clinically
Furthermore, manipulation of volume status and blood pres- with changes in vital signs, with tachycardia and hypotension,
sure is central to the management of conditions such as sub- oliguria, and anuria, but also with more subtle changes in
arachnoid hemorrhage (SAH) and traumatic brain injury mental status, such as restlessness and confusion. Clinical
(TBI) to maintain cerebral perfusion pressure (CPP) or signs of volume excess include weight gain, pulmonary con-
prevent delayed cerebral ischemia (DCI). Whereas neurocriti- gestion or edema, and peripheral edema.
cal care focuses on neurologic monitoring, cardiovascular Basic laboratory tests should be obtained to help determine
monitoring is performed on all neurologically injured patients effective circulating volume. An increase in all plasma electro-
and the data derived are central to all therapeutic decisions. lyte concentrations may indicate volume loss, but creatinine
However, there is no single set of optimal hemodynamic or and blood urea nitrogen (BUN) elevations may be more spe-
cardiac values that applies to all patients. For example, an cific. In particular, a BUN-to-creatinine ratio greater than 15
adequate blood pressure to avoid cerebral hypoperfusion may be more sensitive to indicate an intravascular fluid deficit.
likely is different in a young individual with no preexisting In systemic volume loss, renal sodium retention results in
disease compared with an older patient who has hypertension urinary sodium concentration (UNa) that typically drops
and atherosclerotic disease. Consequently, an understanding below 20 mEq/L. The fractional excretion of sodium (FENa)
of the methods of cardiovascular monitoring, the principles is considered more sensitive and can be calculated by the
behind them, and interpretation of hemodynamic data is a formula
core competence for the neurointensivist. This chapter dis-
cusses techniques to monitor volume status and cardiac func- UNa × PCr/PNa × UCr ×100%
tion. The evaluation of cardiac ischemia, acute coronary
syndromes, and use of biomarkers in these disorders is beyond where U and P are urine and plasma measured concentra-
the scope of this chapter, and the reader is referred to recent tions of sodium and creatinine. A FENa less than 1% indi-
reviews on these topics.1,2 cates a reduced effective circulating volume seen by the
kidney provided no diuretics have been administered in the
previous 24 hours.
Physical Examination and Although these clinical and laboratory evaluations usually
Laboratory Determination are adequate to monitor the surgical or medical patient in the
outpatient or ward setting, they may often be insufficient and
of Effective Circulating Volume inconclusive in the critically ill patient. Instead, more sophis-
Evaluation of volume status, preload, and fluid responsiveness ticated, continuous and real-time methods of hemodynamic
is important in neurocritical care because hypotension is a cardiovascular monitoring become necessary to help deter-
well-known secondary cerebral insult. In addition, volume mine adequate global tissue perfusion.
176 © Copyright 2013 Elsevier Inc. All rights reserved.
Section II—Clinical and Laboratory Assessment 177

Blood Pressure Monitoring Oscillometry


Blood pressure, universally measured in intensive care units Many limitations of manual intermittent blood pressure mea-
(ICUs), reflects myriad factors that maintain the systemic cir- surement have been overcome by automated noninvasive
culation. These include, but are not limited to, circulating blood pressure (NIBP) devices. NIBP devices measure mean
blood volume (both stressed and unstressed) venous return, arterial pressure and use an algorithm to estimate the systolic
splanchnic venular and arteriolar tone, vascular resistance and diastolic blood pressure. The major advantages of auto-
within several microcirculations, heart rate, cardiac compli- mated blood pressure monitoring are reliability, reproduc-
ance, right ventricular afterload, airway pressure, left ventricu- ibility, and built-in low- and high-pressure alarms. In addition,
lar contractility, core body temperature, baroreceptor reflexes in modern devices data are transferred directly into the
and adrenal medullary output. Distilling these factors into a monitor or information system, freeing the bedside caregiver
single figure—the mean arterial pressure—to guide therapy is for other duties. This may result in more frequent sampling
fraught with hazard. In simple terms, blood pressure reflects but perhaps less attentiveness to the results.
stroke volume and peripheral resistance. However, compensa- Most automated NIBP devices are based on oscillometry.
tory reflexes in one of these parameters may balance variabil- This monitoring system consists of pneumatic and electronic
ity in the other. An example is vasoconstriction in the setting components. Modern oscillometers use a single cuff to
of hypovolemia. The result may be misleadingly “normal” occlude the artery and sense the arterial signal. The cuff is
blood pressure despite severe systemic disease. Convention- inflated above systolic pressure and then deflates either con-
ally, blood pressure is measured two ways: noninvasively, using tinuously or in a step-wise manner. As the cuff pressure
manual techniques or oscillometry, and invasively, using direct decreases, at a rate about 4 mm Hg per second, below occlu-
arterial cannulation. sive pressure, blood starts flowing through the artery and
causes detectable oscillation. The pulse pressure wave and the
gauge pressure in the occluding cuff are detected and con-
Noninvasive verted into an electronic signal by a transducer. The pressure
at which the peak amplitude of arterial pulsations occurs cor-
Manual Intermittent Techniques responds closely to directly measured mean arterial pressure
To measure both systolic and diastolic arterial pressure, the (MAP), and values for systolic and diastolic pressure are
most widely used intermittent manual method is the ausculta- derived from proprietary formulas that examine the rate of
tory technique, originally described by Scipione et al.6 but change of the pressure pulsations. A microprocessor uses an
popularized by Korotkoff in 1905. Using a sphygmomanom- algorithm to determine systolic, diastolic, and mean arterial
eter, cuff, and stethoscope, Korotkoff measured blood pressure pressures. Systolic pressure is identified as the pressure at
by auscultation of the sounds generated by arterial blood flow. which pulsations are increasing and are at 25% to 50% of
These sounds are a complex series of audible frequencies pro- maximum. Diastolic pressure is the most difficult value to
duced by turbulent flow, instability of the arterial wall, and determine by oscillometry and is commonly recorded when
shock wave formation as external occluding pressure on the the pulse amplitude has declined from the peak value by
artery is reduced. The pressure at which the first Korotkoff 80%.8 Oscillometers are generally safe and reliable; however,
sound is heard generally is accepted as systolic pressure (phase similar to manual intermittent techniques, care must be taken
I). The sound character progressively changes (phases II and to ensure proper cuff size.
III), becomes muffled (phase IV), and is finally absent (phase
V). Diastolic pressure is recorded at phase IV or V. However,
phase V may never occur in certain pathophysiologic states Invasive
such as aortic regurgitation. In 1905, Harvey Cushing7 recom- Continuous, invasive blood pressure is the gold standard used
mended the routine use of the noninvasive mercury sphygmo- to monitor blood pressure in critically ill patients. First
manometer in intraoperative management of patients. described 60 years ago,9 it is now considered the most
The sphygmomanometer method has several shortcom- reliable method, especially in hypotensive and vasoconstricted
ings. In particular, it is important that measurements are con- patients.10,11 The beat-to-beat and continuity of measurement
sistent from observer to observer in critical care. However, if allow early diagnosis and intervention.
the cuff is released too rapidly or if the observer fails to iden- This technique requires an intra-arterial cannula, fluid-
tify sounds, then incorrect measurements may be recorded. filled tubing, a pressure transducer and a microprocessor with
The sounds may be particularly difficult to hear in shock states a display. The pressure transducer is the key component of the
or with tachyarrhythmias, making inherent interobserver vari- system. It is a low-compliance diaphragm that moves in
ability relevant in this situation. Moreover, if the patient is response to pressure changes transmitted through fluid filled
shivering or edematous, the blood pressure may appear higher tubing. This movement is converted to electrical signals, most
than normal. A slight bend in the limb on which the cuff is commonly using a strain gauge set-up, based on the principle
placed also may lead to falsely elevated recordings, whereas that electrical resistance of a wire changes with changes of its
leaks in the pneumatic system may lead to falsely lower record- length. There are four wires or strain gauges that form a elec-
ings. Finally, cuff size and fit are also of critical importance. trical circuit based on the Wheatstone bridge. In this set-up,
The width of the cuff should be 20% greater than arm diam- with any movement of the diaphragm two wires are com-
eter. If the cuff is too narrow or too short the blood pressure pressed and the two other stretched, thus increasing the
will be overestimated. Many of these problems also are associ- sensitivity to diaphragm movement four times. Modern tech-
ated with continuous blood pressure measurement using nology uses semiconductors or silicone crystals instead of
oscillometry. wires. The MAP is calculated by integrating the area under the
178 Section II—Clinical and Laboratory Assessment

waveform, and the systolic and diastolic pressures calculated 50% of radial lines, with up to 15% resulting in complete
using an algorithm. The arterial pressure waveform is made occlusion.15 The main risk factors for occlusive thrombosis are
of multiple sine waveforms with different frequencies that can catheter size relative to vessel size, the number of arterial
potentially match the natural oscillatory frequency of the punctures needed to insert the cannula, length of catheter,
system and resonance and distort the signal. The monitoring indwelling time, presence of underlying atherosclerosis, and
system is designed to keep its frequency above the arterial use of vasopressors.16 Arterial catheterization also may result
waveform frequencies for accurate measurement. in arterial injury and lead to local hematoma or pseudoaneu-
The major problems associated with invasive blood pres- rysm formation.17 The risk of pseudoaneurysm formation is
sure monitoring are dampening and resonance; these can increased with local infection. Infectious complications of
affect accuracy of the blood pressure waveform and measure- arterial lines are rare but may be related to cannulation site,
ment. Dampening decreases the rate of signal change and and in particular the femoral site, where infection may occur
so leads to low pulse pressure with low systolic and high in up to 18% of cases.18 Therefore all arterial cannulation
diastolic pressure readings. Arterial cannula occlusion, air insertions should be conducted with full sterile technique.
bubbles, and soft tubing are common causes of an over- Because the oscillometric method is not standardized and
dampened signal. Interestingly, MAP accuracy is preserved measuring algorithms are protected by each manufacturer,
because of energy conservation; hence, if the waveform is it is difficult to make accurate comparisons between both
dampened, the mean pressure is accurate but the systolic and methods. Several studies have shown discrepant results
diastolic are not. between methods in critically ill patients. Oscillometry gener-
ally underestimates intra-arterial systolic blood pressure,19,20
as opposed to diastolic blood pressure, which is overestimated,
Strengths, Limitations, and Clinical particularly in hypertensive patients.21 MAP measurement is
Indications for Noninvasive and more reliable.22
The invasive blood pressure monitor must be zeroed and
Invasive Blood Pressure Monitors leveled, in the phlebostatic axis (at the level of the right
Both invasive and noninvasive blood pressure monitors may atrium), regardless of the site of the arterial cannula. Inade-
damage local tissue. Although noninvasive automated blood quate or incorrect zeroing results in incorrect diagnoses and
pressure measurement techniques are considered relatively inappropriate therapy. The bedside clinician should always
safe, repetitive cuff inflation may cause patient discomfort and check the “zero” before initiating antihypertensive or vaso-
persistent arterial occlusion that may result in pain, petechiae pressor therapy.
and ecchymoses, limb edema, venous stasis and thrombophle-
bitis, peripheral neuropathy, and rarely compartment syn-
drome (Table 19.1). These events occur more often after Pulse Pressure and Stroke
prolonged periods of excessively frequent cuff inflation-
deflation cycling and result from local trauma. Other factors
Volume Variability
that may contribute to complications include cuff misplace- A major reason to use invasive rather than noninvasive blood
ment across a joint, or repeated attempts to determine blood pressure monitoring in the ICU is to use respiratory cycle–
pressure in the presence of an artifact-producing condition varying measurements of stroke volume variability (SVV) and
such as involuntary muscle tremors. pulse pressure variation (PPV) during mechanical ventilation
Most invasive arterial lines are placed in the radial artery to help predict fluid responsiveness.23 Indeed some studies
because of easy access and protective collateral flow by the suggest that SVV may be the most accurate predictor of fluid
ulnar artery. This makes digital ischemia an uncommon com- responsiveness, and therapy that is based on SVV appears to
plication.12 Although other arterial sites are described, the be associated with improved hemodynamic stability.24,25 Arte-
femoral artery is the most common alternative, preferred by rial contour analysis has been validated against indirect mea-
some over the radial site because it may better reflect central sures of cardiac output using thermodilution or dye dilution
pressures and its luminal pressure is perhaps less affected by techniques.26 Positive intrathoracic pressure increases right
peripheral vascular disease or arteriosclerosis.13,14 The brachial ventricle (RV) afterload and reduces RV preload. Hence, on
and axillary arteries also may be used but have less collateral inspiration, RV stroke volume falls and results in a decrease
circulation to counteract end limb ischemia. The major com- in the subsequent left ventricle (LV) stroke volume, which
plication of arterial lines is local thrombosis. In the majority manifests during the next expiration. In addition, LV stroke
of arterial lines some degree of nonocclusive thrombosis volume may increase slightly during inspiration as a conse-
occurs without flow compromise. This is seen in more than quence of increased LV filling from enhanced pulmonary
venous return, increased LV compliance because of decreased
RV dimensions, decreased LV afterload, and external pressure
on the LV.27 Thus respiratory variations in arterial pressure
Table 19.1  Complications of Noninvasive reflect in large part the corresponding cyclic variations in LV
Blood Pressure Measurement stroke volume. This relationship has been demonstrated in
Pain directly measured (with transesophageal echocardiography
Petechiae and ecchymoses [TEE]) SVV28 or through measurement of arterial PPV, a sur-
Limb edema rogate of SVV.29 PPV is a good indicator of fluid responsive-
Venous stasis and thrombophlebitis ness as can be demonstrated when plotting stroke volume
Peripheral neuropathy
Compartment syndrome
against end diastolic volume on a Frank-Starling curve. Varia-
tions are exaggerated when the LV is functioning on the steep
Section II—Clinical and Laboratory Assessment 179

portion of the curve, whereby small changes in preload induce pressure contour may not always predict stroke volume
large changes in stroke volume.27 Patients on the flat portion because the pulse contour analysis omits the contributions of
of the curve are insensitive to these cyclic respiratory-related changing vascular tone and vascular bed distribution.39 In
changes in preload (Fig. 19.1).30 addition, several limitations summarized by the acronym SOS
In the mechanically ventilated patient, the effect of respira- need to be considered when using SVV: (1) S, small tidal
tion on blood pressure can be measured by adjusting the speed volume, for example, lung protection strategies and spontane-
of the arterial pressure to match the end-tidal carbon dioxide ous breathing activity; (2) O, open chest conditions; and (3)
(etCO2) (or airway pressure) trace on the monitor. A 15% S, sustained cardiac arrhythmia24,40 because all may influence
decrease in pulse pressure appears to be a sensitive indicator the accuracy of SVV evaluation of volume status or fluid
of fluid responsiveness.29 PPV or SVV has been shown to responsiveness. Thus respiratory cycle-varying measurements
predict fluid responsiveness in mechanically ventilated related to cardiac function may be useful tools in the manage-
patients,31 in cardiac surgery patients,27,32 in septic shock,29,33 ment of neurocritical care unit (NCCU) patients, particularly
with hemorrhage,34 and in elective brain surgery.35 However, those that have large fluctuations in circulating volume (e.g.
these findings are not universal and other studies have failed patients that have diabetes insipidus or those receiving aggres-
to demonstrate a clear relationship between SVV correlates sive osmotherapy with resultant diuresis).
and fluid responsiveness.36-38
Unlike bedside echocardiography, arterial pulse contour
analysis does not require an experienced operator and its use
Monitoring Flow and Volume
is associated with less patient discomfort. However, the arterial To maintain homeostasis, organs require blood flow sufficient
to match metabolic demands. There are two components to
flow: volume and pressure. There must be sufficient volume
in the circulation to ensure that the entire organ tissue receives
distribution, and this must be driven by adequate pressure to
c
ensure its timeliness. So in simple terms, flow is volume
divided by time. In general, perfusion pressure is proportional
d
a to flow, and pressure divided by flow equals resistance.
Stroke volume

However, if only pressure (in this case blood pressure) is mea-


sured, misleading conclusions about flow may arise: high
resistance results in normal pressure but low flow (Fig. 19.2).
Hence, blood pressure alone should be used with caution as a
b surrogate for tissue flow. For example, the kidney, an auto-
regulated organ, is highly sensitive to hypoperfusion, which is
manifested as oliguria. Hence, the combination of a “normal”
blood pressure and a urinary output greater than 0.5 mL/kg/
hr suggests adequate blood flow. However, oliguria may be
End diastolic volume associated with the stress response, and polyuria can result
Fig. 19.1  Stroke volume variability (SVV): on the steep part of the Frank- from diuretic administration, particularly mannitol.
Starling curve, the SV falls considerably on inspiration, from a to b, Evidence of end organ perfusion is helpful if present but
reflecting significant SVV. This is not the case in the “fuller heart,” on means to measure it are relatively limited in routine care.
the flat part of the curve, where the fall in SV, from c to d is much less. What are the alternatives? Tissue blood flow can be measured

Venous side Heart Arterial side Tissues

BP 110/70 mm Hg
Normovolemia
SvO2 = 70% O2 delivery
Normal NORMAL VASCULAR TONE
1200 mL/min
SV = 70 mL

Fig. 19.2  Impact of vasoconstriction on tissue


blood flow. When there is a decrease in cardiac
BP 110/70 mm Hg
Hypovolemia output there is intense vaso- and venoconstriction
SvO2 = 40% O2 delivery
Low INTENSE VASOCONSTRICTION that results in apparently normal blood pressure.
500 mL/min
SV = 70 mL However, tissue blood flow has fallen considerably,
manifested by an increased oxygen-to-extraction
ratio, and a fall in venous oxygen saturation
(SvO2). BP, Blood pressure.
180 Section II—Clinical and Laboratory Assessment

using either specific flow monitors or laboratory analysis of measurements to transpulmonary thermodilution using
end products of metabolism. Flow monitors measure stroke PiCCO-derived CO in patients with SAH. Pooled analysis of
volume and cardiac output or analogous markers of cardiac data showed close agreement between PAC and PiCCO-
function. Laboratory markers include by-products of metabo- derived values of cardiac index.
lism, such as lactate, or oxygen consumption using, for According to the Fick equation, CO (in liters per minute)
example, mixed venous oxygen saturation (SVO2). Other equals oxygen consumption (milliliters of O2 per minute)
monitor types that measure tissue perfusion (such as tissue divided by the arteriovenous O2 difference (milliliters of O2
oxygen electrodes) or metabolic activity (such as carbon per liter of blood). The NiCO (noninvasive cardiac output,
dioxide: capnometry) also may give information about flow. Novametrix) device uses the Fick equation by calculating CO
through changes in capnometry associated with re-breathing
CO2 through a dead space circuit within the breathing system.
Principles of Cardiac Pulse contour analysis is based on the concept that the
contour of the arterial waveform is proportional to the stroke
Output Monitoring volume, which can be estimated by the integral of the change
Because blood pressure monitoring allows for limited infer- in pressure from end systole to end diastole over time. Most
ence about blood flow, there is a widespread interest in mea- arterial contour methods relate an arterial pressure or pressure
suring stroke volume and cardiac output (CO). This, at least difference to a flow or volume change (PiCCO [Pulsion],
theoretically, provides information about circulating volume, PulseCO [LiDCO] and Modelflow [TNO/BMI]). Because the
cardiac preload, afterload, and contractility and its response estimated stroke volume is determined by the elastic and resis-
to volume loading, changes in pressure, and diuresis. This may tance patterns of the aorta, the devices are usually calibrated
be particularly useful in the brain-injured patient in whom using transpulmonary thermodilution, typically with a
cardiovascular dysfunction may accompany neurologic injury. femoral arterial catheter and a central venous line. Output of
Furthermore, the cardiovascular system matches blood flow these pulse contour systems is calculated beat-to-beat, but
to the body’s demands that may vary among patients and presentation of the data is typically with a 30-second window.
within the same patient over time. Therefore there is no single The most sophisticated arterial waveform analysis tool is
“normal” value of CO, and decisions about the adequacy of the esophageal Doppler monitor (EDM), which is placed tran-
CO need to be considered with other variables (e.g., tissue sorally in the esophagus. The Doppler ultrasound measures
perfusion and metabolic demand) in mind. the extent of distension of the thoracic aorta during systole,
There are two types of cardiac output monitors: those that from which stroke volume and CO are derived.
use thermodilution or the Fick principle to measure right
ventricular stroke volume and those that analyze arterial
waveforms to calculate CO. The thermodilution method uses Central Venous Pressure:
a change in pulmonary (or femoral) arterial pressure to cal-
culate CO, associated with injection of below body tempera-
What It Does and Does Not Do
ture fluid or the use of a heating coil. A thermistor in the artery Central venous catheters (CVCs) are 8- to 20-cm flexible cath-
generates a temperature change curve, the area under which eters that are placed in a central vein (subclavian, internal
is proportional to the CO, which can be calculated. This rep- jugular, or femoral), the tips of which lie in the superior vena
resents the gold standard method to measure CO; it is simple cava (SVC), inferior vena cava (IVC), or right atrium. They
and reproducible. Thermodilution CO is the method used to are inserted for a variety of reasons: the administration of
measure cardiac output in pulmonary artery (flotation) cath- vasopressors and hypertonic dextrose solutions (total paren-
eters (PACs, Swan-Ganz catheters) and the PAC-based con- teral nutrition), long-term venous access, and measurement
tinuous thermodilution methods (Vigilance, Edwards Life of central venous pressure (CVP) among them. Peripherally
Sciences [Irvine, CA]; Opti-Q, ICU Medical [San Clemente, inserted central catheters (PICCs) are longer lines typically
CA]; and TruCCOMS, AorTech [Rogers, MN]). Less invasive inserted in antecubital veins with their tips also in the SVC.
devices also using thermodilution (e.g., PiCCO [Pulsion PICC lines have replaced CVCs for drug and nutrition deliv-
Medical Systems, Irving, TX] and VolumeView [Edwards Life ery, but they are considered unreliable to monitor caval pres-
Sciences, Irvine, CA]), ultrasound (e.g., COstatus [Transonic sures. Nonetheless, even CVP measurements obtained from
Systems Inc., Ithaca, NY] or lithium chloride as an indicator CVCs remain a controversial method to estimate effective
(e.g., LiDCO [LiDCO Ltd., Cambridge, UK]).41 These devices circulating volume.45 There is no “normal” CVP, and small
use central venous (for calibration) and peripheral arterial changes in its magnitude may result from large changes in
cannulas (sensor) instead of needing pulmonary artery can- circulating volume. However, CVP measurement may still be
nulation. The PiCCO and VolumeView systems require a valuable because it may indicate a change in patient physiol-
femoral artery catheter. Application of all dilution methods ogy, and changes in the waveforms may reflect intrinsic cardiac
assumes three major conditions: (1) complete mixing of blood abnormalities (Fig. 19.3). CVP is useful as a trend and should
and indicator, (2) no loss of indicator between place of injec- be combined with some form of flow monitoring such as
tion and place of detection, and (3) constant blood flow. The SVO2, stroke volume, urine output, or mentation. In this para-
errors made are primarily related to the violation of these digm, the CVP is used to construct Starling curves.
conditions. Of the mentioned methods the transpulmonary Ventricular performance from volume loading usually
indicator dilution methods are, as the so-called continuous reaches a plateau around a CVP of 10 mm Hg.46 When CVP
cardiac output thermodilution methods, reasonably accepted is greater than 10 mm Hg, hypervolemia may be the cause,
in clinical practice42,43 and have been used in neurocritical but other causes such as acute pulmonary hypertension,
care. For example, Mutoh et al.44 compared PAC CO arrhythmia, RV dysfunction, cardiac tamponade, increased
Section II—Clinical and Laboratory Assessment 181

R arterial compliance is neither consistent nor constant and


there is great inter- and intrapatient variability. In addition,
aortic valve regurgitation may decrease the accuracy of arterial
pressure waveform analysis. Because compliance is the math-
ematical relationship between pressure and volume, external
P T
ECG calibration of the pressure signal with an alternative cardiac
tracing output technique is required. Pulse contour devices—Pulse
Q CO LiDCO (LiDCO+) (LiDCO Ltd.), PiCCO (Pulsion Medical
mm Hg

Systems, Munich, Germany), and VolumeView (Edwards Life


S Sciences)—combine peripheral arterial pulse contour analysis
a c to calculate stroke volume and use dilution or thermodilution
v
y
cardiac output measurement to calibrate the system. Devices
Jugular x
that analyze pulse waveforms also analyze and display pulse
tracing
pressure variability, which may be used to assess dynamic
preload and fluid responsiveness (in mechanically ventilated
Fig. 19.3  Central venous pressure (CVP) waveforms. The central patients).
venous waveform seen on the monitor reflects the events of cardiac PiCCO and VolumeView calculate cardiac output continu-
contraction; the central venous catheter transduces slight variations in ously from pulse contour analysis of the aortic waveform via
pressure that occur in the right atrium during the cardiac cycle and femoral arterial cannula using transpulmonary thermodilu-
transmits them as a characteristic waveform. tion. Both require a CVC (internal jugular or subclavian vein)
a wave: Increased atrial pressure during right atrial contraction. It
to calibrate cardiac output after ice cold fluid injections cen-
correlates with the P wave on an electrocardiogram (ECG).
c wave: Early ventricular contraction. The tricuspid valve bulges into
trally being sensed by the thermistor in the femoral artery.
the right atrium. It correlates with the end of the QRS segment on an The major advantage of this system over a PAC is that there
ECG. is no requirement to catheterize the right heart. The PiCCO
x descent: Atrial relaxation. It occurs before the T wave on an ECG. device measures the area under the aortic waveform; the sys-
v wave: Atrial filling. The right atrium progressively fills while the tolic area is identified as that part of the waveform proximal
tricuspid valve is closed. It occurs as the T wave is ending on an ECG. to the dicrotic notch, and this is proportional to the stroke
y descent: The tricuspid valve opens in diastole; blood flowing into the volume. Although beat-to-beat volumes are measured, these
right ventricle and the atrium empties. It occurs before the P wave on are averaged over 30 seconds to avoid inaccuracy associated
an ECG. with anomalous waveforms, interference, and extrasystoles.
The continued accuracy of these monitors depends on the
intrathoracic or intra-abdominal pressure (auto–positive frequency of calibration using thermodilution. The manufac-
end-expiratory pressure [PEEP], pneumothorax, abdominal turer recommends recalibrations every 8 hours, and more
compartment syndrome) should be considered and frequently in unstable patients.47
evaluated. In PiCCO and VolumeView monitors the temperature dif-
When CVP is elevated, interpretation of CVP waveforms ferential detected using the arterial thermistor is composed of
can be useful. It is important to use electrocardiography to a series of exponential decay curves as the cold injectate passes
identify or time CVP waveforms. For example, the Y descent through the various compartments of the circulatory system.
represents the decrease in atrial pressure at the beginning of Unlike the PAC, the receiver is located at a significant distance
diastole. It follows the V wave. The right atrial Y descent usually from the injector. Consequently, a series of exponential decay
is increased in restrictive physiology, pericardial constriction, curves are constructed as the injectate passes along. Because
volume-overloaded right heart or right heart ischemia, or the injectate is administered centrally and the temperature
infarction. In cardiac tamponade it is small. A large V wave is difference is measured in a proximal artery, the majority of
seen in tricuspid regurgitation; this may be associated with RV the temperature change occurs in the intathoracic compart-
failure secondary to volume overload or intense hypoxic pul- ment. As a consequence, it is possible to obtain an intratho-
monary vasoconstriction. In atrial fibrillation the A wave dis- racic blood volume index (ITBVI) and extravascular lung
appears. In atrioventricular dissociation or junctional rhythm water index (EVLWI). This is a significant advantage to the
a tall cannon A wave is produced. In patients with diastolic PiCCO device because pulmonary edema commonly accom-
dysfunction these arrhythmias can cause sudden hypotension, panies brain injury from acute lung injury, heart failure, or of
and analysis of CVP waveform can be diagnostic. neurogenic origin. Finally, in addition to SVV, the device also
purports to measure global end-diastolic volume, and so
permits the construction of Starling curves with volume titra-
Devices Used to Monitor Cardiac tion. To date, this particular device appears to correlate very
Output: Strengths and Weaknesses well with thermodilution techniques.26,48-50
Pulse Contour Methods
Systolic ejection results in the propulsion of a stoke volume Lithium Dilution
into the arterial tree and distention of the aorta and distal Lithium chloride in low doses is a nontoxic substance that is
arteries. A characteristic waveform is produced that reflects not metabolized. When injected, its concentration is easily
the stroke volume and elastic properties of the arterial wall. measured using a lithium selective electrode. Lithium dilution
The shape of the pulse waveform and the area under the curve cardiac output is calculated from the area under the
can be mathematically related to the cardiac output. However, concentration–time curve when injected from a central line
182 Section II—Clinical and Laboratory Assessment

and measured peripherally. Injection through the antecubital Currently, however, F/V is recommended in situations in
vein appears to be as accurate as a central line. Pulse CO which alternative monitors are unavailable. Another similar,
LiDCO (LiDCO+) combines pulse contour analysis with noncalibrated monitor, the MostCare system (Vytech, Padova,
lithium dilution calibration. Italy) uses the pressure recording analytical method (PRAM)
The major disadvantage of LiDCO+ is the injection of to analyze the arterial wave contour peripherally. Similar to
lithium and the requirement for calibration of cardiac output the other systems MostCare uses a proprietary algorithm to
at least every 8 hours. This is not required in PiCCO. In addi- derive cardiac and flow variables from contour analysis.58
tion, in patients that are hyponatremic or have recently
received neuromuscular blocking agents, the calibration data
may be inaccurate. Data are unreliable with aortic valve disease Esophageal Doppler
or balloon counterpulsation therapy. The major advantage of Esophageal Doppler has been widely used in perioperative
LCO+ is that no specialized central or arterial line is required. medicine (e.g., cardiac surgery), but has made minimal
inroads into ICUs, particularly NCCUs. The thoracic aorta is
located in proximity to the esophagus. The device uses Doppler
Noncalibrated Pulse Contour   ultrasound to measure aortic blood flow velocity, which allows
Waveform Analysis derivation of SV and CO. Insertion of the probe is rapid, and
FloTrac/Vigileo (F/V, Edwards Lifesciences) calculates con- clinically useful derived data are available within seconds. Sub-
tinuous cardiac output (CCO), stroke volume (SV), SVV, and stantial published data support the use of esophageal Doppler
systemic vascular resistance (SVR) using pulse contour analy- to guide goal-directed therapy.59 There are nonetheless several
sis obtained from a single sensor attached to an arterial line potential drawbacks: (1) patient positioning is awkward,
at any peripheral site. Unlike PiCCO or VolumeView, it does (2) there is a steep learning curve and significant interobserver
not require external calibration, a central line, or specialized variability, and (3) the esophageal Doppler, although small in
catheter. Since its introduction in 2006, the device has seen diameter and pliable, cannot be inserted into nonsedated and
several software upgrades, with progressive improvement in nonintubated patients or patients with known esophageal
accuracy. There were concerns that early software versions disease. This device is unlikely to find a place during neuro-
underestimated cardiac output when there was increased arte- surgical procedures because the head may be inaccessible
rial compliance and vasodilatory shock,51,52 but third- during surgery. To be used in NCCUs, placement problems
generation software was recently validated in septic patients.53,54 and observer error need to be overcome.
F/V combines rapid analysis in real time of the arterial pres-
sure waveform with demographic data (such as sex, age,
weight, and height) applied to an evolving algorithm to cal- Other Devices
culate cardiac output. Arterial pulsatility is directly propor- NiCO (noninvasive CO [Novametrix; Wallingford, CT]) is a
tional to stroke volume. The device appears capable of device that uses partial rebreathing of CO2 to derive cardiac
correcting for dynamic changes in vascular tone and compli- output in mechanically ventilated patients. The effects of
ance by analyzing skewness and kurtosis of the arterial wave- unknown ventilation or perfusion inequality in certain
form. These correction variables are updated every 60 seconds patients may explain why the performance of this method
and the arterial waveform is analyzed and averaged over 20 shows a lack of agreement between thermodilution and CO2-
seconds, thus eliminating artifacts, jitter, and premature con- rebreathing cardiac output. Furthermore, it may be unsuitable
tractions. Cardiac output is calculated using the arterial wave- for neurologically injured patients because its use may cause
form and the heart rate yielding apparently reliable measures fluctuation in CBF.
of SVV and hence fluid responsiveness. COstatus (Transonic Systems Inc., Ithaca, NY) calculates
Mayer et al.55 performed a meta-analysis of studies that cardiac output by using ultrasound technology to measure
have investigated F/V. Except for patients with irregular heart changes in blood ultrasound velocity and blood flow follow-
rhythms and aortic stenosis, cardiac output measurements by ing an injection of warm saline solution. Some studies have
F/V are similar to those obtained using transthoracic echocar- demonstrated that CO values measured with ultrasound indi-
diography in critically ill patients.56 Earlier studies demon- cator dilution techniques correlate well with PAC, and the
strated poor correlation between F/V and thermodilution COstatus system provides derived variables including total
methods; with newer software the correlation has improved end diastolic volume index.41,60
and with each software update accuracy continues to improve.57 Pulse dye densiometry (Nihon Kohden, Japan) is a tech-
It should be remembered that thermodilution methods, nique that uses transpulmonary dye dilution with transcuta-
although considered the gold standard, are not ideal devices neous signal detection to measure CO. The dye is injected
to compare with F/V; measurement intervals and averaging intravenously, and although the system is relatively noninva-
times are substantially longer with all thermodilution methods. sive, supporting data for this technology is limited. This
Hence it is possible that F/V is more sensitive to dynamic approach cannot be currently recommended.
changes in cardiovascular activity. Conversely, it is likely that Electrical bioimpedance uses constant electrical current
the F/V is severely limited in patients with aortic valve disease, stimulation to identify thoracic impedance variations associ-
those with indwelling intra-aortic balloon pumps, those ated with blood flow. Bioreactance has evolved from bio-
rewarming from hypothermia, and patients with intracardiac impedance and measures changes in frequency of electrical
shunts. It is conceivable that this particular device will become currents across the chest wall. These concepts are noninvasive
the dominant product in the market in future because of its and elegant in design, but to date there are few data to support
simplicity of placement, lack of observer error, continuity of the use of this approach in critical care.61,62 In addition, electri-
data, and the lack of need for secondary vascular access. cal interference may limit their potential use in the ICU.
Section II—Clinical and Laboratory Assessment 183

CHARACTERISTIC INTRACARDIAC PRESSURE WAVE FORMS


Right Heart Catheterization DURING PASSAGE THROUGH THE HEART

and Pulmonary Artery 40


mm Hg
Pressure Monitoring RA RV PA PCW

The balloon tipped flow-directed PAC was introduced in the


early 1970s and quickly became widely used in critical care, 20
under the moniker of its inventors Swan and Ganz.63 The
bedside ability to “float” a catheter into the heart originated
on a serendipitous observation by cardiologist H. J. Swan when
he observed a sailboat and imagined a parachute-like device 0
catching blood flow and dragging a catheter with it. Swan
subsequently worked with William Ganz to incorporate cardiac
output computation capability using thermodilution into the
catheter itself.64 Despite decades of controversy65,66 about
whether information provided by a PAC can help influence
outcomes in ICU patients, use of the PAC has declined signifi-
cantly, particularly outside of North America. Nevertheless the
PAC remains the gold standard hemodynamic monitor in
clinical use to assess cardiac output, and despite its limitations Fig. 19.4  Placement of a pulmonary artery catheter. The different
most other devices are compared to it.67,68 In addition a PAC pressure waveforms (upper panel) guide the clinician to the location of
also can provide information about pulmonary artery pres- the tip of the catheter. PA, pulmonary artery; PCW, pulmonary capillary
wedge; RA, Right atrium; RV, right ventricle.
sures, right-sided and left-sided filling pressures, and SVO2. In
general, its use should be reserved for more complex patients.
The PAC directly measures intrathoracic, intravascular, and assumptions may not be valid and so can make the use of
intracardiac pressures and is able to display their pressure PCWP estimation of preload faulty.69
waveforms. It also provides measurement of CO and SVO2. Nonetheless, a PAC can reliably measure PAPs and cardiac
Measurement of these hemodynamic indices is continuous output using the thermodilution method, and is the only
and mostly accurate and clearly superior to the clinical assess- device that accurately measures true core temperature and
ment of these indices by physical examination in critically ill mixed SVO2. Moreover, measurements are independent of
patients.65 cardiac pathology, particularly of the aortic valve and cardio-
The PAC can be inserted through a wide bore introducer in vascular anomalies distal to it. Thus PAPs also may provide
any central vein, and “floated” through the right atrium and some indication of volume status on their own. Normal sys-
ventricle into one of the pulmonary arteries using a balloon tolic pulmonary pressures mirror RV systolic pressures in the
attached to the distal part of the catheter. Usually the device absence of pulmonic valve disease and normally range between
is advanced until the balloon is “wedged” into a proximal 20 and 30 mm Hg. The normal mean pulmonary artery pres-
arteriole, at which point flow ceases and the port distal to the sure (mPAP) at rest is 14 plus or minus 3 mm Hg. Elevation
balloon is able to measure the static pressure in the fluid in PAPs may result from hypervolemic states but also from
column that equilibrates across the pulmonary capillaries and pulmonary hypertension, LV failure, and mitral valve disease.70
left atrium. Thus because there are no valves between the In the correct hands the PAC is an invaluable tool to diagnose
pulmonary capillaries and the left atrium, this pulmonary and monitor treatment of pulmonary hypertension and right
capillary wedge pressure (PCWP) is a reflection of left atrial heart failure (Fig. 19.5). However, the correct interpretation
pressure (Fig. 19.4). of PAC-derived hemodynamic data and the subsequent thera-
However, measured pulmonary artery pressures (PAPs) also peutic intervention remain major challenges and are the main
vary depending on the anatomic position of the PAC tip. The reasons for complications associated with these devices.71
lung is anatomically divided in three West zones. The West Many clinicians believe that PCWP reliably reflects preload,
lung zone 1, or the upper, least dependent area or middle area and is useful to construct Starling curves. This is unlikely.72
(zone 2) has alveolar pressures that are greater than PAPs. The pressure-volume relationship of the LV changes dynami-
Thus in these lung regions blood flow may be partially or cally depending on clinical circumstances, and pressure figures
completely interrupted and thus prevent pressure equilibra- are altered by changes in ventricular and atrial compliance,
tion across the pulmonary capillary system during wedging. ventricular systolic and diastolic function, valvular function,
Only in zone 3 does PAP always surpass alveolar pressure, rhythm and heart rate, afterload, intrathoracic pressures, and
making blood flow constant, thus making it the optimal ana- abdominal pressures. They also change with therapeutic inter-
tomic position for the PAC tip to be placed in and wedged. ventions.45,72,73 Furthermore, PAC pressures alone should not
In addition, using PCWP as a surrogate of left atrial pressure be used to guide volume resuscitation, and the inappropriate
assumes a pulmonary capillary and venular resistance of zero. use of these measures may have resulted in the lack of pub-
This assumption is faulty because the capillary accounts for lished beneficial outcome data associated with this device
up to 40% of total pulmonary vascular resistance. In critically to date.
ill patients in whom pulmonary constriction occurs secondary Two types of PAC are available in clinical practice: intermit-
to hypoxemia, this value may rise to even higher levels. In tent bolus (older and less expensive) and continuous cardiac
conditions such as shock, respiratory failure, sepsis, and output devices. The noncontinuous model requires intermit-
acute respiratory distress syndrome (ARDS), the various tent injection of cooled crystalloid to measure cardiac output
184 Section II—Clinical and Laboratory Assessment

Hemodynamically with a thermistor located downstream within the pulmonary


unstable patient artery. The data are averaged over time (about 10 minutes) to
produce an accurate series of measurements; hence there may
be a delay of several minutes before the device records major
Is the patient hypoxic hemodynamic changes (i.e., this approach may be limited in
or is RV dysfunction its use in a very unstable patient). Further, the data may be
suspected? misleading when there are rapid changes of body temperature
such as following cardiopulmonary bypass.
Yes No PAP and PCWP measurements can provide useful informa-
tion. Dynamic PAP changes reflect changes in patient condi-
Is the patient at risk for Yes Is femoral arterial tion or changes induced by therapeutic intervention (e.g., a
malignant arryhthmias? cannulation safe?
pulmonary vasodilator). In the NCCU the PAC may be useful
No to measure global hemodynamic function and the pulmonary
Yes No
arterial response to vasopressor therapy. CBF depends on LV
Insert a pulmonary Insert PiCCO+ Attach FloTrac/
stroke volume, particularly when there is autoregulatory dys-
artery catheter or Lidco+ Vigileo function. Vasopressors increase both systemic and pulmonary
vascular resistance, and profound pulmonary hypertension
Fig. 19.5  Decision tree for placement of a hemodynamic monitor. may result in RV dysfunction, secondary to excessive afterload.
RV, Right ventricle. Thus in patients who require high doses of vasopressors to
maintain CPP, pulmonary arterial catheterization can be used
to guide both fluid and vasopressor therapy. Finally, the pres-
sure waveforms on the PAC can be used to diagnose cardiac
pathology. For example, a large “v” wave on a PAP tracing may
indicate acute mitral regurgitation.

Bedside Cardiac Imaging


The Current Role of Echocardiography  
in Critical Care
Echo reflection, the basis of echocardiography technology was
first described by Lazzaro Spallanzani when evaluating the
effect of sound on navigation practices of bats.74 Langevin
developed sound navigation and ranging (SONAR) based on
the same concept in the search for methods to detect enemy
submarines in World War I.74 In 1946 the French physicist
André Denier suggested that ultrasound could be used to look
at internal organs.75 Inge Edlerm, a Swedish internist, described
the first use of ultrasound on the heart when attempting to
distinguish coexisting mitral regurgitation in stenotic valves.76
Fig. 19.6  Swan-Ganz catheter (note the lack of visible distal heating coil, After collaborating with local physicist Hellmuth Hertz for
which would be used for the continuous cardiac output [CCO] method). several years, they were able to generate the first moving
images of the heart, which were presented at the Lund Royal
Physiological Society in 1954.77 Multiple incremental improve-
ments in ultrasound techniques have occurred since: two-
using the area under the thermodilution curve (Fig. 19.6) and dimensional (B-mode), motion (M-mode), Doppler (velocity
despite its limitations is still considered the standard reference of motion), and color Doppler (direction of motion) technol-
for other devices. ogy. Frazin et al. first introduced transesophageal echocar-
Usually three injections are performed, and an average of diography (TEE) in 1976.78
the three is recorded. The cold bolus is injected into the right In critically ill populations echocardiography now greatly
atrium, and the temperature change in the pulmonary artery expands the capabilities of intensivists to accurately and
is used to calculate cardiac output. This system is highly accu- rapidly diagnose the cause of hemodynamic instability. With
rate, and there is surprisingly little interobserver variability. advances in technology, ultrasound units now are smaller,
SVO2 can be measured but requires a laboratory analysis of multipurpose with improved image quality, and less expen-
the collected sample. This measurement is also highly accurate sive. In the ICU echocardiography is a complementary diag-
and reproducible. nostic tool that, to be fully understood and appropriately used,
Continuous CO PACs were introduced to reduce the work- requires training and experience, but in recent years the use
load of the bedside nurse and provide “continuous data.” This of and training in echocardiography by intensivists has
is misleading because they use a random sequence of tem­ increased79-81 and appropriate use guidelines have been pub-
perature changes generated by thermal filament (Vigilance, lished.82 Furthermore, there is a general agreement among
Edwards Life Sciences) or thermal coil (OptiQ, ICU Medical, critical care societies worldwide that general critical care ultra-
San Clemente, CA) that is located in the superior vena cava, sound and “basic” critical care echocardiography should be
Section II—Clinical and Laboratory Assessment 185

part of the curriculum of ICU physicians.83 This is important majority of patients are already intubated and can be sedated
because even basic knowledge of bedside echocardiography for the procedure.91 However, the possibility of esophageal
may provide life-saving diagnosis in certain conditions (e.g., disease, recent esophageal or gastric surgery, and an unstable
severe heart dysfunction, large pericardial effusion, or severe cervical spine are contraindications to TEE.
hypovolemia).84
The role of echocardiography in neurocritical care is just
beginning to be elucidated, although it is a safe, noninvasive Utility of Echocardiography  
bedside test that provides information about cardiac struc- in Intensive Care Units
ture, function, and hemodynamics. Different physiologic
conditions and use of vasopressors make critical care echocar- Ventricular Function
diography interpretation challenging. Furthermore, most of LV dysfunction in critically ill patients is common and may
the quantitative echocardiographic evaluations have been be caused by ischemia, sepsis, or hyperadrenergic states (“neu-
validated only in cardiac surgery and the cardiology popula- rogenic shock”). When the LV becomes dysfunctional, its
tion. In these patients echocardiography plays a key role to diameter progressively increases and the diastolic volume–to–
help assess LV and RV function, chamber size, and cardiac stroke volume ratio increases: expressed as a percentage, this
valve dysfunction, and in cardiac surgery and high-risk non- means that the fraction of end-diastolic blood that is ejected
cardiac surgery, abnormal echocardiographic findings are an per cycle is reduced. Ejection fraction (EF) can only be reliably
independent predictor of poor outcome.85 Echocardiography measured by echocardiography, and it is useful to correlate
also may be helpful in preload assessment when intravascular filling pressures and stroke volume, using a hemodynamic
volume determination is difficult. In the ICU echocardio- monitor (such as a PAC) with EF. MAP may appear normal
graphic evaluation of unexplained hypoxemia, intracardiac despite a severe reduction in ventricular function because this
versus intrapulmonary shunt, or diagnosis of cardiac tampon- may be impaired further by vasoconstrictor use to increase
ade and aortic dissection is becoming a first-line, bedside CPP. The size of the ventricle is measured using either M-mode
diagnostic tool. or two-dimensional (2D)-mode. Hence, in neurologic disease,
In critical care up to 25% of bedside studies, particularly of when vasodilators cannot be safely used, echocardiography
transthoracic echocardiography (TTE), may be of limited may guide inotropic therapy. Echocardiography also may
diagnostic quality or nondiagnostic. Contrast echocardiogra- unveil regional wall motion abnormalities, usually associated
phy, using microspheres coupled with contrast-specific ultra- with myocardial ischemia.
sound imaging modalities, may help overcome this limitation RV dysfunction also is very common in critically ill patients.
and so avoid the need for TEE.86 Furthermore, the concept of Pulmonary embolism (PE) and ARDS are the main causes.92
limited TTE has evolved in the last few years and this has In addition, excessive mean airway pressure, fat embolism, and
greatly facilitated the use of echocardiography to guide goal- elevated pulmonary vascular resistance also cause increased
directed therapy in the ICU.87,88 Focused, goal-directed echo- RV afterload and RV strain. The right ventricle’s complex
cardiography (TTE or TEE) usually can be performed in a few pyramidal shape makes quantitative and qualitative evalua-
minutes and may include qualitative assessment of LV and RV tion of systolic function very difficult. Although the PAC can
function, an estimate of aortic valve gradient, RV systolic pres- be useful to diagnose RV failure, echocardiography is required
sure, and intravascular volume status among other variables. to diagnose the underlying cause. 2D echocardiography evalu-
ation of RV size, shape, kinetics of septum, and RV free wall
along with the evaluation of severity of tricuspid regurgitation
Transthoracic Versus Transesophageal (TR) by color Doppler are integral components of a complete
Echocardiography evaluation. RV size is generally compared to LV size. In a four-
TTE involves the placement of an ultrasound transducer on chamber view the ratio between RV and LV end-diastolic area
the epigastrium and between ribs to obtain sonographic is measured. A diastolic ventricular ratio greater than 0.6 sug-
images of the heart, aorta, and vena cava. Because of its safety, gests moderate, and a ratio greater than 1, severe dilation.93
reliability, and rapidity, TTE should be the first-line modality
used in most ICU patients to assess hemodynamic instabil-
ity.89 Poor or limited acoustic windows may limit image Hypoxemia and Intracardiac Shunt
quality. However, new technology, harmonic imaging, and The incidence of patent foramen ovale is about 30%. In this
new echo contrast products have significantly improved echo setting of RV dysfunction right atrial pressure can exceed the
signal acquisition.90 TEE is conducted through a miniaturized left atrial pressure. A foramen ovale may open, causing the
transducer mounted laterally on the tip of an endoscopic anatomic substrate for significant intracardiac shunt that can
shaft. Under direct vision and with continuous electrocardi- be detected by contrast or agitated saline echocardiogram.
ography, the endoscope is inserted into the esophagus and After injection of agitated saline in a peripheral or central vein,
positioned behind and under the heart to acquire ultrasono- the appearance of contrast or microbubbles in the left atrium
graphic images of the heart in different angles and planes. within three cardiac cycles is diagnostic of a right-to-left
Most TEE transducers can rotate within their casing by 180 shunt.
degrees (multiplanar) and the shaft may be flexed laterally or
anteroposteriorly. TEE is indicated when a TTE study is not
adequate or in special circumstances, such as when evaluating Assessment of Cardiac Output
aortic dissection or endocarditis of prosthetic valves, or in Thermodilution CO measurement is not always accurate in
excluding intracardiac thrombus presence before elective car- critically ill patients. Very low or very high CO, severe TR,
dioversion. TEE can be used easily in ICUs because the rapid temperature changes, or intracardiac shunt can cause
186 Section II—Clinical and Laboratory Assessment

false readings. In these conditions echocardiography can rela- effective circulating volume is adequate.106,107 Similarly, respi-
tively reliably measure SV and thus CO.94 The most common ratory variation of inferior vena cava diameter (dIVC) mea-
technique is Doppler-derived instantaneous blood flow mea- sured by bedside ultrasonography can help indicate fluid
surement through a conduit (LV outflow tract, pulmonic or responsiveness in mechanically ventilated patients. For
mitral valve). For example, if the ultrasound beam is directed example, in SAH patients Moretti and Pizzi108 observed that a
along the aorta, part of the signal is reflected back by the cutoff dIVC greater than16% was 70% sensitive and 100%
moving red blood cells. The frequency is different, and this specific as a predictor of fluid responsiveness. Esophageal
Doppler shift then is used to calculate the flow velocity and Doppler echocardiography also has yielded indicators of
volume and cardiac output. SV is equal to the product of preload. However, stroke volume as measured by esophageal
cross-sectional area (CSA) of the conduit, determined by 2D Doppler may not be reliable because of significant interob-
echo, and integration of instantaneous blood flow, velocity server variability.109 On the other hand, flow time correction
time integral (VTI), through the conduit. (FTc) or the duration of the aortic velocity signal corrected
for heart rate is considered a promising static indicator of
CSA = Diameter of conduit (D) squared X (π/4)
cardiac preload.110 In a study of neurosurgical patients, FTc
SV = CSA × VTI values were predictive of greater than10% increases in stroke
volume index from a fluid challenge.111 Other studies, however,
SV × Heart rate (HR) = CO are less clear on the correlation between FTc and fluid
status.112,113
CO = CSA × VTI × HR
The technique does not allow continuous monitoring and
may not be valid when there is an intra-aortic balloon pump. Laboratory Analysis of Global
Cardiovascular Function
Volume Status SVO2
Knowledge about volume status and fluid responsiveness is When blood flows through tissues, oxygen is removed and
essential in the NCCU. However, traditional measures of these venous blood returns in an oxygen-depleted state to the right
variables—static preload filling-pressure markers such as CVP heart. The quantity of oxygen in this mixed venous blood has
or pulmonary artery occlusion pressure (PAOP)—may not be been used as a surrogate marker of tissue blood flow, and is
ideal tools to guide fluid management.31 ICU studies show that most easily measured by using an optical probe that continu-
echocardiography, including a transthoracic-focused rapid ously measures mixed SVO2. Ideally SVO2 that reflects the
echocardiographic examination can help estimate volume balance between oxygen delivery (DO2) and consumption
status or fluid responsiveness or guide fluid therapy such as in (VO2) (i.e., the “adequacy” of tissue oxygenation) is measured
septic shock.95-97 Systolic obliteration of the LV is a good indi- with a PAC. If there is no PAC, a CVC can be used to measure
cator of inadequate preload. Besides hypovolemia, right heart oxygen saturation in the superior vena cava (ScVO2). There
dysfunction can be the culprit. On the other hand, ventricle are, however, important differences between SVO2 and
dilation, defined by left ventricular end-diastolic LV diameter, ScVO2.114 Furthermore, the accuracy of ScVO2 depends on the
can be just a sign of chronic heart failure and some patients position of the CVC tip; when located high in the superior
can still respond to volume challenge.98 LV dimensions are vena cava it is less reliable, whereas when adjacent to the right
measured directly to provide the primary preload parameters atrium it is very close to SVO2. Normally, venous blood is 70%
of left ventricular end-diastolic area (LVEDA) or volume to 80% saturated with oxygen. When there is increased oxygen
(LVEDV) with the latter considered a superior determinant of uptake by tissues, or inadequate blood flow, the SVO2 falls.
fluid status. The modified Simpson formula is one of two Hence where there is a clinical suggestion of tissue hypoperfu-
methods to calculate LVEDV by using the assumption that the sion in the absence of hypotension (cold clammy extremities,
cardiac chamber is a sum of multiple cylinders in a truncated oliguria), SVO2 is a robust marker of tissue blood flow. The
ellipse. The alternative method of LVEDV calculation uses use of venous oximetry in shock and severe sepsis is a feature
semiautomated disk summation to calculate volume from of goal-directed resuscitation and has been validated by Rivers,
multiple diameters perpendicular to a longitudinal ventricular Shoemaker, and others.115-117 However, the role of therapy to
axis. The latter method is more expeditious with fewer required increase mixed or central venous oxygen saturation as a
variations in probe position (short-axis view only) and is the marker of adequate tissue perfusion or fluid responsiveness or
method of choice for volume calculation in the presence of as a guide to fluid therapy in ICU patients still needs to be
distorted ventricular anatomy.99 Several studies have demon- fully elucidated.118-121
strated a persistent underestimation of LV volumes by the disk
summation method.100,101 Not all reports favor LVEDV as an
indicator of fluid responsiveness. Whereas some studies indi- Lactate
cate significant LVEDV differences in responders and nonre- The presence or absence of metabolic acidosis has been widely
sponders to a fluid challenge,102,103 others find no difference used as a surrogate endpoint of resuscitation for critically ill
compared with those measured with right arterial pressure or and trauma patients since the 1970s, when it was observed that
PCWP.28,104,105 In ventilated patients, superior vena cava col- hypothermia, coagulopathy, and acidosis constituted a “triad
lapsibility also should be systematically gauged as an indicator of death” in trauma victims.122 The mechanism of acidosis is
of fluid responsiveness. With positive intrathoracic pressure, presumed to be cellular oxygen debt secondary to hypoperfu-
the vena cava demonstrates greater collapsibility or inspira- sion or hypoxia. The anion implicated in this acidosis is
tory diameter decrease when volume status is low than when lactate. The ability to measure serum lactate was not
Section II—Clinical and Laboratory Assessment 187

universally available until the 1990s, and so a surrogate, the


base deficit,123 has been used widely. Care should be taken
Conclusion
using this tool, and inferences about its validity should be Monitoring of volume status and cardiac function plays a fun-
made in the context of quantitative acid-base chemistry.124-127 damental role in the care of critically ill patients including
In the setting of acute trauma there may be multiple acidifying those managed in the NCCU, in part because a variety of acute
and alkalinizing processes that occur simultaneously depend- neurologic disorders can lead to cardiac instability (e.g., Takot-
ing on the volume and nature of intravenous fluids.128 A subo cardiomyopathy) after aneurysm rupture or changes in
“normal” base deficit or excess may hide potentially dangerous volume status (e.g., diabetes insipidus). In addition, hemody-
acid-base disturbances, and base deficit does not reliably namic manipulation (e.g., induced hypertension or hypervol-
reflect lactate in the emergency setting.124,129,130 emia) is an important intervention to regulate CPP and CBF.
Lactic acidosis on admission to the emergency department However, fluid therapy or induced hypertension can aggravate
is a marker of illness severity. The magnitude of acidosis and outcome through exacerbation of lung function that already
of serum lactate elevation is associated with patient out- may be compromised.3,4,148 There are a variety of methods,
comes.122,131-133 The speed of lactate clearance from the circula- both invasive and noninvasive, that can be used to monitor
tion also is a prognostic indicator.131,134-136 This led to the volume and cardiac status including: clinical evaluation and
concept of hyperaggressive volume resuscitation to “wash out laboratory analysis, arterial pressure pulse contour and wave-
lactate,” under the assumption that serum lactate is a marker form analysis, CVCs, PACs, transpulmonary thermodilution,
of tissue hypoperfusion. For example McNelis et al. observed and bedside echocardiography or ultrasound among others.
that lactate clearance was more rapid in surgical intensive care There has been a trend to use less invasive devices to measure
unit (SICU) survivors. However, oxygen delivery and con- hemodynamic variables.41 In addition, TTE and even TEE,
sumption was similar in survivors and nonsurvivors.135 This although traditionally the domain of cardiologists, are used
indicates that although admission lactate and lactate clearance more frequently in critical care and in the operating room in a
may be independent factors associated with mortality, the focused goal-directed manner.88 These various monitors may
mechanism of hyperlactemia may not necessarily be hypoper- be used alone or in combination preferably with integration of
fusion. In addition, several lines of evidence suggest that the variables. For example, a hypotensive patient with low
lactate itself may be harmless137-139 or a necessary intermediate cardiac output differs in diagnostic and therapeutic possibili-
in wound healing and repair because it may enhance collagen ties from a hypotensive patient with increased cardiac output.
production and deposition and angiogenesis.140 Consistent However, the optimal system to monitor a patient may vary
with this, oxygen levels appear to have little effect on wound over the patient’s hospital course and depends on the indi-
lactate137 and whether using serum lactate as a marker of tissue vidual patient, his or her pathology, and the problems that
perfusion to guide therapy helps improve outcome is still to may arise. More important, as with any monitor, the informa-
be fully elucidated.118 Similarly, the role of muscle or subcuta- tion obtained needs to be interpreted and applied correctly.149
neous microdialysis of lactate, pyruvate, and the lactate-to-
pyruvate ratio to guide resuscitation requires further study.141,142
There is emerging evidence that lactic acidosis in trauma Key Points
and critical illness may not be associated with hypoxemia but
also may result from epinephrine-driven aerobic glycoly- 1. Cardiovascular monitoring is an essential component of
sis.139,143 This results from cyclic adenosine monophosphate neurocritical care.
(cAMP)–medicated Na+-K+ ATPase activity that is driven 2. Blood pressure monitoring can be achieved using oscil-
by beta-adrenoceptor activation.144 Administration of beta- lometry or by invasive arterial line placement. Both types
adrenergic agents increases tissue lactate levels; administration of monitoring can produce errors in measurement. Because
of antiadrenergic agents reduces tissue lactate levels.139 of continuity and accuracy of monitoring very low blood
Hyperglycemia can aggravate ischemic brain damage pressures, the intra-arterial measurement represents the
because of increased anaerobic glycolysis, lactate production, gold standard in critically ill patients.
and tissue acidosis.145 However, the brain is glucose depen- 3. Arterial lines reliably measure mean arterial pressure and
dent, and glycolysis supplies energy for metabolism during can be used to estimate fluid responsiveness through pulse
hypoxemia. In addition, lactate may be a source of energy pressure variability. However problems of resonance and
during the postischemia state. Thus there is a “glucose paradox dampening may limit interpretation.
of cerebral ischemia” in which increased energy delivery 4. Blood pressure alone is not always a good measure of tissue
appears to be associated with worsened outcomes.146 However, blood flow. Some form of tissue flow monitoring is
there also is evidence to suggest that elevated brain lactate can required in patients at risk of low flow state.
be associated with better recovery after acute brain injury 5. Several stroke volume monitors that perform pulse contour
predominantly because of hyperglycolysis rather than analysis are available. Each of these devices appears to be
hypoxia.147 reliable and have varying levels of invasiveness.
Alternatively the brain injury may be associated with ele- 6. The pulmonary artery catheter remains the gold standard
vated serum cortisol because elevated blood glucose and device for hemodynamic monitoring. All hemodynamic
lactate is associated with stress and stress is associated with devices require a high level of expertise to correctly inter-
increased blood corticosteroid levels.146 In animal studies the pret acquired data.
timing of hyperglycemia before and after ischemic brain inju- 7. Echocardiography is an important diagnostic tool in hemo-
ries alters outcomes. The application of corticosteroid inhibi- dynamically unstable patients. Transesophageal echocar-
tors may reduce, whereas the application of steroids may diography provides better image quality than transthoracic,
increase the severity of brain injuries.146 but it is more invasive.
188 Section II—Clinical and Laboratory Assessment

8. Mixed venous oxygen saturation is an easily performed 27. Kramer A, Zygun D, Hawes H, et al. Pulse pressure variation predicts fluid
responsiveness following coronary artery bypass surgery. Chest 2004;126(5):
and useful measure of tissue blood flow.
1563–8.
9. The presence of tissue acidosis can help to identify 28. Feissel M, Michard F, Mangin I, et al. Respiratory changes in aortic blood
critically ill patients, and apportion risk. However, it is velocity as an indicator of fluid responsiveness in ventilated patients with
important to avoid over interpreting the mechanism of septic shock. Chest 2001;119(3):867–73.
hyperlactatemia. 29. Michard F, Boussat S, Chemla D, et al. Relation between respiratory changes
in arterial pulse pressure and fluid responsiveness in septic patients with
acute circulatory failure. Am J Respir Crit Care Med 2000;162(1):134–8.
References 30. Michard F, Lopes MR, Auler JO Jr. Pulse pressure variation: beyond the fluid
management of patients with shock. Crit Care 2007;11(3):131.
1. Ramasamy I. Biochemical markers in acute coronary syndrome. Clin Chim 31. Marik PE, Cavallazzi R, Vasu T, et al. Dynamic changes in arterial waveform
Acta 2011;412(15-16):1279–96. Epub 2011, Apr 8. derived variables and fluid responsiveness in mechanically ventilated
2. Noveanu M, Mebazaa A, Mueller C. Cardiovascular biomarkers in the ICU. patients: a systematic review of the literature. Crit Care Med 2009;37(9):
Curr Opin Crit Care 2009;15(5):377–83. 2642–7. Review.
3. Contant CF, Valadka AB, Gopinath SP, et al. Adult respiratory distress 32. Auler JO Jr, Galas F, Hajjar L, et al. Online monitoring of pulse pressure
syndrome: a complication of induced hypertension after severe head injury. variation to guide fluid therapy after cardiac surgery. Anesth Analg 2008;
J Neurosurg 2001;95:560–8. 106(4):1201–6, table of contents.
4. Boyd JH, Forbes J, Nakada TA, et al. Fluid resuscitation in septic shock: a 33. Marx G, Cope T, McCrossan L, et al. Assessing fluid responsiveness by stroke
positive fluid balance and elevated central venous pressure are associated volume variation in mechanically ventilated patients with severe sepsis. Eur J
with increased mortality. Crit Care Med 2011;39(2):259–65. Anaesthesiol 2004;21(2):132–8.
5. Fletcher JJ, Bergman K, Blostein PA, et al. Fluid balance, complications, and 34. Ornstein E, Eidelman LA, Drenger B, et al. Systolic pressure variation
brain tissue oxygen tension monitoring following severe traumatic brain predicts the response to acute blood loss. J Clin Anesth 1998;10(2):
injury. Neurocrit Care 2010;13(1):47–56. 137–40.
6. Riva-Rocci S. Un Nuovo sfigmomanometro. Gazzeta Medica di Torino 1896; 35. Berkenstadt H, Margalit N, Hadani M, et al. Stroke volume variation as a
47:981–96. predictor of fluid responsiveness in patients undergoing brain surgery.
7. Cushing H. On routine determination of arterial tension in operating room Anesth Analg 2001;92(4):984–9.
and clinic. Boston Med Surg J 1903;148:250. 36. Wiesenack C, Prasser C, Rodig G, et al. Stroke volume variation as an
8. Bigatello LM, Schmidt U. Arterial blood pressure monitoring. Minerva indicator of fluid responsiveness using pulse contour analysis in
Anestesiol 2003;69(4):201–9. mechanically ventilated patients. Anesth Analg 2003;96(5):1254–7.
9. Peterson L, Dripps R, Risman G. A method for recording the arterial pulse 37. de Waal EE, Rex S, Kruitwagen CL, et al. Stroke volume variation obtained
and blood pressure in man. Am Heart J 1949;37:771. with FloTrac/Vigileo fails to predict fluid responsiveness in coronary artery
10. Cohn JN. Blood pressure measurement in shock. Mechanism of inaccuracy bypass graft patients. Br J Anaesth 2008;100(5):725–6.
in ausculatory and palpatory methods. JAMA 1967;199(13):118–22. 38. Sander M, von Heymann C, Foer A, et al. Pulse contour analysis after
11. O’Rourke MF. Impact pressure, lateral pressure, and impedance in the normothermic cardiopulmonary bypass in cardiac surgery patients. Crit
proximal aorta and pulmonary artery. J ApplPhysiol 1968;25(5):533–41. Care 2005;9(6):R729–34.
12. Slogoff S, Keats AS, Arlund C. On the safety of radial artery cannulation. 39. Pinsky MR. Probing the limits of arterial pulse contour analysis to predict
Anesthesiology 1983;59(1):42–7. preload responsiveness. Anesth Analg 2003;96(5):1245–7.
13. Chauhan S, Saxena N, Mehrotra S, et al. Femoral artery pressures are more 40. Mahjoub Y, Pila C, Friggeri A, et al. Assessing fluid responsiveness in
reliable than radial artery pressures on initiation of cardiopulmonary bypass. critically ill patients: false-positive pulse pressure variation is detected by
J Cardiothorac Vasc Anesth 2000;14(3):274–6. Doppler echocardiographic evaluation of the right ventricle. Crit Care Med
14. Mignini MA, Piacentini E, Dubin A. Peripheral arterial blood pressure 2009;37(9):2570–5.
monitoring adequately tracks central arterial blood pressure in critically ill 41. Vincent JL, Rhodes A, Perel A, et al. Clinical review: update on
patients: an observational study. Crit Care 2006;10(2):R43. hemodynamic monitoring—a consensus of 16. Crit Care 2011;15(4):229.
15. Martin C, Saux P, Papazian L, et al. Long-term arterial cannulation in ICU 42. Benington S, Ferris P, Nirmalan M. Emerging trends in minimally invasive
patients using the radial artery or dorsalis pedis artery. Chest 2001;119(3): haemodynamic monitoring and optimization of fluid therapy. Eur J
901–6. Anaesthesiol 2009;26(11):893–905. Review.
16. Clark VL, Kruse JA. Arterial catheterization. Crit Care Clin 1992;8(4):687–97. 43. Jabot J, Monnet X, Bouchra L, et al. Cardiac function index provided by
17. Ganchi PA, Wilhelmi BJ, Fujita K, et al. Ruptured pseudoaneurysm transpulmonary thermodilution behaves as an indicator of left ventricular
complicating an infected radial artery catheter: case report and review of the systolic function. Crit Care Med 2009;37(11):2913–8.
literature. Ann Plast Surg 2001;46(6):647–50. 44. Mutoh T, Kazumata K, Ishikawa T, et al. Performance of bedside
18. Band JD, Maki DG. Infections caused by aterial catheters used for transpulmonary thermodilution monitoring for goal-directed
hemodynamic monitoring. Am J Med 1979;67(5):735–41. hemodynamic management after subarachnoid hemorrhage. Stroke 2009;
19. Araghi A, Bander JJ, Guzman JA. Arterial blood pressure monitoring in 40(7):2368–74.
overweight critically ill patients: invasive or noninvasive? Crit Care 2006; 45. Marik PE, Baram M, Vahid B. Does central venous pressure predict fluid
10(2):R64. responsiveness? A systematic review of the literature and the tale of seven
20. Bur A, Herkner H, Vlcek M, et al. Factors influencing the accuracy of mares. Chest 2008;134(1):172–8.
oscillometric blood pressure measurement in critically ill patients. 46. Magder S, Bafaqeeh F. The clinical role of central venous pressure
Crit Care Med 2003;31(3):793–9. measurements. J Intensive Care Med 2007;22(1):44–51.
21. Manios E, Vemmos K, Tsivgoulis G, et al. Comparison of noninvasive 47. Hamzaoui O, Monnet X, Richard C, et al. Effects of changes in vascular tone
oscillometric and intra-arterial blood pressure measurements in hyperacute on the agreement between pulse contour and transpulmonary
stroke. Blood Press Monit 2007;12(3):149–56. thermodilution cardiac output measurements within an up to 6-hour
22. Muecke S, Bersten A, Plummer J. The mean machine: accurate non-invasive calibration-free period. Crit Care Med 2008;36(2):434–40.
blood pressure measurement in the critically ill patient. J Clin Monit 48. Gust R, Gottschalk A, Bauer H, et al. Cardiac output measurement by
Comput 2009;23(5):283–97. transpulmonary versus conventional thermodilution technique in intensive
23. Michard F, Boussat S, Chemla D, et al. Relation between respiratory changes care patients after coronary artery bypass grafting. J Cardiothorac Vasc
in arterial pulse pressure and fluid responsiveness in septic patients with Anesth 1998;12:519–22. doi: 10.1016/S1053-0770(98)90093–3.
acute circulatory failure. Am J Respir Crit Care Med 2000;162:134–8. 49. Ritter S, Rudiger A, Maggiorini M. Transpulmonary thermodilution-derived
24. Michard F. Stroke volume variation: from applied physiology to improved cardiac function index identifies cardiac dysfunction in acute heart failure
outcomes. Crit Care Med 2011;39(2):402–3. and septic patients: an observational study. Crit Care 2009;13(4):R133. doi:
25. Benes J, Chytra I, Altmann P, et al. Intraoperative fluid optimization using 10.1186/cc7994.
stroke volume variation in high risk surgical patients: results of prospective 50. Sakka SG, Ruhl CC, Pfeiffer UJ, et al. Assessment of cardiac preload and
randomized study. Crit Care 2010;14(3):R118. Epub 2010, Jun 16. extravascular lung water by single transpulmonary thermodilution. Intensive
26. Goedje O, Hoeke K, Lichtwarck-Aschoff M, et al. Continuous cardiac output Care Med 2000;26:180–7. doi: 10.1007/s001340050043.
by femoral arterial thermodilution calibrated pulse contour analysis:
comparison with pulmonary arterial thermodilution. Crit Care Med 1999; A complete list of references for this chapter can be found online at
27(11):2407–12. www.expertconsult.com.
Section II—Clinical and Laboratory Assessment 188.e1

References 29. Michard F, Boussat S, Chemla D, et al. Relation between respiratory


changes in arterial pulse pressure and fluid responsiveness in septic patients
1. Ramasamy I. Biochemical markers in acute coronary syndrome. Clin Chim with acute circulatory failure. Am J Respir Crit Care Med 2000;162(1):
Acta 2011;412(15-16):1279–96. Epub 2011, Apr 8. 134–8.
2. Noveanu M, Mebazaa A, Mueller C. Cardiovascular biomarkers in the ICU. 30. Michard F, Lopes MR, Auler JO Jr. Pulse pressure variation: beyond the
Curr Opin Crit Care 2009;15(5):377–83. fluid management of patients with shock. Crit Care 2007;11(3):131.
3. Contant CF, Valadka AB, Gopinath SP, et al. Adult respiratory distress 31. Marik PE, Cavallazzi R, Vasu T, et al. Dynamic changes in arterial waveform
syndrome: a complication of induced hypertension after severe head injury. derived variables and fluid responsiveness in mechanically ventilated
J Neurosurg 2001;95:560–8. patients: a systematic review of the literature. Crit Care Med 2009;37(9):
4. Boyd JH, Forbes J, Nakada TA, et al. Fluid resuscitation in septic shock: a 2642–7. Review.
positive fluid balance and elevated central venous pressure are associated 32. Auler JO Jr, Galas F, Hajjar L, et al. Online monitoring of pulse pressure
with increased mortality. Crit Care Med 2011;39(2):259–65. variation to guide fluid therapy after cardiac surgery. Anesth Analg
5. Fletcher JJ, Bergman K, Blostein PA, et al. Fluid balance, complications, and 2008;106(4):1201–6, table of contents.
brain tissue oxygen tension monitoring following severe traumatic brain 33. Marx G, Cope T, McCrossan L, et al. Assessing fluid responsiveness by
injury. Neurocrit Care 2010;13(1):47–56. stroke volume variation in mechanically ventilated patients with severe
6. Riva-Rocci S. Un Nuovo sfigmomanometro. Gazzeta Medica di Torino sepsis. Eur J Anaesthesiol 2004;21(2):132–8.
1896;47:981–96. 34. Ornstein E, Eidelman LA, Drenger B, et al. Systolic pressure variation
7. Cushing H. On routine determination of arterial tension in operating room predicts the response to acute blood loss. J Clin Anesth 1998;10(2):
and clinic. Boston Med Surg J 1903;148:250. 137–40.
8. Bigatello LM, Schmidt U. Arterial blood pressure monitoring. Minerva 35. Berkenstadt H, Margalit N, Hadani M, et al. Stroke volume variation as a
Anestesiol 2003;69(4):201–9. predictor of fluid responsiveness in patients undergoing brain surgery.
9. Peterson L, Dripps R, Risman G. A method for recording the arterial pulse Anesth Analg 2001;92(4):984–9.
and blood pressure in man. Am Heart J 1949;37:771. 36. Wiesenack C, Prasser C, Rodig G, et al. Stroke volume variation as an
10. Cohn JN. Blood pressure measurement in shock. Mechanism of inaccuracy indicator of fluid responsiveness using pulse contour analysis in
in ausculatory and palpatory methods. JAMA 1967;199(13):118–22. mechanically ventilated patients. Anesth Analg 2003;96(5):1254–7.
11. O’Rourke MF. Impact pressure, lateral pressure, and impedance in the 37. de Waal EE, Rex S, Kruitwagen CL, et al. Stroke volume variation obtained
proximal aorta and pulmonary artery. J ApplPhysiol 1968;25(5):533–41. with FloTrac/Vigileo fails to predict fluid responsiveness in coronary artery
12. Slogoff S, Keats AS, Arlund C. On the safety of radial artery cannulation. bypass graft patients. Br J Anaesth 2008;100(5):725–6.
Anesthesiology 1983;59(1):42–7. 38. Sander M, von Heymann C, Foer A, et al. Pulse contour analysis after
13. Chauhan S, Saxena N, Mehrotra S, et al. Femoral artery pressures are more normothermic cardiopulmonary bypass in cardiac surgery patients. Crit
reliable than radial artery pressures on initiation of cardiopulmonary Care 2005;9(6):R729–34.
bypass. J Cardiothorac Vasc Anesth 2000;14(3):274–6. 39. Pinsky MR. Probing the limits of arterial pulse contour analysis to predict
14. Mignini MA, Piacentini E, Dubin A. Peripheral arterial blood pressure preload responsiveness. Anesth Analg 2003;96(5):1245–7.
monitoring adequately tracks central arterial blood pressure in critically ill 40. Mahjoub Y, Pila C, Friggeri A, et al. Assessing fluid responsiveness in
patients: an observational study. Crit Care 2006;10(2):R43. critically ill patients: false-positive pulse pressure variation is detected by
15. Martin C, Saux P, Papazian L, et al. Long-term arterial cannulation in ICU Doppler echocardiographic evaluation of the right ventricle. Crit Care Med
patients using the radial artery or dorsalis pedis artery. Chest 2001;119(3): 2009;37(9):2570–5.
901–6. 41. Vincent JL, Rhodes A, Perel A, et al. Clinical review: update on
16. Clark VL, Kruse JA. Arterial catheterization. Crit Care Clin 1992;8(4): hemodynamic monitoring—a consensus of 16. Crit Care 2011;15(4):229.
687–97. 42. Benington S, Ferris P, Nirmalan M. Emerging trends in minimally invasive
17. Ganchi PA, Wilhelmi BJ, Fujita K, et al. Ruptured pseudoaneurysm haemodynamic monitoring and optimization of fluid therapy. Eur J
complicating an infected radial artery catheter: case report and review of Anaesthesiol 2009;26(11):893–905. Review.
the literature. Ann Plast Surg 2001;46(6):647–50. 43. Jabot J, Monnet X, Bouchra L, et al. Cardiac function index provided by
18. Band JD, Maki DG. Infections caused by aterial catheters used for transpulmonary thermodilution behaves as an indicator of left ventricular
hemodynamic monitoring. Am J Med 1979;67(5):735–41. systolic function. Crit Care Med 2009;37(11):2913–8.
19. Araghi A, Bander JJ, Guzman JA. Arterial blood pressure monitoring in 44. Mutoh T, Kazumata K, Ishikawa T, et al. Performance of bedside
overweight critically ill patients: invasive or noninvasive? Crit Care 2006; transpulmonary thermodilution monitoring for goal-directed
10(2):R64. hemodynamic management after subarachnoid hemorrhage. Stroke 2009;
20. Bur A, Herkner H, Vlcek M, et al. Factors influencing the accuracy of 40(7):2368–74.
oscillometric blood pressure measurement in critically ill patients. Crit Care 45. Marik PE, Baram M, Vahid B. Does central venous pressure predict fluid
Med 2003;31(3):793–9. responsiveness? A systematic review of the literature and the tale of seven
21. Manios E, Vemmos K, Tsivgoulis G, et al. Comparison of noninvasive mares. Chest 2008;134(1):172–8.
oscillometric and intra-arterial blood pressure measurements in hyperacute 46. Magder S, Bafaqeeh F. The clinical role of central venous pressure
stroke. Blood Press Monit 2007;12(3):149–56. measurements. J Intensive Care Med 2007;22(1):44–51.
22. Muecke S, Bersten A, Plummer J. The mean machine: accurate non-invasive 47. Hamzaoui O, Monnet X, Richard C, et al. Effects of changes in vascular
blood pressure measurement in the critically ill patient. J Clin Monit tone on the agreement between pulse contour and transpulmonary
Comput 2009;23(5):283–97. thermodilution cardiac output measurements within an up to 6-hour
23. Michard F, Boussat S, Chemla D, et al. Relation between respiratory calibration-free period. Crit Care Med 2008;36(2):434–40.
changes in arterial pulse pressure and fluid responsiveness in septic patients 48. Gust R, Gottschalk A, Bauer H, et al. Cardiac output measurement by
with acute circulatory failure. Am J Respir Crit Care Med 2000;162:134–8. transpulmonary versus conventional thermodilution technique in intensive
24. Michard F. Stroke volume variation: from applied physiology to improved care patients after coronary artery bypass grafting. J Cardiothorac Vasc
outcomes. Crit Care Med 2011;39(2):402–3. Anesth 1998;12:519–22. doi: 10.1016/S1053-0770(98)90093–3.
25. Benes J, Chytra I, Altmann P, et al. Intraoperative fluid optimization using 49. Ritter S, Rudiger A, Maggiorini M. Transpulmonary thermodilution-
stroke volume variation in high risk surgical patients: results of prospective derived cardiac function index identifies cardiac dysfunction in acute heart
randomized study. Crit Care 2010;14(3):R118. Epub 2010, Jun 16. failure and septic patients: an observational study. Crit Care
26. Goedje O, Hoeke K, Lichtwarck-Aschoff M, et al. Continuous cardiac 2009;13(4):R133. doi: 10.1186/cc7994.
output by femoral arterial thermodilution calibrated pulse contour analysis: 50. Sakka SG, Ruhl CC, Pfeiffer UJ, et al. Assessment of cardiac preload and
comparison with pulmonary arterial thermodilution. Crit Care Med 1999; extravascular lung water by single transpulmonary thermodilution.
27(11):2407–12. Intensive Care Med 2000;26:180–7. doi: 10.1007/s001340050043.
27. Kramer A, Zygun D, Hawes H, et al. Pulse pressure variation predicts fluid 51. Biancofiore G, Critchley LA, Lee A, et al. Evaluation of an uncalibrated
responsiveness following coronary artery bypass surgery. Chest 2004;126(5): arterial pulse contour cardiac output monitoring system in cirrhotic
1563–8. patients undergoing liver surgery. Br J Anaesth 2009;102:47–54.
28. Feissel M, Michard F, Mangin I, et al. Respiratory changes in aortic blood 52. Monnet X, Anguel N, Naudin B, et al. Arterial pressure-based cardiac
velocity as an indicator of fluid responsiveness in ventilated patients with output in septic patients: different accuracy of pulse contour, uncalibrated
septic shock. Chest 2001;119(3):867–73. pressure waveform devices. Crit Care 2010;14:R109.
188.e2 Section II—Clinical and Laboratory Assessment

53. De Backer D, Marx G, Tan A, et al. Arterial pressure-based cardiac output 81. Vignon P, Mücke F, Bellec F, et al. Basic critical care echocardiography:
monitoring: a multicenter validation of the third- generation software in validation of a curriculum dedicated to noncardiologist residents. Crit Care
septic patients. Intensive Care Med 2011;37(2):233–40. Med 2011;39:636–42.
54. Monnet X, Lahner D. Can the ‘ ‘FloTrac’ ’ really track flow in septic patients? 82. American College of Cardiology Foundation Appropriate Use Criteria Task
Intensive Care Med 2011;37(2):183–5. Force, American Society of Echocardiography, American Heart Association,
55. Mayer J, Boldt J, Poland R, et al. Continuous arterial pressure waveform- et al. ACCF/ASE/AHA/ASNC/HFSA/HRS/SCAI/SCCM/SCCT/SCMR 2011
based cardiac output using the FloTrac/Vigileo: a review and meta-analysis. Appropriate Use Criteria for Echocardiography. A Report of the American
J Cardiothorac Vasc Anesth 2009;23(3):401–6. College of Cardiology Foundation Appropriate Use Criteria Task Force,
56. McLean AS, Huang SJ, Kot M, et al. Comparison of cardiac output American Society of Echocardiography, American Heart Association,
measurements in critically ill patients: FloTrac/Vigileo vs transthoracic American Society of Nuclear Cardiology, Heart Failure Society of America,
Doppler echocardiography. Anaesth Intensive Care 2011;39(4):590–8. Heart Rhythm Society, Society for Cardiovascular Angiography and
57. Mayer J, Boldt J, Beschmann R, et al. Uncalibrated arterial pressure Interventions, Society of Critical Care Medicine, Society of Cardiovascular
waveform analysis for less-invasive cardiac output determination in obese Computed Tomography, and Society for Cardiovascular Magnetic
patients undergoing cardiac surgery. Br J Anaesth 2009;103(2):185–90. Resonance Endorsed by the American College of Chest Physicians. J Am
58. Maj GLG, Landoni G, Bignami E, et al. PRAM (pressure recording Coll Cardiol 2011;57(9):1126–66.
analytical method) for cardiac index measurement in unstable patients. 83. International expert statement on training standards for critical care
Minerva Anestesiol 2008;74:60. ultrasonography. Expert Round Table on Ultrasound in ICU. Intensive Care
59. Funk DJ, Moretti EW, Gan TJ. Minimally invasive cardiac output Med 2011;37(7):1077–83. Epub 2011, May 26.
monitoring in the perioperative setting. Anesth Analg 2009;108(3): 84. Cholley BP, Vieillard-Baron A, Mebazaa A. Echocardiography in the ICU:
887–97. time for widespread use! Intensive Care Med 2006;32(1):9–10.
60. Tsutsui M, Matsuoka N, Ikeda T, et al. Comparison of a new cardiac output 85. Matyal R, Skubas NJ, Shernan SK, et al. Perioperative assessment of
ultrasound dilution method with thermodilution technique in adult diastolic dysfunction. Anesth Analg 2011;113(3):449–72. Epub 2011, Aug 3.
patients under general anesthesia. J Cardiothorac Vasc Anesth 2009;23(6): 86. Platts DG, Fraser JF. Contrast echocardiography in critical care: echoes of
835–40. the future? A review of the role of microsphere contrast echocardiography.
61. Squara P, Denjean D, Estagnasie P, et al. Noninvasive cardiac output Crit Care Resusc 2011;13(1):44–55.
monitoring (NICOM): a clinical validation. Intensive Care Med 2007;33: 87. Faris JG, Veltman MG, Royse CF. Limited transthoracic echocardiography
1191–4. assessment in anaesthesia and critical care. Best Pract Res Clin Anaesthesiol
62. Marque S, Cariou A, Chiche JD, et al. Comparison between Flotrac-Vigileo 2009;23(3):285–98.
and Bioreactance, a totally noninvasive method for cardiac output 88. Cowie BS. Focused transthoracic echocardiography in the perioperative
monitoring. Crit Care 2009;13:R73. period. Anaesth Intensive Care 2010;38(5):823–36.
63. Swan HJ, Ganz W, Forrester J, et al. Catheterization of the heart in man 89. Joseph MX, Disney PJ, Da CR, et al. Transthoracic echocardiography to
with use of a flow-directed balloon-tipped catheter. N Engl J Med 1970; identify or exclude cardiac cause of shock. Chest 2004;126(5):1592–7.
283(9):447–51. 90. Lepper W, Belcik T, Wei K, et al. Myocardial contrast echocardiography.
64. Wagner JG, Leatherman JW. Right ventricular end-diastolic volume as a Circulation 2004;109(25):3132–5.
predictor of the hemodynamic response to a fluid challenge. Chest 1998; 91. Colreavy FB, Donovan K, Lee KY, et al. Transesophageal echocardiography
113(4):1048–54. in critically ill patients. Crit Care Med 2002;30(5):989–96.
65. Connors AF Jr, Speroff T, Dawson NV, et al. The effectiveness of right heart 92. Vieillard-Baron A, Page B, Augarde R, et al. Acute cor pulmonale in massive
catheterization in the initial care of critically ill patients. SUPPORT pulmonary embolism: incidence, echocardiographic pattern, clinical
Investigators. JAMA 1996;276(11):889–97. implications and recovery rate. Intensive Care Med 2001;27(9):1481–6.
66. Sandham JD, Hull RD, Brant RF, et al. A randomized, controlled trial of the 93. Jardin F, Dubourg O, Bourdarias JP. Echocardiographic pattern of acute cor
use of pulmonary-artery catheters in high-risk surgical patients. N Engl J pulmonale. Chest 1997;111(1):209–17.
Med 2003;348(1):5–14. 94. Feinberg MS, Hopkins WE, vila-Roman VG, et al. Multiplane
67. Vincent JL, Pinsky MR, Sprung CL. The pulmonary artery catheter: in transesophageal echocardiographic doppler imaging accurately determines
medio virtus. Crit Care Med 2008;36(11):3093–6. Review. cardiac output measurements in critically ill patients. Chest
68. Squara P, Cecconi M, Rhodes A, et al. Tracking changes in cardiac output: 1995;107(3):769–73.
methodological considerations for the validation of monitoring devices. 95. Griffee MJ, Merkel MJ, Wei KS. The role of echocardiography in
Intensive Care Med 2009;35:1801–8. hemodynamic assessment of septic shock. Crit Care Clin 2010;26(2):365–
69. Pinsky MR. Pulmonary artery occlusion pressure. Intensive Care Med 82, table of contents.
2003;29(1):19–22. 96. Ferrada P, Murthi S, Anand RJ, et al. Transthoracic focused rapid
70. Gayat E, Mebazaa A. Pulmonary hypertension in critical care. Curr Opin echocardiographic examination: real-time evaluation of fluid status in
Crit Care 2011;17(5):439–48. critically ill trauma patients. J Trauma 2011;70(1):56–62; discussion 62–4.
71. Gnaegi A, Feihl F, Perret C. Intensive care physicians’ insufficient knowledge 97. Noritomi DT, Vieira ML, Mohovic T, et al. Echocardiography for
of right-heart catheterization at the bedside: time to act? Crit Care Med hemodynamic evaluation in the intensive care unit. Shock 2010;34
1997;25(2):213–20. (Suppl 1):59–62.
72. Kumar A, Anel R, Bunnell E, et al. Pulmonary artery occlusion pressure and 98. Tousignant CP, Walsh F, Mazer CD. The use of transesophageal
central venous pressure fail to predict ventricular filling volume, cardiac echocardiography for preload assessment in critically ill patients. Anesth
performance, or the response to volume infusion in normal subjects. Crit Analg 2000;90(2):351–5.
Care Med 2004;32(3):691–9. 99. Hofer CK, Ganter MT, Rist A, et al. The accuracy of preload assessment by
73. Michard F, Teboul JL. Predicting fluid responsiveness in ICU patients: a different transesophageal echocardiographic techniques in patients
critical analysis of the evidence. Chest 2002;121(6):2000–8. undergoing cardiac surgery. J Cardiothorac Vasc Anesth 2008;22(2):
74. Singh S, Goyal A. The origin of echocardiography: a tribute to Inge Edler. 236–42.
Tex Heart Inst J 2007;34(4):431–8. 100. Bernard Y, Meneveau N, Boucher S, et al. Lack of agreement between left
75. Edler I, Lindstrom K. The history of echocardiography. Ultrasound Med ventricular volumes and ejection fraction determined by two-dimensional
Biol 2004;30(12):1565–644. echocardiography and contrast cineangiography in postinfarction patients.
76. Nilsson J, Westling H. Ultrasound in Lund: three world premieres. Clin Echocardiography 2001;18(2):113–22.
Physiol Funct Imaging 2004;24(3):137–40. 101. Jensen-Urstad K, Bouvier F, Hojer J, et al. Comparison of different
77. Edler I, Hertz CH. The use of ultrasonic reflectoscope for the continuous echocardiographic methods with radionuclide imaging for measuring left
recording of the movements of heart walls. 1954. Clin Physiol Funct ventricular ejection fraction during acute myocardial infarction treated by
Imaging 2004;24(3):118–36. thrombolytic therapy. Am J Cardiol 1998;81(5):538–44.
78. Frazin L, Talano JV, Stephanides L, et al. Esophageal echocardiography. 102. Cheung AT, Savino JS, Weiss SJ, et al. Echocardiographic and hemodynamic
Circulation 1976;54(1):102–8. indexes of left ventricular preload in patients with normal and abnormal
79. Mayo PH. Training in critical care echocardiography. Ann Intensive Care ventricular function. Anesthesiology 1994;81(2):376–87.
2011;30(1):36. 103. Reuter DA, Felbinger TW, Schmidt C, et al. Stroke volume variations for
80. Quintard H, Philip I, Ichai C. French survey on current use of ultrasound assessment of cardiac responsiveness to volume loading in mechanically
in the critical care unit: ECHOREA. Ann Fr Anesth Reanim 2011;Oct 4. ventilated patients after cardiac surgery. Intensive Care Med 2002;28(4):
Epub ahead of print. 392–8.
Section II—Clinical and Laboratory Assessment 188.e3

104. Bennett-Guerrero E, Kahn RA, Moskowitz DM, et al. Comparison of 128. Fencl V, Jabor A, Kazda A, et al. Diagnosis of metabolic acid-base
arterial systolic pressure variation with other clinical parameters to predict disturbances in critically ill patients. Am J Respir Crit Care Med 2000;
the response to fluid challenges during cardiac surgery. Mt Sinai J Med 162(6):2246–51. Epub 2000, Dec 9.
2002;69(1-2):96–100. 129. Kaplan LJ, Kellum JA. Initial pH, base deficit, lactate, anion gap, strong ion
105. Greim CA, Roewer N, Apfel C, et al. Relation of echocardiographic preload difference, and strong ion gap predict outcome from major vascular injury.
indices to stroke volume in critically ill patients with normal and low Crit Care Med 2004;32(5):1120–4. Epub 2004, Jun 12.
cardiac index. Intensive Care Med 1997;23(4):411–6. 130. Martin MJ, FitzSullivan E, Salim A, et al. Discordance between lactate and
106. Barbier C, Loubieres Y, Schmit C, et al. Respiratory changes in inferior vena base deficit in the surgical intensive care unit: which one do you trust? Am
cava diameter are helpful in predicting fluid responsiveness in ventilated J Surg 2006;191(5):625–30. Epub 2006, May 2. doi: 10.1016/j.amjsurg.
septic patients. Intensive Care Med 2004;30(9):1740–6. 2006.02.014.
107. Vieillard-Baron A, Chergui K, Rabiller A, et al. Superior vena caval 131. Husain FA, Martin MJ, Mullenix PS, et al. Serum lactate and base deficit as
collapsibility as a gauge of volume status in ventilated septic patients. predictors of mortality and morbidity. Am J Surg 2003;185(5):485–91.
Intensive Care Med 2004;30(9):1734–9. Epub 2003, May 3.
108. Moretti R, Pizzi B. Inferior vena cava distensibility as a predictor of fluid 132. Vitek V, Cowley RA. Blood lactate in the prognosis of various forms of
responsiveness in patients with subarachnoid hemorrhage. Neurocrit Care shock. Ann Surg 1971;173(2):308–13. Epub 1971, Feb 1.
2010;13(1):3–9. 133. Guyette F, Suffoletto B, Castillo JL, et al. Prehospital serum lactate as a
109. Roeck M, Jakob SM, Boehlen T, et al. Change in stroke volume in response predictor of outcomes in trauma patients: a retrospective observational
to fluid challenge: assessment using esophageal Doppler. Intensive Care study. J Trauma 2011;70(4):782–6.
Med 2003;29(10):1729–35. 134. Abramson D, Scalea TM, Hitchcock R, et al. Lactate clearance and survival
110. Singer M, Bennett ED. Noninvasive optimization of left ventricular filling following injury. J Trauma 1993;35(4):584–8; discussion 8-9. Epub 1993,
using esophageal Doppler. Crit Care Med 1991;19(9):1132–7. Oct 1.
111. Lee JH, Kim JT, Yoon SZ, et al. Evaluation of corrected flow time in 135. McNelis J, Marini CP, Jurkiewicz A, et al. Prolonged lactate clearance is
oesophageal Doppler as a predictor of fluid responsiveness. Br J Anaesth associated with increased mortality in the surgical intensive care unit. Am J
2007;99(3):343–8. Surg 2001;182(5):481–5. Epub 2002, Jan 5.
112. Monnet X, Rienzo M, Osman D, et al. Esophageal Doppler monitoring 136. Nguyen HB, Rivers EP, Knoblich BP, et al. Early lactate clearance is
predicts fluid responsiveness in critically ill ventilated patients. Intensive associated with improved outcome in severe sepsis and septic shock. Crit
Care Med 2005;31(9):1195–201. Care Med 2004;32(8):1637–42. Epub 2004, Aug 3.
113. Singer M. The FTc is not an accurate marker of left ventricular preload. 137. Gladden LB. Lactate metabolism: a new paradigm for the third millennium.
Intensive Care Med 2006;32(7):1089; author reply 91. J Physiol 2004;558(Pt 1):5–30. Epub 2004, May 8. doi: 10.1113/jphysiol.
114. Reinhart K, Kuhn HJ, Hartog C, et al. Continuous central venous and 2003.058701.
pulmonary artery oxygen saturation monitoring in the critically ill. 138. James JH, Luchette FA, McCarter FD, et al. Lactate is an unreliable
Intensive Care Med 2004;30:1572–8. indicator of tissue hypoxia in injury or sepsis. Lancet 1999;354(9177):505–
115. Gattinoni L, Brazzi L, Pelosi P, et al. A trial of goal-oriented hemodynamic 8. Epub 1999, Aug 28. doi: 10.1016/S0140-6736(98)91132-1.
therapy in critically ill patients. SvO2 Collaborative Group. N Engl J Med 139. Luchette FA, Jenkins WA, Friend LA, et al. Hypoxia is not the sole cause of
1995;333(16):1025–32. Epub 1995/10/19. doi: 10.1056/ lactate production during shock. J Trauma 2002;52(3):415–9. Epub 2002,
NEJM199510193331601. Mar 20.
116. Rivers E, Nguyen B, Havstad S, et al. Early goal-directed therapy in the 140. Constant JS, Feng JJ, Zabel DD, et al. Lactate elicits vascular endothelial
treatment of severe sepsis and septic shock. N Engl J Med 2001;345(19): growth factor from macrophages: a possible alternative to hypoxia. Wound
1368–77. Repair Regen 2000;8(5):353–60. Epub 2000, Dec 15.
117. Shoemaker WC, Appel PL, Kram HB, et al. Prospective trial of supranormal 141. Ohashi H, Kawasaki N, Fujitani S, et al. Utility of microdialysis to detect
values of survivors as therapeutic goals in high-risk surgical patients. Chest the lactate/pyruvate ratio in subcutaneous tissue for the reliable monitoring
1988;94(6):1176–86. Epub 1988, Dec 1. of hemorrhagic shock. J Smooth Muscle Res 2009;45(6):269–78.
118. Gurgel ST, do Nascimento P Jr. Maintaining tissue perfusion in high-risk 142. van den Heuvel I, Vlasselaers D, Wouters PJ, et al. Serial lactate
surgical patients: a systematic review of randomized clinical trials. Anesth measurements using microdialysis of interstitial fluid do not correlate with
Analg 2011;112(6):1384–91. Epub 2010, Dec 14. plasma lactate in children after cardiac surgery. Pediatr Crit Care Med 2009;
119. Velissaris D, Pierrakos C, Scolletta S, et al. High mixed venous oxygen 10(1):66–70.
saturation levels do not exclude fluid responsiveness in critically ill septic 143. James JH, Wagner KR, King JK, et al. Stimulation of both aerobic glycolysis
patients. Crit Care 2011;15(4):R177. Epub ahead of print. and Na(+)-K(+)-ATPase activity in skeletal muscle by epinephrine or
120. Giraud R, Siegenthaler N, Gayet-Ageron A, et al. ScvO2 as a marker to amylin. Am J Physiol 1999;277(1 Pt 1):E176-86. Epub 1999, Jul 17.
define fluid responsiveness. J Trauma 2011;70(4):802–7. 144. Clausen T. Na+-K+ pump regulation and skeletal muscle contractility.
121. Gattinoni L, Brazzi L, Pelosi P, et al. A trial of goal-oriented hemodynamic Physiol Rev 2003;83(4):1269–324. Epub 2003, Sep 25. doi: 10.1152/
therapy in critically ill patients. SvO2 collaborative group. N Engl J Med physrev.00011.2003.
1995;333:1025–32. 145. Schurr A. Lactate, glucose and energy metabolism in the ischemic brain
122. Eddy VA, Morris JA Jr, Cullinane DC. Hypothermia, coagulopathy, and (Review). Int J Mol Med 2002;10(2):131–6. Epub 2002, Jul 18.
acidosis. Surg Clin North Am 2000;80(3):845–54. Epub 2000, Jul 18. 146. Payne RS, Tseng MT, Schurr A. The glucose paradox of cerebral ischemia:
123. Runciman WB, Skowronski GA. Pathophysiology of haemorrhagic shock. evidence for corticosterone involvement. Brain Res 2003;971(1):9–17. Epub
Anaesth Intensive Care 1984;12(3):193–205. Epub 1984, Aug 1. 2003, Apr 15.
124. Brill SA, Stewart TR, Brundage SI, et al Base deficit does not predict 147. Oddo M, Levine JM, Frangos S, et al. Brain lactate metabolism in humans
mortality when secondary to hyperchloremic acidosis. Shock with subarachnoid hemorrhage. Stroke 2012;43(5):1418–21.
2002;17(6):459–62. Epub 2002, Jun 19. 148. Mascia L. Acute lung injury in patients with severe brain injury: a double
125. Fencl V, Leith DE. Stewart’s quantitative acid-base chemistry: applications hit model. Neurocrit Care 2009;11(3):417–26.
in biology and medicine. Respir Physiol 1993;91(1):1–16. Epub 1993, Jan 1. 149. Ospina-Tascon GA, Cordioli RL, Vincent JL. What type of monitoring has
126. Stewart PA. Modern quantitative acid-base chemistry. Can J Physiol been shown to improve outcomes in acutely ill patients? Intensive Care
Pharmacol 1983;61(12):1444–61. Epub 1983, Dec 1. Med 2008;34:800–20.
127. Schlichtig RGA, Severinghaus JW. Current status of acid-base quantitation
in physiology and medicine. Anesthesiol Clin North Am 1998;16:211–33.
Chapter
20  
II

Ventilation and
Pulmonary Function
Maurizio Cereda and Patrick J. Neligan

compliance. Most resistance comes from the airways, but


Introduction small amounts are associated with tissue resistance, stress
Respiratory monitoring is an important aspect of managing relaxation, and gas distribution.
patients admitted to the intensive care unit (ICU), including Respiratory mechanics can be evaluated by several tech-
those with neurologic problems. Obviously, detecting gas niques. Among these the rapid airway occlusion technique that
exchange abnormalities is essential to properly manage many estimates alveolar elastic recoil pressure by measuring the
acute intracranial processes but neurologic patients often have inspiratory plateau airway pressure (Pplat) is a simple bedside
coexisting pulmonary diseases and up to a third of patients test for ICU use. Intrinsic positive end-expiratory pressure
admitted to a neurocritical care unit (NCCU) can develop (PEEPi) is another important variable that usually is measured
acute lung injury (ALI) or acute respiratory distress syndrome using end-expiratory airway occlusion. PEEPi has important
(ARDS).1 Some possible causes for ARDS are listed in Table cardiopulmonary effects including decreased cardiac output,
20.1. These may require complex ventilator management, alveolar overdistention, increased work of breathing, and
which is facilitated by the availability of various types of pul- effects on compliance.
monary physiologic data. Although gas exchange monitoring
is a well-established practice, the bedside measurement of
respiratory mechanics is still relatively underused in the ICU.
Imaging and Diagnostic Studies
This stems from clinicians’ poor understanding of respiratory A variety of imaging and diagnostic tools are available to
physiology, and because the evidence about the impact of this evaluate pulmonary disease in the ICU, to identify high-risk
type of monitoring on outcomes is often confusing. This populations, and as diagnostic tools to optimize mechanical
chapter provides a physiologic background on pulmonary ventilation.2-7 Basic tests include the bedside chest x-ray
function monitoring in the ICU, focusing on bedside mea- (CXR) and arterial blood gas analysis.8 Venous blood gas or
surement of gas exchange and respiratory mechanics, and evaluation of electrolytes and in particular serum bicarbon-
highlights how these data can be used to support sound ven- ate may be useful for ventilator management when arterial
tilator management choices. In addition how to decide who blood gas analysis is not available. In patients who are diffi-
should be ventilated and criteria to determine liberation from cult to wean, assessment of nutritional (e.g., phosphate) and
mechanical ventilation is discussed briefly. endocrine status (e.g., cortisol and thyroid function) can be
helpful. Noninvasive tests include computed tomography
(CT), positron emission tomography (PET) or CT/PET,9,10
Basic Respiratory Physiology CT angiography for pulmonary embolus,11 electrical imped-
and What It Means for ance tomography (EIT), and lung ultrasound (LUS). Invasive
tests for diagnosis include bronchoscopy, bronchoalveolar
Pulmonary Assessment lavage, transbronchial lung biopsy, and open lung biopsies.
There are two compartments to the respiratory system: the These invasive tests can be safely performed even in patients
lung and the chest wall. The chest wall includes the abdomen with ARDS. Common complications include transient hypox-
because abdominal pathology, including obesity or abdomi- emia, respiratory acidosis, and pneumothorax.12 Formal pul-
nal compartment syndrome can alter respiratory mechanics. monary function tests such as spirometry, diffusing capacity
To inflate the respiratory system requires pressure to be gen- of the lung for carbon monoxide (DLCO), exercise testing, or
erated because of its resistive and elastic properties. Elasticity bronchial challenges are infrequently performed on critical
is expressed as compliance (volume change or pressure care patients.13 In the future biomarkers and genomic analy-
change). Esophageal pressure and airway pressure both should sis may become useful tools in diagnosis or identification
be measured to assess how each compartment of the respira- of patients at risk for pulmonary disorders such as ALI
tory system influences respiratory mechanics, particularly or ARDS.14
© Copyright 2013 Elsevier Inc. All rights reserved. 189
190 Section II—Clinical and Laboratory Assessment

Table 20.1  Some Causes of Acute Table 20.2  American-European Consensus


Respiratory Distress Syndrome Definitions of ALI and ARDS Developed  
in 1994
Direct Lung Injury Indirect Lung Injury
Common Pneumonia Sepsis ALI CRITERIA
Aspiration of gastric Severe trauma with Timing: acute onset
contents shock and multiple Oxygenation: PaO2/FiO2 ≤300 mm Hg (regardless of PEEP level)
blood transfusions Chest radiograph: bilateral infiltrates on frontal chest
Less common Pulmonary Drug overdose radiograph
contusion Acute pancreatitis Pulmonary artery wedge pressure: ≤18 mm Hg when
Fat emboli Blood transfusion measured or no clinical evidence of left atrial hypertension
Near-drowning Pregnancy-related ARDS CRITERIA
Inhalational injury ARDS
Reperfusion Same as ALI except oxygenation: PaO2/FiO2 200 mm Hg
pulmonary edema (regardless of PEEP level)

Adapted from Ware LB, Matthay MA. The acute respiratory distress Adapted from Bernard GR, Artigas A, Brigham KL, et al. The American-
syndrome. N Engl J Med 2000;342:1334–48. European consensus conference on ARDS: definitions, mechanisms, relevant
ARDS, Acute respiratory distress syndrome. outcomes, and clinical trial coordination. Am J Respir Crit Care Med
1994;149:818–24.
ALI, Acute lung injury; ARDS, acute respiratory distress syndrome; FiO2,
inspiratory oxygen fraction; PaO2, arterial oxygen tension; PEEP, positive
end-expiratory pressure.
The bedside CXR can reveal abnormalities that may not be
clinically evident. In addition, a CXR can detect the malposi-
tion of tubes, including an endotracheal tube and catheters
and identify complications associated with these devices.15
Today quantitative CT is the gold standard to assess lung mor-
phology, recruitment, and hyperinflation of lung tissue at
Table 20.3  The Murray Lung Injury Score*
different inflation pressures. LUS is a useful, radiation-free, Variable Score
noninvasive bedside lung-imaging tool.16 Like CT it is primar- 1. Chest roentgenogram score
ily a tool to detect changes in density. EIT and PET provide No alveolar consolidation 0
more functional data.9 For example, EIT can provide a non- Alveolar consolidation confined to 1 quadrant 1
invasive continuous image of pulmonary impedance. This can Alveolar consolidation confined to 2 quadrants 2
be useful during mechanical ventilation because it indicates Alveolar consolidation confined to 3 quadrants 3
Alveolar consolidation in all 4 quadrants 4
the distribution of ventilation.3 PET also can be used to assess
lung inflammation10 that along with impaired gas exchange is 2. Hypoxemia score
PaO2/FiO2 >300 0
observed in ARDS, ALI, or ventilator-induced lung injury PaO2/FiO2 225-299 1
(VILI). PaO2/FiO2 175-224 2
PaO2/FiO2 100-174 3
PaO2/FiO2 <100 4
ARDS and ALI
3. PEEP score (when ventilated) (cm H2O)
Many patients in the ICU can develop ALI or ARDS, and PEEP ≤5 0
patients with severe acute brain injury often are at risk for PEEP 6-8 1
developing these conditions. Mortality from ALI and ARDS PEEP 9-11 2
PEEP 12-14 3
remains high and their development adversely affects intra- PEEP >15 4
cranial physiology.17-21 It is important then to know how to
4. Respiratory system compliance score (when
define these conditions, recognize who is at risk for them, and available) (mL/cm H2O)
diagnose their onset (Tables 20.1, 20.2, 20.3, and 20.4). ARDS Compliance >80 0
is a clinical-radiologic diagnosis and includes severe hypox- Compliance 60-79 1
emia assessed by an arterial oxygen tension/fraction of inspired Compliance 40-59 2
oxygen ratio less than 200 and bilateral infiltrates on a CXR Compliance 20-39 3
Compliance <19 4
in the absence of left atrial hypertension. The definition of
ARDS and ALI is provided in Table 20.2.22-24 Other sets of Adapted from Murray JF, Matthay MA, Luce JM, et al. An expanded
definition criteria have been proposed (see Tables 20.3 and definition of the adult respiratory distress syndrome. Am Rev Respir Dis
1988;138:720–3.
20.4). In children ALI and ARDS often are defined by the *The final score is calculated by the addition of the component parts:
oxygenation index defined as the product of mean airway score 0, no lung injury; score 1 to 2.5, mild to moderate lung injury;
pressure (MAP) × FiO2 × 100/PaO2.25 However, the sensitivity score >2.5, severe lung injury.
and specificity of the clinical diagnosis may depend on the FiO2, Inspiratory oxygen fraction; PaO2, arterial oxygen tension; PEEP, positive
end-expiratory pressure.
underlying etiology. Other diagnostic tests are used in large
part to exclude conditions that may mimic ALI or ARDS or
to guide therapy. Chest CT can help evaluate the extent of the
disease and guide ventilation and prognosis. Current therapy
is centered on lung-protective mechanical ventilation and a
restrictive fluid management strategy.
Section II—Clinical and Laboratory Assessment 191

realization that these tracings have profound physiologic


Table 20.4  Comparative Analyses of meanings and that their analysis provides useful clinical
Commonly Used Definitions for ALI   information in ventilated patients.29 A more recent addition
and ARDS is the development of volumetric capnography, which is the
Definition Pros Cons analysis of CO2 over volume waveforms and provides further
pathophysiologic insight.30 Similar to pulse oximetry, cap-
AECC Simple and easy Acute onset: not defined
to use PAOP often not
nometry and capnography have become part of the clinical
Differentiates ALI measured PEEP/ routine in the OR, although their use is still somewhat limited
and ARDS compliance/MAP not in ICU patients.
Prognostic considered
capability based Risk factors not
on ARMA study emphasized Engineering and Technology
Murray score Takes PEEP/ Does not include MAP Pulse oximeters determine oxygen saturation (SpO2) using
compliance into Does not exclude heart
consideration failure
the spectrophotometric characteristics of pulsatile arterial
Differentiates mild Does not identify blood. Oxyhemoglobin absorbs light in the infrared spectrum
to moderate individual risk factors at 940 nm, whereas deoxyhemoglobin absorbs light preferen-
from severe Prognostic ability not tially at 660 nm, corresponding to red light. The pulse oxim-
lung injury validated eter probe consists of two diodes, each emitting red or
Radiologic criteria
more specific infrared light into tissue, and of sensors that measure the
emerging amount of radiation. SpO2 is obtained by process-
Delphi Defines criteria for Excludes P/F >200 and
onset (<72 hr) <300
ing the relative absorption of light by oxygenated and non-
Risk factor Does not include oxygenated hemoglobin. The device reports only the pulsatile
emphasized compliance or MAP component of the recorded signal, and the final result is a
Takes PEEP into continuous measurement of the patient’s oxyhemoglobin
consideration status and pulse rate. Calibration is done in volunteers who
Objectively rules
out heart failure breathe hypoxic mixtures of gas. Standard, bi-wavelength
pulse oximetry does not detect the presence of dyshemoglo-
Oxygenation Takes MAP into Does not take PEEP and
index consideration compliance into
binemias, but multiwave pulse oximeters recently have been
Prognostic ability consideration developed. These instruments also can estimate total hemo-
validated Does not exclude heart globin concentration together with carboxyhemoglobin and
failure Does not methemoglobin levels.31
evaluate radiologic Clinical capnometry is performed using a source of infrared
signs
light and measuring the absorption by CO2 at various wave-
Adapted from Raghavendran K, Napolitano LM. Definition of ALI/ARDS. Crit lengths, using a photodetector within a sampling cell. The
Care Clin 2011;27(3):429–37. measured light absorption is then compared with a CO2-free
AECC, American European Consensus Conference; ALI, acute lung injury;
ARDS, acute respiratory disorder syndrome; ARMA, acute respiratory
reference, and finally CO2 is expressed either as partial
management in ALI/ARDS; MAP, mean arterial pressure; PAOP, pulmonary pressure or as concentration. There are two types of capnom-
artery occlusion pressure; PEEP, positive end-expiratory pressure; P/F, PaO2/FiO2 eters: (1) mainstream capnometers measure CO2 directly in a
ratio.
cuvette placed in the ventilator circuit, close to the endotra-
cheal tube, and (2) sidestream capnometers aspirate gas from
the airway opening using a small-bore tubing line; CO2 is then
Gas Exchange Monitoring measured in a chamber placed within the monitor. Thus CO2
measurements lag behind the respiratory cycle in sidestream
History capnometry. All modern capnometers also provide a capnog-
Blood gas analysis was introduced by Severinghaus in 1958 and raphy tracing on the display screen. Volumetric capnographs
rapidly became an essential part of the clinical routine.26 This combine a mainstream capnometer with a flowmeter within
technique provides direct measurement of arterial blood PO2, the same device,30 thus providing measurements of CO2
PaCO2, and pH and also allows for calculation of serum bicar- production and dead space. In this technique, flow and CO2
bonate, base deficit-excess, and oxyhemoglobin saturation. signals need to be synchronized for measurements to be
The last can be directly measured using co-oximetry, which meaningful.
also measures carboxyhemoglobin and methemoglobin.
The history of pulse oximetry started in the 1930s, with
the first studies on the absorption of light by oxygenated Indications for Use
blood. However, the first clinically acceptable instruments Pulse oximetry is currently part of the basic ICU monitoring
became available only in the 1970s.27 Their introduction and is applied to almost all patients irrespective of their clini-
made bedside monitoring of arterial oxygenation possible in cal status: in fact SpO2 is often referred to as the “fifth vital
the ICU, in the operating room (OR), and in many other sign.” The main purpose of SpO2 monitoring is to provide an
hospital locations. Capnometry, the measurement of carbon early warning of hypoxemia and to avoid its complications. In
dioxide (CO2) in the expired gas, was first made possible by ventilated patients, SpO2 also is used to titrate oxygen therapy32
infrared photometry in the 1940s. A subsequent step was the and positive end-expiratory pressure (PEEP). This decreases
introduction of capnography, which is the analysis of expired the need for frequent arterial blood sampling.33 Capnometry
CO2 versus time waveforms.28 This advancement followed the and capnography are considered a standard in the OR, where
192 Section II—Clinical and Laboratory Assessment

they are mainly used to assess the adequacy of the airway and
of ventilation. In particular, capnometry is the most effective
method to verify successful intubation.34 In the ICU, capnom-
etry is mostly used for select patient populations, such as those
with intracranial hypertension, where it provides a surrogate
for continuous monitoring of arterial PaCO2, the control of
which can be important in management of increased intra-
cranial pressure (ICP). Capnometry also can be used to facili- PaCO2
tate ventilator weaning and at the same time limit blood 3 4
sampling.35 However, there are limitations of this monitor as
an estimate of arterial PaCO2 that should not be disregarded
in patients with lung diseases (see following text). 1 2 5
A

Information Provided  
and Troubleshooting
Pulse oximetry is used to measure arterial saturation nonin-
vasively and accurately predicts arterial PO2.36 However, the
oxyhemoglobin dissociation curve has a sigmoid shape, and
on its flat portion large PO2 decreases may cause only minimal
changes in SpO2 and be underestimated. Furthermore, the
accuracy and precision of pulse oximetry measurements PaCO2
decrease when hemoglobin saturation is less than 90%.37 For
these two reasons, any decrease in the SpO2 below 90% must
be considered a significant clinical event and assumed to
predict hypoxemia. Saturations of less than 80% do not convey
B
any meaningful information other than that the patient is
severely hypoxemic, because no calibration data on human Fig. 20.1  A, Schematic representation of the lungs (top), with the
subjects are available in this range. The use of standard pulse alveolar spaces filled with CO2-rich gas (dark blue) and the airways
oximeters is limited in the presence of dyshemoglobinemias, containing fresh gas (light blue); bottom: normal capnometry tracing
and thus co-oximetry should be performed whenever carbon (blue line) with arterial PaCO2 (red line). The stages of the capnometry
monoxide poisoning is suspected. Hypotension, vasoconstric- tracing are marked with numbers (see text for explanation). B, Schematic
representation of the lungs (top), where low-CO2 gas exhaled by
tion, and hypothermia reduce the pulsatility of capillary blood
hypoperfused alveolar units mixes with CO2-rich gas from well perfused
and the ability of the pulse oximeter to identify a tracing. In units, causing a decrease in the expired PaCO2 to levels that are
addition, dark skin pigmentation decreases the accuracy of significantly lower than in the arterial blood (bottom).
SpO2 measurement.32 This same study also suggested that a
higher SpO2 goal should be used when titrating oxygen therapy
in black or dark-skinned rather than fair-skinned patients. exhaled by such units contains very low or negligible CO2, thus
Capnography tracings are characterized by stages that causing etCO2 to be lower than the arterial PaCO2 (Fig. 20.1,
correspond to specific moments of the respiratory cycle B). The difference between the two variables is proportional
(Fig. 20.1, A). At the start of exhalation, measured PaCO2 is to the fraction of alveolar dead space. Thus in patients with
negligible because the airways contain only fresh gas (point lung disease, etCO2 should never be assumed to accurately
1). The volume of the airways is called anatomic dead space represent arterial PaCO2 unless an association between the
because the gas they contain does not participate in gas two variables can be confirmed by arterial blood gas analysis.
exchange. As expiration proceeds, CO2-rich alveolar gas enters When there is a discrepancy, the trend in etCO2 still can be
the airways, causing a rapid increase in PaCO2 (points 2 to 3). used. Volumetric capnography allows assessment of both ana-
When the dead space has been completely washed out by tomic and alveolar dead space, together with CO2 production.
alveolar gas, the exhaled PaCO2 curve reaches a plateau (points These variables can be used to titrate mechanical ventilation,
3 to 4). During inspiration, fresh gas enters the airways and to detect changes in hemodynamic status and pulmonary
PaCO2 rapidly drops to zero again (point 5). The highest mea- embolism, and to confirm diagnoses and prognoses.30
sured PaCO2, identified at the end of expiration (point 4) is Analysis of capnographic tracings provides further infor-
called end-tidal CO2 (etCO2) and is indicative of the PaCO2 in mation, and the clinician should be aware of a number of
the alveoli. Because the alveolar gas is in equilibrium with the abnormal patterns. In patients with COPD and asthma, the
pulmonary capillary blood, etCO2 is also very close to arterial upstroke of expired CO2 is slower than normal and a plateau
PaCO2 in healthy subjects and it can be used to adjust ventila- stage is never reached. In this case, etCO2 and arterial PaCO2
tion. However, etCO2 may underestimate arterial PaCO2 in the cannot be determined. This phenomenon results from severe
presence of diseases that cause significant alveolar dead space, dishomogeneity of expiration kinetics among different alveo-
such as chronic obstructive pulmonary disease (COPD), pul- lar units. An exponential decrease in etCO2 over minutes is
monary embolism, and ARDS.38 Alveolar dead space is the an ominous sign, because it indicates a rapid decrease in pul-
wasted fraction of inspired gas that does not participate in monary perfusion, which can be due to pulmonary embolism,
CO2 exchange because of the presence of alveolar units with sudden onset of shock, or impending cardiopulmonary arrest.
low or absent perfusion relative to ventilation. In this case, gas Capnography also can be used to detect leaks in the breathing
Section II—Clinical and Laboratory Assessment 193

circuit, disconnection from the ventilator, and a patient’s inspi- assumption that a good SpO2 value also reflects adequate ven-
ratory attempts. tilation. In reality, oxygen therapy in a hypoventilated patient
may effectively correct hypoxemia while masking severe
hypercapnia, thus giving clinicians a false sense of security.
Therapy Based on Gas Exchange This phenomenon could be the cause of adverse respiratory
Monitoring and Its Effect on Outcome events during procedural sedation and can be prevented if
Both pulse oximetry and capnometry became widely accepted capnometry is performed with pulse oximetry.
for clinical use in the mid- to late 1980s. Since then, multiple
medical societies have recommended the use of these moni-
tors, particularly in the OR but also in other settings. It is very Respiratory Mechanics
likely that the introduction of SpO2 and capnometry has con- and Ventilator Variables
tributed to improved perioperative morbidity and mortality.
This statement is based mostly on an analysis of closed anes- History
thetic malpractice claims that suggested that a majority of The conceptual background to respiratory mechanics moni-
intraoperative respiratory mishaps would have been avoided toring is provided by classic pulmonary physiology. Studies on
had pulse oximetry and capnometry been available.39 The pressure-volume loops obtained during inflation and defla-
same dataset also showed a historic trend of reduction in tion of surfactant deficient lungs44 showed characteristic
respiratory-related claims following introduction of these abnormalities45 that later in time also were observed in patients
monitors. However, the strength of clinical evidence that sup- with ARDS.45,46 However, it was the introduction of the rapid
ports the use of capnometry and pulse oximetry in the peri- flow interruption technique46,47 that brought the measurement
operative setting and in the ICU is disappointing, although of respiratory system compliance and resistance to the bedside.
there is evidence to suggest use of pulse oximetry makes a Respiratory mechanics have become more relevant in critical
difference to patient care. First, randomized studies conducted care since the demonstration that excessive alveolar inflation
in the perioperative period show that the use of pulse oxim- leads to ventilator-associated lung injury (VALI)48 and can
etry increases the rate of detection of hypoxemia.33,40 Second, worsen patient outcomes.49
Moller et al.40 in a randomized study that included 28,802 The use of esophageal pressure (Pes) to measure work of
patients observed that patients who received SpO2 monitoring breathing and to separately study the mechanics of the lung
had a lower incidence of intraoperative electrocardiographic and of the chest wall was initially tested in studies on healthy
ischemic changes than control patients who did not have a volunteers and in patients with COPD.50 In the 1990s, this
SpO2 monitor. However, the overall rate of perioperative com- technique was extended to ICU patients and led to an
plications and the mortality were similar in the two groups. understanding of the importance of chest wall mechanics51,52
In a smaller study from the same group, the rate of cognitive and patient-ventilator dyssynchrony (PVD) that can increase
dysfunction was similar in patients who were or were not the work of breathing.53 Impulse oscillometry can be used
monitored with pulse oximetry.41 A 2003 Cochrane review as a noninvasive method to assess pulmonary compliance
concluded that although pulse oximetry improved detection rather than using the esophageal pressure method, which is
of hypoxemia, its utility and cost effectiveness as a tool to invasive.54
improve outcomes is questionable, at least in the perioperative The advent of CT of the chest in ARDS patients led to the
period.42 This observation was confirmed in a subsequent demonstration that ventilation is not homogeneous and that
Cochrane analysis of five randomized trials that included atelectasis is relevant in this syndrome.55 In particular, CT is
22,992 patients and compared pulse oximetry or no pulse able to reveal discrepancies between bedside chest x-rays and
oximetry during the perioperative period (i.e., there was no various clinical and physiologic parameters in ARDS patients.2
difference in mortality although the incidence of hypoxemia For a long time, PEEP was used to achieve goal oxygenation
in the recovery room was less in the pulse oximeter group).43 levels through alveolar recruitment in ARDS and in other
However, given the wide acceptance of this monitoring tech- forms of acute respiratory failure. The discovery of cyclic
nique, it is unlikely that further randomized studies will be alveolar collapse and reopening in human ARDS,56 and animal
performed in the future. evidence that these phenomena contribute to VALI57 led to the
Capnometry, and particularly volumetric capnography formulation of the “open lung concept.” According to this
provides helpful physiologic information that can be used to idea, optimization of alveolar recruitment may improve
guide mechanical ventilation and weaning. Dead space is also patient outcomes and should be a goal of ventilator manage-
an independent predictor of mortality in ARDS patients.38 ment, independent from the oxygenation response. Although
However, there is a lack of outcome evidence that this form the results of randomized studies on this topic have been
of monitoring is helpful in the ICU, other than for the confir- contradictory,58,59 the open lung concept has renewed the
mation of intubation.34 interest in bedside clinical tools to measure alveolar recruit-
ment, lung volumes,60 and regional patterns of ventilation.61
Risks
Pulse oximetry and capnometry are essentially risk-free tech- Engineering and Technology
niques, from the point of view of device safety. Similar to any Monitoring respiratory mechanics requires pneumotacho-
other type of monitoring, it can be argued that patient harm graphs and electromechanical transducers to measure gas
may result from poor interpretation of the data and lack of flows and airway pressures (Paw). Pneumotachographs quan-
understanding of the technique, rather than from the monitor tify flow from the pressure drop generated by the passage of
itself. An example of a common misunderstanding is the gas through a known resistance, which typically includes a
194 Section II—Clinical and Laboratory Assessment

thin metal grid to maintain laminar gas flow. Other designs cmH2O
25
are available, although this is the most frequently used flow-
meter. A differential pressure transducer converts the pres- 20
sure difference across the resistance into an electrical signal,
which then is amplified, calibrated, and processed. Volumes 15
are obtained by mathematical integration of the measured

Paw
10
flow. Transducers typically are embedded in the ventilator
machinery, remotely from the patient and serve the dual 5
purpose of feeding the ventilator servocontrols and of pro-
0
viding data for display. Modern ventilators also display curves,
facilitating detection and interpretation of events. In certain –5
ventilators, flows and pressures are measured at the proximal
opening of the endotracheal tube, bypassing the ventilator Lpm
tubing. This design renders the measurements more accurate
but exposes the transducers to vapor condensation and 100
mucus accumulation. 50
Many ventilators allow the clinician to perform a rapid flow
interruption maneuver at the end of inspiration or expiration 0

Flow
by pushing a dedicated button on the ventilator console. –50
During this maneuver both the inspiratory and the expiratory
valves of the ventilator remain closed for a short time and so –100
provide conditions of zero flow. This maneuver is fundamen- –150
tal to the measurement of respiratory mechanics variables at
the bedside. An algorithm that automatically performs repeti- –200
tive, short-flow interruptions and provides frequently updated
measurements is available, but it has been created specifically L
to be implemented in the proportional assist ventilation 1.50
(PAV+) mode.62 1.25
Pes is measured using a thin, 10-cm-long balloon partially 1.00
filled with air and inserted into the lower third of the esopha-
Volume

gus.50 The use of central venous pressure as a surrogate for Pes 0.75
also has been described.63 Pes measurement is included in 0.50
pulmonary monitoring systems provided with disposable 0.25
esophageal catheters and pneumotachographs.
Pressure volume loops are obtained by step-wise or con- 0
2
tinuous inflations-deflation cycles of the lungs, which are –0.25
performed using either a large-volume syringe or a ventilator Fig. 20.2  An example of patient-ventilator asynchrony. From top
with dedicated software.64 Graphic or mathematical analysis65 to bottom: airway pressure (Paw), flow, and volume versus time tracings
is used to extrapolate clinically useful information from during assist control ventilation. In this example the ventilator does not
these loops. match the patient’s demand for flow and volume. This causes Paw to
EIT is a relatively new bedside imaging technique that decrease and to have a characteristic indentation during inspiration. This
constructs an image of gas distribution within the chest phenomenon can cause patient discomfort unless inspiratory flow and
during the different phases of respiration using a series of volume settings are adjusted. L, Liters; Lpm, liters per minute.
electrode pairs placed around the circumference of the
chest3,61 Potential applications for this technique include
titration of PEEP, recruitment maneuver, and assessment of
regional patterns of ventilation.66-69 Although commercial patient management and are probably sufficient in the major-
devices are available, their clinical application has still to be ity of patients. However, slightly more advanced monitoring
fully elucidated.5 Techniques that measure lung volumes is easy to perform and can be very useful in many cases. First,
through gas dilution or washout techniques are in the process visual inspection and interpretation of Paw and flow versus
of being tested and may be used in the future to help opti- time tracings can help detect asynchronies between patient
mize ventilator parameters.60 and ventilator70 (Fig. 20.2). Furthermore, the flow interrup-
tion method should be used periodically in every ventilated
patient if the machinery allows it. This technique is associated
Indications for Use with no additional risk or cost and allows the measurement
Monitoring of Paw, tidal volume (TV), expired minute volume of compliance, inspiratory resistance, and intrinsic PEEP
(VE), and other basic ventilator variables is essential to the (PEEPi), all variables that can rapidly reveal changes in patient
care of all ventilated patients. These data document the effec- conditions.
tiveness of ventilation, they signal disconnections from the Monitoring inspired volumes and pressures can help detect
ventilator and airway obstruction, and they provide early the presence of excessive alveolar inflation. This is important
warnings of changes in patient condition. In many institutions because alveolar inflation has outcome implications.49 Evi-
these are the only variables that are collected during routine dence suggests that integrating Pes in pulmonary monitoring
Section II—Clinical and Laboratory Assessment 195

V′i Paw − Palv


Ri =
V′i
Paw
∆V
Crs =
∆Palv
Chest
wall PI = Palv − Pes

Palv ∆V ∆V
CI = Ccw =
Pes ∆PI Pes
A

Pip
Pplat
Paw
Paw
PEEPi
V′i PEEP
Flow

TV Flow
Volume

B 1 2 Time C Time
Fig. 20.3  A, The respiratory system is schematically represented as two compartments in series: the lungs and the chest wall. During the delivery of
inspiratory flow (V′i), airway pressure (Paw) is higher than the alveolar pressure (Palv), due to inspiratory resistance (Ri). Palv allows calculation of the
compliance of the respiratory system. If esophageal pressure is known (Pes) it is possible to calculate transpulmonary pressure (Pl) and, finally, the
compliance of the lung (Cl) and of the chest wall (Ccw). B, Paw, flow, and volume tracings during a rapid flow interruption maneuver are shown.
At time 1 the peak inspiratory pressure (Pip) is reached and the inspiratory flow is stopped. After a brief transient stage Paw reaches a plateau pressure
(Pplat) at time 2 and approximates Palv. C, Paw, flow and Palv (blue dashed line) versus time tracings in the presence of intrinsic positive end-expiratory
pressure (PEEPi). At the end of expiration, Palv is abnormally higher than the applied level of PEEP, as also suggested by the presence of residual end-
expiratory flow. When an expiratory flow interruption is performed, Paw equilibrates with Palv and rapidly increases. Intrinsic PEEP (PEEPi) is quantified
as the difference between Paw after the interruption and the applied PEEP. TV, Tidal volume.

provides more accurate estimates of alveolar distention than (Pl), the difference between Palv and pleural pressure. The
Paw alone,71,72 particularly when chest wall mechanics are latter can be estimated by measuring Pes. Furthermore the
abnormal. Pressure volume curves analysis is used in certain value of Crs is determined by the compliance of both the lung
institutions to identify the optimal level of PEEP in ARDS (Cl) and the chest wall (Ccw). In pulmonary physiology, the
patients and has been used in clinical studies that evaluate chest wall also includes the abdominal compartment.
ventilation strategies in ARDS.58,73 However, the precise physi- Normal Crs values are 70 to 80 mL/cm H2O; in normal
ologic meaning of the information obtained is still to be individuals Crs is affected by body size. A Crs value less than
defined. 50 mL/cm H2O is typical in ARDS and can be used to define
this syndrome and to grade its severity.75 Crs also can be lower
than normal in many other acute conditions such as atelecta-
Information Provided   sis, pneumonia, and cardiogenic pulmonary edema. In many
and Troubleshooting patients the change in Crs results from a decrease in Cl.76
The respiratory system requires the generation of pressure for However, in ARDS many patients also have a decrease in
its inflation, as a result of its resistive and elastic properties Ccw.52 This finding typically is from abnormally high intra-
(Fig. 20.3, A). Inspiratory resistance (Ri) is generated mainly abdominal pressure and may be related to a nonpulmonary
in the endotracheal tube and in the bronchial tree, with an etiology of respiratory failure.77 Chest wall compliance is
additional viscoelastic or maldistributive component.46 Ri is decreased in patients with COPD exacerbations.78
calculated as the ratio between the pressure drop across the PEEPi is another important measurement in ventilated
airways and the inspiratory flow (V′i). Ri varies with the size patients. This is the pressure that remains in the alveoli at
of the endotracheal tube and with V′i, but values higher than the end of expiration, above the applied level of PEEP.79
10 cm H2O/L/second typically are abnormal. Ri is abnormally PEEPi is caused by prolonged expiration, by flow-limitation
increased in COPD74 and asthma, in the presence of proximal phenomena that are common in COPD patients, by short
airway and endotracheal tube obstruction, and, to a lesser expiratory times, or more commonly by a combination of
degree, in ARDS.52 The elastic recoil of the respiratory system these factors.74
is quantified as compliance (Crs), which is the ratio between During pressure-controlled modes of mechanical ventila-
inspired volume and the corresponding increase of alveolar tion, inspiratory Paw is preset and it is important to monitor
pressure (Palv), or as its reciprocal, elastance. Because the lung volumes to detect respiratory mechanical abnormalities.
and the chest wall are compartments in series, the actual dis- During volume-controlled ventilation modes, it is TV that is
tending pressure of the lung is the transpulmonary pressure fixed, and any alterations of respiratory mechanics manifest
196 Section II—Clinical and Laboratory Assessment

themselves with changes of Paw. In particular, the peak inspi-


ratory pressure (Pip) is the first variable to alert the clinician
that the patient has a mechanical problem. However, Pip is
measured in the proximal airway and its measurement does
not allow for distinction between elastic and resistive prob-
lems because Palv cannot be directly measured. However, Palv

Volume
UIP
can be estimated using the rapid flow interruption technique,47
which provides conditions of zero flow at the end of inspira-
tion and a stable plateau pressure (Plat) (Fig. 20.3, B). In these
conditions, gas is in equilibrium throughout the airways and
Pplat is close to Palv. Pplat is a function of the elastic recoil of
the respiratory system, if respiratory muscles are inactive. LIP
Knowing Pplat allows the computation of Crs because:
Pressure
Crs = TV/[Pplat − PEEP + PEEPi]
Fig. 20.4  Schematic representation of volume-pressure curves in a
healthy subject (blue line) and in a patient with acute respiratory distress
PEEPi should not be disregarded because it leads to falsely low
syndrome (ARDS) (red line). LIP, Lower inflection point; UIP, upper
compliance values if not accounted for. PEEPi is quantified as inflection point.
the difference between the pressure measured during an expi-
ratory flow interruption and the level of PEEP set on the
ventilator (Fig. 20.3, C). Pplat is important because it is the
only bedside method to estimate alveolar distention at the end pressure volume curves do not seem to predict pulmonary
of inspiration. If Pes is available, it is also possible to measure volume during mechanical ventilation.83
Ccw and Cl with the same maneuver. It must be remembered
that Pes can have a bias in the supine position because the
weight of the heart increases its value. However, tidal changes Therapy Based on Respiratory  
in Pes still are valid.
It is possible to assume that the difference between Pip and Mechanics and Ventilator Variables  
Pplat (see Fig. 20.3, B) is the resistive pressure drop across the and the Effect on Outcome
airways and that it can be used to estimate Ri because Ri = Respiratory mechanics measurements can be used to diagnose
(Pip − Pplat)/V′i. It is important that different measurements new conditions, to follow their clinical course, and to monitor
of Ri are obtained at comparable V′i values because its Ri flow the response to therapy. There is also evidence that Crs is
dependent, due to turbulence. Although Crs can be measured better than oxygenation deficit, as a predictor of mortality in
with any type of ventilator mode and any pattern of flow ARDS.38 In daily practice, respiratory mechanics help distin-
delivery, it is preferable to use a volume-controlled mode with guish between processes that have radically different manage-
a constant flow waveform to measure Ri. ment approaches, particularly when multiple abnormalities
The flow interruption method does not require muscle coexist (i.e., bronchospasm in a patient with ARDS). In addi-
paralysis as long as a constant Paw is observed for at least 0.25 tion, the measurement of Ri can be used to monitor the
second.62 When this is not possible, one can temporarily response to bronchodilator therapy.84 The detection of signifi-
increase TV or inspiratory flow to minimize respiratory drive cant levels of PEEPi is very important because this can cause
at the end of inspiration. Ventilator software applications hemodynamic impairment, elevated work of breathing, and a
automatically calculate Crs and Ri, but they may not recognize patient’s inability to trigger the ventilator.85 If not recognized,
a patient’s muscle activity or gas leaks that render the Pplat PEEPi can lead to very poor clinical choices (i.e., unnecessary
measurement void. Thus visual inspection of the tracings and fluid administration in a hypotensive patient). PEEPi can be
manual computation of the variables are advisable. The flow addressed with changes in ventilator settings, such as decreased
interruption maneuver should be repeated multiple times and respiratory rate and inspiratory times, and only in refractory
the results averaged. cases with increased sedation.
Volume-pressure curves in patients with ARDS have a char- Pplat is used to monitor the level of alveolar distention
acteristic biphasic slope (Fig. 20.4): pressure application at low during mechanical ventilation. A TV reaching a Pplat of 30 cm
lung volume yields little increase in volume until a lower H2O should inflate alveoli approximately to the level of total
inflection point (LIP) is reached. At this point compliance lung capacity. Beyond this value the alveolar walls receive a
improves dramatically and the slope of the curve increases. As pathologic stress that generates mechanical and then inflam-
pressure continues to increase, an upper inflection point matory injury that eventually leads to VALI.86 Patients with
(UIP) is reached, where the curve flattens again. The LIP is ARDS are particularly exposed to VALI because of the typical
thought to represent the pressure required for alveolar recruit- decrease in lung volumes, from alveolar edema or atelectasis.55
ment,80 which is the volume of intrapulmonary gas that is Thus ventilation is distributed to a smaller than normal popu-
gained by reopening alveolar units. The UIP is likely related lation of alveolar units, and each inspiration may cause exces-
to excessive alveolar distention.81 These two variables are used sive alveolar distention and stress, even with TVs that are
to identify optimal PEEP and TV settings, with the goal of acceptable in healthy patients. Maintaining Pplat below 30 cm
maximizing alveolar recruitment and minimizing alveolar dis- H2O should guarantee that inspiratory alveolar inflation is not
tention.82 However, the value of the LIP may not correspond greater than “physiologic.” In fact, Pplat and TV limitation is
to the PEEP required to maximize recruitment, and the part of the lung protective strategy that was shown to have
Section II—Clinical and Laboratory Assessment 197

outcome benefits in ARDS.49 Several points deserve discus- indications can be divided into three broad groups: (1) hypoxic
sion. First, a safety limit for Pplat may not exist because an respiratory failure, (2) hypercarbic ventilatory failure (includ-
outcome benefit from TV limitation is observed even when ing cardiac arrest), and (3) impaired consciousness and airway
Pplat is less than 30 cm H2O.87 Second, elevated TVs probably protection.93 In addition, patients in the NCCU may require
are harmful in non-ARDS populations and even in patients intubation to facilitate specific therapies (e.g., to help reduce
with healthy lungs, irrespective of their Pplat values.88 Third, ICP or manage status epilepticus). The decision to intubate
Cl and Ccw both affect Pplat. When Pes and Pl are measured, a patient may be complicated because there are few data
excessive alveolar distention can be identified even in the pres- to support specific guidelines, and in recent years a variety
ence of acceptable Pplat, as suggested by a large change in Pl.72 of noninvasive forms of ventilation have become available.
In these patients further reductions in TV and Pplat may be Nevertheless, the adage “the indication for intubation and
necessary. By contrast, patients with low Ccw, which is typi- mechanical ventilation is thinking of it”94 is a simple guide to
cally due to intra-abdominal hypertension, can have high remember.
Pplat with acceptable lung distention and may not need ven- In the NCCU intubation and mechanical ventilation are
tilator adjustments.77 indicated in the following conditions: (1) depressed con-
It is still unclear how alveolar recruitment should be mea- sciousness and in particular a Glasgow Coma Scale score less
sured and whether any of the available bedside tools can be than or equal to 8; (2) inability to protect the airway or clear
considered acceptable for clinical practice. Early studies sug- secretions; (3) need to reduce ICP by ventilation control; (4)
gested that Crs could be used to define the optimal PEEP PaO2 less than 60 mm Hg despite supplemental O2; (5) PaCO2
setting.89 However, for reasons that are still incompletely greater than 50 mm Hg, or pH less than 7.2; (6) respiratory
understood, other studies did not always detect a correlation rate greater than 40 breaths per minute or less than 10 per
between compliance and recruitment when PEEP was minute; (7) muscle fatigue; (8) airway compromise (e.g., a
changed90. The appeal of the pressure-volume curve has neck hematoma after anterior cervical surgery); and (9)
increased since it was used to set PEEP in clinical trials that hemodynamic instability.
have tested the open lung concept, although there are concerns Although pulse oximetry can be used to assess for hypox-
about the methodology of these studies.58,73 Other techniques emia, it is best assessed using blood gas analysis because oxim-
to assess recruitment at bedside, such as EIT and gas dilution etry measures the saturation of hemoglobin and not PaO2, and
or washout techniques are promising but have not yet been it can be unreliable in patients with severe anemia, carbon
validated in outcome studies. An innovative strategy that monoxide poisoning, methemoglobinemia, sepsis, or periph-
includes Pes measurement in the setting of PEEP has been eral vasoconstriction.95 Furthermore, normal oxygen satura-
proposed.91 Its rationale is that Pes and Pl can be used to tion does not exclude hypoxemia, particularly in patients on
identify the level of PEEP that guarantees adequate expiratory a high FiO2. In these patients evaluation of the alveolar-arterial
lung distention. With this approach the authors demonstrated oxygen gradient can help because a widening alveolar-arterial
a benefit in improved oxygenation and Crs compared with a oxygen gradient indicates worsening hypoxemia. Bedside
control group in which PEEP was set using a conventional monitors to detect hypercarbia are frequently used in the OR
PEEP/FiO2 nomogram. but are less commonly available in the ICU.35 This can be
important because oxygen saturation obtained on pulse oxim-
etry can be normal when there is significant hypoventilation.
Risks Instead the diagnosis of hypoventilation depends on clinical
Monitoring respiratory mechanics and the use of the flow findings and on arterial blood gas analysis.
interruption technique is associated with little risk to the
patient and can be performed in any condition. Measuring Pes
requires the insertion of esophageal catheters. However, these Weaning and Liberation from
are typically thin and soft tipped, and to these authors’ knowl-
edge no cases of morbidity from this method have been
Mechanical Ventilation
reported. Measuring volume-pressure curves may require dis- Weaning often is used to describe the transition from intuba-
connection from the ventilator and so may expose the patient tion and mechanical ventilation to a spontaneous breathing
to hypoxemia and minor hypercarbia, but the procedure is patient who has a protected airway, but strictly speaking it
overall well tolerated by most patients.92 refers to the progressive reduction in the amount of support
provided by a mechanical ventilator. This and liberation from
mechanical ventilation are fundamental aspects of critical care
Decisions About Intubation, and are important because a longer duration of mechanical
Weaning, and Liberation from ventilation is associated with increased cost and mortal-
ity.19,96,97 In addition, up to half of the time spent on mechani-
Mechanical Ventilation cal ventilation happens after the weaning process is started.19,97
Ventilation and pulmonary function are monitored in the Recent consensus guidelines suggest that the weaning process
ICU, in large part to assess the severity of pulmonary disease, should begin shortly after intubation and be considered in six
its progress, and how a patient responds to therapy. In the ICU discrete steps: (1) treatment of acute respiratory failure, (2)
this often means making decisions about intubation and clinical judgment that weaning may be possible, (3) assess-
mechanical ventilation. ment of the readiness to wean, (4) a spontaneous breathing
Which patient should be intubated? The specific indications trial, (5) extubation, and (6) possible reintubation.98
for endotracheal intubation are difficult to define, and a ICU patients may be classified as: (1) simple to wean (i.e.,
variety of parameters need to be considered. In general, the extubated on the first attempt; about 60% of general ICU
198 Section II—Clinical and Laboratory Assessment

Table 20.5  Factors Associated with Readiness Table 20.6  Spontaneous Breathing Trials:
to Wean Ventilator Support in NCCU Patients Criteria That Indicate Failure
CLINICAL ASSESSMENT CLINICAL ASSESSMENT AND SUBJECTIVE INDICES
• Adequate cough • Agitation and anxiety
• Absence of excessive tracheobronchial secretions • Deterioration in mental status
• Resolution of disease for which the patient was intubated • Diaphoresis
• Normal intracranial pressure (ICP) • Cyanosis
• Patient awake and cooperative • Evidence of increasing effort
• Chest wall pain well controlled • Increased accessory muscle activity
• Dyspnea
OBJECTIVE MEASUREMENTS
OBJECTIVE MEASUREMENTS
• Clinical stability
• Stable cardiovascular status (i.e., heart rate ≤140 beats/ • Agitation and anxiety
min; systolic BP 90-160 mm Hg, no or minimal • PaO2 ≤50-60 mm Hg on FiO2 ≥0.5 or SaO2 <90%
vasopressors) • PaCO2 >50 mm Hg or an increase in PaCO2 by >8 mm Hg
• Stable metabolic status • pH <7.32 or a decrease in pH ≥0.07 pH unit
• Adequate oxygenation • Respiratory frequency >35 breath/min or increased by >50%
• SaO2 >90% on FiO2 ≤0.4 • Rapid shallow breathing index >105 breath/min/L
• PaO2 ≥50-60 mm Hg on FiO2 ≤0.5 (or PaO2/FiO2 • HR >140 breaths/min or increased by >20%
≥150 mm Hg) • Systolic BP >180 mm Hg or increased by >20%
• PAO2-PaO2 gradient <350 mm Hg • Systolic BP <90 mm Hg
• PEEP ≤8 mm Hg • Cardiac arrhythmias
• Adequate pulmonary function
• Respiratory rate ≤35 breaths/min Adapted from Boles J, Bion J, Connors A, et al. Weaning from mechanical
• Maximal inspiratory pressure ≤ –20 to –25 cm H2O ventilation. Eur Respir J 2007;29:1033–56.
• VT >5 mL/kg BP, Blood pressure; FiO2, inspiratory oxygen fraction; HR, heart rate; PaCO2,
• VC >10 mL/kg arterial carbon dioxide tension; PaO2, arterial oxygen tension; SaO2, arterial
oxygen saturation.
• Rapid shallow breathing index (respiratory rate/VT) <105
breaths/min/L
• No significant respiratory acidosis

Adapted from Boles J, Bion J, Connors A, et al. Weaning from mechanical


ventilation. Eur Respir J 2007; 29:1033–56. a subjective determination of cough strength. When a patient
BP, Blood pressure; FiO2, inspiratory oxygen fraction; NCCU, neurocritical is difficult to wean (i.e., fails a spontaneous breathing trial or
care unit; PaO2, arterial oxygen tension; PAO2, alveolar oxygen tension; extubation), a search should be made for factors that reduce
PEEP, positive end-expiratory pressure; SaO2, arterial oxygen saturation;
VC, vital capacity; VT, tidal volume. the chances of successful weaning. Management then has to
balance the need for adequate ventilator support (to reduce
respiratory fatigue) against the need to minimize support
(increase respiratory autonomy) to improve the likelihood of
patients), (2) difficult to wean (i.e., up to three attempts or up successful weaning. The various available strategies are beyond
to 7 days from the onset of weaning), or (3) prolonged wean the scope of this chapter but include assist control ventilation
(i.e., more than three attempts or more than 7 days from the (ACV), synchronized intermittent mechanical ventilation
onset of weaning).98 Decisions about the readiness to wean (SIMV), PSV, T-piece trials, use of noninvasive ventilation or
requires two interdependent steps: assessment of predictors of facilitation therapy, and tracheostomy.100,105-110
weaning and successful completion of a spontaneous breath-
ing trial. Factors associated with readiness to wean are listed in
Table 20.5.98,99 No single finding is specific for weaning success.
Conclusions
Instead the factors listed in Table 20.5 identify patients who Mechanical ventilation is crucial to life support and its correct
are not suitable for weaning (i.e., a spontaneous breathing trial management has significant effects on patient outcome. An
in these patients may have adverse effects). All other patients in-depth understanding of basic pulmonary physiology prin-
should undergo a spontaneous breathing trial that may be ciples is therefore essential to the care of neurologic critically
accomplished using a T-tube or T-piece, pressure support ven- ill patients, with and without coexisting pulmonary problems.
tilation (PSV), or automatic tube compensation (ATC). Each Respiratory monitoring allows assessment of the physiologic
of these may or may not be used with continuous positive effects of ventilator settings and, even more importantly, pro-
airway pressure (CPAP). The criteria that indicate failure of a vides the ability to minimize the iatrogenic injury by mechani-
spontaneous breathing trial are listed in Table 20.6.98,100,101 cal ventilation.
The next step in liberation from mechanical ventilation is
extubation. This requires an assessment of the patient’s ability References
to protect and maintain a patent airway based on: (1) level of 1. Hoesch RE, Lin E, Young M, et al. Acute lung injury in critical neurological
consciousness, (2) cough strength, (3) frequency of secretions, illness. Crit Care Med 2012;40(2):587–93.
and (4) airway patency. A Glasgow Coma Scale score greater 2. Pelosi P, Rocco PR, de Abreu MG. Use of computed tomography scanning to
than 8 is associated with a greater likelihood of successful guide lung recruitment and adjust positive-end expiratory pressure. Curr
extubation. A number of tests evaluate cough strength such as Opin Crit Care 2011;17(3):268–74.
3. Luecke T, Corradi F, Pelosi P. Lung imaging for titration of mechanical
card moistening102 or spirometry103 and assess airway patency ventilation. Curr Opin Anaesthesiol 2012;25(2):131–40.
such as determination of a cuff leak.104 However, the cuff leak 4. Bellani G, Mauri T, Pesenti A. Imaging in acute lung injury and acute
test is neither sensitive nor specific and most physicians make respiratory distress syndrome. Curr Opin Crit Care 2012;18(1):29–34.
Section II—Clinical and Laboratory Assessment 199

5. Lundin S, Stenqvist O. Electrical impedance tomography: potentials and 28. Thompson JE, Jaffe MB. Capnographic waveforms in the mechanically
pitfalls. Curr Opin Crit Care 2012;18(1):35–41. ventilated patient. Respir Care 2005;50:100–8; discussion 108–9.
6. Strumwasser A, Chu E, Yeung L, et al. A novel CT volume index score 29. Carlon GC, Ray C, Jr, Miodownik S, et al. Capnography in mechanically
correlates with outcomes in polytrauma patients with pulmonary contusion. ventilated patients. Crit Care Med 1988;16:550–6.
J Surg Res 2011;170(2):280–5. Epub 2011, Apr 5. 30. Anderson CT, Breen PH. Carbon dioxide kinetics and capnography during
7. Stark P, Greene R, Kott MM, et al. Improved control of image optical density critical care. Crit Care 2000;4:207–15.
with low-dose digital and conventional radiography in bedside imaging. 31. Barker SJ, Badal JJ. The measurement of dyshemoglobins and total
CT-findings in ARDS. Radiologe 1987;27(8):367−9. hemoglobin by pulse oximetry. Curr Opin Anaesthesiol 2008;21:805–10.
8. Eisenhuber E, Schaefer-Prokop CM, Prosch H, et al. Bedside chest 32. Jubran A, Tobin MJ. Reliability of pulse oximetry in titrating supplemental
radiography. Respir Care 2012;57(3):427–43. oxygen therapy in ventilator-dependent patients. Chest 1990;97:1420–5.
9. Musch G. Positron emission tomography: a tool for better understanding of 33. Bierman MI, Stein KL, Snyder JV. Pulse oximetry in the postoperative care of
ventilator-induced and acute lung injury. Curr Opin Crit Care 2011;17(1): cardiac surgical patients: a randomized controlled trail. Chest 1992;102:
7–12. 1367–70.
10. de Prost N, Tucci MR, Melo MF. Assessment of lung inflammation with 34. Knapp S, Kofler J, Stoiser B, et al. The assessment of four different methods
18F-FDG PET during acute lung injury. AJR Am J Roentgenol to verify tracheal tube placement in the critical care setting. Anesth Analg
2010;195(2):292–300. 1999;88:766–70.
11. Magaña M, Bercovitch R, Fedullo P. Diagnostic approach to deep venous 35. Husain T, Gatward JJ, Hambidge OR, et al. Strategies to prevent airway
thrombosis and pulmonary embolism in the critical care setting. Crit Care complications: a survey of adult intensive care units in Australia and New
Clin 2011;27(4):841–67, vi. Zealand. Br J Anaesth 2012;108(5):800–6. Epub 2012, Mar 13.
12. Terminella L, Sharma G. Diagnostic studies in patients with acute 36. Wouters PF, Gehring H, Meyfroidt G, et al. Accuracy of pulse oximeters: the
respiratory distress syndrome. Semin Thorac Cardiovasc Surg 2006;18(1): European multi-center trial. Anesth Analg 2002;94:S13–6.
2–7. 37. Van de Louw A, Cracco C, Cerf C, et al. Accuracy of pulse oximetry in the
13. Ruppel GL, Enright PL. Pulmonary function testing. Respir Care 2012;57(1): intensive care unit. Intensive Care Med 2001;27:1606–13.
165–75. doi: 10.4187.respcare.01640.edu 38. Nuckton TJ, Alonso JA, Kallet RH, et al. Pulmonary dead-space fraction as a
14. Barnett N, Ware LB. Biomarkers in acute lung injury: marking forward risk factor for death in the acute respiratory distress syndrome. N Engl J
progress. Crit Care Clin 2011;27(3):661–83. Med 2002;346:1281–6.
15. Raoof S, Feigin D, Sung A, et al. Interpretation of plain chest roentgenogram. 39. Tinker JH, Dull DL, Caplan RA, et al. Role of monitoring devices in
Chest 2012;141(2):545–58. prevention of anesthetic mishaps: a closed claims analysis. Anesthesiology
16. Xirouchaki N, Magkanas E, Vaporidi K, et al. Lung ultrasound in critically ill 1989;71:541–6.
patients: comparison with bedside chest radiography. Intensive Care Med 40. Moller JT, Johannessen NW, Espersen K, et al. Randomized evaluation of
2011;37(9):1488–93. Epub 2011, Aug 2. pulse oximetry in 20,802 patients. II. Perioperative events and postoperative
17. Monchi M, Bellenfant F, Cariou A, et al. Early predictive factors of survival complications. Anesthesiology 1993;78:445–53.
in the acute respiratory distress syndrome: a multivariate analysis. Am J 41. Moller JT, Svennild I, Johannessen NW, et al. Perioperative monitoring with
Respir Crit Care Med 1998;158(4):1076–81. pulse oximetry and late postoperative cognitive dysfunction. Br J Anaesth
18. Raghavendran K, Napolitano LM. Definition of ALI/ARDS. Crit Care Clin 1993;71:340–7.
2011;27(3):429–37. 42. Pedersen T, Dyrlund Pedersen B, et al. Pulse oximetry for perioperative
19. Esteban A, Anzueto A, Frutos F, et al: Characteristics and outcomes in adult monitoring. Cochrane Database Syst Rev 2003;(3):CD002013.
patients receiving mechanical ventilation: a 28-day international study. 43. Pedersen T, Møller AM, Hovhannisyan K. Pulse oximetry for perioperative
JAMA 2002;287:345–55. monitoring. Cochrane Database Syst Rev 2009;(4):CD002013.
20. Seeley E, McAuley DF, Eisner M, et al. Predictors of mortality in acute lung 44. Mead J, Whittenberger JL, Radford EP, Jr. Surface tension as a factor in
injury during the era of lung protective ventilation. Thorax 2008;63(11): pulmonary volume-pressure hysteresis. J Appl Physiol 1957;10:191–6.
994–8. 45. Matamis D, Lemaire F, Harf A, et al. Total respiratory pressure-volume
21. Oddo M, Nduom E, Frangos S, et al. Lung function and brain oxygen after curves in the adult respiratory distress syndrome. Chest 1984;86:58–66.
severe traumatic brain injury. Neurosurgery 2010;67:338–44. 46. Bates JH, Rossi A, Milic-Emili J. Analysis of the behavior of the respiratory
22. Bernard GR, Artigas A, Brigham KL, et al. The American-European system with constant inspiratory flow. J Appl Physiol 1985;58:1840–8.
consensus conference on ARDS: definitions, mechanisms, relevant 47. D’Angelo E, Calderini E, Torri G, et al. Respiratory mechanics in
outcomes, and clinical trial coordination. Am J Respir Crit Care Med 1994; anesthetized paralyzed humans: effects of flow, volume, and time. J Appl
149:818–24. Physiol 1989;67:2556–64.
23. Murray JF, Matthay MA, Luce JM, et al. An expanded definition of the adult 48. Dreyfuss D, Saumon G. Ventilator-induced lung injury: lessons from
respiratory distress syndrome. Am Rev Respir Dis 1988;138:720–3. experimental studies. Am J Respir Crit Care Med 1998;157:294–323.
24. Ferguson ND, Davis AM, Slutsky AS, et al. Development of a clinical 49. The Acute Respiratory Distress Syndrome Network. Ventilation with lower
definition for acute respiratory distress syndrome using the Delphi tidal volumes as compared with traditional tidal volumes for acute lung
technique. J Crit Care 2005;20(2):147–54. injury and the acute respiratory distress syndrome. the acute respiratory
25. Trachsel D, McCrindle BW, Nakagawa S, et al. Oxygenation index predicts distress syndrome network. N Engl J Med 2000;342:1301–8.
outcome in children with acute hypoxemic respiratory failure. Am J Respir 50. Benditt JO. Esophageal and gastric pressure measurements. Respir Care
Crit Care Med 2005;172:206–11. 2005;50:68–75; discussion 75–7.
26. Severinghaus JW. The invention and development of blood gas analysis
apparatus. Anesthesiology 2002;97:253–6. A complete list of references for this chapter can be found online at
27. Tremper KK, Barker SJ. Pulse oximetry. Anesthesiology 1989;70:98–108. www.expertconsult.com.
Section II—Clinical and Laboratory Assessment 199.e1

References 31. Barker SJ, Badal JJ. The measurement of dyshemoglobins and total
hemoglobin by pulse oximetry. Curr Opin Anaesthesiol 2008;21:805–10.
1. Hoesch RE, Lin E, Young M, et al. Acute lung injury in critical neurological 32. Jubran A, Tobin MJ. Reliability of pulse oximetry in titrating
illness. Crit Care Med 2012;40(2):587–93. supplemental oxygen therapy in ventilator-dependent patients. Chest 1990;
2. Pelosi P, Rocco PR, de Abreu MG. Use of computed tomography scanning 97:1420–5.
to guide lung recruitment and adjust positive-end expiratory pressure. Curr 33. Bierman MI, Stein KL, Snyder JV. Pulse oximetry in the postoperative care
Opin Crit Care 2011;17(3):268–74. of cardiac surgical patients: a randomized controlled trail. Chest 1992;102:
3. Luecke T, Corradi F, Pelosi P. Lung imaging for titration of mechanical 1367–70.
ventilation. Curr Opin Anaesthesiol 2012;25(2):131–40. 34. Knapp S, Kofler J, Stoiser B, et al. The assessment of four different methods
4. Bellani G, Mauri T, Pesenti A. Imaging in acute lung injury and acute to verify tracheal tube placement in the critical care setting. Anesth Analg
respiratory distress syndrome. Curr Opin Crit Care 2012;18(1):29–34. 1999;88:766–70.
5. Lundin S, Stenqvist O. Electrical impedance tomography: potentials and 35. Husain T, Gatward JJ, Hambidge OR, et al. Strategies to prevent airway
pitfalls. Curr Opin Crit Care 2012;18(1):35–41. complications: a survey of adult intensive care units in Australia and New
6. Strumwasser A, Chu E, Yeung L, et al. A novel CT volume index score Zealand. Br J Anaesth 2012;108(5):800–6. Epub 2012, Mar 13.
correlates with outcomes in polytrauma patients with pulmonary 36. Wouters PF, Gehring H, Meyfroidt G, et al. Accuracy of pulse oximeters: the
contusion. J Surg Res 2011;170(2):280–5. Epub 2011, Apr 5. European multi-center trial. Anesth Analg 2002;94:S13–6.
7. Stark P, Greene R, Kott MM, et al. Improved control of image optical 37. Van de Louw A, Cracco C, Cerf C, et al. Accuracy of pulse oximetry in the
density with low-dose digital and conventional radiography in bedside intensive care unit. Intensive Care Med 2001;27:1606–13.
imaging. CT-findings in ARDS. Radiologe 1987;27(8):367−9. 38. Nuckton TJ, Alonso JA, Kallet RH, et al. Pulmonary dead-space fraction as
8. Eisenhuber E, Schaefer-Prokop CM, Prosch H, et al. Bedside chest a risk factor for death in the acute respiratory distress syndrome. N Engl J
radiography. Respir Care 2012;57(3):427–43. Med 2002;346:1281–6.
9. Musch G. Positron emission tomography: a tool for better understanding 39. Tinker JH, Dull DL, Caplan RA, et al. Role of monitoring devices in
of ventilator-induced and acute lung injury. Curr Opin Crit Care 2011; prevention of anesthetic mishaps: a closed claims analysis. Anesthesiology
17(1):7–12. 1989;71:541–6.
10. de Prost N, Tucci MR, Melo MF. Assessment of lung inflammation with 40. Moller JT, Johannessen NW, Espersen K, et al. Randomized evaluation of
18F-FDG PET during acute lung injury. AJR Am J Roentgenol 2010;195(2): pulse oximetry in 20,802 patients. II. Perioperative events and postoperative
292–300. complications. Anesthesiology 1993;78:445–53.
11. Magaña M, Bercovitch R, Fedullo P. Diagnostic approach to deep venous 41. Moller JT, Svennild I, Johannessen NW, et al. Perioperative monitoring with
thrombosis and pulmonary embolism in the critical care setting. Crit Care pulse oximetry and late postoperative cognitive dysfunction. Br J Anaesth
Clin 2011;27(4):841–67, vi. 1993;71:340–7.
12. Terminella L, Sharma G. Diagnostic studies in patients with acute 42. Pedersen T, Dyrlund Pedersen B, et al. Pulse oximetry for perioperative
respiratory distress syndrome. Semin Thorac Cardiovasc Surg 2006;18(1): monitoring. Cochrane Database Syst Rev 2003;(3):CD002013.
2–7. 43. Pedersen T, Møller AM, Hovhannisyan K. Pulse oximetry for perioperative
13. Ruppel GL, Enright PL. Pulmonary function testing. Respir Care 2012; monitoring. Cochrane Database Syst Rev 2009;(4):CD002013.
57(1):165–75. doi: 10.4187.respcare.01640.edu 44. Mead J, Whittenberger JL, Radford EP, Jr. Surface tension as a factor in
14. Barnett N, Ware LB. Biomarkers in acute lung injury: marking forward pulmonary volume-pressure hysteresis. J Appl Physiol 1957;10:191–6.
progress. Crit Care Clin 2011;27(3):661–83. 45. Matamis D, Lemaire F, Harf A, et al. Total respiratory pressure-volume
15. Raoof S, Feigin D, Sung A, et al. Interpretation of plain chest curves in the adult respiratory distress syndrome. Chest 1984;86:58–66.
roentgenogram. Chest 2012;141(2):545–58. 46. Bates JH, Rossi A, Milic-Emili J. Analysis of the behavior of the respiratory
16. Xirouchaki N, Magkanas E, Vaporidi K, et al. Lung ultrasound in critically system with constant inspiratory flow. J Appl Physiol 1985;58:1840–8.
ill patients: comparison with bedside chest radiography. Intensive Care Med 47. D’Angelo E, Calderini E, Torri G, et al. Respiratory mechanics in
2011;37(9):1488–93. Epub 2011, Aug 2. anesthetized paralyzed humans: effects of flow, volume, and time. J Appl
17. Monchi M, Bellenfant F, Cariou A, et al. Early predictive factors of survival Physiol 1989;67:2556–64.
in the acute respiratory distress syndrome: a multivariate analysis. Am J 48. Dreyfuss D, Saumon G. Ventilator-induced lung injury: lessons from
Respir Crit Care Med 1998;158(4):1076–81. experimental studies. Am J Respir Crit Care Med 1998;157:294–323.
18. Raghavendran K, Napolitano LM. Definition of ALI/ARDS. Crit Care Clin 49. The Acute Respiratory Distress Syndrome Network. Ventilation with lower
2011;27(3):429–37. tidal volumes as compared with traditional tidal volumes for acute lung
19. Esteban A, Anzueto A, Frutos F, et al: Characteristics and outcomes in adult injury and the acute respiratory distress syndrome. the acute respiratory
patients receiving mechanical ventilation: a 28-day international study. distress syndrome network. N Engl J Med 2000;342:1301–8.
JAMA 2002;287:345–55. 50. Benditt JO. Esophageal and gastric pressure measurements. Respir Care
20. Seeley E, McAuley DF, Eisner M, et al. Predictors of mortality in acute lung 2005;50:68–75; discussion 75–7.
injury during the era of lung protective ventilation. Thorax 2008;63(11): 51. Loring SH, O’Donnell CR, Behazin N, et al. Esophageal pressures in acute
994–8. lung injury: do they represent artifact or useful information about
21. Oddo M, Nduom E, Frangos S, et al. Lung function and brain oxygen after transpulmonary pressure, chest wall mechanics, and lung stress? J Appl
severe traumatic brain injury. Neurosurgery 2010;67:338–44. Physiol 2010;108(3):515–22. Epub 2009, Dec 17.
22. Bernard GR, Artigas A, Brigham KL, et al. The American-European 52. Pelosi P, Cereda M, Foti G, et al. Alterations of lung and chest wall
consensus conference on ARDS: definitions, mechanisms, relevant mechanics in patients with acute lung injury: effects of positive end-
outcomes, and clinical trial coordination. Am J Respir Crit Care Med 1994; expiratory pressure. Am J Respir Crit Care Med 1995;152:531–7.
149:818–24. 53. Unroe M, MacIntyre N. Evolving approaches to assessing and
23. Murray JF, Matthay MA, Luce JM, et al. An expanded definition of the monitoring patient-ventilator interactions. Curr Opin Crit Care 2010;16(3):
adult respiratory distress syndrome. Am Rev Respir Dis 1988;138:720–3. 261–8.
24. Ferguson ND, Davis AM, Slutsky AS, et al. Development of a clinical 54. Galetke W, Randerath WJ, Feier C, et al. Esophageal pressure method and
definition for acute respiratory distress syndrome using the Delphi impulse oscillometry to assess mechanical properties of the respiratory
technique. J Crit Care 2005;20(2):147–54. system in healthy men. Med Sci Monit 2009;15(8):CR429–35.
25. Trachsel D, McCrindle BW, Nakagawa S, et al. Oxygenation index predicts 55. Gattinoni L, Caironi P, Pelosi P, et al. What has computed tomography
outcome in children with acute hypoxemic respiratory failure. Am J Respir taught us about the acute respiratory distress syndrome? Am J Respir Crit
Crit Care Med 2005;172:206–11. Care Med 2001;164:1701–11.
26. Severinghaus JW. The invention and development of blood gas analysis 56. Crotti S, Mascheroni D, Caironi P, et al. Recruitment and derecruitment
apparatus. Anesthesiology 2002;97:253–6. during acute respiratory failure: a clinical study. Am J Respir Crit Care Med
27. Tremper KK, Barker SJ. Pulse oximetry. Anesthesiology 1989;70:98–108. 2001;164:131–40.
28. Thompson JE, Jaffe MB. Capnographic waveforms in the mechanically 57. Muscedere JG, Mullen JB, Gan K, et al. Tidal ventilation at low airway
ventilated patient. Respir Care 2005;50:100–8; discussion 108–9. pressures can augment lung injury. Am J Respir Crit Care Med 1994;149:
29. Carlon GC, Ray C, Jr, Miodownik S, et al. Capnography in mechanically 1327–34.
ventilated patients. Crit Care Med 1988;16:550–6. 58. Amato MB, Barbas CS, Medeiros DM, et al. Effect of a protective-
30. Anderson CT, Breen PH. Carbon dioxide kinetics and capnography during ventilation strategy on mortality in the acute respiratory distress syndrome.
critical care. Crit Care 2000;4:207–15. N Engl J Med 1998;338:347–54.
199.e2 Section II—Clinical and Laboratory Assessment

59. Brower RG, Lanken PN, MacIntyre N, et al. Higher versus lower positive 84. Dhand R, Duarte AG, Jubran A, et al. Dose-response to bronchodilator
end-expiratory pressures in patients with the acute respiratory distress delivered by metered-dose inhaler in ventilator-supported patients. Am J
syndrome. N Engl J Med 2004;351:327–36. Respir Crit Care Med 1996;154:388–93.
60. Chiumello D, Cressoni M, Chierichetti M, et al. Nitrogen washout/washin, 85. MacIntyre NR, Cheng KC, McConnell R. Applied PEEP during pressure
helium dilution and computed tomography in the assessment of end support reduces the inspiratory threshold load of intrinsic PEEP. Chest
expiratory lung volume. Crit Care 2008;12:R150. 1997;111:188–93.
61. Bodenstein M, David M, Markstaller K. Principles of electrical impedance 86. Marini JJ, Gattinoni L. Ventilatory management of acute respiratory distress
tomography and its clinical application. Crit Care Med 2009;37(2):713–24. syndrome: a consensus of two. Crit Care Med 2004;32:250–5.
62. Younes M, Webster K, Kun J, et al. A method for measuring passive 87. Hager DN, Krishnan JA, Hayden DL, et al. Tidal volume reduction in
elastance during proportional assist ventilation. Am J Respir Crit Care Med patients with acute lung injury when plateau pressures are not high. Am J
2001;164:50–60. Respir Crit Care Med 2005;172:1241–5.
63. Chieveley-Williams S, Dinner L, Puddicombe A, et al. Central venous and 88. Gajic O, Dara SI, Mendez JL, et al. Ventilator-associated lung injury in
bladder pressure reflect transdiaphragmatic pressure during pressure patients without acute lung injury at the onset of mechanical ventilation.
support ventilation. Chest 2002;121:533–8. Crit Care Med 2004;32:1817–24.
64. Sydow M, Burchardi H, Zinserling J, et al. Improved determination of static 89. Suter PM, Fairley B, Isenberg MD. Optimum end-expiratory airway
compliance by automated single volume steps in ventilated patients. pressure in patients with acute pulmonary failure. N Engl J Med 1975;292:
Intensive Care Med 1991;17:108–14. 284–9.
65. Harris RS, Hess DR, Venegas JG. An objective analysis of the pressure- 90. Katz JA, Ozanne GM, Zinn SE, et al. Time course and mechanisms of
volume curve in the acute respiratory distress syndrome. Am J Respir Crit lung-volume increase with PEEP in acute pulmonary failure.
Care Med 2000;161:432–9. Anesthesiology 1981;54:9–16.
66. Grivans C, Lundin S, Stenqvist O, et al. Positive end-expiratory pressure- 91. Talmor D, Sarge T, Malhotra A, et al. Mechanical ventilation guided by
induced changes in end-expiratory lung volume measured by spirometry esophageal pressure in acute lung injury. N Engl J Med 2008;359:2095–104.
and electric impedance tomography. Acta Anaesthesiol Scand 92. Lee WL, Stewart TE, MacDonald R, et al. Safety of pressure-volume curve
2011;55(9):1068–77. doi:10.1111/j.1399–6576.2011.02511.x. Epub 2011, measurement in acute lung injury and ARDS using a syringe technique.
Sep 7. Chest 2002;121:1595–601.
67. Pulletz S, Adler A, Kott M, et al. Regional lung opening and closing 93. American College of Surgeons. ATLS: advanced trauma life support for
pressures in patients with acute lung injury. J Crit Care 2011;Oct 25. Epub doctors. 8th ed. Chicago: ACS; 2010.
ahead of print. 94. Marino PL. Principles of mechanical ventilation. In: Marino PL, editor. The
68. Moerer O, Hahn G, Quintel M. Lung impedance measurements to monitor ICU book. Philadelphia: Lippincott Williams & Wilkins; 2007. p. 461.
alveolar ventilation. Curr Opin Crit Care 2011;17(3):260–7. 95. Wilson BJ, Cowan HJ, Lord JA, et al. The accuracy of pulse oximetry in
69. Lowhagen K, Lindgren S, Odenstedt H, et al. A new non-radiological emergency department patients with severe sepsis and septic shock: a
method to assess potential lung recruitability: a pilot study in ALI patients. retrospective cohort study. BMC Emerg Med 2010;10:9.
Acta Anaesthesiol Scand 2011;55(2):165–74. 96. Cooper LM, Linde-Zwirble WT. Medicare intensive care unit use: analysis
70. Georgopoulos D, Prinianakis G, Kondili E. Bedside waveforms of incidence, cost, and payment. Crit Care Med 2004;32:2247–53.
interpretation as a tool to identify patient-ventilator asynchronies. Intensive 97. Ely EW, Baker AM, Dunagan DP, et al. Effect on the duration of mechanical
Care Med 2006;32:34–47. ventilation of identifying patients capable of breathing spontaneously. N
71. Talmor D, Sarge T, O’Donnell CR, et al. Esophageal and transpulmonary Engl J Med 1996;335:1864–9.
pressures in acute respiratory failure. Crit Care Med 2006;34:1389–94. 98. Boles JM, Bion J, Connors A, et al. Weaning from mechanical ventilation.
72. Chiumello D, Carlesso E, Cadringher P, et al. Lung stress and strain during Eur Respir J 2007;29:1033–56.
mechanical ventilation for acute respiratory distress syndrome. Am J Respir 99. Monaco F, Drummond GB, Ramsay P, et al. Do simple ventilation and gas
Crit Care Med 2008;178:346–55. exchange measurements predict early successful weaning from respiratory
73. Villar J, Kacmarek RM, Perez-Mendez L, et al. A high positive end- support in unselected general intensive care patients? Br J Anaesth 2010;
expiratory pressure, low tidal volume ventilatory strategy improves 105(3):326–33. Epub 2010, Jul 22.
outcome in persistent acute respiratory distress syndrome: a randomized, 100. Esteban A, Frutos F, Tobin MJ, et al. A comparison of four methods of
controlled trial. Crit Care Med 2006;34:1311–18. weaning patients from mechanical ventilation. Spanish Lung Failure
74. Broseghini C, Brandolese R, Poggi R, et al. Respiratory mechanics during Collaborative Group. N Engl J Med 1995;332:345–50.
the first day of mechanical ventilation in patients with pulmonary edema 101. Yang KL, Tobin MJ. A prospective study of indexes predicting the outcome
and chronic airway obstruction. Am Rev Respir Dis 1988;138:355–61. of trials of weaning from mechanical ventilation. N Engl J Med 1991;324:
75. Murray JF, Matthay MA, Luce JM, et al. An expanded definition of the 1445–50.
adult respiratory distress syndrome. Am Rev Respir Dis 1988;138:720–3. 102. Khamiees M, Raju P, DeGirolamo A, et al. Predictors of extubation
76. Polese G, Rossi A, Appendini L, et al. Partitioning of respiratory mechanics outcome in patients who have successfully completed a spontaneous
in mechanically ventilated patients. J Appl Physiol 1991;71:2425–33. breathing trial. Chest 2001;120:1262–70.
77. Gattinoni L, Pelosi P, Suter PM, et al. Acute respiratory distress syndrome 103. Smina M, Salam A, Khamiees M, et al. Cough peak flows and extubation
caused by pulmonary and extrapulmonary disease. different syndromes? outcomes. Chest 2003;124:262–8
Am J Respir Crit Care Med 1998;158:3–11. 104. Fisher MM, Raper RF. The “cuff-leak” test for extubation. Anaesthesia 1992;
78. Musch G, Foti G, Cereda M, et al. Lung and chest wall mechanics in 47:10–2.
normal anaesthetized subjects and in patients with COPD at different PEEP 105. Brochard L, Rauss A, Benito S, et al. Comparison of three methods of
levels. Eur Respir J 1997;10:2545–52. gradual withdrawal from ventilatory support during weaning from
79. Pepe PE, Marini JJ. Occult positive end-expiratory pressure in mechanically mechanical ventilation. Am J Respir Crit Care Med 1994;150:896–903.
ventilated patients with airflow obstruction: the auto-PEEP effect. Am Rev 106. Trevisan CE, Vieira SR. Noninvasive mechanical ventilation may be useful
Respir Dis 1982;126:166–70. in treating patients who fail weaning from invasive mechanical ventilation:
80. Gattinoni L, Pesenti A, Avalli L, et al. Pressure-volume curve of total a randomized clinical trial. Crit Care 2008;12:R51.
respiratory system in acute respiratory failure. computed tomographic scan 107. Agarwal R, Aggarwal AN, Gupta D, et al Role of noninvasive positive-
study. Am Rev Respir Dis 1987;136:730–6. pressure ventilation in postextubation respiratory failure: a meta-analysis.
81. Roupie E, Dambrosio M, Servillo G, et al. Titration of tidal volume and Respir Care 2007;52:1472–9.
induced hypercapnia in acute respiratory distress syndrome. Am J Respir 108. Burns KE, Adhikari NK, Meade MO. A meta-analysis of noninvasive
Crit Care Med 1995;152:121–8. weaning to facilitate liberation from mechanical ventilation. Can J Anaesth
82. Richard JC, Brochard L, Vandelet P, et al. Respective effects of end- 2006;53:305–15.
expiratory and end-inspiratory pressures on alveolar recruitment in acute 109. Rizk EB, Patel AS, Stetter CM, et al. Impact of tracheostomy timing on
lung injury. Crit Care Med 2003;31:89–92. outcome after severe head injury. Neurocrit Care 2011;15(3):481–9.
83. Jonson B, Richard JC, Straus C, et al. Pressure-volume curves and 110. Cabello B, Thille AW, Roche-Campo F, et al. Physiological comparison of
compliance in acute lung injury: evidence of recruitment above the lower three spontaneous breathing trials in difficult-to-wean patients. Intensive
inflection point. Am J Respir Crit Care Med 1999;159:1172–8. Care Med 2010;36(7):1171–9. Epub 2010, Mar 30.
II
Chapter
21  

Endocrine Evaluation
Matthew R. Sanborn and Carrie A. Sims

Introduction particularly those with severe neurologic disorders, sepsis, or


multiorgan dysfunction, are likely to have physiologic changes
The neuroendocrine system is important for body homeosta- rarely, if ever, seen in patients in an outpatient setting. Second,
sis but can undergo significant changes in response to critical it is unclear whether standard tests of endocrine function that
illness. During the acute phase of critical illness or trauma, primarily are designed to assess patients in the outpatient
anabolic hormonal pathways are down-regulated in favor setting (i.e., not under “stress”) are valid in critical care. For
of fulfilling more immediate needs. With prolonged critical example, basal levels of hormones and regulation of hormone
illness, the hypothalamic-pituitary axis (HPA) is subject to responsiveness (i.e., feedback regulation) cannot be assumed
reduced pulsatile secretion of pituitary hormones, reduced to be the same in the typical patient undergoing outpatient
peripheral activity, and global hormonal suppression. If evaluation and one in the ICU. Third, although hormone
unrecognized, HPA dysfunction can lead to problems in both levels in the circulation depend in part on synthesis, they also
the short and long term. However, monitoring the endocrine are influenced by levels of binding proteins1 and tissue-specific
status of critically ill patients can be a challenge. First, pituitary metabolism,2 both of which can be altered by critical illness.
dysfunction often can mimic deficits associated with the neu- Fourth, it is unclear whether adequate serum levels equate
rologically injured patient (Table 21.1). In addition, critical with tissue levels. For example, how some hormone receptors
illness may induce endocrine dysfunction that is not observed respond to their ligands can be altered during sepsis and
outside the intensive care unit (ICU). Therefore a high index hypoxia,3 and hormone metabolism, in particular that of
of suspicion or a screening protocol is necessary. Second, thyroid and cortisol, is altered at the tissue level in critically ill
many of the signs and symptoms of endocrine dysfunction are patients.2,4 Fifth, endocrine assessment depends on measure-
subtle, may be delayed in onset, or may be masked by inter- ment of basal hormone levels and often dynamic tests (i.e.,
ventions common in the acute setting. Third, assays used in stimulation or inhibition of hormone synthesis). These
healthy patients may underestimate hormone levels in the various tests normally are performed under standard condi-
acutely ill, and standards used to define appropriate normal tions such as fasting without the interference of other medica-
values may not be applicable. Although there are few widely tions, and at a particular time. These conditions may be
adopted management guidelines, protocol-driven screening of difficult to replicate in the ICU. Furthermore, several dynamic
the at-risk patient may avoid the potential for misdiagnosis tests (e.g., insulin tolerance test or metyrapone test) used in
and so influence outcome. There are, however, two important the outpatient setting may be dangerous to use in the critical
questions in management: (1) does endocrine intervention care patient. Sixth, there can be a delay in turnaround of
improve outcome when there is critical illness–induced endo- hormone assay results, and hence empiric treatment may be
crine dysfunction, and (2) does endocrine intervention help needed. Seventh, any critical illness is a dynamic process, and
critically ill patients without endocrine dysfunction (e.g., physiologic responses vary and evolve over time. Consistent
steroid use) to improve hemodynamics? with this, cortisol levels are well known to vary in the same
This chapter reviews endocrine deficiencies frequently patient during a 24-hour period. This implies that frequent
encountered in patients in the neurocritical care unit (NCCU) samples may be needed but with that comes an increased risk
and discusses the endocrine consequences of traumatic brain of false-positive results. Finally, various medications and ther-
injury (TBI) and subarachnoid hemorrhage (SAH). This is apies given to patients in the ICU may alter the accuracy of
important because nearly all endocrine axes have close links laboratory testing of endocrine function5-7 (Table 21.2). In
with the central nervous system including regulation of pitu- addition, results may differ across laboratories because stan-
itary hormone production and direct innervation of endo- dardization of assays may vary. Together these limitations
crine glands. Glycemic control and nutrition are addressed in mean that the clinician in the NCCU must pay careful atten-
Chapter 14. tion to the whole patient rather than simply interpreting indi-
vidual hormone levels.
Practicalities of Endocrine The challenges facing laboratory staff, including preana-
lytic, analytic, postanalytic and those associated with assay
Assessment in the Critical Care Unit methodology are reviewed elsewhere.5 A key issue, despite
Several practical difficulties are associated with endocrine considerable effort, is that units used for reporting assay
assessment in the critical care unit. First, patients in the NCCU, results and assay standardizations vary around the world.
200 © Copyright 2013 Elsevier Inc. All rights reserved.
Section II—Clinical and Laboratory Assessment 201

Table 21.1  Signs and Symptoms of Hypothalamic-Pituitary Axis Dysfunction


Hormone Deficiency Symptoms Signs
Adrenocorticotropic Weakness, nausea, fever, shock, abdominal Hypotension, hypoglycemia, hyponatremia,
hormone (ACTH) pain, headache, adrenal crisis myopathy, anemia, eosinophilia
Thyroid hormone Anergy, neuropsychiatric symptoms, cold Bradycardia, hypotension, neuropathy, myopathy
intolerance, memory problems, weight gain
Growth hormone Anergy, poor quality of life Osteoporosis, reduced lean body mass, dyslipidemia
Follicle-stimulating and Oligomenorrhea or amenorrhea, sexual Reduced muscle mass and exercise tolerance,
luteinizing hormone dysfunction, mood disorders osteoporosis, infertility, loss of secondary hair

superior hypophyseal arteries. The anterior pituitary contains


Table 21.2  Common Drugs Used in the glandular cells that respond to inhibitory and releasing factors
Intensive Care Unit That May Affect secreted by the arcuate, periventricular, medial preoptic, and
Endocrine Function paraventricular nuclei from the hypothalamus. The axons of
Adrenal Dysfunction Thyroid Dysfunction the neurons located within these nuclei project to the median
eminence, where they release secretory and inhibitory factors
Heparins Heparins
into a capillary plexus. This median eminence capillary plexus
Warfarin and other Iodinated contrast dye then drains into a second capillary plexus within the anterior
anticoagulants pituitary via the hypophyseal portal veins. The hypothalamic
Phenobarbital, phenytoin Phenobarbital, phenytoin, factors then stimulate or inhibit the glandular cells of the
carbamazepine anterior pituitary and modify secretion of hormones into the
Azole antifungals Amiodarone secondary capillary plexus. Ultimately, the hypophyseal portal
Etomidate Corticosteroids vein drains to the cavernous sinus and to the systemic circula-
tion through the internal jugular vein.
Glucocorticoid therapy Dobutamine, dopamine
Opiates and benzodiazepines Furosemide
Statins Octreotide Corticotroph Dysfunction
Rifampin Lithium, atypical antipsychotics
Rifampin
Etiology
Decreased or inappropriate cortisol secretion is the hall-
mark of adrenal insufficiency. Adrenal insufficiency may be
due to processes that affect: (1) the adrenal gland itself
Furthermore, there can be variation in assay performance (primary adrenal insufficiency), (2) the pituitary gland and
based on the manufacturer. Consequently there are interna- secretion of adrenocorticotropic hormone (ACTH) (second-
tional scientific efforts to standardize assays in which assays ary adrenal insufficiency), or (3) the hypothalamus and
are compared against a candidate reference measurement pro- its secretion of corticotropin-releasing hormone (tertiary
cedure. This means that a given substance, for example, free adrenal insufficiency).
cortisol, measured in a given system such as serum is collected Acute primary adrenal insufficiency may result from
under defined conditions, and the units used to report the bilateral adrenal hemorrhage or infarction secondary to
levels are traceable to a reference measurement procedure trauma, coagulopathy, thromboembolic disease, or sepsis
and a reference preparation that are certified by such organi- (e.g., Waterhouse-Friderichsen syndrome). Waterhouse-
zations as the Joint Committee for Traceability in Laboratory Friderichsen syndrome was first described in the setting of
Medicine. meningococcemia, but also has been reported in several
other systemic bacterial infections including those caused
by Pseudomonas aeruginosa, Streptococcus pneumoniae, Neis-
Anterior Pituitary Dysfunction seria gonorrhoeae, Escherichia coli, Haemophilus influenzae,
and Staphylococcus aureus.8 Several drugs can cause adrenal
Anatomy insufficiency by inhibiting cortisol biosynthesis (e.g., etomi-
The pituitary gland is divided into two functionally, physio- date, ketoconazole, metyrapone, suramin) or by accelerating
logically, and developmentally distinct units: the anterior pitu- its metabolism (e.g., phenytoin, barbiturates, rifampin).9-14
itary, or adenohypophysis, and the posterior pituitary, or Any disease process or medication that interferes with
neurohypophysis. The anterior pituitary develops from ecto- ACTH secretion can cause secondary adrenal insufficiency.
dermal cells that overlie the pharynx that invaginate to form Infection, TBI, tumors, infiltrative diseases, hemorrhage, and
Rathke’s pouch. The posterior wall of Rathke’s pouch also infarction each may damage the pituitary gland.15 Glucocor-
forms a small intermediate lobe that secretes melanocyte- ticoid use is the most common cause of drug-induced second-
stimulating hormone in the fetus, but this regresses in the ary adrenal insufficiency. Exogenous steroids suppress the
adult. The pituitary gland derives its blood supply from two HPA that leads to atrophy of ACTH-producing cells. Second-
branches of the internal carotid artery: the inferior and ary insufficiency can occur when glucocorticoid therapy is
202 Section II—Clinical and Laboratory Assessment

Table 21.3  Recommendations for Perioperative Steroid Coverage


Degree of Surgical Stress Dose* Duration
Minor (e.g., inguinal herniorrhaphy) 25 mg hydrocortisone daily 1 day
Moderate (total knee replacement, abdominal hysterectomy, open 50-75 mg hydrocortisone daily 1 to 2 days
cholecystectomy)
Major (pancreaticoduodenectomy, cardiac surgery with bypass) 100-150 mg hydrocortisone daily 2 to 3 days

Adapted from Salem M, Tainsh RE, Jr., Bromberg J, et al. Perioperative glucocorticoid coverage: a reassessment 42 years after emergence of a problem. Ann Surg
1994;219:416–25.
*Hydrocortisone supplementation should be administered in divided daily doses.

tapered or when physiologic stress exceeds the maintenance poor if glucocorticoid and mineralocorticoid insufficiency is
dose. Consequently, patients who have been on chronic ste- not diagnosed and treated.23,24
roids require a “stress-dose” perioperatively or when admitted
to the NCCU (Table 21.3).
Any process that inhibits the hypothalamic production of Monitoring the Hypothalamic-
corticotropin-releasing hormone (CRH), such as tumors,
infiltrative diseases, e.g., sarcoidosis, cranial irradiation, and
Pituitary-Adrenal Axis in the ICU
chronic high-dose glucocorticoid therapy16 will result in ter- Considerable controversy surrounds the utility, optimal
tiary adrenal insufficiency. Without the ACTH-secretagogue method, and interpretation of hypothalamic-pituitary-
action of CRH on the anterior pituitary, ACTH levels decline, adrenal axis testing in the critically ill patient. In the healthy,
endogenous cortisol levels fall, and the adrenal glands non-ICU patient the initial diagnostic tests for adrenal insuf-
atrophy. ficiency include morning plasma ACTH and serum cortisol
Dysfunction of the HPA in the critically ill patient in the levels. In general, primary adrenal insufficiency can be dis-
absence of glucocorticoid treatment or structural damage to tinguished from secondary disease by an elevation in plasma
the adrenal glands, pituitary, or hypothalamus is an increas- ACTH levels. A cortisol level less than 100 nmol/L (or
ingly recognized phenomenon. The condition has been 3.6 µg/dL) indicates adrenal insufficiency, whereas a value
described as relative adrenal insufficiency (RAI), or critical greater than 500 nmol/L (or 18 µg/dL) suggests an intact
illness-related corticosteroid insufficiency (CIRCI). These dis- hypothalamic-pituitary-adrenal axis.25 However, in the ICU
orders are common in sepsis and are the basis of trials of patient a normal or even elevated cortisol does not mean
glucocorticoids in septic shock. The diagnosis is based on there will be an appropriate response to the stress of surgery
plasma cortisol profiles, but there are currently no tests that or critical illness. The appropriate cortisol level for critically
clearly identify which patient is truly glucocorticoid “defi- ill patients is unknown, and tests used to diagnose adreno-
cient” at a cellular level. A consensus statement defined CIRCI cortical dysfunction have not been validated in the ICU pop-
as inadequate cellular corticosteroid activity for the severity of ulation.26,27 Consensus guidelines suggest a random cortisol
the patient’s illness.17 The pathophysiology of this form of of less than 275 nmol/L (10 µg/dL) or a change in cortisol
adrenal “insufficiency” is not well defined, but critical illness levels of less than 250 nmol/L (9 µg/dL) following 250 µg of
may alter the HPA and down-regulate its feedback responses cosyntropin are the best tests available to diagnose adrenal
resulting in impaired cortisol production and tissue resistance insufficiency in the critically ill patient.17
to glucocorticoids.18 Although the stress of critical illness is Several factors make diagnosis in critical care more chal-
associated with high circulating cortisol levels, there often is a lenging. First, cortisol immunoassays measure protein-bound
blunted response to ACTH. For example, supplemental hydro- cortisol. Both cortisol-binding globulin and albumin are neg-
cortisone may improve hemodynamic stability in septic shock, ative acute-phase reactants and their levels decrease in the
suggesting a relative shortage of steroids or a down-regulation setting of stress.28 This makes the relationship between total
of available receptors.19-21 and free cortisol unpredictable and may give the appearance
of adrenal insufficiency, even though free cortisol levels may
be appropriate. Second, saturation of the corticotropin recep-
Clinical Presentation tor, with a corresponding limit of cortisol release, may con-
Symptoms of acute adrenal insufficiency are often nonspecific found dynamic measurements of glucocorticoid function.
and include vomiting, lethargy, abdominal pain, fever, and Consequently a low, unstimulated free plasma cortisol level
mental status changes. Severe hypotension and shock then has been proposed as a better marker of adrenal insufficiency.29
may develop. This presentation can be confused with, or exac- Although this makes free cortisol an attractive target for
erbate preexisting, septic shock. In some patients, the degree testing, the assay is not routinely available. Furthermore, total
of cardiovascular collapse is associated with mineralocorticoid cortisol has been shown to correlate with free cortisol in
rather than glucocorticoid insufficiency—patients who receive treatment-resistant hypotension in critical illness.30
glucocorticoids may still develop an adrenal crisis if their min- Hyponatremia and hyperkalemia develop in acute adrenal
eralocorticoid requirements are not met.22 Outcome can be insufficiency, so assessment of electrolytes can be a useful
Section II—Clinical and Laboratory Assessment 203

adjunct in the patient who develops hypotension or other a negative feedback system. The thyroid gland primarily pro-
symptoms of acute adrenal insufficiency. duces T4, a biologically inactive hormone that must be con-
verted to the active hormone T3 in peripheral tissues. Because
thyroid hormone is critical for cellular metabolism, both
Adrenal Insufficiency Management overproduction and undersecretion can affect all organ
An acute adrenal crisis can rapidly progress to death; there- systems.
fore, suspected cases should be immediately treated with
100 mg of intravenous (IV) hydrocortisone followed by 100
to 200 mg per day. Because there is relatively little risk associ- Hyperthyroidism and Thyroid Storm
ated with short-term steroid use, treatment of acute adrenal Excess T4 levels characterize hyperthyroidism. An overproduc-
insufficiency should not be delayed pending diagnostic evalu- tive thyroid nodule or gland is the most common etiology, and
ation. Hydrocortisone provides both glucocorticoid and min- TSH production is suppressed. Less commonly, excess TSH
eralocorticoid supplementation without sustained suppression causes elevated thyroid hormone levels. Thyrotoxic crisis, or
of the HPA and is the preferred agent to provide “stress dose” thyroid storm, is a rare but potentially life-threatening state
steroids. Fluid resuscitation with isotonic fluid and vasopres- that most frequently occurs in patients with untreated or par-
sors also are often required. Electrolyte abnormalities and tially treated hyperthyroidism. Trauma or an acute illness can
hypoglycemia should be corrected. precipitate this transition from stable hyperthyroidism to a
Treatment of CIRCI in severe sepsis may be beneficial decompensated crisis. Thyroid function tests are not helpful
but remains controversial. In patients with severe septic to distinguish the two states. Instead the diagnosis of thyroid
shock, Annane et al. demonstrated corticosteroid replacement storm is made on clinical grounds. Signs and symptoms of
reduced the duration of vasopressor support and improved thyroid storm are exaggerated features of hyperthyroidism
survival in patients with RAI.31 However, in another ran­ and include fever (>38.5° C), tachycardia, hypertension, agita-
domized clinical trial, corticosteroid therapy of septic shock tion, delirium, lethargy, seizures, coma, nausea, vomiting,
(CORTICUS), steroid use was associated with improved diarrhea, and, in severe cases, jaundice.
hemodynamic stability but had no effect on overall survival.
In addition, the cosyntropin stimulation test could not help
differentiate who would benefit from steroid supplementa- Treating Thyrotoxicosis
tion.32 Several factors including differences in patient acuity, The therapeutic goals of treating thyroid storm are to: (1)
timing of enrollment, and the addition of fludrocortisone may decrease hormone production and secretion, (2) block the
explain the different outcome results in these two trials.31,32 conversion of T4 to T3, and (3) antagonize the catecholamin-
The 2008 Surviving Sepsis Campaign guidelines do not rec- ergic effects of thyroid hormone. Thionamides such as
ommend using a cosyntropin stimulation test to diagnose propylthiouracil and methimazole can block new thyroid
relative adrenal insufficiency but suggest hydrocortisone hormone synthesis within hours of administration. Propyl-
(<300 mg/day) may be used in any patient with septic shock thiouracil is preferred because it also inhibits the peripheral
that is refractory to fluid and vasopressor support.33 Similarly, conversion of T4 to T3. These agents, however, do not prevent
the consensus statements from an international task force by the release of stored hormone. High-dose iodine (e.g., Lugol’s
the American College of Critical Care Medicine recommend solution or saturated solution of potassium iodide) can acutely
treatment of critically ill patients based on clinical criteria block the release of T4 and T3, but should be given only after
rather than the results of adrenal function testing.17 thyroid synthesis has been blocked. If synthetic function is not
Because hydrocortisone has mineralocorticoid activity, the adequately suppressed, the iodine bolus will enhance thyroid
benefit of adding fludrocortisone as described by Annane hormone synthesis and thus exacerbate the thyrotoxicosis.
et al. remains unclear.31 Nonetheless, oral fludrocortisone Glucocorticoids can reduce the peripheral conversion of T4 to
(50 µg daily) is considered an optional therapy in the Surviv- T3 and, given a high risk of concomitant adrenal insufficiency,
ing Sepsis Campaign guidelines. Supplemental steroids should are considered standard of care in severe thyrotoxicosis. Beta-
be tapered when vasopressors are no longer required to mini- adrenergic blockade, in particular propranolol that has anti-
mize side effects including the risk of secondary adrenal adrenergic effects and inhibits the peripheral conversion of T4
suppression. to T3, remains a mainstay of therapy. Alternatively, esmolol
can be used when rapid titration is needed to reduce potential
side effects. Clinical improvement usually is rapid following
Thyrotroph Dysfunction the initiation of β-blocker therapy. In addition to the expected
cardiac effects, there is marked improvement in agitation, con-
Physiology fusion, psychosis, diaphoresis, diarrhea, and fever.34
Thyroid hormone secretion is tightly regulated by the
hypothalamic-pituitary-thyroid axis. Thyrotropin-releasing Myxedema Coma
hormone (TRH) is released from the hypothalamus and
stimulates the synthesis and secretion of thyroid-stimulating Etiology and Presentation
hormone (TSH). TSH, in turn, controls the synthesis and Myxedema coma is the result of severe, decompensated hypo-
secretion of the thyroid hormones, thyroxine (T4) and triio- thyroidism that can occur insidiously as the result of severe
dothyronine (T3). More than 99.5% of T4 and T3 are protein long-standing hypothyroidism or it can be precipitated by
bound in the serum and are metabolically inactive. The small an acute event such as infection, myocardial infarction,
percentage of free T4 and T3 influence metabolic function head injury, cold exposure, or certain medications (e.g.,
and modulate the release of both TRH and TSH using opiates, lithium, amiodarone). Patients with myxedema coma
204 Section II—Clinical and Laboratory Assessment

demonstrate the classic features of severe hypothyroidism 50 µg. The daily maintenance dose should then be adjusted
including dry skin, thin hair, periorbital swelling, nonpitting based on laboratory findings and clinical improvement. Ini-
edema of the hands and feet, a hoarse voice, macroglossia, and tially, TSH and free T4 levels should be closely followed to
delayed deep tendon reflexes. Progression to myxedema coma, prevent overtreatment. After a stable daily dose is adopted,
however, is characterized by a depressed mental status, hypo- patients should have their thyroid function reevaluated in 4
tension, and hypothermia. Focal and generalized seizures also to 6 weeks.
have been reported.35 Depressed myocardial contractility and
bradycardia result in low cardiac output, profound hypoten-
sion, and diminished cerebral perfusion.36 Mortality can be Nonthyroidal Illness Syndrome
high (30%-60%) even with early diagnosis and appropriate Abnormal circulating levels of thyroid hormone are associated
therapy37,38 and is associated with the degree of hypothermia. with many acute illnesses and do not necessarily indicate
A core temperature less than 32° C is associated with a worse pathology at the level of the hypothalamus or pituitary (Figure
prognosis. 21.1). Under normal conditions, T4 is converted by tissue
5′-monodeiodinase to T3, the metabolically active thyroid
hormone. During periods of carbohydrate deprivation or
Laboratory Findings in Myxedema Coma illness, an alternative pathway of deiodination predominates
Hyponatremia and hypoglycemia are common. Low serum and rT3, a biologically inactive hormone, is created.46,47 The
sodium results from excessive vasopressin secretion and clearance of rT3 also may be impaired because of the inhibited
impaired free water excretion.39,40 Significant hyponatremia 5′-monodeiodinase activity.48 High circulating levels of proin-
(≤125 mEq/L) is common and may contribute to mental flammatory cytokines and increased serum cortisol con­
status changes. Decreased gluconeogenesis and hypoglycemia centrations may promote this shift in deiodination.49-52
may occur with hypothyroidism alone or with adrenal insuf- Medications frequently used in the ICU (e.g., amiodarone,
ficiency.41 Infection is frequently a precipitating cause of glucocorticoids, propranolol) also can contribute to depressed
myxedema coma. However, an elevated white blood cell T3 concentrations by inhibiting tissue 5′-monodeiodinase
(WBC) count often is absent, although a left shift may be activity. With mild illness, only T3 levels decline but with
observed. increasing severity, a decrease in both T3 and T4 is observed
without a concomitant elevation in TSH. These changes once
were considered an adaptive response and the term euthyroid
Management of Myxedema Coma sick syndrome was adopted. Increasing evidence suggests that
When clinically suspected, therapy should be instituted the thyroid abnormalities seen in critical illness reflect a state
without waiting for laboratory confirmation. However, of central hypothyroidism.53 The designation of euthyroid sick
thyroid function tests (serum TSH and free T4) should be syndrome has been abandoned in preference of the more
drawn before starting therapy. A cortisol level also should be metabolically neutral term nonthyroidal illness syndrome
obtained to investigate the possibility of concurrent adrenal (NTIS), which also is referred to as the low T3 syndrome.4
insufficiency. Appropriate hormonal supplementation nor-
malizes the basal metabolic rate and reverses all symptoms TRH
and signs of hypothyroidism.42 Hydrocortisone should be Glucocorticoids
given with thyroid replacement for several days and then Dopamine
tapered based on clinical improvement and assessment of Dobutamine
adrenal function because thyroid replacement may unmask
coexisting adrenal insufficiency and precipitate an adrenal TSH
crisis.43
The deiodinase conversion of T4 to T3 is impaired in severe
hypothyroidism and so IV T3 may be preferable given its
Cytokines, glucocorticoids,
greater biologic availability. T3 rapidly achieves effective T4 amiodarone, propranolol inhibit
tissue levels44 and crosses the blood-brain barrier more readily
than T4 and can hasten the improvement of neurologic symp-
toms.45 The rapid onset of T3, however, also may increase the
risk of treatment-associated complications such as myocar-
dial infarction or arrhythmias. Treatment with IV T4 is associ-
ated with a slower onset of action because the patient’s
inherent tissue deiodinase activity is required to convert T4 to T3 rT3
T3 and clinical improvement may take 1 to 3 days. However,
the slower onset of action may decrease the likelihood of
cardiac complications. A third treatment option is to supple- Cytokines, glucocorticoids,
ment both T4 and T3. This provides T3 at a subtherapeutic amiodarone, propranolol
dose for immediate action and a loading dose of T4. T3 is inhibit
given as an initial IV dose of 10 µg, followed by 10 µg every 8 T2
to 12 hours until there is clinical improvement and the patient Fig. 21.1  Changes in hypothalamic-pituitary-thyroid axis in critical
is able to take maintenance oral doses of T4. In addition, a illness. rT3, Reverse triiodothyronine; TRH, thyrotropin-releasing
loading dose of 200 to 300 µg of T4 is given intravenously, hormone; TSH, thyroid-stimulating hormone; T2, 3,5-Diiodo-L-thyronine;
followed by 100 µg 24 hours later, and then a daily dose of T3, triiodothyronine; T4, thyroxine.
Section II—Clinical and Laboratory Assessment 205

Low T3 and T4 levels and in particular a progressive decline concentrations are generally elevated in hypothyroidism.
in T4 levels54 in critically ill patients may be associated with Although not routinely measured, T3 levels can be useful
higher mortality. However, replacement of low T4 levels in when the diagnosis of NTIS is entertained. Although T3 levels
NTIS with levothyroxine may not improve outcome.55 Van are universally low in NTIS, T4 and TSH values are variable.
den Berghe et al. suggest the thyroid function abnormalities The diagnosis of thyroid dysfunction therefore should be
seen in prolonged critical illness reflect a maladaptive depres- made in the context of the T4-TSH relationship. TSH should
sion of the neuroendocrine drive that should be treated at the be measured using a third-generation assay with a functional
hypothalamic level. In 20 critically ill adult patients, they sensitivity limit of 0.01 to 0.02 mIU/L to differentiate NTIS
observed that coinfusion of TRH and growth hormone– from underlying thyroid dysfunction.60 Using this more sen-
releasing peptide (GHRP)-2 was associated with the return of sitive assay, most hospitalized patients with hyperthyroidism
pulsatile TSH secretion and the normalization of thyroid will have a TSH value less than 0.01 mIU/L and those with
hormone concentrations.56 Further clinical studies are needed NTIS will have values between 0.01 and 0.1 mIU/L.61,62 Both
to establish the efficacy and benefit of this treatment strategy total T4 and free T4 have been used to evaluate NTIS. Assays
in NTIS. Patients with elevated TSH levels, however, have a for free T4 theoretically provide information about the con-
“central” hypothyroidism and should be treated with thyroid centration of biologically active hormone; however, the tech-
replacement therapy. Medications such as glucocorticoids, nical difficulties of measuring free hormone levels in critical
dopamine, and dobutamine suppress TSH production and illness may limit its usefulness.62 T4 levels in NTIS range from
may contribute to a state of central hypothyroidism.57 normal to markedly suppressed.
Untreated central hypothyroidism may produce symptoms
(e.g., decreased energy, neuropsychiatric symptoms, weight
gain) that mimic those seen during long-term recovery after Somatotroph and
TBI, and a high index of suspicion is required to appropriately
diagnose hypothyroidism in these patients. In addition, the
Gonadotroph Dysfunction
low T3 syndrome may be associated with the need for pro- During the acute phase of critical illness the pattern of growth
longed mechanical ventilation.58 Recovery from the underly- hormone (GH) secretion changes. In healthy people, GH is
ing illness is associated with a normalization of thyroid released in a pulsatile fashion with peaks alternating with
function with TSH values recovering before an improvement nearly undetectable troughs. In the acutely ill ICU patient, the
in T4 levels is observed.59 baseline level of GH increases, as does the frequency of peaks
whereas serum levels of insulin-like growth factor (IGF)-1,
growth hormone–binding protein (GHBP), and the expres-
Monitoring the Hypothalamic- sion of GH receptor in peripheral tissues are decreased. It is
postulated that this is an adaptive and desirable response to
Pituitary-Thyroid Axis in the ICU acute illness. The direct effects of the increased levels of GH
Routine thyroid function surveillance is not recommended in include lipolysis, antagonization of insulin, and stimulation
the ICU because the majority of critically ill patients only of the immune system and the decreased levels of IGF-1 and
have transient or nonspecific abnormalities. Thyroid function GH receptor attenuate the somatotropic and anabolic effects
testing should be reserved for at-risk patients or those whose of GH.63
clinical picture suggests thyroid dysfunction. There is signifi- Dysfunction of the somatotrophs and gonadotrophs do not
cant overlap among the laboratory values seen in various produce acute life-threatening illness. However, there is accu-
states of thyroid dysfunction (Table 21.4) and so laboratory mulating evidence that somatotroph dysfunction may play a
abnormalities must be interpreted within the clinical context. role in the “wasting syndrome” of prolonged critical illness in
TSH and free T4 levels can be measured to monitor thyroid which protein wasting occurs despite adequate caloric intake.
function in ICU patients (see Figure 21.1). In general, an In prolonged ICU stays, GH pulsatility remains abnormal and
elevated T4 level suggests hyperthyroidism, whereas a secretion decreases relative to the acute stage. IGF-1 levels also
depressed T4 concentration suggests hypothyroidism. TSH remain low.64 Chronic GH deficiency may lead to a lack of
levels typically are suppressed in hyperthyroidism but can vigor, decreased exercise tolerance, a decrease in lean body
occasionally be elevated in central hyperthyroidism. TSH mass with redistribution of fat, and decreased social function-
ing. This GH deficiency then may limit rehabilitation poten-
tial in patients who survive their ICU stay. Although not
Table 21.4  Diagnosis of Thyroid Axis acutely life threatening, chronic dysfunction of luteinizing or
Dysfunction follicle-stimulating hormone secretion may result in sexual
Disorder T3 T4 TSH
dysfunction, mood disorders, osteoporosis, and alterations in
menstrual function in women.
Thyrotoxicosis High High Low
Hyperthyroidism High High Low (<0.01 mIU/L)
Monitoring Somatotroph and Gonatroph
Myxedema Low Low High (rarely
normal or low) Function in the ICU
NTIS Low Normal to low Normal to low Somatotrophs or gonadotrophs are not routinely tested in the
(0.01-0.1 mIU/L) ICU, in part because some of the observed changes may be
adaptive and there does not appear to be a benefit to correct
NTIS, Nonthyroidal illness syndrome; TSH, thyroid-stimulating hormone;
T3, triiodothyronine; T4, thyroxine.
dysfunction in the acute setting. Guidelines suggest that GH
testing should be delayed until other pituitary defects have
206 Section II—Clinical and Laboratory Assessment

been corrected for a period of at least 3 to 6 months.65 Ongoing insipidus (DI) is a consequence of diminished or absent ADH
research into the role of GH in prolonged critical illness may secretion and results in the excretion of large amounts of free
support a role for testing in the chronic phases of ICU treat- water and should be differentiated from nephrogenic DI,
ment, although there is not enough evidence to support this which results from resistance to the actions of ADH at the
recommendation. kidney. Central DI is frequently encountered in the NCCU.
Approximately 30% of cases are idiopathic, 25% are associated
with tumors of the brain or pituitary, 20% result from intra-
Treating Somatotroph and Gonadotroph cranial surgery, and 16% are post-traumatic. SIADH, on the
Dysfunction in the ICU other hand, occurs when the hypothalamus releases too much
Currently there is no justification for GH replacement or ADH. This results in the retention of free water, hyponatremia,
supplementation in the acute ICU setting. Although several volume expansion, and inappropriately concentrated urine.
small studies have demonstrated improved nitrogen balance
and increased IGF-1 levels with GH replacement in prolonged
illness, two large, randomized trials demonstrated a significant Monitoring Vasopressin Dysfunction  
increase in mortality, length of stay, and length of ventilator in the ICU
dependence with GH supplementation.66 Intervening at the Vasopressin levels are difficult to measure because the mole-
hypothalamic level, however, may offer some benefit in cule is unstable, extensively bound to platelets, and rapidly
patients with prolonged critical illness. Short term (2-5 days) cleared. These difficulties have prevented its routine clinical
infusion of GHRP-2 and TRH in chronically ill ICU patients measurement. Copeptin, on the other hand, is a stable mole-
has been shown to reestablish pulsatility, restore GH and TSH cule co-synthesized with vasopressin and mirrors vasopressin
secretion, improve peripheral responsiveness, and initiate a release. Copeptin has been shown to correlate with vasopres-
shift toward anabolic pathways. Although encouraging, further sin levels across a range of serum osmolarities in healthy vol-
studies are needed to look at the effects of this treatment on unteers and can be useful to help discriminate DI from other
morbidity and mortality.67 similar clinical presentations and SIADH from sodium-
wasting states.68-70 Stimulated copeptin levels measured 45
minutes after an insulin infusion appear also to be helpful.71
Posterior Pituitary Dysfunction However, further validation studies are necessary and as such,
vasopressin dysfunction remains a clinical diagnosis.
Physiology
The posterior pituitary, or neurohypophysis, develops from an
evagination of the embryonic ventricular system. Unlike the Diabetes Insipidus
anterior pituitary, the posterior pituitary does not contain A diagnosis of DI can be made when urine output is high
glandular cells but is composed of terminal axons that origi- (>250 mL/hr or >3 mL/kg/hr in a pediatric population) and
nate in the supraoptic and paraventricular nuclei of the hypo- the urine is dilute (urine osmolality 50-150 mOsm/L or spe-
thalamus. The posterior pituitary secretes two hormones: cific gravity 1.001-1.005). A diagnosis of DI also requires
vasopressin and oxytocin. Vasopressin or antidiuretic hormone serum sodium to be normal or high. DI cannot occur in the
(ADH) is derived from a molecule synthesized in the supra- face of primary adrenal dysfunction, because some degree of
optic and paraventricular nuclei of the hypothalamus. The mineralocorticoid activity is obligatory for the kidney to make
precursor molecule is packaged, cleaved to its active form, and free water. In rare cases in which the diagnosis remains unclear,
transported down axons to the posterior pituitary, where it is a water deprivation test may be used to provoke ADH release.
stored and released into the systemic circulation via a capillary All IVs are stopped and the patient is allowed nothing per
plexus. ADH secretion is tightly regulated by changes in mouth. Urine osmolality is checked every hour. A decrease
plasma osmotic pressure, and plasma volume and acts on the in urine output and a urine osmolality between 600 and
kidney to reduce excretion of free water and on the vasculature 850 mOsm/L indicate a normal response and excludes DI.
to improve vascular tone. At physiologic levels the antidiuretic The test is terminated after 6 to 8 hours or when urine osmo-
effect is prominent. ADH has also been implicated in regulat- lality plateaus (<30 mOsm change in 3 hours) indicating an
ing social behaviors such as aggression and pair bonding. Oxy- abnormal response. If the patient does not respond normally
tocin is a hormone structurally similar to ADH but synthesized to the water deprivation test, then exogenous ADH (5 U
in distinct neurons within the supraoptic and paraventricular aqueous vasopressin [Pitressin] subcutaneously) is adminis-
nuclei. It is responsible for uterine contraction and the lacta- tered. Exogenous ADH should increase urine osmolality to
tion let-down reflex. more than 300 mOsm/L. A less than 5% increase in urine
osmolality following vasopressin indicates normal ADH levels,
a 6% to 67% increase indicates a partial ADH deficiency, and
Disorders of Vasopressin a greater than 67% increase indicates severe ADH deficiency.
The patient must be monitored closely during the water
in the NCCU deprivation test because fatal dehydration can result.
Disorders of vasopressin are encountered frequently in the
NCCU. By contrast, oxytocin has little known physiologic
relevance in the care of NCCU patients. Disorders of vasopres- Syndrome of Inappropriate
sin may result from either too little (diabetes insipidus) or too Antidiuretic Hormone
much (syndrome of inappropriate antidiuretic hormone SIADH is a clinical diagnosis that does not have a single con-
[SIADH]) secretion of the hormone. Central diabetes firmatory test. Bartter and Schwarz proposed the classic
Section II—Clinical and Laboratory Assessment 207

diagnostic criteria in 1967 that includes: (1) hyponatremia


with corresponding hypo-osmolality, (2) continued renal
Endocrine Consequences  
excretion of sodium, (3) urine that is less than maximally of Traumatic Brain Injury
dilute, (4) absence of clinical evidence of volume depletion, Hypopituitarism is a recognized complication of TBI. Al-
(5) absence of other causes of hyponatremia, and (6) correc- though first reported in 1918, hypopituitarism following
tion of hyponatremia by fluid restriction.72 More specifically, TBI was long thought to be a relatively uncommon compli-
serum sodium is generally less than 134 mEq/L, serum osmo- cation. More recent literature, however, suggests that the
lality is less than 280 mOsm, and urinary sodium is greater prevalence of post-traumatic hypopituitarism (specifically
than 18 mEq/L but can often be as high as 50 to 150 mEq/L. anterior pituitary hormone deficiencies) following TBI
If asymptomatic and mild, acute SIADH may be treated by ranges between 28% and 68%. These deficits may manifest
fluid restriction (<1 L/day). More severe cases may require either in the acute period following injury or in a delayed
correction with hypertonic saline and furosemide. The rate of fashion. Some cases of pituitary dysfunction that present in
correction should be monitored closely because central the acute phase spontaneously resolve within 6 months post
pontine myelinolysis may occur with too rapid a correction of injury. Similarly, the delayed onset of anterior pituitary dys-
serum sodium. If the SIADH is chronic in nature, then a more function is generally evident within 6 months following
generous fluid restriction of 1200 to 1800 mL/day with furo- head injury.76,77
semide may be appropriate. Demeclocyline also may be used Endocrine dysfunction in TBI may be associated with
to partially antagonize the effects of ADH on the renal tubules. anterior lobe necrosis, posterior lobe hemorrhage, or stalk
laceration or vascular injury to the hypothalamus. In
addition TBI is hypercatabolic, and nutritional status may
Cerebral Salt Wasting influence endocrine function.78 GH deficiency is the most
It is important to differentiate SIADH from cerebral salt common hormone deficiency seen following TBI. GH-
wasting (CSW). Like SIADH, CSW occurs in the setting secreting somatotroph cells are particularly vulnerable to vas-
of intracranial disease and produces hyponatremia. Unlike cular insult because they are located in the lateral aspect of
SIADH, CSW results in a decrease in extracellular fluid the pituitary gland and derive their blood supply from the
volume. Because laboratory values may be similar or unreveal- hypothalamo-pituitary portal vessels. In contrast ACTH- and
ing, volume status is the principal method to differentiate TSH-producing cells are more protected because of their
SIADH from CSW. Volume status may be measured by central ventral and medial location and their dual blood supply from
venous pressure, a pulmonary capillary wedge pressure, or a the portal vessels and branches of the anterior pituitary
nuclear medicine study of plasma volume. Low volume sup- artery.79,80 However, abnormalities in the ACTH axis may still
ports the diagnosis of CSW. A high potassium in the setting complicate between 4% and 50% of TBI patients. The inci-
of hyponatremia is incompatible with the diagnosis of SIADH dence depends in part on the method used to assess the
and supports a diagnosis of CSW.73 Fluid restriction exacer- ACTH axis and when the tests are performed because hor-
bates CSW and should be avoided. The primary goals of treat- monal function is a dynamic process after TBI.81 Suppression
ment in CSW are sodium and volume replacement. Salt may of gonadotropins is common after TBI but most levels recover
be replaced orally with salt tabs or with IV hypertonic saline. relatively quickly and nearly all within a year.82 However,
Rapid correction should be avoided to reduce the risk of patients who are hypogonadal when they start rehabilitation
central pontine myelinolysis. have lower Functional Independence Measure (FIM) scores at
discharge.83 Posterior pituitary dysfunction in the form of
either DI or SIADH also is seen in TBI but seems unrelated to
Endocrine Consequences of Specific the incidence of anterior hypopituitarism. Although DI or
Diseases Managed in the NCCU SIADH may be present transiently in the acute phase of
injury, SIADH almost always resolves, whereas a small per-
Endocrine Consequences   centage of DI cases may be persistent.82,84,85
of Pituitary Surgery How best to screen patients with moderate or severe TBI
Surgery on or near the pituitary gland can impact endocrine for endocrine dysfunction is still being elucidated. In the acute
function. For example, Fatemi et al. observed a 5% incidence period following TBI, ACTH deficiency can lead to acute
of worse anterior pituitary function, whereas 24% of patients adrenal insufficiency and life-threatening complications. Con-
had improved anterior pituitary function following trans­ sequently, some centers screen all patients with moderate or
sphenoidal resection of pituitary adenomas.74 Pituitary surgery severe TBI with morning cortisol levels for the first 5 post-
also can impact posterior pituitary function and cause shifts injury days.86 If a deficiency of ACTH is discovered, screening
in water and electrolyte balance. Isolated DI is the most for pituitary hormone deficiencies seems reasonable. There is
common deficit seen after transsphenoidal surgery and occurs no evidence, however, to support the routine screening for
in up to 39% of patients who develop hypernatremia postop- thyroid, GH, or gonadal hormone deficiencies in the acute
eratively. Diuresis is typically maximal in the first and second period. Rather, anterior pituitary dysfunction should be
postoperative days. Isolated hyponatremia also can occur, and assessed at either 3 or 6 months after moderate or severe
up to 21% of patients develop SIADH. The development of injury.76 Guidelines released in 2005 suggest prospective and
hyponatremia, however, often is delayed, reaching a nadir on retrospective screening of moderate and severe TBI patients
postoperative day 9 or 10. In the majority of cases, SIADH is with the subsequent replacement with vasopressin, glucocor-
transient and responds to simple fluid restriction. Approxi- ticoid and thyroid hormones. Gonadal and GH replacement
mately 16% of patients have both DI and hyponatremia should be postponed until the existence of these deficits is
during their postoperative recovery.75 confirmed following the correction of other hormone
208 Section II—Clinical and Laboratory Assessment

deficiencies. If gonadal and GH deficiencies are proven, then References


replacement may help improve rehabilitation by increasing 1. Cooper MS, Stewart PM. Adrenal insufficiency in critical illness. J Intensive
muscle mass, energy levels, and emotional stability and reduc- Care Med 2007;22:348–62.
ing the sense of social isolation.80 The role for GH replacement 2. Cohen J, Venkatesh B. Assessment of tissue cortisol activity. Crit Care Resusc
after TBI, however, is still to be elucidated.81 2009;11:287–9.
3. Leonard MO, Godson C, Brady HR, et al. Potentiation of glucocorticoid
activity in hypoxia through induction of the glucocorticoid receptor. J
Endocrine Consequences of   Immunol 2005;174:2250–7.
4. Mebis L, Van den Berghe G. Thyroid axis function and dysfunction in
Subarachnoid Hemorrhage critical illness. Best Pract Res Clin Endocrinol Metab 2011 Oct;25(5):745–57.
5. Clark PM, Gordon K. Challenges for the endocrine laboratory in critical
In the acute phase after SAH hypogonadotropic hypogonad-
illness. Best Pract Res Clin Endocrinol Metab 2011;25(5):847–59.
ism is common, but rates of ACTH and TSH deficiency are 6. Thomas Z, Bandali F, McCowen K, et al. Drug-induced endocrine disorders
rare (7.1% and 1.8%, respectively).87,88 Long-term hypopitu- in the intensive care unit. Crit Care Med 2010;38(6 Suppl):S219–30.
itarism is a known complication following SAH, and anterior Review.
pituitary dysfunction is observed in between 37.5% and 55% 7. Vuong C, Van Uum SH, O’Dell LE, et al. The effects of opioids and opioid
analogs on animal and human endocrine systems. Endocr Rev
of patients following SAH, although in part this depends on 2010;31:98–132.
the evaluation methods used.77 Persistent posterior pituitary 8. Chin R. Adrenal crisis. Crit Care Clin 1991;7:23–42.
dysfunction, however, is less common. However, few studies 9. Cuthbertson BH, Sprung CL, Annane D, et al. The effects of etomidate on
have looked specifically at the effect of treating hypopitu­ adrenal responsiveness and mortality in patients with septic shock. Intensive
Care Med 2009;35:1868–76.
itarism after SAH. Preliminary data suggest that chronic
10. Sonino N. The use of ketoconazole as an inhibitor of steroid production.
neuroendocrine dysfunction contributes to depression, and N Engl J Med 1987;317:812–8.
disturbances in quality of life and sleep in SAH survivors.89 11. Ashby H, DiMattina M, Linehan WM, et al. The inhibition of human
Sodium imbalance in SAH associated with the SIADH adrenal steroidogenic enzyme activities by suramin. J Clin Endocrinol Metab
secretion or CSW syndrome are frequent after SAH. However, 1989;68:505–8.
12. Kyriazopoulou V, Parparousi O, Vagenakis AG. Rifampicin-induced adrenal
there are conflicting reports regarding the incidence of HPA crisis in addisonian patients receiving corticosteroid replacement therapy.
dysfunction in the acute setting following SAH.90 Whereas J Clin Endocrinol Metab 1984;59:1204–6.
some authors have reported a decrease in cortisol and 13. Wagner RL, White PF, Kan PB, et al. Inhibition of adrenal steroidogenesis by
ACTH23,91 others have found no change92 or an increase93 in the anesthetic etomidate. N Engl J Med 1984;310:1415–21.
14. Elias AN, Gwinup G. Effects of some clinically encountered drugs on steroid
cortisol levels. In the subgroup of SAH patients that are refrac-
synthesis and degradation. Metabolism 1980;29:582–95.
tory to vasopressor therapy, however, the incidence of CIRCI 15. Arlt W, Allolio B. Adrenal insufficiency. Lancet 2003;361:1881–93.
after SAH is reported to be as high as 68%.94 In addition there 16. Schmiegelow M, Feldt-Rasmussen U, Rasmussen AK, et al. Assessment of the
is some evidence to suggest levels of cortisol or ACTH may be hypothalamo-pituitary-adrenal axis in patients treated with radiotherapy
associated with global cerebral edema or cerebral ischemia in and chemotherapy for childhood brain tumor. J Clin Endocrinol Metab
2003;88:3149–54.
the first week after SAH.95 This may be particularly important 17. Marik PE, Pastores SM, Annane D, et al. Recommendations for the diagnosis
in management of vasospasm and delayed cerebral ischemia. and management of corticosteroid insufficiency in critically ill adult patients:
In some patients it may be difficult or impossible to maintain consensus statements from an international task force by the American
adequate perfusion pressure to prevent ischemia because of College of Critical Care Medicine. Crit Care Med 2008;36:1937–49.
18. Meduri GU, Yates CR. Systemic inflammation-associated glucocorticoid
adrenal insufficiency.96 As such, testing for adrenal insuffi-
resistance and outcome of ARDS. Ann N Y Acad Sci 2004;1024:24–53.
ciency in patients with SAH with hypotension refractory to 19. Annane D, Sebille V, Charpentier C, et al. Effect of treatment with low doses
vasopressors should be considered. There may be modest of hydrocortisone and fludrocortisone on mortality in patients with septic
outcome benefits when using fludrocortisones in some shock. JAMA 2002;288:862–71.
patients.97 There is limited study on the role of corticosteroids 20. Ligtenberg JJ, Zijlstra JG. The relative adrenal insufficiency syndrome
revisited: which patients will benefit from low-dose steroids? Curr Opin Crit
in SAH; the few published studies suggest that routine corti- Care 2004;10:456–60.
costeroid use is not indicated.98,99 21. Sprung CL, Annane D, Keh D, et al. Hydrocortisone therapy for patients
with septic shock. N Engl J Med 2008;358:111–24.
22. Cronin CC, Callaghan N, Kearney PJ, et al. Addison disease in patients
Conclusion treated with glucocorticoid therapy. Arch Intern Med 1997;157:456–8.
23. Dimopoulou I, Tsagarakis S, Kouyialis AT, et al. Hypothalamic-pituitary-
Endocrine abnormalities often may be overlooked and under- adrenal axis dysfunction in critically ill patients with traumatic brain injury:
diagnosed in the NCCU. Symptoms, both acute and chronic, incidence, pathophysiology, and relationship to vasopressor dependence and
may be masked by or mimic sequelae of neurologic injury. It peripheral interleukin-6 levels. Crit Care Med 2004;32:404–8.
is therefore important that a high index of suspicion be main- 24. Dimopoulou I, Tsagarakis S, Douka E, et al. The low-dose corticotropin
stimulation test in acute traumatic and non-traumatic brain injury:
tained in both the acute setting and in follow-up care of the incidence of hypo-responsiveness and relationship to outcome. Intensive
NCCU population. TBI patients with moderate to severe Care Med 2004;30:1216–9.
injury should undergo testing of the pituitary axis both in the 25. Glowniak JV, Loriaux DL. A double-blind study of perioperative steroid
acute setting and at follow-up. Vasopressin, thyroid hormone, requirements in secondary adrenal insufficiency. Surgery 1997;121:123–9.
26. Cooper MS, Stewart PM. Corticosteroid insufficiency in acutely ill patients.
and cortisol deficiencies must be addressed immediately.
N Engl J Med 2003;348:727–34.
Although there is no evidence to support replacing GH and 27. Nieboer P, van der Werf TS, Beentjes JA, et al. Catecholamine dependency in
sex-steroid hormones in the acute setting, deficiencies in these a polytrauma patient: relative adrenal insufficiency? Intensive Care Med
hormones may impair rehabilitation potential and should be 2000;26:125–7.
considered after other pituitary function has been optimized 28. Beishuizen A, Thijs LG, Vermes I. Patterns of corticosteroid-binding globulin
and the free cortisol index during septic shock and multitrauma. Intensive
at follow-up. However, further research is needed to establish Care Med 2001;27:1584–91.
the benefits and risks of endocrine manipulation in the criti- 29. Klaff LS, Wisse BE. Current controversy related to glucocorticoid and insulin
cally ill. therapy in the intensive care unit. Endocr Pract 2007;13:542–9.
Section II—Clinical and Laboratory Assessment 209

30. Molenaar N, Johan Groeneveld AB, Dijstelbloem HM, et al. Assessing 43. Bigos ST, Ridgway EC, Kourides IA, et al. Spectrum of pituitary alterations
adrenal insufficiency of corticosteroid secretion using free versus total with mild and severe thyroid impairment. J Clin Endocrinol Metab 1978;46:
cortisol levels in critical illness. Intensive Care Med 2011;37:1986–93. 317–25.
31. Annane D, Sebille V, Charpentier C, et al. Effect of treatment with low doses 44. Pereira VG, Haron ES, Lima-Neto N, et al. Management of myxedema coma:
of hydrocortisone and fludrocortisone on mortality in patients with septic report on three successfully treated cases with nasogastric or intravenous
shock. JAMA 2002;288:862–71. administration of triiodothyronine. J Endocrinol Invest 1982;5:331–4.
32. Sprung CL, Annane D, Keh D, et al. Hydrocortisone therapy for patients 45. Chernow B, Burman KD, Johnson DL, et al. T3 may be a better agent than
with septic shock. N Engl J Med 2008;358:111–24. T4 in the critically ill hypothyroid patient: evaluation of transport across the
33. Dellinger RP, Levy MM, Carlet JM, et al. Surviving Sepsis Campaign: blood-brain barrier in a primate model. Crit Care Med 1983;11:99–104.
international guidelines for management of severe sepsis and septic shock: 46. Harris AR, Fang SL, Vagenakis AG, et al. Effect of starvation, nutriment
2008. Crit Care Med 2008;36:296–327. replacement, and hypothyroidism on in vitro hepatic T4 to T3 conversion in
34. Burch HB, Wartofsky L. Life-threatening thyrotoxicosis. thyroid storm. the rat. Metabolism 1978;27:1680–90.
Endocrinol Metab Clin North Am 1993;22:263–77. 47. Davidson MB, Chopra IJ. Effect of carbohydrate and noncarbohydrate
35. Sanders V. Neurologic manifestations of myxedema. N Engl J Med 1962;266: sources of calories on plasma 3,5,3´-triiodothyronine concentrations in man.
599–603 concl. J Clin Endocrinol Metab 1979;48:577–81.
36. Klein I, Ojamaa K. Thyroid hormone and the cardiovascular system. N Engl 48. Chopra IJ. An assessment of daily production and significance of thyroidal
J Med 2001;344:501–9. secretion of 3, 3´, 5´-triiodothyronine (reverse T3) in man. J Clin Invest
37. Hylander B, Noree LO, Rosenqvist U, et al. Myxedema coma: a treatment 1976;58:32–40.
schedule. Lakartidningen 1980;77:1833–5. 49. Nagaya T, Fujieda M, Otsuka G, et al. A potential role of activated NF-kappa
38. Wartofsky L. Myxedema coma. Endocrinol Metab Clin North Am 2006;35: B in the pathogenesis of euthyroid sick syndrome. J Clin Invest
687–98, vii–viii. 2000;106:393–402.
39. Iwasaki Y, Oiso Y, Yamauchi K, et al. Osmoregulation of plasma vasopressin 50. Monig H, Arendt T, Meyer M, et al. Activation of the hypothalamo-
in myxedema. J Clin Endocrinol Metab 1990;70:534–9. pituitary-adrenal axis in response to septic or non-septic diseases:
40. Skowsky WR, Kikuchi TA. The role of vasopressin in the impaired water implications for the euthyroid sick syndrome. Intensive Care Med 1999;25:
excretion of myxedema. Am J Med 1978;64:613–21. 1402–6.
41. Fliers E, Wiersinga WM. Myxedema coma. Rev Endocr Metab Disord
2003;4:137–41. A complete list of references for this chapter can be found online at
42. Zulewski H, Muller B, Exer P, et al. Estimation of tissue hypothyroidism by a www.expertconsult.com.
new clinical score: evaluation of patients with various grades of
hypothyroidism and controls. J Clin Endocrinol Metab 1997;82:771–6.
Section II—Clinical and Laboratory Assessment 209.e1

References 30. Molenaar N, Johan Groeneveld AB, Dijstelbloem HM, et al. Assessing
adrenal insufficiency of corticosteroid secretion using free versus total
1. Cooper MS, Stewart PM. Adrenal insufficiency in critical illness. J Intensive cortisol levels in critical illness. Intensive Care Med 2011;37:1986–93.
Care Med 2007;22:348–62. 31. Annane D, Sebille V, Charpentier C, et al. Effect of treatment with low doses
2. Cohen J, Venkatesh B. Assessment of tissue cortisol activity. Crit Care Resusc of hydrocortisone and fludrocortisone on mortality in patients with septic
2009;11:287–9. shock. JAMA 2002;288:862–71.
3. Leonard MO, Godson C, Brady HR, et al. Potentiation of glucocorticoid 32. Sprung CL, Annane D, Keh D, et al. Hydrocortisone therapy for patients
activity in hypoxia through induction of the glucocorticoid receptor. with septic shock. N Engl J Med 2008;358:111–24.
J Immunol 2005;174:2250–7. 33. Dellinger RP, Levy MM, Carlet JM, et al. Surviving Sepsis Campaign:
4. Mebis L, Van den Berghe G. Thyroid axis function and dysfunction in international guidelines for management of severe sepsis and septic shock:
critical illness. Best Pract Res Clin Endocrinol Metab 2011 Oct;25(5):745–57. 2008. Crit Care Med 2008;36:296–327.
5. Clark PM, Gordon K. Challenges for the endocrine laboratory in critical 34. Burch HB, Wartofsky L. Life-threatening thyrotoxicosis: thyroid storm.
illness. Best Pract Res Clin Endocrinol Metab 2011;25(5):847–59. Endocrinol Metab Clin North Am 1993;22:263–77.
6. Thomas Z, Bandali F, McCowen K, et al. Drug-induced endocrine disorders 35. Sanders V. Neurologic manifestations of myxedema. N Engl J Med
in the intensive care unit. Crit Care Med 2010;38(6 Suppl):S219–30. Review. 1962;266:599–603 concl.
7. Vuong C, Van Uum SH, O’Dell LE, et al. The effects of opioids and opioid 36. Klein I, Ojamaa K. Thyroid hormone and the cardiovascular system.
analogs on animal and human endocrine systems. Endocr Rev 2010;31: N Engl J Med 2001;344:501–9.
98–132. 37. Hylander B, Noree LO, Rosenqvist U, et al. Myxedema coma: a treatment
8. Chin R. Adrenal crisis. Crit Care Clin 1991;7:23–42. schedule. Lakartidningen 1980;77:1833–5.
9. Cuthbertson BH, Sprung CL, Annane D, et al. The effects of etomidate on 38. Wartofsky L. Myxedema coma. Endocrinol Metab Clin North Am 2006;35:
adrenal responsiveness and mortality in patients with septic shock. Intensive 687–98, vii–viii.
Care Med 2009;35:1868–76. 39. Iwasaki Y, Oiso Y, Yamauchi K, et al. Osmoregulation of plasma vasopressin
10. Sonino N. The use of ketoconazole as an inhibitor of steroid production. in myxedema. J Clin Endocrinol Metab 1990;70:534–9.
N Engl J Med 1987;317:812–8. 40. Skowsky WR, Kikuchi TA. The role of vasopressin in the impaired water
11. Ashby H, DiMattina M, Linehan WM, et al. The inhibition of human excretion of myxedema. Am J Med 1978;64:613–21.
adrenal steroidogenic enzyme activities by suramin. J Clin Endocrinol Metab 41. Fliers E, Wiersinga WM. Myxedema coma. Rev Endocr Metab Disord 2003;
1989;68:505–8. 4:137–41.
12. Kyriazopoulou V, Parparousi O, Vagenakis AG. Rifampicin-induced adrenal 42. Zulewski H, Muller B, Exer P, et al. Estimation of tissue hypothyroidism by a
crisis in addisonian patients receiving corticosteroid replacement therapy. new clinical score: evaluation of patients with various grades of
J Clin Endocrinol Metab 1984;59:1204–6. hypothyroidism and controls. J Clin Endocrinol Metab 1997;82:771–6.
13. Wagner RL, White PF, Kan PB, et al. Inhibition of adrenal steroidogenesis by 43. Bigos ST, Ridgway EC, Kourides IA, et al. Spectrum of pituitary alterations
the anesthetic etomidate. N Engl J Med 1984;310:1415–21. with mild and severe thyroid impairment. J Clin Endocrinol Metab 1978;
14. Elias AN, Gwinup G. Effects of some clinically encountered drugs on steroid 46:317–25.
synthesis and degradation. Metabolism 1980;29:582–95. 44. Pereira VG, Haron ES, Lima-Neto N, et al. Management of myxedema coma:
15. Arlt W, Allolio B. Adrenal insufficiency. Lancet 2003;361:1881–93. report on three successfully treated cases with nasogastric or intravenous
16. Schmiegelow M, Feldt-Rasmussen U, Rasmussen AK, et al. Assessment of the administration of triiodothyronine. J Endocrinol Invest 1982;5:331–4.
hypothalamo-pituitary-adrenal axis in patients treated with radiotherapy 45. Chernow B, Burman KD, Johnson DL, et al. T3 may be a better agent than
and chemotherapy for childhood brain tumor. J Clin Endocrinol Metab T4 in the critically ill hypothyroid patient: evaluation of transport across the
2003;88:3149–54. blood-brain barrier in a primate model. Crit Care Med 1983;11:99–104.
17. Marik PE, Pastores SM, Annane D, et al. Recommendations for the diagnosis 46. Harris AR, Fang SL, Vagenakis AG, et al. Effect of starvation, nutriment
and management of corticosteroid insufficiency in critically ill adult patients: replacement, and hypothyroidism on in vitro hepatic T4 to T3 conversion in
consensus statements from an international task force by the American the rat. Metabolism 1978;27:1680–90.
College of Critical Care Medicine. Crit Care Med 2008;36:1937–49. 47. Davidson MB, Chopra IJ. Effect of carbohydrate and noncarbohydrate
18. Meduri GU, Yates CR. Systemic inflammation-associated glucocorticoid sources of calories on plasma 3,5,3´-triiodothyronine concentrations in man.
resistance and outcome of ARDS. Ann N Y Acad Sci 2004;1024:24–53. J Clin Endocrinol Metab 1979;48:577–81.
19. Annane D, Sebille V, Charpentier C, et al. Effect of treatment with low doses 48. Chopra IJ. An assessment of daily production and significance of thyroidal
of hydrocortisone and fludrocortisone on mortality in patients with septic secretion of 3, 3´, 5´-triiodothyronine (reverse T3) in man. J Clin Invest
shock. JAMA 2002;288:862–71. 1976;58:32–40.
20. Ligtenberg JJ, Zijlstra JG. The relative adrenal insufficiency syndrome 49. Nagaya T, Fujieda M, Otsuka G, et al. A potential role of activated NF-kappa
revisited: which patients will benefit from low-dose steroids? Curr Opin Crit B in the pathogenesis of euthyroid sick syndrome. J Clin Invest
Care 2004;10:456–60. 2000;106:393–402.
21. Sprung CL, Annane D, Keh D, et al. Hydrocortisone therapy for patients 50. Monig H, Arendt T, Meyer M, et al. Activation of the hypothalamo-
with septic shock. N Engl J Med 2008;358:111–24. pituitary-adrenal axis in response to septic or non-septic diseases:
22. Cronin CC, Callaghan N, Kearney PJ, et al. Addison disease in patients implications for the euthyroid sick syndrome. Intensive Care Med
treated with glucocorticoid therapy. Arch Intern Med 1997;157:456–8. 1999;25:1402–6.
23. Dimopoulou I, Tsagarakis S, Kouyialis AT, et al. Hypothalamic-pituitary- 51. Corssmit EP, Heyligenberg R, Endert E, et al. Acute effects of interferon-
adrenal axis dysfunction in critically ill patients with traumatic brain injury: alpha administration on thyroid hormone metabolism in healthy men.
incidence, pathophysiology, and relationship to vasopressor dependence and J Clin Endocrinol Metab 1995;80:3140–4.
peripheral interleukin-6 levels. Crit Care Med 2004;32:404–8. 52. Chopra IJ, Williams DE, Orgiazzi J, et al. Opposite effects of
24. Dimopoulou I, Tsagarakis S, Douka E, et al. The low-dose corticotropin dexamethasone on serum concentrations of 3,3´,5´-triiodothyronine (reverse
stimulation test in acute traumatic and non-traumatic brain injury: T3) and 3,3´5-triiodothyronine (T3). J Clin Endocrinol Metab 1975;41:
incidence of hypo-responsiveness and relationship to outcome. Intensive 911–20.
Care Med 2004;30:1216–9. 53. DeGroot LJ. “Non-thyroidal illness syndrome” is functional central
25. Glowniak JV, Loriaux DL. A double-blind study of perioperative steroid hypothyroidism, and if severe, hormone replacement is appropriate in light
requirements in secondary adrenal insufficiency. Surgery 1997;121:123–9. of present knowledge. J Endocrinol Invest 2003;26:1163–70.
26. Cooper MS, Stewart PM. Corticosteroid insufficiency in acutely ill patients. 54. Meyer S, Schuetz P, Wieland M, et al. Low triiodothyronine syndrome: a
N Engl J Med 2003;348:727–34. prognostic marker for outcome in sepsis? Endocrine 2011;39(2):167–74.
27. Nieboer P, van der Werf TS, Beentjes JA, et al. Catecholamine dependency in Epub 2011, Jan 6.
a polytrauma patient: relative adrenal insufficiency? Intensive Care Med 55. Brent GA, Hershman JM. Thyroxine therapy in patients with severe
2000;26:125–7. nonthyroidal illnesses and low serum thyroxine concentration. J Clin
28. Beishuizen A, Thijs LG, Vermes I. Patterns of corticosteroid-binding globulin Endocrinol Metab 1986;63:1–8.
and the free cortisol index during septic shock and multitrauma. Intensive 56. Van den Berghe G, de Zegher F, Baxter RC, et al. Neuroendocrinology of
Care Med 2001;27:1584–91. prolonged critical illness: effects of exogenous thyrotropin-releasing
29. Klaff LS, Wisse BE. Current controversy related to glucocorticoid and insulin hormone and its combination with growth hormone secretagogues. J Clin
therapy in the intensive care unit. Endocr Pract 2007;13:542–9. Endocrinol Metab 1998;83:309–19.
209.e2 Section II—Clinical and Laboratory Assessment

57. Roberts CG, Ladenson PW. Hypothyroidism. Lancet 2004;363:793–803. 79. Kelly DF, Gonzalo IT, Cohan P, et al. Hypopituitarism following traumatic
58. Bello G, Pennisi MA, Montini L, et al. Nonthyroidal illness syndrome and brain injury and aneurysmal subarachnoid hemorrhage: a preliminary
prolonged mechanical ventilation in patients admitted to the ICU. Chest report. J Neurosurg 2000;93:743–52.
2009;135:1448–54. 80. Ghigo E, Masel B, Aimaretti G, et al. Consensus guidelines on screening for
59. Hamblin PS, Dyer SA, Mohr VS, et al. Relationship between thyrotropin and hypopituitarism following traumatic brain injury. Brain Inj 2005;19:
thyroxine changes during recovery from severe hypothyroxinemia of critical 711–24.
illness. J Clin Endocrinol Metab 1986;62:717–22. 81. Hannon MJ, Sherlock M, Thompson CJ. Pituitary dysfunction following
60. Demers LM. Thyroid disease: pathophysiology and diagnosis. Clin Lab Med traumatic brain injury or subarachnoid haemorrhage: in “Endocrine
2004;24:19–28. Management in the Intensive Care Unit.” Best Pract Res Clin Endocrinol
61. Spencer CA, LoPresti JS, Patel A, et al. Applications of a new Metab 2011;25(5):783–98.
chemiluminometric thyrotropin assay to subnormal measurement. J Clin 82. Agha A, Phillips J, O’Kelly P, et al. The natural history of post-traumatic
Endocrinol Metab 1990;70:453–60. hypopituitarism: implications for assessment and treatment. Am J Med
62. Stockigt J. Assessment of thyroid function: towards an integrated 2005;118:1416.
laboratory: clinical approach. Clin Biochem Rev 2003;24:109–22. 83. Carlson NE, Brenner LA, Wierman ME, et al. Hypogonadism on admission
63. Van den Berghe G. Novel insights into the neuroendocrinology of critical to acute rehabilitation is correlated with lower functional status at admission
illness. Eur J Endocrinol 2000;143:1–13. and discharge. Brain Injury 2009;23(4):336–44.
64. Van den Berghe G, de Zegher F, Bouillon R. Clinical review 95: acute and 84. Schneider HJ, Schneider M, Saller B, et al. Prevalence of anterior pituitary
prolonged critical illness as different neuroendocrine paradigms. J Clin insufficiency 3 and 12 months after traumatic brain injury. Eur J Endocrinol
Endocrinol Metab 1998;83:1827–34. 2006;154:259–65.
65. Sonksen PH, Christiansen JS. Consensus guidelines for the diagnosis and 85. Aimaretti G, Ambrosio MR, Di Somma C, et al. Residual pituitary function
treatment of adults with growth hormone deficiency. Growth Hormone after brain injury-induced hypopituitarism: a prospective 12-month study.
Research Society. Growth Horm IGF Res 1998;8(Suppl B):89–92. J Clin Endocrinol Metab 2005;90:6085–92.
66. Takala J, Ruokonen E, Webster NR, et al. Increased mortality associated with 86. Cohan P, Wang C, McArthur DL, et al. Acute secondary adrenal insufficiency
growth hormone treatment in critically ill adults. N Engl J Med 1999; after traumatic brain injury: a prospective study. Crit Care Med
341:785–92. 2005;33:2358–66.
67. Van den Berghe G, Wouters P, Weekers F, et al. Reactivation of pituitary 87. Klose M, Brennum J, Poulsgaard L, et al. Hypopituitarism is uncommon
hormone release and metabolic improvement by infusion of growth after aneurysmal subarachnoid haemorrhage. Clinical Endocrinology (Oxf)
hormone-releasing peptide and thyrotropin-releasing hormone in 2010;73(1):95–101.
patients with protracted critical illness. J Clin Endocrinol Metab 88. Parenti G, Cecchi PC, Ragghianti B, et al. Evaluation of the anterior pituitary
1999;84:1311–23. function in the acute phase after spontaneous subarachnoid hemorrhage.
68. Balanescu S, Kopp P, Gaskill MB, et al. Correlation of plasma copeptin and J Endocrinol Invest 2010;34(5):361–5.
vasopressin concentrations in hypo-, iso-, and hyperosmolar states. J Clin 89. Kreitschmann-Andermahr I, Poll E, Hutter BO, et al. Quality of life and
Endocrinol Metab 2011;96:1046–52. psychiatric sequelae following aneurysmal subarachnoid haemorrhage: does
69. Morgenthaler NG, Struck J, Alonso C, et al. Assay for the measurement of neuroendocrine dysfunction play a role? Clin Endocrinol (Oxf) 2007;66:
copeptin, a stable peptide derived from the precursor of vasopressin. Clin 833–7.
Chem 2006;52:112–9. 90. Vespa P. Endocrine function following acute SAH. Neurocrit Care 2011;15:
70. Fenske W, Stork S, Blechschmidt A, et al. Copeptin in the differential 361–4.
diagnosis of hyponatremia. J Clin Endocrinol Metab 2009;94:123–9. 91. Kelly DF, Gonzalo IT, Cohan P, et al. Hypopituitarism following traumatic
71. Katan M, Morgenthaler NG, Dixit KC, et al. Anterior and posterior pituitary brain injury and aneurysmal subarachnoid hemorrhage: a preliminary
function testing with simultaneous insulin tolerance test and a novel report. J Neurosurg 2000;93:743–52.
copeptin assay. J Clin Endocrinol Metab 2007;92:2640–3. 92. Savaridas T, Andrews PJ, Harris B. Cortisol dynamics following acute severe
72. Bartter FC, Schwartz WB. The syndrome of inappropriate secretion of brain injury. Intensive Care Med 2004;30:1479–83.
antidiuretic hormone. Am J Med 1967;42:790–806. 93. Bendel S, Koivisto T, Ruokonen E, et al. Pituitary-adrenal function in
73. Harrigan MR. Cerebral salt wasting syndrome: a review. Neurosurgery patients with acute subarachnoid haemorrhage: a prospective cohort study.
1996;38:152–60. Crit Care 2008;12:R126.
74. Fatemi N, Dusick JR, Mattozo C, et al. Pituitary hormonal loss and recovery 94. Weant KA, Sasaki-Adams D, Dziedzic K, et al. Acute relative adrenal
after transsphenoidal adenoma removal. Neurosurgery 2008;63: insufficiency after aneurysmal subarachnoid hemorrhage. Neurosurgery
709–18; discussion 18–19. 2008;63(4):645–9; discussion 649–50.
75. Kristof RA, Rother M, Neuloh G, et al. Incidence, clinical manifestations, 95. Zetterling M, Engström BE, Hallberg L, et al. Cortisol and
and course of water and electrolyte metabolism disturbances following adrenocorticotropic hormone dynamics in the acute phase of subarachnoid
transsphenoidal pituitary adenoma surgery: a prospective observational haemorrhage. Br J Neurosurg 2011;25(6):684–92.
study. J Neurosurg 2009;111(3):555–62. 96. Weant KA, Sasaki-Adams D, Dziedzic K, et al. Acute relative adrenal
76. Behan LA, Agha A. Endocrine consequences of adult traumatic brain injury. insufficiency after aneurysmal subarachnoid hemorrhage. Neurosurgery
Horm Res 2007;68(Suppl 5):18–21. 2008;63:645–9; discussion 9–50.
77. Schneider HJ, Kreitschmann-Andermahr I, Ghigo E, et al. 97. Hasan D, Lindsay KW, Wijdicks EF, et al. Effect of fludrocortisone acetate in
Hypothalamopituitary dysfunction following traumatic brain injury and patients with subarachnoid hemorrhage. Stroke 1989;20:1156–61.
aneurysmal subarachnoid hemorrhage: a systematic review. JAMA 2007; 98. Gomis P, Graftieaux JP, Sercombe R, et al. Randomized, double-blind,
298:1429–38. placebo-controlled, pilot trial of high-dose methylprednisolone in
78. Chourdakis M, Kraus MM, Tzellos T, et al. Effect of early compared with aneurysmal subarachnoid hemorrhage. J Neurosurg 2010;112:681–8.
delayed enteral nutrition on endocrine function in patients with traumatic 99. Feigin VL, Anderson N, Rinkel GJ, et al. Corticosteroids for aneurysmal
brain injury: an open-labeled randomized trial. JPEN J Parenter Enteral Nutr subarachnoid haemorrhage and primary intracerebral haemorrhage.
2012;36(1):108–16. Epub 2011, Sep 30. Cochrane Database Syst Rev 2005;3:CD004583.
II
Chapter
22  

Renal, Electrolyte, and


Acid-Base Assessment
Guy M. Dugan

and dopamine; and (3) antidiuretic hormone (ADH). Tech-


Introduction niques to monitor volume status are discussed in Chapter 19.
Between 25% and 75% of critically ill patients are affected by
acute kidney injury (AKI), and AKI is an independent risk
factor for mortality in critically ill patients,1-4 including those Renin-Angiotensin-Aldosterone
with neurologic disorders such as traumatic brain injury (TBI)
or subarachnoid hemorrhage (SAH).5 Even small changes in
System
serum creatinine (SC) are associated with an increased risk of With hypovolemia, the juxtaglomerular apparatus releases
death.5 In addition, the incidence of end-stage renal disease renin that converts angiotensinogen from the liver to angio-
(ESRD) is increasing. These patients, who have significant tensin I. Angiotensin I is converted to angiotensin II by
comorbidities, represent an important group of patients who angiotensin-converting enzyme (ACE), primarily in the lung
require critical care. This chapter reviews renal physiology (Fig. 22.1). Angiotensin II is also produced by the zona glo-
and AKI and its assessment, including consensus definitions merulosa of the adrenal gland in response to hypovolemia or
(RIFLE: risk, injury, failure, loss, end-stage disease) from the hyperkalemia and has the following functions: (1) it increases
Acute Kidney Network (AKIN). In addition, an approach to Na+ and water resorption in the kidney at the proximal con-
acid-base disorders is described. Assessment of volume status voluted tubule (PCT); (2) it increases the production and
and sodium balance and its disorders are reviewed in Chapters release of aldosterone from the adrenal gland that in turn
19 and 21, respectively. causes an increase in Na+ resorption in the collecting tubule;
and (3) it is the most potent vasoconstrictor in the body and
increases blood pressure through its direct vasoconstrictive
Normal Renal Physiology effects.
The kidney maintains the extracellular environment by excret- Aldosterone acts on two types of cells in the collecting
ing the waste products of metabolism (urea, creatinine, and tubule—the principal cell and the intercalated cell. At the
uric acid) while simultaneously regulating the excretion of basolateral membrane of the principal cell, aldosterone
water and solutes (Na+, K+, H+) by causing changes in tubular increases the Na+-K+-adenosine triphosphate (ATPase) pump
reabsorption or secretion. The glomerular filtrate is more than that creates a concentration gradient between the tubular
60 times the volume of plasma, thus almost all of this fluid lumen and the principal cell (Fig. 22.2). This allows Na+ to
must be returned to the systemic circulation by tubular reab- flow down the concentration gradient with the secretion of
sorption. Conversely, solutes move from the peritubular capil- K+.7 The potassium-sparing diuretics, amiloride and triam-
lary through the cell and into the urine in a process called terene, act by directly closing Na+ channels, and spirono­
tubular secretion. Na+, Cl−, and H2O are reabsorbed, H+ is lactone competes with aldosterone. At the level of the
secreted, K+ and uric acid are both absorbed and secreted, and intercalated cell, aldosterone acts at the H+-ATPase pump to
filtered creatinine is excreted virtually unchanged.6 increase the secretion of H+ into the tubule lumen (Fig. 22.3).
Hypovolemia is monitored by baroreceptors (located in the As H+ is secreted into the lumen, HCO3− is formed inside the
carotid sinuses and afferent arterioles of the glomerulus), and cell and then resorbed into the plasma. Thus an increase in
plasma volume is regulated by the resorption or excretion aldosterone can result in a metabolic alkalosis and decreased
of sodium and water. The baroreceptors signal to increase activity results in type 4 renal tubular acidosis.
intravascular volume by three mechanisms: (1) renin- Sympathetic activity increases plasma volume by increas-
angiotensin-aldosterone system (RAAS); (2) sympathetic acti- ing Na+ resorption in the PCT and by stimulating renin
vation with the nerves releasing epinephrine, norepinephrine, release.
210 © Copyright 2013 Elsevier Inc. All rights reserved.
Section II—Clinical and Laboratory Assessment 211

Alternative Third
pathways ventricle
Angiotensinogen 1
e.g. chymase 4

Hypophysiotropic 2
area
Angiotensin I Hypothalamus
Renin Mammillary
Bradykinin body
Negative ACE Median eminence O.C.
Inactive
feedback Portal capillaries Stalk
fragments
INCREASED BLOOD PRESSURE
Anterior
Angiotensin II Posterior lobe
Sodium lobe
retention
AT2 receptor
AVP Pituitary
Aldosterone Oxytocin
(storage) 3

AT1 receptors
in adrenal
gland AT1 receptors in blood LH ACTH Prolactin
vessels → vasoconstriction FSH
TSH GH
Fig. 22.1  Diagram showing the renin-angiotensin pathway.
Fig. 22.4  Illustration of the hypothalamic pituitary axis.

Peritubular
Tubular Na 3Na
capillary Antidiuretic Hormone-Arginine
lumen ATPase
Vasopressin
2K
The hypothalamus regulates osmolality (water content)
through the mechanisms of thirst and antidiuretic hormone
Aldo-R Aldo (ADH). ADH is the generic name given to what is now
K known as arginine vasopressin that is synthesized in the
supraoptic and paraventricular nuclei of the hypothalamus
ANP-R and stored in the posterior lobe of the pituitary (Fig. 22.4).
There are three stimuli for ADH secretion: (1) hyperosmolal-
ity—as little as a 1% increase in osmolality will stimulate
ADH; (2) low circulating volume more than a 10% decrease
Fig. 22.2  Diagram illustrating the effect of aldosterone at the basolateral in volume or more than 10 to 15 mm Hg decrease in blood
membrane of the principal cell where it increases the NA+−K+−ATPase
pressure; and (3) inappropriate release (i.e., syndrome of
pump.
inappropriate antidiuretic hormone [SIADH]) that is dis-
cussed further in Chapter 21.
Tubular Peritubular
lumen capillary Plasma Osmolality
Plasma osmolality refers to the concentration of all the solutes
(electrolytes and nonelectrolytes) in the plasma and is nor-
ATPase H+
mally between 285 and 295 mmol/L. The kidney exerts control
over osmolality at several locations. In the proximal convo-
H2O Cl– luted tubule, sodium and water are resorbed in an isotonic
manner, so the osmolality of the fluid leaving is isotonic. At
the loop of Henle (LOH), Na+-K+-2 Cl− pumps remove elec-
OH– + CO2 HCO3
CA trolytes from the tubular fluid and in the process generate
K+ both a dilute urine and a hypertonic medullary interstitium.
ATPase ADH inserts preformed water channels into the collecting
H+
tubule that allows water to be osmotically reabsorbed and
to concentrate the urine (Fig. 22.5). Osmolality is estimated
by calculating the concentration of three solutes:
Fig. 22.3  Diagram illustrating aldosterone effects at the level of the
intercalated cell, where it acts at the H+−ATPase pump to increase the Osmolality = 2 × Na+ (mmol/L) + Blood urea nitrogen
secretion of H+ into the tubule lumen. (BUN) (mg/dL)/2.8 plus (glucose mg/dL)/18.
212 Section II—Clinical and Laboratory Assessment

Osmolarity of
300 interstitial fluid
300 100 (mosm/L)
300
100
300 300
Cortex H2O NaCl H2O
400 200
H2O H2O
NaCl 400 400
Outer H2O
NaCl H2O
medulla
H2O NaCl H2O
600 400 600 600
H2O
H2O NaCl
Urea
H2O
Inner H2O NaCl Urea
medulla 900 700 900
H2O
H2O NaCl Urea
1200 1200
1200

Active
transport
Passive
transport

Fig. 22.5  Illustration of the Counter Current Mechanism in the kidney and how the kidney affects osmolality at several locations.

BUN and glucose are measured in mg/dL and must be know that ethanol has a molecular weight of 46, so a level of
converted to mmol/L. To convert mg to mmol, divide the mg 230 mg/dL is 50 mmol/L (230 mg/dL/46 mg/mmol) ×10 dL/L
by the MW (MW = mg/mmol). Thus the conversion of BUN: = 50 mmol/L).
Urine concentration is measured by osmolality and by spe-
BUN = 18 mg/dL
cific gravity. Osmolality is the number of particles per liter and
(18 mg/dL/(28 mg/mmol) × 10 dL/L = 6.4 mmol/L. is determined by freezing point depression. It is used in evalu-
ation of hyponatremia and the investigation of polydipsia and
Similar osmolality calculations can be made for commonly polyuria. Specific gravity is the mass (in grams) of 1 mL of
infused intravenous (IV) solutions. D5W has: solution. Thus if there are particularly heavy particles such as
glucose or radiocontrast dye, the specific gravity will overes-
5 g/100 mL (or 5000 mg/dL/180 mg/mmol) × 10 dL/L timate the urine concentration compared with the osmolality.
= 278 mmol/L. There are several circumstances in which the measurement of
Also: urine osmolality may be important8:
0.9% normal saline (NS) = 0.9 g/dL = (900 mg/dL/58.5 mg/
1. Distinguishing between prerenal AKI and acute tubu-
mmol) × 10 dL/L = 154 mmol/L.
lar necrosis (ATN). A urine osmolality greater than
This is balanced by an equal number of Cl−, so the osmolal- 500  mosmol/kg suggests prerenal disease, in which
ity is 154 mmol/L + 154 mmol/L = 308 mmol/L, that is, iso- lower values are not helpful.
tonic. The units of osmolality are expressed as either mOsm/ 2. To distinguish between primary polydipsia and other
kg in conventional units or mmol/L in SI units. causes of hyponatremia. A urine osmolality less than
Because Mg+, Ca+, bilirubin, and albumin are not included 100 mosmol/kg indicates normal suppression of ADH and
in the calculation, there will always be a difference between therefore the presence of primary polydipsia. A higher
the measured and calculated osmolality. This osmolar gap is urine osmolality suggests the presence of at least some
less than 10 mmol/L, and a greater value indicates the accu- ADH usually due to effective volume depletion or SIADH.
mulation of osmoles that are either endogenous (lactic acid, 3. A decreasing urinary osmolality suggests that a spontane-
ketones) or exogenous (ethanol, methanol, ethylene glycol, ous water diuresis has begun to correct hyponatremia. This
isopropyl alcohol). In clinical practice it can be useful to helps to avoid inadvertent overcorrection in hyponatremic
Section II—Clinical and Laboratory Assessment 213

patients with reversible causes of water retention such as


volume depletion, pain, nausea, or drugs. Table 22.1  Causes of Nonrenal Elevations of
4. Urine osmolality helps distinguish between primary Blood Urea Nitrogen and Creatinine
polydipsia and diabetes insipidus (DI) as a cause of poly- Nonrenal causes of elevated urea
uria. After the plasma osmolality is increased by water • Corticosteroids
restriction to stimulate ADH release, the urine becomes • Hyperalimentation
concentrated with primary polydipsia. The urinary • Gastrointestinal bleeding
osmolality remains relatively dilute with both forms of • Tetracyclines
Nonrenal causes of elevated creatinine
DI, though patients with central DI have some concen- • Rhabdomyolysis-increased creatinine release from skeletal
tration after administration of desmopressin (DAVP). muscle
5. In patients who are hypernatremic, a urine osmolality less • Interference with creatinine assay
than plasma osmolality suggests the presence of DI. • Acetone
• Cefoxitin
• Flucytosine
• Methyldopa
AKI in the Intensive Care Unit Blocked tubular creatinine secretion
• Cimetidine
Overview • Trimethoprim
Acute renal failure is common in the intensive care unit (ICU),
with a prevalence of up to 25% to 75%, and it is an independent
risk factor for poor outcome with mortality rates from 28% to RIFLE and AKIN Criteria
90%. These wide ranges reflect that it is a complex, poorly The understanding of AKI has been hindered by the numer-
understood process with more than 30 definitions in the litera- ous definitions used in various studies, and an important
ture. The following section provides a template for evaluation contribution to conceptualization was the development of a
and management. A comprehensive review of therapy, however, consensus definition by the Acute Dialysis Quality Initiative
is beyond the scope of this chapter, and the reader is referred (ADQI) in which they introduced the RIFLE criteria.20 This
to recent reviews on this topic for further information.3,9-14 stratifies renal failure into:
Risk of renal dysfunction: a 1.5 times’ increase in SC;
Epidemiology urine output less than 0.5 mL/kg/hr times 6 hours
(GFR decrease >25%)
Standardization of the definition of AKI has resulted in a
Injury to the kidneys: twofold increase in SC; urine
better understanding of its incidence. Population-based
output less than 0.5 mL/kg/hr times 12 hours
studies suggest it is nearly as common as myocardial infarc-
(GFR decrease >50%)
tion.15,16 The causes of AKI are several. For example, Liano et
Failure of kidney function: threefold increase in SC;
al. collected data on 748 cases of AKI in a prospective, multi-
urine output less than 0.5 mL/kg/hr times 24 hours
center study from Madrid. The following causes were identi-
(GFR decrease 75%)
fied: ATN, 45%; prerenal, 21%; acute on chronic renal failure,
Loss of kidney function: defined as the need for renal
13%; urinary tract obstruction, 10%; glomerulonephritis or
replacement therapy (RRT) for more than 4 weeks
vasculitis, 4%; acute interstitial nephritis, 2%; and atheroem-
End-stage kidney disease: need for dialysis for longer than
boli, 1%.17 In the United States, epidemiologic data from the
3 months
Program to Improve Care in Acute Renal Disease (PICARD)
examined the etiology of AKI in 618 patients from five medical A modification of the RIFLE criteria was subsequently pro-
centers. The most common causes were ischemic acute tubular posed by the AKIN, which included representatives from the
necrosis (including sepsis and hypotension), prerenal disease ADQI group and from other nephrology and intensive care
(hypovolemia, hemorrhage), nephrotoxicity (radiocontrast societies. Their proposal for kidney failure resulted in a three-
media [RCM], rhabdomyolysis), AKI with cardiac disease stage classification that corresponds to risk (stage 1), injury
(heart failure and shock), AKI with liver disease (hepatorenal (stage 2), and failure (stage 3) of the RIFLE criteria. Loss and
syndrome, cirrhosis), and multifactorial etiologies.18 Multi- ESRD are removed form the staging system and defined as
center epidemiologic studies suggest that overall sepsis is now outcomes.
the most common cause of AKI in the ICU.1,3
Stage 1: Increase in SC of 0.3 mg/dL or an increase in 1.5
to 2 from baseline; urine output less than 0.5 mL/
Diagnosis kg/hr for more than 6 hours
Stage 2: Increase in SC more than two- to threefold from
Conceptually AKI refers to the acute decline in glomerular and
baseline; urine output less than 0.5 mg/kg/hr for
tubular function that results in azotemia (the accumulation of
more than 12 hours
nitrogenous waste products), and the inability to maintain
Stage 3: Increase in SC more than threefold from baseline
fluid and electrolyte homeostasis. The diagnosis of renal
or SC greater than or equal to 4 mg/dL with an
failure is based on a history and physical examination along
acute increase of at least 0.5 mg/dL; urine output
with general supporting chemistries, the urinalysis, assess-
less than 0.5 mg/kg/hr for 24 hours or anuria for
ment of the glomerular filtration rate (GFR; primarily the
12 hours
BUN and SC), and urine output. However, not all elevations
in BUN and creatinine indicate renal injury,19 and oliguria The addition of an absolute change in SC of more than or
(<400 mL/24 hr) is not a universal finding (Table 22.1). equal to 0.3 mL/kg/hr is based on data that show an 80%
214 Section II—Clinical and Laboratory Assessment

increase in mortality associated with changes in SC concen-


tration of as little as 0.3 to 0.5 mL/kg/hr.21 The last two cri- Table 22.2  Causes of Prerenal Acute
teria are identical to the RIFLE risk criteria. The criteria Kidney Injury
should be applied after volume resuscitation and urinary Intravascular volume depletion
tract obstruction excluded if oliguria is the sole diagnostic Hemorrhage, GI volume loss (diarrhea, vomiting), skin losses
criterion. These criteria may have their greatest applicability (sweat, burns), renal losses (diuretics, osmotic diuresis),
in helping to standardize definitions in clinical studies. third space fluid; e.g., pancreatitis, bowel surgery, crush
AKIN also recommended replacing the term acute renal injury
Decreased “effective” circulating volume
failure with acute kidney injury to represent the entire spec- CHF, nephritic syndrome, cirrhosis, sepsis
trum of acute renal failure. Renal vasoconstriction
However, these modifications may still be misleading in Liver failure, hypercalcemia, ACE inhibitors, ARB, NSAIDs
assessing AKI. The SC does not accurately reflect the GFR in Increased renal vein pressure/IVC obstruction
Abdominal compartment syndrome
a patient who is not in steady state. Thus a patient may be Hypotension from shock—cardiogenic, septic
developing AKI with a normal creatinine, and a rising creati- Selective ischemia—bilateral renal artery stenosis, often in the
nine may not imply ongoing kidney injury. Furthermore, cre- context of ACEI or ARB
atinine is removed by dialysis, so it is not possible to assess
ACE, Angiotensin-converting enzyme; ACEI, angiotensin-converting enzyme
kidney function by measuring it once dialysis is initiated. inhibitor; ARB, angiotensin receptor blocker; CHF, congestive heart failure;
Moreover, AKI may exist despite a normal urine output. GI, gastrointestinal; IVC, inferior vena cava; NSAIDs, nonsteroidal
anti-inflammatory drugs.
This has led to investigation into a number of biomarkers—
N-acetyl-glucosaminidase (NAG), neutrophil gelatinase–
associated lipocalin (NGAL), cystatin C, or kidney injury
molecule 1 (KIM-1) among others—that may better indicate
the state of kidney injury.3,22-26 Finally, RIFLE and AKIN cri- Table 22.3  Laboratory Findings That Help
teria rely in large part on SC and measurement of urine output Differentiate Prerenal AKI and ATN
for the diagnosis of AKI. Changes in SC may be delayed and
Laboratory Prerenal AKI ATN
in most ICUs urine output is measured manually, so these
measurements may be subject to human error. Newer tech- Urine specific gravity >1.020 ≈1.012
niques that provide continuous minute-to-minute monitor- Urine sodium, mEq/L <20 >30
ing of urine output may allow more accurate evaluation of BUN/SC ratio 20 10
urine output and hence early identification of AKI.27
Urine osmolality >350 ≈300
Urine/plasma osmolality >1.5 1.0
Classification of AKI
FENa <1% >1%
A standard strategy to classify AKI is to divide it into prerenal
FE urea <35% >50%
disease, postrenal disease, and intrinsic disease based on the
primary site of injury—the tubules, glomerulus, interstitium, AKI, Acute kidney injury; ATN, acute tubular necrosis; BUN, blood urea
nitrogen; FE, fractional excretion; FENa, fractional excretion of sodium;
or vessels. Prerenal disease may progress to tubular injury, and SC, serum creatinine
ATN may result from ischemic or toxic causes. These catego-
ries may have some clinical utility in organizing thinking, but
they imply an insight into pathophysiology (prerenal) or his-
tology (ATN) that is either greatly simplified or misleading.28 increase of BUN compared with creatinine with a BUN-
A complementary approach is to examine the injury in differ- to-SC ratio of greater than 10 : 1 to 15 : 1. As tubular function
ent disease states. An integrative approach using both of these is preserved, there is a high urine osmolality, greater than
strategies is used in this outline. 350 mOsm/L.
Of the tests available (see Table 22.3), the fractional excre-
tion of sodium (FENa) may be the preferred screening test to
Prerenal AKI and ATN differentiate between prerenal AKI and ATN (Table 22.4).
Prerenal AKI may result from intravascular volume depletion, FENa = (urine Na × serum creatinine)/
an alteration of glomerular hemodynamics, or vascular com- (serum Na × urine creatinine) ×100
promise (Table 22.2). Prerenal AKI may evolve into an entity
traditionally referred to as ATN; the distinctions based on Classically, a FENa less than 1% is characteristic of prerenal
urine chemistries are outlined in Table 22.3. Physiologically, AKI, whereas a FENa greater than 1% to 2% signifies ATN.
the reduction in blood pressure activates cardiac and arterial However, a low FENa is not unique to prerenal disease. It can
receptors that increases sympathetic neural tone (improving occur in disorders associated with normal tubular function
blood pressure but causing renal vasoconstriction), and but a low GFR such as acute glomerulonephritis (GN), vascu-
releasing both renin, which leads to the production of angio- litis, and acute urinary tract obstruction. It also can be seen
tensin II, and ADH. Angiotensin II and adrenergic activation when ATN is superimposed on a chronic sodium-retaining
stimulate the proximal reabsorption of sodium, and aldoste- state as may occur with aminoglycoside therapy, congestive
rone increases the sodium reabsorption in the distal tubule. heart failure (CHF), or cirrhosis.29 The confounding effects of
This results in a urinary sodium concentration of less than diuretics may be obviated by the use of the fractional excretion
20 mEq/L. There is enhanced reabsorption of urea from the of urea, which when less than 35% accurately indicates prer-
medullary collecting ducts, resulting in a disproportionate enal azotemia.30
Section II—Clinical and Laboratory Assessment 215

Table 22.4  Interpretation of the Fractional


Excretion of Sodium
FENa <1%
Prerenal AKI
Low GFR with normal tubular function
Acute glomerulonephritis
Vasculitis
Acute urinary tract obstruction
ATN
Chronic disease with sodium-retaining states
Aminoglycosides
Cirrhosis
CHF
Rhabdomyolysis
Contrast nephropathy
FENa >1%-2%
ATN
Prerenal AKI with: Fig. 22.7  Urine sediment showing free red cells and a red cell cast
Glucosuria that is tightly packed with red cells. Red cell casts are virtually
Diuretics diagnostic of glomerulonephritis or vasculitis. (Courtesy Harvard Medical
Acute on chronic disease School.)
Mannitol

AKI, Acute kidney injury; ATN, acute tubular necrosis; CHF, congestive heart
failure; FENa, fractional excretion of sodium; GFR, glomerular filtration rate.
Glomerular Disease in AKI
Acute glomerulonephritis may be the cause of AKI in the
ICU due to bacterial endocarditis, staphylococcal sepsis,
visceral abscesses, hepatitis B, systemic lupus erythematosus
(SLE), Goodpasture syndrome, or rapidly progressive glo-
merulonephritis (RPGN). Red cells and red blood cell casts,
and proteinuria are seen on urinalysis31 (Fig. 22.7). An alter-
ation of glomerular hemodynamics also may contribute to
AKI. Afferent vasoconstriction may be seen in hepatorenal
syndrome and efferent vasodilation from ACE inhibitors.
Vasodilators such as nitroprusside and nifedipine may con-
tribute to AKI by altering intrarenal hemodynamics without
systemic hypotension.39

Interstitial Disease in AKI


Drugs most often induce acute interstitial nephritis, but infec-
tions such as from Legionella, Leptospira, and streptococcal
Fig. 22.6  Urine sediment on urinalysis showing multiple muddy brown organisms, and autoimmune disorders may also be responsi-
granular casts that suggest acute tubular necrosis. (Courtesy Harvard
ble. The most commonly implicated drugs are beta-lactam
Medical School.)
antibiotics, sulfonamides, rifampin, and nonsteroidal anti-
inflammatory drugs (NSAIDs). The complete clinical spec-
The urinalysis in prerenal disease is normal (this may trum presents as fever, rash, arthralgias, and eosinophilia, and
include granular casts), whereas ATN has muddy brown casts31 a urine analysis shows sterile pyuria with white blood cell
(Fig. 22.6). Pathologically, tubular necrosis is usually not seen casts, and eosinophiluria (except NSAIDs). Hematuria and
with ATN, and there is often limited histologic evidence of proteinuria also are common. The FENa usually is greater
injury despite marked functional impairment.32 ATN may be than 1%, indicating tubular damage, but lower values may be
induced by those factors that cause prerenal AKI as well as by seen in mild disease.40
drugs, rhabdomyolysis, tumor lysis, and vascular insults.
Diuretics may convert ATN from an oliguric to a nonoliguric
state and may be tried for volume control, but they have no AKI in Cirrhosis
effect on renal recovery or survival.33,34 Loop diuretics work by Patients with cirrhosis or acute hepatitis can develop renal
inhibition of a Na+−K+−2Cl− transporter in the loop of Henle. failure without proteinuria and with a reduced urinary
A secretory isoform of this transporter is present in the inner sodium. The hallmark is intense renal vasoconstriction with
ear endolymph, and thus high-dose diuretics may lead to per- peripheral vasodilation. The kidneys in patients with hepa-
manent hearing loss.35 Dopamine does not provide protection torenal syndrome (HRS) are normal on autopsy and func-
in early ATN and may cause harm by reducing renal blood tioned normally when transplanted into patients without
flow in postischemic ATN.36-38 Prevention of ATN by optimiz- cirrhosis. Moreover, HRS can be reversed following liver
ing volume status and avoiding nephrotoxic agents remain the transplantation.41-43 In 1996 the International Ascites Club
mainstay of management. published a consensus paper that subdivided HRS into two
216 Section II—Clinical and Laboratory Assessment

types. Type 1 HRS is characterized by a rapid decline in renal


function with a doubling of SC to more than 2.5 mg/dL or
having a creatinine clearance to less than 20 mL/min within
2 weeks. In type 2 HRS the presentation is of stable renal
failure in a patient with refractory ascites in which the SC
increases to more than 1.5 mg/dL or a creatinine clearance of
less than 40 mL/min.44 Untreated type 1 has mortality as high
as 80% in 2 weeks, whereas type 2 has a median survival of
approximately 6 months.
The precipitating factors for HRS have been identified
as bacterial infection (48%), gastrointestinal (GI) bleeding
(33%), aggressive paracentesis (27%), and drugs (diuretics,
aminoglycosides, nonsteroidal anti-inflammatory drugs
[NSAIDs], and angiotensin-converting enzyme inhibitors
[ACEIs]/angiotensin II receptor blockers [ARBs]). However,
24% develop type 1 HRS without an obvious precipitating
factor.45 A high index of suspicion is needed to assess AKI in
the context of advanced liver disease because reduction in
muscle may render a SC within the normal range even in the
context of a decreased GFR, and GI bleeding and the amount
of protein in the diet may affect the BUN. Sepsis should be
suspected in any cirrhotic patient with AKI even in the absence
of leukocytosis and fever. Clinically the syndrome presents
with prerenal physiology (i.e., oliguria, a benign urine sedi-
ment, and low urine sodium).
Treatment may include midodrine (an alpha agonist) and
octreotide (a long-acting form of somatostatin that inhibits
endogenous vasodilator release), with albumin, and terlipres-
Fig. 22.8  Cutaneous manifestations of cholesterol emboli syndrome.
sin (a vasopressin analog used in Europe).46,47 Dialysis should
be offered only if there is a chance for liver transplantation in
the short term. Transjugular intrahepatic portosystemic shunt
(TIPS) involves placing a stent between the hepatic vein and 22.8). Anticoagulation is dangerous in patients with CES but
the intrahepatic portion of the portal vein. It is primarily may be necessary in patients with a thrombotic event.50
designed to treat bleeding varices, but it has been used for
HRS, though it may worsen encephalopathy. It may serve as a
bridge to liver transplantation, which is the only effective Abdominal Compartment Syndrome
treatment.48,49 Abdominal compartment syndrome (ACS) refers to symp-
tomatic organ dysfunction arising from an increase in intra-
abdominal pressure (IAP; See also Chapter 23). A pressure
Vascular Diseases Causing AKI of 20 with organ dysfunction is the operational definition
A number of vascular disorders of both the large and small used in studies, but in clinical practice a single IAP cannot
vessels may give a prerenal picture of AKI. Large vessel disease predictably diagnose ACS in all patients. ACS arises from
may arise from operative arterial cross-clamping, renal artery tissue edema or third-spaced fluid and may occur in diverse
thrombosis or embolism, or renal artery stenosis. Renal infarc- clinical states such as abdominal surgery, pancreatitis, and
tion may present with fever, hematuria, acute flank pain, ileus, bleeding. Hypotension may arise from the ensuing inferior
and leukocytosis that may mimic an acute abdomen. Hyper- vena cava (IVC) obstruction, and be accompanied by a high
tensive crises may disrupt the vascular endothelium and be pulmonary artery occlusion pressure or elevated femoral vein
associated with thrombocytopenia and microangiopathy, reti- pressure despite a reduced venous return. Clinically the kidney
nopathy, and AKI. Systemic vasculitis such as Wegener’s gran- dysfunction looks similar to prerenal kidney disease with a low
ulomatosis, polyarteritis nodosa, hypersensitivity vasculitis, urinary sodium, and an increase in plasma renin, aldosterone,
and Henoch-Schönlein purpura often cause AKI. and ADH level. Oliguria may develop at an IAP of 15 mm Hg
Cholesterol emboli syndrome (CES) arises following aortic and anuria at 30 mm Hg.51 A high index of suspicion is needed
manipulation from percutaneous intervention, surgery, or because this condition may look like evolution of the underly-
systemic anticoagulation. The crystals incite an inflammatory ing disease process with hypotension, acute respiratory dis-
reaction and adventitial fibrosis may obliterate the vessel tress syndrome (ARDS), and AKI from multiorgan failure.52
lumen. Hypocomplementemia and eosinophilia are seen, and
any organ may be affected including the kidney, the GI tract
with bleeding from microinfarcts, the skin, and the central Drugs and AKI
nervous system (CNS) with stroke. The cutaneous manifesta- Many medications can induce kidney failure and may lead to
tions may include a blue toe or livedo reticularis that results a picture of prerenal AKI, ATN, or acute interstitial nephritis.
from a superficial infarct and compensatory dilation of the A complete discussion is beyond the scope of this chapter, but
surrounding vessels to give a livedo or “lacelike” pattern (Fig. some common drugs are discussed.
Section II—Clinical and Laboratory Assessment 217

NSAIDs and Acetaminophen-Induced AKI Recovery to baseline generally occurs spontaneously within
Prostacyclin and prostaglandin E2 preserve renal blood flow 1 to 4 weeks, and acetylcysteine has no protective effect on the
and glomerular filtration and inhibition of prostaglandin syn- kidney as it does with the liver.58 Drugs that increase cyto-
thesis with a NSAID can lead to a reversible AKI with an SC chrome P450 activity such as phenytoin, phenobarbital,
increase within the first 3 to 7 days.53,54 This hemodynamically rifampin, and carbamazepine enhance the risk of acetamino-
mediated injury may be less with aspirin because there is only phen nephrotoxicity.59
partial inhibition of glomerular cyclooxygenase.55 Acute inter-
stitial nephritis and the nephritic syndrome also may occur
with NSAIDs and it presents with hematuria, pyuria, white Radiocontrast Media–Induced AKI
cell casts,31 and proteinuria along with an acute rise in SC (Fig. Radiocontrast media (RCM) are either ionic or nonionic and
22.9). The complete syndrome of fever, rash, eosinophilia, and are of variable osmolality. Risk factors for RCM–induced AKI
eosinophiluria may be absent. Spontaneous recovery usually include underlying renal insufficiency (SC >1.5 mg/dL), dia-
occurs within weeks to months after discontinuation of betic nephropathy, heart failure, volume depletion, PCI (pro-
therapy.56 Membranous nephropathy with proteinuria has motes the development of atheroemboli), and the dose and
been reported to occur with diclofenac, but probably any tonicity of the RCM agent. Renal injury is observed within
NSAID may be involved.57 the first 12 to 24 hours with recovery typically beginning
Acetaminophen-induced acute renal failure may occur with within 3 to 5 days.60,61 Patients with diabetes and heart failure
an overdose, but some alcoholics are at increased risk from are at increased risk of AKI from RCM, possibly due to
therapeutic doses. The pathologic changes reveal ATN, and the impaired nitric oxide (NO) generation.62 Many of the com-
uric acid shows granular and epithelial cell casts31 (Fig. 22.10). monly used RCM agents induce a false-positive result when
either a dipstick or sulfosalicylic acid is used to detect pro-
teinuria, and this should not be tested for at least 24 hours
after a contrast study.63 ATN may result; however, the FENa is
often less than 1%. Prevention is best accomplished with
hydration, often with sodium bicarbonate. N-acetylcysteine
may also have protective benefit.64

Aminoglycosides
These are concentrated in the proximal tubular cells, and AKI
can become evident several days after the drug has been
stopped. Distal tubular damage also occurs with a loss in
renal concentrating ability resulting in polyuria and hypo-
magnesemia. Risk factors include volume contraction, age,
hypokalemia, and a short dosing interval. The routine moni-
toring of peak and tough levels does not decrease the likeli-
hood of nephrotoxicity.

Fig. 22.9  White cell cast in which blue-stained white cells (arrow) are
contained within a granular cast. (Courtesy Frances Andrus, BA, Victoria Lithium
Hospital, London, Ontario, Canada.) Lithium may cause a nephrogenic DI by causing ADH resis-
tance in up to 20% of patients. Some data suggest lithium
levels may increase with concomitant ACE inhibitors.65,66

ACE Inhibitors
The primary side effects occur by one of two mechanisms. A
reduction in angiotensin II formation may cause hypoten-
sion, AKI, hyperkalemia, and problems during pregnancy.
Cough, edema, and anaphylactoid reactions are related to
increased kinins because ACE is also a kininase. The side
effects related to reduced angiotensin II, but not those related
to kinins, are also seen with the angiotensin receptor block-
ers. AKI is seen predominantly in patients with bilateral renal
artery stenosis, hypertensive nephrosclerosis, CHF, polycystic
kidney disease, or chronic renal failure. In each of these the
GFR is partially maintained by efferent arteriolar vasocon-
striction mediated by angiotensin II.67 The rise in
Fig. 22.10  Epithelial cell cast with free epithelial cells (arrow) in SC usually occurs in 3 to 5 days. Hyperkalemia may result
the urine sediment. Renal tubular epithelial cells are larger than white from ACEI/ARBs because angiotensin II increases aldoste-
cells and have a single, large central nucleus. (Courtesy Frances Andrus, BA, rone that is the major hormonal stimulus to urinary potas-
Victoria Hospital, London, Ontario, Canada.) sium excretion.
218 Section II—Clinical and Laboratory Assessment

Fig. 22.11  Skin involvement is characteristic with scleromyxedema-like fibrosis in nephrogenic systemic fibrosis (NSF) that can complicate Gadolinium
administration.

Table 22.5  Causes of Rhabdomyolysis


• Trauma and compartment syndromes
• Seizures, dystonic reactions
• Impaired energy supply—carbon monoxide poisoning,
mitochondrial myopathies
• Malignant hyperthermia and neuroleptic malignant
syndrome
• Near-drowning with hypothermia
• Drugs
Anticholinergics due to the loss of impaired sweating
Statins, colchicine—direct myotoxins
Macrolides, cyclosporine, gemfibrozil, and protease
inhibitors interfere with statin clearance and potentiate
toxicity
• Cocaine-induced hyperthermia
• Carbon monoxide—insufficient energy production
• Infections—ehrlichiosis, falciparum malaria
Fig. 22.12  Uric acid crystals seen on the urinalysis. These crystals • Electrolyte disorders—hypokalemia, hypophosphatemia (in
are pleomorphic, most often appearing as rhombic plates or rosettes. alcoholics receiving total parenteral nutrition without
They are yellow or reddish-brown and form only in an acid urine. (Courtesy phosphate supplementation)
Harvard Medical School.)

Amphotericin B (>15 mg/dL) in patients with tumors associated with high cell


Amphotericin B results in renal insufficiency with K+ and turnover or with chemotherapy (e.g., after treatment of leu-
Mg++ wasting and metabolic acidosis due to a type 1 or distal kemias and lymphoma, or other malignancies including
RTA and polyuria due to a nephrogenic DI. medulloblastomas). AKI results from tubular obstruction by
precipitated uric acid crystals or depiction of calcium phos-
phate complexes in the tubular lumen and interstitium.69
Gadolinium Patients have decreased urine output with uric acid crystals
Gadolinium may be contraindicated in patients with a GFR seen on the urinalysis31 (Fig. 22.12). Treatment includes ade-
less than 30 mL/min or any degree of renal insufficiency in quate hydration and allopurinol. The role of urinary alkalini-
patients with hepatorenal syndrome because its use is associ- zation is controversial.70
ated with nephrogenic systemic fibrosis (NSF). The latency
of NSF is usually 2 to 4 weeks, but may be between 2 days
and 18 months. Skin involvement is characteristic with Pigment-Induced AKI
scleromyxedema-like fibrosis that may lead to disabling joint Myoglobin from rhabdomyolysis and hemoglobin from intra-
contractures (Fig. 22.11). Systemic involvement may include vascular hemolysis are heme pigments that may induce AKI.
the lungs, pleura, myocardium, pericardium, and dura. Scleral Hemolysis leading to AKI is uncommon and usually arises
plaques are common.68 from a transfusion reaction due to ABO incompatible blood.
Rhabdomyolysis is more common and may arise from a
variety of causes (Table 22.5). Patients with rhabdomyolysis
Acute Tumor Lysis Syndrome, Acute Uric present with the triad of pigmented granular casts, elevated
Acid Nephropathy, and AKI creatine kinase (CK), and myoglobinuria—a red to brown
Acute tumor lysis syndrome consists of hyperphosphatemia urine sediment that tests positive for heme but without red
(>8 mg/dL), hypocalcemia, hyperkalemia, and hyperuricemia blood cells. Myoglobin is rapidly cleared, so one may see
Section II—Clinical and Laboratory Assessment 219

elevated CK levels without myoglobinuria. The plasma is


normal color unless its excretion is impaired from renal insuf- Table 22.6  Causes of Postrenal Failure
ficiency. Patients with hemolysis have a similar finding in their Bladder outlet obstruction
urine but have a reduced haptoglobin and abnormal periph- Prostate enlargement
eral smear with red plasma.71 Pelvic pathology
The renal injury results from obstruction of the tubules Crystalluria
from heme pigment casts, tubular injury from free chelatable Acyclovir
Indinavir
iron, and volume depletion. In contrast to other forms of Uric acid
ATN, the FENa is often less than 1%. In rhabdomyolysis, Papillary tip necrosis
abnormalities in serum electrolytes are common and include Diabetes mellitus with pyelonephritis
metabolic acidosis, hyperuricemia, hyperkalemia, and hyper- Analgesic abuse
Sickle cell disease
phosphatemia. Initial hypocalcemia, caused from the deposi-
tion of calcium in damaged muscle and decreased bone
responsiveness to parathyroid hormone (PTH), may subse-
quently be followed by hypercalcemia during recovery when Postrenal AKI
this calcium is released and secondary hyperparathyroidism Although an uncommon cause of AKI in the ICU, postrenal
from AKI.72 obstruction represents a reversible form of renal injury.
Treatment is with saline, potentially up to 1 to 2 L/hr to Imaging of the renal system is required for proper manage-
maintain a urine output of 200 to 300 mL/hr. After diuresis is ment of AKI as well as measuring postvoid residuals (>50 mL
established, urinary alkalinization with 75 mmol of sodium is abnormal). As intratubular pressure increases, glomerular
bicarbonate in 1 L 0.45 NS with a goal of a urine pH above filtration pressure decreases. Postrenal AKI can be divided into
6.5 can be initiated. This may mitigate some of the deleteri- renal and extrarenal causes (Table 22.6). Intrarenal causes
ous effect of myoglobin (or hemoglobin), but it may induce include crystal deposition that may occur with ethylene glycol,
hypocalcemia by precipitating calcium phosphate with uric acid nephropathy that may occur in tumor lysis syn-
ensuing tetany, seizures, or arrhythmias. Close monitoring of drome, or tubular obstruction from light chain casts that may
serum bicarbonate, calcium, potassium, and urine pH is occur with multiple myeloma. Extrarenal causes may include
required. A loop diuretic or mannitol may be added if diure- prostate disease, pelvic malignancy, and retroperitoneal disor-
sis is not established after volume repletion. Mannitol may be ders.75 It is important to recognize that patients with a partial
beneficial with a CK greater than 20,000 to 30,000 U/L, but it obstruction may have a normal or increased urine output due
is recommended that the plasma osmolality and osmolal gap to a secondary concentrating defect.
be measured every 4 to 6 hours. Therapy is continued until The commonly discussed “postobstructive” diuresis is
the urine discoloration clears and the plasma CK is less than usually a physiologic response to excrete the fluid retained
5000 to 10,000 U/L or if the urine pH does not increase during the obstruction, and assiduous replacement of this can
above 6.5 after 4 hours or the osmolal gap rises above lead to volume overload. There may be the need for some
55 mosmol/kg.73 fluid above baseline due to a mild sodium-wasting tendency
and a mild concentrating defect due to the down-regulation
of water channels. Half normal saline at 75 mL/hr, with atten-
AKI in Pregnancy tion to volume status, is suggested as an appropriate initial
AKI may occur in association with microangiopathic hemo- therapy.76
lytic anemia and thrombocytopenia in late pregnancy. Two
entities of principal concern are thrombotic thrombocytope-
nic purpura and hemolytic uremic syndrome (TTP-HUS)
Renal Replacement Therapy in AKI
and severe preeclampsia, usually with the HELLP syndrome The acute indications for RRT include volume overload, life-
(hemolysis, elevated liver enzymes, low platelets). TTP-HUS threatening metabolic abnormalities such as hyperkalemia
probably exists as a continuum, but TTP is usually seen ante- and acidosis, acute pericarditis, and encephalopathy. A detailed
partum and is diagnosed when neurologic abnormalities pre- description of RRT is beyond the scope of this chapter; the
dominate, whereas HUS is characterized by more severe renal reader is referred to recent reviews for further informa-
failure and is seen postpartum. These distinctions are fre- tion.10,12,77-80 The objectives of RRT are to remove fluid by
quently unclear and may not be important for manage- ultrafiltration and control azotemia by removing excess solute.
ment.74 The therapy of TTP-HUS in pregnancy is plasma Although peritoneal dialysis is still used, RRT is almost always
infusion with or without plasma exchange as in the nonpreg- accomplished by intravascular access in the ICU. Intermittent
nant patient. Delivery is indicated for the HELLP variant of hemodialysis (IHD) is the most frequently used form of RRT
preeclampsia. in the ICU and is accomplished by a central venous double-
Acute fatty liver of pregnancy results from fatty infiltration lumen catheter that passes the blood through a dialysis mem-
of hepatocytes with inflammation and is associated with AKI brane. Studies have demonstrated improved clinical outcomes
in 60% of cases. Patients have elevated liver function tests with the use of synthetic biocompatible membranes (polysul-
(LFTs) with disseminated intravascular coagulation (DIC) fone, polyamide, polyacrylonitrile, and polymethyl methacry-
and delivery is indicated. Bilateral renal cortical necrosis is a late) compared to cellulose-based membranes (cellulose
complication of pregnancy caused by abruptio placentae, pla- acetate, cuprophane.) Hypoxemia with a PaO2 decrease of
centa previa, intrauterine fetal death, or amniotic fluid embo- 10 torr may occur, but the major complication of IHD is
lism. Patients present with a triad of anuria, gross hematuria, hypotension that may prompt consideration for different
and flank pain. There is no specific therapy. continuous renal replacement therapy (CRRT) strategies.
220 Section II—Clinical and Laboratory Assessment

However, there is no survival advantage with CRRT compared


with IHD, and the RRT choice may depend on a variety of Table 22.7  Antibiotic Dosing
factors. When rapid solute control is necessary such as hyper- Recommendations in CRRT85
kalemia, IHD is most suitable. Slow continuous ultrafiltration Drug CVVH CVVHD (HDF)
(SCUF) may be preferred if volume overload without
Amikacin 7.5 mg/kg q24h Same
azotemia is present. Continuous veno-venous hemofiltration
(CVVH) is indicated with hepatic failure, head trauma, or Amphotericin
septic shock.19   Deoxycholate 0.4-1 mg/kg q24h Same
  Lipid complex 3-5 mg/kg q24h Same
Neurologic complications can be common in patients   Liposomal 3-5 mg/kg q24h Same
undergoing hemodialysis. The dialysis disequilibrium syn-
Acyclovir 5-7.5 mg/kg q24h Same
drome refers to headache, nausea, delirium, and asterixis that
may progress to seizures, coma, or death. It is thought to result Ampicillin-sulbactam 3 g q12h 3 g q8h
from cerebral edema, and risk factors include older age, severe Aztreonam 1-2 g q12h 2 g q12h
acidosis, and new hemodialysis patients with markedly ele- Cefazolin 1-2 g q12h 2 g q12h
vated BUN. Milder symptoms such as muscle cramps and
anorexia are part of this syndrome. Erythropoietin may cause Cefepime 1-2 g q12h 2 g q12r
a rapid increase in blood pressure and result in encephalopa- Cefotaxime 1-2 g q12h 2 g q12h
thy and seizures. Other medications may precipitate seizures Ceftazidime 1-2 g q12 h 2 g q12h
in the hemodialysis population including beta-lactam antibi-
Ceftriaxone 2 g q12-24h Same
otics, meperidine, metoclopramide, and acyclovir. Hypoten-
sion or a reactive hypertension from a reactive vasoconstriction Cefuroxime 1.5 g q8-12h 1.5 g q8h
from a reactive hypotension may precipitate seizures. Dialysis Clindamycin 600-900 mg q8h Same
patients are at increased risk for intracerebral hemorrhage Ciprofloxacin 200 mg q12h 200-400 mg q12h
(ICH) and SAH, and subdural hematoma and ischemic
Colistin 2.5 mg/kg q48h Same
strokes.81 CRRT may affect the dosing of antibiotics, and Table
22.7 reflects the changes that may be needed. Daptomycin 4 or 6 mg/kg q48h Same
Doxycycline 100-200 mg q12h Same

Prognosis in AKI Fluconazole 200-400 mg q24h 400-800 mg q24h

Nonoliguric AKI, as well as AKI that is associated with ami- Imipenem-cilastatin 250 mg q6h or 250 mg q6h or
500 mg q8h 500 mg q8h
noglycosides and RCM, may have a mortality of 25% to 30%. 500 mg q6h
AKI occurring in the context of septic shock and burns has a
Itraconazole No change No change
mortality rate of 70% to 90%, usually as a result of infection.50
If the patient survives, oliguria averages 10 to 14 days. Over Levofloxacin 250 mg q24h Same
the next 3 to 7 days is an increase in urinary volume, but a Linezolid 600 mg q12h Same
sustained rise in the BUN and creatinine. Fluid administration Meropenem 1 g q12h Same
is often needed, and dialysis can usually be discontinued. Most
survivors regain function within 30 days, although rarely Metronidazole 500 mg q6-8h Same
recovery may extend to 90 days.19 Moxifloxacin 400 mg q24h Same
Nafcillin or oxacillin 2 g q4-6h Same
Acid-Base Balance in Critical Care Piperacillin- 2.25 g q6h 2.25-3.375 g q6h
tazobactam
A number of rules should be learned, but an important prin-
Rifampin 300-600 mg q12h Same
ciple is that all acid-base assessment must be accompanied
by a clinical history because this will help elucidate which Ticarcillin-clavulanate 2 g q6-8h 3.1 g q6h
of several possible patterns reflect clinical reality.7,82,83 An Tobramycin and Load 2 mg/kg then
approach to analysis of acid-base disorders is provided in gentamicin 1.5-2 mg/kg q24h
Table 22.8. Also, because an arterial blood gas (ABG) is and check levels
obtained in the evaluation of these disorders, a brief statement Vancomycin 1 g q48h (check 1 g q24h (check
is made about normal gas exchange. Monitoring ventilation levels) levels)
and pulmonary function is discussed in Chapter 20. Voriconazole 4 mg/kg PO q12h Same

CRRT, Continuous renal replacement therapy; CVVH, continuous veno-venous


Normal Gas Exchange hemofiltration; CVVHD, continuous veno-venous hemodialysis.

Respiration is a complex process involving integration between


the pons, medulla, spinal cord, carotid body, and muscles. primary stimulus for respiration. Delivering high concentra-
Increases in PaCO2 are sensed in the medulla, causing stimula- tions of oxygen to these patients may suppress the hypoxic
tion of respiration. Hypoxemia is detected in the carotid body respiratory drive. The alveolar-arterial gradient (A-a gradient)
(located at the bifurcation of the internal and external carotid is important in evaluating disorders of respiration (Table
artery). In chronic respiratory acidosis the pH is nearly 22.9). The A-a gradient tells us that the presence of carbon
normal, despite a high PaCO2, and the medulla is no longer dioxide (CO2) in the alveoli reduces the alveolar oxygen
sensitive to elevations in PaCO2. This makes hypoxemia the content. Traditionally, when a clinician is confronted with
Section II—Clinical and Laboratory Assessment 221

Table 22.8  An Approach to Acid-Base Disorders (59)


STAGE 1: IDENTIFY THE PRIMARY ACID-BASE DISORDER
Rule 1
Examine the pHa and PaCO2. If either is abnormal, an acid-base abnormality is present.
Rule 2
If they are both abnormal, a primary disorder is present. If they are abnormal in the same direction, the primary disorder is
metabolic. If they are abnormal in opposite directions, the primary disorder is respiratory.
Example: 7.23/23 Both are decreased, so this is a primary metabolic acidosis.
Rule 3
If only one is abnormal, a mixed disorder is present.
Example: 7.37/55 Because the PaCO2 is elevated and there is a normal pH, a mixed disorder is present: a respiratory acidosis with a
metabolic alkalosis. Compensatory processes do not normalize the pH.
STAGE 2: EVALUATE THE COMPENSATORY RESPONSE
If only one of either the PaCO2 or pH is abnormal, a mixed disorder is present; skip to stage 3 for further analysis. When the pH
and PaCO2 are both abnormal and a primary disorder is present, evaluate the degree of compensation to determine if another
process is present.
Rule 4
If the pH and PaCO2 change in the same direction, a metabolic process is present, and the HCO3 is used to determine the
appropriate PaCO2.
Compensation for metabolic acidosis:  PaCO2 = (1.5 × HCO3) + 8 ± 2
Compensation for metabolic alkalosis  PaCO2 = (0.7 × HCO3) + 20 ± 5
If the PaCO2 is greater than expected, a concomitant respiratory acidosis is present, whereas if it is lower than predicted, a
respiratory alkalosis exists.
Example: pH = 7.23, PaCO2 = 23, HCO3 = 15 mEq/L
When the pH and PaCO2 are both abnormal in the same lower direction, this indicates a primary metabolic acidosis. The expected
PaCO2 would be 30.5 ± 2. Because the PaCO2 is lower than this expected value, a superimposed respiratory alkalosis is present.
Rule 5
If the pH and PaCO2 change in opposite directions, a respiratory disorder is present. Using the history and the equations for the
expected pH and HCO3, determine if the condition is acute, partially compensated, or fully compensated.
Example: pH = 7.54, PaCO2 = 23
If the history suggests an acute process, the equation for the expected change in pH is:
Expected pH = 7.40 + (0.008 × [ΔPaCO2])
The calculated value, 7.54, is the same as the measured value, so this represents an acute respiratory alkalosis. If the pH were
higher, a metabolic alkalosis would also be present. If the pH were lower, a metabolic acidosis would be present.
STAGE 3: CHECK THE ANION GAP AND GAP–GAP TO EVALUATE METABOLIC ACIDOSIS
An anion gap (AG) acidosis may be accompanied by a normal AG acidosis or a metabolic alkalosis.
This is called the gap–gap and is detected by examining the ratio:
AG excess/HCO3 deficit = (AG-12)/(24-HCO3)
If the increase in the AG due to added H+ is balanced by the decrease in HCO3, the ratio is 1, and only an AG acidosis exists.
However, if a hyperchloremic acidosis also is present, the decrease in HCO3 will be greater than the increase in the AG, and the
ratio will be <1. Alternatively, if a metabolic alkalosis is present, as may occur with nasogastric suctioning, the decrease in HCO3 is
less than the increase in AG, and the ratio is >1.
Interpretation of gap–gap:
<1: Additional non-AG acidosis
1-2: Typical pattern in high anion gap acidosis
>2: Additional metabolic alkalosis, or perhaps a coexisting chronic respiratory acidosis, which has resulted in a compensatory
elevation of HCO3

the application of 100% oxygen). The A-a gradient increases


Table 22.9  Alveolar Gas Equation with age according to the following formula:
PAO2 = PiO2 − (PaCO2/R), where PiO2 = FiO2 (PB − PH2O)
or using common values: Normal A-a gradient = (Age + 10) / 4.
PAO2 = (FiO2 × [760 − 47]) − (PaCO2/0.8) = 150 − PaCO2/0.8

Abbreviations: PiO2 = partial pressure of O2 in the central airways;


Respiratory Acid-Base Disorders
FiO2 (fraction of inspired oxygen); FiO2 on room air = 0.21, PaCO2 (value from
your arterial blood gas [ABG]); PB = barometric pressure (760 mm Hg at sea Acute Respiratory Acidosis
level, PB = PN2 + PO2 + PaCO2 + PH2O); PH2O = water vapor pressure (47 mm Hg at
37° C); R = respiratory quotient (ratio of carbon dioxide production to oxygen
Respiratory acidosis normally is buffered by an increase in
consumption) = VCO2/VO2 = 0.8 (usual). HCO3−. In an acute respiratory acidosis, the HCO3 will increase
by 1 mEq/L for each 10 mm Hg elevation in PaCO2 greater
than 40 mm Hg. The expected HCO3 is described by the fol-
hypoxemia, the A-a gradient is calculated. If it is normal, the lowing equation:
low arterial oxygen tension (PaO2) is caused by hypoventila-
HCO3 = 24 + ([ΔPaCO2]/10)
tion. If it is increased, it may be the result of ventilation-
perfusion (V/Q) mismatching (PaO2 increases with application If the PaCO2 increases from 40 to 60 mm Hg, the HCO3
of 100% oxygen) or of shunting (PaO2 does not increase with therefore will increase from 24 to 26 mm Hg. An increase
222 Section II—Clinical and Laboratory Assessment

greater than this suggests the concomitant presence of a meta- Cerebral blood flow can be partially regulated by CO2-
bolic alkalosis. The expected change in pH during this process induced changes in the pH. An alkalosis causes cerebral vaso-
is expressed by the following equation: constriction and there may be a 40% decrease in cerebral
blood flow with a decrease in the PaCO2 from 40 to 20 mm Hg.
Expected pH = 7.40 − (0.008 × [ ∆PaCO2 ])
This technique to decrease intracranial pressure (ICP) is mod-
Hence if the PaCO2 increases acutely from 40 to 60 mm Hg, ulated by pH and not the PaCO2.
the pH will decrease to 7.24 (7.40 − [0.008 × 20]).

Chronic Respiratory Acidosis Metabolic Acid-Base Disorders


In a chronic respiratory acidosis that develops over 2 to 3 days, The physiology underlying acid-base disorders is discussed
the HCO3− will increase by 4 mEq/L for every 10 mm Hg below but the equations for compensation are summarized
change in PaCO2 described by the following equation: first. In metabolic disorders, compensation results in an ele-
vated PaCO2 for a metabolic alkalosis and a lower PaCO2 in a
Expected HCO3− = 24 + 4([ΔPaCO2]/10)
metabolic acidosis. The expected changes can be calculated by
If the PaCO2 increases from 40 to 60 mm Hg, the HCO3− the following equations:
will increase from 24 to 32 mm Hg. The pH is similarly Metabolic alkalosis:
affected, changing 0.003 unit for each 1 mm Hg change in Expected PaCO2 = (0.7 × HCO3) + 20 ± 5
PaCO2 according to the following equation: Metabolic acidosis:
Expected PaCO2 = (1.5 × HCO3) + 8 ± 2
Expected pH = 7.40 − (0.003 × [ΔPaCO2])
In a patient who has chronic obstructive pulmonary disease
(COPD) with a PaCO2 that equals 60 mm Hg, the expected Metabolic Alkalosis
pH is 7.34. Metabolic alkalosis can result from nasogastric (NG) suction-
In addition to the elevated HCO3−, there may also be a lower ing or diuretics from the loss of chloride. For every molecule
chloride level. As new HCO3− is generated in the collecting of Cl− that is excreted, one molecule of HCO3− is reabsorbed.
duct from hydrolysis, hydrogen is secreted into the tubular Though there is a loss of both H+ and Cl−, it is chloride deple-
lumen. Na+ flows in to maintain electrical neutrality. This tion, stimulating HCO3− reabsorption, which is the major con-
influx leaves less Na+ available to be resorbed with Cl− and so tributing factor to the alkalosis. Thiazide and loop diuretics
Cl− excretion is increased. promote natriuresis and Cl− is excreted with the sodium.
Additionally, volume depletion promotes metabolic alkalosis
in two ways. The increase in Na+ resorption is accompanied
Acute Respiratory Alkalosis by an increase in bicarbonate reabsorption. Volume depletion
Respiratory alkalosis is buffered by a decrease in HCO3−. In an also stimulates renin, promoting the formation of aldosterone,
acute respiratory alkalosis the HCO3− decreases by 2 mEq/L which causes H+ excretion in the distal tubules. Volume resus-
for each 10 mm Hg decrease in PaCO2 less than 40 mm Hg citation is not effective in reversing the alkalosis unless Cl− is
according to the following equation: also replaced. Hypokalemia is associated with alkalosis and a
shift of H+ into the cells and an increase in H+ secretion in the
Expected HCO3− = 24 − 2(ΔPaCO2/10)
distal tubules. Aldosterone and other mineralocorticoids act
If the PaCO2 decreases from 40 to 20 mm Hg, the HCO3 on the collecting tubule to stimulate the resorption of Na+,
will decrease from 24 to 20 mm Hg. In practice, a HCO3− of accompanied by the excretion of H+ and K+, which gives rise
less than 18 mEq/L indicates a coexisting metabolic acidosis. to hypokalemia, hypertension, and hypernatremia. Metabolic
The expected change in pH during this process is expressed alkalosis also may result from the administration of exogenous
by the following equation: HCO3 such as HCO3 infusions, from acetate in TPN, or from
the administration of antacids in renal failure. Citrate in
Expected pH = 7.40 + (0.008 × [ΔPaCO2])
banked blood may produce a metabolic alkalosis, but a
If the PaCO2 acutely decreases from 40 to 15 mm Hg, the minimum of 8 units must be transfused.
pH will rise to 7.56. Clinically, metabolic alkalosis can be divided into two
categories: (1) saline-responsive, associated with volume de-
pletion; and (2) saline-resistant, associated with excess miner-
Chronic Respiratory Alkalosis alocorticoid activity. In saline-responsive metabolic alkalosis,
In chronic respiratory alkalosis the HCO3− will decrease by volume depletion stimulates the resorption of Na+ that also
5  mEq/L for every 10  mm  Hg decrease in PaCO2 Described causes the retention of Cl− and HCO3−. However, some extra
by the equation: HCO3− is excreted with Na+ to maintain electrical neutrality,
so a urinary Cl− less than 20 mEq/L is a better indicator of
Expected HCO3− = 24 − 5(ΔPaCO2/10)
volume depletion in metabolic alkalosis.
Thus if the PaCO2 decreases from 40 to 20 mmHg, the HCO3−
will decrease from 24 to approximately 14 mm Hg. The HCO3
generally does not fall below 12 for compensation. The change Anion Gap
in pH can be expressed by: A normal anion-gap acidosis occurs because the lost HCO3− is
replaced by a Cl−. Causes for this are listed in Table 22.10. The
Expected pH = 7.40 + (0.003 × [ΔPaCO2]) anion gap (AG) is defined as:
Section II—Clinical and Laboratory Assessment 223

absorb dietary sugar, leaving carbohydrates to be metabolized


Table 22.10  Causes for Normal by bacteria in the colon. Lactobacilli metabolism of carbohy-
Anion-Gap Acidosis drate produces D lactic acid. It presents with neurologic
• Gastrointestinal loss—secretions from below the stomach
changes (confusion, ataxia, slurred speech, memory loss) sim-
are rich in bicarbonate because they neutralize the acid ulating intoxication without alcohol ingestion after a high-
from the stomach. carbohydrate meal.
• Renal loss of HCO3−—proximal RTA
• Early renal failure of a distal RTA
• Ingestions—HCO3−—acetate in TPN Ketoacidosis
• Carbonic anhydrase inhibitors—acetazolamide
• Recovery of ketoacidosis when given normal saline Glucose and ketones are the two fuels available for the body.
In the fed state, glucose is the primary fuel source; ketones
RTA, Renal tubular acidosis; TPN, total parenteral nutrition.
assume this role in the fasting state. Ketones are produced
in the liver from triglycerides, and there are three types:
(1) B-hydroxybutyrate, the predominant ketone of diabetic
ketoacidosis (DKA) and alcoholic ketoacidosis (AKA), but not
Table 22.11  Causes of an Anion-
measured by the standard urine assay; (2) acetoacetate, mea-
Gap Acidosis
sured by the lab, but accounts for only 25% or less of the total
Propylene glycol Ethanol ketone load; and (3) acetone, which does not cause an acidosis.
Lactic acidosis Ethylene glycol Ketoacidosis becomes clinically significant when insulin activ-
Uremia Diabetic ketoacidosis (DKA) ity is reduced in the following conditions: (1) DKA, the body
Methanol Starvation
Salicylate Isoniazid
is unable to produce or release insulin; (2) AKA, alcohol
induces lipolysis (increases the supply of fatty acids for ketone
production) and consumes nicotinamide adenine dinucleo-
tide (NAD+) (decreasing gluconeogenesis, forcing the liver
AG = Na+ − (Cl− + HCO3−)
to produce ketones); and (3) starvation-insulin release is
An anion gap exists because there are anions such as sulfate, suppressed.
phosphate, organic anions, and proteins that are not measured In AKA and starvation ketoacidosis, the treatment is glucose,
on the routine chemistry panel, where there are very few which stimulates endogenous insulin production to suppress
unmeasured cations. The normal AG is between 7 and ketogenesis. In DKA are insulin deficiency, hyperglycemia,
13 mEq/L, but newer autoanalyzers report a higher Cl−, so the ketosis (causing AG), and hyponatremia (often pseudohypo-
normal may be between 3 and 11 mEq/L. Hypoalbuminemia natremia), and treatment with insulin and fluids is needed.
may spuriously lower the AG and mask an underlying anion- Hyperglycemic hyperosmolar nonketotic (HHNK) coma is
gap acidosis. Causes of an anion-gap acidosis are listed in seen in people with type 2 diabetes and is characterized
Table 22.11 and each is briefly described. by extremely high blood sugar (≈1000 mg/dL) but without
ketosis (or acidosis) because insulin is present. The primary
treatment is volume replacement. Uremia causes an anion gap
Propylene Glycol acidosis from an impaired ability to clear the daily acid load
Propylene glycol is an alcohol used to increase the water solu- of daily metabolism, but the GFR is usually less than 40 mL/
bility of lorazepam, diazepam, esmolol, nitroglycerin, and min before this occurs.
phenytoin, and it is metabolized in the liver to lactate and
pyruvate. Signs of toxicity are agitation, seizures, coma, tachy-
cardia, and hypotension. The lactate may exceed 10 mEq/L Alcohols
and clinically mimic sepsis. The metabolites of alcohols, generated by alcohol and alde-
hyde dehydrogenase are responsible for the toxicity associated
with alcohol ingestion. Thus it can take 12 to 24 hours for
Lactic Acidosis acidosis and organ failure to occur, and this may be delayed
A normal AG may accompany lactic acidosis. Iberti et al. further when there also is ethanol ingestion. Consequently,
observed that 50% of patients with a lactate level between 5 management is to prevent metabolite formation until the
and 9.9 mmol/L had AG less than 12 mEq/L.84 Lactic acidosis parent compound can be excreted or dialyzed. Methanol and
results from mitochondrial dysfunction and has three forms. ethylene glycol intoxication present with a high anion gap
(1) Type A arises from tissue hypoxia—septic, cardiogenic, or acidosis, but the measurements of these may be off-site and
hemorrhagic shock, or respiratory failure. (2) Type B is caused will not be available to assist immediate clinical decision
by impaired mitochondrial oxygen utilization, such as from making. The osmolal gap defined as the difference between
toxins; that is, there is no systemic hypoperfusion. These the calculated osmolality (osmolality = 2 × Na+[mmol/L] +
toxins can include: metformin (Glucophage); cyanide poison- BUN [mg/dL]/2.8 + [glucose mg/dL]/18) and the measured
ing, for example, a nitroprusside infusion; thiamine (B1) defi- osmolality may help clinical decisions. The normal osmolal
ciency that can be associated with loss of body stores, increased gap is 10 mOsm/kg H2O or less. Causes of an elevated osmolal
excretion by furosemide (Lasix), or magnesium depletion that gap are listed in Table 22.12.
results in a functional form of thiamine deficiency; malignan- As one integrates the history and laboratory findings, it is
cies, alcoholism, or AZT (zidovudine). (3) Type D occurs in important to remember that the alcohols contribute to the
patients with jejunoileal bypass or in patients with short osmolal gap and the toxic metabolites contribute to the
bowel syndrome. The shortened small intestine is unable to acidosis. Thus toxicity is not excluded on a gap less than
224 Section II—Clinical and Laboratory Assessment

10 mOsm/kg. A small elevation isn’t necessarily from a toxic


Table 22.12  Causes of an Elevated Osmolal metabolite, but a gap greater than 25 is presumptive evidence
Gap with an Anion-Gap Acidosis in the appropriate setting. 100 mg/dL of ethanol (MW = 46)
Ethylene glycol Elevated osmolal gap without acidosis
adds 22 mOsm/kg. Isopropyl alcohol does not cause an acido-
Methanol Ethanol sis, but its primary metabolite, acetone, manifests by positive
Formaldehyde Isopropanol serum and urinary ketones.
ESRD without dialysis Diethyl ether
Paraldehyde Mannitol
DKA Severe hyperproteinemia Methanol
AKA Severe hyperlipidemia
Lactic acidosis Methanol poisoning occurs with the ingestion of shellac,
varnish, wiper fluids and deicing solutions, or denatured
AKA, Alcoholic ketoacidosis; DKA, diabetic ketoacidosis; ESRD, end-stage
renal disease. gelled alcohol (Sterno). It is converted by alcohol dehydroge-
nase into the toxic formaldehyde and then formic acid.

A B

C D

Fig. 22.13  Brain MRI obtained on day 15 after methanol intoxication. A, T2-weighted image showed high signal abnormalities in bilateral basal
ganglia (arrows), frontal, and occipital subcortical white matter (arrowheads), consistent with edematous change. B, T2-weighted image showed
edematous change involving bilateral optic tracts and optic radiations (arrows). High signal edematous change was also noted in the optic disc of
the left eye (arrowheads). C, T1-weighted image showed slightly high signal component in bilateral basal ganglia, indicating the hemorrhage (arrows).
D, T1-weighted image with gadolinium administration showed marginal enhancement in bilateral putamen, indicating breakdown of the blood-brain
barrier.
Section II—Clinical and Laboratory Assessment 225

Malaise, nausea, and headache occur within 12 to 24 hours of 3. Bellomo R. Acute renal failure. Semin Respir Crit Care Med 2011;32(5):639–
50. Epub 2011, Oct 11.
ingestion, followed by acidosis, visual disturbances (formalde-
4. Mandelbaum T, Scott DJ, Lee J, et al. Outcome of critically ill patients with
hyde’s retinal toxicity), seizures, and coma. Serious toxicity acute kidney injury using the Acute Kidney Injury Network criteria. Crit
occurs with one teaspoon—50 mL may be lethal. Optic nerve Care Med 2011;39(12):2659–64.
edema and basal ganglia infarcts can be observed (Fig. 22.13). 5. Li N, Zhao WG, Zhang WF. Acute kidney injury in patients with severe
traumatic brain injury: implementation of the acute kidney injury network
stage system. Neurocrit Care 2011;14(3):377–81.
Ethylene Glycol 6. Rose BD, Post TW. Introduction to renal function. Available at www.upto
date.com (accessed August 2009).
Ethylene glycol is found in antifreeze and solvents, and inges- 7. Rose BD, Post TW. Collecting tubules. Available at www.uptodate.com
tion manifests initially by neurologic symptoms, including (accessed August 2009).
8. Rose BD. Urine osmolality versus specific gravity. Available at www.uptodate
cranial nerve palsies and tetany (from oxalate-induced hypo-
.com (accessed August 2009).
calcemia) and later by pulmonary edema. Renal failure is a late 9. Pasticci F, Fantuzzi AL, Pegoraro M, et al. Nutritional management of stage 5
finding due to the effects of oxalate crystals. Lactic acidosis chronic kidney disease. J Ren Care 2012;38(1):50–8.
can be seen with ethylene glycol, but it is insufficient to 10. Patel P, Nandwani V, McCarthy PJ, et al. Continuous renal replacement
account for the degree of acidosis. therapies: a brief primer for the neurointensivist. Neurocrit Care 2010;13(2):
286–94.
11. Arulkumaran N, Montero RM, Singer M. Management of the dialysis patient
in general intensive care. Br J Anaesth 2012;108(2):183–92. Epub 2012, Jan 4.
Treatment of Alcohol or Methanol Toxicity 12. Basu RK, Wheeler DS, Goldstein S, et al. Acute renal replacement therapy in
For each of these disorders time is crucial, and important pediatrics. Int J Nephrol 2011;2011:785392. Epub 2011, Jun 1.
13. Chuasuwan A, Kellum JA. Acute kidney injury and its management. Contrib
decisions may need to be made without all the data. Therapy
Nephrol 2011;171:218–25. Epub 2011, May 23.
includes sodium bicarbonate and fomepizole. Fomepizole 14. Claure-Del Granado R, Mehta RL. Withholding and withdrawing renal
(or ethanol) inhibits alcohol dehydrogenase and blocks support in acute kidney injury. Semin Dial 2011;24(2):208–14. doi:10.1111/
metabolite production, but early treatment is crucial because j.1525-139X.2011.00832.x.
it does not prevent toxicity if metabolism to an acid species 15. Srisawat N, Kellum JA. Acute kidney injury: definition, epidemiology, and
outcome. Curr Opin Crit Care 2011;17(6):548–55.
has already occurred. It is useful for both methanol and eth- 16. Singbartl K, Kellum JA. AKI in the ICU: definition, epidemiology, risk
ylene glycol poisoning. Hemodialysis should be used if meta- stratification, and outcomes. Kidney Int 2011;Oct 5. Epub ahead of print.
bolic acidosis is present or there is evidence of end-organ 17. Liano F, Pascual J. Madrid Acute Renal Failure Study Group. Epidemiology
damage. All methanol intoxications should also receive cofac- of acute renal failure: a prospective, multicenter, community-based study.
Kidney Int 1996;50:811.
tor therapy: either folinic acid (50 mg IV) or folic acid (50 mg
18. Mehta RL, Pascual MT, Soroko S, et al. Spectrum of acute renal failure in the
every 6 hours IV) and probably thiamine (100 mg IV). intensive care unit: the 1 PICARD experience. Kidney Int 2004;66:1613.
19. Muther RS. Acute renal failure in the critically ill. In: ACCP, editor. ACCP
Critical Care Board Review 2007: Course Syllabus. Northbrook, Ill: American
Salicylate College of Chest Physicians; 2007.
20. Bellomo R, Ronco C, Kellum JA, et al. Acute renal failure-definition,
Aspirin overdose is characterized by the accumulation of sal- outcome measures, animal models, fluid therapy and information
icylic acid, the active metabolite of acetylsalicylic acid toxic- technology needs: the Second International Consensus Conference of the
ity is characterized by two primary acid-base disorders. Acute Dialysis Quality Initiative ADQI. Group Crit Care 2004;8:R204–12.
Respiratory alkalosis initially ensues from direct CNS stimu- 21. Chertow GM, Burick E, Honour M, et al. Acute kidney injury, mortality,
length of stay, and costs in hospitalized patients. J Am Soc Nephrol 2005;
lation and is followed by a metabolic acidosis. Salicylic acid
16:3365–70.
itself contributes little to the anion gap (due to its high 22. Palevsky PM. Definition of acute kidney injury (acute renal failure).
molecular weight), but the anion gap results from the Available at www.uptodate.com (accessed August 2009).
buildup of lactic acid and ketones by unclear mechanisms. 23. Waring WS, Moonie A. Earlier recognition of nephrotoxicity using novel
The treatment is alkalinization of the urine. Increasing the biomarkers of acute kidney injury. Clin Toxicol (Phila) 2011;49(8):
720–8.
pH from 7.2 to 7.5 halves the tissue level of salicylic acid. 24. Payen D, Legrand M. Can we identify prerenal physiology and does it
Dialysis is indicated for patients in coma or with levels matter? Contrib Nephrol 2011;174:22–32. Epub 2011, Sep 9.
80 mg/dL or greater. 25. Srisawat N, Wen X, Lee M, et al. Urinary biomarkers and renal recovery in
critically ill patients with renal support. Clin J Am Soc Nephrol
2011;6(8):1815–23. Epub 2011, Jul 14.
Conclusion 26. Doi K, Negishi K, Ishizu T, et al. Evaluation of new acute kidney injury
biomarkers in a mixed intensive care unit. Crit Care Med 2011;39(11):
Electrolyte and acid-base abnormalities and acute and chronic 2464–9.
kidney injury are ubiquitous in neurocritical care. An under- 27. Panagiotou A, Garzotto F, Gramaticopolo S, et al. Continuous real-time
standing of basic renal physiology assists in providing a frame- urine output monitoring for early detection of acute kidney injury. Contrib
Nephrol 2011;171:194–200. Epub 2011, May 23.
work for understanding these disorders and a context for their 28. Bellomo R. Defining, quantifying, and classifying acute renal failure. Crit
management. Care Clin 2005;21:223–7.
29. Muther RS. Drug interference with renal function tests. Am J Kidney Dis
1983;3:118–20.
References 30. Kaplan AA, Kohn OF. Fractional excretion of urea as a guide to renal
dysfunction. Am J Nephrol 1992:12:54–9.
1. Piccinni P, Cruz DN, Gramaticopolo S, et al. Prospective multicenter study 31. Post TW, Rose BD. Urinalysis in the diagnosis of renal disease. Available at
on epidemiology of acute kidney injury in the ICU: a critical care www.uptodate.com (accessed August 2009).
nephrology Italian collaborative effort (NEFROINT). Minerva Anestesiol 32. Bohle A, Christensen J, Kokot F, et al. Acute renal failure in man: new aspects
2011;77(11):1072–83. Epub 2011, May 11. concerning pathogenesis. A morphometric study. Am J Nephrol 1990;10:
2. Clec’h C, Gonzalez F, Lautrette A, et al. Multiple-center evaluation of 374–88.
mortality associated with acute kidney injury in critically ill patients: a 33. Lameire N, Vanholder R, Van Biesen W. Loop diuretics for patients with
competing risks analysis. Crit Care 2011;15(3):R128. Epub 2011, May 17. acute renal failure: helpful or harmful. JAMA 2002;288:2599.
226 Section II—Clinical and Laboratory Assessment

34. Cantarovich F, Rangoonwala B, Lorenz H, et al. High-dose furosemide for 43. Iwatsuke S, Popovtzer MM, Corman JI, et al. Recovery from hepatorenal
established ARF: a prospective, randomized, double-blind, placebo- syndrome after orthotopic liver transplantation. N Engl J Med 1973;289:
controlled, multicenter trial. Am J Kidney Dis 2004;44:402. 1155–9.
35. Delpire E, Lu J, England R, et al. Deafness and imbalance associated with 44. Arroyo V, Gines P, Gerbes AL, et al. Definition and diagnostic criteria of
inactivation of the secretory Na-K-2Cl co-transporter. Nat Genet 1999; refractory ascites and hepatorenal syndrome in cirrhosis. Int Ascites Club
22:192. Hepatol 1996;23:164.
36. Marik PE, Iglesias J. Low-dose dopamine does not prevent acute renal failure 45. Watt K, Uhanova J, Minuk GY. Hepatorenal syndrome: diagnostic accuracy,
in patients with septic shock and oliguria. NORASEPT II Study Investigators. clinical features, and outcome in a tertiary care centre. Am J Gastroenterol
Am J Med 1999;107:387–90. 2002;97:2046–50.
37. Bellomo R, Chapman M, Finfer S, et al. Low-dose dopamine in patients 46. Esrailian E, Pantangco ER, Kyulo NL, et al. Octreotide/Midodrine therapy
with early renal dysfunction: a placebo-controlled randomized trial. Lancet significantly improves renal function and 30-day survival in patients with
2000;356:2139. type 1 hepatorenal syndrome: a large retrospective series. Dig Dis Sci
38. Lauschke A, Teichgraber UK, Frei U, et al. Low-dose dopamine worsens 2007;52:742.
renal perfusion in patients with acute renal failure. Kidney Int 2006;69: 47. Uriz J, Gines P, Cardenas A, et al. Terlipressin plus albumin infusion: an
1669. effective and safe therapy of hepatorenal syndrome. J Hepatol 2000;22:43–8.
39. Reid GM, Muther RS. Nitroprusside-induced acute azotemia. Am J Nephrol 48. Wong F, Pantea L, Sniderman K. Midodrine, octreotide, albumin, and TIPS
1987;7:313–15. in selected patients with cirrhosis and type 1 hepatorenal syndrome.
40. Rose B, Appel GB. Clinical manifestations and diagnosis of acute interstitial Hepatology 2004;40:55–64.
nephritis. Available at www.uptodate.com (accessed August 2009). 49. Olson JC, Wendon JA, Kramer DJ, et al. Intensive care of the patient with
41. Hecker R, Sherlock S. Electrolyte and circulatory changes in terminal liver cirrhosis. Hepatology 2011;54(5):1864–72. doi:10.1002/hep.24622.
failure. Lancet 1956;2:1221–5. 50. McCarthy JT. Prognosis of patients with acute renal failure in the intensive
42. Koppel MH, Coburn JN, Mims MM, et al. Transplantation of cadaveric care unit: a tale of two eras. Mayo Clin Proc 1996;7:117–26.
kidneys from patients with hepatorenal syndrome. Evidence for the
functional nature of renal failure in advanced liver disease. N Engl J Med A complete list of references for this chapter can be found online at
1969;280:1267–71. www.expertconsult.com.
Section II—Clinical and Laboratory Assessment 226.e1

References 31. Post TW, Rose BD. Urinalysis in the diagnosis of renal disease. Available at
www.uptodate.com (accessed August 2009).
1. Piccinni P, Cruz DN, Gramaticopolo S, et al. Prospective multicenter study 32. Bohle A, Christensen J, Kokot F, et al. Acute renal failure in man: new aspects
on epidemiology of acute kidney injury in the ICU: a critical care concerning pathogenesis. A morphometric study. Am J Nephrol 1990;10:
nephrology Italian collaborative effort (NEFROINT). Minerva Anestesiol 374–88.
2011;77(11):1072–83. Epub 2011, May 11. 33. Lameire N, Vanholder R, Van Biesen W. Loop diuretics for patients with
2. Clec’h C, Gonzalez F, Lautrette A, et al. Multiple-center evaluation of acute renal failure: helpful or harmful. JAMA 2002;288:2599.
mortality associated with acute kidney injury in critically ill patients: a 34. Cantarovich F, Rangoonwala B, Lorenz H, et al. High-dose furosemide for
competing risks analysis. Crit Care 2011;15(3):R128. Epub 2011, May 17. established ARF: a prospective, randomized, double-blind, placebo-
3. Bellomo R. Acute renal failure. Semin Respir Crit Care Med 2011;32(5):639– controlled, multicenter trial. Am J Kidney Dis 2004;44:402.
50. Epub 2011, Oct 11. 35. Delpire E, Lu J, England R et al. Deafness and imbalance associated with
4. Mandelbaum T, Scott DJ, Lee J, et al. Outcome of critically ill patients with inactivation of the secretory Na-K-2Cl co-transporter. Nat Genet 1999;
acute kidney injury using the Acute Kidney Injury Network criteria. Crit 22:192.
Care Med 2011;39(12):2659–64. 36. Marik PE, Iglesias J. Low-dose dopamine does not prevent acute renal failure
5. Li N, Zhao WG, Zhang WF. Acute kidney injury in patients with severe in patients with septic shock and oliguria. NORASEPT II Study Investigators.
traumatic brain injury: implementation of the acute kidney injury network Am J Med 1999;107:387–90.
stage system. Neurocrit Care 2011;14(3):377–81. 37. Bellomo R, Chapman M, Finfer S, et al. Low-dose dopamine in patients with
6. Rose BD, Post TW. Introduction to renal function. Available at early renal dysfunction: a placebo-controlled randomized trial. Lancet
www.uptodate.com (accessed August 2009). 2000;356:2139.
7. Rose BD, Post TW. Collecting tubules. Available at www.uptodate.com 38. Lauschke A, Teichgraber UK, Frei U, et al. Low-dose dopamine worsens renal
(accessed August 2009). perfusion in patients with acute renal failure. Kidney Int 2006;69:1669.
8. Rose BD. Urine osmolality versus specific gravity. Available at www.uptodate. 39. Reid GM, Muther RS. Nitroprusside-induced acute azotemia. Am J Nephrol
com (accessed August 2009). 1987;7:313–15.
9. Pasticci F, Fantuzzi AL, Pegoraro M, et al. Nutritional management of stage 5 40. Rose B, Appel GB. Clinical manifestations and diagnosis of acute interstitial
chronic kidney disease. J Ren Care 2012;38(1):50–8. nephritis. Available at www.uptodate.com (accessed August 2009).
10. Patel P, Nandwani V, McCarthy PJ, et al. Continuous renal replacement 41. Hecker R, Sherlock S. Electrolyte and circulatory changes in terminal liver
therapies: a brief primer for the neurointensivist. Neurocrit Care failure. Lancet 1956;2:1221–5.
2010;13(2):286–94. 42. Koppel MH, Coburn JN, Mims MM, et al. Transplantation of cadaveric
11. Arulkumaran N, Montero RM, Singer M. Management of the dialysis patient kidneys from patients with hepatorenal syndrome. Evidence for the
in general intensive care. Br J Anaesth 2012;108(2):183–92. Epub 2012, Jan 4. functional nature of renal failure in advanced liver disease. N Engl J Med
12. Basu RK, Wheeler DS, Goldstein S, et al. Acute renal replacement therapy in 1969;280:1267–71.
pediatrics. Int J Nephrol 2011;2011:785392. Epub 2011, Jun 1. 43. Iwatsuke S, Popovtzer MM, Corman JI, et al. Recovery from hepatorenal
13. Chuasuwan A, Kellum JA. Acute kidney injury and its management. Contrib syndrome after orthotopic liver transplantation. N Engl J Med 1973;289:
Nephrol 2011;171:218–25. Epub 2011, May 23. 1155–9.
14. Claure-Del Granado R, Mehta RL. Withholding and withdrawing renal 44. Arroyo V, Gines P, Gerbes AL, et al. Definition and diagnostic criteria of
support in acute kidney injury. Semin Dial 2011;24(2):208–14. doi:10.1111/j refractory ascites and hepatorenal syndrome in cirrhosis. Int Ascites Club
.1525-139X.2011.00832.x. Hepatol 1996;23:164.
15. Srisawat N, Kellum JA. Acute kidney injury: definition, epidemiology, and 45. Watt K, Uhanova J, Minuk GY. Hepatorenal syndrome: diagnostic accuracy,
outcome. Curr Opin Crit Care 2011;17(6):548–55. clinical features, and outcome in a tertiary care centre. Am J Gastroenterol
16. Singbartl K, Kellum JA. AKI in the ICU: definition, epidemiology, risk 2002;97:2046–50.
stratification, and outcomes. Kidney Int 2011;Oct 5. Epub ahead of print. 46. Esrailian E, Pantangco ER, Kyulo NL, et al. Octreotide/midodrine therapy
17. Liano F, Pascual J, Madrid Acute Renal Failure Study Group. Epidemiology significantly improves renal function and 30-day survival in patients with
of acute renal failure: a prospective, multicenter, community-based study. type 1 hepatorenal syndrome: a large retrospective series. Dig Dis Sci
Kidney Int 1996;50:811. 2007;52:742.
18. Mehta RL, Pascual MT, Soroko S, et al. Spectrum of acute renal failure in the 47. Uriz J, Gines P, Cardenas A, et al. Terlipressin plus albumin infusion:
intensive care unit: the 1 PICARD experience. Kidney Int 2004;66:1613. an effective and safe therapy of hepatorenal syndrome. J Hepatol 2000;22:
19. Muther RS. Acute renal failure in the critically ill. In: ACCP, editor. ACCP 43–8.
Critical Care Board Review 2007: Course Syllabus. Northbrook, Ill: American 48. Wong F, Pantea L, Sniderman K. Midodrine, octreotide, albumin, and TIPS
College of Chest Physicians; 2007. in selected patients with cirrhosis and type 1 hepatorenal syndrome.
20. Bellomo R, Ronco C, Kellum JA, et al. Acute renal failure-definition, Hepatology 2004;40:55–64.
outcome measures, animal models, fluid therapy and information 49. Olson JC, Wendon JA, Kramer DJ, et al. Intensive care of the patient with
technology needs: the Second International Consensus Conference of the cirrhosis. Hepatology 2011;54(5):1864–72. doi:10.1002/hep.24622.
Acute Dialysis Quality Initiative ADQI. Group Crit Care 2004;8:R204–12. 50. McCarthy JT. Prognosis of patients with acute renal failure in the intensive
21. Chertow GM, Burick E, Honour M, et al. Acute kidney injury, mortality, care unit: a tale of two eras. Mayo Clin Proc 1996;7:117–26.
length of stay, and costs in hospitalized patients. J Am Soc Nephrol 2005; 51. Richards WO, Scovill W, Shin B, et al. Acute renal failure associate with
16:3365–70. increased intra-abdominal pressure. Ann Surg 1983;197:183.
22. Palevsky PM. Definition of acute kidney injury (acute renal failure). 52. Mohmand H, Goldfarb S. Renal dysfunction associated with intra-
Available at www.uptodate.com (accessed August 2009). abdominal hypertension and the abdominal compartment syndrome. J Am
23. Waring WS, Moonie A. Earlier recognition of nephrotoxicity using novel Soc Nephrol 2011;22(4):615–21. Epub 2011, Feb 10.
biomarkers of acute kidney injury. Clin Toxicol (Phila) 2011;49(8):720–8. 53. Oates JA, Fitzgerald GA, Branch RA, et al. Clinical implication of
24. Payen D, Legrand M. Can we identify prerenal physiology and does it prostaglandin and thromboxane A2 formation. N Engl J Med 1988;319:761.
matter? Contrib Nephrol 2011;174:22–32. Epub 2011, Sep 9. 54. Huerta C, Castellsague J, Baras-Lorenzo C, et al. Nonsteroidal anti-
25. Srisawat N, Wen X, Lee M, et al. Urinary biomarkers and renal recovery in inflammatory drugs and risk of ARF in the general population. Am J Kidney
critically ill patients with renal support. Clin J Am Soc Nephrol 2011;6(8): Dis 2005;45:531.
1815–23. Epub 2011, Jul 14. 55. Patrono C, Dunn MJ. The clinical significance of inhibition of renal
26. Doi K, Negishi K, Ishizu T, et al. Evaluation of new acute kidney injury prostaglandin synthesis. Kidney Int 1987;32:1.
biomarkers in a mixed intensive care unit. Crit Care Med 2011;39(11):2464–9. 56. Clive DM, Stoff JS. Renal syndromes associated with nonsteroidal
27. Panagiotou A, Garzotto F, Gramaticopolo S, et al. Continuous real-time anti-inflammatory drugs. N Engl J Med 1984;310:563.
urine output monitoring for early detection of acute kidney injury. Contrib 57. Radford MG, Holley KE, Grande JP, et al. Reversible membranous
Nephrol 2011;171:194–200. Epub 2011, May 23. nephropathy associated with the use of nonsteroidal anti-inflammatory
28. Bellomo R. Defining, quantifying, and classifying acute renal failure. Crit drugs. JAMA 1996;276:466.
Care Clin 2005;21:223–7. 58. Kaysen GA, Pond SM, Roper MH, et al. Combined hepatic and renal injury
29. Muther RS. Drug interference with renal function tests. Am J Kidney Dis in alcoholics during therapeutic use of acetaminophen. Arch Intern Med
1983;3:118–20. 1985;145:2019–23.
30. Kaplan AA, Kohn OF. Fractional excretion of urea as a guide to renal 59. Blakely P, McDonald BR. Acute renal failure de to acetaminophen ingestion:
dysfunction. Am J Nephrol 1992:12:54–9. A case report and review of the literature. J Am Soc Nephrol 1995;6:48.
226.e2 Section II—Clinical and Laboratory Assessment

60. Weisbord SD, Palevsky PM. Radiocontrast-induced acute renal failure. 74. August P, George JN. Acute renal failure in pregnancy. www.uptodate.
J Intensive Care Med 2005;20:63. com (accessed August 2009).
61. Lakhal K, Ehrmann S, Chaari A, et al. Acute Kidney Injury Network 75. Poole BD, Schrier RW. Acute renal failure. In Fink MP, Abraham E, et al.
definition of contrast-induced nephropathy in the critically ill: incidence and Textbook of critical care, ed 5. Elsevier, New York, 2005, p. 1139–50.
outcome. J Crit Care 2011;26(6):593–9. Epub 2011, Jul 6. 76. Rose BD. Urine output in urinary tract obstruction and postobstructive
62. Agmon Y, Peleg H, Greenfield A, et al. Nitric oxide and prostanoids protect diuresis. Available at www.uptodate.com (accessed August 2009).
the renal outer medulla from radiocontrast toxicity in the rat. J Clin Invest 77. Negash DT, Dhingra VK, Copland M, et al. Intensity of continuous renal
1994;94:1069. replacement therapy in acute kidney injury in the intensive care unit: a
63. Morcos DK, El-Nahas AM, Broen P, et al. Effect of iodinated water soluble systematic review and meta-analysis. Vasc Endovascular Surg
contrast media on urinary protein assays. BMJ 1992;305:29. 2011;45(6):504–10. Epub 2011, Jun 5.
64. Tepel M, van der Giet M, Schwarzfeld C, et al. Prevention of radiographic- 78. Graham P, Lischer E. Nursing issues in renal replacement therapy:
contrast-agent-induces reductions in renal function by acetylcysteine. N Engl organization, manpower assessment, competency evaluation and quality
J Med 2000;343:180–4. improvement processes. Semin Dial 2011;24(2):183–7. doi:10.1111/j.1525-
65. Boton R, Gaviria M, Batlle DC. Prevalence, pathogenesis, and treatment of 139X.2011.00835.x.
renal dysfunction associated with chronic lithium therapy. Am J Kidney Dis 79. Prowle JR, Schneider A, Bellomo R. Clinical review: optimal dose of
1987;10:329. continuous renal replacement therapy in acute kidney injury. Crit Care
66. Lehmann K, Ritz E. Angiotensin-converting enzyme inhibitors may cause 2011;15(2):207. Epub 2011, Mar 18.
renal dysfunction in patients on long-term lithium treatment. Am J Kidney 80. Lines SW, Cherukuri A, Murdoch SD, et al. The outcomes of critically ill
Dis 1995;25:82. patients with acute kidney injury receiving renal replacement therapy. Int J
67. Kaplan NM, Rose BD. Major side effects of ACI inhibitors. Available at Artif Organs 2011;34(1):2–9.
www.uptodate.com (accessed August 2009). 81. Barri YM, Golper TA, Seizures in patients undergoing hemodialysis.
68. Miskulin D, Gul A, Rudnick MR, et al. Nephrogenic systemic fibrosis/ Available at www.uptodate.com (accessed August 2009).
nephrogenic fibrosing dermopathy in advanced renal failure. Available at 82. Marino PL. The ICU book. 3rd ed. Lippincott Williams & Wilkins,
www.uptodate.com (accessed August 2009). Philadelphia, 2007.
69. Haas M, Ohler L, Watzke H, et al. The spectrum of acute renal failure in 83. Gauthier PM, Szerlip HM. Metabolic acidosis in the intensive care unit.
tumor lysis syndrome. Nephrol Dial Transplant 1999;13:776–9. Critical care clinics. Crit Care Clin 2002;18(2):289–308.
70. Fernandez PC, Larson RA, Agus ZS. Tumor lysis syndrome. Available at 84. Iberti TJ, Leibowitz AB, Papadakos PJ. Low sensitivity of the anion gap as a
www.uptodate.com (accessed August 2009). screen to detect hyperlactatemia in critically ill patients. Crit Care Med
71. Gabow PA, Kaehny WD, Kelleher SP. The spectrum of rhabdomyolysis. 1990;18:275–7.
Medicine (Baltimore) 1982;61:141. 85. Trotman RL, Williamson JC, Shoemaker DM, Salzer WL. Antibiotic dosing
72. Miller ML. Rhabdomyolysis. Available at www.uptodate.com (accessed in critically ill adult patients receiving continuous renal replacement therapy.
August 2009). Clin Infect Dis 2005;41(8):1159–66.
73. Rose BD. Clinical features and prevention of heme pigment-induced acute
tubular necrosis. Available at www.uptodate.com (accessed August 2009).
Chapter
23  
II

Gastrointestinal and
Hepatic Disorders
Christiana E. Hall and Aashish R. Patel

Introduction hemodynamic monitoring techniques.2 These noninvasive


Gastrointestinal (GI) issues and maladies and in particular GI devices can provide data on cardiac performance, preload, and
bleeding, liver dysfunction, and pancreatitis can complicate systemic vascular resistance, similar to that provided by Swan-
the primary course of critically ill neurologic patients. In addi- Ganz catheterization (see Chapter 19).
tion, patients with hepatic encephalopathy may be cared for Laboratory assessment includes hemoglobin and hemato-
in the neurocritical care unit (NCCU). The neurointensivist crit, and to exclude an occult bleeding diathesis platelet count,
therefore must be prepared to recognize, coordinate specialty coagulation indices, and if necessary disseminated intravascu-
care, and provide local monitoring and support when these lar coagulation (DIC). The brain contains high levels of tissue
issues arise. This chapter reviews disorders of the GI system factor (TF), and in severe injury or associated with a pro-
including the liver and pancreas that are of importance in the longed or complex surgery (e.g., for a large arteriovenous
NCCU. A comprehensive review of GI and hepatic function is malformation [AVM]), significant TF release may occur and
beyond the scope of this chapter. Nutrition is reviewed in induce the DIC cascade. The optimal platelet count after an
Chapter 14. injury to or a disease of the nervous system may depend on
pathology and time after the initial insult. There is general
agreement that a platelet count greater than 100,000 is a rea-
Gastrointestinal Bleeding sonable goal after TBI, after neurosurgery, or for anticipated
GI bleeding is relatively common in acutely ill hospitalized intervention or instrumentation of the central nervous system
patients, and the risk may be augmented in the NCCU associ- (CNS). This includes for placement of an intracranial monitor
ated with severe sympathetic stress such as in acute subarach- or external ventricular drain. By contrast for other patients
noid hemorrhage (SAH) or neurotrauma patients, including without CNS disorders or likely surgery, a platelet count
both traumatic brain injury (TBI) and spinal cord injury greater than 50,000 is reasonable perhaps even when there is
(SCI) and in patients in whom steroids are administered. The GI bleeding. For the neurointensivist this means that platelet
bleeding source may be from the upper or lower GI tract. The support may differ depending on whether GI bleeding occurs
ligament of Treitz is the landmark that distinguishes the upper in a patient with a disorder such as myasthenia gravis or severe
from lower GI tract. Common causes for upper GI bleeding TBI or is a postoperative neurosurgical patient. The various
(UGIB) are peptic ulcer disease, including stress-induced ulcer laboratory indices should be repeated at intervals until they
bleeding,1 variceal hemorrhage, and esophagitis. Common are stabilized. This typically includes assessment of complete
causes for lower GI bleeding (LGIB) are diverticulitis, angio- blood count with platelets and if necessary coagulation studies
dysplasia, colitis that can have several causes, tumors, and local every 6 hours.
anorectal pathologies. UGIB presents with hematemesis, GI bleeding requires an assessment of the source of bleed-
melena, hematochezia, and occasionally hypotension when ing and should include an early consultation with a gastroen-
more severe. In patients with a nasogastric tube in place, the terology service. Endoscopy typically is the initial means to
bleeding may manifest in the drainage contents (e.g., coffee- identify the source of UGIB, determine whether active bleed-
ground gastric contents). LGIB presents with hematochezia ing still is present, and guide the frequency of monitoring,
and melena, and can be a reason for anemia. including repeat endoscopy or other diagnostic tests. In addi-
When GI bleeding occurs the initial focus is to ensure tion, the source of bleeding may be treated through endo-
hemodynamic stability with resuscitation using crystalloid or scopic techniques. An intravenous bolus of proton pump
blood products as appropriate. Patient monitoring therefore inhibitors followed by an infusion is recommended to prepare
is aimed at recognition and avoidance of hemorrhagic shock the patient for endoscopy associated with nonvariceal bleed-
using reliable real-time continuous blood pressure and heart ing.3,4 Somatostatin or octreotide can be considered for vari-
rate monitoring. In more severe GI bleeding, resuscitation can ceal bleeding. The Blatchford score, which is based on several
be facilitated using central venous pressure (CVP), mixed factors including urea and hemoglobin levels, systolic blood
venous oxygen saturation (ScvO2), and newer noninvasive pressure, heart rate, hepatic disease, cardiac failure, melena,
© Copyright 2013 Elsevier Inc. All rights reserved. 227
228 Section II—Clinical and Laboratory Assessment

and presentation with syncope, can be used to determine feasible when LGIB is identified,7 but whether urgent colonos-
which patients require urgent endoscopy.5 Once endoscopy is copy is associated with better outcome than a standard care
complete the Rockall score, which includes the endoscopic algorithm based on angiography and expectant colonoscopy
diagnosis and evidence for hemorrhage among other vari- is unclear.8 Current guidelines for evaluation, monitoring, and
ables, can be used to stratify patients into high or low risk for management of UGIB, variceal bleeding, and LGIB are pro-
recurrent hemorrhage or mortality.6 Bedside colonoscopy is vided in Table 23.19,10; Table 23.211; and Figure 23.1.12

Table 23.1  Summary of Recommendations from 2010 Guidelines of the International Consensus
Upper Gastrointestinal Bleeding Conference Group*
Resuscitation, risk assessment, and pre-endoscopy management:
1. Urgent/emergent evaluation and initiation of resuscitation
2. Use appropriate prognostic scales for mortality and rebleeding
to stratify patients as low or high risk.
3. Consider nasogastric tube placement.
4. PRBC transfusion for Hgb 7 g/dL
5. Correct coagulopathy, but do not delay endoscopy.
6. Promotility agents are not recommended as routine for
pre-endoscopy.
7. Selected patients determined at low risk for rebleed after acute
ulcer bleeding may be considered for discharge after endoscopy.
8. Pre-endoscopic PPI therapy may be useful to downstage the
lesion and decrease need for endoscopic management, but its
administration should not delay endoscopic evaluation.
Endoscopic management: Pharmacologic management:
1. Institution-specific protocols for multidisciplinary UGIB 1. Histamine receptor antagonists are not recommended for
are recommended; availability of an endoscopist trained treatment in acute UGIB.
in endoscopic hemostasis is recommended. 2. Somatostatin and octreotide are not recommended for
2. Urgent availability of endoscopic support staff treatment in acute UGIB.
3. Endoscopy within 24 hours for most cases 3. PPI infusion protocol (bolus plus continuous infusion) should be
4. Hemostatic treatment not indicated for low-risk lesions given post–successful endoscopic treatment to all high-risk
5. Clots in the ulcer bed should be irrigated with the goal lesion patients to reduce rebleeding and mortality.
of revealing the underlying lesion for treatment. 4. Patients should be discharged on once-daily regimen of oral PPI
6. Removing adherent clots is controversial; consider for a duration appropriate to the treated etiology.
endoscopic therapy vs. intensive PPI therapy.
7. Hemostatic endoscopic treatment is indicated for
high-risk lesions.
8. Epinephrine injection alone is suboptimal: best used as
part of a combined therapy
9. Endoscopic coaptive therapy methods are equivalent.
10. Clips, thermocoagulation, or sclerosant injection should
be used for high-risk lesions, alone or combined with
epinephrine.
11. Second-look endoscopy as a routine is not recommended.
12. In case of rebleeding, a second endoscopic therapy
attempt is generally recommended.
Nonendoscopic and nonpharmacologic in-hospital Postdischarge ASA and NSAIDs:
management: 1. For patients with history of ulcer bleeding who require chronic
1. Following endoscopy, patients at low risk may be fed treatment with NSAIDs, it must be recognized that whether
within 24 hours. treatment is with traditional NSAID plus PPI or with COX-2
2. Patients with high-risk lesions should remain hospitalized inhibitor alone, the risk for recurrent ulcer bleeding remains
for 72 hours after endoscopy. clinically significant.
3. Surgical consultation is advised when endoscopic therapy 2. For patients with history of ulcer bleeding, the combination of
has failed. a COX-2 antagonist plus PPI is recommended to reduce risk of
4. Percutaneous embolization may provide an alternative to rebleed compared with COX-2 agent alone.
surgery when endoscopic therapy has failed. 3. For patients on low-dose ASA for cardiovascular risk who then
5. Patients with bleeding ulcers require testing for develop UGIB, the ASA therapy should be resumed as soon as
Helicobacter pylori and should receive eradication therapy risk for cardiovascular complication is thought to outweigh risk
if positive; eradication should be confirmed with a for rebleed.
negative test result. 4. In patients who require cardiovascular prophylaxis after ulcer
6. Negative acute testing for H. pylori should be repeated. bleeding, it should be noted that clopidogrel alone has a higher
risk for rebleeding than ASA plus PPI.

Adapted from Barkun AN, Bardou M, Kuipers EJ, et al. International consensus recommendations on the management of patients with nonvariceal upper
gastrointestinal bleeding. Ann Intern Med 2010;152:101–13; Barkun A, Bardou M, Marshall JK, et al. Consensus recommendations for managing patients with
nonvariceal upper gastrointestinal bleeding. Ann Intern Med 2003;139:843–57.
*Nonvariceal UGIB.
ASA, Acetylsalicylic acid; COX, cyclooxygenase; Hgb, hemoglobin; NSAIDs, nonsteroidal anti-inflammatory drugs; PRBC, packed red blood cell; PPI, proton pump
inhibitor; UGIB, upper gastrointestinal bleeding.
Section II—Clinical and Laboratory Assessment 229

Table 23.2  Summary of Recommendations for Acute Variceal Hemorrhage from the Practice
Parameters Committee of the American College of Gastroenterology
HEMORRHAGE FROM ESOPHAGEAL VARICES
1. Acute UGIB in patients with cirrhosis requires prompt attention with judicious intravascular volume support and blood
transfusion aimed at maintenance of Hgb at ≈8 g/dL.
2. Short, up to 1 week of antibiotic prophylaxis of SBP and other infections is recommended for all cirrhotic patients with
gastrointestinal hemorrhage: oral norfloxacin 400 mg bid or intravenous (IV) ciprofloxacin or IV ceftriaxone 1 g/day in advanced
cirrhotic patients or when quinolone resistance may be suspected.
3. Pharmacologic therapy using somatostatin or its analogs octreotide, vapreotide and terlipressin (latter not available in United
States) should be initiated immediately on suspicion of variceal bleeding and continued 3 to 5 days after diagnosis.
4. Endoscopy should be performed within 12 hours to diagnose variceal hemorrhage and treat by endoscopic variceal ligation (EVL)
or sclerotherapy.
5. Transjugular intrahepatic portosystemic shunt (TIPS) is indicated for patients with uncontrolled bleeding, or who have failed
combined pharmacologic and endoscopic treatment.
6. Balloon tamponade should be used as a temporizing measure maximally for 24 hours in patients with uncontrollable bleeding
while awaiting definitive endoscopic or TIPS treatment.
HEMORRHAGE FROM GASTRIC VARICES
1. In patients bleeding from gastric fundal varices, endoscopic variceal obturation using tissue adhesives such as cyanoacrylate is
preferred if available; EVL is an option.
2. TIPS is indicated for patients with uncontrolled bleeding or who have failed combined pharmacologic and endoscopic treatment.

Adapted from Garcia-Tsao G, Sanyal AJ, Grace ND, et al. Prevention and management of gastroesophageal varices and variceal hemorrhage in cirrhosis. Am J
Gastroenterol 2007;102:2086–102.
Hgb, Hemoglobin; SBP, spontaneous bacterial peritonitis; UGIB, upper gastrointestinal bleeding.

Bloody diarrhea
Hematochezia

Evaluate, resuscitate • Stool culture


consult GI endoscopist • Stool E. coli O157:H7
• Stool C. diff toxin
• Immunocompromised
consider CMV
NGT aspirate • Consider IBD
• Treat as appropriate
to identified cause
• If pain consider
ischemia

No blood Any blood


normal gastric/ or concern for UGIB, or
duodenal fluids hemodynamic instability

Bowel prep →
(–) (+)
Treated as
Colonoscopy EGD
appropriate

Source No source Technically


identified identified impossible

Treated as Continued Severe Arteriography


Fig. 23.1  Algorithm for lower gastrointestinal
appropriate bleeding Nuclear scanning
CT/CTA bleeding (LGIB). C. diff, Clostridium difficile; CMV,
Surgical consult cytomegalovirus; CT, computed tomography; CTA,
No or lesser Small bowel
bleeding suspected computed tomography angiography; E. coli,
Escherichia coli; EGD, esophagogastroduodenoscopy;
GI, gastrointestinal; IBD, inflammatory bowel disease;
Small Bowel Studies MRI, magnetic resonance imaging; NGT, nasogastric
Capsule enteroscopy tube; UGIB, upper gastrointestinal bleeding. (Modified
Push enteroscopy from Barnert J, Messmann H. Diagnosis and management of
Double balloon enteroscopy
lower gastrointestinal bleeding. Nat Rev Gastroenterol
CT or MRI enterography techniques
Hepatol 2009;6:637–46.)
230 Section II—Clinical and Laboratory Assessment

Hepatic Failure Table 23.3  West Haven Criteria for


Few patients with hepatic failure are cared for in the NCCU. Semiquantitative Grading of Mental State
However, patients with hepatic encephalopathy and cerebral in Acute Liver Failure
edema frequently require the expertise of neurointensivists GRADE I
and neurosurgeons and management in the NCCU, for
example, when therapeutic hypothermia is used for increased • Trivial lack of awareness
• Euphoria or anxiety
intracranial pressure (ICP) associated with hepatic encepha- • Shortened attention span
lopathy.13,14 The pathology of hepatic encephalopathy is • Impaired performance of addition
incompletely understood but appears to be associated with GRADE II
nitrogenous GI products and in particular ammonia that
bypass liver metabolism in acute liver failure (ALF). These • Lethargy or apathy
• Minimal disorientation for time or place
substances cross the blood-brain barrier, where they are • Subtle personality change
thought to increase gama-aminobutyric acid (GABAergic) • Inappropriate behavior
transmission and injure astrocytes that subsequently swell • Impaired performance of subtraction
(i.e., cytotoxic edema). For this reason, corticosteroids are of GRADE III
no use. On the other hand, several lines of evidence suggest
• Somnolence to semistupor, but responsive to verbal stimuli
that proinflammatory mechanisms are involved in the patho- • Confusion
genesis of brain edema in ALF; this proposed mechanism • Gross disorientation
provides a basis for using induced hypothermia to bridge GRADE IV
patients with hepatic encephalopathy to transplant.15,16
• Coma (unresponsive to verbal or noxious stimuli)
ALF, also know as fulminant liver failure, is the abrupt loss
of liver function in a patient without previous liver disease. It Adapted from Atterbury CE, Maddrey WC, Conn HO. Neomycin-sorbitol and
typically is associated with coagulopathy (international nor- lactulose in the treatment of acute portal-systemic encephalopathy: a
controlled, double-blind clinical trial. Am J Dig Dis 1978;23:398–406.
malized ratio [INR] >0.5) and encephalopathy. In the United
States, acetaminophen accounts for approximately 50% of
ALF cases: other causes include hepatitis, drug-induced liver
injury, and viral or autoimmune hepatitis. Initial evaluation The Acute Physiology and Chronic Health Evaluation
includes a liver function panel, prothrombin time [PT]/INR, (APACHE) II score or Sequential Organ Failure Assessment
complete blood count, fibrinogen, D-dimer, acetaminophen (SOFA) score also can help predict outcome in acetaminophen-
level, toxicology screen, electrolytes, and creatinine. In addi- induced ALF.20,21 These scores (SOFA, MELD) and Simplified
tion, a search for an etiology is necessary; this may include Acute Physiology Score (SAPS) II also can be used to predict
alpha-fetoprotein, ceruloplasmin, serum protein electropho- outcome among patients with chronic liver insufficiency and
resis, virology (cytomegalovirus; Epstein-Barr virus; hepatitis cirrhosis who require ICU admission.22 Some studies suggest
A, B, or C virus antigens; or antibodies), or antinuclear anti- that ICU prognostic models such as SOFA may perform better
body among others. An abdominal computed tomography than liver specific scores such as Child-Turcotte-Pugh (CTP)
(CT) scan to evaluate liver volume is useful, but if not feasible or MELD for outcome prediction among cirrhotic patients
bedside ultrasound may be obtained. A head CT scan is neces- who are admitted to the ICU.23
sary for patients with grade III or IV encephalopathy (see The average overall mortality in ALF is 80%. By contrast,
following text). During an ICU stay, patient follow-up evalu- 60% to 80% survive after transplant. Thus once a patient is
ation may include arterial blood gas, arterial lactate, arterial considered eligible for a transplant, the goals of care are to
blood oxygen (ABO) analysis (two separate tests), and every- provide a bridge until this occurs. In 2007, the U.S. Acute
6-hour PT/INR, transaminase level, total and direct bilirubin, Liver Failure Study Group (ALFSG) published guidelines to
and serum sodium. Frequent assessment of serum osmolarity codify ICU management of ALF patients in their participat-
or osmolality gap (when mannitol is used), and coagulation ing centers.24 Key points associated with the neurologic
status also is required. aspects and in particular patient monitoring of fulminant
There are two important approaches to classification of ALF liver failure is summarized later in this chapter.14 Other
patients: (1) grading of hepatic encephalopathy and (2) deter- aspects of ICU care—hemodynamic support, mechanical
mining whether a patient is eligible for transplant and whether ventilation, renal support, prevention of infection, and nutri-
transplant is needed to prevent death. There are several clas- tion and management of end-stage liver disease—are dis-
sification scales for hepatic encephalopathy, but the West cussed elsewhere.25-27
Haven Simplified Criteria17 (Table 23.3) is used most fre-
quently. The severity of encephalopathy is the main barometer
of disease severity and grades III and IV often mark the Hepatic Encephalopathy
threshold for escalation of therapies. Determining who and Hyperammonemia
requires a liver transplant to prevent death is complex and Hepatic encephalopathy (HE) is a feature of ALF. However,
requires consideration of ethical factors and an anticipation patients with chronic liver failure who acutely deteriorate
of future needs to ensure that definitive therapy is available in (acute on chronic liver failure) may also experience HE.28 In
time to rescue the patient. The King’s College Criteria18 and these patients HE manifests by confusion, poor judgment, or
the Model for End-Stage Liver Disease (MELD)19 are widely personality change among other clinical features that may be
used scales to predict mortality in ALF and to help triage exacerbated by the disease that requires ICU admission or
patients with severe liver disease for transplant (Table 23.4). the admission itself. The terminology associated with HE is
Section II—Clinical and Laboratory Assessment 231

Table 23.4  Common Prognostic Scales for Mortality Risk Assessment in Acute Liver Failure
Model for End-Stage
King’s College Criteria18 Modified King’s College18 Liver Disease (MELD)19
Non-APAP all cause prothrombin time (PT) >100 seconds Acetaminophen-induced disease MELD score =
(INR >6.5) Arterial pH <7.3 (irrespective of 3.78 [Ln serum bilirubin mg/dL]
Or grade of encephalopathy) +
Any three of the following: Or 11.2 [Ln INR]
Age <10 or >40 years Grade III or IV encephalopathy +
Etiology: non-A, non-B hepatitis, halothane hepatitis, and 9.57 [Ln serum creatinine mg/dL]
idiosyncratic drug reactions PT >100 seconds (INR >6.5) +
Duration of jaundice before onset of encephalopathy and 6.43
>7 days Serum creatinine >3.4 mg/dL
PT >50 seconds ____________________________
Serum bilirubin >18 mg/dL (all of scale is irrespective of Interpretation:
the grade of encephalopathy)
40 or more → 71.3% mortality
30-39 → 52.6% mortality
20-29 → 19.6% mortality
10-19 → 6.0% mortality
<9 → 1.9% mortality
PPV mortality = 98% PPV mortality = 84% 3-month mortality
NPV mortality = 82% NPV mortality = 86%

APAP, N-acetyl-p-aminophenol; INR, international normalized ratio; Ln, natural logarithm; NPV, negative predictive value; PPV, positive predictive value.

defined in a report of the Hepatic Encephalopathy Consensus Cerebral Edema


Group at the World Congress of Gastroenterology in 1998.29,30 Cerebral edema is most prominent in patients with the fastest
Brain astrocytes internally detoxify ammonia to glutamine, onset of ALF and is associated with increased morbidity and
an osmotically active moiety. Although increased brain am­ mortality. Cerebral edema also may be observed in patients
monia and its detoxification product, glutamine, appear to with severe chronic liver disease or with portosystemic shunts.
play a role in the pathogenesis of HE and brain edema patho- For all HE grade III or IV patients and for those with an acute
genesis,31 the evidence for ammonia treatment is insufficient change in mental status, a baseline brain CT is recommended.
for a high-level recommendation in consensus guidelines. A normal CT scan does not exclude the likelihood of increased
Similarly, the role of probiotics or rifaximin for HE is still ICP. Intracranial hypertension is more likely in patients
being elucidated.32,33 If ammonia-reducing agents are admin- with hyperacute and acute causes of ALF, grade III or IV HE,
istered, lactulose is preferred. Neomycin also may be used but those who develop pupillary abnormalities, seizures, systemic
generally is not recommended because its use is associated inflammation, an arterial ammonia greater than 150 µmol/L,
with a risk for nephrotoxicity. Potential adverse effects of hyponatremia, and those on vasopressor support.38 Whether
inhibitors of brain glutamine synthesis (e.g., methionine sulf- to place an ICP monitor is controversial largely because of
oximine) to prevent brain edema also have limited their use bleeding concerns and a lack of randomized trials to guide
in ALF. When lactulose is used, the following should be moni- clinicians. However, ICP monitoring can help guide manage-
tored: (1) stool output and (2) the grade of HE and develop- ment of cerebral edema. The ALFSG group24 recommends an
ment of harmful side effects including abdominal distention, ICP monitor be considered for grade III or IV HE patients
increased risk for aspiration, and dehydration. Interval assess- being bridged to liver transplant, and those with a potential
ment of serum ammonia levels may aid in the optimal titra- for survival without transplant who receive medical manage-
tion of the therapy, although the relationship between ment. Because the bleeding risks are 10% to 20%, mild HE
ammonia levels and severity of HE across patients varies (grade I and II) patients should not have an invasive ICP
because of differences in ammonia metabolism and analytic monitor. Subarachnoid and intraparenchymal fiber-optic ICP
methods. Newer methods such as quantification of ammo- monitors are preferred because the risk associated with ven-
nium in the breath have been described.34 triculostomy use is greater, whereas epidural monitors have
limited accuracy. Treatments to optimize coagulation should
be given immediately before the procedure. Alternatively non-
Seizures invasive ICP monitors may be considered or used to decide
Seizures, including subtle manifestations are common in ALF about the value of invasive monitors (see Chapters 34 and 46).
patients with grade III or IV HE.35 Although the available data The optimal ICP or cerebral perfusion pressure (CPP) asso-
are conflicting, they do not support routine phenytoin pro- ciated with increased survival and recovery in grade III or IV
phylaxis. Sedation using benzodiazepines or propofol can HE is not known; however, recommended targets are to main-
confer antiepileptic benefits. Electroencephalography (EEG) is tain ICP less than 25 and CPP from 50 to 80 mm Hg. Patient
recommended for grade III or IV HE when there is: (1) an volume status needs to be monitored to help maintain nor-
unexplained episodic change in neurologic examination, (2) motension and to guide correction of hypovolemia where
myoclonus, and (3) a need to titrate barbiturate coma or guide present. Vasopressors may be used as a second-line therapy
induced hypothermia to control increased ICP.36,37 when systolic blood pressure (SBP) is less than 90 mm Hg or
232 Section II—Clinical and Laboratory Assessment

mean arterial pressure (MAP) is less than 65 mm Hg to help


keep CPP between 50 and 80 mm Hg. Table 23.5  Ranson Criteria in Acute
ICP management in HE is similar to that for other condi- Pancreatitis
tions that cause increased ICP. The following general features Prognosis for
should be monitored: (1) head of bed position, (2) head in At Admission Initial 48 Hours Mortality
line position, (3) level of agitation (see Chapter 11), (4)
Age >55 years Fall in Hct >10% <3 signs = 1%
PaCO2 (ideally 30-40 mm Hg, and (5) systemic temperature WBC >16,000/mm3 Calcium <8 mg/dL 3-4 signs = 16%
to target normothermia and avoid hyperpyrexia. When ICP is Glucose >200 mg/dL PaO2 <60 mm Hg 5-6 signs = 40%
monitored, these variables can be optimized against the ICP LDH >350 IU/L Base deficit >4 mEq/L >7 signs = 100%
measurement. Cerebral hyperemia is considered to be one of SGOT >250 SF units BUN increase by
>5 mg/dL
several factors associated with cerebral edema in ALF. The Third space losses >6 L
role of therapies such as hyperventilation, N-acetylcysteine,
thiopentone sodium, and propofol still has to be fully From Ranson JH, Rifkind KM, Roses DF, et al. Prognostic signs and the
role of operative management in acute pancreatitis. Surgery, Gynecology &
elucidated.39 Obstetrics 1974;139(1):69-81
The ALFSG recommends mannitol as the first-line osmotic BUN, Blood urea nitrogen; Hct, hematocrit; LDH, lactate dehydrogenase;
agent for elevated ICP in ALF. Whether a scheduled or PaO2, arterial oxygen tension; SGOT, serum glutamic-oxaloacetic transaminase;
as-needed dose is preferred is not indicated but because of a WBC, white blood cell.

better side effect profile, lower bolus doses where effective


(e.g., 0.25-0.50 g/kg) are recommended. These bolus doses
should continue while ICP is greater than 25 provided serum pancreatitis the median prevalence for organ failure is about
osomolarity is less than 320 mOsm/L; this should be checked 50%. Overall mortality for pancreatitis is 5% with a range
every 6 hours. The interval calculation of the osmolar gap may between 3% for interstitial and 17% for necrotizing pancre-
help determine timing of subsequent doses to sustain a thera- atitis. In the absence of organ failure, mortality is zero but
peutic osmotic effect. Hypertonic saline as a bolus treatment increases to 3% for single-organ failure and up to 50% for
or a continuous infusion also may be an option to help control multiple-organ failure.41
elevated ICP. When used, serum sodium should be checked In patients with pancreatitis initial laboratory evaluation
every 6 hours to target serum sodium of 145 to 155. should include complete blood count (CBC) with differential,
Second-line therapies for increased ICP include induced complete serum chemistry including calcium, liver function,
hypothermia or barbiturate coma. Cooling does lower ICP in lactate dehydrogenase, amylase, lipase, and triglycerides. The
HE and can help bridge a patient to liver transplant; an ICP Ranson Criteria, particularly those based on 48-hour labora-
monitor should guide the depth of cooling.13 Temperature tory data can be used to estimate mortality risk and for risk
needs to be closely monitored with a target temperature in the stratification (Table 23.5). In addition, APACHE II scores are
mild to moderate hypothermia range, but not less than 32° C. useful: scores that increase during the first 48 hours are associ-
Modern indwelling catheter-based systems or surface cooling ated with worse course, whereas improving scores are associ-
methods that have tight thermostatic regulation in a feedback ated with a benign course. Hemoconcentration at admission
loop from a reliable core temperature sensor work best. Older may be a poor prognostic indicator and particularly if the
devices are more difficult to regulate and often yield undesir- hematocrit then increases. In the absence of hemoconcentra-
ably large temperature fluctuations. When induced hypother- tion, pancreatic necrosis is unlikely. Serum C-reactive protein
mia is used, electrolytes and coagulation parameters should (CRP) levels tend to peak at 36 to 72 hours, and when greater
be assessed frequently and urine output and hydration status than 150 mg/L may suggest pancreatic necrosis.
must be closely monitored because “cold diuresis” can occur. Except where there is need to evaluate for alternative diag-
Initiation and maintenance of barbiturate coma require nostic considerations, a CT of the abdomen is more useful
intensive hemodynamic monitoring to titrate therapies for when obtained after a delay of 2 to 4 days than at admission.
hemodynamic stability. Continuous EEG looking for a burst By that time a more severe clinical course has declared itself,
suppression pattern helps to titrate the barbiturate dose. For and the CT then can distinguish between simple interstitial
example, a progressive increase in the time between bursts disease or the presence of necrosis and microcirculation dis-
may indicate a higher than necessary dose and thus a need to ruption. This distinction requires contrast administration,
reduce the infusion rate of a barbiturate maintenance dose. and the study is often called a dynamic CT of the pancreas or
Daily serum levels of pentobarbital should be followed. contrast-enhanced multidetector CT (MDCT). The timing of
However, the adequacy of therapy is demonstrated by success- this study should be considered carefully to avoid contrast
ful reduction of ICP as indicated by an ICP monitor. with a rising serum creatinine and evolving renal failure.
When findings of necrotizing pancreatitis are present, inter-
mittent serial CT examinations may be required to monitor
Pancreatitis for secondary intra-abdominal complications, pseudocyst
Pancreatitis, usually in its milder forms, occasionally may development, organized necrosis, and vascular complications
complicate the course of patients under care for primary such as thrombosis or pseudoaneurysms. However, the cumu-
neurologic illness. Guidelines provide an overview of the dis- lative dose of radiation with serial CT scans needs to be con-
tribution of severity in acute pancreatitis.40 Simple interstitial sidered.42 The Balthazar CT criteria for acute pancreatitis43
pancreatitis accounts for 85% of cases, whereas 15% are nec- divide the severity of pancreatitis into five grades, A through
rotizing and about a third of these develop infected necrosis. E: normal, enlarged pancreas, inflamed and or peripancreatic
One in 10 patients with interstitial pancreatitis develops any fat, single fluid collection, and two or more fluid collections.
organ failure, which tends to be reversible. In necrotizing Grade A indicates the least and grade E the most severe form
Section II—Clinical and Laboratory Assessment 233

of pancreatitis. Most patients with severe pancreatitis have one intermittent pressure transduction—every hour in the at-risk
or several pancreatic fluid collections (grades D and E). population—is used most frequently.50,51
Increasing grade is associated with increasing morbidity and Abdominal perfusion pressure (APP) is described by:
mortality. The CT severity grade is an attempt to improve the
APP = MAP − IAP
prognostic accuracy of CT. In each grade patients are assigned
points for the amount of necrosis; higher numerical scores Abdominal compartment syndrome and organ function
indicate greater extent of necrosis and worse prognosis.44,45 In compromise occur when IAP approaches MAP and visceral
general, CT grading scales more accurately diagnose clinically perfusion is compromised. The optimal target APP is greater
severe disease and better identify pancreatic infection or the than or equal to 60 mm Hg. Normal IAP in critically ill
need for intervention than do clinical criteria.46 patients is thought to be 5 to 7 mm Hg. It may be near zero
Patients with more severe pancreatitis or those with altered in the healthy population. IAP is graded according to sever-
vital signs, diminishing urine output (UOP), increasing ity50,51: grade I, 12-15 mm Hg; grade II, 16 to 20 mm Hg;
volume requirements, tachypnea or tachycardia, encephalopa- grade III, 21 to 25 mm Hg; grade IV, greater than 25 mm Hg.
thy, or any indication of early organ failure require ICU care. There is no single pressure threshold at which reductions in
Initial care focuses on adequate fluid resuscitation and avoid- microcirculatory blood flow sufficient to initiate organ dys-
ance of hypoxemia. In more severe cases third spacing can be function, are certain to occur. Oliguria is one of the first
profound, and aggressive fluid support may be required. outward signs of organ compromise.52-54 Critical IAP levels
Emesis, diaphoresis, and vascular permeability all contribute seem to begin in the range of 10 to 15 mm Hg. Abdominal
to the volume challenges, and the development of hypovole- compartment syndrome therefore should not defined by a
mia increases the risk of a necrotizing process if the microcir- single number but rather as sustained IAP greater than
culation is inadequately supplied. Fluid status therefore should 20 mm Hg (with or without APP <60 mm Hg) that is associ-
be monitored closely using continuous monitoring of blood ated with new organ dysfunction or failure.50 Management of
pressure, heart rate, and urine output. In more severe cases, intra-abdominal hypertension (abdominal compartment syn-
management may be best guided by CVP and ScvO2 monitor- drome) includes: (1) correction of positive fluid balance, (2)
ing and noninvasive monitoring techniques of cardiac func- evacuation of intraluminal contents, (3) percutaneous drain-
tion that provide data on cardiac performance, preload, and age of peritoneal free fluid, (4) improvement of abdominal
systemic vascular resistance (see Chapter 19). wall compliance such as prone position and alternating (asyn-
Infected pancreatic necrosis occurs in about one third of chronous) ventilation, or (5) decompression of the abdomen
patients with necrotizing pancreatitis and generally develops by laparotomy.55,56
after the first week. Clinically, patients with or without infected
necrotizing pancreatitis appear similar. An abdominal CT may
help make the diagnosis because air bubbles may be observed
Conclusion
in the retroperitoneum with infection but not in sterile necro- Gastrointestinal (GI) disorders, GI bleeding, hepatic dysfunc-
sis. Diagnosis is made by CT-guided percutaneous aspiration tion, pancreatitis, and intra-abdominal compartment syn-
that if negative may be repeated every 5 to 7 days when sys- drome can develop in patients in the NCCU. In addition,
temic toxicity persists. Surgical debridement, often repeated, some patients with hepatic encephalopathy require care in the
is the gold standard for treatment. NCCU to manage increased ICP (e.g., using therapeutic
hypothermia).13,14 The neurointensivist therefore should have
knowledge of these disorders and how to care for the various
Abdominal Compartment Syndrome conditions. A number of grading scales (e.g., the West Haven
Increased abdominal pressure may arise in many disorders, Criteria,17 MELD criteria,19 or Ranson Criteria)57 among
but some more common conditions include cirrhosis with others, that are specific to GI, liver, and pancreatic disease or
ascites, following major trauma, pancreatitis, after massive general ICU scales (e.g., APACHE, SOFA, or SAPs)20-22 can
resuscitation or massive transfusion, sepsis, acidosis, or respi- help guide care. In addition, groups such as the U.S. Acute
ratory failure that requires high positive end-expiratory pres- Liver Failure Study Group (ALFSG) and the Practice Param-
sure (PEEP) levels for treatment. A high index of clinical eters Committee of the American College of Gastroenterology
suspicion in the appropriate circumstances, and willingness to have published guidelines to codify ICU management of
initiate IAP monitoring, usually by bladder pressure measure- patients with GI, hepatic, or pancreatic disease.9-12,24
ment, is needed to diagnose dangerous, but often externally
occult, elevations in intra-abdominal pressure (IAP).47,48
Failure to recognize this condition and failure to monitor it References
can compromise respiratory function and intra-abdominal 1. Ali T, Harty RF. Stress-induced ulcer bleeding in critically ill patients.
organ function and lead to multiorgan failure and death.49 Gastroenterol Clin North Am 2009;38(2):245–65.
IAP is the steady-state pressure inside the abdominal cavity. 2. Vincent JL, Rhodes A, Perel A, et al. Clinical review: update on
hemodynamic monitoring: a consensus of 16. Crit Care 2011;15(4):229.
It is measured in millimeters of mercury (mm Hg). The usual 3. Barkun A, Bardou M, Marshall JK, et al. Consensus recommendations for
method to measure IAP is through fluid column pressure managing patients with nonvariceal upper gastrointestinal bleeding. Ann
transduction from the port of a Foley catheter into which Intern Med 2003;139(10):843–57.
25 mL of sterile saline are instilled into the bladder with the 4. Greenspoon J, Barkun A, Bardou M, et al. Management of patients with
Foley catheter clamped just distal to the measurement port. nonvariceal upper gastrointestinal bleeding. Clin Gastroenterol Hepatol
2012;10(3):234–9. Epub 2011, Aug 4.
Steady-state pressure is obtained at end expiration in a relaxed 5. Stanley AJ, Ashley D, Dalton HR, et al. Outpatient management of patients
patient with the transducer zeroed at the midaxillary line. with low-risk upper-gastrointestinal haemorrhage: multi-centre validation
Continuous measurement protocols are described, but and prospective evaluation. Lancet 2009;373:42–7.
234 Section II—Clinical and Laboratory Assessment

6. Packham CJ, Rockall TA, Logan RF. Outpatient care for selected patients 29. Ferenci P, Lockwood A, Mullen K, et al. Hepatic encephalopathy: definition,
with acute upper gastrointestinal bleeding. Lancet 1995;345:659–60. nomenclature, diagnosis, and quantification: final report of the working
7. Kim BC, Cheon JH, Kim TI, et al. Risk factors and the role of bedside party at the 11th World Congresses of Gastroenterology, Vienna, 1998.
colonoscopy for lower gastrointestinal hemorrhage in critically ill patients. Hepatology 2002;35(3):716–21.
Hepatogastroenterology 2008;55(88):2108–11. 30. Mullen KD, Prakash RK. New perspectives in hepatic encephalopathy. Clin
8. Green BT, Rockey DC, Portwood G, et al. Urgent colonoscopy for evaluation Liver Dis 2012;16(1):1–5. Epub 2012, Jan 16.
and management of acute lower gastrointestinal hemorrhage: a randomized 31. Desjardins P, Du T, Jiang W, et al. Pathogenesis of hepatic encephalopathy
controlled trial. Am J Gastroenterol 2005;100:2395–402. and brain edema in acute liver failure: role of glutamine redefined.
9. Barkun AN, Bardou M, Kuipers EJ, et al. International consensus Neurochem Int 2012 Feb 21. Epub ahead of print.
recommendations on the management of patients with nonvariceal upper 32. McGee RG, Bakens A, Wiley K, et al. Probiotics for patients with hepatic
gastrointestinal bleeding. Ann Intern Med 2010;152:101–13. encephalopathy. Cochrane Database Syst Rev 2011;11:CD008716.
10. Barkun A, Bardou M, Marshall JK, et al. Consensus recommendations for 33. Bass NM, Mullen KD, Sanyal A, et al. Rifaximin treatment in hepatic
managing patients with nonvariceal upper gastrointestinal bleeding. Ann encephalopathy. N Engl J Med 2010;362(12):1071–81.
Intern Med 2003;139:843–57. 34. Blanco Vela CI, Bosques Padilla FJ. Determination of ammonia
11. Garcia-Tsao G, Sanyal AJ, Grace ND, et al. Prevention and management of concentrations in cirrhosis patients: still confusing after all these years? Ann
gastroesophageal varices and variceal hemorrhage in cirrhosis. Am J Hepatol 2011;10(Suppl 2):S60–5.
Gastroenterol 2007;102:2086–102. 35. Ellis AJ, Wendon JA, Williams R: Subclinical seizure activity and prophylactic
12. Barnert J, Messmann H. Diagnosis and management of lower phenytoin infusion in acute liver failure: a controlled clinical trial.
gastrointestinal bleeding. Nat Rev Gastroenterol Hepatol 2009;6:637–46. Hepatology 2000;32:536–41.
13. Stravitz RT, Larsen FS. Therapeutic hypothermia for acute liver failure. Crit 36. Kaplan PW, Rossetti AO. EEG patterns and imaging correlations in
Care Med 2009;37(7 Suppl):S258–64. encephalopathy: encephalopathy part II. J Clin Neurophysiol
14. Frontera JA, Kalb T. Neurological management of fulminant hepatic failure. 2011;28(3):233–51.
Neurocrit Care 2011;14(2):318–27. 37. Marchetti P, D’Avanzo C, Orsato R, et al. Electroencephalography in
15. Jiang W, Desjardins P, Butterworth RF. Direct evidence for central patients with cirrhosis. Gastroenterology 2011;141(5):1680–9.e1-2. Epub
proinflammatory mechanisms in rats with experimental acute liver failure: 2011, Jul 18.
protective effect of hypothermia. J Cereb Blood Flow Metab 2009;29(5): 38. Shawcross DL, Wendon JA. The neurological manifestations of acute liver
944–52. Epub 2009, Mar 4. failure. Neurochem Int 2011 Nov 2. Epub ahead of print.
16. Stravitz RT, Lee WM, Kramer AH, et al. Therapeutic hypothermia for acute 39. Mpabanzi L, Jalan R. Neurological complications of acute liver failure:
liver failure: toward a randomized, controlled trial in patients with advanced pathophysiological basis of current management and emerging therapies.
hepatic encephalopathy. Neurocrit Care 2008;9(1):90–6. Neurochem Int 2011 Nov 13. Epub ahead of print.
17. Atterbury CE, Maddrey WC, Conn HO. Neomycin-sorbitol and lactulose in 40. Banks PA, Freeman ML, the Practice Guidelines Committee of the American
the treatment of acute portal-systemic encephalopathy: a controlled, College of Gastroenterology. Practice guidelines in acute pancreatitis. Am J
double-blind clinical trial. Am J Dig Dis 1978;23:398–406. Gastroenterol 2006;101:2379–400.
18. O’Grady JG, Alexander GJ, Hayllar KM, et al. Early indicators of prognosis 41. Vege SS, Gardner TB, Chari ST, et al. Low mortality and high morbidity in
in fulminant hepatic failure. Gastroenterology 1989;97:439–55. severe acute pancreatitis without organ failure: a case for revising the Atlanta
19. Weisner R, Edwards E, Freeman R, et al. Model for end-stage liver disease classification to include “moderately severe acute pancreatitis.” Am J
(MELD) and allocation of donor livers. Gastroenterology 2003;124:91–6. Gastroenterol 2009;104(3):710–5. Epub 2009, Feb 3.
20. Larson AM, Polson J, Fontana RJ, et al. Acetaminophen-induced acute liver 42. Ball CG, Correa-Gallego C, Howard TJ, et al. Radiation dose from computed
failure: results of a United States multicenter, prospective study. Hepatology tomography in patients with necrotizing pancreatitis: how much is too
2005;42:1364–72. much? J Gastrointest Surg 2010;14(10):1529–35. Epub 2010, Sep 8.
21. Craig DG, Reid TW, Wright EC, et al. The sequential organ failure 43. Balthazar EJ, Ranson JHC, Naidich DP, et al. Acute pancreatitis: prognostic
assessment (SOFA) score is prognostically superior to the model for value of CT. Radiology 1985;156:767–72.
end-stage liver disease (MELD) and MELD variants following paracetamol 44. Balthazar EJ, Robinson DL, Megibow AJ, et al. Acute pancreatitis: value of
(acetaminophen) overdose. Aliment Pharmacol Ther 2012;35(6):705–13. CT in establishing prognosis. Radiology 1990;174:331–6.
doi: 10.1111/j.1365-2036.2012.04996.x. Epub 2012, Jan 20. 45. Balthazar EJ. Acute pancreatitis: assessment of severity with clinical and CT
22. Levesque E, Hoti E, Azoulay D, et al. Prospective evaluation of the prognostic evaluation. Radiology 2002;223(3):603–13. Review.
scores for cirrhotic patients admitted to an intensive care unit. J Hepatol 46. Bollen TL, Singh VK, Maurer R, et al. Comparative evaluation of the
2012;56(1):95–102. Epub 2011, Aug 9. modified CT severity index and CT severity index in assessing severity of
23. Cholongitas E, Senzolo M, Patch D, et al. Review article: scoring systems for acute pancreatitis. AJR Am J Roentgenol 2011;197(2):386–92.
assessing prognosis in critically ill adult cirrhotics. Aliment Pharmacol Ther 47. Malbrain ML, Chiumello D, Pelosi P, et al. Incidence and prognosis
2006 1;24(3):453–64. of intraabdominal hypertension in a mixed population of critically ill
24. Stravitz RT, Kramer AH, Davern T, et al. Intensive care of patients with acute patients: a multiple-center epidemiological study. Crit Care Med 2005;33:
liver failure: recommendations of the U.S. Acute Liver Failure Study Group. 315–22.
Crit Care Med 2007;35:2498–508. 48. Malbrain ML. Abdominal pressure in the critically ill: measurement and
25. Findlay JY, Fix OK, Paugam-Burtz C, et al. Critical care of the end-stage liver clinical relevance. Intensive Care Med 1999;25:1453–8.
disease patient awaiting liver transplantation. Liver Transpl 2011;17(5): 49. Hedenstierna G, Larsson A. Influence of abdominal pressure on respiratory
496–510. doi: 10.1002/lt.22269. and abdominal organ function. Curr Opin Crit Care 2012;18(1):80–5.
26. Al-Khafaji A, Huang DT. Critical care management of patients with Review.
end-stage liver disease. Crit Care Med 2011;39(5):1157–66. 50. Malbrain MLNG, Cheatham ML, Kirkpatrick A, et al. Results from the
27. Bémeur C, Desjardins P, Butterworth RF. Role of nutrition in the international conference of experts on intra-abdominal hypertension and
management of hepatic encephalopathy in end-stage liver failure. J Nutr compartment syndrome. I Definitions. Intensive Care Med 2006;32:
Metab 2010;489823. Epub 2010, Dec 22. 1722–32.
28. Wlodzimirow KA, Eslami S, Abu-Hanna A, et al. A systematic review on
prognostic indicators of acute on chronic liver failure and their predictive A complete list of references for this chapter can be found online at
value for mortality. Liver Int 2012 Mar 19. Epub ahead of print. www.expertconsult.com.
Section II—Clinical and Laboratory Assessment 234.e1

References 27. Bémeur C, Desjardins P, Butterworth RF. Role of nutrition in the


management of hepatic encephalopathy in end-stage liver failure. J Nutr
1. Ali T, Harty RF. Stress-induced ulcer bleeding in critically ill patients. Metab 2010;489823. Epub 2010, Dec 22.
Gastroenterol Clin North Am 2009;38(2):245–65. 28. Wlodzimirow KA, Eslami S, Abu-Hanna A, et al. A systematic review on
2. Vincent JL, Rhodes A, Perel A, et al. Clinical review: update on prognostic indicators of acute on chronic liver failure and their predictive
hemodynamic monitoring: a consensus of 16. Crit Care 2011;15(4):229. value for mortality. Liver Int 2012 Mar 19. Epub ahead of print.
3. Barkun A, Bardou M, Marshall JK, et al. Consensus recommendations for 29. Ferenci P, Lockwood A, Mullen K, et al. Hepatic encephalopathy: definition,
managing patients with nonvariceal upper gastrointestinal bleeding. Ann nomenclature, diagnosis, and quantification: final report of the working
Intern Med 2003;139(10):843–57. party at the 11th World Congresses of Gastroenterology, Vienna, 1998.
4. Greenspoon J, Barkun A, Bardou M, et al. Management of patients with Hepatology 2002;35(3):716–21.
nonvariceal upper gastrointestinal bleeding. Clin Gastroenterol Hepatol 30. Mullen KD, Prakash RK. New perspectives in hepatic encephalopathy. Clin
2012;10(3):234–9. Epub 2011, Aug 4. Liver Dis 2012;16(1):1–5. Epub 2012, Jan 16.
5. Stanley AJ, Ashley D, Dalton HR, et al. Outpatient management of patients 31. Desjardins P, Du T, Jiang W, et al. Pathogenesis of hepatic encephalopathy
with low-risk upper-gastrointestinal haemorrhage: multi-centre validation and brain edema in acute liver failure: role of glutamine redefined.
and prospective evaluation. Lancet 2009;373:42–7. Neurochem Int 2012. Epub ahead of print.
6. Packham CJ, Rockall TA, Logan RF. Outpatient care for selected patients 32. McGee RG, Bakens A, Wiley K, et al. Probiotics for patients with hepatic
with acute upper gastrointestinal bleeding. Lancet 1995;345:659–60. encephalopathy. Cochrane Database Syst Rev 2011;11:CD008716.
7. Kim BC, Cheon JH, Kim TI, et al. Risk factors and the role of bedside 33. Bass NM, Mullen KD, Sanyal A, et al. Rifaximin treatment in hepatic
colonoscopy for lower gastrointestinal hemorrhage in critically ill patients. encephalopathy. N Engl J Med 2010;362(12):1071–81.
Hepatogastroenterology 2008;55(88):2108–11. 34. Blanco Vela CI, Bosques Padilla FJ. Determination of ammonia
8. Green BT, Rockey DC, Portwood G, et al. Urgent colonoscopy for evaluation concentrations in cirrhosis patients: still confusing after all these years? Ann
and management of acute lower gastrointestinal hemorrhage: a randomized Hepatol 2011;10(Suppl 2):S60–5.
controlled trial. Am J Gastroenterol 2005;100:2395–402. 35. Ellis AJ, Wendon JA, Williams R: Subclinical seizure activity and prophylactic
9. Barkun AN, Bardou M, Kuipers EJ, et al. International consensus phenytoin infusion in acute liver failure: a controlled clinical trial.
recommendations on the management of patients with nonvariceal upper Hepatology 2000;32:536–41.
gastrointestinal bleeding. Ann Intern Med 2010;152:101–13. 36. Kaplan PW, Rossetti AO. EEG patterns and imaging correlations in
10. Barkun A, Bardou M, Marshall JK, et al. Consensus recommendations for encephalopathy: encephalopathy part II. J Clin Neurophysiol
managing patients with nonvariceal upper gastrointestinal bleeding. Ann 2011;28(3):233–51.
Intern Med 2003;139:843–57. 37. Marchetti P, D’Avanzo C, Orsato R, et al. Electroencephalography in patients
11. Garcia-Tsao G, Sanyal AJ, Grace ND, et al. Prevention and management of with cirrhosis. Gastroenterology 2011;141(5):1680–9.e1-2. Epub 2011, Jul 18.
gastroesophageal varices and variceal hemorrhage in cirrhosis. Am J 38. Shawcross DL, Wendon JA. The neurological manifestations of acute liver
Gastroenterol 2007;102:2086–102. failure. Neurochem Int 2011 Nov 2. Epub ahead of print.
12. Barnert J, Messmann H. Diagnosis and management of lower 39. Mpabanzi L, Jalan R. Neurological complications of acute liver failure:
gastrointestinal bleeding. Nat Rev Gastroenterol Hepatol 2009;6:637–46. pathophysiological basis of current management and emerging therapies.
13. Stravitz RT, Larsen FS. Therapeutic hypothermia for acute liver failure. Crit Neurochem Int 2011 Nov 2. Epub ahead of print.
Care Med 2009;37(7 Suppl):S258–64. 40. Banks PA, Freeman ML, the Practice Guidelines Committee of the American
14. Frontera JA, Kalb T. Neurological management of fulminant hepatic failure. College of Gastroenterology. Practice guidelines in acute pancreatitis. Am J
Neurocrit Care 2011;14(2):318–27. Gastroenterol 2006;101:2379–400.
15. Jiang W, Desjardins P, Butterworth RF. Direct evidence for central 41. Vege SS, Gardner TB, Chari ST, et al. Low mortality and high morbidity in
proinflammatory mechanisms in rats with experimental acute liver failure: severe acute pancreatitis without organ failure: a case for revising the Atlanta
protective effect of hypothermia. J Cereb Blood Flow Metab 2009;29(5): classification to include “moderately severe acute pancreatitis.” Am J
944–52. Epub 2009, Mar 4. Gastroenterol 2009;104(3):710–5. Epub 2009, Feb 3.
16. Stravitz RT, Lee WM, Kramer AH, et al. Therapeutic hypothermia for acute 42. Ball CG, Correa-Gallego C, Howard TJ, et al. Radiation dose from computed
liver failure: toward a randomized, controlled trial in patients with advanced tomography in patients with necrotizing pancreatitis: how much is too
hepatic encephalopathy. Neurocrit Care 2008;9(1):90–6. much? J Gastrointest Surg 2010;14(10):1529–35. Epub 2010, Sep 8.
17. Atterbury CE, Maddrey WC, Conn HO. Neomycin-sorbitol and lactulose in 43. Balthazar EJ, Ranson JHC, Naidich DP, et al. Acute pancreatitis: prognostic
the treatment of acute portal-systemic encephalopathy: a controlled, value of CT. Radiology 1985;156:767–72.
double-blind clinical trial. Am J Dig Dis 1978;23:398–406. 44. Balthazar EJ, Robinson DL, Megibow AJ, et al. Acute pancreatitis: value of
18. O’Grady JG, Alexander GJ, Hayllar KM, et al. Early indicators of prognosis CT in establishing prognosis. Radiology 1990;174:331–6.
in fulminant hepatic failure. Gastroenterology 1989;97:439–55. 45. Balthazar EJ. Acute pancreatitis: assessment of severity with clinical and CT
19. Weisner R, Edwards E, Freeman R, et al. Model for end-stage liver disease evaluation. Radiology 2002;223(3):603–13. Review.
(MELD) and allocation of donor livers. Gastroenterology 2003;124:91–6. 46. Bollen TL, Singh VK, Maurer R, et al. Comparative evaluation of the
20. Larson AM, Polson J, Fontana RJ, et al. Acetaminophen-induced acute liver modified CT severity index and CT severity index in assessing severity of
failure: results of a United States multicenter, prospective study. Hepatology acute pancreatitis. AJR Am J Roentgenol 2011;197(2):386–92.
2005;42:1364–72. 47. Malbrain ML, Chiumello D, Pelosi P, et al. Incidence and prognosis of
21. Craig DG, Reid TW, Wright EC, et al The sequential organ failure assessment intraabdominal hypertension in a mixed population of critically ill patients:
(SOFA) score is prognostically superior to the model for end-stage liver a multiple-center epidemiological study. Crit Care Med 2005;33:315–22.
disease (MELD) and MELD variants following paracetamol 48. Malbrain ML. Abdominal pressure in the critically ill: measurement and
(acetaminophen) overdose. Aliment Pharmacol Ther 2012;35(6):705–13. doi: clinical relevance. Intensive Care Med 1999;25:1453–8.
10.1111/j.1365-2036.2012.04996.x. Epub 2012, Jan 20. 49. Hedenstierna G, Larsson A. Influence of abdominal pressure on respiratory
22. Levesque E, Hoti E, Azoulay D, et al. Prospective evaluation of the prognostic and abdominal organ function. Curr Opin Crit Care 2012;18(1):80–5.
scores for cirrhotic patients admitted to an intensive care unit. J Hepatol Review.
2012;56(1):95–102. Epub 2011, Aug 9. 50. Malbrain MLNG, Cheatham ML, Kirkpatrick A, et al. Results from the
23. Cholongitas E, Senzolo M, Patch D, et al. Review article: scoring systems for international conference of experts on intra-abdominal hypertension and
assessing prognosis in critically ill adult cirrhotics. Aliment Pharmacol Ther compartment syndrome. I Definitions. Intensive Care Med 2006;32:1722–32.
2006 1;24(3):453–64. 51. Malbrain MLNG, Cheatham ML, Kirkpatrick A, et al. Results from the
24. Stravitz RT, Kramer AH, Davern T, et al. Intensive care of patients with acute international conference of experts on intra-abdominal hypertension and
liver failure: recommendations of the U.S. Acute Liver Failure Study Group. compartment syndrome. II. Recommendations. Intensive Care Med
Crit Care Med 2007;35:2498–508. 2007;33:951–62.
25. Findlay JY, Fix OK, Paugam-Burtz C, et al. Critical care of the end-stage liver 52. Sugrue M, Jones F, Janjua KJ, et al. Temporary abdominal closure: a
disease patient awaiting liver transplantation. Liver Transpl 2011;17(5): prospective evaluation of its effects on renal and respiratory physiology.
496–510. doi: 10.1002/lt.22269. J Trauma 1998;45:914–21.
26. Al-Khafaji A, Huang DT. Critical care management of patients with 53. Sugrue M, Jones F, Deane SA, et al. Intra-abdominal hypertension is an
end-stage liver disease. Crit Care Med 2011;39(5):1157–66. independent cause of postoperative renal impairment. Arch Surg
1999;134:1082–5.
234.e2 Section II—Clinical and Laboratory Assessment

54. Malbrain MLNG, Deeren DH, De Potter T, et al. Abdominal compartment 56. De Keulenaer BL, De Waele JJ, Malbrain ML. Nonoperative management of
syndrome following rectus sheath hematoma: bladder-to-gastric pressure intra-abdominal hypertension and abdominal compartment syndrome:
difference as a guide to treatment. ANZ J Surg 2005;75:A8. evolving concepts. Am Surg 2011;77(Suppl 1):S34–41.
55. Anand RJ, Ivatury RR. Surgical management of intra-abdominal 57. Ranson JH, Rifkind KM, Roses DF, et al. Prognostic signs and the role of
hypertension and abdominal compartment syndrome. Am Surg operative management in acute pancreatitis. Surgery, Gynecology &
2011;77(Suppl 1):S42–5. Obstetrics 1974;139(1):69–81.
III
Chapter
24  

Evoked Potentials
Emmanuel Carrera, Ronald G. Emerson, and Jan Claassen

Introduction brainstem, to the ventroposterior lateral thalamus, to the


Neuromonitoring can help determine the extent of neurologic primary somatosensory receiving cortex, and then to a wide
injury, can detect secondary injury at a potentially reversible network of cortical areas involved in somatosensory process-
stage, can help assess the effect of clinical care on an individual ing (secondary somatosensory cortex, posterior parietal
patient, and may aid in predicting outcome. This can be par- cortex, posterior and mid-insula and mid-cingulate cortex).
ticularly important in the neurocritical care unit (NCCU), Median and tibial nerves are most often stimulated in SSEP
where the neurologic examination may be difficult because of testing, although others (e.g., ulnar, peroneal) may be used
depressed consciousness, hypothermia, or use of sedatives. when appropriate.
Somatosensory evoked potentials (SSEPs) have been used in
clinical practice since they were first described in the late 1940s
in patients with myoclonus,1 and after electroencephalogra- Stimulation
phy (EEG) are the most frequently used neurophysiologic tool The stimulus for SSEPs is a brief electrical pulse delivered by
in the NCCU. In general, SSEPs are less affected by sedation a pair of electrodes placed on the skin above the nerve. The
or hypothermia than EEG.2 Brainstem auditory evoked poten- median nerve is stimulated at the lateral-anterior wrist; the
tials (BAEPs) have been used since the 1960s to study nonin- tibial nerve is stimulated at the ankle, with the cathode
vasively and objectively the function of the brainstem auditory between the Achilles tendon and the medial malleolus. To
pathways.3 Other tools such as motor evoked potentials and reduce artifact produced by electrical stimulation, a ground
visual evoked potentials are less frequently used in the NCCU. electrode is placed between the stimulation site and the
Use of evoked potential (EP) monitoring in neurocritical care recording site. The stimulus is delivered as a constant mono-
practice varies regionally and even between intensive care phasic square-wave pulse that lasts approximately 100 to
units (ICUs), and the ready availability of neuroimaging has 200 msec, at a rate of typically 4 per second. The intensity of
limited the application of EPs in many ICUs. However, the the stimulation is two to four times the sensory threshold as
diagnostic and prognostic value of EPs is embraced by national evidenced by the contraction of the muscle innervated by the
and international organizations.4 Advantages of EPs include: stimulated nerve.
(1) they are noninvasive, (2) they may be used serially, (3) they
give objective quantitative values that can be followed over
time and compared between patients, (4) they are relatively Recording
stable in the presence of mild hypothermia and relatively resis- Both standard surface disk electrodes and needle electrodes
tant to many commonly used sedatives, and (5) they are inex- can be used. Recording electrodes for upper limb SSEPs are
pensive.5 In addition, modern invasive neuromonitoring and placed at the clavicle between the heads of the sternocleido-
treatment devices such as intravascular cooling catheters can mastoid muscles (Erb’s point) and on the skin overlying cer-
limit patient transport or use of magnetic resonance imaging vical bodies 6 to 7. On the scalp, electrodes are placed at
(MRI). In the absence of portable imaging devices, diagnostic CP3 and CP4 (respectively midway between C3 and C4) of
tools such as EPs that can be brought to the patient’s bedside the international 10-20 system. Bipolar recording between
provide an alternative diagnostic tool. The technical feasibility CP3 and CP4 detects the cortical component of the SSEP;
of continuous EP monitoring along with continuous EEG referential recording, to earlobe or noncephalic locations
monitoring has been demonstrated.6 (e.g., the shoulder), is used to detect subcortial (brainstem)
components. For tibial SSEPs, electrodes are placed in the
popliteal fossa (peripheral component) and over the lumbar
Engineering and Technology vertebra (lumbar component). At least two bipolar channels
(e.g., CPz-Fpz and CP3-CP4) (see Chapter 25 for a review of
Somtatosensory Evoked Potentials EEG montage nomenclature) should be used to record the
SSEP testing assesses the integrity of the dorsal column– cortical component; as with the median SSEPs, referential
lemniscal system.5 This pathway projects via the dorsal column scalp recording is used to detect subcortical components.
of the spinal cord to the cuneate nucleus in the lower The SSEP waveform is obtained by averaging typically from
236 © Copyright 2013 Elsevier Inc. All rights reserved.
Section III—Electrophysiology 237

500 to 2000 stimuli. It is is necessary to repeat at least two


independent averages to demonstrate reproducibility. For Table 24.1  Normal Values for Evoked
clinical purposes, analysis times of 50 msec (median nerve) Potentials from Healthy Volunteers
or 100 msec (tibial nerve) are used. Latency, Latency,
Mean Upper Limit
Recording Site (msec) (msec)
Filters
MEDIAN SSEP
SSEPs are recorded using a broad pass-band with high-pass
N9 Erb’s point 9.8 11.5
and low-pass filters set typically to 30 Hz and 2000 Hz,
respectively. Notch filters to eliminate power line noise (50 or N13 Cervical spine (C7) 13.3 14.5
60 Hz) should be used with caution because of their ten- N20 Contralateral cortex 19.8 23.0
dency to create “ringing” oscillatory artifact. Digital postpro- (CP3 or CP4)
cessing of the recorded signal may be appropriate in specific INTERVALS MEDIAN
situations. CCT (P14-N20) — 5.6 6.6
Tibial SSEP
Brainstem Auditory Evoked Potentials N8 Popliteal fossa 8.5 10.5
BAEPs are generated using brief auditory stimuli that activate N22 Lumbar spine (L1) 21.8 25.2
the cochlea, the auditory nerve, and then the brainstem audi-
P30 Fz-Cv7 29.2 34.7
tory pathways.7
P39 Cz-Fz 38.0 43.9
INTERVALS TIBIAL
Stimulation
N22-P30 — 7.4 10.2
The stimulus used to generate a BAEP consists of a brief (less
P30-P39 — 8.7 13.4
than 200 μsec) “click,” typically delived at a rate of approxi-
mately 10 Hz. The stimuli are presented to one ear at a time CCT, Central conduction time; SSEP, somatosensory evoked potential.
using headphones or ear inserts while the nonstimualted ear
is “masked” with white hissing noise to prevent stimulation by
sound conducted through the cranium.

indentified initially based on the effects of clinical lesions and


Recording direct recording from intracranial structures; more recently,
Either standard surface electrodes or needle electrodes may be source modeling based on three-dimensional MRI has been
used. BAEPs are typically recorded using a two-channel used to revisit these generators.8
montage: ipsilateral ear to vertex (channel 1) and contralateral Clinical interpretation is based primarily on the analysis of
ear to vertex (channel 2). Averaging of 2000 to 4000 stimuli is “short latency” SSEP signals, up to and including those cor-
typically required; at least two independent averages must be responding to the activation of the somatosensory receiving
recorded to prove reproducibility of waveforms. cortex (Table 24.1). The N9 is a compound action potential
reflecting the peripheral afferent volley and is used as a refer-
ence point from which to calculate conduction time for
Filters the centrally recorded potentials. Peripheral neuropathy may
BAEPs are recorded using a broad pass-band, with the high- increase the latency and decrease the amplitude of this wave-
pass filter typically set to 100 Hz or 150 Hz and the low-pass form. N13 is generated by dorsal horn cells of the cervical
filter set to 3000 Hz. spinal cord. P14 is generated in the caudal medial lemniscus
within the lower medulla, with possible additional presynaptic
contribution from the upper cervical cord. N18 is attributed
Information Provided and What to postsynaptic activity in multiple brainstem gray matter
the Information Means structures. N20 reflects activation of the primary somatosen-
sory receiving area, located in the posterior bank of the
Somtatosensory Evoked Potentials rolandic fissure in Brodmann’s area 3b.9,10 Although actually
SSEP recordings consist of signals recorded from different measuring a composite of white and gray matter events in the
sites along the afferent sensory pathways. By convention, nega- large fiber sensory system between the cervicomedullary junc-
tive singals are displayed as upward deflections, and positive tion and somatosensory cortex, the P14 to N20 interpeak
signals downward. Physiologic signals are recorded with dif- latency (calculated by subtracting the P14 peak latency
ferential amplifiers, devices that amplify the difference between from the N20 peak latency) is commonly referred to as the
two inputs. A negative signal in the first, or “noninverting” central conduction time (CCT).5 Absent or delayed central
input produces an upward inflection; a negative signal in the conduction with present peripheral conduction may permit
second, or “inverting” input produces a downward inflection. lesions to be localized by SSEPs.
The time between stimulation and the recorded potential is Principal components of the tibial SSEP include the N8
called the latency, and is usually expressed in milliseconds. A recorded from the popliteal fossa, the N22 generated by the
normal median SSEP waveform is shown in Figure 24.1. dorsal gray matter of the lumbosacral cord, the P30 generated
Generator sources for specific components of the SSEP were at the cervicomedullary junction, and the P39 generated in the
238 Section III—Electrophysiology

N20

N18

P14

5 msec

Fig. 24.1  Normal median nerve somatosensory evoked potential (SSEP) recording. The four anatomical cross-sections (from bottom to top
of illustration) are as follows: cervical spinal cord, brainstem, midbrain, and brain (cerebral cortex, coronal section).

somatosensory cortex.5 In contrast to the median nerve SSEP, I III


the P39 cortical signal9 is positive in polarity and variably
maximal in amplitude either near the vertex or over the cen-
troparietal scalp ipsilateral to the stimulated leg. This interin- V
dividual variability in the scalp topography of P39, due to the A2-Cz
variations in the normal location of the leg and foot area
within the interhemispheric fissure, necessitates use of at least 0.2 uV
two bipolar scalp-scalp channels, one in the midline and
another using laterally placed electrodes, to ensure reliable 1 msec
detection.
Middle- and long-latency SSEP components are not com- A1-Cz
monly used clinically in the ICU because they are less reliable
and more susceptible than short-latency SSEPs to the effects
of medication and to depressed levels of consciousness.5

Fig. 24.2  Normal brainstem auditory evoked potential (BAEP) bilaterally


Brainstem Auditory Evoked Potentials (click 90 db, noise 60 db; recorded from the right ear) in a 49-year-old
Clinical interpretation of the BAEP is based on signals gener- woman with a right frontal brain tumor.
ated within approximately 7 milliseconds following the stim-
ulus (Fig. 24.2). Studies suggest that BAEPs have multiple
generators, with nonsequential interaction between second-
and third-order ipsilateral and contralateral neuronal cells nucleus or axons of the lateral lemniscus (wave IV), and the
and axons. Although the exact identity of short-latency BAEP inferior colliculus and ventral lateral lemniscus (wave V).11
waveforms remains somewhat uncertain, commonly recog- Because waves II and IV are less reliably recorded across indi-
nized generators include the distal auditory nerve (wave I), viduals, clinical interpretation is based primarily on assess-
the auditory nerve as it exits the porus acousticus or the ment of waves I, III, and V (see Fig. 24.2).
cochlear nucleus (wave II), the cochlear nucleus or ipsilateral BAEPs are resistant to the effects of sedative medication and
superior olivary nucleus (wave III), the superior olivary to a patient’s level of consciousness depression, adding to their
Section III—Electrophysiology 239

utility in the ICU setting. Although the amplitude and latency


of these waves are affected substantially by click stimulus
Postanoxic Coma (Hypoxic-Ischemic
intensity, the I to V interpeak latency (as well as I to III and Encephalopathy)
III to V interpeak latencies) is relatively unaffected by intensity After cardiopulmonary resuscitation, the neurologic examina-
changes and interpeak latency and is therefore relied on for tion does not reliably predict the presence or absence of
clinical interpretion. specific SSEP patterns, particularly absent or present N20
Delays in wave V with a normal wave I latency suggest responses.16 Nevertheless, EPs can help predict outcome,
a conduction abnormality somewhere central to the distal particularly poor outcome rather than good outcome, after
portion of cranial nerve VIII. Absence of wave I with pre- cardiac arrest. Normal SSEP responses have less predictive
served wave V could reflect a problem with the peripheral power than abnormal or absent responses. It does not appear
hearing apparatus or with the auditory nerve, but commonly that induced hypothermia after cardiac arrest (e.g., 33° C for
reflects technical difficulty recording wave I. Functional dis- 24 hours) affects the predictive power of SSEPs, although
ruption of the brainstem may cause loss of waves II to V with withdrawal of care may bias these observations.17,18 In particu-
preservation of wave I.12 Unilaterally abnormal BAEPs most lar, bilaterally absent cortical SSEP responses 3 days after
often reflect ipsilateral brainstem damage. resuscitation are associated with a poor prognosis.19-30 Short-
More rostral structures, including generators within the latency SSEPs (N20) may be the most reliable method to
primary auditory cortex and surrounding areas, are respon- predict poor outcome in anoxic-ischemic encephalopathy.
sible for longer latency components.13 However, as with SSEPs, This is included in practice parameters on outcome prediction
the later components of the BAEP are affected by conscious- in comatose patients after cardiopulmonary resuscitation
ness state and attention,14 and are less commonly used clini- from the Quality Standards Subcommittee of the American
cally in the ICU setting. Academy of Neurology and regarded as Level II evidence (i.e.,
probably disease specific).27 For example, in a prospective
study of 407 cardiac arrest patients, Zandbergen et al. observed
Indications for Use bilaterally absent N20s in 45% of patients that were comatose
In the ICU, EPs are primarily used for prognostication, par- at 72 hours; all these had poor outcome19 (Fig. 24.3). In a
ticularly in patients after cardiac arrest and those with trau- subsequent systematic literature review that included 4500
matic brain injury (TBI). Diagnostically, EPs have largely been anoxic patients, Zandbergen et al. observed that bilaterally
replaced by imaging studies (see Chapters 26 and 28), invasive absent N20s within the first week were 100% specific for
neuromonitoring (e.g., brain oxygen monitoring), and con- poor outcome.27 However, among those patients with poor
tinuous EEG (see Chapter 25). However, serial and even con- outcome, 46% may have preserved N20 responses within the
tinuous EP monitoring is feasible2,6 and may be considered first week of cardiac arrest28 (Fig. 24.4). SSEPs perform favor-
when continuous noninvasive neuromonitoring is warranted ably when compared with other outcome predictors such as
and EEG may be of limited value, such as in evolving spinal the pupillary light response, the Glasgow Coma Scale (GCS),
cord or brainstem injury. In addition EPs may be used to or EEG.26 Combining SSEPs with other outcome predictors
complement or supplement the information provided by such as EEG or serum markers of neuronal injury provides
imaging or other monitors. Consensus recommendations to high prognostic accuracy.24-26,31 Although SSEPs may allow the
guide optimal use of electrophysiologic tests including SSEP clinician to predict poor prognosis with some degree of accu-
for diagnosis, follow-up, and prognosis in the ICU have been racy, good outcome is much more difficult to predict. Discrep-
published.15 ancies between SSEP findings and outcome (i.e., recovery of

CP4-CP3

CP3-CP4
CP3-chin
CP4-chin

SC5-FPZ SC5-FPZ

0.7 uV

5 msec
Right Erbs-chin

0.7 uV
Left Erbs-chin
A 5 msec B
Fig. 24.3  Somatosensory evoked potentials (SSEPs) after cardiac arrest in a 65-year-old-man with about 15 minutes of pulseless electrical
activity who underwent hypothermia. The tracings show SSEPs recorded after (A) right and (B) left median nerve stimulation. There are bilaterally
present peripheral and cervical and subcortical median nerve SSEP potentials and absent cortical responses.
240 Section III—Electrophysiology

C4'-C3' C3'-C4'

C3'-right Erbs C4'-left Erbs

0.7 uV
0.7 uV
5 msec
5 msec
Left-right Erbs Right-left Erbs

A B
Fig. 24.4  Somatosensory evoked potentials (SSEPs) in a 59-year-old man with multiple cardiac arrests and possible hypoxic brain injury.
Right (A) and left (B) median nerve SSEPs show present cortical responses and mildly prolonged central conduction times bilaterally.

SSEPs but absence of clinical improvement)19,32 may be focal lesions, have subdural or extradural fluid collections, or
explained by hypoxic injury sparing the somatosensory tracts, have undergone decompressive craniotomy in the 48 hours
or secondary brain injury acquired after the SSEPs were before the recording. Overall, SSEPs perform favorably when
obtained (i.e., recurring cardiac arrests or cardiac output compared with other TBI outcome predictors such as the
failure). GCS, pupillary or motor responses, computed tomography
Several other SSEP parameters including the CCT, the N20- (CT), and EEG findings,42 and the the false-positive rate of
to-P25 amplitude ratio,26,33 and the N70 latency34 are associ- bilaterally absent SSEPs after TBI is less than 0.5%.
ated with outcome but are less robust than absent cortical Repeated SSEPs can help detect secondary injury (e.g.,
SSEP responses. When the N20s are present or questionable, hematoma enlargement or increased intracranial presure
late SSEP components such as the N70 (absent or delayed [ICP]), brainstem herniation, or cerebral ischemia.38,45,46 In
>130 msec) can help identify patients likely to have a poor some patients a change in SSEPs may precede an ICP increase.46
outcome.34 However, false positives are common (between 4% Serial SSEPs also may suggest recovery6,43 (Fig. 24.6). For
and 15%) and make it impractical to base treatment decisions example, reduction of latency and normalization of amplitude
on this finding. Late components may be better associated may occur earlier than recovery in the clinical examination.43
with long-term cognitive outcome than short-latency compo- Poor long-term functional outcome (death or vegetative state
nents35 but are not routinely used for patients with cardiac at 2 years) is seen in comatose patients with traumatic brain-
arrest. The role of BAEPs after cardiac arrest is less well studied. stem lesions and absent N20s that do not recover within 48
However, middle-latency auditory evoked responses are absent hours of the injury. However, recovery of the N20 does not
in patients who die or remain in a persistent vegetative state.22 necessarily mean recovery of brain function.47 Prolongation
Finally, EPs can be used to guide care in neonatal encepha- or absent CCT is associated with poor 1-year cognitive and
lopathy or childhood asphyxia.36 behavioral function.48
Although rare, reappearance of previously absent bilateral
SSEPs may occur after TBI. This is described in some patients
Traumatic Brain Injury with good outcome after infratentorial hemorrhage or follow-
Following TBI, SSEPs can help predict short-term mortality ing hemicraniectomy.49-51 In addition, circumscribed bilateral
and long-term outcome.34,35,37-44 Bilaterally absent cortical sensory tract contusions in the brainstem may mimic bilateral
SSEP responses with preserved peripheral and spinal poten- SSEP loss from supratentorial injury. This may not necessarily
tials are associated with poor outcome.37-40 In general, SSEPs mean a fatal prognosis.50 Finally, marked attenuation of the
more accurately predict poor outcome than good outcome.41 cortical N20-P25 must be distinguished from persistent loss
For example, in a review of 44 studies, Carter and Butt41 of these signals.43 BAEPs are used less frequently than SSEPs
observed that 71% of patients with normal SSEPs (N = 553) for TBI outcome prediction,52-54 but when performed, BAEP
have a favorable outcome (normal or moderate disability; abnormalities are more frequently seen in TBI patients with
Fig. 24.5). By contrast when SSEPs are absent bilaterally (n = poor outcome.52,55
777), 99% have an unfavorable outcome (severe disability,
vegetative state, or death). The positive predictive value and
sensitivity are 71% and 59%, respectively, for normal SSEP Acute Ischemic Stroke
and a favorable outcome; and 99% and 46.2%, respectively, Diagnosis of acute ischemic stroke (AIS), is done by neuro-
for bilaterally absent SSEP and an unfavorable outcome. logic examination and neuroimaging. EPs, however, may be
Patients who have a favorable outcome despite bilaterally used in a patient with infratentorial ischemia if MRI is not
absent SSEPs (N = 12) are more commonly children, have available or in a patient with a pacemaker who cannot have
Section III—Electrophysiology 241

A B

C3-C4
C4-C3
C4-left Erbs

C4-right Erbs C3-left Erbs

C3-right Erbs
0.5 uV CS6-left Erbs

5 msec
CS5-right Erbs
Right-left Erbs

0.5 uV
Left-right Erbs
5 msec
C D
Fig. 24.5  Somatosensory evoked potential (SSEPs) in a comatose 76-year-old man with a traumatic right subdural hematoma (A) and contralateral
parietal contusion (B). Symmetric bilateral median nerve cortical and subcortial SSEPs are present. The left median nerve SSEP (C) (Erb’s 11 msec, P14
15 msec, N20 22 msec) and the right median nerve SSEP (D) (Erb’s 11 msec, P14 16 msec, N20 23 msec) are present.

an MRI, and can be used for continuous monitoring in select infarction,62 and those with basilar thrombosis63 and can be
patients at risk for herniation (e.g., a large cerebellar stroke). particularly helpful when making decisions about prognosis
Ischemia of the pontine tegmentum results in both BAEP (loss when consciousness is impaired. Good outcome (moderate
of wave V) and SSEP abnormalities (absent N20). Ischemia disability or better) is more likely when BAEPs and SSEPs are
restricted to the cerebellar peduncles only affects the BAEPs.56 normal.63 EP abnormalities are of indeterminate predictive
False-negative EP results may occur—brainstem strokes may value in patients with locked-in syndrome.60 Cerebellar
have normal SSEPs with medial or lateral strokes in up to 60% infarcts associated with coma may be accompanied by prolon-
and 75% patients, respectively.57 EPs may be useful in patients gation of the I to V interpeak latencies,64 although this abnor-
with locked-in syndrome to provide objective evidence of mality may persist despite clinical improvement.
brainstem involvement although there is no specific pattern There are conflicting data on the association between EP
to the abnormalities,58,59 and depending on the affected struc- findings and outcome after supratentorial AIS. On the one
tures, BAEP and SSEP findings may vary from unilaterally hand, normal BAEPs but not SSEPs obtained before a hemi-
normal to bilaterally absent.58-60 Finally, BAEP obtained within craniectomy are associated with survival and good functional
24 hours of large middle cerebral artery infarction may help outcome (Barthel Index ≥60) after large hemispheric strokes.65
identify patients at risk for malignant edema61 and thus the On the other hand, SSEP findings after small internal capsule
need for a hemicraniectomy. or corona radiate strokes66 do not correlate with 3-month
The role of EPs in outcome prediction after AIS is less well outcome (Barthel Index). Others have found that serial SSEPs
studied than TBI or anoxia and is mainly limited to case series and BAEPs are associated with 30-day Glasgow Outcome
of patients with posterior fossa or massive hemispheric infarc- Scale independent of stroke type and location, with the
tion. Bilateral BAEP and SSEP abnormalities are seen in exception that BAEPs are not associated with outcome in
patients with poor outcome after pontine and mesencephalic those with infratentorial lesions.67 In part these differences in
242 Section III—Electrophysiology

Vertebra 7 Left cortical response Vertebra 7 Right cortical response

Day 1 Day 1
N13 N13
N20

1 µV
1 µV P25

Day 10 Day 10
N13 N13
N20

P25
N20 P25

Day 304 Day 304


N13 N13

N20 N20

P25

10 20 30 40 msec 10 20 30 40 msec 10 20 30 40 msec 10 20 30 40 msec


Fig. 24.6  Recovery of somatosensory evoked potential (SSEP) after traumatic brain injury (TBI). This 14-year-old boy was admitted in coma
(Glasgow Coma Scale score 3). He developed elevated intracranial pressure, and a clinical improvement was first seen on day 13 after the injury. He
did not move his left side purposefully until day 26. The initial median nerve SSEP is abnormal. Left cortical recordings were absent, and right
hemispheric responses were prolonged with a severely decreased amplitude. For the left cortical projections, the first indication of recovery was
observed with SSEPs on day 10. Over the next few days, these SSEP findings improved. Twenty-three days after injury, latencies of the right cortical
projection were within normal limits, whereas amplitudes remained reduced. One year after the accident, he had a minimal residual speech impairment
but had returned to his former activities. (Reprinted with permission from Claassen J, Hansen HC. Early recovery after closed traumatic head injury: somatosensory
evoked potentials and clinical findings. Crit Care Med 2001;29:494–502.)

results may stem from differences in study design and small ICH in the putamen or thalamus generally parallels the nor-
sample size. malization of SSEP components.69
Several EP observations are associated with stroke location. SSEPs and BAEPs can provide prognostic information in
AIS of the ventroposterior thalamus leads to an absent patients with an ICH located in the brainstem, particularly in
response, decrease in amplitude, delay in peak latency, or the pons70,71 or in deep supratentorial structures including the
attenuation of median N20-P25 and tibial P40 components.68 putamen72,73 and thalamus.72 In particular, prolongation of I
Lateral ventroposterior lesions preferentially affect tibial to V interpeak latencies in the BAEP and bilaterally absent
SSEPs, whereas medial lesions affect median SSEPs. Strokes N20s in the SSEP are are associated with poor prognosis.70,74
that affect the thalamocortical pathways lead to less consistent SSEPs may have a lower false-negative rate for poor outcome
SSEP abnormalities. Although large strokes of the corona than BAEPs after putaminal ICHs.73 Good functional outcome
radiate may lead to similar SSEP abnormalities as thalamic is likely when BAEPs are normal and SSEPs are normal after
strokes,68 smaller lesions of the corona radiate or internal putaminal ICH.72 Following a pontine ICH, good outcome
capsule often have an inconsistent relationship between EP may be seen if normal amplitudes and latencies of waves I to
findings and sensory symptoms.66 This inconsistency is likely V in BAEPs are at least unilaterally present.70
from incomplete involvement of the thalamocortical pathways
in small strokes.
Subarachnoid Hemorrhage
There is an unclear relationship between the severity of sub-
Intracerebral Hemorrhage arachnoid hemorrhage (SAH) and EP findings; some studies
Evoked potentials (EPs) have little if any role in the diagno- suggest an association between CCT prolongation75 or absence
sis of an intracerebral hemorrhage (ICH). In addition EP of SSEP and BAEP responses76 and the Hunt Hess grade,
localization value is lim­ited after thalamic or putaminal whereas others found no association between CCT abnor-
ICH.69 However, BAEP abnormalities may provide informa- malities and clinical severity.77,78 Xenon single photon emis-
tion about small posterior circulation bleeds, particularly sion computed tomography (SPECT)79,80 studies suggest CCT
when tegmental structures are affected.70 Serial SSEPs may prolongation is associated with impaired cerebral blood flow
provide an alternative to frequent brain imaging to examine (CBF) after SAH, particularly when CBF is less than
potential for neurologic recovery, particularly for ICHs in 30 mL/100 g/min.81 However, EP studies do not have a role in
the posterior circulation. Recovery of motor function after vasospasm diagnosis in clinical practice.
Section III—Electrophysiology 243

Studies in the 1970s did not find an association between EP an ICP monitor (i.e., determining the consequence of an ICP
findings and outcome.82 Subsequent studies describe an asso- increase).46
ciation between BAEP and SSEP abnormalities including
bilateral CCT prolongation (>6 msec) or bilaterally absent
cortical responses and poor outcome or death after SAH.77,83,84
Outside the Intensive Care Unit
Tibial rather than median nerve SSEP may be more robust in A large body of literature describes the use of intraoperative
outcome prediction.84 Patients with normal SSEPs have the neurophysiologic monitoring including use of SSEPs, BAEPs,
potential for a good outcome.77 These differences may be asso- transcranial motor evoked potentials (tcMEPs), and direct
ciated with advances in SAH management particularly for electromygraphy, and evidence-based guidelines were pub-
poor-grade SAH patients. lished on the use of intraoperative monitoring.98 The reader
is referred to reviews on the topic.99-101 SSEPs can: (1) help
identify cerebral ischemia during cerebrovascular surgery
Spinal Cord Injury including carotid endarterectomy or aneurysm surgery,102 or
EPs have an established role during spinal surgery but are not during repair of the thoracic aorta,103 (2) identify spinal cord
frequently used to monitor patients with spinal cord injury injury during spine surgery,104 or (3) help guide resection of
(SCI) in the ICU.85 Although EP abnormalities after SCI, epileptic foci,105 tumors,106 or arteriovenous malformations
including spinal cord ischemia, are associated with long-term (AVMs)107 in select cortical areas or intramedullary spinal
outcome,86-88 they do not appear to add any prognostic accu- lesions,108 whereas BAEPs can be useful during posterior fossa
racy to the neurologic examination but can be used to supple- surgery.109 In general, neurologic abnormalities such as para-
ment it.86,89,90 SSEPs may have a role in injury assessment for plegia after spinal surgery never or very rarely occur if there
comatose patients with suspected SCI.88 In these patients the are no changes in EPs during intraoperative monitoring. EPs
extent of neurologic deficits is associated with the degree of also are used in studies of cognition and behavior and to
SSEP abnormalities: N9 and N20 latencies for the upper limbs evaluate conditions such as multiple sclerosis, Friedreich’s
and P40 and P60 for the lower limbs.91 ataxia, and Alzheimer’s disease.

Brain Death How to Use Evoked Potential


There is no single method to confirm brain death, the diag- Monitoring, and Troubleshooting
nosis of which depends in large part on the clinical examina-
tion (see Chapter 13) and strict protocols. However, Wijdicks Electrical Artifact
in a review conducted a decade ago, found there was a lack of Electrical artifact (“noise”) may frequently interfere with EP
consensus on the exact criteria in many published practice recordings in the ICU setting. This is of particular concern in
parameters and guidelines and that 28 of 70 (40%) of the SSEP recordings because notch filters are discouraged. The
practice guidelines he studied mandated the use of confirma- noise level influences the accuracy of the recording and thus
tory testing. Many of these recommended use of EPs.4 When interobserver agreement. This can limit the use of SSEPs in
repeat studies are performed, patients who progress to brain prognosis decisions.110 Therefore every effort should be made
death lose subcortical SSEP and all BAEP responses.12,92 Loss to minimize the artifact level (<0.25 microV). If this is not
of the median nerve SSEP noncephalic P14 and of its cephalic possible, studies should be interpreted with caution. Neuro-
referenced reflection N14 and the N18 is seen in brain muscular junction blockade also may be considered when
death.92,93 No specific BAEP pattern is seen in patients who noise is excessive.111
progress to brain death.94,95 Diagnostic accuracy is improved
by combining BAEPs and SSEPs.96 EPs can also be helpful in
patients when there is doubt about the clinical findings or Medication Effects
cardiopulmonary instability limits the ability to perform an Subcortical EP components are relatively unaffected by most
apnea test. medications that are used in the ICU. By contrast, cortical EP
components are more susceptible to medication effects. Seda-
tive medications, including benzodiazepines, propofol, barbi-
Coma Associated with Metabolic turates, nitrous oxide, and halogenated inhalational agents all
or Toxic Encephalopathy depress the cortical components of EPs in a dose-related
The International Society for Hepatic Encephalopathy and manner.112-115 However, in these circumstances cortical EP
Nitrogen Metabolism (ISHEN) published guidelines for the components are more robust than the EEG, and consequently
use of neurophysiologic investigations in hepatic encepha- SSEPs may be used to monitor patients with barbiturate-
lopathy.97 There are three main recommendations. First, the induced coma and an isoelectric EEG.114
choice of neurophysiologic test depends in part on the severity A severe overdose with central nervous system (CNS)
of encephalopathy. Quantitative EEG may be used for mild depressants (e.g., amitriptyline, barbiturates, meprobamate,
forms, but SSEPs should be considered for more severe or nitrazepam) may affect SSEP and BAEP latencies. These
encephalopathy. Second, when N20 responses are bilaterally abnormalities may include CCT prolongation and N20 dis-
absent, further evaluation is recommended to determine the persion of the SSEP, and interpeak delays of the BAEP.35
extent of irreversible brain injury before liver transplantation. Pheny­toin may increase latencies and decrease amplitudes of
Third, SSEPs and BAEPs may be used to help make decisions brainstem BAEP components and abnormal BAEPs, includ-
about ICP and its management, for example, when an ICP ing increasing latency of wave I, and I to V and I to III inter-
monitor is not feasible or to complement the information of peak latencies may be observed in up to a third of patients
244 Section III—Electrophysiology

taking phenytoin, particularly with supratherapeutic phenyt-


oin levels.116 In phenytoin intoxication, all waves except BAEP
Conclusion
waves I and V may be lost.117 Clinical neurophysiology including SSEPs and BAEPs can be
useful in the NCCU for diagnosis (e.g., brain death or neuro-
muscular disorders), for follow-up, and as a guide to manage-
Hypothermia-Induced Changes ment (e.g., hepatic encephalopathy), and in outcome prediction
Therapeutic hypothermia is increasingly used in the ICU, and (e.g., anoxic-ischemic encephalopathy) after cardiac arrest,
so the effects of temperature on EP studies require careful TBI, and toxic and metabolic encephalopathy. In general, the
consideration. Much of the knowledge about EPs during information provided by EPs is better able to predict poor
hypothermia comes from its use during cardiac and neurovas- outcome than good outcome, and the significance of the find-
cular surgery. However, reports are variable and depend in ings depends in part on the patient’s pathology.
part on what was measured and at what temperature. There is
a linear relationship between decreasing temperature and
increasing cortical and peripheral SSEP latencies (N10, P14,
Acknowledgments
and N19).118 The N19 is consistently recordable at tempera- We thank Noeleen Ostapkovich for her careful review of this manuscript
tures greater than 26° C, usually disappearing between 20° C and Edward Gallo, REEGT, for help with reproducing figures.
and 25° C. In general the more peripherally generated compo-
nents, N10 and P14, are more robust to the effect of hypother- References
mia. P14 is elicitable at more than 21° C, whereas N10 persists 1. Dawson G. Investigations on a patient subject to myoclonic seizure after
even when the temperature is less than 20° C. Subcortical stimulation. J Neurol Neurosurg Psychiatry 1947;10:141–62.
SSEP components (P13 and N14) become more consistently 2. Moulton RJ, Brown JI, Konasiewicz SJ. Monitoring severe head injury: a
recognized at profound hypothermia (<20° C) compared with comparison of EEG and somatosensory evoked potentials. Can J Neurol Sci
normothermia, whereas cortical components disappear.119 1998;25:S7–11.
3. Celesia G, Puletti F. Auditory cortical areas of man. Neurology
Hypothermia increases BAEP latency of wave I interpeak 1969;19:211–20.
intervals.113 There is a linear increase of latencies and inter- 4. Wijdicks EF. Brain death worldwide: accepted fact but no global consensus
peak intervals by approximately 7% per 1° C decrease in tem- in diagnostic criteria. Neurology 2002;58:20–5.
perature.120 At 26° C, BAEP values are approximately doubled. 5. Cruccu G, Aminoff MJ, Curio G, et al. Recommendations for the clinical use
of somatosensory-evoked potentials. Clin Neurophysiol 2008;119:1705–19.
These temperature-induced changes are reversible with 6. Fossi S, Amantini A, Grippo A, et al. Continuous EEG-SEP monitoring of
rewarming, but during the rewarming phase latencies may severely brain injured patients in NICU: methods and feasibility.
differ compared with the cooling phase (“hysteresis”).118 BAEP Neurophysiol Clin 2006;36:195–205.
amplitudes may initially increase when temperature is between 7. Celesia GC, Brigell MG. Auditory evoked potentials. In: Niedermeyer E,
25° C and 30° C and then decrease at lower temperatures.120-122 Lopes da Silva F, editors. Electroencephalography: basic principles, clinical
applications, and related fields. Baltimore: Lippincott Williams & Wilkins;
Longer latency components may disappear with moderate 2005. p. 1137–64.
degrees of hypothermia, but ultimately all components disap- 8. Mauguiere F. Somatosensory evoked potentials: normal responses, abnormal
pear when temperature is less than 14° C to 20° C.120,123 waveforms, and clinical applications in neurological diseases. In:
Induced hypothermia is commonly used after cardiac Niedermeyer E, Lopes da Silva F, editors. Electroencephalography: basic
applications, and related fields. Baltimore: Lippincott Williams & Wilkins;
arrest. When SSEP and BAEPs are studied between 24 and 28 2005. p. 1067–119.
hours after the arrest and hypothermia (33° C),17 established 9. Goff GD, Matsumiya Y, Allison T, et al. The scalp topography of human
EP parameters associated with outcome (i.e., poor outcome somatosensory and auditory evoked potentials. Electroencephalogr Clin
with bilaterally absent N20 in SSEPs) remain reliable. BAEPs Neurophysiol 1977;42:57–76.
had no additional value in outcome prediction. 10. Desmedt JE, Ozaki I. SEPs to finger joint input lack the N20-P20 response
that is evoked by tactile inputs: contrast between cortical generators in areas
3b and 2 in humans. Electroencephalogr Clin Neurophysiol 1991;80:513–21.
Risks 11. Scherg M, von Cramon D. A new interpretation of the generators of BAEP
waves I-V: results of a spatio-temporal dipole model. Electroencephalogr
BAEP and SSEP studies are safe. However, several ICU- Clin Neurophysiol 1985;62:290–9.
12. Starr A. Auditory brain-stem responses in brain death. Brain
specific considerations about electrical safety should be kept 1976;99:543–54.
in mind.124 In particular, ICU patients may be at increased 13. Yvert B, Crouzeix A, Bertrand O, et al. Multiple supratemporal sources of
risk for electrical injury, in particular cardiac arrhythmias. magnetic and electric auditory evoked middle latency components in
Fluid paths, especially central lines, may allow currents to humans. Cereb Cortex 2001;11:411–23.
enter a patient’s body. Weak or comatose patients are unable 14. Linden DE. The p300: where in the brain is it produced and what does it tell
us? Neuroscientist 2005;11:563–76.
to withdraw from current sources. To reduce these risks, all 15. Guerit JM, Amantini A, Amodio P, et al. Consensus on the use
electrical equipment used in the patient area should be the of neurophysiological tests in the intensive care unit (ICU):
three-wire grounded type with “hospital grade” plugs. Equip- electroencephalogram (EEG), evoked potentials (EPs), and
ment should be tested on a regular schedule for excessive electroneuromyography (ENMG). Neurophysiol Clin 2009;39:71–83.
16. Bouwes A, Binnekade JM, Verbaan BW, et al. Predictive value of neurological
current leakage and electrical outlets tested for integrity of the examination for early cortical responses to somatosensory evoked potentials
ground connection. Devices connected to one patient should in patients with postanoxic coma. J Neurol 2012;259(3):537–41. Epub 2011,
be plugged into the same group of outlets. Equipment or Sep 2.
outlets that show any sign of malfunction or damage should 17. Tiainen M, Kovala TT, Takkunen OS, et al. Somatosensory and brainstem
not be used. Likewise patient ground connections should auditory evoked potentials in cardiac arrest patients treated with
hypothermia. Crit Care Med 2005;33:1736–40.
never be improvised; patient connections on clinical equip- 18. Bouwes A, Binnekade JM, Zandstra DF, et al. Somatosensory evoked
ment use special isolators or current limiters to reduce the potentials during mild hypothermia after cardiopulmonary resuscitation.
risk of shock. Neurology 2009;73:1457–61.
Section III—Electrophysiology 245

19. Zandbergen EG, Hijdra A, Koelman JH, et al. Prediction of poor outcome 36. Lori S, Bertini G, Molesti E, et al. The prognostic role of evoked potentials
within the first 3 days of postanoxic coma. Neurology 2006;66:62–8. in neonatal hypoxic-ischemic insult. J Matern Fetal Neonatal Med 2011;
20. Madl C, Kramer L, Domanovits H, et al. Improved outcome prediction in 24 Suppl 1:69–71. Epub 2011, Aug 31.
unconscious cardiac arrest survivors with sensory evoked potentials 37. Sleigh JW, Havill JH, Frith R, et al. Somatosensory evoked potentials in
compared with clinical assessment. Crit Care Med 2000;28:721–6. severe traumatic brain injury: a blinded study. J Neurosurg 1999;91:
21. Gendo A, Kramer L, Hafner M, et al. Time-dependency of sensory evoked 577–80.
potentials in comatose cardiac arrest survivors. Intensive Care Med 2001;27: 38. Rumpl E, Prugger M, Battista HJ, et al. Short latency somatosensory evoked
1305–11. potentials and brain-stem auditory evoked potentials in coma due to CNS
22. Logi F, Fischer C, Murri L, et al. The prognostic value of evoked responses depressant drug poisoning. Preliminary observations. Electroencephalogr
from primary somatosensory and auditory cortex in comatose patients. Clin Clin Neurophysiol 1988;70:482–9.
Neurophysiol 2003;114:1615–27. 39. Procaccio F, Polo A, Lanteri P, et al. Electrophysiologic monitoring in
23. Berek K, Lechleitner P, Luef G, et al. Early determination of neurological neurointensive care. Curr Opin Crit Care 2001;7:74–80.
outcome after prehospital cardiopulmonary resuscitation. Stroke 1995;26: 40. Chiappa KH, Hoch DB. Electrophysiologic monitoring. In: Ropper AH,
543–9. editor. Neurological and neurosurgical intensive care. New York: Raven Press;
24. Young GB, Doig G, Ragazzoni A. Anoxic-ischemic encephalopathy: clinical 1993. p. 147–83.
and electrophysiological associations with outcome. Neurocrit Care 41. Carter BG, Butt W. Review of the use of somatosensory evoked potentials in
2005;2:159–64. the prediction of outcome after severe brain injury. Crit Care Med 2001;29:
25. Chen R, Bolton CF, Young B. Prediction of outcome in patients with anoxic 178–86.
coma: a clinical and electrophysiologic study. Crit Care Med 1996;24:672–8. 42. Carter BG, Butt W. Are somatosensory evoked potentials the best predictor
26. Bassetti C, Bomio F, Mathis J, et al. Early prognosis in coma after cardiac of outcome after severe brain injury? A systematic review. Intensive Care
arrest: a prospective clinical, electrophysiological, and biochemical study of Med 2005;31:765–75.
60 patients. J Neurol Neurosurg Psychiatry 1996;61:610–5. 43. Claassen J, Hansen HC. Early recovery after closed traumatic head injury:
27. Zandbergen EG, de Haan RJ, Stoutenbeek CP, et al. Systematic review of somatosensory evoked potentials and clinical findings. Crit Care Med 2001;
early prediction of poor outcome in anoxic-ischaemic coma. Lancet 1998; 29:494–502.
352:1808–12. 44. Houlden DA, Taylor AB, Feinstein A, et al. Early somatosensory evoked
28. Wijdicks EF, Hijdra A, Young GB, et al. Practice parameter: prediction of potential grades in comatose traumatic brain injury patients predict
outcome in comatose survivors after cardiopulmonary resuscitation (an cognitive and functional outcome. Crit Care Med 2010;38:167–74.
evidence-based review): report of the Quality Standards Subcommittee of 45. Moulton RJ, Shedden PM, Tucker WS, et al. Somatosensory evoked potential
the American Academy of Neurology. Neurology 2006;67:203–10. monitoring following severe closed head injury. Clin Invest Med 1994;17:
29. Zhang Y, Su YY, Haupt WF, et al. Application of electrophysiologic 187–95.
techniques in poor outcome prediction among patients with severe focal and 46. Amantini A, Fossi S, Grippo A, et al. Continuous EEG-SSEP monitoring in
diffuse ischemic brain injury. J Clin Neurophysiol 2011;28(5):497–503. severe brain injury. Clin Neurophysiol 2009;39:85–93.
30. Rossetti AO, Oddo M, Liaudet L, et al. Predictors of awakening from 47. Christophis P. The prognostic value of somatosensory evoked potentials in
postanoxic status epilepticus after therapeutic hypothermia. Neurology 2009; traumatic primary and secondary brain stem lesions. Zentralbl Neurochir
72(8):744–9. 2004;65:25–31.
31. Meynaar IA, Oudemans-van Straaten HM, van der Wetering J, et al. Serum 48. Furlonger AJ, Sleigh JW, Havill JH, et al. Cognitive and psychosocial
neuron-specific enolase predicts outcome in post-anoxic coma: a prospective outcome in survivors of severe traumatic brain injury: correlations with
cohort study. Intensive Care Med 2003;29:189–95. cerebral perfusion pressure, frontal lobe damage and somatosensory evoked
32. Krieger DW. Evoked potentials not just to confirm hopelessness in anoxic potentials. Crit Care Resusc 2000;2:246–52.
brain injury. Lancet 1998;352:1796–7. 49. Rumpl E, Prugger M, Gerstenbrand F, et al. Central somatosensory
33. Walser H, Mattle H, Keller HM, et al. Early cortical median nerve conduction time and short latency somatosensory evoked potentials in
somatosensory evoked potentials: prognostic value in anoxic coma. Arch post-traumatic coma. Electroencephalogr Clin Neurophysiol 1983;56:
Neurol 1985;42:32–8. 583–96.
34. Zandbergen EG, Koelman JH, de Haan RJ, et al. SSEPs and prognosis in 50. Pohlmann-Eden B, Dingethal K, Bender HJ, et al. How reliable is the
postanoxic coma: only short or also long latency responses? Neurology 2006; predictive value of SEP (somatosensory evoked potentials) patterns in severe
67:583–6. brain damage with special regard to the bilateral loss of cortical responses?
35. Prohl J, Rother J, Kluge S, et al. Prediction of short-term and long-term Intensive Care Med 1997;23:301–8.
outcomes after cardiac arrest: a prospective multivariate approach combining
biochemical, clinical, electrophysiological, and neuropsychological A complete list of references for this chapter can be found online at
investigations. Crit Care Med 2007;35:1230–7. www.expertconsult.com.
Section III—Electrophysiology 245.e1

References 26. Bassetti C, Bomio F, Mathis J, et al. Early prognosis in coma after cardiac
arrest: a prospective clinical, electrophysiological, and biochemical study of
1. Dawson G. Investigations on a patient subject to myoclonic seizure after 60 patients. J Neurol Neurosurg Psychiatry 1996;61:610–5.
stimulation. J Neurol Neurosurg Psychiatry 1947;10:141–62. 27. Zandbergen EG, de Haan RJ, Stoutenbeek CP, et al. Systematic review of
2. Moulton RJ, Brown JI, Konasiewicz SJ. Monitoring severe head injury: a early prediction of poor outcome in anoxic-ischaemic coma. Lancet 1998;
comparison of EEG and somatosensory evoked potentials. Can J Neurol Sci 352:1808–12.
1998;25:S7–11. 28. Wijdicks EF, Hijdra A, Young GB, et al. Practice parameter: prediction of
3. Celesia G, Puletti F. Auditory cortical areas of man. Neurology 1969;19: outcome in comatose survivors after cardiopulmonary resuscitation (an
211–20. evidence-based review): report of the Quality Standards Subcommittee of
4. Wijdicks EF. Brain death worldwide: accepted fact but no global consensus the American Academy of Neurology. Neurology 2006;67:203–10.
in diagnostic criteria. Neurology 2002;58:20–5. 29. Zhang Y, Su YY, Haupt WF, et al. Application of electrophysiologic
5. Cruccu G, Aminoff MJ, Curio G, et al. Recommendations for the clinical techniques in poor outcome prediction among patients with severe focal
use of somatosensory-evoked potentials. Clin Neurophysiol 2008;119: and diffuse ischemic brain injury. J Clin Neurophysiol 2011;28(5):497–503.
1705–19. 30. Rossetti AO, Oddo M, Liaudet L, et al. Predictors of awakening from
6. Fossi S, Amantini A, Grippo A, et al. Continuous EEG-SEP monitoring of postanoxic status epilepticus after therapeutic hypothermia. Neurology
severely brain injured patients in NICU: methods and feasibility. 2009;72(8):744–9.
Neurophysiol Clin 2006;36:195–205. 31. Meynaar IA, Oudemans-van Straaten HM, van der Wetering J, et al. Serum
7. Celesia GC, Brigell MG. Auditory evoked potentials. In: Niedermeyer E, neuron-specific enolase predicts outcome in post-anoxic coma: a
Lopes da Silva F, editors. Electroencephalography: basic principles, clinical prospective cohort study. Intensive Care Med 2003;29:189–95.
applications, and related fields. Baltimore: Lippincott Williams & Wilkins; 32. Krieger DW. Evoked potentials not just to confirm hopelessness in anoxic
2005. p. 1137–64. brain injury. Lancet 1998;352:1796–7.
8. Mauguiere F. Somatosensory evoked potentials: normal responses, 33. Walser H, Mattle H, Keller HM, et al. Early cortical median nerve
abnormal waveforms, and clinical applications in neurological diseases. In: somatosensory evoked potentials: prognostic value in anoxic coma. Arch
Niedermeyer E, Lopes da Silva F, editors. Electroencephalography: basic Neurol 1985;42:32–8.
applications, and related fields. Baltimore: Lippincott Williams & Wilkins; 34. Zandbergen EG, Koelman JH, de Haan RJ, et al. SSEPs and prognosis in
2005. p. 1067–119. postanoxic coma: only short or also long latency responses? Neurology
9. Goff GD, Matsumiya Y, Allison T, et al. The scalp topography of human 2006;67:583–6.
somatosensory and auditory evoked potentials. Electroencephalogr Clin 35. Prohl J, Rother J, Kluge S, et al. Prediction of short-term and long-term
Neurophysiol 1977;42:57–76. outcomes after cardiac arrest: a prospective multivariate approach
10. Desmedt JE, Ozaki I. SEPs to finger joint input lack the N20-P20 response combining biochemical, clinical, electrophysiological, and
that is evoked by tactile inputs: contrast between cortical generators in neuropsychological investigations. Crit Care Med 2007;35:1230–7.
areas 3b and 2 in humans. Electroencephalogr Clin Neurophysiol 1991;80: 36. Lori S, Bertini G, Molesti E, et al. The prognostic role of evoked potentials
513–21. in neonatal hypoxic-ischemic insult. J Matern Fetal Neonatal Med 2011;
11. Scherg M, von Cramon D. A new interpretation of the generators of BAEP 24 Suppl 1:69–71. Epub 2011, Aug 31.
waves I-V: results of a spatio-temporal dipole model. Electroencephalogr 37. Sleigh JW, Havill JH, Frith R, et al. Somatosensory evoked potentials in
Clin Neurophysiol 1985;62:290–9. severe traumatic brain injury: a blinded study. J Neurosurg 1999;91:
12. Starr A. Auditory brain-stem responses in brain death. Brain 577–80.
1976;99:543–54. 38. Rumpl E, Prugger M, Battista HJ, et al. Short latency somatosensory evoked
13. Yvert B, Crouzeix A, Bertrand O, et al. Multiple supratemporal sources of potentials and brain-stem auditory evoked potentials in coma due to CNS
magnetic and electric auditory evoked middle latency components in depressant drug poisoning. Preliminary observations. Electroencephalogr
humans. Cereb Cortex 2001;11:411–23. Clin Neurophysiol 1988;70:482–9.
14. Linden DE. The p300: where in the brain is it produced and what does it 39. Procaccio F, Polo A, Lanteri P, et al. Electrophysiologic monitoring in
tell us? Neuroscientist 2005;11:563–76. neurointensive care. Curr Opin Crit Care 2001;7:74–80.
15. Guerit JM, Amantini A, Amodio P, et al. Consensus on the use of 40. Chiappa KH, Hoch DB. Electrophysiologic monitoring. In: Ropper AH,
neurophysiological tests in the intensive care unit (ICU): editor. Neurological and neurosurgical intensive care. New York: Raven
electroencephalogram (EEG), evoked potentials (EPs), and Press; 1993. p. 147–83.
electroneuromyography (ENMG). Neurophysiol Clin 2009;39:71–83. 41. Carter BG, Butt W. Review of the use of somatosensory evoked potentials
16. Bouwes A, Binnekade JM, Verbaan BW, et al. Predictive value of in the prediction of outcome after severe brain injury. Crit Care Med 2001;
neurological examination for early cortical responses to somatosensory 29:178–86.
evoked potentials in patients with postanoxic coma. J Neurol 2012;259(3): 42. Carter BG, Butt W. Are somatosensory evoked potentials the best predictor
537–41. Epub 2011, Sep 2. of outcome after severe brain injury? A systematic review. Intensive Care
17. Tiainen M, Kovala TT, Takkunen OS, et al. Somatosensory and brainstem Med 2005;31:765–75.
auditory evoked potentials in cardiac arrest patients treated with 43. Claassen J, Hansen HC. Early recovery after closed traumatic head injury:
hypothermia. Crit Care Med 2005;33:1736–40. somatosensory evoked potentials and clinical findings. Crit Care Med 2001;
18. Bouwes A, Binnekade JM, Zandstra DF, et al. Somatosensory evoked 29:494–502.
potentials during mild hypothermia after cardiopulmonary resuscitation. 44. Houlden DA, Taylor AB, Feinstein A, et al. Early somatosensory evoked
Neurology 2009;73:1457–61. potential grades in comatose traumatic brain injury patients predict
19. Zandbergen EG, Hijdra A, Koelman JH, et al. Prediction of poor outcome cognitive and functional outcome. Crit Care Med 2010;38:167–74.
within the first 3 days of postanoxic coma. Neurology 2006;66:62–8. 45. Moulton RJ, Shedden PM, Tucker WS, et al. Somatosensory evoked
20. Madl C, Kramer L, Domanovits H, et al. Improved outcome prediction in potential monitoring following severe closed head injury. Clin Invest Med
unconscious cardiac arrest survivors with sensory evoked potentials 1994;17:187–95.
compared with clinical assessment. Crit Care Med 2000;28:721–6. 46. Amantini A, Fossi S, Grippo A, et al. Continuous EEG-SSEP monitoring in
21. Gendo A, Kramer L, Hafner M, et al. Time-dependency of sensory evoked severe brain injury. Clin Neurophysiol 2009;39:85–93.
potentials in comatose cardiac arrest survivors. Intensive Care Med 2001; 47. Christophis P. The prognostic value of somatosensory evoked potentials in
27:1305–11. traumatic primary and secondary brain stem lesions. Zentralbl Neurochir
22. Logi F, Fischer C, Murri L, et al. The prognostic value of evoked responses 2004;65:25–31.
from primary somatosensory and auditory cortex in comatose patients. 48. Furlonger AJ, Sleigh JW, Havill JH, et al. Cognitive and psychosocial
Clin Neurophysiol 2003;114:1615–27. outcome in survivors of severe traumatic brain injury: correlations with
23. Berek K, Lechleitner P, Luef G, et al. Early determination of neurological cerebral perfusion pressure, frontal lobe damage and somatosensory evoked
outcome after prehospital cardiopulmonary resuscitation. Stroke 1995;26: potentials. Crit Care Resusc 2000;2:246–52.
543–9. 49. Rumpl E, Prugger M, Gerstenbrand F, et al. Central somatosensory
24. Young GB, Doig G, Ragazzoni A. Anoxic-ischemic encephalopathy: clinical conduction time and short latency somatosensory evoked potentials in
and electrophysiological associations with outcome. Neurocrit Care 2005;2: post-traumatic coma. Electroencephalogr Clin Neurophysiol 1983;56:
159–64. 583–96.
25. Chen R, Bolton CF, Young B. Prediction of outcome in patients with anoxic 50. Pohlmann-Eden B, Dingethal K, Bender HJ, et al. How reliable is the
coma: a clinical and electrophysiologic study. Crit Care Med 1996;24:672–8. predictive value of SEP (somatosensory evoked potentials) patterns in
245.e2 Section III—Electrophysiology

severe brain damage with special regard to the bilateral loss of cortical 76. Haupt WF, Hojer C, Pawlik G. Prognostic value of evoked potentials and
responses? Intensive Care Med 1997;23:301–8. clinical grading in primary subarachnoid haemorrhage. Acta Neurochir
51. Krieger D, Jauss M, Schwarz S, et al. Serial somatosensory and brainstem (Wien) 1995;137:146–50. discussion 50.
auditory evoked potentials in monitoring of acute supratentorial mass 77. Ritz R, Schwerdtfeger K, Strowitzki M, et al. Prognostic value of SSEP in
lesions. Crit Care Med 1995;23:1123–31. early aneurysm surgery after SAH in poor-grade patients. Neurol Res
52. Cant BR, Hume AL, Judson JA, et al. The assessment of severe head injury 2002;24:756–64.
by short-latency somatosensory and brain-stem auditory evoked potentials. 78. Szabo S, Miko L, Novak L, et al. Correlation between central
Electroencephalogr Clin Neurophysiol 1986;65:188–95. somatosensory conduction time, blood flow velocity, and delayed cerebral
53. Lindsay KW, Carlin J, Kennedy I, et al. Evoked potentials in severe head ischemia after aneurysmal subarachnoid hemorrhage. Neurosurg Rev 1997;
injury—analysis and relation to outcome. J Neurol Neurosurg Psychiatry 20:188–95.
1981;44:796–802. 79. Rosenstein J, Wang AD, Symon L, et al. Relationship between hemispheric
54. Lindsay K, Pasaoglu A, Hirst D, et al. Somatosensory and auditory brain cerebral blood flow, central conduction time, and clinical grade in
stem conduction after head injury: a comparison with clinical features in aneurysmal subarachnoid hemorrhage. J Neurosurg 1985;62:25–30.
prediction of outcome. Neurosurgery 1990;26:278–85. 80. Ikeda K, Yamashita J. Postoperative central conduction time and cerebral
55. Rappaport M, Hemmerle AV, Rappaport ML. Short and long latency blood flow in patients with aneurysmal subarachnoid hemorrhage:
auditory evoked potentials in traumatic brain injury patients. Clin relationship with prognosis and ischemic conditions. Surg Neurol 1992;38:
Electroencephalogr 1991;22:199–202. 445–53.
56. Shimbo Y, Sakata M, Hayano M, et al. Topographical relationships between 81. Fazl M, Houlden DA, Weaver K. Correlation between cerebral blood flow,
the brainstem auditory and somatosensory evoked potentials and the somatosensory evoked potentials, CT scan grade and neurological grade
location of lesions in posterior fossa stroke. Neurol Med Chir (Tokyo) in patients with subarachnoid hemorrhage. Can J Neurol Sci 1991;18:
2003;43:282–91. discussion 92. 453–7.
57. Kalita J, Misra UK. Motor and sensory evoked potential studies in 82. Symon L, Hargadine J, Zawirski M, et al. Central conduction time as an
brainstem strokes. Electromyogr Clin Neurophysiol 1997;37:379–83. index of ischaemia in subarachnoid haemorrhage. J Neurol Sci
58. Gutling E, Isenmann S, Wichmann W. Electrophysiology in the locked-in- 1979;44:95–103.
syndrome. Neurology 1996;46:1092–101. 83. Schick U, Dohnert J, Meyer JJ, et al. Prognostic significance of SSEP, BAEP
59. Towle VL, Maselli R, Bernstein LP, et al. Electrophysiologic studies on and serum S-100B monitoring after aneurysm surgery. Acta Neurol Scand
locked-in patients: heterogeneity of findings. Electroencephalogr Clin 2003;108:161–9.
Neurophysiol 1989;73:419–26. 84. Wachter D, Christophis P, Stein M, et al. Use of multimodal
60. Bassetti C, Mathis J, Hess CW. Multimodal electrophysiological studies electrophysiological monitoring to predict outcome after subarachnoid
including motor evoked potentials in patients with locked-in syndrome: hemorrhage? A prospective series. J Neurosurg Sci 2011;55(3):179–87.
report of six patients. J Neurol Neurosurg Psychiatry 1994;57:1403–6. 85. Niimi Y, Deltis V, Berenstein A. Neurophysiological monitoring during
61. Burghaus L, Liu WC, Dohmen C, et al. Evoked potentials in acute ischemic spinal endovascular procedures. In: Nuwer M, editor. Handbook of clinical
stroke within the first 24 h: possible predictor of a malignant course. neurophysiology. Amsterdam: Elsevier; 2008. p. 651–70.
Neurocrit Care 2008;9:13–6. 86. Iseli E, Cavigelli A, Dietz V, et al. Prognosis and recovery in ischaemic and
62. Stern BJ, Krumholz A, Weiss HD, et al. Evaluation of brainstem stroke traumatic spinal cord injury: clinical and electrophysiological evaluation.
using brainstem auditory evoked responses. Stroke 1982;13:705–11. J Neurol Neurosurg Psychiatry 1999;67:567–71.
63. Krieger D, Adams HP, Rieke K, et al. Prospective evaluation of the 87. Jacobs SR, Yeaney NK, Herbison GJ, et al. Future ambulation prognosis as
prognostic significance of evoked potentials in acute basilar occlusion. Crit predicted by somatosensory evoked potentials in motor complete and
Care Med 1993;21:1169–74. incomplete quadriplegia. Arch Phys Med Rehabil 1995;76:635–41.
64. Krieger D, Adams HP, Rieke K, et al. Monitoring therapeutic efficacy of 88. Curt A, Dietz V. Ambulatory capacity in spinal cord injury: significance of
decompressive craniotomy in space occupying cerebellar infarcts using somatosensory evoked potentials and ASIA protocol in predicting outcome.
brain-stem auditory evoked potentials. Electroencephalogr Clin Arch Phys Med Rehabil 1997;78:39–43.
Neurophysiol 1993;88:261–70. 89. Katz RT, Toleikis RJ, Knuth AE. Somatosensory-evoked and dermatomal-
65. Steiner T, Jauss M, Krieger DW. Hemicraniectomy for massive cerebral evoked potentials are not clinically useful in the prognostication of acute
infarction: evoked potentials as presurgical prognostic factors. J Stroke spinal cord injury. Spine 1991;16:730–5.
Cerebrovasc Dis 1998;7:132v8. 90. Zörner B, Blanckenhorn WU, Dietz V, et al. Clinical algorithm for improved
66. Kalita J, Misra UK. Somatosensory evoked potential studies in internal prediction of ambulation and patient stratification after incomplete spinal
capsule and corona radiata infarction. J Neurol 1998;245:531–6. cord injury. J Neurotrauma 2010;27(1):241–52.
67. Haupt WF, Birkmann C, Halber M. Serial evoked potentials and outcome in 91. Cheliout-Heraut F, Loubert G, Masri-Zada T, et al. Evaluation of early
cerebrovascular critical care patients. J Clin Neurophysiol 2000;17:326–30. motor and sensory evoked potentials in cervical spinal cord injury.
68. Chu NS. Median and tibial somatosensory evoked potentials. Changes in Neurophysiol Clin 1998;28:39–55.
short- and long-latency components in patients with lesions of the 92. Buchner H, Ferbert A, Hacke W. Serial recording of median nerve
thalamus and thalamo-cortical radiations. J Neurol Sci 1986;76:199–219. stimulated subcortical somatosensory evoked potentials (SEPs) in
69. Kato H, Sugawara Y, Ito H, et al. Somatosensory evoked potentials developing brain death. Electroencephalogr Clin Neurophysiol
following stimulation of median and tibial nerves in patients with localized 1988;69:14–23.
intracerebral hemorrhage: correlations with clinical and CT findings. J 93. Sonoo M, Tsai-Shozawa Y, Aoki M, et al. N18 in median somatosensory
Neurol Sci 1991;103:172–8. evoked potentials: a new indicator of medullary function useful for the
70. Ferbert A, Buchner H, Bruckmann H. Brainstem auditory evoked potentials diagnosis of brain death. J Neurol Neurosurg Psychiatry 1999;67:374–8.
and somatosensory evoked potentials in pontine haemorrhage. Correlations 94. Goldie WD, Chiappa KH, Young RR, et al. Brainstem auditory and
with clinical and CT findings. Brain 1990;113(Pt 1):49–63. short-latency somatosensory evoked responses in brain death. Neurology
71. Lumenta CB. Measurements of brain-stem auditory evoked potentials in 1981;31:248–56.
patients with spontaneous intracerebral hemorrhage. J Neurosurg 1984;60: 95. Machado C, Valdes P, Garcia-Tigera J, et al. Brain-stem auditory evoked
548–52. potentials and brain death. Electroencephalogr Clin Neurophysiol
72. Liu CW, Chu NS, Ryu SJ. CT somatosensory and brainstem auditory 1991;80:392–8.
evoked potentials in the early prediction of functional outcome in 96. Facco E, Munari M, Gallo F, et al. Role of short latency evoked potentials in
putaminal hemorrhage. Acta Neurol Scand 1991;84:28–32. the diagnosis of brain death. Clin Neurophysiol 2002;113:1855–66.
73. Shigemori M, Yuge T, Kawasaki K, et al. Evaluation of brain dysfunction in 97. Guerit JM, Amantini A, Fischer C, et al. Neurophysiological investigations
hypertensive putaminal hemorrhage with multimodality evoked potentials. of hepatic encephalopathy: ISHEN practice guidelines. Liver Int
Stroke 1987;18:72–6. 2009;29:789–96.
74. Facco E, Behr AU, Munari M, et al. Auditory and somatosensory evoked 98. Nuwer MR, Emerson RG, Galloway G, et al. Evidence-based guideline
potentials in coma following spontaneous cerebral hemorrhage: early update: intraoperative spinal monitoring with somatosensory and
prognosis and outcome. Electroencephalogr Clin Neurophysiol 1998;107: transcranial electrical motor evoked potentials. J Clin Neurophysiol 2012;
332–8. 29(1):101–8.
75. Wang AD, Cone J, Symon L, et al. Somatosensory evoked potential 99. Gonzalez AA, Jeyanandarajan D, Hansen C, et al. Intraoperative
monitoring during the management of aneurysmal SAH. J Neurosurg neurophysiological monitoring during spine surgery: a review. Neurosurg
1984;60:264–8. Focus 2009;27(4):E6.
Section III—Electrophysiology 245.e3

100. Resnick DK, Anderson PA, Kaiser MG, et al. Electrophysiological 112. Sloan TB, Ronai AK, Toleikis JR, et al. Improvement of intraoperative
monitoring during surgery for cervical degenerative myelopathy and somatosensory evoked potentials by etomidate. Anesth Analg 1988;67:
radiculopathy. J Neurosurg Spine 2009;11(2):245–52. 582–5.
101. McGarvey ML, Cheung AT. Neuologic intraoperative electrophysiologic 113. Legatt A. BAEP in surgery. In: Nuwer M, editor. Intraoperative monitoring
monitoring. In: Reich DL, Kahn RA, Mittnacht AJC, et al, editors. of neural function handbook of clinical neurophysiology. Elsevier; 2008.
Monitoring in anesthesia and perioperative care. New York: Cambridge p. 334–49.
University Press; 2011. p. 199–217. 114. Drummond JC, Todd MM, Schubert A, et al. Effect of the acute
102. Wicks RT, Pradilla G, Raza SM, et al. Impact of changes in intraoperative administration of high dose pentobarbital on human brain stem auditory
somato-sensory evoked potentials (SSEPs) on stroke rates after clipping of and median nerve somatosensory evoked responses. Neurosurgery 1987;20:
intracranial aneurysms. Neurosurgery 2011;Nov 3. Epub ahead of print. 830–5.
103. Shinzawa M, Yoshitani K, Minatoya K, et al. Changes of motor evoked 115. Liu EH, Wong HK, Chia CP, et al. Effects of isoflurane and propofol on
potentials during descending thoracic and thoracoabdominal aortic surgery cortical somatosensory evoked potentials during comparable depth of
with deep hypothermic circulatory arrest. J Anesth 2011;Dec 27. Epub anaesthesia as guided by bispectral index. Br J Anaesth 2005;94:193–7.
ahead of print. 116. Chan YW, Woo E, Yu YL. Chronic effects of phenytoin on brain-stem
104. Quraishi NA, Lewis SJ, Kelleher MO, et al. Intraoperative multimodality auditory evoked potentials in man. Electroencephalogr Clin Neurophysiol
monitoring in adult spinal deformity: analysis of a prospective series of one 1990;77:119–26.
hundred two cases with independent evaluation. Spine 2009;34(14):1504–12. 117. Hirose G, Kitagawa Y, Chujo T, et al. Acute effects of phenytoin on
105. Simon MV, Cole AJ, Chang EC, et al. An intraoperative multimodal brainstem auditory evoked potentials: clinical and experimental study.
neurophysiologic approach to successful resection of precentral gyrus Neurology 1986;36:1521–4.
epileptogenic lesions. Epilepsia 2012;Feb 6. Epub ahead of print. 118. Markand ON, Warren C, Mallik GS, et al. Effects of hypothermia on short
106. Krieg SM, Shiban E, Droese D, et al. Predictive value and safety of latency somatosensory evoked potentials in humans. Electroencephalogr
intraoperative neurophysiological monitoring using motor evoked Clin Neurophysiol 1990;77:416–24.
potentials in glioma surgery. Neurosurgery 2011;Nov 3. Epub ahead of print. 119. Guerit JM, Soveges L, Baele P, Dion R. Median nerve somatosensory evoked
107. Lepski G, Honegger J, Liebsch M, et al. Safe resection of arteriovenous potentials in profound hypothermia for ascending aorta repair.
malformations in eloquent motor areas aided by functional imaging and Electroencephalogr Clin Neurophysiol 1990;77:163–73.
intraoperative monitoring. Neurosurgery 2011;Sep 23. Epub ahead of print. 120. Markand ON, Lee BI, Warren C, et al. Effects of hypothermia on brainstem
108. Constantini S, Miller DC, Allen JC, et al. Radical excision of intramedullary auditory evoked potentials in humans. Ann Neurol 1987;22:507–13.
spinal cord tumors: surgical morbidity and long-term follow-up evaluation 121. Rodriguez RA, Audenaert SM, Austin EH, 3rd, et al. Auditory evoked
in 164 children and young adults. J Neurosurg 2000;93(2 Suppl):183–93. responses in children during hypothermic cardiopulmonary bypass: report
109. Simon MV. Neurophysiologic intraoperative monitoring of the of cases. J Clin Neurophysiol 1995;12:168–76.
vestibulocochlear nerve. J Clin Neurophysiol 2011;28(6):566–81. 122. Kusakari J, Inamura N, Sakurai T, et al. Effect of hypothermia upon the
110. Zandbergen EG, Hijdra A, de Haan RJ, et al. Interobserver variation in the electrocochleogram and auditory evoked brainstem response. Tohoku J Exp
interpretation of SSEPs in anoxic-ischaemic coma. Clin Neurophysiol Med 1984;143:351–9.
2006;117:1529–35. 123. Rosenblum SM, Ruth RA, Gal TJ. Brain stem auditory evoked potential
111. Hakimi K, Kinney G, Kraft G, et al. Reliability in interpretation of median monitoring during profound hypothermia and circulatory arrest. Ann Otol
somatosensory evoked potentials in the setting of coma: factors and Rhinol Laryngol 1985;94:281–3.
implications. Neurocrit Care 2009;11(3):353–61. 124. Seaba P. Electrical safety. Am J EEG Technol 1980;20:1–13.
III
Chapter
25  

Electroencephalography
Peter Horn, Mauro Oddo, and Sarah E. Schmitt

Introduction others. However, the integration of EEG recording into the


The German psychiatrist Hans Berger made the first recording environment of neurocritical care requires a refined technical
of the electric field of the human brain in 1924 in Jena. He setup and highly trained technical and medical staff. These
gave this recording the name electroencephalogram (EEG).1 necessities require a mutidisciplinary team. This chapter
His crucial observations served as the basis for the later appli- focuses on the application of intermittent (iEEG) or cEEG
cation of the EEG in clinical practice and promoted the fast monitoring in the NCCU.
development of clinical neurology and neurophysiology. The
EEG itself represents the spontaneous electrical activity of the
brain. Most brain electrical activity in a healthy individual is Physiologic Electroencephalogram
between 20 and 40 microvolts, although it may reach up to
100 microvolts. There are differences when measured on the
Background Activity
scalp or directly on the brain surface. The bandwidth of the In a healthy individual, typical EEG background activity
signal ranges between 0.4 and about 50 Hz and is highly vari- consists of a well-organized and predictable spectrum of
able. In contrast to spontaneous EEG background activity, waveforms. However, numerous factors may influence the fre-
evoked potentials (EPs) represent the components of the EEG quency and organization of these waveforms. There exist
that arise in response to a certain stimulus (e.g., visual, tactile, ultra-slow and ultra-fast frequencies that in general do not
or auditory) (see Chapter 24). These signals are usually below play a significant role in clinical practice in the intensive care
the noise level and are thus not readily distinguished in the unit (ICU). However, ultra-fast frequencies can be clinically
EEG background activity. Therefore one must use repetition useful in predicting outcomes from epilepsy surgery. Very low-
of the stimuli and signal averaging to improve the signal-to- frequency changes usually are filtered out and although they
noise ratio to get appropriate information.2 may be observed in patients in coma or preterminal condi-
In clinical practice, the recording of EEG background activ- tions, they usually do not provide clinically useful informa-
ity and the application of EPs are broadly accepted. Thus tion. In clinical practice the frequency range of the EEG is
EEG, including both traditional recordings and modifications broken down as follows:
such as compressed spectral analysis, is a cornerstone of Delta rhythm: 0.1 to 3.5 Hz
neurologic diagnostics, clinical neurophysiology, and related Theta rhythm: 4 to 7.5 Hz
research. Before 1996, EEGs were performed on analog Alpha rhythm: 8 to 13 Hz
machines with paper printouts. Technical advances since 2000 Beta rhythm: 14 to 30 Hz
have led to a step-wise introduction of the EEG into neuro- Gamma rhythm: >30 Hz (often filtered out on scalp EEG)
critical care. The era of digitization has broadened the Several aspects are essential for EEG interpretation: (1) fre-
spectrum for EEG recordings on the neurocritical care unit quency, (2) amplitude, (3) rhythmicity, (4) morphology, and
(NCCU) (EEG information can be recorded continuously (5) spatial distribution. These aspects are highly variable
and stored digitally).3 EEG and EPs have become common under normal conditions and show a strong dependence
and valuable tools in the NCCU. In particular, different on numerous factors such as age, level of consciousness
EEG-derived parameters can help predict individual clinical (alertness), and physical or mental activity. In addition, many
prognosis and outcome.4,5 Continuous EEG (cEEG) allows external or intrinsic stimuli can influence EEG background
real-time monitoring of the underlying cerebral physiology activity.7,8
noninvasively and cost effectively6 in critically ill patients, and
it can be used to gauge the efficacy of certain medications or
therapeutic maneuvers. For example, cEEG can be used for Pathologic Electroencephalogram
seizure diagnosis in comatose patients after traumatic brain
injury (TBI), detection of delayed cerebral ischemia (DCI),
Activity
after subarachnoid hemorrhage (SAH), and titration of thera- EEG background activity is very sensitive to changes in brain
pies for status epilepticus or barbiturate-induced coma among homeostasis and physiology. EEG may show manifestations of
246 © Copyright 2013 Elsevier Inc. All rights reserved.
Section III—Electrophysiology 247

Fig. 25.1  Right temporal seizure with evolution in morphology, amplitude, frequency, and spatial distribution.

numerous conditions, including metabolic encephalopathy, patients with impaired consciousness in whom clinical diag-
systemic hypoxia, brain trauma, ischemic and hemorrhagic nosis of a seizure may be difficult. In this group of patients
stroke, brain tumor, and infectious disease to name a few. EEG also can help assess status epilepticus (SE), nonconvul-
Any change in electrical brain activity, either focal or gen- sive status epilepticus (NCSE), and seizure-like activity such
eralized, that leads to a failure of local compensation may as periodic epileptiform discharges (PEDs) or generalized
cause epileptic discharges. The extent and duration of EEG periodic epileptiform discharges (G-PEDs).9-11
alterations are highly variable. An electrographic seizure is SE is a neurologic emergency with high mortality and mor-
defined as a pattern lasting more than 10 seconds with (1) bidity. There are several types of SE, and consequently it has
epileptiform waveforms at 3 Hz or greater, (2) epileptiform several definitions. The International League Against Epilepsy
waveforms at less than 3 Hz with clear evidence of evolution defines SE as “a seizure that persists for a sufficient length of
in time or space and (3) evidence of clinical or electrographic time or repeated frequently enough that recovery between
improvement following administration of an antiepileptic attacks does not occur.”11a SE can be subdivided based on clini-
drug (AED. Fig. 25.1). Furthermore, a seizure is likely to be cal and EEG criteria into generalized convulsive status (tonic-
present if the pathologic EEG is characterized by an evolution clonic, tonic, clonic, and myoclonic), generalized nonconvulsive
in frequency, morphology, and location.3 (absence) status, elementary partial status (with several sub-
types), and complex partial status. Official definitions have
not included the exact duration of time, but based on animal
Types of Seizures, Related Phenomena, studies that demonstrate significant brain damage after 30
and Differential Diagnosis minutes, convulsive status epilepticus (CSE) traditionally is
In the clinical setting many seizures are easily recognized and considered as one continuous unremitting seizure that lasts
therefore amenable to direct intervention and treatment. Con- greater than 30 minutes. CSE may be observed in between 1%
vulsive seizures (either primary or secondarily generalized) and 30% of ICU patients.12-14 However, it is impractical and
are usually clinically obvious and do not require an EEG for unsafe to wait 30 minutes before initiating treatment and
diagnosis. However, many focal seizures are associated with because it is rare for generalized convulsive seizures to spon-
few or no outward clinical manifestations. These seizures, taneously cease after 5 minutes, some suggest that a duration
which are referred to as nonconvulsive seizures or subclinical of 5 or 10 minutes may be a reasonable operational defini-
seizures require an EEG for their diagnosis, including among tion.15,16 NCSE is defined as an impairment of consciousness
248 Section III—Electrophysiology

or behavior from baseline state for at least 30 minutes without necessary for at least 48 hours to detect 85% of the CSE and
convulsive movements, and the presence of one or more of NCSE in comatose ICU patients. Thus the latency for the
the following: (1) repetitive focal or generalized epileptiform detection of NCSE or CSE might be as long as 48 to 72 hours,
activity (spikes, sharp waves, spike-and-wave, sharp-and-slow or even up to 120 hours.10,22
wave complexes) or rhythmic theta or delta activity at more Up to 50% of individuals with a history of generalized
than two per second; (2) the EEG patterns described in 1 at tonic-clonic seizures who fail to return to baseline despite
less than one per second, but with improvement after intrave- adequate treatment have evidence of NCSE on EEG.23 There
nous (IV) AED injection (e.g., a benzodiazepine); or (3) a is evidence that NCSE may exacerbate neuronal injury. For
temporal evolution of epileptiform or rhythmic activity example, NCSE is known to lead to significant hippocampal
greater than 1 Hz with change in location or frequency over atrophy after TBI indicative for severe neuronal injury.24 It
time and normalization (either clinically or electrographi- can also exacerbate midline shift in intracerebral hemorrhage
cally) in response to AED administration. If a patient experi- (ICH) patients.25 In addition, nonconvulsive electrographic
ences frequent recurrent seizures without a clear return to post-traumatic seizures have been observed to result in
baseline mental status between seizures, then he or she is in increases in ICP and microdialysis markers of cellular distress
NCSE by definition. Many of the patients with NCSE have (e.g., lactate pyruvate ratio or glutamate).26,27
subtle motor manifestations; these manifestations may be A variety of other periodic EEG patterns can be observed
focal or generalized.17,18 NCSE may be classified according to in critically ill patients such as triphasic waves, periodic later-
patient age or subdivided into: (1) complex partial SE (CPSE) alized epileptiform discharges (PLEDs), or burst suppression.
or (2) generalized nonconvulsive SE. PLEDs are considered traditionally an interictal epileptiform
Nonconvulsive seizures (NCS) are common in critically ill disturbance (Fig. 25.2). They are observed most frequently
patients, and many studies suggest that the majority of sei- with acute focal destructive lesions (e.g., infarct, infection, or
zures in these patients have limited clinical manifestations and tumor). The presence of PLEDs may predict a delayed time to
can only be recognized with EEG monitoring.19 After primary first seizure among ICU patients who undergo cEEG monitor-
brain injury, NCS occur in 11% to 55% of patients.20,21 The ing; that is, if PLEDs are present a longer duration of cEEG
variable prevalence of NCS reported in the studies may reflect may be indicated. A spontaneous burst-suppression pattern
different injury types and different levels of sedation present indicates severe brain dysfunction particularly in comatose
during the EEG recordings. The criteria for diagnosis are listed patients.18 This condition is associated with bursts of cortical
in Table 25.1. The clinical spectrum of NCS is broad, and activity that alternate with near complete suppression of back-
without prolonged monitoring the majority of NCS will not ground activity. The most common context in which burst
be identified. However, it is unclear what constitutes an suppression occurs is in pharmacologically induced comas for
adequate duration of monitoring. Several studies suggest that the control of elevated ICP (see following text) or refractory
the continuous monitoring of EEG background activity is status epilepticus.28 However, a nonpharmacologically induced
burst-suppression pattern may be seen in patients with severe
diffuse brain injury, particularly in severe diffuse anoxic brain
Table 25.1  Diagnostic Criteria for a injury. In addition, focal or generalized nonseizure activity,
Nonconvulsive Seizure like the aforementioned rhythmic or periodic sharp tran-
sients, is frequently observed in critically ill patients. The inci-
Any pattern lasting at least 10 seconds satisfying any one of dence of G-PED or PLED ranges between 50% and 58%.10,23
the following three primary criteria: Patients with PEDs are at high risk of seizures. G-PEDS rep-
Primary criteria
Repetitive generalized or focal spikes, sharp waves, resent a rare phenomenon that was observed in 37 of 3000
spike-and-wave complexes at ≥3/second EEG recordings only in one center.29 In this series, 89% of 37
Repetitive generalized or focal spikes, sharp waves, patients had either a myoclonic seizure (35.2%), a generalized
spike-and-wave or sharp-and-slow wave complexes at convulsive seizure (GCS) (21.6%), or generalized status epi-
<3/second with a secondary criterion
Sequential rhythmic, periodic, or quasi-periodic waves at
lepticus (GSE) (32.4%) within 48 hours of the detection of
≥1/second and clear evolution in frequency (increasing or the G-PEDs. The occurrence of G-PEDS and its variants is
decreasing by at least 1/second), morphology, or location associated with subacute sclerosing panencephalitis (SSPE)
(gradual spread into or out of a region that involves at and sporadic Creutzfeldt-Jakob disease but most frequently
least two electrodes). Change in sharpness without observed acutely in patients with diffuse anoxic brain injury.30
another morphology change is not enough to be
considered evolution in morphology. Evolution in The application of high-frequency recording by means
amplitude alone is not sufficient for diagnosis. of subdural grid electrodes allows the detection of self-
Secondary criterion propagating waves of neural and glial depolarization. This
Significant improvement in clinical state or appearance of slow progressing wave that spreads at 2 to 5 mm/minute over
previously absent normal EEG patterns (such as posterior-
dominant “alpha” rhythm) that is temporally associated
the cortex is known as cortical spreading depression (CSD).
with administration of a rapidly acting AED. Resolution of In the normal brain the EEG shows a period of transient
the “epileptiform” discharges that leaves diffuse slowing depression of cortical electrical activity that is associated
without clinical improvement and without appearance of with an increase in regional CBF and oxygen delivery (intact
previously absent normal EEG patterns would not satisfy neurovascular coupling). This phenomenon has a multifac-
the secondary criterion.
torial etiology and is known to occur in migraine. Under
Modified from Chong DJ, Hirsch LJ. Which EEG patterns warrant treatment in pathologic conditions, however, neurovascular coupling is
the critically ill? Reviewing the evidence for treatment of periodic epileptiform impaired and the depolarization wave causes a reduction in
discharges and related patterns. J Clin Neurophysiol 2005;22:79–91.
AED, Antiepileptic drug; EEG, electroencephalogram.
rCBF (spreading ischemia, neurovascular uncoupling). The
increased energy demands imposed by CSD may worsen
Section III—Electrophysiology 249

Fig. 25.2  Left central temporal periodic lateralized epileptiform discharges (PLEDs) at approximately 1 Hz.

neuronal injury under these conditions.31 Thus after aneu-


rysmal SAH the number and duration of clusters of CSD are Electrode Placement and Montage
temporally linked to the development of delayed infarct.32,33 Scalp pathology or other cranial injuries can limit electrode
The number of clusters is highly variable and may last placement38,39 and data quality. For monitoring of generalized
between 3 and 60 hours. activity the standard 21 electrodes40 according to the 10-20
system may be adequate. A few studies41 suggest 10 electrode
positions and a structured 14-channel montage with referen-
Electroencephalogram tial and bipolar derivations for monitoring in patients with
Methodology TBI. For cEEG monitoring Labar et al.42 successfully used 5
electrodes and 2 channels to analyze trends in compressed
Duration of Electroencephalogram spectral arrays in SAH patients. However, it is generally
Monitoring accepted that reducing the number of electrodes limits resolu-
The occurrence and time course of seizures, NCS, NCSE, and tion of waveform morphology and distribution. Therefore
PLEDs among others is highly variable and disease or pathol- focal abnormalities may be more difficult to recognize. Fur-
ogy dependent. Routine iEEG recordings, which last less than thermore, full electrode configuration improves the ability to
30 minutes, fail to capture a seizure in more than half of all distinguish brain signals from artifacts, aids in spatial localiza-
seizing patients. However, up to 50% of the seizures occur tion of pathologic activity, and provides redundancy.
within the first 60 minutes of EEG recording2,34,35 and 80% of Alternative methods are amplitude-integrated EEG (aEEG)
comatose patients have seizures within 24 hours of monitor- monitoring using two electrodes for seizure detection, a tech-
ing. Thus in comatose patients, exclusion of NCS or NCSE nique used in purpose-built devices that are common in neo-
requires at least 24 to 48 hours of recording time, whereas the natal ICUs. Most studies suggest a sensitivity of about 60%,
duration of routine iEEG should be at least 30 to 60 minutes but in some studies using this technique a third or fewer of
long for seizure detection. When EEG is used to detect isch- seizures identified using conventional electrode arrangements
emia and in particular DCI after aneurysmal SAH, monitoring are detected with aEEG.43,44 Similarly, a six-electrode configu-
should continue for the whole period of risk, up to 14 days ration placed at the hairline and compared to a full electrode
after insult.36,37 montage may miss a third of seizures and up to one half of
250 Section III—Electrophysiology

PLEDs. Because ictal arrhythmias, including ictal asystole recognize seizures and to detect reasonably rapid (seconds to
and ictal bradycardia, are relatively common phenomena, a minutes) changes in EEG content on an expert level. The
channel dedicated to electrocardiographic monitoring, prefer- obvious disadvantages are the time efforts needed for data
ably using two noncephalic electrodes, is nearly always needed review and the low sensitivity to gradual trends in the EEG
and may reveal important changes in the cardiac rhythm that (loss of faster frequencies or changes in variability of fre-
may be seen in association with some seizures.45-47 A nonce- quency content). In addition, this method is highly subjec-
phalic electrocardiographic channel can help distinguish tive, and interpretation of EEG patterns can vary between
between certain artifacts and rhythmic EEG discharges. In different electroencephalographers. Therefore visual analysis
daily practice, skin breakdown can be a problem, particularly of the raw EEG signal alone limits the application and bene-
when cup electrodes are used for prolonged periods. Alterna- ficial impact of cEEG monitoring technology in the ICU.49
tively needle electrodes can be used, but there are risks associ- Various approaches to process the raw EEG improve the
ated with this such as needlesticks or broken subdermal wires. diagnostic accuracy and cut down the time needed for data
Recently, intracortical electrodes that are placed stereotacti- interpretation. The aEEG method depicts time-compressed
cally into deep brain regions have been used in clinical and rectified EEG amplitude derived from raw EEG on a
research. Using this approach, Stuart et al., were able to dem- semi-logarithmic scale, whereas frequency domain methods
onstrate the potential value of depth electrodes to detect DCI of EEG present an alternative approach of signal simplifica-
associated with vasospasm in poor-grade SAH patients.48 tion.50,51 The aEEG provides an accurate measure of EEG
background activity52 that can be useful for clinical applica-
tion. Color density spectral array (CDSA) applies fast-Fourier
Data Analysis transformation (FFT) to convert raw EEG into a time-
Recording of EEG background activity requires the inspec- compressed and color-coded display (Fig. 25.3).53 Clinical
tion of the raw EEG. The visual analysis remains the stan- applications of quantitative EEG such as CDSA and related
dard method of EEG assessment. It is the best method to techniques include monitoring the depth of sedation,54

Fig. 25.3  Changes in color spectral array correspond to left temporal seizure activity in this patient with recurrent left temporal complex partial
seizures.
Section III—Electrophysiology 251

detection of cerebral ischemia,55 and identification of sei- diagnostic tool for coma of unknown etiology. In addition,
zures in adults, children, and neonates.56,57 The use of cEEG EEG recordings help monitor and direct therapeutic maneu-
may be most useful in patients that require long-term moni- vers in the NCCU. This includes the monitoring of the burst-
toring, for example, after poor-grade SAH when patients are suppression pattern during treatment of refractory intracranial
at risk for both seizures and ischemia. Claassen et al. reported hypertension, the monitoring of treatment efficacy for NCSE
that 19% of patients who had cEEG after poor-grade SAH and CSE, and the detection of DCI.4,68-70
(n = 108) had seizures, and 95% of these seizures were NCS
without any detectable clinical correlate. The application of
quantitative analysis to the cEEG (relative alpha variability, Traumatic Brain Injury
post-stimulation alpha-to-delta ratio [ADR]) enhances its In TBI, seizures are identified more frequently when EEG is
use in detection of DCI and increases its sensitivity for isch- performed than through clinical observation. However, the
emia detection.55 However, this does not improve the sensi- exact incidence varies according to the methodology used—
tivity for seizure detection, but its use can accelerate the AEDs, sedatives, and duration of monitoring. For example, in
process of screening the EEG for seizures. When using quan- a prospective study of 94 severe TBI patients who underwent
titative cEEG, raw EEG data should be available for individ- cEEG (on average for 7 days), Vespa et al. observed seizures in
ual review. The application of video EEG recordings (vEEG) 22% of the subjects, half of which were NCS: 50% of seizures
can help increase the likelihood for seizure detection on the occurred within day 1, despite phenytoin prophylaxis.71 Brain
ICU,58 particularly in patients with suspected NCSE.59 contusions and skull fractures were not associated with
increased seizures. When no antiepileptic prophylaxis was
given, Ronne-Engstrom and Winkler observed NCS in 28%
Electroencephalogram Artifacts and NCSE in 11% of TBI patients (n = 70) who had cEEG
and Artifact Detection monitoring more than 24 hours.72 NCS were associated with
Typical artifacts of the EEG are either non-neural physiologic extracerebral or intracerebral hematomas and older age (>60
activities of the subject or caused by external technical years). The majority of these events lasted less than 30 minutes
problems.60 Physiologic artifact sources are eye blinks, eye and all responded to benzodiazepines and phenytoin. On the
movements, muscle activity in the vicinity of the head (e.g., other hand, Amantini et al. observed far fewer seizures in TBI
face muscles, jaws, tongue, neck), heartbeat, pulse, and Mayer patients (3%). These differences may be associated with use
waves. Most problematic in clinical practice are ocular (electro- of a much greater dose of medications such as midazolam,
oculogram [EOG]) and muscle (electromyogram [EMG]) propofol, thiopental, or a combination, that can have antisei-
artifacts. EOG activity is caused by lateral eye movements or zure activity.73
by eye blinks (f = 20 times/min).61 This results in low-frequency EEG recordings have been used to predict outcome in TBI
activity most prominent over the anterior head regions (f max patients. Several factors are associated with outcome, includ-
= 4 Hz), whereas EMG activity usually shows as high-frequency ing (1) an increase or decrease in the absolute and relative
activity (>20 Hz), with a wide range of amplitudes.62 A wide amplitudes in the alpha and theta bands in the EEG can help
variety of technical artifacts can be seen, including induction differentiate survivors from nonsurvivors74; (2) a delta EEG
artifact, electrostatic artifact, capacitative artifact, and 60 Hz pattern is associated with death and poor functional outcome75;
artifact. In particular, the presence of an electrical appliance (3) a reduced percentage of alpha variability is associated with
running on an AC circuit can create a 60-Hz artifact.60 Artifact poor prognosis41; and (4) the presence of EEG reactivity to
rejection remains challenging in practice where hand- painful stimuli is associated with a good outcome.76 However,
optimized, semiautomatic, and fully automatic approaches prognostic errors based on EEG may be observed in 20% of
exist.61 Semiautomatic approaches, although less time con- patients.74
suming, require user interaction for ambiguous or outlier
components.63 Fully automated methods are proposed for the
classification of eye artifacts but are used only in some non- Aneurysmal Subarachnoid Hemorrhage
standardized research protocols.64 These methods, although The incidence of seizures after aneurysmal SAH (aSAH) varies
robust for the detection of eye artifacts, are not easily usable from 4% to 24%. Most cases occur at the time of aneurysm
for non–eye artifacts and may require the additional recording rupture or shortly after the initial hemorrhage.77 Risk factors
of the EOG. Future developments include EEGLAB plug-ins for seizures include a history of epilepsy, middle cerebral
and support vector machine (SVM)–based systems that in artery aneurysms, a cerebral infarct, and an intracerebral
a fully automatic mode recognize and reject major artifacts hematoma.78 NCS also are identified after SAH. The incidence
like eye blinks, eye movements, and heartbeats; the detection varies with the method of detection. For example, the inci-
of muscular or more subtle artifacts has not been reported dence of NCS is 3%, when using repeated standard (i.e.,
so far.65-67 iEEG).79 When using cEEG, and in particular for more than
24 hours, seizures are detected in 19% of patients, and the
majority (95%) of them are NCS.34 Up to 8% of patients have
Indications for NCSE, despite antiepileptic prophylaxis (fosphenytoin).80 Risk
Electroencephalogram Monitoring factors for NCSE include (1) older age, (2) female sex, (3)
poor-grade SAH, (4) the amount of cisternal blood, (5) hydro-
General Considerations cephalus, and (6) the presence of structural brain lesions
EEG is increasingly recognized as a useful tool to detect sei- including focal or global cerebral edema. NCSE can be refrac-
zures and seizure-like activity in different pathologies involv- tory to therapy, and many of these patients have a poor
ing the central nervous system (CNS) and to serve as a outcome. In many of these series cEEG was used in patients
252 Section III—Electrophysiology

Bilateral frontal ischemia — decrease in alpha variability

Fig. 25.4  Decrease in alpha variability in subarachnoid hemorrhage (SAH) associated with bifrontal ischemia.

flow (rCBF); these changes can be used to help differentiate


Table 25.2  Relationship Between Cerebral reversible from irreversible ischemia. When the EEG wave-
Blood Flow and Electroencephalogram form pattern is suppressed, rCBF is less than 8 to 10 mL/100 g/
Changes minute and indicates irreversible tissue damage.
CBF mL/100 g/min EEG Change Reversibility
In 2009, the occurrence of CSD after aSAH was demon-
strated.84,85 This may be present in more than 70% of the
35-70 Normal No injury patients studied and often is associated with epileptic seizures.
25-35 Loss of beta Reversible There is growing evidence that the presence of CSD, the
18-25 Theta slowing Reversible number of CSD clusters, and their duration may precede or
lead to DCI and potentially infarction.31,86
12-18 Delta slowing Reversible
There are conflicting data on the effect of seizures on SAH
<8-10 Suppression Irreversible patients’ outcome.79,87 Relatively limited data exist on the
CBF, Cerebral blood flow; EEG, electroencephalogram. prognostic value of EEG after SAH; the presence of PEDs, the
absence of normal sleep architecture and reactivity to stimuli,
and NCSE on cEEG are associated with poor outcome in this
with convulsive seizure activity, unexplained neurologic dete- population.78
rioration, or persistent and unexplained coma, but NCS are
still common even without any detectable clinical correlate.34
EEG including iEEG, aEEG, and cEEG recordings can help Intracerebral Hemorrhage
detect DCI.42,81-83 For example, Rivierez et al. studied 151 Seizures can be identified in 31% of patients with ICH. NCS
SAH patients on day 1 and day 5 after SAH with standard occur in between 10% and 30% of patients and may be
EEG, and found that patients (n = 78) with bilateral bursts of detected within 48 hours of cEEG initiation, regardless of
slow waves were more likely to have DCI.81 Artifacts and the antiepileptic phrophylaxis.25,88 This information may be
need for manual postprocessing of the raw data can limit the important because there is a described independent associa-
use of EEG to detect DCI. However, quantitative EEG analy- tion between phenytoin prophylaxis and worse cognitive
sis (qEEG) holds promise as a method to detect DCI (Fig. outcome after ICH.89
25.4). A decrease in the ADR extracted from the cEEG shows
a strong association with DCI in poor-grade SAH patients.83
The median decrease of ADR for patients with DCI was 24%, Hypoxic-Ischemic Encephalopathy
whereas an increase of 3% was observed in patients without EEG can be used for seizure and NCS detection and for
DCI associated with vasospasm.42,82,83 In a cohort of 32 outcome prediction after hypoxic-ischemic encephalopathy
mainly good-grade patients monitored with cEEG and trend- (HIE). In adult cardiac arrest (CA) patients, EEG seizures are
ing of a quantitative measure, the relative alpha variability, identified in 17% to 36% of patients.13,90,91 Electrographic sei-
Vespa et al.82 found that in all 19 patients with angiographi- zures also are observed when therapeutic hypothermia (TH)
cally proven vasospasm, relative alpha variability was signifi- is used and occur both during TH and after rewarming.92,93
cantly decreased and preceded the diagnosis of vasospasm by Because TH and sedation have antiepileptic properties, the
2 to 3 days. Table 25.2 lists the relation between EEG changes occurrence of electrical seizures during TH may reflect wide-
and the corresponding changes in regional cerebral blood spread severe brain injury, and prolonged seizures that occur
Section III—Electrophysiology 253

during TH are associated with poor outcome.91,94 However, a in Europe, Zoons et al. observed seizures in 17% of patients.
subset of patients who develop postanoxic seizures can have a These were associated with severe CNS and systemic inflam-
favorable outcome.95 mation, structural CNS lesions and pneumococcal meningitis.
A dichotomized scaling of postanoxic EEG patterns, “malig- Overall mortality rate of patients with epilepsy was 41%.105 In
nant” versus “benign” was described before the use of TH. a retrospective review of NCCU patients who had cEEG,
Malignant patterns include nonreactive background and Claassen et al. observed that 29% of those with CNS infections
spontaneous burst suppression and are a powerful EEG had seizures. Of those, half were NCS, which accounted for
pattern (false-positive rate, 0%) to identify patients with poor 10% of all NCS in this population.34 Other studies show that
outcome.96 For example, a nonreactive EEG pattern is associ- 6% to 12% of adult patients with ABM have seizures, but the
ated with in-hospital mortality,91,97 whereas the presence of a relationship between outcome and seizures is unclear.106,107 In
reactive EEG background98 or continuous activity90 predicts infants with bacterial meningitis, abnormal EEG findings,
awakening from postanoxic coma. These data, however, are including a continuous low voltage or flat trace on aEEG, EEG
not validated in patients who receive TH. In the posthypother- background activity and overall EEG description are associ-
mia era, there are also some data to suggest that the absence ated with adverse outcome.108 Infants with normal or mildly
of an N20 cortical response on somatosensory evoked poten- abnormal EEGs generally have a good neurologic status at
tials (SSEPs) in a postanoxic patient is still associated with a discharge.109
very poor prognosis.91
In neonates with HIE, therapeutic hypothermia can help
improve clinical outcome.99 EEG can be used to detect seizures Sepsis and Metabolic Disorders
during and after therapeutic hypothermia. For example, Multiple-organ failure can lead to seizures, and NCS may be
Wusthoff et al. observed seizures in 65% of neonates (n = 26) particularly common in these patients.110 For example, Oddo
with HIE who were studied; NCS were present in 47% and SE et al., in a retrospective analysis of 201 patients without a
in 23%. Seizure onset was observed within 48 hours in 76% history of brain injury who were admitted to a medical ICU,
of the patients.100 Unlike in adults, the background pattern observed NCS in 10% of the patients.36 Sepsis was an inde-
does not enhance the predictive value of EEG among infants pendent risk factor for NCS. In patients with liver failure, EEG
with HIE.101 can help detect seizures, particularly NCS, which are frequent
in this setting.111 After toxic-metabolic encephalopathy, NCS
are observed in 20% of patients admitted to the ICU who
Brain Tumors undergo cEEG.34
Brain tumors are a common cause of epilepsy, accounting for
10% to 15% of all causes of seizures in adult patients.102 Electroencephalographic
However, the exact incidence of seizures in patients with brain
tumors admitted to the ICU is poorly understood, in part
Monitoring to Guide Therapy
because cEEG monitoring often is not part of a prospective The recording of EEG background activity can be used to
protocol but rather is obtained for suspected cases based on assess and guide the depth of sedation in patients with
clinical judgment. Among 158 patients admitted to an NCCU, pharmacologically induced coma, for example, the induction
Bladin et al. observed seizures in patients, 6 of whom had of “burst-suppression-EEG” to control ICP and or control
brain tumors. Most of the patients (5/6) had clinical sei- refractory status epilepticus (RSE). In these patients monitor-
zures.103 Claassen et al. observed seizures in 23% of patients ing is recommended to guide dosing, with careful titration to
with brain tumors admitted to an NCCU who had cEEG; all maintain a sufficient burst-suppression pattern on the EEG
presented as NCS.34 Seizures were detected during the first 24 while minimizing adverse drug effects. It is not clear the
hours of monitoring in 88% of patients, whereas cEEG number of bursts per minute considered standard; however,
detected seizures more than 24 hours after cEEG monitoring values of 3 to 5 or 4 to 6 bursts per minute are targeted during
was started in patients who were comatose at the time moni- barbiturate therapy. Alternatively, the bispectral index (BIS)
toring began. Patients with seizures: (1) were younger, (2) monitor may be used to titrate barbiturate therapy because
were more likely to be comatose, and (3) had a more frequent the BIS and suppression ratio (SR) values correlate with the
history of seizures than patients without seizures at cEEG. It standard EEG-based method.112-116
is unclear if risk factors for seizure development in all brain EEG also may serve, in combination with other studies and
tumors (e.g., cortical localization or low-grade tumors) may clinical exam findings, as a surrogate marker of clinical
have the same effect on seizure occurrence in those patients outcome in patients after CA and after TH, although current
admitted in the ICU. Therefore EEG monitoring may be con- practice parameters do not recommend the use of EEG alone
sidered in brain tumor patients with altered mental status or for predicting prognosis. In cardiac arrest patients, a continu-
coma when admitted to the ICU. The duration of monitoring ous or reactive EEG background is associated with a good
should be 24 hours in noncomatose patients without evidence recovery.117 On the other hand, irreversible coma is usually
of ictal activity and more than 48 to 72 hours in comatose inevitable with a spontaneous burst-suppression EEG pattern,
patients. an unresponsive EEG pattern, or absent N20 on SSEPs.117,118
Similar observations have been made in children.119
Meningoencephalitis
Seizures can complicate bacterial, viral, or tuberculous men- Refractory Status Epilepticus
ingitis or encephalitis.104 Malaria also can cause seizures. In a Status epilepticus resistant to benzodiazepines and at least
prospective cohort of adult acute bacterial meningitis (ABM) one antiepileptic drug is defined as refractory, and guidelines
254 Section III—Electrophysiology

advise coma induction with midazolam, propofol, or barbitu-


rates.120 Coma induction itself may cause severe side effects.37 Table 25.3  Advantages and Disadvantages
So far, there are no randomized trials that show an outcome of Electroencephalography as a Monitor in
benefit for patients with RSE who undergo EEG monitoring; the Intensive Care Unit
the data are derived from prospective cohort trials, observa- Advantages Disadvantages
tional databases, and expert opinion. Nevertheless, iEEG
Sensitive to changes in Nonspecific as to etiology of
and cEEG can help detect seizure activity and manage the level neurologic functioning abnormality technical issues
of sedation. cEEG monitoring in RSE helps obtain early from variety of causes Difficult to implement
seizure suppression, maintain burst-suppression pattern if it Structural Requires skilled personnel
is refractory, and withdraw anesthetic drugs safely while mon- Metabolic 24 hr/day
itoring the possibility of seizure reappearance.3 It is not clear Large amount of data (data
storage and review)
whether complete suppression of cerebral electrogenesis,
simple seizure suppression, or burst-suppression pattern is Good spatial resolution Susceptible to artifacts
Monitors many brain areas Electrical
more effective.121,122 Some suggest it is advisable to maintain simultaneously Eye movements, ECG, EMG
a burst-to-suppression ratio of approximately 1 : 4. Others Excellent temporal resolution Electrode placement
advise a burst-suppression pattern of about 10 seconds of 2-4 ms
suppression for each burst of activity, ideally for at least 24 Bedside, noninvasive
hours.37,120,123 Barbiturates tend to be associated with higher ECG, Electrocardiogram; EMG, electromyogram.
likelihood of achieving burst suppression on EEG than pro-
pofol or midazolam, but their use is associated with longer
mechanical ventilation and hospital stay.121
review tools will further facilitate the integration of EEG mon-
itoring in the ICU. In addition, there are efforts to develop
Monitoring of Sedation Depth practical definitions of clinical relevant EEG findings in the
and Burst Suppression ICU.18 Table 25.3 summarizes the typical advantages and dis-
The BIS monitor was developed to assess the hypnotic com- advantages of EEG methodology as a monitor in the ICU.
ponent of general anesthesia.124 A good correlation between
the cEEG burst rate per minute and the BIS and suppression
ratio to assess the depth of propofol anesthesia in the treat-
Conclusion
ment of RSE is described, but the BIS monitor does recognize In summary, EEG and especially cEEG is a promising and
regional epileptic activity and epileptic activity during the evolving technique to assess neurologic status in the critically
burst-suppression pattern. Therefore cEEG should be consid- ill. It allows the noninvasive assessment of brain function over
ered the primary monitoring technique to assess the depth of a large surface area. Based on the knowledge gathered so far,
anesthesia in RSE. If cEEG is not available, a BIS monitor may EEG should be considered in those with acute brain injury,
be considered an alternative to assess the level of sedation impaired mental status, and unexplained alteration in con-
although its use in RSE still is controversial.69,124 BIS monitor- sciousness. Bedside EEG can help titrate therapy and repre-
ing has been used to guide sedation depth in patients with sents a valuable tool to detect secondary insults, such as
severe trauma. However, despite a high specificity (90%) for cerebral ischemia in at-risk patients. However, EEG monitor-
the detection of sedation changes, the interpretation of the BIS ing in the ICU is still an expensive cost-intensive technology
readings remains subjective and appears to be even more that requires well-trained, dedicated medical and technical
complex in TBI patients.125 staff. Continued education and research are necessary to
determine whether EEG leads to management and treatment
decisions that are associated with outcomes. Advances in
Logistics and Costs information technology may soon make real-time bedside
The routine application of EEG monitoring and especially monitoring of brain activity using cEEG possible.
continuous monitoring in the NCCU is still limited by the
high costs for technical equipment and personnel. Because
high-quality data recording and interpretation are necessary References
for any clinical application, a specialized EEG technician and 1. Tudor M, Tudor L, Tudor KI. [Hans Berger (1873-1941)—The history of
an experienced electroencephalographer are essential.126 In electroencephalography]. Acta Med Croatica 2005;59(4):307–13.
2. Claassen J, Mayer SA. Continuous electroencephalographic monitoring in
addition the integration of EEG monitoring into ICU care neurocritical care. Curr Neurol Neurosci Rep 2002;2:534–40.
may trigger EEG-driven clinical decision making and there- 3. Friedman D, Claassen J, Hirsch LJ. Continuous electroencephalogram
fore reduce the need for other diagnostic or imaging modali- monitoring in the intensive care unit. Anesth Analg 2009;109:506–23.
ties.4 Therefore cEEG implementation requires specific 4. Vespa PM, Nenov V, Nuwer MR. Continuous EEG monitoring in the
intensive care unit: early findings and clinical efficacy. J Clin Neurophysiol
training for nurses, technicians, and physicians.6 Because any 1999;16:1–13.
intervention can influence the EEG signal and there are 5. Horst ter HJ, van Olffen M, Remmelts HJ, et al. The prognostic value of
numerous sources for artifacts, the medical staff at bedside amplitude integrated EEG in neonatal sepsis and/or meningitis. Acta
should be trained to be able to detect and interpret common Paediatr 2010;99:194–200.
artifacts and to recognize basic EEG patterns and their dynam- 6. Kull LL, Emerson RG. Continuous EEG monitoring in the intensive care
unit: technical and staffing considerations. J Clin Neurophysiol 2005;22:
ics. Developments such as magnetic resonance imaging 107–18.
(MRI)–compatible electrodes127 and smaller recording and 7. Brenner RP, Schaul N. Periodic EEG patterns: classification, clinical
data storage systems and Web-based data access and data correlation, and pathophysiology. J Clin Neurophysiol 1990;7:249–67.
Section III—Electrophysiology 255

8. Andraus MEC, Andraus CF, Alves-Leon SV. Periodic EEG patterns: 28. Oertel M, Kelly DF, Lee JH, et al. Metabolic suppressive therapy as a
importance of their recognition and clinical significance. Arq Neuropsiquiatr treatment for intracranial hypertension: why it works and when it fails. Acta
2012;70:145–51. Neurochir Suppl 2002;81:69–70.
9. Holtkamp M, Meierkord H. Nonconvulsive status epilepticus: a diagnostic 29. Yemisci M, Gurer G, Saygi S, et al. Generalised periodic epileptiform
and therapeutic challenge in the intensive care setting. Ther Adv Neurol discharges: clinical features, neuroradiological evaluation and prognosis in
Disord 2011;4:169–81. 37 adult patients. Seizure 2003;12:465–72.
10. García-Morales I, García MT, Galán-Dávila L, et al. Periodic lateralized 30. Chong DJ, Hirsch LJ. Which EEG patterns warrant treatment in the critically
epileptiform discharges: etiology, clinical aspects, seizures, and evolution in ill? Reviewing the evidence for treatment of periodic epileptiform discharges
130 patients. J Clin Neurophysiol 2002;19:172–7. and related patterns. J Clin Neurophysiol 2005;22:79–91.
11. Snodgrass SM, Tsuburaya K, Ajmone-Marsan C. Clinical significance of 31. Strong AJ, Hartings JA, Dreier JP. Cortical spreading depression: an adverse
periodic lateralized epileptiform discharges: relationship with status but treatable factor in intensive care? Curr Opin Crit Care 2007;13:126–33.
epilepticus. J Clin Neurophysiol 1989;6:159–72. 32. Dreier JP: The role of spreading depression, spreading depolarization and
11a.  International League Against Epilepsy. Proposal for revised clinical spreading ischemia in neurological disease. Nat Med 2011;1–9.
and electroencephalographic classification of epileptic seizures. From the 33. Fabricius M, Fuhr S, Willumsen L, et al. Association of seizures with cortical
Commission on Classification and Terminology of the International League spreading depression and peri-infarct depolarisations in the acutely injured
Against Epilepsy. Epilepsia 1981;22(4):489–501. human brain. Clin Neurophysiol 2008;119:1973–84.
12. Rossetti AO, Kaplan PW. Seizure semiology: an overview of the ‘inverse 34. Claassen J, Mayer SA, Kowalski RG, et al. Detection of electrographic
problem’. Eur Neurol 2010;63:3–10. seizures with continuous EEG monitoring in critically ill patients. Neurology
13. Rossetti AO, Urbano LA, Delodder F, et al. Prognostic value of continuous 2004;62:1743–8.
EEG monitoring during therapeutic hypothermia after cardiac arrest. Crit 35. Hirsch LJ. Classification of EEG patterns in patients with impaired
Care 2010;14:R173. consciousness. Epilepsia 2011;52 (Suppl. 8):S21–4.
14. Meierkord H, Holtkamp M. Status epilepticus: an independent outcome 36. Oddo M, Carrera E, Claassen J, et al. Continuous electroencephalography in
predictor after cerebral anoxia. Neurology 2008;70:2015–6; author reply the medical intensive care unit. Crit Care Med 2009;37:2051–6.
2015–6. 37. Rossetti AO, Oddo M. The neuro-ICU patient and electroencephalography
15. Alldredge BK, Gelb AM, Isaacs SM, et al. A comparison of lorazepam, paroxysms: if and when to treat. Curr Opin Crit Care 2010;16:105–9.
diazepam, and placebo for the treatment of out-of-hospital status 38. Jordan D, Schneider G, Hock A, et al. EEG parameters and their
epilepticus. N Engl J Med 2001;345:631–7. combination as indicators of depth of anaesthesia. Biomed Tech (Berl)
16. Treiman DM, Meyers PD, Walton NY, et al. A comparison of four treatments 2006;51:89–94.
for generalized convulsive status epilepticus. N Engl J Med 1998;339:792–8. 39. Jordan KG. Continuous EEG monitoring in the neuroscience intensive care
17. Brenner RP. The interpretation of the EEG in stupor and coma. Neurologist unit and emergency department. J Clin Neurophysiol 1999;16:14–39.
2005;11:271–84. 40. Klem GH, Lüders HO, Jasper HH, et al. The ten-twenty electrode system
18. Hirsch LJ, Brenner RP, Drislane FW, et al. The ACNS Subcommittee On of the International Federation. The International Federation of Clinical
Research Terminology for Continuous EEG Monitoring. Proposed Neurophysiology. Electroencephalogr Clin Neurophysiol Suppl 1999;52:3–6.
standardized terminology for rhythmic and periodic EEG patterns 41. Vespa PM, Boscardin WJ, Hovda DA, et al. Early and persistent impaired
encountered in critically ill patients. J Clin Neurophysiol 2005;22:128–35. percent alpha variability on continuous electroencephalography monitoring
19. Jirsch J, Hirsch LJ. Nonconvulsive seizures: developing a rational approach to as predictive of poor outcome after traumatic brain injury. J Neurosurg
the diagnosis and management in the critically ill population. Clin 2002;97:84–92.
Neurophysiol 2007;118:1660–70. 42. Labar DR, Fisch BJ, Pedley TA, et al. Quantitative EEG monitoring for
20. Jordan KG. Neurophysiologic monitoring in the neuroscience intensive care patients with subarachnoid hemorrhage. Electroencephalogr Clin
unit. Neurol Clin 1995;13:579–626. Neurophysiol 1991;78:325–32.
21. Lowenstein DH, Aminoff MJ. Clinical and EEG features of status epilepticus 43. Wusthoff CJ, Shellhaas RA, Clancy RR. Limitations of single-channel EEG
in comatose patients. Neurology 1992;42:100–4. on the forehead for neonatal seizure detection. J Perinatol 2009;29:237–42.
22. Pohlmann-Eden B, Hoch DB, Cochius JI, et al. Periodic lateralized 44. Shellhaas RA, Soaita AI, Clancy RR. Sensitivity of amplitude-integrated
epileptiform discharges: a critical review. J Clin Neurophysiol electroencephalography for neonatal seizure detection. Pediatrics
1996;13:519–30. 2007;120:770–7.
23. Walker M, Cross H, Smith S, et al. Nonconvulsive status epilepticus: 45. Opherk C, Coromilas J, Hirsch LJ. Heart rate and EKG changes in 102
Epilepsy Research Foundation workshop reports. Epileptic Disord seizures: analysis of influencing factors. Epilepsy Res 2002;52:117–27.
2005;7(3):253–96. 46. Opherk C, Hirsch LJ. Ictal heart rate differentiates epileptic from non-
24. Vespa PM, McArthur DL, Xu Y, et al. Nonconvulsive seizures after traumatic epileptic seizures. Neurology 2002;58:636–8.
brain injury are associated with hippocampal atrophy. Neurology 2010;75: 47. Zijlmans M, Flanagan D, Gotman J. Heart rate changes and ECG
792–8. abnormalities during epileptic seizures: prevalence and definition of an
25. Vespa PM, O’Phelan K, Shah M, et al. Acute seizures after intracerebral objective clinical sign. Epilepsia 2002;43:847–54.
hemorrhage: a factor in progressive midline shift and outcome. Neurology 48. Stuart RM, Waziri A, Weintraub D, et al. Intracortical EEG for the detection
2003;60:1441–6. of vasospasm in patients with poor-grade subarachnoid hemorrhage.
26. Vespa P, Martin NA, Nenov V, et al. Delayed increase in extracellular Neurocrit Care 2010;13:355–8.
glycerol with post-traumatic electrographic epileptic activity: support for 49. Scheuer ML, Wilson SB. Data analysis for continuous EEG monitoring in the
the theory that seizures induce secondary injury. Acta Neurochir Suppl 2002; ICU: seeing the forest and the trees. J Clin Neurophysiol 2004;21:353–78.
81:355–7. 50. Rampil IJ. Elements of EEG signal processing. Int J Clin Monit Comput
27. Vespa PM, Miller C, McArthur D, et al. Nonconvulsive electrographic 1987;4:85–98.
seizures after traumatic brain injury result in a delayed, prolonged increase
in intracranial pressure and metabolic crisis. Crit Care Med 2007;35(12): A complete list of references for this chapter can be found online at
2830–6. www.expertconsult.com.
Section III—Electrophysiology 255.e1

References 26. Vespa P, Martin NA, Nenov V, et al. Delayed increase in extracellular
glycerol with post-traumatic electrographic epileptic activity: support for
1. Tudor M, Tudor L, Tudor KI. [Hans Berger (1873-1941)—The history of the theory that seizures induce secondary injury. Acta Neurochir Suppl
electroencephalography]. Acta Med Croatica 2005;59(4):307–13. 2002;81:355–7.
2. Claassen J, Mayer SA. Continuous electroencephalographic monitoring in 27. Vespa PM, Miller C, McArthur D, et al. Nonconvulsive electrographic
neurocritical care. Curr Neurol Neurosci Rep 2002;2:534–40. seizures after traumatic brain injury result in a delayed, prolonged increase
3. Friedman D, Claassen J, Hirsch LJ. Continuous electroencephalogram in intracranial pressure and metabolic crisis. Crit Care Med 2007;35(12):
monitoring in the intensive care unit. Anesth Analg 2009;109:506–23. 2830–6.
4. Vespa PM, Nenov V, Nuwer MR. Continuous EEG monitoring in the 28. Oertel M, Kelly DF, Lee JH, et al. Metabolic suppressive therapy as a
intensive care unit: early findings and clinical efficacy. J Clin Neurophysiol treatment for intracranial hypertension: why it works and when it fails.
1999;16:1–13. Acta Neurochir Suppl 2002;81:69–70.
5. Horst ter HJ, van Olffen M, Remmelts HJ, et al. The prognostic value of 29. Yemisci M, Gurer G, Saygi S, et al. Generalised periodic epileptiform
amplitude integrated EEG in neonatal sepsis and/or meningitis. Acta discharges: clinical features, neuroradiological evaluation and prognosis in
Paediatr 2010;99:194–200. 37 adult patients. Seizure 2003;12:465–72.
6. Kull LL, Emerson RG. Continuous EEG monitoring in the intensive care 30. Chong DJ, Hirsch LJ. Which EEG patterns warrant treatment in the
unit: technical and staffing considerations. J Clin Neurophysiol 2005;22: critically ill? Reviewing the evidence for treatment of periodic epileptiform
107–18. discharges and related patterns. J Clin Neurophysiol 2005;22:79–91.
7. Brenner RP, Schaul N. Periodic EEG patterns: classification, clinical 31. Strong AJ, Hartings JA, Dreier JP. Cortical spreading depression: an adverse
correlation, and pathophysiology. J Clin Neurophysiol 1990;7:249–67. but treatable factor in intensive care? Curr Opin Crit Care 2007;13:126–33.
8. Andraus MEC, Andraus CF, Alves-Leon SV. Periodic EEG patterns: 32. Dreier JP. The role of spreading depression, spreading depolarization and
importance of their recognition and clinical significance. Arq spreading ischemia in neurological disease. Nat Med 2011;1–9.
Neuropsiquiatr 2012;70:145–51. 33. Fabricius M, Fuhr S, Willumsen L, et al. Association of seizures with
9. Holtkamp M, Meierkord H. Nonconvulsive status epilepticus: a diagnostic cortical spreading depression and peri-infarct depolarisations in the acutely
and therapeutic challenge in the intensive care setting. Ther Adv Neurol injured human brain. Clin Neurophysiol 2008;119:1973–84.
Disord 2011;4:169–81. 34. Claassen J, Mayer SA, Kowalski RG, et al. Detection of electrographic
10. García-Morales I, García MT, Galán-Dávila L, et al. Periodic lateralized seizures with continuous EEG monitoring in critically ill patients.
epileptiform discharges: etiology, clinical aspects, seizures, and evolution in Neurology 2004;62:1743–8.
130 patients. J Clin Neurophysiol 2002;19:172–7. 35. Hirsch LJ. Classification of EEG patterns in patients with impaired
11. Snodgrass SM, Tsuburaya K, Ajmone-Marsan C. Clinical significance of consciousness. Epilepsia 2011;52 (Suppl. 8):S21–4.
periodic lateralized epileptiform discharges: relationship with status 36. Oddo M, Carrera E, Claassen J, et al. Continuous electroencephalography
epilepticus. J Clin Neurophysiol 1989;6:159–72. in the medical intensive care unit. Crit Care Med 2009;37:2051–6.
11a.  International League Against Epilepsy. Proposal for revised clinical 37. Rossetti AO, Oddo M. The neuro-ICU patient and electroencephalography
and electroencephalographic classification of epileptic seizures. From the paroxysms: if and when to treat. Curr Opin Crit Care 2010;16:105–9.
Commission on Classification and Terminology of the International League 38. Jordan D, Schneider G, Hock A, et al. EEG parameters and their
Against Epilepsy. Epilepsia 1981;22(4):489–501. combination as indicators of depth of anaesthesia. Biomed Tech (Berl)
12. Rossetti AO, Kaplan PW. Seizure semiology: an overview of the ‘inverse 2006;51:89–94.
problem’. Eur Neurol 2010;63:3–10. 39. Jordan KG. Continuous EEG monitoring in the neuroscience intensive care
13. Rossetti AO, Urbano LA, Delodder F, et al. Prognostic value of continuous unit and emergency department. J Clin Neurophysiol 1999;16:14–39.
EEG monitoring during therapeutic hypothermia after cardiac arrest. Crit 40. Klem GH, Lüders HO, Jasper HH, et al. The ten-twenty electrode system of
Care 2010;14:R173. the International Federation. The International Federation of Clinical
14. Meierkord H, Holtkamp M. Status epilepticus: an independent outcome Neurophysiology. Electroencephalogr Clin Neurophysiol Suppl 1999;52:3–6.
predictor after cerebral anoxia. Neurology 2008;70:2015–6; author reply 41. Vespa PM, Boscardin WJ, Hovda DA, et al. Early and persistent impaired
2015–6. percent alpha variability on continuous electroencephalography monitoring
15. Alldredge BK, Gelb AM, Isaacs SM, et al. A comparison of lorazepam, as predictive of poor outcome after traumatic brain injury. J Neurosurg
diazepam, and placebo for the treatment of out-of-hospital status 2002;97:84–92.
epilepticus. N Engl J Med 2001;345:631–7. 42. Labar DR, Fisch BJ, Pedley TA, et al. Quantitative EEG monitoring for
16. Treiman DM, Meyers PD, Walton NY, et al. A comparison of four patients with subarachnoid hemorrhage. Electroencephalogr Clin
treatments for generalized convulsive status epilepticus. N Engl J Med Neurophysiol 1991;78:325–32.
1998;339:792–8. 43. Wusthoff CJ, Shellhaas RA, Clancy RR. Limitations of single-channel EEG
17. Brenner RP. The interpretation of the EEG in stupor and coma. Neurologist on the forehead for neonatal seizure detection. J Perinatol 2009;29:237–42.
2005;11:271–84. 44. Shellhaas RA, Soaita AI, Clancy RR. Sensitivity of amplitude-integrated
18. Hirsch LJ, Brenner RP, Drislane FW, et al. The ACNS Subcommittee electroencephalography for neonatal seizure detection. Pediatrics 2007;120:
on Research Terminology for Continuous EEG Monitoring. Proposed 770–7.
standardized terminology for rhythmic and periodic EEG patterns 45. Opherk C, Coromilas J, Hirsch LJ. Heart rate and EKG changes in 102
encountered in critically ill patients. J Clin Neurophysiol 2005;22: seizures: analysis of influencing factors. Epilepsy Res 2002;52:117–27.
128–35. 46. Opherk C, Hirsch LJ. Ictal heart rate differentiates epileptic from
19. Jirsch J, Hirsch LJ. Nonconvulsive seizures: developing a rational approach non-epileptic seizures. Neurology 2002;58:636–8.
to the diagnosis and management in the critically ill population. Clin 47. Zijlmans M, Flanagan D, Gotman J. Heart rate changes and ECG
Neurophysiol 2007;118:1660–70. abnormalities during epileptic seizures: prevalence and definition of an
20. Jordan KG. Neurophysiologic monitoring in the neuroscience intensive care objective clinical sign. Epilepsia 2002;43:847–54.
unit. Neurol Clin 1995;13:579–626. 48. Stuart RM, Waziri A, Weintraub D, et al. Intracortical EEG for the detection
21. Lowenstein DH, Aminoff MJ. Clinical and EEG features of status of vasospasm in patients with poor-grade subarachnoid hemorrhage.
epilepticus in comatose patients. Neurology 1992;42:100–4. Neurocrit Care 2010;13:355–8.
22. Pohlmann-Eden B, Hoch DB, Cochius JI, et al. Periodic lateralized 49. Scheuer ML, Wilson SB. Data analysis for continuous EEG monitoring in
epileptiform discharges: a critical review. J Clin Neurophysiol 1996;13: the ICU: seeing the forest and the trees. J Clin Neurophysiol 2004;21:
519–30. 353–78.
23. Walker M, Cross H, Smith S, et al. Nonconvulsive status epilepticus: 50. Rampil IJ. Elements of EEG signal processing. Int J Clin Monit Comput
Epilepsy Research Foundation workshop reports. Epileptic Disord 1987;4:85–98.
2005;7(3):253–96. 51. Rampil IJ. A primer for EEG signal processing in anesthesia. Anesthesiology
24. Vespa PM, McArthur DL, Xu Y, et al. Nonconvulsive seizures after 1998;89:980–1002.
traumatic brain injury are associated with hippocampal atrophy. Neurology 52. Naqeeb al N, Edwards AD, Cowan FM, et al. Assessment of neonatal
2010;75:792–8. encephalopathy by amplitude-integrated electroencephalography. Pediatrics
25. Vespa PM, O’Phelan K, Shah M, et al. Acute seizures after intracerebral 1999;103:1263–71.
hemorrhage: a factor in progressive midline shift and outcome. Neurology 53. Wilson SB, Scheuer ML, Emerson RG, et al. Seizure detection: evaluation of
2003;60:1441–6. the Reveal algorithm. Clin Neurophysiol 2004;115:2280–91.
255.e2 Section III—Electrophysiology

54. Bricolo A, Turazzi S, Faccioli F, et al. Clinical application of compressed 80. Dennis LJ, Claassen J, Hirsch LJ, et al. Nonconvulsive status epilepticus
spectral array in long-term EEG monitoring of comatose patients. after subarachnoid hemorrhage. Neurosurgery 2002;51:1136–43; discussion
Electroencephalogr Clin Neurophysiol 1978;45:211–25. 1144.
55. Claassen J, Mayer SA, Hirsch LJ. Continuous EEG monitoring in 81. Rivierez M, Landau-Ferey J, Grob R, et al. Value of electroencephalogram in
patients with subarachnoid hemorrhage. J Clin Neurophysiol 2005;22: prediction and diagnosis of vasospasm after intracranial aneurysm rupture.
92–8. Acta Neurochir 1991;110:17–23.
56. Abend NS, Dlugos D, Herman S. Neonatal seizure detection using 82. Vespa PM, Nuwer MR, Juhász C, et al. Early detection of vasospasm after
multichannel display of envelope trend. Epilepsia 2008;49:349–52. acute subarachnoid hemorrhage using continuous EEG ICU monitoring.
57. Friedman DE, Schevon C, Emerson RG, et al. Cyclic electrographic seizures Electroencephalogr Clin Neurophysiol 1997;103:607–15.
in critically ill patients. Epilepsia 2008;49:281–7. 83. Claassen J, Hirsch LJ, Kreiter KT, et al. Quantitative continuous EEG
58. Pandian JD, Cascino GD, So EL, et al. Digital video-electroencephalographic for detecting delayed cerebral ischemia in patients with poor-grade
monitoring in the neurological-neurosurgical intensive care unit: clinical subarachnoid hemorrhage. Clin Neurophysiol 2004;115:2699–710.
features and outcome. Arch Neurol 2004;61:1090–4. 84. Dreier JP, Major S, Manning A, et al. Cortical spreading ischaemia is a
59. Sutter R, Fuhr P, Grize L, et al. Continuous video-EEG monitoring novel process involved in ischaemic damage in patients with aneurysmal
increases detection rate of nonconvulsive status epilepticus in the ICU. subarachnoid haemorrhage. Brain 2009;132:1866–81.
Epilepsia 2011;52:453–7. 85. Mutch WAC. New concepts regarding cerebral vasospasm: glial-centric
60. Winkler I, Haufe S, Tangermann M. Automatic classification of artifactual mechanisms. Can J Anesth/J Can Anesth 2010;57:479–89.
ICA-components for artifact removal in EEG signals. Behav Brain Funct 86. Dreier JP, Woitzik J, Fabricius M, et al. Delayed ischaemic neurological
2011;7:30. deficits after subarachnoid haemorrhage are associated with clusters of
61. Iwasaki M, Kellinghaus C, Alexopoulos AV, et al. Effects of eyelid closure, spreading depolarizations. Brain 2006;129:3224–37.
blinks, and eye movements on the electroencephalogram. Clin 87. Rosengart AJ, Huo JD, Tolentino J, et al. Outcome in patients with
Neurophysiol 2005;116:878–85. subarachnoid hemorrhage treated with antiepileptic drugs. J Neurosurg
62. Goncharova II, McFarland DJ, Vaughan TM, et al. EMG contamination of 2007;107:253–60.
EEG: spectral and topographical characteristics. Clin Neurophysiol 2003; 88. Claassen J, Jetté N, Chum F, et al. Electrographic seizures and periodic
114:1580–93. discharges after intracerebral hemorrhage. Neurology 2007;69:1356–65.
63. Barbati G, Porcaro C, Zappasodi F, et al. Optimization of an independent 89. Messé SR, Sansing LH, Cucchiara BL, et al. Prophylactic antiepileptic drug
component analysis approach for artifact identification and removal in use is associated with poor outcome following ICH. Neurocrit Care 2009;
magnetoencephalographic signals. Clin Neurophysiol 2004;115:1220–32. 11:38–44.
64. Gao JF, Yang Y, Lin P, et al. Automatic removal of eye-movement and blink 90. Rundgren M, Westhall E, Cronberg T, et al. Continuous amplitude-
artifacts from EEG signals. Brain Topogr 2010;23:105–14. integrated electroencephalogram predicts outcome in hypothermia-treated
65. Choi D, Ryu Y, Lee Y, et al. Performance evaluation of a motor-imagery- cardiac arrest patients. Critical Care Medicine 2010;38:1838–44.
based EEG-Brain computer interface using a combined cue with 91. Rossetti AO, Oddo M, Logroscino G, et al. Prognostication after cardiac
heterogeneous training data in BCI-naive subjects. Biomed Eng Online arrest and hypothermia: a prospective study. Ann Neurol 2010;67:
2011;10:91. 301–7.
66. Furdea A, Ruf CA, Halder S, et al. A new (semantic) reflexive brain- 92. Legriel S, Bruneel F, Sediri H, et al. Early EEG monitoring for detecting
computer interface: in search for a suitable classifier. J Neurosci Methods postanoxic status epilepticus during therapeutic hypothermia: a pilot study.
2012;203:233–40. Neurocrit Care 2009;11:338–44.
67. Brunner C, Billinger M, Vidaurre C, et al. A comparison of univariate, 93. Abend NS, Topjian A, Ichord R, et al. Electroencephalographic monitoring
vector, bilinear autoregressive, and band power features for brain-computer during hypothermia after pediatric cardiac arrest. Neurology 2009;72:
interfaces. Med Biol Eng Comput 2011;49:1337–46. 1931–40.
68. Scheuer ML. Continuous EEG monitoring in the intensive care unit. 94. Rossetti AO, Logroscino G, Liaudet L, et al. Status epilepticus: an
Epilepsia 2002;43(Suppl. 3):S114–27. independent outcome predictor after cerebral anoxia. Neurology 2007;69:
69. Musialowicz T, Mervaala E, Kälviäinen R, et al. Can BIS monitoring be used 255–60.
to assess the depth of propofol anesthesia in the treatment of refractory 95. Rossetti AO, Oddo M, Liaudet L, et al. Predictors of awakening from
status epilepticus? Epilepsia 2010;51:1580–6. postanoxic status epilepticus after therapeutic hypothermia. Neurology
70. Shellhaas RA, Gallagher PR, Clancy RR. Assessment of neonatal 2009;72:744–9.
electroencephalography (EEG) background by conventional and two 96. Rothstein TL, Thomas EM, Sumi SM. Predicting outcome in hypoxic-
amplitude-integrated EEG classification systems. J Pediatr 2008;153: ischemic coma: a prospective clinical and electrophysiologic study.
369–74. Electroencephalogr Clin Neurophysiol 1991;79:101–7.
71. Vespa PM, Nuwer MR, Nenov V, et al. Increased incidence and impact of 97. Fugate JE, Wijdicks EFM, Mandrekar J, et al. Predictors of neurologic
nonconvulsive and convulsive seizures after traumatic brain injury as outcome in hypothermia after cardiac arrest. Ann Neurol 2010;68:
detected by continuous electroencephalographic monitoring. J Neurosurg 907–14.
1999;91:750–60. 98. Kaplan PW, Genoud D, Ho TW, et al. Etiology, neurologic correlations,
72. Ronne-Engstrom E, Winkler T. Continuous EEG monitoring in patients and prognosis in alpha coma. Clin Neurophysiol 1999;110:205–13.
with traumatic brain injury reveals a high incidence of epileptiform 99. Simbruner G, Mittal RA, Rohlmann F, et al. Systemic hypothermia after
activity. Acta Neurol Scand 2006;114:47–53. neonatal encephalopathy: outcomes of neo.nEURO.network RCT. Pediatrics
73. Amantini A, Fossi S, Grippo A, et al. Continuous EEG-SEP monitoring in 2010;126:e771–8.
severe brain injury. Neurophysiol Clin 2009;39:85–93. 100. Wusthoff CJ, Dlugos DJ, Gutierrez-Colina A, et al. Electrographic seizures
74. Steudel WI, Krüger J. Using the spectral analysis of the EEG for prognosis during therapeutic hypothermia for neonatal hypoxic-ischemic
of severe brain injuries in the first post-traumatic week. Acta Neurochir encephalopathy. J Child Neurol 2011;26:724–8.
Suppl 1979;28:40–2. 101. Shankaran S, Pappas A, McDonald SA, et al. Predictive value of an early
75. Beridze M, Khaburzania M, Shakarishvili R, et al. Dominated EEG patterns amplitude integrated electroencephalogram and neurologic examination.
and their prognostic value in coma caused by traumatic brain injury. Pediatrics 2011;128:e112–20.
Georgian Med News 2010;28–33. 102. Brogna C, Gil Robles S, Duffau H: Brain tumors and epilepsy. Expert Rev
76. Gütling E, Gonser A, Imhof HG, et al. EEG reactivity in the prognosis of Neurother 2008;8:941–55.
severe head injury. Neurology 1995;45:915–18. 103. Bladin CF, Alexandrov AV, Bellavance A, et al. Seizures after stroke:
77. Butzkueven H, Evans AH, Pitman A, et al. Onset seizures independently a prospective multicenter study. Arch Neurol 2000;57:1617–22.
predict poor outcome after subarachnoid hemorrhage. Neurology 2000;55: 104. Edmond K, Clark A, Korczak VS, et al. Global and regional risk of disabling
1315–20. sequelae from bacterial meningitis: a systematic review and meta-analysis.
78. Claassen J, Hirsch LJ, Frontera JA, et al. Prognostic significance of Lancet Infect Dis 2010;10:317–28.
continuous EEG monitoring in patients with poor-grade subarachnoid 105. Zoons E, Weisfelt M, de Gans J, et al. Seizures in adults with bacterial
hemorrhage. Neurocrit Care 2006;4:103–12. meningitis. Neurology 2008;70:2109–15.
79. Little AS, Kerrigan JF, McDougall CG, et al. Nonconvulsive status 106. Vibha D, Bhatia R, Prasad K, et al. Clinical features and independent
epilepticus in patients suffering spontaneous subarachnoid hemorrhage. prognostic factors for acute bacterial meningitis in adults. Neurocrit Care
J Neurosurg 2007;106:805–11. 2010;13:199–204.
Section III—Electrophysiology 255.e3

107. Flores-Cordero JM, Amaya-Villar R, Rincón-Ferrari MD, et al. Acute 116. Tobias JD. Bispectral index monitoring documents burst suppression
community-acquired bacterial meningitis in adults admitted to the during pentobarbital coma. J Intensive Care Med 2008;23:258–62.
intensive care unit: clinical manifestations, management and prognostic 117. Oddo M, Rossetti AO. Predicting neurological outcome after cardiac arrest.
factors. Intensive Care Med 2003;29:1967–73. Current Opinion in Critical Care 2011;17:254–9.
108. Horst ter HJ, Jongbloed-Pereboom M, van Eykern LA, et al. Amplitude- 118. Rabinstein AA, Wijdicks EFM. The value of EEG monitoring after cardiac
integrated electroencephalographic activity is suppressed in preterm arrest treated with hypothermia. Neurology 2012;78:774–5.
infants with high scores on illness severity. Early Hum Dev 2011;87: 119. Kessler SK, Topjian AA, Gutierrez-Colina AM, et al. Short-term outcome
385–90. prediction by electroencephalographic features in children treated with
109. Klinger G, Chin CN, Otsubo H, et al. Prognostic value of EEG in neonatal therapeutic hypothermia after cardiac arrest. Neurocrit Care 2011;14:37–43.
bacterial meningitis. Pediatr Neurol 2001;24:28–31. 120. Meierkord H, Boon P, Engelsen B, et al. EFNS guideline on the
110. Bergen DC, Ristanovic R, Gorelick PB, et al. Seizures and renal failures. Int management of status epilepticus. Eur J Neurol 2006;13:445–50.
J Artif Organs 1994;17:247–51. 121. Rossetti AO, Logroscino G, Bromfield EB. Refractory status epilepticus:
111. Guerit J-M, Amantini A, Fischer C, et al. Neurophysiological investigations effect of treatment aggressiveness on prognosis. Arch Neurol
of hepatic encephalopathy: ISHEN practice guidelines. Liver Int 2005;62:1698–702.
2009;29:789–96. 122. Krishnamurthy KB, Drislane FW. Depth of EEG suppression and outcome
112. Seder DB, Fraser GL, Robbins T, et al. The bispectral index and suppression in barbiturate anesthetic treatment for refractory status epilepticus.
ratio are very early predictors of neurological outcome during therapeutic Epilepsia 1999;40:759–62.
hypothermia after cardiac arrest. Intensive Care Med 2010;36:281–8. 123. Meierkord H, Boon P, Engelsen B, et al. EFNS guideline on the
113. Chollet-Xémard C, Combes X, Soupizet F, et al. Bispectral index management of status epilepticus in adults. Euro J Neurol 2010;17(3):
monitoring is useless during cardiac arrest patients’ resuscitation. 348–55.
Resuscitation 2009;80:213–16. 124. Johansen JW. Update on bispectral index monitoring. Best Pract Res Clin
114. Young GB, Jordan KG, Doig GS. An assessment of nonconvulsive seizures Anaesthesiol 2006;20:81–99.
in the intensive care unit using continuous EEG monitoring: an 125. Ogilvie MP, Pereira BMT, Ryan ML, et al. Bispectral index to monitor
investigation of variables associated with mortality. Neurology propofol sedation in trauma patients. J Trauma 2011;71:1415–21.
1996;47:83–9. 126. Young GB. Continuous EEG monitoring in the ICU. Acta Neurol Scand
115. Riker RR, Fraser GL, Wilkins ML. Comparing the bispectral index and 2006;114:67–8.
suppression ratio with burst suppression of the electroencephalogram 127. Mirsattari SM, Lee DH, Jones D, et al. MRI compatible EEG electrode
during pentobarbital infusions in adult intensive care patients. system for routine use in the epilepsy monitoring unit and intensive care
Pharmacotherapy 2003;23:1087–93. unit. Clin Neurophysiol 2004;115:2175–80.
IV
Chapter
26  

Computed Tomography
Asako Miyakoshi and Wendy A. Cohen

is a standard tool to image intracranial and extracranial vas-


Introduction culature in nontraumatized patients and is frequently used in
Computed tomography (CT)–based imaging technology con- patients with acute ischemic infarcts who also undergo MRI
tinues to be the principal imaging modality in patients who when being evaluated for urgent intra-arterial thrombolysis.
require neurocritical care. CT imaging can monitor intracra- In these patients magnetic resonance perfusion (MRP) is
nial events, CT angiography (CTA) can demonstrate intracra- used to define areas of abnormal perfusion not yet infarcted
nial and extracranial vasculature, and CT perfusion (CTP) can (diffusion-perfusion mismatch).3 In addition, MRA can be
provide measures of intracranial blood flow. All of these useful to assess vascular dissections.4 However, MRA and MRP
studies are done on multislice CT scanners using commer- have the technical limitations of MRI in a patient on life
cially available software and can be performed on portable support.
scanners housed in the neurocritical care unit (NCCU) (i.e., Cerebral perfusion also is measured with nonradioactive
point-of-care assessment).1 Noncontrast head CT shows ana- xenon-CT (Xe-CT). The advantage of stable Xe-CT is the
tomic intracranial injury: hemorrhage, hydrocephalus, contu- possibility to measure regional cerebral blood flow in a quan-
sion, mass effect, shifts, ischemia, infarction, shear injury, and titative manner. During a Xe-CT, xenon gas is inhaled in
diffuse axonal injury (DAI). Contrast-enhanced head CT facil- combination with oxygen over 5 minutes during CT scan-
itates identification of medium to large vascular abnormalities ning. Xenon is lipid-soluble and passes freely though the
(although typically not aneurysms), masses, and blood-brain blood-brain barrier. Therefore brain, venous, and end-tidal
barrier disruption. Contrast also is required for CTA and CTP. xenon concentrations are assumed to be similar. Xenon is
CTA may show vascular occlusion in a patient with an acute radiodense, and the density of xenon on a CT image has
infarct, the source of an intraparenchymal hemorrhage, or been shown to correlate with end-tidal xenon concentration.
vasospasm following subarachnoid hemorrhage (SAH). CTP Calculation of cerebral blood flow (CBF) is based on use of
is used to evaluate infarct core versus penumbra in a patient the Fick equation. Accuracy of CBF measurement using
with an acute infarct, to show areas of reduced blood flow in stable xenon has been confirmed by comparison to radiola-
a patient with vasospasm, and to demonstrate the presence or beled microsphere studies.5,6 Xe-CT has some inherent limi-
absence of autoregulation following traumatic brain injury tations. Side effects of xenon inhalation include sedation,
(TBI). bronchospasm, and respiratory depression. Xenon blood
flow measurements require specialized equipment to deliver
the gas and to measure the end-tidal xenon concentration.
A Brief Comparison Because the gas is sedating, some patients have difficulty
with Other Techniques lying still for the 5-minute scan acquisition. Availability of
xenon gas is variable in the United States and at the time of
Other imaging and monitoring techniques can provide similar publication is used under research protocols.7,8
information to that obtained by CT (See also Chapters Transcranial Doppler (TCD) is used to evaluate cerebral
27-29). This information can complement or supplement the perfusion by measurement of flow velocities in first- and
CT data. The presence of an ischemic infarct or the extent of second-order vessels. Access depends on adequate acoustic
injury with DAI is more accurately determined using windows and accuracy may be operator dependent.7 An
magnetic resonance imaging (MRI). Disadvantages of MRI advantage to TCD is ease of use (typically bedside) and
when compared with CT include the limitations in identi­ repeatability.
fication of acute intracranial hemorrhage, difficulty moni­ Single photon emission computed tomography (SPECT)
toring an unstable patient in the MRI suite because of using Tc-99m ethyl cysteinate dimer (ECD) or Tc-99m hexa-
requirements for nonferromagnetic life-support devices, methyl propyleneamine oxime (HMPAO) provides a qualita-
longer imaging times with MRI, and the relatively greater tive evaluation of cerebral perfusion. The isotope is bound on
expense of MRI equipment. The additional information the first pass through the brain. Evaluation of supratentorial
from an MRI does not usually change acute management in flow is made in relation to cerebellar flow. Repeat evaluation
patients with TBI.2 Magnetic resonance angiography (MRA) cannot occur until after the technetium isotope has decayed.
258 © Copyright 2013 Elsevier Inc. All rights reserved.
Section IV—Radiology 259

Techniques Evaluation of CTA examinations should include the source


images as well as the MIPs and the 3-D reformations.11,12
Computed Tomography (Nonenhanced CT) Source images show small vascular wall abnormalities. The
Cross-sectional images of the head from a nonenhanced head reformations show the anatomic relationships of the vessels.
CT scan show the major intracranial anatomy (ventricles, cis- Atherosclerotic narrowing of a vessel may be more easily
terns, sulci, gray matter vs. white matter borders) and the skull appreciated on the source image and may be averaged out on
anatomy. A head CT demonstrates the presence of acute intra- an MIP or 3-D reformation. Conversely the caudal-pointing
cranial hemorrhage more reliably than any other imaging dome of an anterior communicating artery aneurysm or
examination. Other pathologic processes that can be identi- cranial-directed dome of a basilar tip aneurysm may be dif-
fied include midline shift, mass effect, hydrocephalus, intra- ficult to define on axial source images but will be clearly
cranial edema, and encephalomalacia. Skull and facial fractures shown on the reformations. The common artifacts in a CTA
are shown from the same scan acquisition using windows and examination arise from patient motion, which causes misreg-
reconstruction algorithms maximized for bone. The only con- istration in the MIPs and in the 3-D reconstructions. An inad-
traindication to a head CT is an inability of the patient to lie equate contrast bolus caused by a mistimed contrast injection
on the CT table. In the setting of acute intracranial decom- or due to poor cardiac output can occur but is less common.
pensation, pregnancy or pediatric age become relative consid- Numerous studies have documented the accuracy of CTA for
erations or contraindications because CT requires the use of aneurysms greater than 4 to 5 mm in diameter. Aneurysms
ionizing radiation. A head CT can be acquired in 1 to 2 3 mm or less are missed 20% of the time.13,14
minutes using current-generation scanners. Life support
equipment, including intracranial monitoring systems, is
easily accommodated. Scans are typically displayed in an axial Computed Tomography Perfusion
projection but can be reconstructed into other planes, usually Calculation of CTP parameters is based on the central volume
coronal or sagittal. principle, represented by the following equation:
A head CT does not show vascular structures, may not
CBF = CBV/MTT
show masses, may not easily aid identification of subacute-
to-chronic hemorrhage, may not demonstrate demyelination, where CBF equals cerebral blood flow (mL/100 g/min−1),
and may not identify intracranial infection. Small infarcts or CBV equals cerebral blood volume (mL/100 g), and MTT
new infarcts in patients with prior ischemic areas may be hard equals mean transit time (seconds, time of the bolus passing
to visualize. Depending on the clinical question CT with iodin- through a volume of tissue). One commonly used method
ated contrast, MRI without or with gadolinium-based contrast, uses deconvolution, a mathematical process, to derive these
or vascular imaging using either CTA, MRA, or catheter angi- parameters from the time density curve generated by a first-
ography may be a more informative imaging procedure. pass measurement of contrast through the brain. MTT is cal-
culated from the arterial input time-density curve, CBV is
calculated as the area under time-density curve in the tissue
Computed Tomography Angiography of interest, and CBF is determined using the central volume
CTA scans follow the first pass of a contrast load as it moves principle.15,16 The density of the iodinated contrast is corre-
through the vascular tree of the scanned anatomy. The scan lated to the volume of contrast in a region. Another CT-based
acquisition is typically timed to be 1 to 2 seconds behind the technique uses a maximum slope model from this calcula-
arterial peak of the contrast bolus. The smallest available slice tion.17 There is also variation between commercial vendors in
is used for the scan acquisition to improve detection of small the implementation of the deconvolution algorithm. This
vascular abnormalities and to improve the three-dimensional results in variability of the quantitative results from CT per-
(3-D) reformations of the vascular volumes. Scans can be fusion calculations.18 Older techniques to measure cerebral
viewed as the initial acquisition (source images), as maximum perfusion used tracers that were freely diffusible across the
intensity projections (MIPs) often thicker than the initial scan blood-brain barrier and not limited to the intravascular space
acquisition, or as true 3-D volumes. MIPs usually include the (such as xenon in radioactive and nonradioactive forms).
surrounding brain and bone. This is a disadvantage when Iodinated contrast in contradistinction to xenon remains
evaluating the cavernous segments of the internal carotid intravascular under normal circumstances. Actual perfusion
arteries or the vertebral arteries within the foramen transver- of brain parenchyma is not shown. Although data obtained
saria of the cervical vertebrae. MIPs facilitate visualization of from Xe-CT cannot be directly applied to CTP due to techni-
longer segments of the vessel because the entire diameter of cal differences, generally good correlation between CTP and
the vessel is included in the volume and MIPs can be obtained Xe-CT is reported.19
in any desired orientation. Intracranial vessels are well seen The advantage of CTP is widespread availability and rela-
with a combination of coronal, axial, and sagittal projections. tively low cost. CTP can be obtained on any multislice CT
Extracranial carotid and vertebral arteries may be better scanner with appropriate software availability. A disadvantage
evaluated using coronal, oblique, and sagittal reformations. of CTP is the limited anatomic coverage of the brain using
Three-dimensional volume reformations eliminate the sur- 16- to 128-slice scanners. The length of the CT detector deter-
rounding bone and brain and are rotated in space, either as mines the volume of brain evaluated on a single CTP acquisi-
single images or as a cine loop. Fuzzy connectedness also may tion. Attempts to move the table during the CTP acquisition
be used to eliminate bony structures at the skull base.9 However, are difficult to implement and have not been generally adopted
the diagnostic accuracy of bone-subtracted CTA often is no (toggle table). The posterior fossa is difficult to evaluate during
better than careful analysis of 3-D volume-rendered CTA a CTP study because of artifact from the bone in the skull base
when source data are assessed in multiplanar reformats.10 and because it is difficult to image the posterior fossa without
260 Section IV—Radiology

scanning through the lens of the eye. Gantry angulation can tomography (SPECT), is higher.31,32 For comparison, back-
eliminate the direct dose to the eyes while including the top ground radiation dose over a year in approximately 3 mSv, the
of the cerebellum and the occipital lobes in the imaging plane. dose from a chest x-ray is 0.1 mSv, and the dose during a
This angulation is usually not able to include the lower por- roundtrip flight from Los Angeles to New York is 0.03 mSv.
tions of the posterior fossa. Iodinated contrast intravenously administered increases
Although CTP is described as quantitative, postprocessing serum creatinine.33 Patients with compromised renal function
techniques can affect the measurements. Issues of input vessel may be at greater risk.34 CTA usually requires 60 to 100 mL
selection, vessel opacification in the area being evaluated, and of contrast, CTP 40 to 50 mL per anatomic level evaluated.
variation in acquisition software remain.20-22 Postprocessing The significance of these contrast doses depends on the
can be done manually or automatically. Interobserver agree- patient’s underlying renal function, total amount of contrast
ment in measurements of CBF, CBV, MTT and volume of given, and the timing of those doses. For example, if a patient
infarct core and penumbra is better with automatically post- has an acute SAH, the following studies may happen in rapid
processed data.23,24 CTP calculations also are based on the succession: CTA and diagnostic catheter angiography without
assumption of an intact blood-brain barrier. Many studies or with aneurysm coiling, each with a significant dye load. In
use internal comparisons to evaluate CBF, CBV, and MTT a patient admitted for polytrauma, admission studies may
rather than the numerical values and reproducibility of include CTA of the cranial and extracranial cervical vessels,
quantitative assessment has been questioned, especially in CTA of aorta and peripheral vessels, abdominal CT with con-
processes such as acute ischemic infarct.25 trast to evaluate for abdominal injury, and possibly cranial
Blood flow in normal mixed gray and white matter is 50 to CTP to evaluate the presence of absence of cerebral autoregu-
80 mL/100 g brain tissue/min. CBF is higher in the gray lation. A patient with an acute infarct is likely to have a more
matter compared with white matter. CBF is kept relatively limited set of studies: CTA and CTP. Only the minority who
constant by autoregulation mechanisms. In the absence of might benefit from intra-arterial treatment are likely to have
changes to the autoregulatory system CBF changes with altera- angiography. Additionally, in a stroke patient, MRI, MRA, and
tions in PaCO2, functional requirements, and administration MRP may be used acutely, possibly diminishing the volume
of vasoactive substances. Under normal conditions CBF is not of iodinated contrast. This is, however, the patient population
changed by changes in systemic blood pressure. Low-flow most likely to have compromised renal function secondary to
states are of concern because of the risk of stroke. Nerve cell systemic processes such as diabetes and hypertension.
function deteriorates at CBF less than 20 mL/100 g/min. Cell
death occurs at CBF less than 5-10 mL/100 g/min.26
Indications
Patient Safety Routine Head Computed Tomography Scan
Radiation dose and contrast load are significant patient safety The indications for noncontrast head CT are well established
issues with CT-based imaging. CTA scans are similar to other and described, and the reader is referred to recent reviews or
CT studies in that the scanner acquires single slices at multiple textbooks that describe this it in detail. Here is provided a brief
levels. The usual issue for these patients is the number of synopsis of how head CT can aid the neurointensivist.
diagnostic cranial CT exams obtained per patient rather than A head CT is the initial emergent study for patients with
the dose delivered per study. acute neurologic injury, whether stroke, trauma, confusion, or
CTP studies require acquisition of 40 to 50 scans over 1 unknown neurologic event. It also is the primary study to
minute at a single anatomic level. The scanning techniques follow the neurologically compromised patient in an intensive
used for a perfusion CT are lower than those for a diagnostic care unit (ICU) setting. A typical example is the patient
head study, typically 80 to 100 kVp/200 mA for CTP com- with an acute intracranial hemorrhage. The acute and sub-
pared with 120 to 130 kVp/100 to 250 mA for a head CT. The acute management questions can involve resolution of the
lower kVp of CTP results in a lower radiation dose and hemorrhage with diminishing mass effect, increasing paren-
improved visualization of the iodinated contrast. Radiation chymal vasogenic edema in response to intraparenchymal
dose measures ionizing energy absorbed per unit of mass and blood products, increased volume of blood if delayed hemor-
is measured in gray (Gy) or milligray (mGy); 1 Gy = 1 joule rhage occurs, or hydrocephalus if blood within the arachnoid
per kilogram. Alternative units are sieverts (Sv) or millisieverts granulations causes diminished cerebrospinal fluid (CSF)
(mSv). For x-ray radiation in the ranges produced by CT scan- resorption. Other issues addressed by head CT include iden-
ners, 1 mSv = 1 mGy. When CTP is done at the basal ganglia tification of generalized brain swelling such as might occur in
level, avoiding the orbits, intracranial radiation dose is 180 to a patient with DAI, midbrain compression (unilateral from
363 mGy, surface dose is 327 to 713 mGy, and lens dose (from uncal herniation or bilateral from diffuse swelling), midline
scatter) is 11 m to 22 mGy.27 This is higher than the average shift, new or evolving brain infarction, postoperative issues, or
surface dose of 66 to 83 mGy at the tuber frontale by conven- new fluid collections. Identification of possible infection or
tional head CT scan using 130 to 135 kVp and 105 to 270 mA.28 vascular injury requires a contrast study.
If CTP is performed at multiple levels, the dose is increased
in proportion to number of acquisitions. The threshold dose
for cataract formation is 2 to 10 Gy. The overall effective dose Imaging Appearence
equivalent for CTP is 2 plus or minus 2.5 mSv. The dose from INTRACRANIAL HEMORRHAGE
a CTA of the brain is 2 to 4 mSv.27,29,30 The dose from other Acute intraparenchymal hemorrhage is denser than brain with
blood flow measurement techniques, such as positron emis- CT. The density reflects the concentration of the protein com-
sion tomography (PET) and single photon emission computed ponent of the hemoglobin molecule and diminishes with
Section IV—Radiology 261

A B C
Fig. 26.1  Computed tomography (CT) and computed tomography angiography (CTA) images to illustrate an intraparenchymal hemorrhage
and spot sign on the CTA. A, Axial noncontrast head CT. Hemorrhage is present in the left temporal lobe with extension into the occipital horn of
the lateral ventricle. B, CTA source image. A punctate focus of enhancement on the lateral margin of the hemorrhage is seen (spot sign); this suggests
ongoing hemorrhage (arrow). C, Delayed contrast enhanced CT. Irregular collection of contrast in the lateral margin of the clot is caused by ongoing
hemorrhage (arrow). This scan was obtained 1 to 2 minutes after the contrast injection.

breakdown of the hemoglobin molecule. An uncomplicated in the falx and tentorium, and may extend across suture lines.
clot within the brain becomes more lucent on its margins, An acute SDH tends to be higher density than brain. Chronic
continues to decrease in density over time (2-3 weeks de­ SDH is of lower density than brain because of hemoglobin
pending on the initial size of the clot), and ultimately breakdown similar to the process in intraparenchymal hemor-
resorbs, leaving an area of encephalomalacia (low density with rhage. Acute bleeding into a chronic SDH results in mixed
focal volume loss). When contrast is given, contrast extravasa- density and blood-fluid levels. More chronic SDH may not
tion is associated with subsequent intracranial hemorrhage conform as clearly to the curve of the skull.
growth.35 This is similar to the “spot sign,” a bright spot on
CTA source images that predicts hematoma growth (Fig. 26.1). ACUTE EPIDURAL HEMATOMA
SAH, unlike intraparenchymal hemorrhage, remains within An acute epidural hematoma (EDH) is hyperdense to brain,
the sulci and cisterns. It does not undergo the breakdown bows away from the curve of the skull (sometimes described
process seen with intraparenchymal clots. Density is depen- as lens shaped), and does not cross sutures but can cross the
dent on the concentration of blood in the subarachnoid space midline (crosses dural sinuses). An EDH often is associated
(i.e., the volume of the hemorrhage within the volume of with skull fractures, classically in the squamous temporal bone
CSF). Small hemorrhages may be poorly seen because of across the path of the middle meningeal artery. EDHs, which
mixing of the blood with CSF. Resolution of subarachnoid lie superficial to dura, are less likely than SDH to be associated
blood is dependent on clearing by CSF circulatory processes. with an adjacent brain contusion.
Attempts have been made to quantitate the volume of SAH in
relation to the likelihood of developing vasospasm.36,37 The INCREASED INTRACRANIAL PRESSURE
Fisher scale was an early method to correlate the volume and Increased intracranial pressure (ICP) causes a decrease in the
distribution of subarachnoid blood to the development of size of CSF spaces in the brain followed by increasing severity
vasospasm.38 Subsequent modifications or the development of of herniation. Diagnostic clues include a generalized decrease
newer grading scales have improved the predictive reliability. in the size of sulci and cisterns. In particular, compression or
In the Hijdra scale the volume of blood in each of 10 basal absence of the perimesencephalic cisterns is an indication that
cisterns and four ventricles is separately evaluated and summed ICP may be elevated. However, head CT findings do not always
for a total score.39 reliably predict intracranial physiology (e.g., ICP and brain
Acute blood within a more chronic fluid collection may be oxygen).40 If the increase in ICP is due to diffuse swelling, the
seen as a blood-fluid level with the denser blood (both in ventricles also may decrease in size. If it is due to hydrocepha-
terms of visual appearance and true fluid density) in a depen- lus, the ventricles may increase in size. When it is associated
dent part of the collection. This often is seen as blood in with local mass effect and herniation, there may be compres-
the occipital horns (blood layering in the dependent part of sion of the ventricles on the side of the lesion, displacement
the ventricular system in a supine patient), but also occurs of midline away from the lesion, and effacement of cisterns.
in chronic subdural hematoma (SDH) and in other cystic An example is subfalcine and uncal herniation in a patient
cavities. with a subacute, large, middle cerebral artery infarct with
marked cytotoxic edema.
ACUTE SUBDURAL HEMATOMA
Acute SDH is found within the subdural space, superficial to HERNIATION
the arachnoid. On imaging, acute SDH conforms to the cur- The appearance of herniation depends on the location of the
vature of the skull, is confined by dural boundaries (does not mass lesion or mass effect within the brain. Uncal herniation
cross the midline), may extend between the leaves of the dura and subfalcine herniation occur with mass lesions on one side
262 Section IV—Radiology

A B

C D
Fig. 26.2  Head computed tomography (CT) scan illustrating subfalcine and uncal herniation associated with an acute-on-chronic subdural
hematoma (SDH). A, Uncal herniation. Right sided holohemispheric mixed density SDH. There is right uncal herniation with flattening of the
suprasellar cistern and midbrain compression. B, Comparison: normal brain CT at the level of the midbrain. C, Subfalcine herniation. A more superior
slice in the same patient that shows compression of the right lateral ventricle and subfalcine herniation. D, Comparison: normal brain CT at the level
of the basal ganglia or lateral ventricles.

of the brain (Fig. 26.2). The uncus is displaced toward the Posterior fossa mass effect can be seen as compression and
opposite side of the brain from the intracranial mass, the displacement of the fourth ventricle, compression of the
suprasellar cistern is distorted, and the midbrain is displaced cerebellopontine-angle cisterns and cisterns anterior to the
away from the mass and compressed. Subfalcine herniation is brainstem, and displacement of the vermis upward with flat-
usually part of this constellation with compression of the ipsi- tening and distortion of the quadrigeminal plate cistern
lateral ventricle, compression of the third ventricle, and dis- (tectum) (i.e., “upward herniation”; Fig. 26.3). Also possible,
placement of the third ventricle and septum pellucidum across but more difficult to appreciate using CT, is downward dis-
midline away from the mass lesion. In general, when there is placement of the cerebellar tonsils into the foramen magnum.
more than 5 mm of midline shift, a surgical solution for the
increase in ICP often is necessary. Uncal herniation can be INFARCT
bilateral. This might occur if the patient had diffuse swelling. Ischemic injury to the brain on CT (Fig. 26.4) is seen as areas
In this case all supratentorial ventricles are compressed, the of low density with poor to absent identification of the differ-
suprasellar cistern and perimesencephalic cisterns are effaced, ence between gray matter and white matter (loss of gray-white
and the midbrain is flattened. differentiation). Examples are the insular ribbon sign with
Section IV—Radiology 263

A B

C D
Fig. 26.3  Head computed tomography (CT) scan that illustrates progressive upward herniation. A, Admission head CT scan that demonstrates
and early infarct (low density) in the left cerebellum. The fourth ventricle is midline. B, More superior slice from the admission head CT scan of the
same patient. The quadrigeminal plate cistern (arrow) and suprasellar cistern (arrowhead) are open. C, Day 1 head CT scan. There is partial effacement
of the quadrigeminal plate cistern (arrow). The suprasellar cistern and temporal horns are normal (arrowheads). D, Head CT scan obtained on day 2.
The quadrigeminal plate cistern is effaced (arrow) and the temporal horns are dilated (arrowheads). There is increased mass effect in the posterior
fossa caused by swelling of the subacute cerebellar infarct.

absence of a visible insula on the CT scan, generalized loss of contrast within a venous sinus on a contrast-enhanced CT
gray-white differentiation in an arterial vascular territory, or (delta sign), or absence of a sinus on a magnetic resonance
loss of gray-white differentiation in the hemispheres in a venogram (Fig. 26.5). Venous sinus thrombosis is more reli-
patient with diffuse anoxic injury. Acutely there may be ably diagnosed than is the thrombosis of a superficial vein
minimal mass effect. With time (12-48 hours) there can be because of the variation in the distribution of superficial
marked swelling of the areas of cytotoxic edema with possible cerebral veins.
herniation. Occasionally hemorrhagic transformation (i.e.,
the development of hemorrhage within an acute or subacute
infarct) may occur. Computed Tomography Angiography
The appearance of a venous infarct differs from that of an Indications for CTA include diagnosis and evaluation of intra-
arterial infarct in the distribution of the low-density region cranial aneurysm, either ruptured or unruptured, diagnosis of
and the propensity toward hemorrhage. The areas of low cerebral vasospasm, diagnosis of intracranial vascular malfor-
density do not correspond to conventional arterial distribu- mations, evaluation of extracranial and intracranial vascular
tions but may involve deep structures, the temporal lobe, structures in a patient with ischemic infarct or chronic cere-
and paramidline structures. The diagnosis depends on brovascular disease, evaluation of underlying pathology in a
appreciating high density in a venous sinus, absence of patient with acute intracranial hemorrhage, diagnosis of
264 Section IV—Radiology

A B

C D

Fig. 26.4  Computed tomography (CT) imaging showing evolution of acute to subacute infarct. A, Noncontrast head CT scan obtained 2
hours after symptom onset. B, Noncontrast head CT scan obtained 9 hours later demonstrates loss of gray-white differentiation in the left hemisphere.
C, Noncontrast head CT scan obtained 3 days after symptom onset illustrating completed left middle cerebral artery infarct. D, Coronal image of
computed tomography angiography shows occlusion of left middle cerebral artery.

vascular injury following blunt or penetrating trauma, and puncture may still be required to exclude the uncommon
diagnosis of cerebral venous thrombosis (CVT). but possible false-negative CT. Some suggest when there is
strong clinical suspicion of SAH that the combination of CT
and CTA may offer a less invasive diagnostic paradigm than
Computed Tomography Perfusion lumbar puncture.43 The quality of CTA to detect aneurysms is
CTP is a developing technology, and its use is still to be fully almost comparable to digital subtraction angiography (DSA).
elucidated. Indications include diagnosis of acute infarction, However, DSA remains superior to CTA to detect small aneu-
evaluation of patients with acute infarction for possible inter- rysms (<5 mm), especially with 3-D rotational angiography.
vention, diagnosis of vasospasm, assessment of autoregulation Three-dimensional rotational angiography also allows for
in TBI patients, evaluation of blood-brain barrier permeabil- more precise evaluation and measurement of aneurysms.13,44
ity in acute infarction to assess for the risk of hemorrhagic CTA can assist in the rapid diagnosis or exclusion of aneu-
transformation, measurement of cerebral vascular reserve in rysms and arteriovenous malformations in the acutely ill
patients with chronic vascular occlusions, and evaluation of patient, such as the patient with a large intracerebral hema-
tumor vascularity.41,42 toma who is rapidly deteriorating. When used as a first screen-
ing test for intracranial hemorrhage, CTA is often as accurate
as DSA for more proximal lesions but has a lower sensitivity
Subarachnoid Hemorrhage for smaller more distal lesions and dural arterio-venous
The sensitivity of noncontrast-enhanced CT for SAH within fistulas.45
12 hours of ictus is 98% to 100%. If there is a high suspicion CT angiography can be used in the initial work-up of
for SAH or if the patient has a subacute hemorrhage, lumbar patients with a suspicion of an intracranial aneurysm
Section IV—Radiology 265

A B

Fig. 26.5  Computed tomography (CT) and magnetic resonance imaging (MRI) for comparison illustrating venous sinus thrombosis.
A, Contrast-enhanced CT. A thrombosis of the right sigmoid sinus is illustrated with a delta sign (nonopacification of the sinus) (arrow). There is
contrast enhancement of the left sigmoid sinus (arrowhead). B, T1-weighted noncontrast axial MRI. Subacute clot seen in the right sigmoid sinus is
high signal (arrow). There is a flow void in the left sigmoid sinus (arrowhead). The high signal material in the mastoid resection cavity (anterior to the
sinus thrombosis) is surgically implanted fat.

(Fig. 26.6). Romijn et al. compared CTA with bone elimina- (oxygenation, infection, metabolic abnormalities). Diagnosis
tion to DSA and 3-D rotational angiography in 108 patients of cerebral vasospasm can be made with many different
with clinically suspected SAH. Specificity, sensitivity, positive imaging modalities, TCD, CTA, SPECT, CTP, and catheter
predictive value, and negative predictive value of CTA with angiography. Catheter angiography, the diagnostic gold stan-
bone removal was 0.99, 0.90, 0.98, and 0.95, respectively. Sen- dard, is invasive and has a stroke risk, although in the SAH
sitivity was 0.99 for aneurysms greater than or equal to 3 mm population this is very rare.61,62 SPECT correlates with angio-
and 0.38 for aneurysms less than 3 mm.44 Other authors have graphically demonstrated spasm 65% of the time.63,64 TCD
found similar results without bone removal.13,46-48 When CTA may not demonstrate distal spasm but is a very useful tool to
is negative the false-negative rate is reported by some to be identify spasm in proximal or basal vessels (see Chapter 30).
zero after DSA.49 In particular, negative CTA findings are reli- CTA is complementary to TCD, which is performed bedside.
able to exclude an aneurysm with a perimesencephalic distri- In various studies CTA has 75% to 90% accuracy to detect
bution of blood. When the hemorrhage is diffuse or more cerebral vasospasm when compared with catheter angiogra-
peripheral, DSA should be considered when CTA is negative.50 phy. The diagnosis of vasospasm is concordant between CTA
However, the sensitivity of on-call resident interpretation of a and catheter angiography for no spasm (96%) and severe
CTA after SAH is lower than an attending,51 and among spasm (100%) and usually adequate when the patient is symp-
patients who undergo subsequent digital subtraction angiog- tomatic.65 However, the accuracy using CTA for mild and
raphy there is a treatment plan change in about 20%.52 CTA moderate spasm is 57% and 64%. Concordance between CTA
may be used to decide whether to occlude the aneurysm using and catheter angiography is greater for proximal than for
endovascular or surgical techniques, and in some cases, guide distal vessels.65-67 Catheter angiography is necessary if intra-
operative management. The need for catheter angiography arterial treatment of vasospasm is contemplated. It is helpful,
before decisions of management (coil vs. clip) and to deter- if using CTA to diagnose vasospasm, to have a baseline study
mine surgical approach are a balance between the immediate for comparison of vessel size. CTA measures of vascular diam-
surgical concerns, the requirement for rapid intervention, eter slightly overestimate the size of larger vessels and under-
and the experience of the involved physicians.50,53,54 Discrepan- estimate the size of the smaller distal branches.68 Other authors
cies between CTA and DSA frequently are associated with suggest the correlation between CTA and DSA measured
location, for example, cavernous vs. supraclinoid, or differen- diameters is between 0.45 and 0.76 and that underestimation
tiation of a vascular loop from a small aneurysm. Use of CTA of diameter of large and overestimation of diameter of small
to evaluate aneurysms after occlusion (clip or coil) is less clear. arteries occurs with CTA. The major limitation of CTA,
The artifact from the metal coil or clip can prevent accurate however, is the inability to measure some vessels because of
visualization of possible residual aneurysm.55,56 Despite this, artifact.68 In addition, CTA cannot demonstrate changes in
there are reports of CTA use to evaluate treated aneurysms flow dynamics that are visible with catheter angiography.
with 87% to 97% sensitivity and specificity, although intrao- CTA shows the anatomy of the vascular tree. Perfusion
bserver concordance was less robust.57 methods (i.e., CTP) show areas of abnormal CBF. Timely
Symptomatic vasospasm occurs in 17% to 40% of SAH evaluation of low perfusion status is important because angio-
patients.37,59 Diagnosis and management of cerebral vaso- plasty of vasospasm can be performed to prevent secondary
spasm can be facilitated using CTA.59,60 Neurologic decline in infarction when low-flow states are identified. SPECT, TCD,
a patient with aneurysmal SAH has many potential causes, PET, Xe-CT, and CTP all can show reduced CBF associated
both intracranial (cerebral vasospasm and ischemia, hydro- with vasospasm. SPECT also has been used to evaluate brain
cephalus, increasing edema, new hemorrhage) and systemic perfusion. For SPECT, patients are required to lie still longer
266 Section IV—Radiology

A B

C D E

F G
Fig. 26.6  Basilar bifurcation aneurysm imaged and displayed using seven different methods. A, Noncontrast head computed tomography
(CT). Diffuse subarachnoid hemorrhage. Lower density focus in the suprasellar cistern is the basilar aneurysm (arrow). B, Computed tomography
angiography (CTA)—coronal maximum intensity projection (MIP). The basilar bifurcation aneurysm projects superior to the posterior cerebral arteries. 
C, CTA—sagittal MIP. The basilar bifurcation aneurysm projects superior to the dorsum sella. D, CTA—three-dimensional (3-D) reconstruction.
E, Catheter angiogram. The left vertebral artery injection (AP projection) shows the basilar aneurysm. F, 3-D MIP reconstruction of the rotational
angiographic acquisition-catheter angiogram. The left vertebral artery was injected. The small human figure in the lower right shows the patient
orientation. G, 3-D MIP reconstruction of the rotational angiographic acquisition-catheter angiogram. The hollow-vessel reconstruction can help guide
intra-arterial (endovascular) treatment.

than for CTP, and it can only be repeated after 24 hours. MRP CTP studies in patients with intracranial vasospasm have
is not commonly used to evaluate cerebral vasospasm in the examined CBF, CBV, and MTT. In broad terms MTT is very
critically ill patient following SAH because of difficulty moni- sensitive to changes in cerebral perfusion but is not specific
toring the patient’s physiologic status in the vicinity of a high for infarction. CBV is specific for areas of brain with irrevers-
field magnet, the distortion caused by metal within the head ible ischemic damage. CBF and CBV are significantly lower
(clip, coil), and the qualitative nature of the study. and MTT longer in patients with moderate to severe
Section IV—Radiology 267

vasospasm. CBF and CBV show biphasic decrease at 1 to 3 ischemic stroke88,89 or increase the risk of contrast-induced
days and at 10 to 17 days after ictus, more apparent in the nephropathy90 and is cost effective91 (i.e., multimodal CT
more vascular gray matter than in white matter. In one study, imaging [CT, CTA, and CTP] can play a significant role in
MTT showed early areas of diminished perfusion, but could acute stroke neuroimaging).42,92,93 In addition, combined CT
not be used to separate mild, moderate, and severe vaso- and CTP studies may be used to select some patients
spasm.29 CBF reduction is closely related to clinical outcome. for endovascular reperfusion94 or intravenous thrombolysis95
Sviri et al. found that CBF reduction and prolongation of rather than selection solely on time-based criteria, or if the
MTT were closely correlated with delayed ischemic deficit, time of stroke onset is not known combined CT and CTP can
severity of vasospasm seen by catheter angiography, vaso- be used to help predict the likelihood of hemorrhagic trans-
spasm seen on TCD, perfusion estimated on SPECT, and clini- formation or development of malignant edema.96
cal course.69,70 In the presence of delayed cerebral ischemia CTA for acute ischemic infarcts should include the extra-
(DCI), threshold values for CTP-determined CBF and MTT cranial as well as intracranial vessels, typically the aortic
are considered to be 36.5 mL/100 g/min (95% sensitivity, 70% arch through the top of the head. CTA may be more spe-
specificity) and 5.4 seconds (78% sensitivity, 70% specificity), cific for identification of small, slow flow but not occluded
respectively. However, absolute threshold values may not be vessels than is MRA. Neither is completely specific. However,
generalizable because of scanner equipment and postprocess- if acute occlusion is suspected, either study can be used to
ing differences.71 Others have confirmed the prolongation of guide therapy, including of suspected basilar artery throm-
MTT and its correlation to CBF and vasospasm, and some bosis.97 CTA also can be used as an indicator of severity
suggest that in DCI qualitative assessment of CTP may be a of ischemic injury. Smith et  al. in 72 patients with acute
better imaging modality than head CT or CTA for quick stroke found that baseline National Institutes of Health
decision making.72-76 Because of some questions about the (NIH) stroke score and intracranial large vessel occlusion
reliability of absolute flow values in a CTP calculation, Van de predicted poor final neurologic outcome at discharge.
Schaaf used side-to-side ratios to evaluate territorial abnor- Similar outcome is found with posterior circulation occlu-
malities in perfusion. CBF ratio of 0.72 for abnormal to sions86,98 and when large vessel occlusion is seen on CTA
normal appeared to best correlate with delayed ischemic after transient ischemic attack (TIA), a decline in neurologic
changes.77 Noncontrast CT should be done as necessary to function is more likely.99
exclude other causes of clinical deterioration including recur- Early imaging in the patient with an acute ischemic infarct
rent hemorrhage, hydrocephalus, and edema. Relative perfu- is directed to identify tissue at risk of infarction compared with
sion changes on admission CTP studies, that is, before DCI tissue with irreversible injury (penumbra vs. core). The under-
develops (CBF, MTT, and time to peak) may help identify lying assumption is that acute intervention may be more effec-
patients who will go on to delayed ischemia following SAH.78- tive if there is tissue at risk but does not have irreversible injury.
80
The absolute flow values at admission may not always be Perfusion studies can be used to make this distinction.100,101
helpful. Good correlation is seen between MRP and CTP for For CTP the parameters used to define penumbra versus isch-
MTT.3 Finally, changes in CTP, particularly in poor-grade emic core remain under discussion. In most series the penum-
patients, may be used as a surrogate marker to monitor the bra is considered to have reduced CBF, normal or elevated
success of treatment strategies for vasospasm81 or guide man- CBV due to cerebral autoregulatory response, and elevated
agement decisions in the comatose SAH patient.82 MTT (Fig. 26.7). Tan et al. evaluated 113 patients, 55 of whom
went on to experience infarction. CBV was the most accurate
predictor of final infarct volume (97% specificity), whereas
Acute Cerebral Infarction increased MTT was predictive of tissue at risk for infarction
Acute stroke imaging starts with CT. The first consideration is in the presence of vascular occlusion.90 Others have found
differentiation of ischemic infarct from intracranial hemor- similar results. The irreversible core will have decreased CBF
rhage. If ischemic infarct is present and the patient is within and CBV with elevated MTT.15,16,102 Volume of the infarct core
the time window for emergent treatment (3 hours for intra- on CTP correlates well with volume of the infarct on diffusion-
venous tissue plasminogen activator [tPA] and 6 to 8 hours weighted MRI. However, CBV is not always low in patients
for intra-arterial methods in the anterior circulation) the size with acute infarction103 and some suggest CTP-measured CBF
of the infarct, the anatomic location, and the presence of corresponds with the acute diffusion-weighted imaging lesion
occluded vessels in the neck or brain should be defined.83,84 If better than CBV.104
there is a hemorrhage, the location and source of the bleed is There have been attempts to quantify the measures of CBF,
an immediate question as is midline shift and possible hernia- CBV, and MTT in patients with acute infarction. For preserva-
tion. CT is the primary imaging modality to diagnose acute tion of cerebral function CBF needs to be greater than 18 to
intracranial hemorrhage. CT is less sensitive than MRI for 19 mL/100 g/min; minimum CBF to preserve tissue viability
diagnosis of acute ischemic infarct, particularly in the poste- is 10 to 15 mL/100 g/min.105,106 Viability of tissue within the
rior fossa and brainstem. Despite this, protocols around the penumbra with diminished blood flow is time dependent,
use and timing of MRI in a patient with an acute infarct seen although clear time thresholds have not been established. CTP
within the treatment window vary among centers. Protocols has been used as a predictor of thrombolytic therapy and
may include immediate CT only, immediate CT, CTA plus or clinical outcome.102,107-109 Wintermark et al. reported using a
minus CTP, CT followed by MRI, MRA, and MRP or any 34% decrease in CBF relative to clinically normal areas as a
number of permutations of this. CTA, CTP, MRA, and MRP threshold for ischemia and infarction, and a CBV threshold of
provide similar information in the acute period.85-87 The com- 2.5 mL/100 g to differentiate infarction from penumbra.
bination of CT, CTA, and CTP does not appear to adversely There was good correlation of the results of CT perfusion and
increase the time to thrombolysis in patients with acute final infarcted area after recanalization, which reflected
268 Section IV—Radiology

A B

C D E
Fig. 26.7  Computed tomography perfusion study used to demonstrate the ischemic penumbra. The images, all obtained at the level of the
centrum semiovale, show mismatch of cerebral blood flow (CBF) or mean transit time to cerebral blood volume. A, CBF map. Large area of diminished
CBF in the distribution of the middle cerebral artery (blue is low flow). B, Mean transit time (MTT) map. Large area of prolonged MTT in the distribution
of the middle cerebral artery (blue is slow flow). C, Cerebral blood volume map. Cortical blood volume is normal to minimally increased (blue is
low blood volume). D, Noncontrast CT. Mild diffuse sulcal effacement is illustrated in the right hemisphere with a focal area of low density (arrow).
E, magnetic resonance imaging (MRI) diffusion-weighted image. Punctate areas of restricted diffusion in a watershed distribution on the right.

recovery of the penumbra after arterial recanalization.109 primary diagnostic imaging study. DAI is the one process that
Indeed reperfusion (with an MTT reperfusion index >75%) can be difficult to diagnose with CT, because hemorrhage may
may be a more accurate predictor of infarct volume than not be present and the small foci of abnormal low density may
recanalization.110 However, generalized clinical application of be difficult to see with CT. The progression of these processes
numerical values requires further study, because the values in the acute phase and the secondary complications (e.g.,
may vary with different perfusion acquisition techniques and swelling, shift, hydrocephalus, delayed hemorrhage, infarct)
pretreatment CTP prognostic maps may not always be associ- provide the indications for follow-up CT scans. Routine CT
ated with recovery or not follow early reperfusion.111 Whether also can be used to grade the head injury through the Mar-
threshold perfusion values might be used in other disorders, shall112 or Rotterdam scales.113 There is good interobserver
including TBI, remains unclear. In addition, threshold values reproducibility between neuroradiologists and neurosurgeons
may not be generalizable because of differences in equipment when calculating these scales,114 both of which can be prog-
and postprocessing methods. nostically important.115,116

Traumatic Brain Injury Computed Tomography Angiography


CTA in trauma is of most use to screen for or identify vascular
Routine Computed Tomography injury in blunt or penetrating trauma4,117-119 although DSA
Traumatic intracranial injury includes contusion, subarach- may still be needed in some patients.120 Incidence of injury to
noid or intraventricular hemorrhage, hematoma (intraparen- the carotid or vertebral arteries following blunt trauma is
chymal/SDH/EDH), and DAI. This can occur from a variety approximately 30% in a subset of patients who have specific
of mechanisms, have secondary injury from local cellular findings, including cervical spine fracture or dislocation, frac-
process or from associated vascular injury with infarct, and be ture through the foramen transversarium, upper cervical
seen in combination. Because most cranial injury is associated spine injury with a high-force mechanism, LeFort II and
with hemorrhage, mass lesions, or shift, CT has become the LeFort III facial injuries, bilateral mandibular condyle
Section IV—Radiology 269

fractures, fractures through the carotid canal in the skull base, study where brain death was defined as absence of intracranial
expanding hematoma in the neck, and unexplained acute flow on catheter angiography.136
infarct. This compares an incidence of less than 1% in the
larger population of patients with blunt injury.121-125 Penetrat-
ing injury also places vascular structures at risk and requires
Portable Computed Tomography
either exploration if in zone 2 or imaging if in zone 1 or zone Mobile CT scanners for cranial imaging have evolved since
3. Although some discussion remains, in the majority of these the late 1990s. This point of care technology has the advantage
patients CTA is the initial study of choice because of rapidity of bringing the scanner to the ICU patient. These are the
of acquisition, low risk, and accuracy.126-130 As in the other patients who are most difficult to transport because of ventila-
applications of CTA, the development of x-ray tubes that have tory requirements, extensive physiologic monitoring, and
sufficient heat capacity to image larger areas of the body (i.e., uncertainty of physiologic changes that may occur during
arch to vertex), thin isovoxel slices, and rapid, flexible refor- transport.137 The best images from a mobile scanner are
mations has expedited the conversation from catheter angiog- similar in detail to the average image from fixed CT scanners,
raphy to CTA. and the radiation dose is comparable.1,138-140 If needed, extra
shielding can be added when the device is used in an open
ward environment to restrict radiation exposure to others.141
Computed Tomography Perfusion The scanners have less flexibility in angulation of the image
Use of cerebral perfusion techniques to modify treatment of with respect to the patient’s head. There also are potential
TBI patients, especially closed head injury, is less uniformly limitations in anatomy that can be imaged. For example, the
embraced than is CTA to identify vascular injuries or CTP to length of the patient’s neck determines the most caudal slice
evaluate an acute ischemic infarct. Xe-CT and CTP have been on one scanner available in 2011. This may limit evaluation
used to estimate CBF in relation to cerebral perfusion pressure of the patient’s neck or cervical spine. Portable CT scanners
(CPP), evaluate the status of autoregulatory functions, and are slower than conventional fixed CT. However, the major
possibly predict the extent of intracranial injury. Conceptu- advantage is that patients do not have to be removed from
ally, elevation of mean arterial pressure does not change CPP their beds. This means patients can be continually monitored
if autoregulation is intact. In contrast, if autoregulation is and ventilated during the scan without having first to be
absent, elevation of mean arterial pressure will increase CBF disconnected.142
and cerebral perfusion. The aim of the treatment is to main-
tain CPP to minimize secondary intracranial injury.
In general, target values should be ICP less than 20 mm Hg
Conclusion
and CPP of 50 to 70 mm Hg. CPP values less than 50 mm Hg Noncontrast CT is an important and common tool to evaluate
carry a two- to three-times higher risk of cerebral ischemia patients with acute neurologic disorders and those in the
and cerebral hypoperfusion, whereas CPP values greater than NCCU. CTA and CTP both provide noninvasive imaging.
70 mm Hg may be associated with an increased incidence of CTA is more firmly established as a method to evaluate the
systemic complications such as cardiorespiratory failure. CPP- intracranial vasculature. CTP is a developing technology and
directed therapy versus ICP-directed therapy depends on the requires further evaluation to fully define its use and accuracy.
state of the individual’s cerebral vascular autoregulation. If Both can provide useful information that aids care of patients
autoregulation is intact, a CPP-directed therapy could be used in the NCCU. In an era of increasing concern with radiation
with a higher probability of favorable outcome, whereas if dosage, the use of either has to be weighed against the biologic
autoregulation is absent ICP-guided therapy may yield better risks, particularly when multiple follow-up CT scans are per-
results. formed. The advent of portable CT scans has allowed CT
Cerebral contusion can enlarge and cause increased ICP imaging to be performed in NCCU.
and neurologic deterioration during a patient’s ICU course.
Early identification of the patient at risk for contusion enlarge- References
ment could prevent further tissue damage. CTP may allow
1. Peace K, Maloney E, Frangos S, et al. The use of a portable head CT scanner
better evaluation of tissue viability than noncontrast CT. in the ICU. J Neurosci Nurs 2010;42(2):109–16.
Compared with noncontrast routine CT, the areas with 2. Manolakai D, Velmahos GC, Spaniolas K, et al. Early magnetic resonance
decreased CBF, CBV, and prolonged MTT correlate with imaging is unnecessary in patients with traumatic brain injury. J Trauma
development of brain damage seen on later follow-up CTs.131 2009;66(4):1008–12.
Wintermark et al. also reported a good correlation between 3. Eastwood JD, Lev MH, Wintermark M, et al. Correlation of early dynamic
CT perfusion imaging with whole-brain MR diffusion and perfusion
the extent of CTP abnormalities with CPP. Preservation imaging in acute hemispheric stroke. Am J Neuroradiol 2003;24(9):1869–75.
of autoregulation may be a good predictor of the patients’ 4. Heuer G, Hurst R, Le Roux P. Traumatic carotid artery injury. In: Winn HR,
outcome.132,133 editor. Youmans neurological surgery. 6th ed. Philadelphia: Elsevier; 2011.
5. Gur D, Yonas H, Jackson DL, et al. Simultaneous measurements of cerebral
blood flow by the xenon/CT method and the microsphere method: a
Brain Death comparison. Invest Radiol 1985;20(7):672–7.
6. Yonas H, Pindzola RP, Johnson DW. Xenon/computed tomography cerebral
CTA can be used to aid in the diagnosis of brain death, blood flow and its use in clinical management. Neurosurg Clin N Am
although it may not always be reliable.134 In one study of 21 1996;7(4):605–16.
patients, the sensitivity of CTA compared with electroenceph- 7. Coles JP. Imaging of cerebral blood flow and metabolism. Curr Opin
Anesthesiol 2006;19:473–80.
alography was 52.4%. Brain death on CTA was defined as an 8. Horn P, Vajkoczy P, Thome C, et al. Xenon-induced flow activation in
absence of intracranial vascular opacification.135 Divergence patients with cerebral insult who undergo xenon-enhanced CT blood flow
between catheter angiography and CTA was 30.2% in another studies. Am J Neuroradiol 2001;22:1543–9.
270 Section IV—Radiology

9. Abrahams J, Saha P, Hurst R, et al. Three dimensional bone-free rendering of 30. Lee EJ, Lee SK, Agid R, et al. Comparison of image quality and radiation
the cerebral circulation by use of computed tomography and fuzzy dose between fixed tube current and combined automatic tube current
connectedness. Neurosurgery 2002;51:264–9. modulation in craniocervical CT angiography. Am J Neuroradiol 2009;30:
10. Ramasundara S, Mitchell PJ, Dowling RJ. Bone subtraction CT angiography 1754–9.
for the detection of intracranial aneurysms. J Med Imaging Radiat Oncol 31. Huda W, Sandison GA. The use of the effective dose equivalents, HE, for
2010;54(6):526–33. doi:10.1111/j.1754-9485.2010.02211.x. 99mTc labelled radiopharmaceuticals. Eur J Nucl Med 1989;15(4):174–9.
11. Anderson GB, Steinke DE, Petruk KC, et al. Computed tomographic 32. Huda W, Sandison GA. Estimates of the effective dose equivalent, HE, in
angiography versus digital subtraction angiography for the diagnosis and positron emission tomography studies. Eur J Nucl Med 1990;17(3-4):116–20.
early treatment of ruptured intracranial aneurysms. Neurosurgery 33. Langner S, Stumpe S, Kirsch M, et al. No increased risk for contrast-induced
1999;45(6):1315–20. nephropathy after multiple CT perfusion studies of the brain with a
12. van der Schaaf IC, Velthuis BK, Wermer MJH, et al. New detected aneurysms nonionic, dimeric, iso-osmolal contrast medium. Am J Neuroradiol
on follow-up screening in patients with previously clipped intracranial 2008;29(8):1525–9.
aneurysms comparison with DSA or CTA at the time of SAH. Stroke 34. Newhouse JH, Kho D, Qasim A, et al. Frequency of serum creatinine changes
2005;36:1753–8. in the absence of iodinated contrast material: implications for studies of
13. Dammert S, Krings T, Moller-Hartmann W, et al. Detection of intracranial contrast nephrotoxicity. Am J Radiol 2008;191:376–82.
aneurysms with multislice CT: comparison with conventional angiography. 35. Hallevi H, Abraham AT, Barreto AD, et al. The spot sign in intracerebral
Neuroradiology 2004;46(6):427–34. hemorrhage: the importance of looking for contrast extravasation. Stroke
14. Agid R, Lee SK, Willinsky RA, et al. Acute subarachnoid hemorrhage: using 2010;41(1):e34–40. Epub 2009, Nov 12.
64-slice multidetector CT angiography to “triage” patients’ treatment. 36. Rosen DS, Macdonald RL. Subarachnoid hemorrhage grading scales: a
Neuroradiology 2006;48(11):787–94. systematic review. Neurocrit Care 2005;2:110–18.
15. Nabavi DG, Cenic A, Craen RA, et al. Quantitative assessment of cerebral 37. Frontera JA, Claassen J, Schmidt JM, et al. Prediction of symptomatic
hemodynamics using computed tomography: stability, accuracy, and vasospasm after subarachnoid hemorrhage: the modified fisher scale.
precision studies in dogs. J Comput Assist Tomogr 1999;23:506–15. Neurosurgery 2006;59(1):21–7.
16. Wintermark M, Maeder P, Thiran JP, et al. Quantitative assessment of 38. Fisher CM, Kistler JP, Davis JM. Relation of cerebral vasospasm to
regional cerebral blood flow by perfusion CT studies at low injection rates: subarachnoid hemorrhage visualized by computerized tomographic
a critical review of the underlying theoretical models. Eur Radiol 2001;11: scanning. Neurosurgery 1980;6:1–9.
1220–30. 39. Hijdra A, Brouwers PJ, Vermeulen M, et al. Grading the amount of blood on
17. Abels B, Klotz E, Tomandl BF, et al. Perfusion CT in acute ischemic stroke: computed tomograms after subarachnoid hemorrhage. Stroke 1990;21:
a qualitative and quantitative comparison of deconvolution and maximum 1156–61.
slope approach. Am J Neuroradiol 2010;31:1690–8. 40. Katsnelson M, Mackenzie L, Frangos S, et al. Are initial radiographic and
18. Kudo K, Sasaki M, Yamada K, et al. Differences in CT perfusion maps clinical scales associated with subsequent intracranial pressure and brain
generated by different commercial software: quantitative analysis by using oxygen levels after severe traumatic brain injury? Neurosurgery 2011;Nov 9.
identical source data of acute stroke patients. Radiology 2010;254:200–9. Epub ahead of print.
19. Sase S, Honda M, Machida K, et al. Comparison of cerebral blood flow 41. Hoeffner EG, Case I, Jain R, et al. Cerebral perfusion CT: technique and
between perfusion computed tomography and xenon-enhanced computed clinical applications. Radiology 2004;231:632–44.
tomography for normal subjects: territorial analysis. J Comput Assist 42. Leiva-Salinas C, Wintermark M, Kidwell CS. Neuroimaging of cerebral
Tomogr 2005;29(2):270–7. ischemia and infarction. Neurotherapeutics 2011;8(1):19–27.
20. Sanelli PC, Lev MH, Eastwood JD, et al. The effect of varying user-selected 43. McCormack RF, Hutson A. Can computed tomography angiography of the
input parameters on quantitative values in CT perfusion maps. Acad Radiol brain replace lumbar puncture in the evaluation of acute-onset headache
2004;11(10):1085–92. after a negative noncontrast cranial computed tomography scan? Acad
21. Wintermark M, Lau BC, Chien J, et al. The anterior cerebral artery is an Emerg Med 2010;17(4):444–51.
appropriate arterial input function for perfusion-CT processing in patients 44. Romijn M, Gratama van Andel HA, van Walderveen MA, et al. Diagnostic
with acute stroke. Neuroradiology 2008;50:227–36. accuracy of CT angiography with matched mask bone elimination for
22. Waaijera A, van der Schaaf IC, Velthuisa BK, et al. Reproducibility of detection of intracranial aneurysms: comparison with digital subtraction
quantitative CT brain perfusion measurements in patients with symptomatic angiography and 3D rotational angiography. Am J Neuroradiol 2008;29(1):
unilateral carotid artery stenosis. Am J Neuroradiol 2007;28:927–32. 134–9.
23. Fiorella D, Heiserman J, Prenger E, et al. Assessment of the reproducibility of 45. Yeung R, Ahmad T, Aviv RI, et al. Comparison of CTA to DSA in
postprocessing dynamic CT perfusion data. Am J Neuroradiol 2004;25(1): determining the etiology of spontaneous ICH. Can J Neurol Sci 2009;36(2):
97–107. 176–80.
24. Soares BP, Dankbaar JW, Bredno J, et al. Automated versus manual post- 46. Nijjar S, Patel B, McGinn G, et al. Computed tomographic angiography as
processing of perfusion-CT data in patients with acute cerebral ischemia: the primary diagnostic study in spontaneous subarachnoid hemorrhage.
influence on interobserver variability. Neuroradiology 2009;51(7):445–51. J Neuroimaging 2007;17(4):295–9.
25. Imielinska C, Liu X, Rosiene J, et al. Toward objective quantification of 47. Westerlaan HE, Gravendeel J, Fiore D, et al. Multislice CT angiography in
perfusion-weighted computed tomography in subarachnoid hemorrhage: the selection of patients with ruptured intracranial aneurysms suitable for
quantification of symmetry and automated delineation of vascular clipping or coiling. Neuroradiology 2007;49(12):997–1007.
territories. Acad Radiol 2005;12(7):874–87. 48. Mohan S, Lee W, Tan JT, et al. Multi-detector computer tomography
26. Ito H, Kanno I, Kato C, et al. Database of normal human cerebral blood angiography in the initial assessment of patients acutely suspected of having
flow, cerebral blood volume, cerebral oxygen extraction fraction and cerebral intracranial aneurysm rupture. Ann Acad Med (Singaore) 2009;38(9):769–73.
metabolic rate of oxygen measured by positron emission tomography 49. Prestigiacomo CJ, Sabit A, He W, et al. Three dimensional CT angiography
with 15O-labelled carbon dioxide or water, carbon monoxide and oxygen: versus digital subtraction angiography in the detection of intracranial
a multicentre study in Japan. Eur J Nucl Med Mol Imaging 2004;31(5): aneurysms in subarachnoid hemorrhage. J Neurointerv Surg 2010;2(4):
635–43. 385–9. Epub 2010, Jun 24.
27. Hirata M, Sugawara Y, Fukutomi Y, et al. Measurement of radiation dose in 50. Agid R, Andersson T, Almqvist H, et al. Negative CT angiography findings
cerebral CT perfusion study. Radiat Med 2005;23(2):97–103. in patients with spontaneous subarachnoid hemorrhage: when is digital
28. Cohnen M, Fischer H, Hamacher J, et al. CT of the head by use of reduced subtraction angiography still needed? AJNR Am J Neuroradiol 2010;31(4):
current and kilovoltage: relationship between image quality and dose 696–705. Epub 2009, Nov 26.
reduction. Am J Neuroradiol 2000;21(9):1654–60.
29. Nabavi DG, LeBlanc LM, Baxter B, et al. Monitoring cerebral perfusion after A complete list of references for this chapter can be found online at
subarachnoid hemorrhage using CT. Neuroradiology 2001;43:7–16. www.expertconsult.com.
Section IV—Radiology 270.e1

References 24. Soares BP, Dankbaar JW, Bredno J, et al. Automated versus manual
post-processing of perfusion-CT data in patients with acute cerebral
1. Peace K, Maloney E, Frangos S, et al. The use of a portable head CT ischemia: influence on interobserver variability. Neuroradiology 2009;51(7):
scanner in the ICU. J Neurosci Nurs 2010;42(2):109–16. 445–51.
2. Manolakai D, Velmahos GC, Spaniolas K, et al. Early magnetic resonance 25. Imielinska C, Liu X, Rosiene J, et al. Toward objective quantification of
imaging is unnecessary in patients with traumatic brain injury. J Trauma perfusion-weighted computed tomography in subarachnoid hemorrhage:
2009;66(4):1008–12. quantification of symmetry and automated delineation of vascular
3. Eastwood JD, Lev MH, Wintermark M, et al. Correlation of early dynamic territories. Acad Radiol 2005;12(7):874–87.
CT perfusion imaging with whole-brain MR diffusion and perfusion 26. Ito H, Kanno I, Kato C, et al. Database of normal human cerebral blood
imaging in acute hemispheric stroke. Am J Neuroradiol 2003;24(9): flow, cerebral blood volume, cerebral oxygen extraction fraction and
1869–75. cerebral metabolic rate of oxygen measured by positron emission
4. Heuer G, Hurst R, Le Roux P. Traumatic carotid artery injury. In: Winn tomography with 15O-labelled carbon dioxide or water, carbon monoxide
HR, editor. Youmans neurological surgery. 6th ed. Philadelphia: Elsevier; and oxygen: a multicentre study in Japan. Eur J Nucl Med Mol Imaging
2011. 2004;31(5):635–43.
5. Gur D, Yonas H, Jackson DL, et al. Simultaneous measurements of cerebral 27. Hirata M, Sugawara Y, Fukutomi Y, et al. Measurement of radiation dose in
blood flow by the xenon/CT method and the microsphere method: a cerebral CT perfusion study. Radiat Med 2005;23(2):97–103.
comparison. Invest Radiol 1985;20(7):672–7. 28. Cohnen M, Fischer H, Hamacher J, et al. CT of the head by use of reduced
6. Yonas H, Pindzola RP, Johnson DW. Xenon/computed tomography cerebral current and kilovoltage: relationship between image quality and dose
blood flow and its use in clinical management. Neurosurg Clin N Am reduction. Am J Neuroradiol 2000;21(9):1654–60.
1996;7(4):605–16. 29. Nabavi DG, LeBlanc LM, Baxter B, et al. Monitoring cerebral perfusion
7. Coles JP. Imaging of cerebral blood flow and metabolism. Curr Opin after subarachnoid hemorrhage using CT. Neuroradiology 2001;43:
Anesthesiol 2006;19:473–80. 7–16.
8. Horn P, Vajkoczy P, Thome C, et al. Xenon-induced flow activation in 30. Lee EJ, Lee SK, Agid R, et al. Comparison of image quality and radiation
patients with cerebral insult who undergo xenon-enhanced CT blood flow dose between fixed tube current and combined automatic tube current
studies. Am J Neuroradiol 2001;22:1543–9. modulation in craniocervical CT angiography. Am J Neuroradiol
9. Abrahams J, Saha P, Hurst R, et al. Three dimensional bone-free rendering 2009;30:1754–9.
of the cerebral circulation by use of computed tomography and fuzzy 31. Huda W, Sandison GA. The use of the effective dose equivalents, HE,
connectedness. Neurosurgery 2002;51:264–9. for 99mTc labelled radiopharmaceuticals. Eur J Nucl Med 1989;15(4):
10. Ramasundara S, Mitchell PJ, Dowling RJ. Bone subtraction CT angiography 174–9.
for the detection of intracranial aneurysms. J Med Imaging Radiat Oncol 32. Huda W, Sandison GA. Estimates of the effective dose equivalent, HE, in
2010;54(6):526–33. doi:10.1111/j.1754-9485.2010.02211.x. positron emission tomography studies. Eur J Nucl Med 1990;17(3-4):
11. Anderson GB, Steinke DE, Petruk KC, et al. Computed tomographic 116–20.
angiography versus digital subtraction angiography for the diagnosis and 33. Langner S, Stumpe S, Kirsch M, et al. No increased risk for contrast-
early treatment of ruptured intracranial aneurysms. Neurosurgery induced nephropathy after multiple CT perfusion studies of the brain with
1999;45(6):1315–20. a nonionic, dimeric, iso-osmolal contrast medium. Am J Neuroradiol
12. van der Schaaf IC, Velthuis BK, Wermer MJH, et al. New detected 2008;29(8):1525–9.
aneurysms on follow-up screening in patients with previously clipped 34. Newhouse JH, Kho D, Qasim A, et al. Frequency of serum creatinine
intracranial aneurysms comparison with DSA or CTA at the time of SAH. changes in the absence of iodinated contrast material: implications for
Stroke 2005;36:1753–8. studies of contrast nephrotoxicity. Am J Radiol 2008;191:376–82.
13. Dammert S, Krings T, Moller-Hartmann W, et al. Detection of intracranial 35. Hallevi H, Abraham AT, Barreto AD, et al. The spot sign in intracerebral
aneurysms with multislice CT: comparison with conventional angiography. hemorrhage: the importance of looking for contrast extravasation. Stroke
Neuroradiology 2004;46(6):427–34. 2010;41(1):e34–40. Epub 2009, Nov 12.
14. Agid R, Lee SK, Willinsky RA, et al. Acute subarachnoid hemorrhage: using 36. Rosen DS, Macdonald RL. Subarachnoid hemorrhage grading scales: a
64-slice multidetector CT angiography to “triage” patients’ treatment. systematic review. Neurocrit Care 2005;2:110–18.
Neuroradiology 2006;48(11):787–94. 37. Frontera JA, Claassen J, Schmidt JM, et al. Prediction of symptomatic
15. Nabavi DG, Cenic A, Craen RA, et al. Quantitative assessment of cerebral vasospasm after subarachnoid hemorrhage: the modified Fisher scale.
hemodynamics using computed tomography: stability, accuracy, and Neurosurgery 2006;59(1):21–7.
precision studies in dogs. J Comput Assist Tomogr 1999;23:506–15. 38. Fisher CM, Kistler JP, Davis JM. Relation of cerebral vasospasm to
16. Wintermark M, Maeder P, Thiran JP, et al. Quantitative assessment of subarachnoid hemorrhage visualized by computerized tomographic
regional cerebral blood flow by perfusion CT studies at low injection rates: scanning. Neurosurgery 1980;6:1–9.
a critical review of the underlying theoretical models. Eur Radiol 39. Hijdra A, Brouwers PJ, Vermeulen M, et al. Grading the amount of blood
2001;11:1220–30. on computed tomograms after subarachnoid hemorrhage. Stroke
17. Abels B, Klotz E, Tomandl BF, et al. Perfusion CT in acute ischemic stroke: 1990;21:1156–61.
a qualitative and quantitative comparison of deconvolution and maximum 40. Katsnelson M, Mackenzie L, Frangos S, et al. Are initial radiographic and
slope approach. Am J Neuroradiol 2010;31:1690–8. clinical scales associated with subsequent intracranial pressure and brain
18. Kudo K, Sasaki M, Yamada K, et al. Differences in CT perfusion maps oxygen levels after severe traumatic brain injury? Neurosurgery 2011;Nov 9.
generated by different commercial software: quantitative analysis by Epub ahead of print.
using identical source data of acute stroke patients. Radiology 2010;254; 41. Hoeffner EG, Case I, Jain R, et al. Cerebral perfusion CT: technique and
200–9. clinical applications. Radiology 2004;231:632–44.
19. Sase S, Honda M, Machida K, et al. Comparison of cerebral blood flow 42. Leiva-Salinas C, Wintermark M, Kidwell CS. Neuroimaging of cerebral
between perfusion computed tomography and xenon-enhanced computed ischemia and infarction. Neurotherapeutics 2011;8(1):19–27.
tomography for normal subjects: territorial analysis. J Comput Assist 43. McCormack RF, Hutson A. Can computed tomography angiography of the
Tomogr 2005;29(2):270–7. brain replace lumbar puncture in the evaluation of acute-onset headache
20. Sanelli PC, Lev MH, Eastwood JD, et al. The effect of varying user-selected after a negative noncontrast cranial computed tomography scan? Acad
input parameters on quantitative values in CT perfusion maps. Acad Radiol Emerg Med 2010;17(4):444–51.
2004;11(10):1085–92. 44. Romijn M, Gratama van Andel HA, van Walderveen MA, et al. Diagnostic
21. Wintermark M, Lau BC, Chien J, et al. The anterior cerebral artery is an accuracy of CT angiography with matched mask bone elimination for
appropriate arterial input function for perfusion-CT processing in patients detection of intracranial aneurysms: comparison with digital subtraction
with acute stroke. Neuroradiology 2008;50:227–36. angiography and 3D rotational angiography. Am J Neuroradiol 2008;29(1):
22. Waaijera A, van der Schaaf IC, Velthuisa BK, et al. Reproducibility of 134–9.
quantitative CT brain perfusion measurements in patients with 45. Yeung R, Ahmad T, Aviv RI, et al. Comparison of CTA to DSA in
symptomatic unilateral carotid artery stenosis. Am J Neuroradiol 2007;28: determining the etiology of spontaneous ICH. Can J Neurol Sci 2009;
927–32. 36(2):176–80.
23. Fiorella D, Heiserman J, Prenger E, et al. Assessment of the reproducibility 46. Nijjar S, Patel B, McGinn G, et al. Computed tomographic angiography as
of postprocessing dynamic CT perfusion data. Am J Neuroradiol the primary diagnostic study in spontaneous subarachnoid hemorrhage.
2004;25(1):97–107. J Neuroimaging 2007;17(4):295–9.
270.e2 Section IV—Radiology

47. Westerlaan HE, Gravendeel J, Fiore D, et al. Multislice CT angiography in 68. Ferguson SD, Rosen DS, Bardo D, et al. Arterial diameters on catheter and
the selection of patients with ruptured intracranial aneurysms suitable for computed tomographic angiography. Surg Neurol 2010;73(3):165–73.
clipping or coiling. Neuroradiology 2007;49(12):997–1007. 69. Sviri GE, Britz GW, Lewis DH, et al. Dynamic perfusion computed
48. Mohan S, Lee W, Tan JT, et al. Multi-detector computer tomography tomography in the diagnosis of cerebral vasospasm. Neurosurgery 2006;59:
angiography in the initial assessment of patients acutely suspected of 319–25.
having intracranial aneurysm rupture. Ann Acad Med (Singaore) 70. Sviri GE, Mesiwala AH, Lewis DH, et al. Dynamic perfusion computerized
2009;38(9):769–73. tomography in cerebral vasospasm following aneurysmal subarachnoid
49. Prestigiacomo CJ, Sabit A, He W, et al. Three dimensional CT angiography hemorrhage: a comparison with technetium-99m–labeled ethyl cysteinate
versus digital subtraction angiography in the detection of intracranial dimer–single-photon emission computerized tomography. J Neurosurg
aneurysms in subarachnoid hemorrhage. J Neurointerv Surg 2010;2(4): 2006;104:404–10.
385–9. Epub 2010, Jun 24. 71. Sanelli PC, Ugorec I, Johnson CE, et al. Using quantitative CT perfusion for
50. Agid R, Andersson T, Almqvist H, et al. Negative CT angiography findings evaluation of delayed cerebral ischemia following aneurysmal subarachnoid
in patients with spontaneous subarachnoid hemorrhage: when is digital hemorrhage. AJNR Am J Neuroradiol 2011;Sep 29. Epub ahead of print.
subtraction angiography still needed? AJNR Am J Neuroradiol 72. Harrigan MR, Magnano CR, Guterman LR, et al. Computed tomographic
2010;31(4):696–705. Epub 2009, Nov 26. perfusion in the management of aneurysmal subarachnoid hemorrhage:
51. Hochberg AR, Rojas R, Thomas AJ, et al. Accuracy of on-call resident new application of an existent technique. Neurosurgery 2005;56(2):304–17.
interpretation of ct angiography for intracranial aneurysm in subarachnoid 73. Kanazawa R, Kato M, Ishikawab K, et al. Convenience of the computed
hemorrhage. AJR Am J Roentgenol 2011;197(6):1436–41. tomography perfusion method for cerebral vasospasm detection after
52. Tomycz L, Bansal NK, Hawley CR, et al. “Real-world” comparison of subarachnoid hemorrhage. Surg Neurol 2007;67(6):604–11.
non-invasive imaging to conventional catheter angiography in the 74. Pham M, Johnson A, Bartsch AJ, et al. CT perfusion predicts secondary
diagnosis of cerebral aneurysms. Surg Neurol Int 2011;2:134. Epub 2011, cerebral infarction after aneurysmal subarachnoid hemorrhage. Neurology
Sep 30. 2007;69:762–5.
53. Kershenovich A, Rappaport ZH, Maimon S. Brain computed tomography 75. Dankbaar JW, Rijsdijk M, van der Schaaf IC, et al. Relationship between
angiographic scans as the sole diagnostic examination for excluding vasospasm, cerebral perfusion, and delayed cerebral ischemia after
aneurysms in patients with perimesencephalic subarachnoid hemorrhage. aneurysmal subarachnoid hemorrhage. Neuroradiology
Neurosurgery 2006;59(4):798–801. 2009;51(12):813–19.
54. Miley JT, Taylor RA, Janardhan V, et al. The value of computed tomography 76. Dankbaar JW, de Rooij NK, Velthuis BK, et al. Diagnosing delayed cerebral
angiography in determining treatment allocation for aneurysmal ischemia with different CT modalities in patients with subarachnoid
subarachnoid hemorrhage. Neurocrit Care 2008;9(3):300–6. hemorrhage with clinical deterioration. Stroke 2009;40(11):3493–8. Epub
55. Wermer MJH, van der Schaaf IC, Velthuis BK, et al. Follow-up screening 2009, Sep 17.
after subarachnoid haemorrhage: frequency and determinants of new 77. Van de Schaaf IC, Wermer MJ, van der Graaf Y, et al. CT after subarachnoid
aneurysms and enlargement of existing aneurysms. Brain 2005 (128): hemorrhage: relation of cerebral perfusion to delayed cerebral ischemia.
2421–9. Neurology 2006;66:1533–8.
56. Wallace RC, Karis JP, Partovi S, et al. Noninvasive imaging of treated 78. van der Schaaf IC, Wermer MJ, van der Graaf YA, et al. Prognostic value of
cerebral aneurysms, part II: CT angiographic follow-up of surgically cerebral perfusion-computed tomography in the acute stage after
clipped aneurysms. Am J Neuroradiol 2007;28:1207–12. subarachnoid hemorrhage for the development of delayed cerebral
57. Uysal E, Ozel A, Erturk SM, et al. Comparison of multislice computed ischemia. Stroke 2006;37:409–13.
tomography angiography and digital subtraction angiography in the 79. Dankbaar JW, de Rooij NK, Smit EJ, et al. Changes in cerebral perfusion
detection of residual or recurrent aneurysm after surgical clipping with around the time of delayed cerebral ischemia in subarachnoid hemorrhage
titanium clip. Acta Neurochir (Wien) 2009;151(2):131–5. patients. Cerebrovasc Dis 2011;32(2):133–40. Epub 2011, Jul 19.
58. Charpentier C, Audibert G, Guillemin F, et al. Multivariate analysis of 80. Sanelli PC, Jou A, Gold R, et al. Using CT perfusion during the early
predictors of cerebral vasospasm occurrence after aneurysmal subarachnoid baseline period in aneurysmal subarachnoid hemorrhage to assess for
hemorrhage. Stroke 1999;30(7):1402–8. development of vasospasm. Neuroradiology 2011;53(6):425–34. Epub 2010,
59. Diringer MN, Bleck TP, Claude Hemphill J 3rd, et al. Critical care Aug 7.
management of patients following aneurysmal subarachnoid 81. Nogueira RG, Lev MH, Roccatagliata L, et al. Intra-arterial nicardipine
hemorrhage: recommendations from the Neurocritical Care Society’s infusion improves CT perfusion-measured cerebral blood flow in patients
Multidisciplinary Consensus Conference. Neurocrit Care 2011;Jul 20. Epub with subarachnoid hemorrhage-induced vasospasm. AJNR Am J
ahead of print. Neuroradiol 2009;30(1):160–4. Epub 2008, Oct 22.
60. Washington CW, Zipfel GJ, Participants in the International Multi- 82. Huang AP, Arora S, Wintermark M, et al. Perfusion computed
disciplinary Consensus Conference on the Critical Care Management of tomographic imaging and surgical selection with patients after poor-grade
Subarachnoid Hemorrhage. Detection and monitoring of vasospasm and aneurysmal subarachnoid hemorrhage. Neurosurgery 2010;67(4):964–74;
delayed cerebral ischemia: a review and assessment of the literature. discussion 975.
Neurocrit Care 2011;15(2):312–7. 83. The National Institute of Neurological Disorders and Stroke rt-PA Stroke
61. Willensky RA, Taylor SM, terBrugge K, et al. Neurologic complications of Study Group. Tissue plasminogen activator for acute ischemic stroke.
cerebral angiography: prospective analysis of 2,899 procedures and review N Engl J Med 1995;333:1581–8.
of the literature. Radiology 2003;227:522–8. 84. Hacke W, Kaste M, Fieschi C, et al. Randomised double-blind placebo-
62. Le Roux P, Elliott JP, Eskridge JM, et al. Risks and benefits of diagnostic controlled trial of thrombolytic therapy with intravenous alteplase in acute
angiography following aneurysm surgery: a retrospective analysis of 597 ischaemic stroke (ECASS II). Lancet 1998;352:1245–51.
studies. Neurosurgery 1998;42:1248–55. 85. Schramm P, Schellinger PD, Klotz E, et al. Comparison of perfusion
63. Soucy JP, McNamara D, Mohr G, et al. Evaluation of vasospasm secondary computed tomography and computed tomography angiography source
to subarachnoid hemorrhage with technetium 99m hexamethyl- images with perfusion-weighted imaging and diffusion-weighted imaging
propyleneamine oxime (HMPAO) tomoscintigraphy. J Nucl Med 1990;31: in patients with acute stroke of less than 6 hours duration. Stroke 2004;35:
972–7. 1652–8.
64. Rajendran JG, Lewis DH, Newell DW, et al. Brain SPECT used to evaluate 86. Smith WS, Tsao JW, Billings ME, et al. Prognostic significance of
vasospasm after subarachnoid hemorrhage: correlation with angiography angiographically confirmed large vessel intracranial occlusion in patients
and transcranial Doppler. Clin Nucl Med 2001;26(2):125–30. presenting with acute brain ischemia. Neurocrit Care 2006;4(1):14–7.
65. Shankar JJ, Tan IY, Krings T, et al. CT angiography for evaluation of 87. Tan JC, Dillon WP, Liu S, et al. Systematic comparison of perfusion-CT
cerebral vasospasm following acute subarachnoid haemorrhage. and CT-angiography in acute stroke patients. Ann Neurol 2007;61(6):
Neuroradiology 2011;May 4. Epub ahead of print. 533–43.
66. Anderson GB, Ashforth R, Steinke DE, et al. CT angiography for the 88. Salottolo KM, Fanale CV, Leonard KA, et al. Multimodal imaging does not
detection of cerebral vasospasm in patients with acute subarachnoid delay intravenous thrombolytic therapy in acute stroke. AJNR Am J
hemorrhage. Am J Neuroradiol 2000;21(6):1011–15. Neuroradiol 2011;32(5):864–8. Epub 2011, Feb 10.
67. Otawara Y, Ogasawara K, Ogawa A, et al. Evaluation of vasospasm after 89. Gentile NT, Cernetich J, Kanamalla US, et al. Expedited computed
subarachnoid hemorrhage by use of multislice computed tomographic tomography perfusion and angiography in acute ischemic stroke: a
angiography. Neurosurgery 2002;51(4):939–42. feasibility study. J Emerg Med 2011;Apr 15. Epub ahead of print.
Section IV—Radiology 270.e3

90. Lima FO, Lev MH, Levy RA, et al. Functional contrast-enhanced CT for combinations of computed tomographic predictors. Neurosurgery
evaluation of acute ischemic stroke does not increase the risk of contrast- 2005;57(6):1173–82.
induced nephropathy. AJNR Am J Neuroradiol 2010;31(5):817–21. Epub 114. Chun KA, Manley GT, Stiver SI, et al. Interobserver variability in the
2009, Dec 31. assessment of CT imaging features of traumatic brain injury.
91. Young KC, Benesch CG, Jahromi BS. Cost-effectiveness of multimodal CT J Neurotrauma 2010;27(2):325–30.
for evaluating acute stroke. Neurology 2010;75(19):1678–85. Epub 2010, 115. Lingsma HF, Roozenbeek B, Steyerberg EW, et al. Early prognosis in
Oct 6. traumatic brain injury: from prophecies to predictions. Lancet Neurol
92. Ledezma CJ, Wintermark M. Multimodal CT in stroke imaging: new 2010;9(5):543–54.
concepts. Radiol Clin North Am 2009;47(1):109–16. 116. Maas AI, Steyerberg EW, Butcher I, et al. Prognostic value of computerized
93. Leiva-Salinas C, Wintermark M. Imaging of acute ischemic stroke. tomography scan characteristics in traumatic brain injury: results from the
Neuroimaging Clin N Am 2010;20(4):455–68. IMPACT study. J Neurotrauma 2007;24(2):303–14.
94. Turk A, Magarik JA, Chaudry I, et al. CT perfusion-guided patient selection 117. Burlew CC, Biffl WL. Blunt cerebrovascular trauma. Curr Opin Crit Care
for endovascular treatment of acute ischemic stroke is safe and effective. 2010;Aug 31. Epub ahead of print.
J Neurointerv Surg 2011;Sep 19. Epub ahead of print. 118. Löhrer L, Vieth V, Nassenstein I, et al. Blunt cerebrovascular injuries in
95. Michel P, Ntaios G, Reichhart M, et al. Perfusion-CT guided intravenous acute trauma care: a screening protocol. Eur Spine J 2011;Sep 6. Epub
thrombolysis in patients with unknown-onset stroke: a randomized, ahead of print.
double-blind, placebo-controlled, pilot feasibility trial. Neuroradiology 119. Kaye D, Brasel KJ, Neideen T, et al. Screening for blunt cerebrovascular
2011;Aug 2. Epub ahead of print. injuries is cost-effective. J Trauma 2011;70(5):1051–6; discussion
96. Hom J, Dankbaar JW, Soares BP, et al. Blood-brain barrier permeability 1056–7.
assessed by perfusion CT predicts symptomatic hemorrhagic 120. DiCocco JM, Emmett KP, Fabian TC, et al. Blunt cerebrovascular injury
transformation and malignant edema in acute ischemic stroke. AJNR Am J screening with 32-channel multidetector computed tomography: more
Neuroradiol 2011;32(1):41–8. Epub 2010, Oct 14. slices still don’t cut it. Ann Surg 2011;253(3):444–50.
97. Bonatti G, Ferro F, Haglmüller T, et al. Basilar artery thrombosis: imaging and 121. York G, Barboriak D, Petrella J, et al. Association of internal carotid artery
endovascular therapy. Radiol Med 2010;115(8):1219–33. Epub 2010, Jul 31. injury with carotid canal fractures in patients with head trauma. Am J
98. Sylaja PN, Puetz V, Dzialowski I, et al. Prognostic value of CT angiography Roentgenol 2005;184(5):1672–8.
in patients with suspected vertebrobasilar ischemia. J Neuroimaging 122. Cothren CC, Moore EE, Ray CE Jr., et al. Screening for blunt
2008;18(1):46–9. cerebrovascular injuries is cost-effective. Am J Surg 2005;190(6):845–9.
99. Poisson SN, Nguyen-Huynh MN, Johnston SC, et al. Intracranial large 123. Cothren CC, Moore EE, Ray CE Jr., et al. Cervical spine fracture patterns
vessel occlusion as a predictor of decline in functional status after transient mandating screening to rule out blunt cerebrovascular injury. Surgery
ischemic attack. Stroke 2011;42(1):44–7. Epub 2010, Nov 24. 2007;141(1):76–82.
100. Konstas AA, Wintermark M, Lev MH. CT perfusion imaging in acute 124. Biffl WL, Moore EE, Ryu RK, et al. The unrecognized epidemic of blunt
stroke. Neuroimaging Clin N Am 2011;21(2):215–38, ix. carotid arterial injuries: early diagnosis improves neurologic outcome. Ann
101. Lui YW, Tang ER, Allmendinger AM, et al. Evaluation of CT perfusion in Surg 1998;228(4):462–70.
the setting of cerebral ischemia: patterns and pitfalls. AJNR Am J 125. Biffl WL, Moore EE, Offner PJ, et al. Optimizing screening for blunt
Neuroradiol 2010;31(9):1552–63. Epub 2010, Feb 25. cerebrovascular injuries. Am J Surg 1999;178(6):517–22.
102. Schaefer PW, Roccatagliata L, Ledezma C, et al. First-pass quantitative CT 126. Berne JD, Reuland KS, Villarreal DH, et al. Sixteen-slice multi-detector
perfusion identifies thresholds for salvageable penumbra in acute stroke computed tomographic angiography improves the accuracy of screening for
patients treated with Intra-arterial therapy. Am J Neuroradiol 2006;27(1): blunt cerebrovascular injury. J Trauma 2006;60(6):1204–9.
20–5. 127. Biffl WL, Egglin T, Benedetto B, et al. Sixteen-slice computed tomographic
103. Eastwood JD, Lev MH, Azhari T, et al. CT perfusion scanning with angiography is a reliable noninvasive screening test for clinically significant
deconvolution analysis: pilot study in patients with acute middle cerebral blunt cerebrovascular injuries. J Trauma 2006;60(4):745–51.
artery stroke. Radiology 2002;222(1):227–36. 128. Eastman AL, Chason DP, Perez CL, et al. Computed tomographic
104. Campbell BCV, Christensen S, Levi CR, et al. Cerebral blood flow is the angiography for the diagnosis of blunt cervical vascular injury: is it ready
optimal CT perfusion parameter for assessing infarct core. Stroke 2011;42: for primetime? J Trauma 2006;60(5):925–9.
3435–40. 129. Utter GH, Hollingworth W, Hallam DK, et al. Sixteen-slice CT angiography
105. Hossman KA. Neuronal survival and revival during and after cerebral in patients with suspected blunt carotid and vertebral artery injuries. J Am
ischemia. Am J Emerg Med 1983;1(2):191–7. Coll Surg 2006;203(6):838–48.
106. Powers WJ, Grubb RL Jr., Darriet D, et al. Cerebral blood flow and cerebral 130. Malhotra AK, Camacho M, Ivatury RR, et al. Computed tomographic
metabolic rate of oxygen requirements for cerebral function and viability in angiography for the diagnosis of blunt carotid/vertebral artery injury: a
humans. J Cereb Blood Flow Metab 1985;5(4):600–8. note of caution. Ann Surg 2007;246:632–43.
107. Silvennoinen HM, Hamberg LM, Lindsberg PJ, et al. CT perfusion identifies 131. Soustiel JF, Mahamid E, Goldsher D, et al. Perfusion-CT for early
increased salvage of tissue in patients receiving intravenous recombinant assessment of traumatic cerebral contusions. Neuroradiology
tissue plasminogen activator within 3 hours of stroke onset. Am J 2008;50:189–96.
Neuroradiol 2008;29(6):1118–23. 132. Wintermark M, Chioléro R, van Melle G, et al. Relationship between
108. Knoepfli AS, Sekoranja L, Bonvin C, et al. Evaluation of perfusion CT and brain perfusion computed tomography variables and cerebral perfusion
TIBI grade in acute stroke for predicting thrombolysis benefit and clinical pressure in severe head trauma patients. Crit Care Med 2004;32(7):
outcome. J Neuroradiol 2009;36(3):131–7. 1579–87.
109. Wintermark M, Reichhart M, Cuisenaire O, et al. Comparison of admission 133. Wintermark M, Chiolero R, Van Melle G, et al. Cerebral vascular
perfusion computed tomography and qualitative diffusion- and perfusion- autoregulation assessed by perfusion CT in severe head trauma patient.
weighted magnetic resonance imaging in acute stroke patients. Stroke J Neuroradiol 2006;33(1):27–37.
2002;33(8):2025–31. 134. Berenguer CM, Davis FE, Howington JU. Brain death confirmation:
110. Soares BP, Tong E, Hom J, et al. Reperfusion is a more accurate predictor of comparison of computed tomographic angiography with nuclear medicine
follow-up infarct volume than recanalization: a proof of concept using CT perfusion scan. J Trauma 2010;68(3):553–9.
in acute ischemic stroke patients. Stroke 2010;41(1):e34–40. Epub 2009, 135. Quesnel C, Fulgencio JP, Adrie C, et al. Limitations of computed
Nov 12. tomographic angiography in the diagnosis of brain death. Intensive Care
111. Zhao L, Barlinn K, Bag AK, et al. Computed tomography perfusion Med 2007;33(12):2129–35.
prognostic maps do not predict reversible and irreversible neurological 136. Combes JC, Chomel A, Ricolfi F, et al. Reliability of computed tomographic
dysfunction following reperfusion therapies. Int J Stroke 2011;6(6):544–6. angiography in the diagnosis of brain death. Transplant Proc
doi:10.1111/j.1747-4949.2011.00681.x. 2007;39(1):16–20.
112. Marshall LF, Bowers-Marshall S, Klauber MR, et al. A new classification of 137. Swanson E, Mascitelli J, Stiefel M, et al. The effect of patient transport on
head injury based on computerized tomography. J Neurosurg 1991;75 brain oxygen in comatose patients. Neurosurgery 2010;66:925–32.
(Suppl):S14–20. 138. Larovere KL, Brett MS, Tasker RC, et al. Head computed tomography
113. Maas AI, Hukkelhoven CW, Marshall LF, et al. Prediction of outcome in scanning during pediatric neurocritical care: diagnostic yield and the
traumatic brain injury with computed tomographic characteristics: a utility of portable studies. Neurocrit Care 2011;Sep 21. Epub ahead of
comparison between the computed tomographic classification and print.
270.e4 Section IV—Radiology

139. Carlson AP, Yonas H. Portable head computed tomography scanner- 141. Stevens GC, Rowles NP, Foy RT, et al. The use of mobile computed
technology and applications: experience with 3421 scans. tomography in intensive care: regulatory compliance and radiation
J Neuroimaging 2011;Jun 23. doi:10.1111/j.1552-6569.2011.00621.x. Epub protection. J Radiol Prot 2009;29(4):483–90. Epub 2009, Nov 18.
ahead of print. 142. Peace K, Maloney E, Frangos S, et al. Portable head CT scan and its effect
140. Rumboldt Z, Huda W, All JW. Review of portable CT with assessment of a on intracranial pressure, cerebral perfusion pressure and brain oxygen.
dedicated head CT scanner. AJNR Am J Neuroradiol 2009;30(9):1630–6. J Neurosurgery 2011;114:1479–84.
Epub 2009, Aug 6.
Chapter
27  
IV

Xenon-Enhanced Computed
Tomography
Andrew P. Carlson and Howard Yonas

Studies with xenon 133 use the “wash-out” curve of this


Introduction tracer, whereas studies with stable xenon focus on the “wash-
Xenon-enhanced computed tomography (Xe-CT) is a power- in” curve. Use of a wash-in curve reduces any theoretical
ful physiologic imaging tool that provides a clinician with alteration of flow measurement due to an activation of CBF
quantitative cerebral blood flow (CBF) data, allows the physi- after xenon inhalation and has been validated in other quan-
ologic response to various interventions to be measured, titative CBF studies.3-9 In 2000 the dose of xenon was reduced
and provides prognostic data to guide patient management. to 28% because newer CT scanners had a lower noise level.
Though the data provided are in a tomographic format, This helped reduce the few sensorial effects associated with
it provides quantitative information for critical decision 33% xenon and reduced cost. With 28% xenon, studies can be
making when needed. In this way Xe-CT CBF is similar to the routinely performed in cooperative outpatients and in venti-
CBF monitors discussed in subsequent chapters rather than lated ICU patients.
imaging technologies. This chapter discusses the theory and To calculate CBF using Xe-CT requires a modification of
physics behind the data provided, the practical aspects of the Kety-Schmidt equation. Two variables are needed to solve
Xe-CT, and the clinical decision-making process in several the equation: (1) the arterial concentration curve of the agent
neurologic conditions encountered in the neurocritical care and (2) the extent and time course of tissue arrival that
unit (NCCU) based on quantitative CBF data. depends on the blood flow and the blood-brain partition
coefficient, or lambda. The arterial concentration is measured
indirectly from the end-tidal xenon concentration; this is a
Technology very good approximation of a highly diffusible gas such as
xenon. The brain partition coefficient and flow is calculated
Theory and Physiology for each CT pixel by solving the Kety-Schmidt for both vari-
CBF can be measured by a number of diffusible tracers. Xenon ables using an iterative mathematical approach:
133 was an ideal early tracer because xenon is highly lipid Cxe br (t) = λk o ∫ Cxe Art (u) e − k (t −u )du
t

soluble and its 133 isotope can be readily tracked with external
scintillation counters. Using this technology, early studies where Cxebr (t) is the time dependent brain xenon concentra-
showed that hemispheric and then superficial regional flow tion, λ is the blood-brain partition coefficient, k is the brain
could be measured in mL/100 g/min. However, because radio- uptake flow rate constant, CxeArt(u) is the time-dependent
isotope precautions were needed and resolution in the depth arterial xenon concentration. This calculation is performed
of the brain was limited, routine intensive care unit (ICU) typically at 4 CT levels; however, 8 to 10 levels can be used
application was not possible. In the late 1970s, once it was with newer multidetector scanners.
reported that stable xenon was radiodense, similar to iodine, The calculated CBF values then are represented graphically
CT imaging techniques and the ability to tomographically in a standardized color-coded fashion for each pixel (Figs.
image CBF were reported.1,2 Tens of thousands of Xe-CT CBF 27.1 to 27.3). A bell-shaped filter is applied to aid visual
studies have been done since then without significant compli- display. To obtain useful regional values, the cortical mantle
cations. When first available clinically, use of xenon in the can automatically be divided into 20 2-cm-deep regions of
United States received “grandfather” approval as a contrast interest (ROI), and the average for each region displayed.
agent from the U.S. Food and Drug Administration (FDA). Alternatively, any ROI of regular or irregular shape and size
When Praxair, the company that supplied medical-grade can be selected by the user on either the CT or the CBF image
xenon, withdrew from the rare-gas market, xenon lost clinical (see Fig. 27.1). Additional analysis can be performed to assess
approval, and currently its use in the United States occurs the validity and further manipulate the data (Figs. 27.2 to
under investigational new drug (IND) and research protocols. 27.7). For mixed cortical sources, CBF in the range of 54 plus
A reapplication for clinical approval has been submitted to or minus 10 mL/100 g/min is normal. Values less than 15 to
the FDA. 20 mL/100 g/min represent ischemia and are associated with
© Copyright 2013 Elsevier Inc. All rights reserved. 271
272 Section IV—Radiology

Fig. 27.1  Standard xenon-enhanced


computed tomography (Xe-CT)
study with quantitative cerebral
blood flow (CBF) values for each of
the approximately 24,000 CT voxels.
The baseline scan is displayed on the
left, and the CBF coded image on the
right. The image on the right is color
coded to the range of CBF values
displayed on the right of the image. The
cortical mantle can be automatically
divided into 20 regions of interest. The
mean CBF within each region is
displayed in the box below. The tissue
heterogeneity (TH) is a measure of the
variability of CBF values within the
region. The number of voxels (area) and
mean Hounsfield unit (mean HU)
enhancement also are displayed for
each region of interest. The user also
can define regions of interest as shown
in areas 21 (right deep frontal white
matter) and 22 (left basal ganglia).

Fig. 27.2  Xenon-enhanced computed tomography (Xe-CT) image showing cerebral blood flow (CBF) values at each of the four levels
typically studied. The top row shows the baseline images, the middle row visually displays the color-coded CBF values (with the scale on the right).
The bottom row is a measure of the data reliability. Low confidence indicates that movement or other factors may have interfered with data acquisition.
High confidence values (blue) indicate that the values obtained are valid.
Section IV—Radiology 273

Fig. 27.3  This xenon-enhanced computed tomography (Xe-CT) display shows the same image divided into six regions of interest with
the average cerebral blood flow (CBF) values displayed in the left box. The upper left scan is the baseline image, the upper right image is
the CBF data, the lower right is the confidence of the study, and the graph displays the frequency of occurrence of each numeric CBF value in the
scan slice.

BL 1 Base BL 1 Base
BL 2 1.09% BL 2 1.04%
CT 1 4.09% CT 1 3.15%
CT 2 4.33% CT 2 4.60%
CT 3 5.28% CT 3 7.38%
CT 4 7.38% CT 4 9.83%
CT 5 13.37% CT 5 12.24%
CT 6 20.49% CT 6 11.88%

0 15 30 45 0 15 30 45
(%) (%)

Tolerance level
Below tolerance level
Above tolerance level

Fig. 27.4  To further ensure the validity of the cerebral blood flow (CBF) study, this graph displays the consistency in position of each
of the six serial scans required for the data acquisition (CT 1-6). The computer compares the relative position of the inner table from scan to
scan, and movement of less than 15% from the baseline images (BL1 and BL2) is considered within tolerance. The graph on the left shows significant
movement in the last image, making the data less reliable, whereas the graph on the right shows a study with low movement and good reliability.
274 Section IV—Radiology

IMG. INCLUDED: 1 2 3 4 5 6 160


15 HU 150
10 140
130
5
120
0 110
0 2 4
Time (min) 100
90
80
Fig. 27.5  The raw data acquisition data can be
reviewed by the user for any data point in the image 70
as a graph of increasing Hounsfield unit (HU) 60
15 HU
enhancement over time. The six data points represent 50
the six images acquired to make the cerebral blood flow 10 40
(CBF) calculation. The upper graph represents the normal 30
5
enhancement in a region of gray matter, as indicated by 20
the upper arrow, and the lower graph represents the lack 0 10
0 2 4
of enhancement as seen in the ventricle as indicated by the Time (min) 0
lower arrow.

Fig. 27.6  A comparison cerebral blood flow (CBF) study. The upper images are the two studies compared side by side. The first study is done
with the patient sedated and the PaCO2 35 mm Hg. In the second study, the patient was allowed to respire normally, which brought her PaCO2 to
28 mm Hg. This was done to determine whether to allow the patient to continue to hyperventilate or sedate her to bring her PaCO2 to a more
normal level. The lower right image shows quantitatively the difference between the first and second studies—the red values showing an increase in
CBF and the blue values showing a decrease. These data show that the patient had improved CBF with a lower PaCO2. She was therefore allowed
to continue with the lower values, avoiding the complications of sedation or paralytics, and objectively improving her CBF.

neurologic deficit and infarction.2,10-14 Normal gray matter parameters such as arterial carbon dioxide pressure (PaCO2)
blood flow is 84 mL/100 g/min and white matter is 20 mL/ or blood pressure (see Fig. 27.6). Today a four-level study,
100 g/min. The mixed cortical flow and gray matter flow which measures almost 100,000 flow values on four CT levels
decrease with age, but white matter flow remains constant requires less than 10 seconds. This quick study time allows
(Fig. 27.8).13 Xe-CT CBF–derived flow information to be used in prospec-
The half-life of arterial xenon concentration is about 30 tive clinical decision making. For example, each mm Hg
seconds, and 99% of the agent is washed out of the body increase of PaCO2 should increase CBF 3% in the normal
within 5 minutes (see Fig. 27.7). This makes it possible to patient. The amount of flow augmentation associated with
repeat the study within 10 minutes while varying physiologic changes in PaCO2 provides insight about the vasodilation
Section IV—Radiology 275

CO2
60

45

30

15

0
–2 –1 0 1 2 3 4 5 6
Time (min)

XENON
40 Fig. 27.7  These two graphs are the respiratory data
followed during a xenon cerebral blood flow (CBF)
30 study. The top graph shows the end-tidal CO2 during the
study. The bottom graph shows the end-tidal xenon, with
20 the patient breathing the xenon gas starting at time 0. The
curve of xenon wash-in is a very good approximation of
arterial concentration of xenon, given its high diffusibility.
10
This makes obtaining this input function into the CBF
calculation possible without arterial sampling, and with
0
accurate data of the wash-in curve rather than relying on 
–2 –1 0 1 2 3 4 5 6
a standard curve. The xenon washes out very quickly 
Time (min) (~1 minute) after the gas is turned off.

120 PURE GRAY MATTER Standard deviation Devices and Engineering


100 The Xe-CT CBF data depend on several components: (1) soft-
90 ware that uses CT images to document the arrival of xenon
CBF (mL/100 g/min)

80 within the brain; (2) xenon end-tidal data recorded by thermal


*y = (–
70 .45) x conductivity as an indirect measure of arterial levels of xenon;
+ 106.5
MIXED CORTICAL
60 (3) a computer to handle the complex mathematical process;
50 and (4) hardware that maintains and delivers fixed concentra-
40 **y = (–.21) x tions of xenon and oxygen. Significant advances have been
+ 61.13 made in each of these components. In the early 1980s, the
30
PURE WHITE MATTER delivery system was a large bag filled with the desired gas
20
10 mixture. Today a closed loop system that can mix and main-
N.S. tain the desired concentrations while scrubbing CO2 is used
0
10 20 30 40 50 60 70 80
in part because of cost. This reduced the amount of xenon
used from 20 to 4.5 L/study. The initial computers required
Age (years) an hour to compute a few-level study and the transfer of data
*p < .0001 required moving reel-to-reel stored data. Today data transfer
**p < .0025
is instantaneous and the calculation of 24,000 flow values on
Fig. 27.8  Normative cerebral blood flow (CBF) values and the each of four computer tomography levels requires a few
variation with age are displayed. Gray matter CBF decreases with seconds.
age, whereas white matter CBF remains relatively constant. (From Yonas The evolution of CT scanners means that a multilevel high-
H, Darby JM, Marks EC, et al. CBF measured by Xe-CT: approach to analysis and
resolution CBF study is now possible with lower concentra-
normal values. J Cereb Blood Flow Metab 1991;11:716–25.)
tions of xenon (28% rather than 33%). Because there is
variation between CT scanners, a “phantom” has been devel-
oped to standardize data if needed (Fig. 27.9). The goal is to
status of brain regions being examined. This response may be maintain a signal-to-noise ratio of at least 8 : 1; the lower
altered in various clinical situations (see following). Anxiety xenon concentration is possible because modern scanners
and arrhythmias may result when CO2 is used as a vasodila- have significantly reduced the noise level that does not com-
tory agent. Consequently acetazolamide, which is a carbonic promise the confidence in the numerical validity of the tech-
anhydrase inhibitor that does not have as many side effects nology. The lowest concentration of xenon that provides valid
but that causes tissue acidosis similar to 5% carbon dioxide data is the goal because xenon, although it is an inert noble
(CO2) inhalation, is used more frequently in clinical practice. gas, has properties that may alter neuron function. At 80%
When tissue loses its ability to augment CBF in response to a xenon is a very safe and effective anesthetic agent with very
vasodilatory challenge, it implies maximal vasodilation and so rapid onset and offset of action. At 33% xenon may cause
no further reserve. This concept is helpful in deciding about dysesthesias and altered consciousness in about 3% to 5% of
hemodynamic reserve and subsequent stroke risk.15-23 patients. These complications are observed in less than 1% of
276 Section IV—Radiology

CBF Histogram CBF


5%
Frequency in percentage Confidence
4% Worst

3% 1%
3%
2%
6%
1% 90%

0% 0%
0 40 80 120 160
CC/100 grams/Min Best

Fig. 27.9  Xenon-enhanced computed tomography (Xe-CT) of a phantom device, allowing for standardization of cerebral blood flow (CBF) data with
any CT scanner.

patients with 28% xenon. Apnea is a concern with exposure


to 100% xenon, but medically significant apnea does not occur
using 33% xenon. Only brief episodes of respiratory irregular-
ity (<1% of patients) that is reversible with coaching may be
observed using 28% xenon. Patients occasionally describe
some anxiety or mild euphoria, which clears within minutes
of study cessation. Vital signs are not typically affected.
The only component of the Xe-CT CBF system that has
not changed over the past 20 years is the mathematical
approach to calculate CBF. This methodology has undergone
extensive validation studies with multiple technologies, for
example, microspheres,3 iodoantipyrine4 and in vivo xenon
133,9 positron emission tomography (PET),6 and thermal
dilution flow probes.8 Consequently the early literature that
describes the Xe-CT CBF method remains relevant to current
technology. This methodology also is being integrated within Fig. 27.10  Portable computed tomography (CT) scanner and xenon
modern portable CT scanning technology (e.g., CereTom- delivery system stored in hallway of the neurocritical care unit.
NeuroLogica: Danvers, MA; Fig. 27.10). Even though epi-
sodic, this bedside tomographic quantitative data can help
guide patient care in the NCCU24-26 and complement the con-
tinuous data from invasive monitors that provide more local (Fig. 27.11). The patient should be cooperative, amenable to
information.27 In particular, portable imaging eliminates intravenous (IV) sedation, or mechanically ventilated because
many of the risks associated with hospital transport of the the head position must be stable during the 4.5 minutes of
critically ill. The image quality is reliable.28-30 In addition, data acquisition. The xenon delivery system is attached to the
information from Xe-CT permits correlation of anatomy and patient’s mask or to the ventilator circuit. The end-tidal CO2
physiology. and end-tidal xenon concentrations are monitored continu-
ously and displayed on the system monitor. The baseline CT
levels for the study are identified and imaged before starting
Xe-CT Cerebral Blood Flow Procedure xenon inhalation. Next, xenon inhalation is started (28%) and
A typical Xe-CT study is described. Once the patient is identi- the scanner begins serial imaging at the same preset levels as
fied for the study (see the following text for indications), he the arterial xenon concentration increases. The first 2 minutes
or she is brought to the CT scanner, or a portable scanner is are the most critical phase of data acquisition because head
taken to the patient’s bedside. The xenon technology can be movement at this time will make the study quantitatively
used with most CT scanners with only minimal adaptation unreliable, while movement later allows for salvage of the
Section IV—Radiology 277

Neurocritical Care Unit Applications


Patient management in the NCCU can be guided by published
recommendations and guidelines.34,35 Ideally management
needs to be patient specific and targeted, and it is in these
circumstances that information provided by Xe-CT can be
useful. There are many potential clinical scenarios in which
Xe-CT can be useful, including identification of otherwise
unrecognized problems, confirmation of clinical diagnoses,
confirmation of information from other monitors, titration of
specific treatments, and avoidance of adverse treatment
effects.26 In a recent prospective study of 2003 Xe CBF studies
at seven centers, 93% of studies were considered clinically
useful.33 The majority of studies were performed for ischemic
stroke and occlusive vascular disease.
Fig. 27.11  A xenon-enhanced computed tomography (Xe-CT)
system at a patient’s bedside. The xenon computer is in the lower left
of the picture in front of the chair. The control console for the portable
Subarachnoid Hemorrhage and Vasospasm
CT scanner is in the foreground on the right. The scanner itself is seen Following aneurysmal subarachnoid hemorrhage (SAH),
in the background in the upper left of the frame at the patient’s head. vasospasm, or more appropriately, delayed ischemic neuro-
The xenon delivery system is attached to the ventilator circuit (not seen). logic deficit (DIND) can adversely affect patient outcome.
Many patients (~70%) develop some angiographic evidence
for vasospasm,36,37 but far fewer develop clinical ischemia.37,38
study by exclusion of later poor quality data. Movement In these patients Xe-CT is the only imaging or blood flow
during image acquisition degrades the study because the base- technique that can identify tissue at risk for infarction14 and
line images are subtracted from the subsequent images to get measure clinical response to an intervention. In addition,
a measure of the change of Hounsfield units. The CT data are Xe-CT has been used to assess and compare therapies for
then transferred to the xenon workstation that contains the DIND, for example, normovolemia or hypervolemia,39 or
time-linked air (arterial) curve data. cardiac output augmentation compared with hypertension
At the workstation, the computer uses the Hounsfield only.37,40 In particular Xe-CT is very predictive of infarction in
enhancement and the end-tidal xenon data to solve the inte- SAH patients with vasospasm. Those who demonstrate hemo-
gral equation for each pixel of the CT scan. The program also dynamically significant vasospasm (i.e., CBF between 6 and
analyzes for the numerical reliability of the data, which needs 15 mL/100 g/min) likely will develop infarction in that terri-
to be factored into the clinical analysis of the CBF data. The tory,41,42 whereas in patients in whom CBF is greater than
data then can be manipulated using the software. ROIs can be 19 mL/100 g/min, infarction is very rare.41
studied where each pixel’s flow is averaged over the entire ROI. CBF may be reduced after SAH often independent of
intracranial pressure (ICP) and cerebral perfusion pressure
(CPP)43; this decrease, however, is usually associated with the
Limitations severity of SAH. In these patients, Xe-CT may also be used
There are several potential limitations when Xe-CT scans are to answer routine management questions such as identify
performed. First are inherent risks of radiation exposure. the optimal blood pressure or PaCO2 level. This allows the
There are multiple serial slices; these should be directed above physician to determine which interventions will be beneficial
the orbit and lens (the most radiosensitive structure of the in a specific patient and what the best “threshold” for treat-
head). Second, ventilation may be altered, but at 28% xenon ment is in an individual. This can be very useful in SAH
rarely if ever causes any significant alteration of ventilation because large fluid volumes often may be given, and here
pattern. Third, the study takes longer than a standard CT scan Xe-CT can help differentiate hyperemia from vasospasm-
to set up and requires experienced personnel to operate the causing ischemia because in both circumstances transcranial
equipment. Fourth, data can be limited by motion artifact Doppler flow velocities can be increased.44-47 Use of Xe-CT to
during scanning. Fifth, the technology and equipment are not examine the efficacy of hypertonic saline48,49 and identify
available at many institutions. Finally, the FDA currently does patients in whom vasopressor-induced hypertension may
not approve xenon for clinical use other than in approved increase ischemia rather than help it50,51 also is described.
research protocols. Recently, 80% xenon was reported to be an Because Xe-CT studies can be repeated in quick succession,
ideal anesthetic agent because even after hours of inhalation patients also may be examined to assess their response to
it was not associated with any physiologic side effects and had vasopressor agents.
rapid awakening.31 Other studies have demonstrated that Quantitative CBF data also may be used to assess the effi-
xenon can be neuroprotective because it can block N-methyl- cacy of specific treatments for vasospasm including intra-
d-aspartate (NMDA) receptors.32 Carlson et al. examined the arterial infusion of papaverine, angioplasty or even intra-aortic
safety of 28% xenon in 2003 Xe-CT examinations, including balloon counter pulsation.52 In these studies it was found that
1486 non­ventilated patients.33 Respiratory suppression (>20 papaverine, although it reduced vasospasm, increased CBF
seconds) occurred in 39 (1.9%) studies; all resolved spontane- in less than half the patients.53,54 By contrast Xe-CT studies
ously. No adverse event that resulted in a persistent neurologic have shown that angioplasty (mechanical rather than phar­
change or other sequelae were observed. macologic) improves CBF55 and can reverse ischemia.56 This
278 Section IV—Radiology

information can be used to guide decisions about the risks and establish the state of autoregulation and the impact of treat-
benefits of angioplasty.57 ments on brain physiology and validate the information pro-
vided by more local monitors such as an ICP or brain oxygen
monitor.69 Consistent with this, in a small pilot study it appears
Carotid Endarterectomy or Arteriovenous that there is a reasonable correlation between xenon CBF
Malformation Surgery and oxygen saturation measured using an INVOS cerebral
Vascular hyperemia that can cause intracerebral hemorrhage oximeter (Somanetics; Troy, MI).70
after a critically stenotic vessel is opened during carotid end- Measurements with Xe-CT have provided insight into the
arterectomy (CEA) is a rare but well-known complication. pathophysiology after TBI and may detect ischemia not oth-
Xe-CT studies with or without an acetazolamide challenge erwise seen on CT, and which may not be associated with
may help identify and guide management of these patients.58,59 other abnormalities; this may help direct early medical and
Following removal of arteriovenous malformations (AVMs) surgical management.71,72 For example, the CBF patterns
various patterns of CBF redistribution may occur, including around traumatic intracerebral hemorrhage or contusions can
increased regional CBF, or more commonly, decreased CBF, be used to determine who requires surgery73,74 or guide blood
which can be responsible for a neurologic deficit.60 A quantita- pressure and CO2 levels to ensure adequate CBF to the peri-
tive CBF study may assist with management of postoperative contusional tissue.75,76 However, it does not appear that xenon
blood pressure in a patient with new neurologic deficit not CBF studies can be used to predict which contusions will
explained by other imaging. enlarge.77 Xe-CT studies also may help with outcome predic-
tion in adults and children78 and in particular when there is
reduced brainstem CBF, outcome usually is poor.79 Robertson
Vascular Occlusion et al. also have used xenon CBF to examine how genetic varia-
Xe-CT has been used frequently to evaluate hemodynamic tion in endothelial nitric oxide synthase (NOS3), that plays a
sufficiency in patients with occlusive carotid vascular disease role in CBF regulation, contributes to the brain’s response to
or who may be undergoing surgery in which the carotid is at injury.80
risk or needs to be sacrificed and so predict the need for a In children the CBF response after TBI differs from adults.
bypass or risk of stroke.61 In addition, patients with moya­ Hyperemia with increased ICP can occur, and in these patients
moya disease can be studied to examine direct or indirect quantitative CBF data will show no need to increase CPP.78 On
revascularization techniques and the status of collateral the other hand, the CBF data can help identify tissue at risk
perfusion.62-64 To do this the Xe-CT study is performed before for ischemia. This may be important because an ICP monitor
and after acetazolamide administration or as part of a balloon and plain head CT will not differentiate the cause of the ICP
test occlusion. Reduced CBF values less than 30 mL/100 g/ increase.
min, an absolute CBF decrease, and significant asymmetry in
middle cerebral artery CBF during balloon test occlusion
suggest a high risk of stroke during carotid occlusion.21 Lack Ischemic Stroke
of an appropriate vasodilatory response to acetazolamide Acute stroke is common, and apart from intravenous tissue
indicates a vasculature that has already maximally dilated; this plasminogen activator (tPA) given within 3 to 4.5 hours of
decreased hemodynamic reserve is very predictive of subse- symptom onset, there are few specific treatment options. Data
quent infarct65 in patients with atherosclerotic occlusive gained from Xe-CT and other CT modalities (e.g., CT angi-
disease17 or those about to undergo surgery. The quantitative ography) may help clarify patient physiology and identify a
values obtained with Xe-CT appear to be better able to guide group of patients who have potentially salvageable tissue.81-85
management in these patients than the relative changes seen This is important because there is a great variability in the
with modalities such as single photon emission computed proportion of the ischemic core compared with salvageable
tomography (SPECT).21 For example, Chaer et al. found that penumbra that depends on individual vascular variability and
among 179 patients with carotid stenosis more than 50% who collateral flow (Fig. 27.12).86 Recanalization cannot re­cover
underwent an acetazolamide-activated xenon CBF study, infracted core, but may recover penumbral tissue (i.e., it is
those older than 70 years of age had reduced cerebrovascular possible to predict patients who will respond based on the
reserve, which may explain the increased risk of stroke in amount of salvageable tissue rather than time frame alone).87
older adult patients.66 In addition, the Xe-CT identified abnormality can be useful
in outcome prediction.86 Furthermore, if a patient has a neu-
rologic deficit but a normal CBF study, he or she will be likely
Traumatic Brain Injury to recover normal function without intervention. By contrast,
Traumatic brain injury (TBI) management in the NCCU is a patient with a very large volume of already infracted tissue
centered on the prevention of secondary cerebral insults, and (large core with CBF values less than 9 mL/100 g/min) is at
in part this depends on control of adequate CPP to maintain risk for edema and herniation.88 The progression to life-
CBF. Current guidelines recommend a CPP between 50 and threatening swelling and herniation can be accurately pre-
70 mm Hg; in this range Xe-CT studies show that CBF is fairly dicted (e.g., early Xe-CT can predict patients at risk for
constant, but that even lower CPP (50 mm Hg) can provide herniation after stroke and identify patients who may benefit
adequate CBF in some patients.67 Autoregulation and CO2 from decompressive craniectomy before herniation occurs).84,89
reactivity68 frequently are abnormal in patients with severe In addition, Xe-CT has been used to define supratentorial
TBI and so a one-to-one relationship between CPP and CBF hypoperfusion after brainstem strokes, particularly larger
is not always present. TBI pathophysiology is complex and pontine infarcts and guide therapy to prevent supratentorial
heterogeneous, and in these patients Xe-CT studies can help infarction.90
Section IV—Radiology 279

100%

90%

80%

70%
% of MCA territory
60%

50%

40%

30%

20%

10%

0%
28 35 33 15 9 6 12 1 3 22 23 24 11 7 34 17 16 13 26 21 25 4 19 30 14 36 2 8 18 20 5 29 27 10 32 31

Patient #
Core Penumbra NC/NP

Fig. 27.12  A series of patients with ischemic middle cerebral artery (MCA) stroke, showing the variability in infarcted core. (From Jovin TG, Yonas H,
Gebel JM, et al: The cortical ischemic core and not the consistently present penumbra is a determinant of clinical outcome in acute middle cerebral artery occlusion. Stroke
2003;34:2426–33.)

Intracerebral Hemorrhage increased CBF.103 These Xe-CT data may help guide possible
Intracerebral hemorrhage (ICH), although less common than intervention.
ischemic stroke, is devastating but difficult to treat. In particu-
lar the role of surgery is unclear.91,92 Xe-CT studies may offer
a method to select patients who may benefit from surgical Brain Death
evacuation of an ICH by demonstration of a persistent pen- Brain death is discussed in Chapter 13. It is a clinical diagnosis
umbra around the ICH.93,94 but confirmatory tests (e.g., absence of flow on catheter angi-
ography or brain scintigraphy imaging) can be useful in select
patients. Xe-CT studies suggest that global CBF less than
Tumor 5 mL/100 g/min is associated with brain death.104,105
There are several potential applications for Xe-CT in the eval-
uation and treatment of intracranial tumors. First, blood flow
studies have identified CBF gradients that extend peripherally Other Conditions
from a tumor and heterogeneity of blood flow within various Xe-CT data can be used to assess patient physiology in a
tumor types. The CBF levels adjacent to the tumor, including variety of other conditions such as in ventilation response to
both malignant and benign tumors, may even be at ischemic bronchodilators in asthma and chronic obstructive pulmo-
levels.95-97 In some patients dexamethasone may improve nary disease (COPD),106 to provide quantitative information
regional CBF around malignant glial tumors, but in others it on pulmonary ventilation and function,107-110 as an objective
may decrease. Surgical removal of the tumor on the other method to assess the state of the liver in chronic liver disease,111
hand reliably improves CBF.98 Second, Xe-CT CBF studies can and to assess patient physiology in carbon monoxide poison-
be used to help define tumor types or degree of malig- ing.112 For example, following cardiac arrest, return of reduced
nancy.99,100 However, these studies may only be required in CBF to normal levels during the first 48 hours is associated
patients who are not able to undergo magnetic resonance with a good prognosis.113
imaging (MRI) studies because advanced MRI techniques can
be used to define tumor pathology and anatomy.101
Conclusion
Xe-CT CBF studies have a long history of use in patients with
Epilepsy cerebrovascular disease because the studies provide quantita-
During interictal periods CBF has been shown to increase tive CBF data and can be repeated easily to assess hemody-
then return to normal in the postictal period.102 In status namic reserve or treatment response. These physiologic data
epilepticus CBF can decrease and reach potentially ischemic can be used to guide management decisions in the NCCU,
levels. This is followed by a period of several weeks of where management often is focused on efforts to improve CBF
280 Section IV—Radiology

to prevent secondary brain injuries due to persistent or late- 21. Witt JP, Yonas H, Jungreis C. Cerebral blood flow response pattern during
balloon test occlusion of the internal carotid artery. AJNR Am J Neuroradiol
onset ischemia. These studies may be best achieved with the
1994;15(5):847–56.
combination of a portable CT scanner and Xe-CT CBF analy- 22. Yonas H, Kromer H, Jungreis C. Compromised vascular reserves does predict
sis. However, xenon currently needs to be used in research subgroups with carotid occlusion and an increased stroke risk. J Stroke
protocols in the United States because it does not have FDA Cerebrovasc Dis 1997;6:458.
approval for clinical use. Recent safety analysis studies in 2003 23. Yonas H, Smith HA, Durham SR, et al. Increased stroke risk predicted by
compromised cerebral blood flow reactivity. J Neurosurg 1993;79:483–9.
scans using 28% xenon suggest that Xe-CT should be made 24. Gunnarsson T, Theodorsson A, Karlsson P, et al. Mobile computerized
widely available because it has a very low risk of adverse events tomography scanning in the neurosurgery intensive care unit: increase in
and no permanent morbidity while it provides useful clinical patient safety and reduction of staff workload. J Neurosurg 2000;93:432–6.
information.33 25. Hillman J, Sturnegk P, Yonas H, et al. Bedside monitoring of CBF with
xenon-CT and a mobile scanner: a novel method in neurointensive care. Br J
Neurosurg 2005;19:395–401.
References 26. Sturnegk P, Mellergard P, Yonas H, et al. Potential use of quantitative bedside
CBF monitoring (Xe-CT) for decision making in neurosurgical intensive
1. Drayer BP, Gur D, Yonas H, et al. Abnormality of the xenon brain:blood care. Br J Neurosurg 2007;21:332–9.
partition coefficient and blood flow in cerebral infarction: an in vivo 27. Schroder ML, Muizelaar JP. Monitoring of regional cerebral blood flow
assessment using transmission computed tomography. Radiology 1980;135: (CBF) in acute head injury by thermal diffusion. Acta Neurochir Suppl
349–54. (Wien) 1993;59:47–9.
2. Gur D, Good WF, Wolfson SK, Jr, et al. In vivo mapping of local cerebral 28. Carlson AP, Yonas H. Portable head computed tomography scanner-
blood flow by xenon-enhanced computed tomography. Science 1982;215: technology and applications: experience with 3421 scans. J Neuroimaging.
1267–8. 2011; Jun 23. Epub ahead of print.
3. Gur D, Yonas H, Jackson DL, et al. Simultaneous measurements of cerebral 29. Peace K, Maloney E, Frangos S, et al. The use of a portable head CT scanner
blood flow by the xenon/CT method and the microsphere method. in the ICU. J Neurosci Nurs 2010;42(2):109–16.
A comparison. Invest Radiol 1985;20:672–7. 30. Peace K, Maloney E, Frangos S, et al. Portable head CT scan and its effect on
4. Wolfson SK, Jr, Clark J, Greenberg JH, et al. Xenon-enhanced computed intracranial pressure, cerebral perfusion pressure and brain oxygen.
tomography compared with [14C]iodoantipyrine for normal and low J Neurosurgery 2011;114:1479–84.
cerebral blood flow states in baboons. Stroke 1990;21:751–7. 31. Hecker K, Baumert JH, Horn N, et al. Xenon, a modern anaesthesia gas.
5. Tone O, Ito U, Tomita H, et al. Correlation between cerebral blood flow Minerva Anestesiol 2004;70:255–60.
values obtained by Xe-CT and Kety-Schmidt (N2O) methods. Acta Neurol 32. Dickinson R, Peterson BK, Banks P, et al. Competitive inhibition at the
Scand Suppl 1996;166:18–21. glycine site of the N-methyl-D-aspartate receptor by the anesthetics xenon
6. Nariai T. Comparison of measurement between Xe/CT CBF and PET in and isoflurane: evidence from molecular modeling and electrophysiology.
cerebrovascular disease and brain tumor. Acta Neurol Scand Suppl 1996; Anesthesiology 2007;107:756–67.
166:10–2. 33. Carlson AP, Brown AM, Zager E, et al. Xenon-enhanced cerebral blood flow
7. Bergholt B, Ostergaard L, von Oettingen G, et al. Cerebral blood flow in at 28% xenon provides uniquely safe access to quantitative, clinically useful
volunteers measured by PET and Xe CT/CBF: a comparison. Keio J Med cerebral blood flow information: a multicenter study. AJNR Am J
2000;49(Suppl 1):A55–7. Neuroradiol 2011;32(7):1315–20. Epub 2011 Jun 23.
8. Valadka AB, Hlatky R, Furuya Y, et al. Brain tissue PO2: correlation with 34. The Brain Trauma Foundation. The American Association of Neurological
cerebral blood flow. Acta Neurochir Suppl 2002;81:299–301. Surgeons. The Joint Section on Neurotrauma and Critical Care. Initial
9. Matsuda M, Lee H, Kuribayashi K, et al. Comparative study of regional management. J Neurotrauma 2000;17:463–9.
cerebral blood flow values measured by Xe CT and Xe SPECT. Acta Neurol 35. Adelson PD, Bratton SL, Carney NA, et al. Guidelines for the acute medical
Scand Suppl 1996;166:13–6. management of severe traumatic brain injury in infants, children, and
10. Gur D, Yonas H, Good WF. Local cerebral blood flow by xenon-enhanced adolescents. Chapter 1: introduction. Pediatr Crit Care Med 2003;4:S2–4.
CT: current status, potential improvements, and future directions. 36. Haley EC, Jr, Kassell NF, Torner JC. The International Cooperative Study on
Cerebrovasc Brain Metab Rev 1989;1:68–86. the Timing of Aneurysm Surgery. The North American experience. Stroke
11. Johnson DW, Stringer WA, Marks MP, et al. Stable xenon CT cerebral blood 1992;23:205–14.
flow imaging: rationale for and role in clinical decision making. AJNR Am J 37. Joseph M, Ziadi S, Nates J, et al. Increases in cardiac output can reverse flow
Neuroradiol 1991;12:201–13. deficits from vasospasm independent of blood pressure: a study using xenon
12. Yonas H. Cerebral blood measurements in vasospasm. Neurosurg Clin N Am computed tomographic measurement of cerebral blood flow. Neurosurgery
1990;1:307–18. 2003;53:1044–51; discussion 51–2.
13. Yonas H, Darby JM, Marks EC, et al. CBF measured by Xe-CT: approach to 38. Fisher CM, Kistler JP, Davis JM. Relation of cerebral vasospasm to
analysis and normal values. J Cereb Blood Flow Metab 1991;11:716–25. subarachnoid hemorrhage visualized by computerized tomographic
14. Yonas H, Sekhar L, Johnson DW, et al. Determination of irreversible scanning. Neurosurgery 1980;6:1–9.
ischemia by xenon-enhanced computed tomographic monitoring of cerebral 39. Lennihan L, Mayer SA, Fink ME, et al. Effect of hypervolemic therapy on
blood flow in patients with symptomatic vasospasm. Neurosurgery 1989; cerebral blood flow after subarachnoid hemorrhage: a randomized
24:368–72. controlled trial. Stroke 2000;31:383–91.
15. Gur AY, Bova I, Bornstein NM. Is impaired cerebral vasomotor reactivity a 40. Tone O, Tomita H, Tamaki M, et al. Correlation between cardiac output and
predictive factor of stroke in asymptomatic patients? Stroke 1996;27: cerebral blood flow following subarachnoid hemorrhage. Keio J Med
2188–90. 2000;49(Suppl 1):A151–3.
16. Horowitz M, Yonas H, Albright AL. Evaluation of cerebral blood flow and 41. Tanaka A, Kumate S, Nakayama Y, et al. Postoperative subarachnoid clots
hemodynamic reserve in symptomatic moyamoya disease using stable and the pattern of cerebral ischemia associated with symptomatic
xenon-CT blood flow. Surg Neurol 1995;44:251–61; discussion 62. vasospasm. Surg Neurol 1998;49:164–8; discussion 8–9.
17. Nemoto EM, Yonas H, Kuwabara H, et al. Identification of hemodynamic 42. Compagnone C, Tagliaferri F, Fainardi E, et al. Diagnostic impact of the
compromise by cerebrovascular reserve and oxygen extraction fraction in spectrum of ischemic cerebral blood flow thresholds in sedated
occlusive vascular disease. J Cereb Blood Flow Metab 2004;24:1081–9. subarachnoid hemorrhage patients. Acta Neurochir Suppl 2006;96:53–6.
18. Pindzola RR, Sashin D, Nemoto EM, et al. Identifying regions of 43. Schubert GA, Seiz M, Hegewald AA, et al. Acute hypoperfusion immediately
compromised hemodynamics in symptomatic carotid occlusion by after subarachnoid hemorrhage: a xenon contrast-enhanced CT study.
cerebrovascular reactivity and oxygen extraction fraction. Neurol Res 2006; J Neurotrauma 2009;26(12):2225–31.
28:149–54. 44. Clyde BL, Resnick DK, Yonas H, et al. The relationship of blood velocity as
19. Tarr RW, Johnson DW, Rutigliano M, et al. Use of acetazolamide-challenge measured by transcranial Doppler ultrasonography to cerebral blood flow as
xenon CT in the assessment of cerebral blood flow dynamics in patients determined by stable xenon computed tomographic studies after aneurysmal
with arteriovenous malformations. AJNR Am J Neuroradiol 1990;11: subarachnoid hemorrhage. Neurosurgery 1996;38:896–904.
441–8. 45. Chieregato A, Sabia G, Tanfani A, et al. Xenon-CT and transcranial Doppler
20. Webster MW, Makaroun MS, Steed DL, et al. Compromised cerebral blood in poor-grade or complicated aneurysmatic subarachnoid hemorrhage
flow reactivity is a predictor of stroke in patients with symptomatic carotid patients undergoing aggressive management of intracranial hypertension.
artery occlusive disease. J Vasc Surg 1995;21:338–44; discussion 44–5. Intensive Care Med 2006;32:1143–50.
Section IV—Radiology 281

46. Rothoerl RD, Woertgen C, Brawanski A. Hyperemia following aneurysmal 49. Al-Rawi PG, Tseng MY, Richards HK, et al. Hypertonic saline in patients
subarachnoid hemorrhage: incidence, diagnosis, clinical features, and with poor-grade subarachnoid hemorrhage improves cerebral blood flow,
outcome. Intensive Care Med 2004;30:1298–302. brain tissue oxygen, and pH. Stroke 2010;41(1):122–8. Epub 2009 Nov 12.
47. Firlik KS, Firlik AD, Yonas H. Xenon-enhanced computed tomography in the 50. Darby JM, Yonas H, Marks EC, et al. Acute cerebral blood flow response to
management of cerebral vasospasm following aneurysmal subarachnoid dopamine-induced hypertension after subarachnoid hemorrhage.
hemorrhage. Keio J Med 2000;49(Suppl 1):A148–50. J Neurosurg 1994;80:857–64.
48. Tseng MY, Al-Rawi PG, Pickard JD, et al. Effect of hypertonic saline on
cerebral blood flow in poor-grade patients with subarachnoid hemorrhage. A complete list of references for this chapter can be found online at
Stroke 2003;34:1389–96. www.expertconsult.com.
Section IV—Radiology 281.e1

References 26. Sturnegk P, Mellergard P, Yonas H, et al. Potential use of quantitative


bedside CBF monitoring (Xe-CT) for decision making in neurosurgical
1. Drayer BP, Gur D, Yonas H, et al. Abnormality of the xenon brain:blood intensive care. Br J Neurosurg 2007;21:332–9.
partition coefficient and blood flow in cerebral infarction: an in vivo 27. Schroder ML, Muizelaar JP. Monitoring of regional cerebral blood flow
assessment using transmission computed tomography. Radiology 1980; (CBF) in acute head injury by thermal diffusion. Acta Neurochir Suppl
135:349–54. (Wien) 1993;59:47–9.
2. Gur D, Good WF, Wolfson SK, Jr, et al. In vivo mapping of local cerebral 28. Carlson AP, Yonas H. Portable head computed tomography scanner-
blood flow by xenon-enhanced computed tomography. Science 1982;215: technology and applications: experience with 3421 scans. J Neuroimaging
1267–8. 2011; Jun 23. Epub ahead of print.
3. Gur D, Yonas H, Jackson DL, et al. Simultaneous measurements of cerebral 29. Peace K, Maloney E, Frangos S, et al. The use of a portable head CT
blood flow by the xenon/CT method and the microsphere method. A scanner in the ICU. J Neurosci Nurs 2010;42(2):109–16.
comparison. Invest Radiol 1985;20:672–7. 30. Peace K, Maloney E, Frangos S, et al. Portable head CT scan and its effect
4. Wolfson SK, Jr, Clark J, Greenberg JH, et al. Xenon-enhanced computed on intracranial pressure, cerebral perfusion pressure and brain oxygen.
tomography compared with [14C]iodoantipyrine for normal and low J Neurosurgery 2011;114:1479–84.
cerebral blood flow states in baboons. Stroke 1990;21:751–7. 31. Hecker K, Baumert JH, Horn N, et al. Xenon, a modern anaesthesia gas.
5. Tone O, Ito U, Tomita H, et al. Correlation between cerebral blood flow Minerva Anestesiol 2004;70:255–60.
values obtained by Xe-CT and Kety-Schmidt (N2O) methods. Acta Neurol 32. Dickinson R, Peterson BK, Banks P, et al. Competitive inhibition at the
Scand Suppl 1996;166:18–21. glycine site of the N-methyl-D-aspartate receptor by the anesthetics xenon
6. Nariai T. Comparison of measurement between Xe/CT CBF and PET in and isoflurane: evidence from molecular modeling and electrophysiology.
cerebrovascular disease and brain tumor. Acta Neurol Scand Suppl 1996; Anesthesiology 2007;107:756–67.
166:10–2. 33. Carlson AP, Brown AM, Zager E, et al. Xenon-enhanced cerebral blood flow
7. Bergholt B, Ostergaard L, von Oettingen G, et al. Cerebral blood flow in at 28% xenon provides uniquely safe access to quantitative, clinically useful
volunteers measured by PET and Xe CT/CBF: a comparison. Keio J Med cerebral blood flow information: a multicenter study. AJNR Am J
2000;49(Suppl 1):A55–7. Neuroradiol 2011;32(7):1315–20. Epub 2011 Jun 23.
8. Valadka AB, Hlatky R, Furuya Y, et al. Brain tissue PO2: correlation with 34. The Brain Trauma Foundation. The American Association of Neurological
cerebral blood flow. Acta Neurochir Suppl 2002;81:299–301. Surgeons. The Joint Section on Neurotrauma and Critical Care. Initial
9. Matsuda M, Lee H, Kuribayashi K, et al. Comparative study of regional management. J Neurotrauma 2000;17:463–9.
cerebral blood flow values measured by Xe CT and Xe SPECT. Acta Neurol 35. Adelson PD, Bratton SL, Carney NA, et al. Guidelines for the acute
Scand Suppl 1996;166:13–6. medical management of severe traumatic brain injury in infants,
10. Gur D, Yonas H, Good WF. Local cerebral blood flow by xenon-enhanced children, and adolescents. Chapter 1: introduction. Pediatr Crit Care Med
CT: current status, potential improvements, and future directions. 2003;4:S2–4.
Cerebrovasc Brain Metab Rev 1989;1:68–86. 36. Haley EC, Jr, Kassell NF, Torner JC. The International Cooperative Study on
11. Johnson DW, Stringer WA, Marks MP, et al. Stable xenon CT cerebral blood the Timing of Aneurysm Surgery. The North American experience. Stroke
flow imaging: rationale for and role in clinical decision making. AJNR Am J 1992;23:205–14.
Neuroradiol 1991;12:201–13. 37. Joseph M, Ziadi S, Nates J, et al. Increases in cardiac output can reverse
12. Yonas H. Cerebral blood measurements in vasospasm. Neurosurg Clin N flow deficits from vasospasm independent of blood pressure: a study using
Am 1990;1:307–18. xenon computed tomographic measurement of cerebral blood flow.
13. Yonas H, Darby JM, Marks EC, et al. CBF measured by Xe-CT: approach to Neurosurgery 2003;53:1044–51; discussion 51–2.
analysis and normal values. J Cereb Blood Flow Metab 1991;11:716–25. 38. Fisher CM, Kistler JP, Davis JM. Relation of cerebral vasospasm to
14. Yonas H, Sekhar L, Johnson DW, et al. Determination of irreversible subarachnoid hemorrhage visualized by computerized tomographic
ischemia by xenon-enhanced computed tomographic monitoring of scanning. Neurosurgery 1980;6:1–9.
cerebral blood flow in patients with symptomatic vasospasm. Neurosurgery 39. Lennihan L, Mayer SA, Fink ME, et al. Effect of hypervolemic therapy on
1989;24:368–72. cerebral blood flow after subarachnoid hemorrhage: a randomized
15. Gur AY, Bova I, Bornstein NM. Is impaired cerebral vasomotor reactivity a controlled trial. Stroke 2000;31:383–91.
predictive factor of stroke in asymptomatic patients? Stroke 1996;27: 40. Tone O, Tomita H, Tamaki M, et al. Correlation between cardiac output
2188–90. and cerebral blood flow following subarachnoid hemorrhage. Keio J Med
16. Horowitz M, Yonas H, Albright AL. Evaluation of cerebral blood flow and 2000;49(Suppl 1):A151–3.
hemodynamic reserve in symptomatic moyamoya disease using stable 41. Tanaka A, Kumate S, Nakayama Y, et al. Postoperative subarachnoid clots
xenon-CT blood flow. Surg Neurol 1995;44:251–61; discussion 62. and the pattern of cerebral ischemia associated with symptomatic
17. Nemoto EM, Yonas H, Kuwabara H, et al. Identification of hemodynamic vasospasm. Surg Neurol 1998;49:164–8; discussion 8–9.
compromise by cerebrovascular reserve and oxygen extraction fraction in 42. Compagnone C, Tagliaferri F, Fainardi E, et al. Diagnostic impact of the
occlusive vascular disease. J Cereb Blood Flow Metab 2004;24:1081–9. spectrum of ischemic cerebral blood flow thresholds in sedated
18. Pindzola RR, Sashin D, Nemoto EM, et al. Identifying regions of subarachnoid hemorrhage patients. Acta Neurochir Suppl 2006;96:53–6.
compromised hemodynamics in symptomatic carotid occlusion by 43. Schubert GA, Seiz M, Hegewald AA, et al. Acute hypoperfusion
cerebrovascular reactivity and oxygen extraction fraction. Neurol Res 2006; immediately after subarachnoid hemorrhage: a xenon contrast-enhanced
28:149–54. CT study. J Neurotrauma 2009;26(12):2225–31.
19. Tarr RW, Johnson DW, Rutigliano M, et al. Use of acetazolamide-challenge 44. Clyde BL, Resnick DK, Yonas H, et al. The relationship of blood velocity as
xenon CT in the assessment of cerebral blood flow dynamics in patients measured by transcranial Doppler ultrasonography to cerebral blood flow
with arteriovenous malformations. AJNR Am J Neuroradiol 1990;11:441–8. as determined by stable xenon computed tomographic studies after
20. Webster MW, Makaroun MS, Steed DL, et al. Compromised cerebral blood aneurysmal subarachnoid hemorrhage. Neurosurgery 1996;38:896–904.
flow reactivity is a predictor of stroke in patients with symptomatic carotid 45. Chieregato A, Sabia G, Tanfani A, et al. Xenon-CT and transcranial Doppler
artery occlusive disease. J Vasc Surg 1995;21:338–44; discussion 44–5. in poor-grade or complicated aneurysmatic subarachnoid hemorrhage
21. Witt JP, Yonas H, Jungreis C. Cerebral blood flow response pattern during patients undergoing aggressive management of intracranial hypertension.
balloon test occlusion of the internal carotid artery. AJNR Am J Intensive Care Med 2006;32:1143–50.
Neuroradiol 1994;15(5):847–56. 46. Rothoerl RD, Woertgen C, Brawanski A. Hyperemia following aneurysmal
22. Yonas H, Kromer H, Jungreis C. Compromised vascular reserves does subarachnoid hemorrhage: incidence, diagnosis, clinical features, and
predict subgroups with carotid occlusion and an increased stroke risk. outcome. Intensive Care Med 2004;30:1298–302.
J Stroke Cerebrovasc Dis 1997;6:458. 47. Firlik KS, Firlik AD, Yonas H. Xenon-enhanced computed tomography in
23. Yonas H, Smith HA, Durham SR, et al. Increased stroke risk predicted by the management of cerebral vasospasm following aneurysmal subarachnoid
compromised cerebral blood flow reactivity. J Neurosurg 1993;79:483–9. hemorrhage. Keio J Med 2000;49(Suppl 1):A148–50.
24. Gunnarsson T, Theodorsson A, Karlsson P, et al. Mobile computerized 48. Tseng MY, Al-Rawi PG, Pickard JD, et al. Effect of hypertonic saline on
tomography scanning in the neurosurgery intensive care unit: increase in cerebral blood flow in poor-grade patients with subarachnoid hemorrhage.
patient safety and reduction of staff workload. J Neurosurg 2000;93:432–6. Stroke 2003;34:1389–96.
25. Hillman J, Sturnegk P, Yonas H, et al. Bedside monitoring of CBF with 49. Al-Rawi PG, Tseng MY, Richards HK, et al. Hypertonic saline in patients
xenon-CT and a mobile scanner: a novel method in neurointensive care. Br with poor-grade subarachnoid hemorrhage improves cerebral blood flow,
J Neurosurg 2005;19:395–401. brain tissue oxygen, and pH. Stroke 2010;41(1):122–8. Epub 2009 Nov 12.
281.e2 Section IV—Radiology

50. Darby JM, Yonas H, Marks EC, et al. Acute cerebral blood flow response to 74. von Oettingen G, Bergholt B, Gyldensted C, et al. Blood flow and ischemia
dopamine-induced hypertension after subarachnoid hemorrhage. within traumatic cerebral contusions. Neurosurgery 2002;50:781–8;
J Neurosurg 1994;80:857–64. discussion 8–90.
51. Ract C, Vigue B. Comparison of the cerebral effects of dopamine and 75. Chieregato A, Fainardi E, Tanfani A, et al. Induced acute arterial
norepinephrine in severely head-injured patients. Intensive Care Med hypertension and regional cerebral flow in intracontusional low density
2001;27:101–6. area. Acta Neurochir Suppl 2003;86:361–5.
52. Nussbaum ES, Sebring LA, Ganz WF, et al. Intra-aortic balloon 76. McLaughlin MR, Marion DW. Cerebral blood flow and vasoresponsivity
counterpulsation augments cerebral blood flow in the patient with cerebral within and around cerebral contusions. J Neurosurg 1996;85:871–6.
vasospasm: a xenon-enhanced computed tomography study. Neurosurgery 77. Depreitere B, Aviv R, Symons S, et al. Study of perfusion in and around
1998;42:206–13; discussion 13–4. cerebral contusions by means of computed tomography. Acta Neurochir
53. Firlik KS, Kaufmann AM, Firlik AD, et al. Intra-arterial papaverine for the Suppl 2008;102:259–62.
treatment of cerebral vasospasm following aneurysmal subarachnoid 78. Adelson PD, Clyde B, Kochanek PM, et al. Cerebrovascular response in
hemorrhage. Surg Neurol 1999;51:66–74. infants and young children following severe traumatic brain injury:
54. Clyde BL, Firlik AD, Kaufmann AM, et al. Paradoxical aggravation of a preliminary report. Pediatr Neurosurg 1997;26:200–7.
vasospasm with papaverine infusion following aneurysmal subarachnoid 79. Ritter AM, Muizelaar JP, Barnes T, et al. Brain stem blood flow, pupillary
hemorrhage. Case report. J Neurosurg 1996;84:690–5. response, and outcome in patients with severe head injuries. Neurosurgery
55. Murai Y, Kominami S, Kobayashi S, et al. The long-term effects of 1999;44:941–8.
transluminal balloon angioplasty for vasospasms after subarachnoid 80. Robertson CS, Gopinath SP, Valadka AB, et al. Variants of the endothelial
hemorrhage: analyses of cerebral blood flow and reactivity. Surg Neurol nitric oxide gene and cerebral blood flow after severe traumatic brain
2005;64:122–6; discussion 7. injury. J Neurotrauma 2011;28(5):727–37. Epub 2011 May 4.
56. Firlik AD, Kaufmann AM, Jungreis CA, et al. Effect of transluminal 81. Kilpatrick MM, Yonas H, Goldstein S, et al. CT-based assessment of acute
angioplasty on cerebral blood flow in the management of symptomatic stroke: CT, CT angiography, and xenon-enhanced CT cerebral blood flow.
vasospasm following aneurysmal subarachnoid hemorrhage. J Neurosurg Stroke 2001;32:2543–9.
1997;86:830–9. 82. Meyer JS, Rauch GM. Why emergency XeCT-CBF should become routine
57. Linskey ME, Horton JA, Rao GR, et al. Fatal rupture of the intracranial in acute ischemic stroke before thrombolytic therapy. Keio J Med 2000;
carotid artery during transluminal angioplasty for vasospasm induced by 49(Suppl 1):A25–8.
subarachnoid hemorrhage. Case report. J Neurosurg 1991;74:985–90. 83. Firlik AD, Kaufmann AM, Wechsler LR, et al. Quantitative cerebral blood
58. Penn AA, Schomer DF, Steinberg GK. Imaging studies of cerebral flow determinations in acute ischemic stroke: relationship to computed
hyperperfusion after carotid endarterectomy. Case report. J Neurosurg tomography and angiography. Stroke 1997;28:2208–13.
1995;83:133–7. 84. Firlik AD, Yonas H, Kaufmann AM, et al. Relationship between cerebral
59. Yamane K, Shima T, Nishida M, et al. Changes in cerebral blood flow after blood flow and the development of swelling and life-threatening herniation
carotid endarterectomy. Keio J Med 2000;49(Suppl 1):A80–2. in acute ischemic stroke. J Neurosurg 1998;89:243–9.
60. Van Roost D, Schramm J, Solymosi L, et al. Presence and removal of 85. Hughes RL, Yonas H, Gur D, et al. Cerebral blood flow determination
arteriovenous malformation: impact of regional cerebral blood flow, as within the first 8 hours of cerebral infarction using stable xenon-enhanced
assessed with xenon/CT. Acta Neurol Scand Suppl 1996;166:136–8. computed tomography. Stroke 1989;20:754–60.
61. Schubert GA, Weinmann C, Seiz M, et al. Cerebrovascular insufficiency as 86. Jovin TG, Yonas H, Gebel JM, et al. The cortical ischemic core and not the
the criterion for revascularization procedures in selected patients: consistently present penumbra is a determinant of clinical outcome in
a correlation study of xenon contrast-enhanced CT and PWI. Neurosurg acute middle cerebral artery occlusion. Stroke 2003;34:2426–33.
Rev 2009;32(1):29–35; discussion 35–6. Epub 2008 Sep 13. 87. Gupta R, Jovin TG, Yonas H. Xenon CT cerebral blood flow in acute stroke.
62. Czabanka M, Peña-Tapia P, Scharf J, et al. Characterization of direct and Neuroimaging Clin N Am 2005;15:531–42, x.
indirect cerebral revascularization for the treatment of European patients 88. Firlik AD, Yonas H. The utility of XeCT cerebral blood flow in the
with moyamoya disease. Cerebrovasc Dis 2011;32(4):361–9. Epub 2011 management of acute stroke. Keio J Med 2000;49(Suppl 1):A129–30.
Sep 15. 89. Kalia KK, Yonas H. An aggressive approach to massive middle cerebral
63. Zaharchuk G, Do HM, Marks MP, et al. Arterial spin-labeling MRI can artery infarction. Arch Neurol 1993;50:1293–7.
identify the presence and intensity of collateral perfusion in patients with 90. Miyazawa N, Uchida M, Fukamachi A, et al. Xenon contrast-enhanced CT
moyamoya disease. Stroke 2011;42(9):2485–91. Epub 2011 Jul 28. imaging of supratentorial hypoperfusion in patients with brain stem
64. Huang AP, Liu HM, Lai DM, et al. Clinical significance of posterior infarction. AJNR Am J Neuroradiol 1999;20:1858–62.
circulation changes after revascularization in patients with moyamoya 91. Mendelow AD, Gregson BA, Fernandes HM, et al. Early surgery versus
disease. Cerebrovasc Dis 2009;28(3):247–57. Epub 2009 Jul 14. initial conservative treatment in patients with spontaneous supratentorial
65. Field M, Jungreis CA, Chengelis N, et al. Symptomatic cavernous sinus intracerebral haematomas in the International Surgical Trial in
aneurysms: management and outcome after carotid occlusion and selective Intracerebral Haemorrhage (STICH): a randomised trial. Lancet
cerebral revascularization. AJNR Am J Neuroradiol 2003;24:1200–7. 2005;365:387–97.
66. Chaer RA, Shen J, Rao A, et al. Cerebral reserve is decreased in elderly 92. Bhattathiri PS, Gregson B, Prasad KS, et al. Intraventricular hemorrhage
patients with carotid stenosis. J Vasc Surg 2010;52(3):569–74; discussion and hydrocephalus after spontaneous intracerebral hemorrhage: results
574–5. from the STICH trial. Acta Neurochir Suppl 2006;96:65–8.
67. Chieregato A, Tanfani A, Compagnone C, et al. Global cerebral blood flow 93. Kitahara T, Yamashita T, Kashiwagi S, et al. Hemodynamics of hypertensive
and CPP after severe head injury: a xenon-CT study. Intensive Care Med putaminal hemorrhage evaluated by xenon-enhanced computed
2007;33:856–62. tomography and acetazolamide test. Acta Neurol Scand Suppl
68. Kushi H, Moriya T, Saito T, et al. Importance of metabolic monitoring 1996;166:139–43.
systems as an early prognostic indicator in severe head injured patients. 94. Ding HY, Han X, Lv CZ, et al. Xenon-CT study of regional cerebral blood
Acta Neurochir Suppl 1999;75:67–8. flow around hematoma in patients with basal ganglia hemorrhage. Acta
69. Menzel M, Doppenberg EM, Zauner A, et al. Cerebral oxygenation in Neurochir Suppl 2008;105:161–4.
patients after severe head injury: monitoring and effects of arterial 95. Mirzai S, Samii M. Current status and future challenges in cerebral blood
hyperoxia on cerebral blood flow, metabolism and intracranial pressure. flow mapping in intracranial tumors. Keio J Med 2000;49(Suppl 1):
J Neurosurg Anesthesiol 1999;11:240–51. A16–24.
70. Shafer R, Brown A, Taylor C. Correlation between cerebral blood flow and 96. Behrens PF, Ostertag CB, Warnke PC. Regional cerebral blood flow in
oxygen saturation in patients with subarachnoid hemorrhage and traumatic peritumoral brain edema during dexamethasone treatment: a xenon-
brain injury. J Neurointerv Surg 2011;Mar 25. Epub ahead of print. enhanced computed tomographic study. Neurosurgery 1998;43:235–40;
71. Bouma GJ, Muizelaar JP, Stringer WA, et al. Ultra-early evaluation of discussion 40–1.
regional cerebral blood flow in severely head-injured patients using 97. Tatagiba M, Mirzai S, Samii M. Peritumoral blood flow in intracranial
xenon-enhanced computerized tomography. J Neurosurg 1992;77:360–8. meningiomas. Neurosurgery 1991;28:400–4.
72. Latchaw RE, Yonas H, Darby JM, et al. Xe-CT cerebral blood flow 98. Van Roost D, Hartmann A, Quade G. Changes of cerebral blood flow
determination following cranial trauma. Acta Radiol Suppl 1986;369:370–3. following dexamethasone treatment in brain tumour patients: a Xe/CT
73. Chieregato A, Fainardi E, Servadei F, et al. Centrifugal distribution of study. Acta Neurochir (Wien) 2001;143:37–43.
regional cerebral blood flow and its time course in traumatic intracerebral 99. Nakatsuka M, Mizuno S. Blood flow study of meningothelial and fibrous
hematomas. J Neurotrauma 2004;21:655–66. meningiomas by xenon-CT. Keio J Med 2000;49(Suppl 1):A164–7.
Section IV—Radiology 281.e3

100. Nasel C, Rossler K, Heimberger K, et al. Tumour blood flow and partition 108. Park EA, Goo JM, Park SJ, et al. Chronic obstructive pulmonary disease:
coefficients: correlation with grade of cerebral gliomas using xenon- quantitative and visual ventilation pattern analysis at xenon ventilation CT
enhanced computed tomography. Neuroradiology 1997;39:627–32. performed by using a dual-energy technique. Radiology 2010;256(3):985–
101. Wang S, Kim S, Chawla S, et al. Differentiation between glioblastomas, 97. Epub 2010 Jul 22.
solitary brain metastases, and primary cerebral lymphomas using diffusion 109. Thieme SF, Hoegl S, Nikolaou K, et al. Pulmonary ventilation and perfusion
tensor and dynamic susceptibility contrast-enhanced MR imaging. AJNR imaging with dual-energy CT. Eur Radiol 2010;20(12):2882–9. Epub 2010
Am J Neuroradiol 2011;32(3):507–1. Jun 23.
102. Johnson DW, Hogg JP, Dasheiff R, et al. Xe-CT cerebral blood flow studies 110. Goo HW, Yang DH, Hong SJ, et al. Xenon ventilation CT using dual-source
during continuous depth electrode monitoring in epilepsy patients. AJNR and dual-energy technique in children with bronchiolitis obliterans:
Am J Neuroradiol 1993;14:245–52. correlation of xenon and CT density values with pulmonary function test
103. Okuchi K, Nagata K, Otsuka H, et al. Regional cerebral blood flow after results. Pediatr Radiol 2010;40(9):1490–7. Epub 2010 Apr 22.
status epilepticus. Keio J Med 2000;49(Suppl 1):A75–6. 111. Takahashi H, Suzuki M, Ikeda H, et al. Evaluation of quantitative portal
104. Pistoia F, Johnson DW, Darby JM, et al. The role of xenon CT venous, hepatic arterial, and total hepatic tissue blood flow using xenon CT
measurements of cerebral blood flow in the clinical determination of brain in alcoholic liver cirrhosis-comparison with liver cirrhosis related to
death. AJNR Am J Neuroradiol 1991;12:97–103. hepatitis C virus and nonalcoholic steatohepatitis. Alcohol Clin Exp Res
105. Darby JM, Yonas H, Gur D, et al. Xenon-enhanced computed tomography 2010;34 Suppl 1:S7–S13. Epub 2009 Nov 5.
in brain death. Arch Neurol 1987;44:551–4. 112. Sesay M, Bidabe AM, Guyot M, et al. Regional cerebral blood flow
106. Goo HW, Yu J. Redistributed regional ventilation after the administration measurements with Xenon-CT in the prediction of delayed encephalopathy
of a bronchodilator demonstrated on xenon-inhaled dual-energy CT in a after carbon monoxide intoxication. Acta Neurol Scand Suppl
patient with asthma. Korean J Radiol 2011;12(3):386–9. Epub 2011 Apr 25. 1996;166:22–7.
107. Sase S, Nakano H, Suzuki H, et al. Subtraction lung image for evaluating 113. Inoue Y, Shiozaki T, Irisawa T, et al. Acute cerebral blood flow variations
pulmonary ventilation in xenon-enhanced CT. Med Phys after human cardiac arrest assessed by stable xenon enhanced computed
2010;37(8):4464–74. tomography. Curr Neurovasc Res 2007;4:49–54.
IV
Chapter
28  

MRI in Neurocritical Care


Damien Galanaud and Louis Puybasset

Introduction not been established. MR examinations often are possible


at 1.5 tesla in patients with pacemakers, neurostimulators,
Knowing the extent of brain damage in the context of acute and implantable defibrillators but remain dangerous, and
cerebral injury can be of paramount importance, especially require extra caution. The presence of ferromagnetic
when the patient is at crossroads, when a decision should be metals inside the eye or central nervous system, such as
made about whether or not to continue care. Although clinical older aneurysm clips, is a contraindication to MRI.
or electrophysiologic examinations provide some informa-  Patient monitoring is hampered because only limited
tion, they are not always reliable because of the degree of visual observation of the subject is possible, and the moni-
sedation these patients receive. Computed tomography (CT) toring devices, designed for use in a high magnetic field
is the first line of imaging examination to rule out a neuro- are not as reliable as their counterparts used in the inten-
surgical emergency (e.g., extra- or subdural hematoma) or sive care unit (ICU). Thus MRI examination should not
suggest increased intracranial pressure (ICP). However, CT be attempted in respiratory or hemodynamically unstable
cannot provide a reliable evaluation of the prognosis because patients, unless the procedure is absolutely needed.
of its poor sensitivity (especially in the posterior fossa) and  The MRI examination should not be attempted in patients
lack of specificity. Magnetic resonance imaging (MRI) has with intracranial hypertension because a prolonged flat
excellent sensitivity to demonstrate brain lesions and charac- position can increase ICP.
terize different pathologic processes and so can be the neuro-  Electric syringes do not operate properly in the magnetic
imaging procedure of choice in many patients. This chapter field and have to be located away from the patient and the
reviews MRI brain imaging, MRI findings in select patholo- magnet. This requires long intravenous (IV) tubing that
gies encountered in the neurocritical care unit (NCCU) (e.g., may slow or alter drug or fluid delivery.
traumatic brain injury, stroke, infection, anoxic injury, and  MRI contrast media injections should be avoided in
hemorrhage among others), and use of MRI including patients with severe renal insufficiency because this may
advanced techniques in estimating prognosis. MRI imaging of lead to nephrogenic systemic fibrosis.
the spine, extracranial vasculature, or other organ systems
(e.g., cardiac MRI) is beyond the scope of this chapter,
although all can be relevant to patient care.1-4 MRI Sequences
An ever-increasing number of sequences can be performed to
MRI in a Brain-Injured Patient: examine the brain. Because the duration of the examination
the Procedure is limited, great care should be taken to select the most appro-
priate sequence for the clinical question. Routinely performed
Contraindications and Safety Rules MRI sequences include the following:
Performing an MRI in a critically brain-injured patient is a
challenging task. The MRI scanner is mostly a very powerful  T1-weighted sequences are the “anatomic” sequences.
magnet (1.5-3 tesla), inside which the subject is almost com- They are used mostly to detect morphologic changes, such
pletely inaccessible during the 30- to 60-minute examination. as ventricle enlargement or brain atrophy, and to detect
There are several consequences of these technical hurdles: subacute parenchymal blood (methemoglobin, areas of
high signal) and areas of contrast enhancement.
 All medical devices associated with the patient must be  T2-weighted sequences are used to demonstrate lesions
MR compatible. In general, since 1995 implantable devices (contusions, ischemia, edema), which generally appear as
(e.g., stents, cardiac valves, neurosurgical clips, or coils areas of increased signal. In neuroimaging the most widely
among others) are almost always MR compatible at used T2-weighted sequence is the fluid-attenuated inver-
1.5 tesla. At higher field strength, however, their safety has sion recovery (FLAIR) sequence in which the signal of the
282 © Copyright 2013 Elsevier Inc. All rights reserved.
Section IV—Radiology 283

cerebrospinal fluid (CSF) is suppressed. In addition to


its superb ability to demonstrate areas of parenchymal MRI of Major Pathologies
increased signal, FLAIR plays an important role in dem-
onstrating acute subarachnoid hemorrhage (SAH). Ischemic Stroke
 T2*-weighted sequences are specially designed to demon- Acute ischemic stroke (AIS) rarely alters consciousness, except
strate intraparenchymal blood, both acute (deoxyhemo- in massive hemispheric infarcts with brain herniation or
globin) and chronic (hemosiderin), which appears as areas basilar artery occlusion. However, the diagnosis of AIS should
of decreased signal, the latter persisting for years. be considered when an ICU patient develops a new neurologic
 MR angiography (MRA) can be performed to examine the deficit, especially if an early CT scan is normal or precipitating
arteries of the circle of Willis or MR venography (MRV) factors such as cardiac or vascular surgery are present, or a
to evaluate the intracranial veins and sinuses. brainstem or posterior fossa stroke is likely.13,14 MRI is the
 Diffusion-weighted imaging studies the random move- examination of choice to confirm the diagnosis. A rapid (less
ments of water molecules. It is represented as the apparent than an hour after symptom onset) decrease of the ADC of
diffusion coefficient (ADC), which is a physical character- the affected brain area is seen on MRI.15,16 FLAIR is normal
istic of the tissue. Only a very limited number of patho- within the first 4 to 6 hours and then shows areas of increased
logic processes can decrease the ADC, the most widely signal. MRA may demonstrate the level of vascular obstruc-
encountered being acute ischemia. tion. The presence of an area of decreased ADC and normal
FLAIR is highly sensitive and specific of AIS (Fig. 28.1). MRI
These sequences are available on all MR scanners and can be findings in acute stroke are listed in Table 28.1.
routinely performed. More advanced acquisitions, where The gold standard imaging technique to assess cerebral
available, can be used to better characterize the pathologic hemodynamics (i.e., flow and metabolism) is positron emis-
processes and the extent of brain damage. Indeed, normally sion tomography (PET) (see Chapter 29), but novel MRI
appearing brain tissue on T2-weighted sequences can be either approaches provide methods to measure cerebral blood flow
normal or abnormal according to what other sequences (CBF), cerebral blood volume (CBV), and cerebral metabolic
are used: rate of oxygen (CMRO2).17

 Diffusion tensor imaging (DTI) is a variant of diffusion  When perfusion-weighted imaging (PWI) is used with
imaging that studies the spatial restriction of diffusion. diffusion-weighted imaging (DWI), the ischemic penum-
The presence of white matter tracts “channels” the water bra often can be identified.18-20 In PWI, protons in arterial
molecules, which move in a preferential direction. The blood to the brain are magnetically tagged; this creates an
preferential restriction of diffusivity is measured by the intra-arterial contrast with the intracranial contents and
fractional anisotropy (FA). This sequence can also be used so allows for calculation of perfusion maps (i.e., cerebral
to follow individual white matter tracts, such as the pyra- blood flow). PWI is relatively time consuming to obtain,
midal and spinothalamic. This processing is called fiber but a PWI-DWI mismatch in which there is still viable
tracking. tissue but inadequate flow identifies tissue at risk for
 MR spectroscopy (MRS) studies brain metabolism. This infarction. This information can be used to guide throm-
sequence quantifies molecules specific to some cell types bolytic therapy. Alternatively, a DWI-FLAIR mismatch can
or physiologic processes that may assist in differentiating be used to guide thrombolytic therapy.21 Compared with
brain tumors from radiation necrosis or from an abscess PET studies MRI evaluation may overestimate the size of
among others.5 The most easily detected are as follows: the penumbra.22
 N-acetyl aspartate (NAA) is a neuronal marker synthe-  Advances in phase contrast MRA allow noninvasive assess-
sized in the mitochondria of neurons from the amino ment of blood flow rates and flow direction through quan-
acid aspartic acid, which decreases where there is titative magnetic resonance angiography (qMRA). This
neuron damage or death. For example, a reduction in permits evaluation of CBF and collateral circulation in
NAA is observed after stroke reaching a nadir about 2 patients with stroke, transient ischemic attacks (TIAs), or
weeks after symptom onset.6 occlusive cerebral vascular disease, or after bypass surgery.23
 Creatine (Cr), a ubiquitous molecule involved in ener-  Arterial spin labeling (ASL) MRI, including pulsed ASL
getic metabolism. It is often used as a reference. (PASL) or continuous ASL (CASL), remains the most
 Choline (Cho) increases in cases of membrane synthe- popular MRI method used to measure CBF.24 ASL does not
sis (e.g., brain tumors) or catabolism. require exogenous contrast and is best used to measure
 Blood oxygen level–dependent (BOLD) functional MRI CBF in the cerebral cortex. Results are less robust in white
(fMRI) is less commonly used because it is difficult to matter.25 A newer ASL technique, vessel-selective ASL (VS-
perform in a sedated patient. It has mostly been used ASL) permits separate labeling of feeding arteries such that
after the acute phase, in vegetative or minimally con- perfusion territories can be measured.26
scious patients, to search for the presence of residual  CBV can be measured by modified ASL approaches.27 In
high order functional activities.7-11 The coherent sponta- addition, CBV may be noninvasively measured using vas-
neous BOLD fluctuations, observed with the resting state cular space occupancy (VASO) MRI and its variant “inflow
fMRI method, which do not require the patient’s cooper- VASO” (iVASO),28 or through quantitative STAR labeling
ation, have been shown to be still present under general of arterial regions (QUASAR).27
anesthesia and could possibly be useful to evaluate the  Decisions about CBF require an understanding about
persistence of structured brain activities in comatose metabolism (CMRO2). For example, brain tissue remains
patients.10,12 viable when CMRO2 is preserved through increased oxygen
284 Section IV—Radiology

A B C

D E F G
Fig. 28.1  This 52-year-old male presented with rapid onset of ataxia and then coma. Magnetic resonance imaging (MRI) in diffusion-weighted
(A, B, with corresponding apparent diffusion coefficient [ADC] maps, D, E) and fluid-attenuated inversion recovery (FLAIR) (C) sequences. MR
angiography (F) and conventional digital substracted angiography (G). Areas of increased signal on diffusion imaging in the pons, cerebellum, and
thalami, with a decreased ADC corresponding to an acute ischemic stroke are seen. The relatively normal FLAIR sequence indicates that this stroke
is recent (<3-4 hours). MR angiography and digital subtraction angiography (DSA) show occlusion of the basilar artery.

this technique can be applied in a quantitative fashion to


Table 28.1  Magnetic Resonance Imaging patients in the ICU.
Findings in Acute Stroke
Hyperintensity on diffusion-weighted imaging Intracranial Hematoma
Little or no signal change on T2 or fluid-attenuated inversion CT scan is the imaging modality of choice to diagnose an
recovery (FLAIR) sequences
Hypointensity on apparent diffusion coefficient acute intracranial hematoma. However, MRI can be per-
Hypointense artery sign of acute intravascular thrombus on formed as a diagnostic tool to determine the etiology of the
gradient-recalled echo sequences intracranial hematoma30 and evaluate the extent of surround-
Absent arterial flow void, i.e., vessel occlusion, on ing edema or as a prognostic tool to determine the extent of
T2-weighted or FLAIR sequences
Hyperintense vessel sign, which suggests slow or collateral
brain injury (see later section on The Prognostic Value of
flow, on FLAIR sequences MRI). Hemorrhagic lesions are best detected with T2*-
Arterial occlusion on magnetic resonance angiography may be weighted sequences, on which they appear as areas of decreased
visible signal. This sequence also may show associated small hypoin-
Reduced or absent contrast on dynamic perfusion-weighted tensities scattered in the brain (or “blebs”), resulting from
imaging source images
Reduced or absent perfusion on perfusion-weighted imaging previous microvascular injuries. FLAIR sequences can help
perfusion parameter maps define the extent of other vascular risk factor–related brain
lesions (e.g., lacunar infarcts and leukoaraiosis).
Modified from Merino JG, Warach S. Imaging of acute stroke. Nat Rev Neurol
2010;6(10):560–71. Epub 2010, Sep 14.

Anoxic Brain Injury


extraction fraction (OEF) even when CBF is reduced. OEF Anoxic brain injury, for example cardiac arrest, is common.
is best evaluated using PET, but BOLD provides an indirect Lesions can involve the pallidum, the cerebellum, the thala-
marker of neuronal activity and so can provide qualitative mus, the striatum, and the cortex (laminar necrosis), more
results that relate CBF and CMRO2 in patients with chronic specifically the occipital and perirolandic areas (Figs. 28.2 and
cerebrovascular disease.29 More research is required before 28.3).31,32 The brainstem usually is spared. Lesions appear as
Section IV—Radiology 285

A B C
Fig. 28.2  Intracranial hemorrhage complicated by severe cardiomyopathy; 3 weeks later the patient remains in coma. Magnetic resonance
imaging (MRI) in T1 (A), fluid-attenuated inversion recovery (FLAIR) (B), and diffusion-weighted (C) sequences. There is a diffuse hyperintensity of
the cortex on T1 and FLAIR sequences that suggest laminar necrosis. The atrophy and increased signal of the striatum seen on diffusion imaging also
are associated with the prolonged low cardiac output.

A B
1494 Units RM 1805 Units RM
1400
Control
1200
1500 NAA

1000
NAA Cho
800 1000

600 Cr

400
500
200

0
0
–200

–490 –400
4.30 3.71 3.09 2.48 1.87 1.25 0.64 0.39 ppm 4.30 3.71 3.09 2.48 1.87 1.25 0.64 0.39 ppm
C D
Fig. 28.3  A 42-year-old male with altered consciousness 1 week after cardiac arrest. Magnetic resonance imaging (MRI) fluid-attenuated
inversion recovery (FLAIR) sequence (A, B) and MR spectroscopy (box in A) on the thalamus (C) and of a control subject (D). There are lesions of the
lenticular nuclei and occipital and frontal motor (arrows) cortices on the FLAIR sequence. N-acetyl aspartate (NAA) in the thalamus is decreased
compared with the control subject. These findings suggest anoxic ischemic injury. Cho, Choline; Cr, creatine.
286 Section IV—Radiology

hyperintense first on FLAIR and diffusion imaging (usually a However, the potential prognostic value of MRI in this
decreased ADC), then on T1 sequences.33 The time course of context remains to be fully elucidated.
these changes is still poorly known; they can appear within the
first 24 hours or be delayed for as much as 1 week. The pres-
ence of hyperintensity in the cortex (particularly the motor
Posterior Reversible
cortex) and striatum suggests poor neurologic prognosis.34 Leukoencephalopathy Syndrome
Diffusion MRI also may help predict long-term outcome after Posterior reversible leukoencephalopathy syndrome (PRESS)
cardiac arrest.35 This can be particularly valuable when induced is a complication of uncontrolled hypertension and of some
hypothermia is used.36 In the most severe forms, brain atrophy treatments such as immunosuppressors or some chemothera-
is observed in the following weeks.37,38 MR spectroscopy, if peutic agents (e.g., intrathecal methotrexate). Its diagnosis is
performed early, may show the presence of lactate.39,40 Later, a easily made with MRI, which shows areas of subcortical edema
decrease in NAA can be observed (see Fig. 28.3). Its prognostic that predominate in the posterior areas. The lesions also can
value has been well established in children, especially in cases involve the frontal white matter or posterior fossa. Contrast
of near drowning and perinatal anoxia.41,42 Susceptibility- enhancement is uncommon. Diffusion imaging should be
weighted imaging also may be used to help predict outcome performed to evaluate PRESS: two different patterns can be
after neonatal hypoxic-ischemic injuries.43 observed51,52:
 Areas of increased ADC correspond to vasogenic edema.
Subarachnoid Hemorrhage These lesions usually are reversible and associated with
good prognosis.
CT is considered the imaging modality of choice to detect  Areas of decreased ADC correspond to cytotoxic edema;
acute SAH. MRI may be useful, depending on what sequences
these lesions generally are not reversible and suggest a
are performed to diagnose SAH if CT is negative and in the
more reserved prognosis.
subacute phase. The presence of blood in the subarachnoid
spaces gives a hyperintensity on the FLAIR sequence.44 False
negatives may occur and a normal examination does not rule Traumatic Brain Injury
out the diagnosis, especially if the MRI is performed several CT is the initial imaging study for traumatic brain injury
days after the onset of symptoms45 because of CSF washing of (TBI) because it permits rapid and accurate diagnosis of
blood. In addition the diagnostic accuracy of FLAIR may be intracranial hematoma and mass effect on which to base
reduced when the SAH is in compressed cisterns.46 Higher- management decisions. MRI is useful for follow-up evalua-
field magnets (3 tesla and more) tend to have a higher rate of tion especially of diffuse axonal injury (DAI), examination
both false positives and negatives. MRA or MRI can be used of brainstem pathology, and to aid in decisions about
to demonstrate the causative aneurysm and can help evaluate prognosis.53-55 Newer MRI sequences (e.g., gradient-recalled
ischemic complications secondary to vasospasm. The results echo, susceptibility-weighted imaging (SWI), and DWI) have
of MRA are most accurate when there is severe arterial nar- greater sensitivity for diffuse brain injury, whereas diffusion
rowing but less reliable for moderate or mild vasospasm.47 In tensor imaging provides information about white matter
addition, the dynamics of blood flow are altered when there injury.53,55 Multiple mechanisms lead to cerebral injury in
are multiple areas of vessel narrowing; this compromises the TBI; hence the imaging pattern can be varied and heteroge-
accuracy of time-of-flight sequences. Finally, admission DWI neous. This chapter focuses on pathology observed in the
imaging may help guide management decisions in poor-grade brain and what is most relevant to follow up during ICU
SAH patients; those with no or only spotty DWI lesions tend care. The reader is referred to recent reviews for a full dis-
to have a good outcome.48 cussion of imaging in TBI including of the skull and cranio-
facial structures and for the common data elements on TBI
imaging.53-56
Infectious Brain Diseases
MRI can be extremely useful in some infectious brain  Cerebral contusions can be edematous or hemorrhagic.
diseases. They appear as areas of hyperintensity on the FLAIR
sequence, with associated hypointensity on the T2*
 The lesions of herpetic meningoencephalitis predominate sequence when hematomas are present. Their most
in the temporal lobes, insula, and basi-frontal areas. Con- frequent locations are the basi-frontal areas and the
trast-enhanced, ischemic or hemorrhagic foci may be temporal lobes.
present.  DAI (Fig. 28.5). This injury results usually from decelera-
 Bacterial meningitis usually does not require an MRI tion and is associated with shearing of the white matter
examination. This examination is sometimes performed in fibers. Three different kinds of DAI can be observed:
patients who develop focal signs or altered consciousness,  Hemorrhagic DAI is the most easily detected: the white
in search for an abscess, ventriculitis, or vasculitis. matter lesions are associated with lesions of the small
 Brain abscesses appear as cystic, contrast-enhancing arteries, causing very small areas of bleeding, which
lesions. The capsule shows hypointensity on T2 or FLAIR give hypointense dots (or blebs) on the T2* sequence.
and slight hypersignal on T1-weighted sequences, the They have been associated with persistent cognitive
content of the abscess has decreased ADC, and amino acids impairment in patients with an otherwise normal
can be detected on MR spectroscopy49,50 (Fig. 28.4). MRI.57 SWI is a very sensitive method to identify
 Both edematous and ischemic lesions of the white matter microbleeds, which are a marker of traumatic axonal
may be seen in septic shock–induced brain dysfunction. injury.58-60
Section IV—Radiology 287

A B C D
Aa

Aa

4 3 2 1 0 4 3 2 1 0
E F
Fig. 28.4  This 45-year-old patient with a history of intravenous drug abuse and alcoholic cirrhosis presented with fever and a rapidly
altering consciousness. Magnetic resonance imaging (MRI) images: T1 with gadolinium (A), fluid-attenuated inversion recovery (FLAIR) with
gadolinium (B), diffusion-weighted sequences (C), and apparent diffusion coefficient (ADC) map (D). MR spectroscopy at short (E) and long (F) echo
times. The contrast-enhancing cystic lesion is high signal on diffusion imaging with a decreased ADC, and the presence of amino acids (Aa) on MR
spectroscopy are typical of an intracranial abscess. The associated meningitis and ventriculitis are diagnosed by the presence of increased signal of
the sulci and the frontal horns of the lateral ventricles on the FLAIR sequence.

Ischemic DAI occurs when the lesions of the small


 where the compression and displacement of this structure
arteries lead to vascular occlusion and ischemia. They give microhemorrhagic infarctions detectable on the T2*
are best detected with the diffusion sequence that sequence (Duret’s hemorrhages).62
shows punctiform areas of decreased ADC.  Intracranial hypertension (IH) is common in TBI patients
 DAI without vascular lesions is the most difficult to admitted to the NCCU. The underlying mechanism can
detect. These lesions do not give any sign on MR include disruption of the blood-brain barrier, space-
sequences, except atrophy of relatively late onset. DTI occupying lesions (hematomas, contusions), and cytotoxic
is probably the best method to detect them; interrup- edema from ischemic lesions. Although indirect signs of
tion of the white matter tract leads to a decrease IH such as reduced ventricles and sulci can be detected on
in the fractional anisotropy of the area, or can be CT or MRI, the severity of IH is difficult to quantify on
detected by fiber tracking. However, although some imaging. The consequences of IH can be evaluated at the
studies have shown marked difference in fractional subacute phase with DTI; the global injury to the brain
anisotropy values between TBI subjects with a good parenchyma leads to a diffuse decrease of supratentorial
and poor prognosis, the role of DTI to determine FA (see Fig. 28.5).
prognosis of individual patients remains to be fully  Extracranial complications also may adversely affect the
defined.59 brain parenchyma. The most common is hypotension or
 Acute vascular lesions can arise from direct compression, hypovolemia, which is associated with lesions similar to
commonly the posterior cerebral artery between the tem- those observed in anoxia or ischemia.
poral lobe and the midbrain (e.g., during temporal hernia-  Although patients with mild TBI and concussion rarely
tion). Carotid or vertebral dissection can lead to secondary require care in the NCCU, there is continued debate on
ischemic strokes that may occur several days after the initial the diagnosis and management and in particular whether
injury.61 These mechanisms lead to “classic” strokes, easily the extent of tissue-based impairments or other stressors
demonstrated with diffusion imaging. Other processes can causes disability. Conventional MRI may be insensitive to
lead to a more insidious form of ischemia. Subdural and microstructural changes that may be present. Advanced
extradural hematomas stretch the vascular structures, and imaging techniques, including SWI that is very sensitive to
can lead to a devascularization of brain parenchyma. These microbleeds, DTI using different measures (i.e., FA, mean
lesions may be difficult to detect, except in the midbrain, diffusivity [MD], and axial diffusivity [AD] to examine
288 Section IV—Radiology

A B C

Control

D E
Fig. 28.5  Prolonged intracranial hypertension in a 17-year-old female after head injury. The patient remained unconscious 1 month later.
Magnetic resonance imaging (MRI) images: fluid-attenuated inversion recovery (FLAIR) (A), T2* (B), diffusion-weighted (C), and diffusion tensor
(D, fractional anisotropy map) sequences in the patient and an fractional anisotropy (FA) map at the same level of a control subject (E). There are
patchy areas of hyperintensity on FLAIR sequence, with areas of decreased signal on T2* and increased signal on diffusion imaging that correspond
to ischemic and hemorrhagic diffuse axonal injury. The diffuse alteration of the white matter tracks on the fractional anisotropy map results from
both diffuse axonal injuries and prolonged intracranial hypertension.

white matter), MR spectroscopy, and resting state fMRI to and involves the determination of: (1) the reversibility of
evaluate the default mode network (DMN), however, can lesions, (2) their bilaterality and symmetry, and (3) the
help in evaluation of mild TBI and provide objective bio- involvement of critical functional brain areas.
markers that underlie long-term cognitive deficits.59,63-69
The DTI maps can be normalized to the International
Consortium for Brain Mapping atlas for atlas-based analy- Reversibility of Traumatic
sis and can be assessed using global methods (i.e., white Brain Injury Lesions
matter histogram or voxel-based approaches) or a regional On conventional MRI images, the evolution and reversibility
approach (i.e., tractography).70 of most lesions is well described. Vasogenic edema, whether
local (e.g., in a contusion or surrounding a hematoma) or
diffuse generally resolves over time. The natural evolution of
the hematomas is spontaneous regression over several weeks,
The Prognostic Value of MRI with the ultimate persistence of a “scar” that is hyperintense
Advanced MRI techniques can be used to help determine the on FLAIR and hypointense on T2*. The reversibility of these
prognosis of the severely brain-injured patient, but this is still lesions means that although they may appear extensive on
under investigation. Anoxic-ischemic disease and TBI have the initial MRI, they may not always mean poor prognosis,
been best studied.71-73 In a prospective cohort of severe TBI, it especially if they do not involve major functional areas. By
was observed in 106 patients that the use of quantitative DTI contrast, other lesions may be considered “irreversible”
in the acute setting enhanced the sensitivity and specificity of (e.g., ischemic lesions, and most DAI lesions). In these “irre-
12-month functional outcome prediction compared with the versible” lesions there is a slow progression toward brain
International Mission for Prognosis and Analysis of Clinical atrophy, increasing sometimes even months after the initial
Trials in TBI (IMPACT) score alone74 in preparation. Given event.75 Most structural brain imaging studies after TBI that
the complexity of this examination, there are less available incorporate volumetric measures and a longitudinal design
data in some pathologies such as intracranial hemorrhage. demonstrate some degree of brain atrophy that progresses
Establishing the prognosis of TBI patients is relatively complex, over time.76 This pattern of atrophy often is regionally
Section IV—Radiology 289

selective and associated with the severity of the injury and  The prognostic value of unilateral lesions of the thalamus,
outcome.77 pons or mesencephalon, and of metabolic alterations seen
The reversibility of the alterations seen on DTI and MRS on MR spectroscopy without associated lesions on conven-
remains to be fully elucidated. An NAA reduction on MRS tional sequences remains to be fully elucidated. However,
maybe reversible because it can be associated with transient a significant decrease in NAA in these areas usually is asso-
neuronal dysfunction rather than death. Whether there is a ciated with a poor prognosis.85
threshold value in NAA that differentiates between reversible
and irreversible neuronal damages has yet to be established. Lesions of critical areas associated with high-order functions
Similar limits exist with diffusion tensor imaging; whereas do not per se impair consciousness but can lead to significant
major reductions in FA correspond to white matter destruc- deficits that must be taken into account when decisions about
tion, late, localized, re-increases in FA values are described. prognosis and care are made. The deleterious effects of lesions
This FA increase may result from axonal regrowth or in cortical areas are relatively easy to predict. However, the
remyelination.78 prognostic significance of alterations in the white matter
tracts that originate from or connect with these areas, is less
studied and remains a challenge. DTI, using fractional anisot-
Involvement of Major Brain Structures ropy maps and fiber tracking algorithms, will probably be the
Evaluation of the integrity or impairment of major brain method of choice, but the application of this postprocessing
structures is a critical step in the prognostic evaluation of TBI method to an individual patient’s diagnosis and to outcome
patients. From a practical viewpoint these can be divided into prediction is still in its infancy although promising in its utility
areas critical for regaining consciousness, and structures including for patients who remain unconsciousness several
involved in high-order brain functions. Alternatively, lesions days after injury.72,86-89 fMRI studies of functional connectivity
may be considered according to depth—hemisphere, central, in the default mode network in the chronic phase after TBI
and brainstem injury. In severe TBI, bilateral brainstem lesions also may help in outcome prediction in the chronic phase after
are strongly predictive of poor outcome, but the relationship TBI.90 The prognostic value of lesions in some cortical or
of lesion location to outcome in moderate TBI is less clear.79 subcortical areas is as follows:
The mechanisms of consciousness and their imaging cor-  Lesions of the primary motor cortex and pyramidal
relates remain incompletely understood. Dehaene et al. and
tract are associated with hemiplegia or hemiparesia, but
Parvizi and Damasio among others, have developed and
extensive destruction of the motor cortex is uncommon.
studied the concept of the “global workspace,” an interconnec-
The pyramidal tract is more commonly involved, by
tion of brainstem nuclei, white matter tracts, deep brain struc-
hematomas of the centrum semiovale or internal capsule.
tures, and cortical areas, the integrity of which is critical to
There can be delayed recovery in some patients of this
consciousness. The following network is considered to be
function.
important for consciousness: the substantia reticulata, located  Lesions of the hippocampi and medial temporal lobe are
in the posterior pons and connected to the posterointernal
associated with short-term memory difficulties. Distinct
thalamic nuclei through the mesencephalon.80,81 The thalamic
fMRI patterns during working memory also may provide
nuclei in turn are associated with the hypothalamus, the basal
insights into recovery in the chronic stage.91
forebrain, and various cortical areas including (but not limited  Extensive lesions of the frontal lobes and cingular cortex
to) the cingulum (see also Chapter 10 for a discussion on
can cause behavioral problems such as akinetic mutism,
consciousness). Lesions in these areas are associated with a
which may limit full recovery and integration into a
delayed recovery of consciousness, a vegetative state (VS), or
normal social life.
minimally conscious state (MCS). Consistent with this, studies  Unilateral lesions, even extensive, may be relatively well
have demonstrated that functional connectivity within the
tolerated with late recovery, whereas this is not true if the
well-characterized default mode network is associated with
lesions are bilateral. Lesions of the language areas (Broca’s
the level of arousal in subjects with severe brain injury.82,83 In
and Wernicke’s) could carry a less favorable prognosis
addition, arterial spin labeling (ASL) techniques to examine
related to asymmetry of this brain function.
CBF in different brain regions may provide further insight
into functional recovery84 in the subacute stage. What remains
to be determined is exactly how the extent of lesions in these
areas of the brain relates to poor prognosis.
Conclusion
MRI in critically ill NCCU patients can be a challenge, for
 Hemorrhagic lesions of the posterior pons, bilateral tha- example, patient transport to the MRI facility or the pres-
lamic lesions (either ischemic or hemorrhagic) that are ence of monitors that are not MRI compatible and need to
visible on conventional MR sequences, appear to be associ- be replaced. However, MRI can provide very useful diagnos-
ated with poor prognosis. tic and prognostic information for patient management. The
 Bilateral lesions of the mesencephalon often are associated imaging protocol should routinely include both conven-
with VS or MCS. These lesions in TBI often result from a tional (T1 and T2 weighted) and advanced (DTI, MR spec-
shearing mechanism that occurs during deceleration troscopy) imaging techniques. Imaging technologies and
because of the different mass between the telencephalon techniques continue to evolve and in the future, multivariate
and posterior fossa. They may be difficult to detect with analysis of the information including that derived from
conventional MRI sequences, but are easily visible on dif- other advanced imaging techniques such as volumetric MRI
fusion tensor images. However, quantitative assessment of analysis, SWI, DTI, magnetization transfer imaging (MTI),
mesencephalic injury with DTI still is a challenge. ASL, fMRI (including resting state and connectivity MRI),
290 Section IV—Radiology

and hyperpolarization scanning may help objectively and 27. Petersen ET, Lim T, Golay X. Model-free arterial spin labeling quantification
approach for perfusion MRI. Magn Reson Med 2006;55:219–32.
reliably predict the outcome of the patient.
28. Hua J, Qin Q, Donahue MJ, et al. Inflow-based vascular-space occupancy
(IVASO) MRI. Magn Reson Med 2011;66:40–56.
References 29. Heyn C, Poublanc J, Crawley A, et al. Quantification of cerebrovascular
reactivity by blood oxygen level-dependent MR imaging and correlation
1. Looby S, Flanders A. Spine trauma. Radiol Clin North Am 2011;49(1):129– with conventional angiography in patients with Moyamoya disease. Am J
63. Review. Neuroradiol 2010;31:862–7.
2. Mughal MM, Khan MK, DeMarco JK, et al. Symptomatic and asymptomatic 30. Kamel H, Navi BB, Hemphill JC 3rd. A rule to identify patients who require
carotid artery plaque. Expert Rev Cardiovasc Ther 2011;9(10):1315–30. magnetic resonance imaging after intracerebral hemorrhage. Neurocrit Care
3. Chiribiri A, Ishida M, Nagel E, et al. Coronary imaging with cardiovascular 2011;Jul 15. Epub ahead of print.
magnetic resonance: current state of the art. Prog Cardiovasc Dis 31. Wijdicks EF, Campeau NG, Miller GM. MR imaging in comatose survivors
2011;54(3):240–52. of cardiac resuscitation. AJNR Am J Neuroradiol 2001;22(8):1561–5.
4. Jiji RS, Kramer CM. Cardiovascular magnetic resonance: applications in 32. Tha KK, Terae S, Yamamoto T, et al. Early detection of global cerebral
daily practice. Cardiol Rev 2011;19(5):246–54. Review. anoxia: improved accuracy by high-b-value diffusion-weighted imaging with
5. Elias AE, Carlos RC, Smith EA, et al. MR spectroscopy using normalized and long echo time. AJNR Am J Neuroradiol 2005;26(6):1487–97.
non-normalized metabolite ratios for differentiating recurrent brain tumor 33. Arbelaez A, Castillo M, Mukherji SK. Diffusion-weighted MR imaging of
from radiation injury. Acad Radiol 2011;18(9):1101–8. global cerebral anoxia. AJNR Am J Neuroradiol 1999;20(6):999–1007.
6. Muñoz Maniega S, Cvoro V, Chappell FM, et al. Changes in NAA and lactate 34. Barrett KM, Freeman WD, Weindling SM, et al. Brain injury after
following ischemic stroke: a serial MR spectroscopic imaging study. cardiopulmonary arrest and its assessment with diffusion-weighted magnetic
Neurology 2008;71(24):1993–9. resonance imaging. Mayo Clin Proc 2007;82(7):828–35.
7. Owen AM, Coleman MR, Menon DK, et al. Residual auditory function in 35. Wijman CA, Mlynash M, Caulfield AF, et al. Prognostic value of brain
persistent vegetative state: a combined PET and fMRI study. Neuropsychol diffusion-weighted imaging after cardiac arrest. Ann Neurol
Rehabil 2005;15(3–4):290–306. 2009;65(4):394–402.
8. Siva N. Patient in a coma plays tennis. Lancet Neurol 2006;5(11):906. 36. Oddo M, Rossetti AO. Predicting neurological outcome after cardiac arrest.
9. Schiff ND. fMRI reveals large-scale network activation in minimally Curr Opin Crit Care 2011;17(3):254–9.
conscious patients. Neurology 2005;64(3):514–23. 37. Allen JS, Tranel D, Bruss J, et al. Correlations between regional brain
10. Salvador R, Suckling J, Coleman MR, et al. Neurophysiological architecture volumes and memory performance in anoxia. J Clin Exper Neuropsychol
of functional magnetic resonance images of human brain. Cereb Cortex 2006;28(4):457–76.
2005;15(9):1332–42. 38. Ammermann H, Kassubek J, Lotze M, et al. MRI brain lesion patterns in
11. Owen AM, Coleman MR, Boly M, et al. Detecting awareness in the vegetative patients in anoxia-induced vegetative state. J Neurol Sci
state. Science 2006;313(5792):1402. 2007;260(1-2):65–70.
12. Vincent JL, Patel GH, Fox MD, et al. Intrinsic functional architecture in the 39. Geraghty MC, Torbey MT. Neuroimaging and serologic markers of
anaesthetized monkey brain. Nature 2007;447(7140):83–6. neurologic injury after cardiac arrest. Neurol Clin 2006;24(1):107–21, vii.
13. Hwang DY, Silva GS, Furie KL, et al. Comparative sensitivity of computed 40. Berek K, Lechleitner P, Luef G, et al. Early determination of neurological
tomography vs. magnetic resonance imaging for detecting acute posterior outcome after prehospital cardiopulmonary resuscitation. Stroke
fossa infarct. J Emerg Med 2012;Feb 2. Epub ahead of print. 1995;26(4):543–9.
14. Merino JG, Warach S. Imaging of acute stroke. Nat Rev Neurol 41. Kreis R, Arcinue E, Ernst T, et al. Hypoxic encephalopathy after near-
2010;6(10):560–71. Epub 2010, Sep 14. drowning studied by quantitative 1H-magnetic resonance spectroscopy.
15. Schlaug G, Siewert B, Benfield A, et al. Time course of the apparent diffusion J Clin Invest 1996;97(5):1142–54.
coefficient (ADC) abnormality in human stroke. Neurology 42. Zarifi MK, Astrakas LG, Poussaint TY, et al. Prediction of adverse outcome
1997;49(1):113–9. with cerebral lactate level and apparent diffusion coefficient in infants with
16. Oppenheim C, Grandin C, Samson Y, et al. Is there an apparent diffusion perinatal asphyxia. Radiology 2002;225(3):859–70.
coefficient threshold in predicting tissue viability in hyperacute stroke? 43. Kitamura G, Kido D, Wycliffe N, et al. Hypoxic-ischemic injury: utility of
Stroke 2001;32(11):2486–91. susceptibility-weighted imaging. Pediatr Neurol 2011;45(4):220–4.
17. Donahue MJ, Strother MK, Hendrikse J. Novel MRI approaches for assessing 44. Noguchi K, Seto H, Kamisaki Y, et al. Comparison of fluid-attenuated
cerebral hemodynamics in ischemic cerebrovascular disease. Stroke inversion-recovery MR imaging with CT in a simulated model of acute
2012;43(3):903–15. Epub 2012, Feb 16. subarachnoid hemorrhage. AJNR Am J Neuroradiol 2000;21(5):923–7.
18. Nour M, Liebeskind DS. Brain imaging in stroke: insight beyond diagnosis. 45. Mohamed M, Heasly DC, Yagmurlu B, et al. Fluid-attenuated inversion
Neurotherapeutics 2011;8(3):330–9. recovery MR imaging and subarachnoid hemorrhage: not a panacea. AJNR
19. Yoo AJ, González RG. Clinical applications of diffusion MR imaging for Am J Neuroradiol 2004;25(4):545–50.
acute ischemic stroke. Neuroimaging Clin N Am 2011;21(1):51–69, vii. 46. Shimoda M, Hoshikawa K, Shiramizu H, et al. Problems with diagnosis by
20. Copen WA, Schaefer PW, Wu O. MR perfusion imaging in acute ischemic fluid-attenuated inversion recovery magnetic resonance imaging in patients
stroke. Neuroimaging Clin N Am 2011;21(2):259–83, x. with acute aneurysmal subarachnoid hemorrhage. Neurol Med Chir (Tokyo)
21. Thomalla G, Cheng B, Ebinger M, et al. DWI-FLAIR mismatch for the 2010;50(7):530–7.
identification of patients with acute ischaemic stroke within 4·5 h of 47. Hattingen E, Blasel S, Dumesnil R, et al. MR angiography in patients with
symptom onset (PRE-FLAIR): a multicentre observational study. Lancet subarachnoid hemorrhage: adequate to evaluate vasospasm-induced vascular
Neurol 2011;10(11):978–86. Epub 2011, Oct 4. narrowing? Neurosurg Rev 2010;33(4):431–9. Epub 2010, Jun 8.
22. Heiss WD. The ischemic penumbra: correlates in imaging and implications 48. Sato K, Shimizu H, Fujimura M, et al. Acute-stage diffusion-weighted
for treatment of ischemic stroke. The Johann Jacob Wepfer Award 2011. magnetic resonance imaging for predicting outcome of poor-grade
Cerebrovasc Dis 2011;32(4):307–20. Epub 2011, Sep 15. aneurysmal subarachnoid hemorrhage. J Cereb Blood Flow Metab
23. Amin-Hanjani S, Charbel FT, Malisch T, et al. Use of quantitative magnetic 2010;30(6):1110–20. Epub 2010, Jan 6.
resonance angiography to stratify stroke risk in symptomatic vertebrobasilar 49. Grand S, Passaro G, Ziegler A, et al. Necrotic tumor versus brain abscess:
disease. Stroke 2005;36:1140–5. importance of amino acids detected at 1H MR spectroscopy-initial results.
24. Hernandez DA, Bokkers RP, Mirasol RV, et al. Pseudocontinuous arterial Radiology 1999;21(3):785–93.
spin labeling quantifies relative cerebral blood flow in acute stroke. Stroke 50. Luthra G, Parihar A, Nath K, et al. Comparative evaluation of fungal,
2012;43(3):753–8. Epub 2012, Feb 16. tubercular, and pyogenic brain abscesses with conventional and diffusion
25. van Osch MJ, Teeuwisse WM, van Walderveen MA, et al. Can arterial spin MR imaging and proton MR spectroscopy. AJNR Am J Neuroradiol
labeling detect white matter perfusion signal? Magn Reson Med 2007;28(7):1332–8.
2009;62:165–73.
26. Wong EC. Vessel-encoded arterial spin-labeling using pseudocontinuous A complete list of references for this chapter can be found online at
tagging. Magn Reson Med 2007;58:1086–91. www.expertconsult.com.
Section IV—Radiology 290.e1

References 30. Kamel H, Navi BB, Hemphill JC 3rd. A rule to identify patients who require
magnetic resonance imaging after intracerebral hemorrhage. Neurocrit Care
1. Looby S, Flanders A. Spine trauma. Radiol Clin North Am 2011;49(1):129– 2011;Jul 15. Epub ahead of print.
63. Review. 31. Wijdicks EF, Campeau NG, Miller GM. MR imaging in comatose survivors
2. Mughal MM, Khan MK, DeMarco JK, et al. Symptomatic and asymptomatic of cardiac resuscitation. AJNR Am J Neuroradiol 2001;22(8):1561–5.
carotid artery plaque. Expert Rev Cardiovasc Ther 2011;9(10):1315–30. 32. Tha KK, Terae S, Yamamoto T, et al. Early detection of global cerebral
3. Chiribiri A, Ishida M, Nagel E, et al. Coronary imaging with cardiovascular anoxia: improved accuracy by high-b-value diffusion-weighted imaging with
magnetic resonance: current state of the art. Prog Cardiovasc Dis long echo time. AJNR Am J Neuroradiol 2005;26(6):1487–97.
2011;54(3):240–52. 33. Arbelaez A, Castillo M, Mukherji SK. Diffusion-weighted MR imaging of
4. Jiji RS, Kramer CM. Cardiovascular magnetic resonance: applications in global cerebral anoxia. AJNR Am J Neuroradiol 1999;20(6):999–1007.
daily practice. Cardiol Rev 2011;19(5):246–54. Review. 34. Barrett KM, Freeman WD, Weindling SM, et al. Brain injury after
5. Elias AE, Carlos RC, Smith EA, et al. MR spectroscopy using normalized and cardiopulmonary arrest and its assessment with diffusion-weighted magnetic
non-normalized metabolite ratios for differentiating recurrent brain tumor resonance imaging. Mayo Clin Proc 2007;82(7):828–35.
from radiation injury. Acad Radiol 2011;18(9):1101–8. 35. Wijman CA, Mlynash M, Caulfield AF, et al. Prognostic value of brain
6. Muñoz Maniega S, Cvoro V, Chappell FM, et al. Changes in NAA and lactate diffusion-weighted imaging after cardiac arrest. Ann Neurol
following ischemic stroke: a serial MR spectroscopic imaging study. 2009;65(4):394–402.
Neurology 2008;71(24):1993–9. 36. Oddo M, Rossetti AO. Predicting neurological outcome after cardiac arrest.
7. Owen AM, Coleman MR, Menon DK, et al. Residual auditory function in Curr Opin Crit Care 2011;17(3):254–9.
persistent vegetative state: a combined PET and fMRI study. Neuropsychol 37. Allen JS, Tranel D, Bruss J, et al. Correlations between regional brain
Rehabil 2005;15(3–4):290–306. volumes and memory performance in anoxia. J Clin Exper Neuropsychol
8. Siva N. Patient in a coma plays tennis. Lancet Neurol 2006;5(11):906. 2006;28(4):457–76.
9. Schiff ND. fMRI reveals large-scale network activation in minimally 38. Ammermann H, Kassubek J, Lotze M, et al. MRI brain lesion patterns in
conscious patients. Neurology 2005;64(3):514–23. patients in anoxia-induced vegetative state. J Neurol Sci
10. Salvador R, Suckling J, Coleman MR, et al. Neurophysiological architecture 2007;260(1–2):65–70.
of functional magnetic resonance images of human brain. Cereb Cortex 39. Geraghty MC, Torbey MT. Neuroimaging and serologic markers of
2005;15(9):1332–42. neurologic injury after cardiac arrest. Neurol Clin 2006;24(1):107–21, vii.
11. Owen AM, Coleman MR, Boly M, et al. Detecting awareness in the vegetative 40. Berek K, Lechleitner P, Luef G, et al. Early determination of neurological
state. Science 2006;313(5792):1402. outcome after prehospital cardiopulmonary resuscitation. Stroke
12. Vincent JL, Patel GH, Fox MD, et al. Intrinsic functional architecture in the 1995;26(4):543–9.
anaesthetized monkey brain. Nature 2007;447(7140):83–6. 41. Kreis R, Arcinue E, Ernst T, et al. Hypoxic encephalopathy after near-
13. Hwang DY, Silva GS, Furie KL, et al. Comparative sensitivity of computed drowning studied by quantitative 1H-magnetic resonance spectroscopy.
tomography vs. magnetic resonance imaging for detecting acute posterior J Clin Invest 1996;97(5):1142–54.
fossa infarct. J Emerg Med 2012;Feb 2. Epub ahead of print. 42. Zarifi MK, Astrakas LG, Poussaint TY, et al. Prediction of adverse outcome
14. Merino JG, Warach S. Imaging of acute stroke. Nat Rev Neurol with cerebral lactate level and apparent diffusion coefficient in infants with
2010;6(10):560–71. Epub 2010, Sep 14. perinatal asphyxia. Radiology 2002;225(3):859–70.
15. Schlaug G, Siewert B, Benfield A, et al. Time course of the apparent diffusion 43. Kitamura G, Kido D, Wycliffe N, et al. Hypoxic-ischemic injury: utility of
coefficient (ADC) abnormality in human stroke. Neurology 1997;49(1):113–9. susceptibility-weighted imaging. Pediatr Neurol 2011;45(4):220–4.
16. Oppenheim C, Grandin C, Samson Y, et al. Is there an apparent diffusion 44. Noguchi K, Seto H, Kamisaki Y, et al. Comparison of fluid-attenuated
coefficient threshold in predicting tissue viability in hyperacute stroke? inversion-recovery MR imaging with CT in a simulated model of acute
Stroke 2001;32(11):2486–91. subarachnoid hemorrhage. AJNR Am J Neuroradiol 2000;21(5):923–7.
17. Donahue MJ, Strother MK, Hendrikse J. Novel MRI approaches for assessing 45. Mohamed M, Heasly DC, Yagmurlu B, et al. Fluid-attenuated inversion
cerebral hemodynamics in ischemic cerebrovascular disease. Stroke recovery MR imaging and subarachnoid hemorrhage: not a panacea. AJNR
2012;43(3):903–15. Epub 2012, Feb 16. Am J Neuroradiol 2004;25(4):545–50.
18. Nour M, Liebeskind DS. Brain imaging in stroke: insight beyond diagnosis. 46. Shimoda M, Hoshikawa K, Shiramizu H, et al. Problems with diagnosis by
Neurotherapeutics 2011;8(3):330–9. fluid-attenuated inversion recovery magnetic resonance imaging in patients
19. Yoo AJ, González RG. Clinical applications of diffusion MR imaging for with acute aneurysmal subarachnoid hemorrhage. Neurol Med Chir (Tokyo)
acute ischemic stroke. Neuroimaging Clin N Am 2011;21(1):51–69, vii. 2010;50(7):530–7.
20. Copen WA, Schaefer PW, Wu O. MR perfusion imaging in acute ischemic 47. Hattingen E, Blasel S, Dumesnil R, et al. MR angiography in patients with
stroke. Neuroimaging Clin N Am 2011;21(2):259–83, x. subarachnoid hemorrhage: adequate to evaluate vasospasm-induced vascular
21. Thomalla G, Cheng B, Ebinger M, et al. DWI-FLAIR mismatch for the narrowing? Neurosurg Rev 2010;33(4):431–9. Epub 2010, Jun 8.
identification of patients with acute ischaemic stroke within 4·5 h of 48. Sato K, Shimizu H, Fujimura M, et al. Acute-stage diffusion-weighted
symptom onset (PRE-FLAIR): a multicentre observational study. Lancet magnetic resonance imaging for predicting outcome of poor-grade
Neurol 2011;10(11):978–86. Epub 2011, Oct 4. aneurysmal subarachnoid hemorrhage. J Cereb Blood Flow Metab
22. Heiss WD. The ischemic penumbra: correlates in imaging and implications 2010;30(6):1110–20. Epub 2010, Jan 6.
for treatment of ischemic stroke. The Johann Jacob Wepfer Award 2011. 49. Grand S, Passaro G, Ziegler A, et al. Necrotic tumor versus brain abscess:
Cerebrovasc Dis 2011;32(4):307–20. Epub 2011, Sep 15. importance of amino acids detected at 1H MR spectroscopy-initial results.
23. Amin-Hanjani S, Charbel FT, Malisch T, et al. Use of quantitative magnetic Radiology 1999;21(3):785–93.
resonance angiography to stratify stroke risk in symptomatic vertebrobasilar 50. Luthra G, Parihar A, Nath K, et al. Comparative evaluation of fungal,
disease. Stroke 2005;36:1140–5. tubercular, and pyogenic brain abscesses with conventional and diffusion
24. Hernandez DA, Bokkers RP, Mirasol RV, et al. Pseudocontinuous arterial MR imaging and proton MR spectroscopy. AJNR Am J Neuroradiol
spin labeling quantifies relative cerebral blood flow in acute stroke. Stroke 2007;28(7):1332–8.
2012;43(3):753–8. Epub 2012, Feb 16. 51. Covarrubias DJ, Luetmer PH, Campeau NG, Posterior reversible
25. van Osch MJ, Teeuwisse WM, van Walderveen MA, et al. Can arterial spin encephalopathy syndrome: prognostic utility of quantitative diffusion-
labeling detect white matter perfusion signal? Magn Reson Med weighted MR images. AJNR Am J Neuroradiol 2002;23(6):1038–48.
2009;62:165–73. 52. Lamy C, Oppenheim C, Méder JF, et al. Neuroimaging in posterior reversible
26. Wong EC. Vessel-encoded arterial spin-labeling using pseudocontinuous encephalopathy syndrome. J Am Soc Neuroimaging 2004;14(2):89–96.
tagging. Magn Reson Med 2007;58:1086–91. 53. Provenzale JM. Imaging of traumatic brain injury: a review of the recent
27. Petersen ET, Lim T, Golay X. Model-free arterial spin labeling quantification medical literature. AJR Am J Roentgenol 2010;194(1):16–9.
approach for perfusion MRI. Magn Reson Med 2006;55:219–32. 54. Gean AD, Fischbein NJ. Head trauma. Neuroimaging Clin N Am
28. Hua J, Qin Q, Donahue MJ, et al. Inflow-based vascular-space occupancy 2010;20(4):527–56.
(IVASO) MRI. Magn Reson Med 2011;66:40–56. 55. Kubal WS. Updated imaging of traumatic brain injury. Radiol Clin North
29. Heyn C, Poublanc J, Crawley A, et al. Quantification of cerebrovascular Am 2012;50(1):15–41.
reactivity by blood oxygen level-dependent MR imaging and correlation 56. Haacke EM, Duhaime AC, Gean AD, et al. Common data elements in
with conventional angiography in patients with Moyamoya disease. Am J radiologic imaging of traumatic brain injury. J Magn Reson Imaging
Neuroradiol 2010;31:862–7. 2010;32(3):516–43.
290.e2 Section IV—Radiology

57. Scheid R, Walther K, Guthke T, et al. Cognitive sequelae of diffuse axonal 75. Tomaiuolo F, Worsley KJ, Lerch J, et al. Changes in white matter in
injury. Arch Neurol 2006;63(3):418–24. long-term survivors of severe non-missile traumatic brain injury: a
58. Colbert CA, Holshouser BA, Aaen GS, et al. Value of cerebral computational analysis of magnetic resonance images. J Neurotrauma
microhemorrhages detected with susceptibility-weighted MR imaging for 2005;22(1):76–82.
prediction of long-term outcome in children with nonaccidental trauma. 76. Ross DE. Review of longitudinal studies of MRI brain volumetry in patients
Radiology 2010;256(3):898–905. with traumatic brain injury. Brain Inj 2011;25(13–14):1271–8.
59. Sharp DJ, Ham TE. Investigating white matter injury after mild traumatic 77. Warner MA, Youn TS, Davis T, et al. Regionally selective atrophy after
brain injury. Curr Opin Neurol 2011;24(6):558–63. traumatic axonal injury. Arch Neurol 2010;67(11):1336–44. Epub 2010, Jul
60. Beauchamp MH, Ditchfield M, Babl FE, et al. Detecting traumatic brain 12.
lesions in children: CT versus MRI versus susceptibility weighted imaging 78. Voss HU, Uluc AM, Dyke JP, et al. Possible axonal regrowth in late recovery
(SWI). J Neurotrauma 2011;28(6):915–27. Epub 2011, Jun 9. from the minimally conscious state. J Clin Invest 2006;116(7):2005–11.
61. Tawil I, Stein DM, Mirvis SE, et al. Posttraumatic cerebral infarction: 79. Skandsen T, Kvistad KA, Solheim O, et al. Prognostic value of magnetic
incidence, outcome, and risk factors. J Trauma 2008;64(4):849–53. resonance imaging in moderate and severe head injury: a prospective study
62. Alexander E, Kushner J, Six EG. Brainstem hemorrhages and increased of early MRI findings and one-year outcome. J Neurotrauma
intracranial pressure: from Duret to computed tomography. Surg Neurol 2011;28(5):691–9. Epub 2011, Apr 26.
1982;17(2):107–10. 80. Dehaene S, Kerszberg M, Changeux JP. A neuronal model of a global
63. Niogi SN, Mukherjee P. Diffusion tensor imaging of mild traumatic brain workspace in effortful cognitive tasks. Proc Natl Acad Sci U S A
injury. J Head Trauma Rehabil 2010;25(4):241–55. 1998;95(24):14529–34.
64. Kasahara K, Hashimoto K, Abo M, et al. Voxel- and atlas-based analysis of 81. Parvizi J, Damasio AR. Neuroanatomical correlates of brainstem coma. Brain
diffusion tensor imaging may reveal focal axonal injuries in mild traumatic 2003;126(Pt 7):1524–36.
brain injury: comparison with diffuse axonal injury. Magn Reson Imaging 82. Soddu A, Vanhaudenhuyse A, Bahri MA, et al. Identifying the default-mode
2012;Jan 26. Epub ahead of print. component in spatial IC analyses of patients with disorders of consciousness.
65. Gonzalez PG, Walker MT. Imaging modalities in mild traumatic brain injury Hum Brain Mapp 2012;33(4);778–96.
and sports concussion. PM R 2011;3(10 Suppl 2):S413–24. 83. Vanhaudenhuyse A, Noirhomme Q, Tshibanda LJ, et al. Default network
66. Henry LC, Tremblay J, Tremblay S, et al. Acute and chronic changes in connectivity reflects the level of consciousness in non-communicative
diffusivity measures after sports concussion. J Neurotrauma brain-damaged patients. Brain 2010;133:161–71.
2011;28(10):2049–59. Epub 2011, Oct 4. 84. Liu AA, Voss HU, Dyke JP, et al. Arterial spin labeling and altered cerebral
67. Henry LC, Tremblay S, Leclerc S, et al. Metabolic changes in concussed blood flow patterns in the minimally conscious state. Neurology
American football players during the acute and chronic post-injury phases. 2011;77(16):1518–23. Epub 2011, Sep 21.
BMC Neurol 2011;11:105. 85. Carpentier A, Galanaud D, Puybasset L, et al. Early morphologic and
68. Johnson B, Zhang K, Gay M, et al. Alteration of brain default network in spectroscopic magnetic resonance in severe traumatic brain injuries can
subacute phase of injury in concussed individuals: resting-state fMRI study. detect “invisible brain stem damage” and predict “vegetative states.”
Neuroimage 2012;59(1):511–8. Epub 2011, Aug 7. J Neurotrauma 2006;23(5):674–85.
69. Mayer AR, Mannell MV, Ling J, et al. Functional connectivity in mild 86. Betz JF, Zhuo J, Roy A, et al. Prognostic value of diffusion tensor imaging
traumatic brain injury. Hum Brain Mapp 2011;32(11):1825–35. parameters in severe traumatic brain injury. J Neurotrauma 2012;Feb 24.
70. Marquez de la Plata CD, Yang FG, Wang JY, et al. Diffusion tensor imaging Epub ahead of print.
biomarkers for traumatic axonal injury: analysis of three analytic methods. 87. Wang JY, Bakhadirov K, Abdi H, et al. Longitudinal changes of structural
J Int Neuropsychol Soc 2011;17(1):24–35. Epub 2010, Nov 12. connectivity in traumatic axonal injury. Neurology 2011;77(9):818–26. Epub
71. Tollard E, Galanaud D, Perlbarg V, et al. Experience of diffusion tensor 2011, Aug 3.
imaging and 1H spectroscopy for outcome prediction in severe traumatic 88. Newcombe V, Chatfield D, Outtrim J, et al. Mapping traumatic axonal injury
brain injury: preliminary results. Crit Care Med 2009;37:1448–55. using diffusion tensor imaging: correlations with functional outcome. PLoS
72. Perlbarg V, Puybasset L, Tollard E, et al. Relation between brain lesion One 2011;6(5):e19214.
location and clinical outcome in patients with severe traumatic brain injury: 89. Matsushita M, Hosoda K, Naitoh Y, et al. Utility of diffusion tensor imaging
a diffusion tensor imaging study using voxel-based approaches. Hum Brain in the acute stage of mild to moderate traumatic brain injury for detecting
Mapp 2009;30(12):3924–33. white matter lesions and predicting long-term cognitive function in adults.
73. Warner MA, Marquez de la Plata C, Spence J, et al. Assessing spatial J Neurosurg 2011;115(1):130–9. Epub 2011, Mar 18.
relationships between axonal integrity, regional brain volumes, and 90. Sharp DJ, Beckmann CF, Greenwood R, et al. Default mode network
neuropsychological outcomes after traumatic axonal injury. J Neurotrauma functional and structural connectivity after traumatic brain injury. Brain
2010;27:2121–30. 2011;134(Pt 8):2233–47.
74. Galanaud G, Perlbarg V, Gupta R, et al. for the NICER (Neuro Imaging in 91. Kasahara M, Menon DK, Salmond CH, et al. Traumatic brain injury alters
Coma Emergence and Recovery). Diffusion tensor imaging for outcome the functional brain network mediating working memory. Brain Inj
prediction in severe traumatic brain injury: results of a prospective 2011;25(12):1170–87.
multi-center trial cohort. N Engl J Med 2012 (submitted).
Chapter
29  
IV

PET and SPECT


Thomas Geeraerts and David K. Menon

increases and N-acetyl aspartate (NAA) reductions in normal-


Introduction appearing tissue in head injury7 probably represent ischemia
Computed tomography (CT) and magnetic resonance imaging and neuronal loss, respectively,8 although acute, reversible
(MRI) provide structural images of the brain. CT allows early NAA reductions may represent mitochondrial dysfunction.9
diagnosis of surgically treatable intracranial space-occupying Changes in choline, creatine, and myoinositol also have been
lesions in traumatic brain injury (TBI) and intracranial hem- demonstrated, but their significance is less certain. NAA
orrhage and exclusion of intracranial hemorrhage before reductions appear to be an important marker of poor
thrombolysis. MRI is very sensitive at detecting white matter outcome,7,10 but their potential reversibility implies that late
abnormalities, and provides early diagnostic and prognostic studies may be required to define long-term significance. In
information in stroke and TBI.1-3 However, therapeutic strate- addition, although some high-resolution studies have been
gies based solely on structural images are limited because published,11 current implementation of the technique is gen-
structural changes are late and irreversible. Moreover, conven- erally limited by relatively poor spatial resolution,12 limited
tional imaging is poor at assessing functional recovery. Func- brain coverage, and long imaging times.11
tional imaging allows early documentation of reversible The major advantage of radioligand techniques such as
ischemia in vivo, validation and calibration of bedside clinical positron emission tomography (PET) and single photon emis-
monitoring tools, characterization of pathophysiology in indi- sion computed tomography (SPECT) is the opportunity to
vidual patients, and early outcome prediction. specifically aim at a target pathophysiologic process, and espe-
cially with PET, the high sensitivity to detect physiologic
changes in brain physiology. To date, PET is probably the most
Functional CT and MRI Versus selective and sensitive method to visualize and measure in vivo
Radioligand Techniques receptor density and oxidative metabolism.13 However, unlike
MRI, the main limitation of radioligand techniques is the
for Imaging Physiology exposure to radiation. For PET and SPECT, the radiation
Functional imaging approaches in clinical and experimental exposure typically ranges between 2 and 10 mSv (depending
stroke have traditionally used cerebral blood flow (CBF) on the specific radioligand use), corresponding to between
thresholds for ischemia and have succeeded in identifying one and four times annual natural background.14 Another
useful predictive values for tissue survival or death. limitation is the importance of partial volume effect for small
Although fewer studies are available in the context of head structure that limits the spatial resolution to 3 to 5 mm for
injury, similar approaches now attempt to identify critical PET and to 10 mm for SPECT. Moreover, the short half-life
physiologic thresholds for tissue survival in TBI.4-6 Xenon CT of positron emitters used for PET as 11C or 15O requires the
can provide absolute quantification of CBF, but this technique presence of a cyclotron in the site where it is used: this increases
does not provide whole brain coverage, with imaging typically costs.
being limited to two to four selected slices. Although absolute
quantification of physiologic parameters allows the most
rational application of these thresholds to clinical contexts,
Methodologic Issues
clinically useful data can be obtained from assessment of rela- In both PET and SPECT, a radioactive isotope is administered
tive changes in cerebrovascular physiology. Perfusion CT or and external detectors measure brain radioactivity. To avoid
MRI with intravenous contrast media allows estimation of prolonged radioactive exposure, relatively short-lived isotopes
relative CBF and volume (rCBF and rCBV, respectively). At a are used to image cerebral physiology and metabolism or to
qualitative level, functional MRI (fMRI) using blood oxygen label proteins, receptors, or neurotransmitters (Table 29.1).
level dependent (BOLD) contrast allows identification of The synthesized labeled compound must have the same meta-
regional brain activity and the assessment of functional con- bolic or pharmacokinetic proprieties as the unlabeled com-
nectivity, but no direct measure of cerebrovascular adequacy. pound. However, they typically are administered in such small
Proton magnetic resonance spectroscopy (1H-MRS) studies amounts that pharmacodynamic effects are not important.
show changes in absolute or relative levels of metabolites, and Isotopes of universal atoms such as carbon or oxygen can be
so can document the consequences of ischemia. Lactate used to label molecules without inducing any modification in
© Copyright 2013 Elsevier Inc. All rights reserved. 291
292 Section IV—Radiology

Table 29.1  Examples of Possible Applications of Radioligand Methods to Study Brain Physiology
Methods Atoms Isotopes Half-life for Emission Examples of Labeled Tracer Applications
11 11
PET Carbon C 20.3 min C-radopride D2 dopamine receptors
11
C-flumazenil GABA-A receptor
11
C-PK11195 Activated microglia
(brain inflammation)
11
C-PiB or 11C-SB13 Amyloid plaques
15
Oxygen O 2.1 min H215O, C15O, and 15O2 CBF, CBV, OEF and CMRO2
18 18
Fluorine F 109 min Fluorodeoxyglucose (FDG) Glucose metabolism
18
Fluoromisonidazole Tissue hypoxia
99m
SPECT Technetium Tc99m 6.01 hr Tc-hexamethylpropyleneamine CBF
oxide
123
Iodine I 13.1 hr Iodoamphetamine CBF

CBF, Cerebral blood flow; CBV, cerebral blood volume; CMRO2, cerebral metabolic rate of oxygen; GABA, gamma-aminobutyric acid; OEF, oxygen extraction fraction;
PET, positron emission tomography; PiB, Pittsburg compound; SPECT, single photon emission computed tomography.

POSITRON EMISSION TOMOGRAPHY SINGLE PHOTON EMISSION


(PET) COMPUTED TOMOGRAPHY (SPECT)

Detectors

Positron emitting
tracer
Single photon emitting
5 mm

β+
tracer
e–
γ ray photon e+
511 keV
γ ray photon
511 keV γ ray photon
140 keV

Scan reconstruction Scan reconstruction

Fig. 29.1  Schematic of detection in positron emission tomography (PET, left panel) and single photon emission computed tomography (SPECT using
99m
Tc, right panel).

the chemical structure. Importantly most PET and SPECT form of two photons (gamma rays) released at an angle of 180
studies are performed away from the neurocritical care unit degrees to each other (Fig. 29.1). This annihilation energy can
(NCCU), and so careful management and anesthetic tech- be detected externally using coincidence detectors, and the
nique if necessary are required because the study environment locus of each reaction localized within the object by computer
can be unfamiliar.15 algorithms. The tracer distributes to different compartments,
according to its pharmacokinetic proprieties. Kinetic model-
ing that often requires measurement of radioactivity input in
Positron Emission Tomography plasma is used to derive quantitative measures of the cerebral
PET is a technique that measures the accumulation of positron- concentration. Typically this process requires placement of an
emitting radioisotopes within a three-dimensional object. The arterial catheter and rapid sampling of arterial blood during
production of positron-emitting tracer requires a cyclotron. the tracer injection and equilibration, so as to provide an
The positrons that are emitted are the antimatter equivalent input function for modeling tracer distribution. With some
of electrons. The collision of an electron and a positron anni- tracers, however, an alternative approach makes use of the
hilates both particles, resulting in a release of energy in the inhomogeneous distribution of the tracer in the brain. This
Section IV—Radiology 293

Parametric
(functional)
Normalization map
Voxel-by-voxel to standardized
analysis space

25 Reference tissue models


20 or
15 Plasma input function
10 40
5 35
0
30
25

kB/mL
Plasma activity
20
Metaboltes corrected plasma activity
15
10
5
0
Select 0 1000 2000 3000 4000
pertinent Plasma sample time [sec]
radiotracer
Appropriate
pharmacokinetic model

Emission
scan

Scan reconstruction
Computer processing

Fig. 29.2  The complex process of analyzing an image from positron emission tomography using 11C-flumazenil.

property can be used to define a brain area without specific PET clearly can define many complex aspects of cerebral
tracer binding, which can be used as a reference area to provide physiology and pathophysiology, it is a research tool that is
a tissue input function.16 For example 11C-flumazenil PET can relatively expensive and not universally available.
be used to estimate central benzodiazepine receptor gamma-
aminobutyric acid (GABA)-A density, with the pons as a
tissue reference without specific binding; this avoids the need Single Photon Emission
to sample arterial blood.17 Computed Tomography
Although PET often is referred to as the gold standard in SPECT relies on the imaging of gamma-emitting tracers
physiologic or metabolic imaging, the absolute values that it in which a single photon is emitted (see Fig. 29.2).
produces result from complex detection and computing Tc99m-hexamethylpropyleneamine oxide (HMPAO)18 and
123
systems, both of which are prone to error (Fig. 29.2). Perhaps I-amphetamine have been used to map CBF.19 SPECT does
most important, the accuracy of figures derived from PET is not require a cyclotron to be produced. SPECT is a relatively
crucially dependent on the validity of kinetic models, many simple and inexpensive technique that can be used to assess
of which have not been tested in the injured brain. Although three-dimensional cerebral perfusion. However, the images
the data obtained using the technique are extremely valuable, produced are of relatively low resolution and generally
and probably not obtainable by other methods, it is essential nonquantitative.
that the physiologic insights provided by the PET are assessed Although recent papers have reported on quantitative
critically in the context of these considerations. Although implementations of SPECT,20 this requires further validation.
294 Section IV—Radiology

rCBF CMRO2 OEF CMRgluc OGR

0-30 ml/100 g/min 0-100 µm/100 g/min 0-75% 0-40 µm/100 g/min 0-10

Fig. 29.3  Triple oxygen positron emission tomography (PET) at day 1 after removal of a traumatic left subdural hematoma. On the side
of subdural hematoma is reduced relative cerebral blood flow (rCBF) and cerebral metabolic rate of oxygen (CMRO2) with high oxygen extraction
fraction (OEF) that suggests compensation for ischemia. The decrease in oxygen glucose ratio (OGR) indicates a disproportionate reduction in oxidative
metabolism. Nonoxidative glucose use is inefficient at generating adenosine triphosphate (ATP); this helps explain the perilesional hyperglycolysis
(increased fluorodeoxyglucose [FDG] uptake). CMRgluc, Cerebral metabolic rate for glucose.

One interesting development has been the use of analysis tech- Tc99m-HMPAO SPECT to detect symptomatic vasospasm is
niques such as statistical parametric mapping, derived from only 50% and perhaps less in predicting vessel narrowing in
functional brain imaging, to examine changes in patterns of some vascular distributions.34 Newer semiquantitative SPECT
blood flow across populations of patients with head injury.21 studies using Hermes brain registration and analysis software
(BRASS) may improve on this but requires further study.35
PET CBF in symptomatic patients also has been found to be
Imaging of Cerebral Blood Flow less than that observed in asymptomatic patients.36 The cor-
relation between metabolic markers of brain ischemia mea-
and Volume sured by microdialysis and a decrease in PET CBF is not
Patient evaluation in neurocritical care often may be con- excellent, suggesting other causes for metabolic disorders.36,37
founded by the use of sedative agents and by the metabolic Nevertheless, PET or the combination of techniques such as
effects of trauma or injury,22 which may cause primary reduc- PET, perfusion CT and microdialysis can provide insights into
tions in cerebral metabolism. A decrease in CBF that is coupled the role of cellular hypoxia after SAH,38 or the response to
with the depressed metabolism would not represent ischemia. therapies such as normal saline boluses to induce hypervol-
Under these circumstances the only true measure of the ade- emia39 or blood transfusion to improve oxygen delivery.40
quacy of CBF is a measurement of the oxygen extraction frac- Several groups describe the use of PET and SPECT to map
tion (OEF). A robust identification of ischemia depends on TBI pathophysiology. Perhaps the most interesting of these are
the identification of areas with increased OEF. Triple oxygen studies in the acute phase, which aim to elucidate early patho-
PET (using 15O-labeled tracers in three separate scans (H215O, physiology. In patients with mild to moderate TBI within the
C15O, and 15O2) provides valid quantitative data for CBF, cere- first 3 months after injury, CBF measured by SPECT is reduced
bral blood volume (CBV), OEF, and the cerebral metabolic by 40% to 70% predominantly in the frontal and temporal
rate of oxygen (CMRO2) (Fig. 29.3).23-25 lobes, basal ganglia, and thalamus.41 This reduced CBF was
PET can play an important role in understanding the associated with unfavorable outcome and poor performance
pathophysiologic interplay among CBF brain cellular function on neuropsychological testing and had a better relationship
in ischemic stroke, carotid artery disease, vascular dementia, with long-term outcome than CT or conventional MRI.41-43 At
intracerebral hemorrhage, and aneurysmal subarachnoid 6 months after TBI the extent of regional brain atrophy cor-
hemorrhage (SAH).26 In clinical stroke, PET identifies a core relates best with PET measurements of CMRO2 and CBF and
area with very low CBF, CBV, and oxidative metabolism, that less so lobar OEF.44 SPECT-detected improvements in CBF
corresponds to irreversible damage.27 The surrounding area also have been observed to occur when there is cognitive
with normal or elevated CBV and relative CBF preservation recovery after concussion45 or can help explain flow patterns
with an increase in OEF is termed the penumbra28 and repre- associated with cognitive fatigue.46 However, SPECT as a
sents tissue at risk, that may be rescued by physiologic or stand-alone test may not be sufficient to confirm that a head
pharmacologic intervention. The extent of the penumbra injury did or did not occur.47,48
measured with PET has been shown in stroke patients to help PET has been used to precisely define the ischemic thresh-
predict tissue outcome.29,30 old after TBI. A recent whole brain voxel-based study demon-
After SAH, PET and SPECT have helped to document the strated the presence of distributed ischemia within 24 hours
modification of CBF and CBV that may show a significant of TBI, as defined by an increased OEF.49,50 In contrast, previ-
decrease during vasospasm.31-33 However, the sensitivity of ous studies have found low global OEF values after TBI,
Section IV—Radiology 295

suggesting that global ischemia is uncommon.51,52 These dif- some regions. Interestingly, these OEF increases are not only
ferences may reflect difficulties in demonstrating ischemia at from reduced oxygen delivery but also associated with increases
different times after injury, when early compensated ischemia in oxygen demand from hyperventilation. The burden of early
is associated with a high OEF. By contrast, progressive cell ischemia measured using these techniques correlates well with
death within the voxel results in progressive reductions in eventual outcome.32
OEF that culminate in very low values in voxels where most Other studies have correlated PET-derived measures of
neurons have died. Averaged CBF in areas that eventually cerebrovascular physiology with bedside monitoring tools
progressed to structural abnormality was significantly reduced such as microdialysis and brain tissue oxygen49,59 (Fig. 29.4).
(median CBF 16.9 mL/100 g/min) compared with nonlesion These studies help validate and calibrate bedside monitors,
areas. These variations may be confounded by spatial averag- define the limits or reliability of a given technique, and provide
ing of OEF within structurally defined regions or across the insights into pathophysiology.60
whole brain, possibly due to the dilution of small regions Blood flow and metabolism vary dramatically across the
with increased OEF by larger regions with normal or low traumatized brain, and different regions may require different
OEF. It also is possible that there are mechanisms of ischemic therapeutic approaches. This is difficult to achieve in the clini-
cell death that are not characterized by OEF increases, such as cal environment because medical therapies are “global” in
microvascular ischemia49 and mitochondrial dysfunction.53,54 nature. However, it is best to measure the effect of common
Indeed hypoperfused pericontusional regions where there is therapeutic interventions in which benefit is demonstrated to
oxygen hypometabolism in the subacute stage after TBI may decide about continuing that therapy. A potential advantage
not always result in tissue necrosis, emphasizing the need to of PET and monitoring of brain metabolism may be detection
know about CBF and metabolism (and from any monitoring of abnormalities in brain regions without structural change
modality) before conclusions about ischemia are made.55 on other imaging modalities.
Both the extent and duration of ischemia contribute to the
evolution of irreversible tissue damage. This time dependence
may mean that it is not possible to predict tissue outcome Imaging of Oxygen
based on measurement of CBF, CMRO2, or OEF at a single
time point after injury. It also is likely that tissue survival will
and Glucose Metabolism
be better predicted by complex interactions of physiologic 18-Fluorodeoxyglucose (18FDG) PET can be used to estimate
variables rather than simple univariate thresholds. Initial mul- glucose metabolism. Brain-injured patients commonly dem-
tivariate analysis appears to provide better separation of onstrate evidence of global hypometabolism and metabolic
damaged and nonlesion tissue, and further investigation may stress that fails to recover in patients who have a poor outcome.
involve using multivariate statistical analysis or neural network Bergsneider et al.61 demonstrated increased global and perile-
techniques to determine if there are combinations of physio- sional 18FDG uptake in acute head injury using combined
logic variables that better predict tissue outcome. Indeed tech- microdialysis FDG PET. Comparison with global measure-
niques using multivariate algorithms (using data from acute ments suggested that these represented hyperglycolysis (imply-
perfusion-weighted and diffusion-weighted MRI scans), to ing anaerobic glucose utilization) rather than simple
generate voxel-based maps predicting tissue outcome, have hypermetabolism. The pathophysiology underlying these
been used with some success in stroke studies.56 abnormalities is beginning to be elucidated, but astrocyte glu-
PET studies can be repeated and used to assess changes in tamate reuptake, relative ischemia, and obligate hyperglycoly-
physiology with therapeutic maneuvers, such as osmotherapy, sis in inflammatory cells have all been considered as possible
hyperventilation, hyperoxia, transfusion, and CPP augmenta- causes. In addition ictal FDG PET can show increased hip-
tion.40,49,57,58 Triple-oxygen PET has been used to address the pocampal glucose uptake in TBI patients with seizures or non-
metabolic effects of hyperventilation in TBI. Hyperventilation convulsive seizures.62 Further insight into pathophysiology
to an arterial carbon dioxide tension (PaCO2) of 25 mm Hg can be obtained when the FDG PET studies are combined
produces some increase in OEF, but does cause a significant with microdialysis studies of brain glucose.60 However, there
reduction in global CMRO252 or in regions of interest with low is some concern that changes in the handling of FDG in the
baseline CBF values.51 It is interesting that these studies did injured brain may make standard kinetic models for this
not detect regions with extremely high OEF values that would tracer inappropriate and therefore contribute to an apparent
suggest true ischemia, even within 24 hours of head injury. FDG increase uptake that is an artifact.63
Other groups have used PET to show that moderate reduc- The oxygen-glucose ratio (OGR; CMRO2/CMRgluc, both
tions in PaCO2 (to 31 mm Hg) can result in increases in the measured in µmol/100 g/min) is calculated using CMRO2 and
volume of brain tissue with CBF values that are less than CMRgluc from triple oxygen and FDG PET.64 This ratio has
ischemic thresholds (<20 mL/100 g/min).32 Importantly, the been used to support the concept of nonischemic metabolic
development of these ischemic areas that typically are peri- crisis.65 Reduction in OGR indicates a disproportionate reduc-
contusional or in white matter may not be detected by reduc- tion in oxidative metabolism, probably related to impaired
tions in jugular bulb oxygen saturations below commonly mitochondrial function and in TBI is abnormal in three quar-
accepted thresholds for ischemia (<55%). Subsequent studies ters of the patients during the early phase, suggesting that
have shown that ischemia, defined as cerebral venous oxygen metabolic crisis without ischemia is common after TBI.65
18
content of less than 3.5 mL per 100 mL (that equates to OEF FDG PET also has been used to quantify recovery from
values of 75%-80%), are common in early head injury and head injury. Although 18FDG uptake in the acute phase cor­
may be observed beyond 24 hours post injury. The volume of relates poorly with clinical state,22 sequential changes in
brain defined as ischemic using these criteria shows an increase FDG uptake parallel functional recovery.66 For example, Garcia-
with hyperventilation associated with reduced CMRO2 in Panach et al.67 studied 49 severe TBI patients who remained
296 Section IV—Radiology

50
45 r = 0.69
40

Lactate/pyruvate ratio
35
30
25
20
15
10
5
0
0 10 20 30 40 50 60 70
B Oxygen extraction fraction (%)

5 r = 0.46

PbtO2 (kPa)
3

2
OEF
1

0
0-75% 20 25 30 35 40 45
A
C Oxygen extraction fraction (%)

Fig. 29.4  Positron emission tomography (PET) validation and calibration of bedside clinical monitoring tools. A region of interest that
corresponds to catheter location is identified (gray circle). Oxygen extraction fraction (OEF) measured by PET (A) correlates with metabolic parameter
of oxidative metabolism as lactate-to-pyruvate ratio measured by microdialysis (B) and brain tissue oxygen (PbtO2) measured with Neurotrend sensor
(C). (Redrawn from Johnston AJ, Steiner LA, Coles JP, et al. Effect of cerebral perfusion pressure augmentation on regional oxygenation and metabolism after head injury.
Crit Care Med 2005;33[1]:189–95; Hutchinson PJ, Gupta AK, Fryer TF, et al. Correlation between cerebral blood flow, substrate delivery, and metabolism in head injury:
a combined microdialysis and triple oxygen positron emission tomography study. J Cereb Blood Flow Metab 2002;22[6]:735–45.)

vegetative or minimally conscious, had recovered but had post- postconcussive symptoms compared with normal controls. In
traumatic amnesia, or were fully recovered, and 10 controls addition, functional imaging studies such as PET may have
using FDG PET. The patients with the least recovery had the important ethical implications for the care of brain-injured
most severe hypometabolism. In addition, greater levels of patients, for example, enhance the ability to diagnose a vegeta-
activation of the corticocortical connections were associated tive or minimally conscious state.71,72
with better neurologic recovery, and thalamic metabolism The use of FDG PET is not limited to understanding brain
was least in the vegetative and minimally conscious patients.67 metabolism. Impaired gas exchange and inflammation are
Similarly, in boxers with suspected chronic TBI decreased hallmarks of lung disorders such as acute respiratory distress
18-fluorodeoxyglucose (18FDG) uptake is observed in select syndrome (ARDS), acute lung injury (ALI), and ventilator-
brain regions.68 In patients who remain in coma 3 months after induced lung injury (VILI). PET can be used to examine the
injury there appears to be good concordance between FDG relationship between the distributions of pulmonary perfu-
PET and fMRI.69 sion, ventilation and aeration, and the effect of positive end-
Other studies have used nonquantitative H215O PET with expiratory pressure, recruitment maneuvers, and prone
cognitive paradigms to assess patterns of cortical activation in positioning on these pulmonary changes in ALI, whereas FDG
the recovery period of head injury. These functional activation PET can be used to study regional neutrophil metabolic acti-
studies do not address acute pathophysiology, but may provide vation in ALI and VILI.73-75 In addition, SPECT studies can
important information about the neural substrate of cognitive help diagnose pulmonary emboli.76 PET and SPECT also can
and mnemonic deficits after head injury. One area where this be used to evaluate cardiovascular disease including early
has the potential to be helpful is to differentiate structural or detection of pathophysiologic processes before the disease
functional brain damage from comorbid depression or post- manifests,77 whereas FDG PET may play a role in the evalua-
traumatic stress disorder (PTSD) in patients with repetitive tion of a fever of unknown origin in which it has a very high
mild TBI, especially combat veterans.70 FDG PET studies show negative predictive value.78 Finally, FDG PET has been used as
decreased cerebral metabolic rate of glucose in the cerebellum, a surrogate endpoint in trials to evaluate therapies for
vermis, pons, and medial temporal lobe in TBI patients with Alzheimer’s disease.79
Section IV—Radiology 297

Brain Receptor Imaging cerebral beta-amyloid in patients with Down syndrome and
Alzheimer’s disease.100-102 Currently [(18)F]3’-F-PiB (flute-
Dopamine and Serotonin Receptors metamol), (18)F-AV-45 (florbetapir), and (18)F-AV-1 (florbe-
Modification in dopaminergic neurotransmission is involved taben) are undergoing phase II and III clinical trials. Ideally
in many neurodegenerative diseases, as well as in schizophre- these ligands may be able to help detect amyloid plaque in vivo
nia. Different subtypes of dopamine receptors can be imaged in the brains of Alzheimer’s disease patients or subjects with
with PET and SPECT.13 In vivo quantification of functional mild cognitive impairment.103
effect of drugs like neuroleptics is of interest in such patholo-
gies, and is a major source of PET development. Abnormal
function of serotoninergic neurotransmission is involved in
Conclusion
depression and compulsive disorders. Radioligand-labeled PET and less so SPECT are relatively expensive and not uni-
molecules with specific binding to serotonin receptor sub- versally available, but they represent powerful research tools
types using PET or SPECT appears to be of great interest for functional imaging of the brain of patients in the NCCU
and allows study of pathophysiology and therapeutic inter­ and for diverse pathologies such as Parkinson’s disease,
ventions.80-82 The potential list of receptors that can be imaged Alzheimer’s disease, depression and other mood disorders,
using radioligand techniques—cholinergic, opioid, and ade- cancer including evaluation of glioblastoma aggressiveness,
nosine receptors—is in constant evolution.13 This can provide lung dysfunction including detection of pulmonary embolus,
insight into pathophysiologic processes of specific diseases or cardiac disease, and fever.73,76-78,104-111 PET provides quantitative
recovery; for example, Östberg et al.83 observed that choliner- and SPECT qualitative data, and in neurocritical care both are
gic function assessed with [methyl-(11)C]N-methylpiperidyl- reliable techniques to monitor brain physiology. Their excel-
4-acetate that reflects acetylcholinesterase (AChE) activity was lent sensitivity at detecting small changes in cerebral physiol-
less in TBI survivors than controls, particularly when there ogy has helped increase knowledge about the processes of
was cognitive impairment. brain injury and recovery in a variety of neurologic conditions
including TBI and stroke. Furthermore, use of these techniques
in relatively small patient samples has provided insight into
GABA-A Receptors how commonly used therapeutic strategies (e.g., induced
GABA receptors are expressed in the cortex and deep gray hypertension or hyperventilation) affect brain physiology and
matter and can be imaged with 11C-flumazenil (11C-FMZ) have so informed better therapeutic approaches.
PET, a neuronal benzodiazepine or GABA receptor ligand.84,85
11
C-FMZ binding is very low in white matter. This marker
can be used to measure neuronal integrity or loss.25,86-88
Acknowledgments
SPECT imaging with [123I] iomazenil (IMZ)) also suggests Dr. Thomas Geeraerts is supported by grants from the Société Française
that imaging with this radioligand for the central benzodiaz- d’Anesthésie et de Réanimation (SFAR) and from Journées d’Enseignement
epine receptor in the cortex may have the potential to dis- Post-Universitaire d’Anesthésie-Réanimation (JEPU)-Novo Nordisk.
close a reversible vulnerability of neurons following TBI.89 In Professor David K. Menon is part of the Neuroscience Theme at the
ischemic stroke 11C-FMZ PET is very sensitive at detecting Cambridge Biomedical Research Centre, and is supported by grants from
irreversible tissue damage and can be used to select patients the National Institute of Health Research UK, Medical Research Council
who may benefit from reperfusion therapy.90,91 11C-FMZ also (UK), Royal College of Anaesthetists, Wellcome Trust and Queens’
is used with high sensitivity in epilepsy to study the modifi- College Cambridge.
cations of GABAergic neurotransmission induced by the
pathology and by its treatment with antiepileptic drugs.92,93 References
When combined with FDG PET, 11C-FMZ PET can help 1. Kampfl A, Franz G, Aichner F, et al. The persistent vegetative state after
provide insights into the coma and the vegetative state.72 closed head injury: clinical and magnetic resonance imaging findings in 42
patients. J Neurosurg 1998;88(5):809–16.
2. Kampfl A, Schmutzhard E, Franz G, et al. Prediction of recovery from
Specific Pathophysiologic Processes post-traumatic vegetative state with cerebral magnetic-resonance imaging.
Lancet 1998;351(9118):1763–7.
PK11195 is a ligand for peripheral benzodiazepine receptors 3. Lovblad KO, Baird AE, Schlaug G, et al. Ischemic lesion volumes in acute
expressed by microglia and up-regulated in case of microglia stroke by diffusion-weighted magnetic resonance imaging correlate with
activation.94 PK11195 PET has been used to image, quantify, clinical outcome. Ann Neurol 1997;42(2):164–70.
and localize microglia activation in experimental models of 4. Heiss WD, Huber M, Fink GR, et al. Progressive derangement of periinfarct
viable tissue in ischemic stroke. J Cereb Blood Flow Metab 1992;12(2):193–203.
ischemia95 and in humans in multiple sclerosis, Alzheimer’s 5. Marchal G, Beaudouin V, Rioux P, et al. Prolonged persistence of substantial
disease, and herpes encephalitis.96-98 Because microglia activa- volumes of potentially viable brain tissue after stroke: a correlative PET-CT
tion is thought to be involved in the many pathologic pro- study with voxel-based data analysis. Stroke 1996;27(4):599–606.
cesses that contribute to neurodegeneration, the quantification 6. Powers WJ, Grubb RL, Jr., Darriet D, et al. Cerebral blood flow and cerebral
metabolic rate of oxygen requirements for cerebral function and viability in
of the effect of therapeutic intervention on reactive microglia humans. J Cereb Blood Flow Metab 1985;5(4):600–8.
is of particular interest. For example, studies in TBI patients 7. Garnett MR, Blamire AM, Rajagopalan B, et al. Evidence for cellular damage
using PK11195 PET show that increased microglial activation in normal-appearing white matter correlates with injury severity in patients
may be present up to 17 years after TBI and that high PK following traumatic brain injury: a magnetic resonance spectroscopy study.
binding in the thalamus is associated with greater cognitive Brain 2000;123(Pt 7):1403–9.
8. Higuchi T, Fernandez EJ, Maudsley AA, et al. Mapping of lactate and
impairment.99 N-acetyl-L-aspartate predicts infarction during acute focal ischemia: in vivo
More recently PET studies with carbon 11-labeled Pitts- 1H magnetic resonance spectroscopy in rats. Neurosurgery 1996;38(1):121–
burgh compound B ([(11)C]PiB) have been used to measure 9; discussion 129–30.
298 Section IV—Radiology

9. Signoretti S, Marmarou A, Tavazzi B, et al. N-Acetylaspartate reduction as a 32. Coles JP, Minhas PS, Fryer TD, et al. Effect of hyperventilation on cerebral
measure of injury severity and mitochondrial dysfunction following diffuse blood flow in traumatic head injury: clinical relevance and monitoring
traumatic brain injury. J Neurotrauma 2001;18(10):977–91. correlates. Crit Care Med 2002;30(9):1950–9.
10. Friedman SD, Brooks WM, Jung RE, et al. Quantitative proton MRS predicts 33. Jabre A, Babikian V, Powsner RA, et al. Role of single photon emission
outcome after traumatic brain injury. Neurology 1999;52(7):1384–91. computed tomography and transcranial Doppler ultrasonography in clinical
11. Beauchamp NJ, Jr, Barker PB, Wang PY, et al. Imaging of acute cerebral vasospasm. J Clin Neurosci 2002;9(4):400–3.
ischemia. Radiology 1999;212(2):307–24. 34. Kincaid MS, Souter MJ, Treggiari MM, et al. Accuracy of transcranial
12. Garnett MR, Corkill RG, Blamire AM, et al. Altered cellular metabolism Doppler ultrasonography and single-photon emission computed
following traumatic brain injury: a magnetic resonance spectroscopy study. tomography in the diagnosis of angiographically demonstrated cerebral
J Neurotrauma 2001;18(3):231–40. vasospasm. J Neurosurg 2009;110(1):67–72.
13. Heiss WD, Herholz K. Brain receptor imaging. J Nucl Med 35. Cooke D, Koppula B, Seiler D, et al. Semiquantitative software SPECT
2006;47(2):302–12. analysis in aneurysmal subarachnoid hemorrhage-related vasospasm. Nucl
14. Small GW, Bookheimer SY, Thompson PM, et al. Current and future uses of Med Commun 2010;31(1):53–8.
neuroimaging for cognitively impaired patients. Lancet Neurol 36. Sarrafzadeh AS, Haux D, Ludemann L, et al. Cerebral ischemia in
2008;7(2):161–72. aneurysmal subarachnoid hemorrhage: a correlative microdialysis-PET
15. Veenith T, Coles JP. Anaesthesia for magnetic resonance imaging and study. Stroke 2004;35(3):638–43.
positron emission tomography. Curr Opin Anaesthesiol 2011;24(4):451–8. 37. Enblad P, Valtysson J, Andersson J, et al. Simultaneous intracerebral
16. Gunn RN, Lammertsma AA, Hume SP, et al. Parametric imaging of microdialysis and positron emission tomography in the detection of
ligand-receptor binding in PET using a simplified reference region model. ischemia in patients with subarachnoid hemorrhage. J Cereb Blood Flow
Neuroimage 1997;6(4):279–87. Metab 1996;16(4):637–44.
17. Klumpers UM, Veltman DJ, Boellaard R, et al. Comparison of plasma input 38. Sarrafzadeh AS, Nagel A, Czabanka M, et al. Imaging of hypoxic-ischemic
and reference tissue models for analysing [(11)C]flumazenil studies. J Cereb penumbra with (18)F-fluoromisonidazole PET/CT and measurement of
Blood Flow Metab 2008;28(3):579–87. related cerebral metabolism in aneurysmal subarachnoid hemorrhage.
18. Bullock R, Statham P, Patterson J, et al. The time course of vasogenic J Cereb Blood Flow Metab 2010;30(1):36–45. Epub 2009, Sep 23.
oedema after focal human head injury—evidence from SPECT mapping of 39. Jost SC, Diringer MN, Zazulia AR, et al. Effect of normal saline bolus on
blood brain barrier defects. Acta Neurochir Suppl (Wien) 1990;51:286–8. cerebral blood flow in regions with low baseline flow in patients with
19. Shiina G, Onuma T, Kameyama M, et al. Sequential assessment of cerebral vasospasm following subarachnoid hemorrhage. J Neurosurg 2005;103(1):
blood flow in diffuse brain injury by 123I-iodoamphetamine single-photon 25–30.
emission CT. AJNR Am J Neuroradiol 1998;19(2):297–302. 40. Dhar R, Zazulia AR, Videen TO, et al. Red blood cell transfusion increases
20. Della Corte F, Giordano A, Pennisi MA, et al. Quantitative cerebral blood cerebral oxygen delivery in anemic patients with subarachnoid hemorrhage.
flow and metabolism determination in the first 48 hours after severe head Stroke 2009;40(9):3039–44. Epub 2009, Jul 23.
injury with a new dynamic SPECT device. Acta Neurochir (Wien) 1997; 41. Bavetta S, Nimmon CC, White J, et al. A prospective study comparing SPET
139(7):636–41; discussion 41–2. with MRI and CT as prognostic indicators following severe closed head
21. Stamatakis EA, Wilson JT, Hadley DM, et al. SPECT imaging in head injury injury. Nucl Med Commun 1994;15(12):961–8.
interpreted with statistical parametric mapping. J Nucl Med 2002;43(4): 42. Jacobs A, Put E, Ingels M, et al. Prospective evaluation of technetium-99m-
476–83. HMPAO SPECT in mild and moderate traumatic brain injury. J Nucl Med
22. Bergsneider M, Hovda DA, Lee SM, et al. Dissociation of cerebral glucose 1994;35(6):942–7.
metabolism and level of consciousness during the period of metabolic 43. Prayer L, Wimberger D, Oder W, et al. Cranial MR imaging and cerebral
depression following human traumatic brain injury. J Neurotrauma 2000; 99mTc HM-PAO-SPECT in patients with subacute or chronic severe closed
17(5):389–401. head injury and normal CT examinations. Acta Radiol 1993;34(6):593–9.
23. Frackowiak RS, Jones T, Lenzi GL, et al. Regional cerebral oxygen utilization 44. Xu Y, McArthur DL, Alger JR, et al. Early nonischemic oxidative metabolic
and blood flow in normal man using oxygen-15 and positron emission dysfunction leads to chronic brain atrophy in traumatic brain injury. J Cereb
tomography. Acta Neurol Scand 1980;62(6):336–44. Blood Flow Metab 2010;30(4):883–94. Epub 2009, Dec 23.
24. Frackowiak RS, Lenzi GL, Jones T, et al. Quantitative measurement of 45. Amen DG, Wu JC, Taylor D, et al. Reversing brain damage in former NFL
regional cerebral blood flow and oxygen metabolism in man using 15O and players: implications for traumatic brain injury and substance abuse
positron emission tomography: theory, procedure, and normal values. rehabilitation. J Psychoactive Drugs 2011;43(1):1–5.
J Comput Assist Tomogr 1980;4(6):727–36. 46. Hattori N, Swan M, Stobbe GA, et al. Differential SPECT activation patterns
25. Sette G, Baron JC, Mazoyer B, et al. Local brain haemodynamics and oxygen associated with PASAT performance may indicate frontocerebellar functional
metabolism in cerebrovascular disease: positron emission tomography. Brain dissociation in chronic mild traumatic brain injury. J Nucl Med
1989;112(Pt 4):931–51. 2009;50(7):1054–61. Epub 2009, Jun 12.
26. Powers WJ, Zazulia AR. PET in cerebrovascular disease. PET Clin 2010;5(1): 47. Wortzel HS, Filley CM, Anderson CA, et al. Forensic applications of cerebral
83106. single photon emission computed tomography in mild traumatic brain
27. Baron JC. Mapping the ischaemic penumbra with PET: implications for injury. J Am Acad Psychiatry Law 2008;36(3):310–22.
acute stroke treatment. Cerebrovasc Dis 1999;9(4):193–201. 48. Lewine JD, Davis JT, Bigler ED, et al. Objective documentation of traumatic
28. Moustafa RR, Baron JC. Clinical review: imaging in ischaemic stroke— brain injury subsequent to mild head trauma: multimodal brain imaging
implications for acute management. Crit Care 2007;11(5):227. with MEG, SPECT, and MRI. J Head Trauma Rehabil 2007;22(3):141–55.
29. Heiss WD. Best measure of ischemic penumbra: positron emission 49. Johnston AJ, Steiner LA, Coles JP, et al. Effect of cerebral perfusion pressure
tomography. Stroke 2003;34(10):2534–5. augmentation on regional oxygenation and metabolism after head injury.
30. Heiss WD, Kracht LW, Thiel A, et al. Penumbral probability thresholds of Crit Care Med 2005;33(1):189–95.
cortical flumazenil binding and blood flow predicting tissue outcome in 50. Abate MG, Trivedi M, Fryer TD, et al. Early derangements in oxygen and
patients with cerebral ischaemia. Brain 2001;124(Pt 1):20–9. glucose metabolism following head injury: the ischemic penumbra and
31. Leclerc X, Fichten A, Gauvrit JY, et al. Symptomatic vasospasm after pathophysiological heterogeneity. Neurocrit Care 2008;9(3):319–25.
subarachnoid haemorrhage: assessment of brain damage by diffusion and
perfusion-weighted MRI and single-photon emission computed tomography. A complete list of references for this chapter can be found online at
Neuroradiology 2002;44(7):610–6. www.expertconsult.com.
Section IV—Radiology 298.e1

References 25. Sette G, Baron JC, Mazoyer B, et al. Local brain haemodynamics and
oxygen metabolism in cerebrovascular disease: positron emission
1. Kampfl A, Franz G, Aichner F, et al. The persistent vegetative state after tomography. Brain 1989;112(Pt 4):931–51.
closed head injury: clinical and magnetic resonance imaging findings in 42 26. Powers WJ, Zazulia AR. PET in cerebrovascular disease. PET Clin 2010;5(1):
patients. J Neurosurg 1998;88(5):809–16. 83106.
2. Kampfl A, Schmutzhard E, Franz G, et al. Prediction of recovery from 27. Baron JC. Mapping the ischaemic penumbra with PET: implications for
post-traumatic vegetative state with cerebral magnetic-resonance imaging. acute stroke treatment. Cerebrovasc Dis 1999;9(4):193–201.
Lancet 1998;351(9118):1763–7. 28. Moustafa RR, Baron JC. Clinical review: imaging in ischaemic stroke—
3. Lovblad KO, Baird AE, Schlaug G, et al. Ischemic lesion volumes in acute implications for acute management. Crit Care 2007;11(5):227.
stroke by diffusion-weighted magnetic resonance imaging correlate with 29. Heiss WD. Best measure of ischemic penumbra: positron emission
clinical outcome. Ann Neurol 1997;42(2):164–70. tomography. Stroke 2003;34(10):2534–5.
4. Heiss WD, Huber M, Fink GR, et al. Progressive derangement of periinfarct 30. Heiss WD, Kracht LW, Thiel A, et al. Penumbral probability thresholds of
viable tissue in ischemic stroke. J Cereb Blood Flow Metab 1992;12(2): cortical flumazenil binding and blood flow predicting tissue outcome in
193–203. patients with cerebral ischaemia. Brain 2001;124(Pt 1):20–9.
5. Marchal G, Beaudouin V, Rioux P, et al. Prolonged persistence of substantial 31. Leclerc X, Fichten A, Gauvrit JY, et al. Symptomatic vasospasm after
volumes of potentially viable brain tissue after stroke: a correlative PET-CT subarachnoid haemorrhage: assessment of brain damage by diffusion and
study with voxel-based data analysis. Stroke 1996;27(4):599–606. perfusion-weighted MRI and single-photon emission computed
6. Powers WJ, Grubb RL, Jr., Darriet D, et al. Cerebral blood flow and cerebral tomography. Neuroradiology 2002;44(7):610–6.
metabolic rate of oxygen requirements for cerebral function and viability in 32. Coles JP, Minhas PS, Fryer TD, et al. Effect of hyperventilation on cerebral
humans. J Cereb Blood Flow Metab 1985;5(4):600–8. blood flow in traumatic head injury: clinical relevance and monitoring
7. Garnett MR, Blamire AM, Rajagopalan B, et al. Evidence for cellular correlates. Crit Care Med 2002;30(9):1950–9.
damage in normal-appearing white matter correlates with injury severity in 33. Jabre A, Babikian V, Powsner RA, et al. Role of single photon emission
patients following traumatic brain injury: a magnetic resonance computed tomography and transcranial Doppler ultrasonography in
spectroscopy study. Brain 2000;123(Pt 7):1403–9. clinical vasospasm. J Clin Neurosci 2002;9(4):400–3.
8. Higuchi T, Fernandez EJ, Maudsley AA, et al. Mapping of lactate and 34. Kincaid MS, Souter MJ, Treggiari MM, et al. Accuracy of transcranial
N-acetyl-L-aspartate predicts infarction during acute focal ischemia: in vivo Doppler ultrasonography and single-photon emission computed
1H magnetic resonance spectroscopy in rats. Neurosurgery 1996;38(1):121– tomography in the diagnosis of angiographically demonstrated cerebral
9; discussion 129–30. vasospasm. J Neurosurg 2009;110(1):67–72.
9. Signoretti S, Marmarou A, Tavazzi B, et al. N-Acetylaspartate reduction as a 35. Cooke D, Koppula B, Seiler D, et al. Semiquantitative software SPECT
measure of injury severity and mitochondrial dysfunction following diffuse analysis in aneurysmal subarachnoid hemorrhage-related vasospasm. Nucl
traumatic brain injury. J Neurotrauma 2001;18(10):977–91. Med Commun 2010;31(1):53–8.
10. Friedman SD, Brooks WM, Jung RE, et al. Quantitative proton MRS 36. Sarrafzadeh AS, Haux D, Ludemann L, et al. Cerebral ischemia in
predicts outcome after traumatic brain injury. Neurology aneurysmal subarachnoid hemorrhage: a correlative microdialysis-PET
1999;52(7):1384–91. study. Stroke 2004;35(3):638–43.
11. Beauchamp NJ, Jr, Barker PB, Wang PY, et al. Imaging of acute cerebral 37. Enblad P, Valtysson J, Andersson J, et al. Simultaneous intracerebral
ischemia. Radiology 1999;212(2):307–24. microdialysis and positron emission tomography in the detection of
12. Garnett MR, Corkill RG, Blamire AM, et al. Altered cellular metabolism ischemia in patients with subarachnoid hemorrhage. J Cereb Blood Flow
following traumatic brain injury: a magnetic resonance spectroscopy study. Metab 1996;16(4):637–44.
J Neurotrauma 2001;18(3):231–40. 38. Sarrafzadeh AS, Nagel A, Czabanka M, et al. Imaging of hypoxic-ischemic
13. Heiss WD, Herholz K. Brain receptor imaging. J Nucl Med penumbra with (18)F-fluoromisonidazole PET/CT and measurement of
2006;47(2):302–12. related cerebral metabolism in aneurysmal subarachnoid hemorrhage.
14. Small GW, Bookheimer SY, Thompson PM, et al. Current and future uses J Cereb Blood Flow Metab 2010;30(1):36–45. Epub 2009, Sep 23.
of neuroimaging for cognitively impaired patients. Lancet Neurol 39. Jost SC, Diringer MN, Zazulia AR, et al. Effect of normal saline bolus on
2008;7(2):161–72. cerebral blood flow in regions with low baseline flow in patients with
15. Veenith T, Coles JP. Anaesthesia for magnetic resonance imaging and vasospasm following subarachnoid hemorrhage. J Neurosurg
positron emission tomography. Curr Opin Anaesthesiol 2011;24(4):451–8. 2005;103(1):25–30.
16. Gunn RN, Lammertsma AA, Hume SP, et al. Parametric imaging of 40. Dhar R, Zazulia AR, Videen TO, et al. Red blood cell transfusion increases
ligand-receptor binding in PET using a simplified reference region model. cerebral oxygen delivery in anemic patients with subarachnoid hemorrhage.
Neuroimage 1997;6(4):279–87. Stroke 2009;40(9):3039–44. Epub 2009, Jul 23.
17. Klumpers UM, Veltman DJ, Boellaard R, et al. Comparison of plasma input 41. Bavetta S, Nimmon CC, White J, et al. A prospective study comparing SPET
and reference tissue models for analysing [(11)C]flumazenil studies. J Cereb with MRI and CT as prognostic indicators following severe closed head
Blood Flow Metab 2008;28(3):579–87. injury. Nucl Med Commun 1994;15(12):961–8.
18. Bullock R, Statham P, Patterson J, et al. The time course of vasogenic 42. Jacobs A, Put E, Ingels M, et al. Prospective evaluation of technetium-99m-
oedema after focal human head injury—evidence from SPECT mapping of HMPAO SPECT in mild and moderate traumatic brain injury. J Nucl Med
blood brain barrier defects. Acta Neurochir Suppl (Wien) 1990;51:286–8. 1994;35(6):942–7.
19. Shiina G, Onuma T, Kameyama M, et al. Sequential assessment of cerebral 43. Prayer L, Wimberger D, Oder W, et al. Cranial MR imaging and cerebral
blood flow in diffuse brain injury by 123I-iodoamphetamine single-photon 99mTc HM-PAO-SPECT in patients with subacute or chronic severe
emission CT. AJNR Am J Neuroradiol 1998;19(2):297–302. closed head injury and normal CT examinations. Acta Radiol 1993;34(6):
20. Della Corte F, Giordano A, Pennisi MA, et al. Quantitative cerebral blood 593–9.
flow and metabolism determination in the first 48 hours after severe head 44. Xu Y, McArthur DL, Alger JR, et al. Early nonischemic oxidative metabolic
injury with a new dynamic SPECT device. Acta Neurochir (Wien) 1997; dysfunction leads to chronic brain atrophy in traumatic brain injury.
139(7):636–41; discussion 41–2. J Cereb Blood Flow Metab 2010;30(4):883–94. Epub 2009, Dec 23.
21. Stamatakis EA, Wilson JT, Hadley DM, et al. SPECT imaging in head injury 45. Amen DG, Wu JC, Taylor D, et al. Reversing brain damage in former NFL
interpreted with statistical parametric mapping. J Nucl Med 2002;43(4): players: implications for traumatic brain injury and substance abuse
476–83. rehabilitation. J Psychoactive Drugs 2011;43(1):1–5.
22. Bergsneider M, Hovda DA, Lee SM, et al. Dissociation of cerebral glucose 46. Hattori N, Swan M, Stobbe GA, et al. Differential SPECT activation
metabolism and level of consciousness during the period of metabolic patterns associated with PASAT performance may indicate frontocerebellar
depression following human traumatic brain injury. J Neurotrauma 2000; functional dissociation in chronic mild traumatic brain injury. J Nucl Med
17(5):389–401. 2009;50(7):1054–61. Epub 2009, Jun 12.
23. Frackowiak RS, Jones T, Lenzi GL, et al. Regional cerebral oxygen utilization 47. Wortzel HS, Filley CM, Anderson CA, et al. Forensic applications of
and blood flow in normal man using oxygen-15 and positron emission cerebral single photon emission computed tomography in mild traumatic
tomography. Acta Neurol Scand 1980;62(6):336–44. brain injury. J Am Acad Psychiatry Law 2008;36(3):310–22.
24. Frackowiak RS, Lenzi GL, Jones T, et al. Quantitative measurement of 48. Lewine JD, Davis JT, Bigler ED, et al. Objective documentation of
regional cerebral blood flow and oxygen metabolism in man using 15O and traumatic brain injury subsequent to mild head trauma: multimodal
positron emission tomography: theory, procedure, and normal values. brain imaging with MEG, SPECT, and MRI. J Head Trauma Rehabil 2007;
J Comput Assist Tomogr 1980;4(6):727–36. 22(3):141–55.
298.e2 Section IV—Radiology

49. Johnston AJ, Steiner LA, Coles JP, et al. Effect of cerebral perfusion pressure 72. Heiss WD. PET in coma and in vegetative state. Eur J Neurol 2011;Jul 28.
augmentation on regional oxygenation and metabolism after head injury. Epub ahead of print.
Crit Care Med 2005;33(1):189–95. 73. Musch G. Positron emission tomography: a tool for better understanding
50. Abate MG, Trivedi M, Fryer TD, et al. Early derangements in oxygen and of ventilator-induced and acute lung injury. Curr Opin Crit Care 2011;
glucose metabolism following head injury: the ischemic penumbra and 17(1):7–12.
pathophysiological heterogeneity. Neurocrit Care 2008;9(3):319–25. 74. de Prost N, Tucci MR, Melo MF. Assessment of lung inflammation with
51. Diringer MN, Videen TO, Yundt K, et al. Regional cerebrovascular and 18F-FDG PET during acute lung injury. AJR Am J Roentgenol 2010;195(2):
metabolic effects of hyperventilation after severe traumatic brain injury. 292–300.
J Neurosurg 2002;96(1):103–8. 75. Bellani G, Messa C, Guerra L, et al. Lungs of patients with acute respiratory
52. Diringer MN, Yundt K, Videen TO, et al. No reduction in cerebral distress syndrome show diffuse inflammation in normally aerated regions:
metabolism as a result of early moderate hyperventilation following severe a [18F]-fluoro-2-deoxy-D-glucose PET/CT study. Crit Care Med 2009;
traumatic brain injury. J Neurosurg 2000;92(1):7–13. 37(7):2216–22.
53. Verweij BH, Muizelaar JP, Vinas FC, et al. Improvement in mitochondrial 76. Morris TA, Gerometta M, Yusen RD, et al. Detection of Pulmonary
dysfunction as a new surrogate efficiency measure for preclinical trials: Emboli With 99mTc-labeled anti-D-dimer (DI-80B3)Fab’ fragments
dose-response and time-window profiles for administration of the calcium (ThromboView). Am J Respir Crit Care Med 2011;Jun 16. Epub ahead of
channel blocker Ziconotide in experimental brain injury. J Neurosurg 2000; print.
93(5):829–34. 77. Dobrucki LW, Sinusas AJ. PET and SPECT in cardiovascular molecular
54. Clausen T, Zauner A, Levasseur JE, et al. Induced mitochondrial failure in imaging. Nat Rev Cardiol 2010;7(1):38–47. Epub 2009, Nov 24.
the feline brain: implications for understanding acute post-traumatic 78. Keidar Z, Gurman-Balbir A, Gaitini D, et al. Fever of unknown origin: the
metabolic events. Brain Res 2001;908(1):35–48. role of 18F-FDG PET/CT. J Nucl Med 2008;49(12):1980–5. Epub 2008, Nov 7.
55. Kawai N, Nakamura T, Tamiya T, et al. Metabolic disturbance without brain 79. Tzimopoulou S, Cunningham VJ, Nichols TE, et al. A multi-center
ischemia in traumatic brain injury: a positron emission tomography study. randomized proof-of-concept clinical trial applying [18F]FDG-PET for
Acta Neurochir Suppl 2008;102:241–5. evaluation of metabolic therapy with rosiglitazone XR in mild to moderate
56. Jiang Q, Chopp M, Zhang ZG, et al. The temporal evolution of MRI tissue Alzheimer’s disease. J Alzheimers Dis 2010;22(4):1241–56.
signatures after transient middle cerebral artery occlusion in rat. J Neurol 80. Tauscher J, Bagby RM, Javanmard M, et al. Inverse relationship between
Sci 1997;145(1):15–23. serotonin 5-HT(1A) receptor binding and anxiety: a [(11)C]WAY-100635
57. Diringer MN, Scalfani MT, Zazulia AR, et al. Cerebral hemodynamic and PET investigation in healthy volunteers. Am J Psychiatry 2001;158(8):
metabolic effects of equi-osmolar doses mannitol and 23.4% saline in 1326–8.
patients with edema following large ischemic stroke. Neurocrit Care 2011; 81. Stamatakis EA, Hetherington MM. Neuroimaging in eating disorders. Nutr
14(1):11–7. Neurosci 2003;6(6):325–34.
58. Nortje J, Coles JP, Timofeev I, et al. Effect of hyperoxia on regional 82. Parsey RV, Kegeles LS, Hwang DR, et al. In vivo quantification of brain
oxygenation and metabolism after severe traumatic brain injury: serotonin transporters in humans using [11C]McN 5652. J Nucl Med
preliminary findings. Crit Care Med 2008;36(1):273–81. 2000;41(9):1465–77.
59. Hutchinson PJ, Gupta AK, Fryer TF, et al. Correlation between cerebral 83. Östberg A, Virta J, Rinne JO, et al. Cholinergic dysfunction after traumatic
blood flow, substrate delivery, and metabolism in head injury: a combined brain injury: preliminary findings from a PET study. Neurology
microdialysis and triple oxygen positron emission tomography study. 2011;76(12):1046–50.
J Cereb Blood Flow Metab 2002;22(6):735–45. 84. Heiss WD, Kracht L, Grond M, et al. Early [(11)C]flumazenil/H(2)O
60. Hutchinson PJ, O’Connell MT, Seal A, et al. A combined microdialysis and positron emission tomography predicts irreversible ischemic cortical
FDG-PET study of glucose metabolism in head injury. Acta Neurochir damage in stroke patients receiving acute thrombolytic therapy. Stroke
(Wien) 2009;151(1):51–61; discussion 61. Epub 2008, Dec 20. 2000;31(2):366–9.
61. Bergsneider M, Hovda DA, Shalmon E, et al. Cerebral hyperglycolysis 85. Heiss WD, Kracht LW, Thiel A, et al. Penumbral probability thresholds of
following severe traumatic brain injury in humans: a positron emission cortical flumazenil binding and blood flow predicting tissue outcome in
tomography study. J Neurosurg 1997;86(2):241–51. patients with cerebral ischaemia. Brain 2001;124(Pt 1):20–9.
62. Vespa PM, McArthur DL, Xu Y, et al. Nonconvulsive seizures after 86. Heiss WD, Graf R, Fujita T, et al. Early detection of irreversibly damaged
traumatic brain injury are associated with hippocampal atrophy. Neurology ischemic tissue by flumazenil positron emission tomography in cats. Stroke
2010;75(9):792–8. 1997;28(10):2045–51.
63. Richards HK, Simac S, Piechnik S, et al. Uncoupling of cerebral blood flow 87. Baron JC. Mapping the ischaemic penumbra with PET: a new approach.
and metabolism after cerebral contusion in the rat. J Cereb Blood Flow Brain 2001;124(Pt 1):2–4.
Metab 2001;21(7):779–81. 88. Geeraerts T, Coles JP, Aigbirhio FI, et al. Validation of reference tissue
64. Wu HM, Huang SC, Hattori N, et al. Selective metabolic reduction in gray modelling for [11C]flumazenil positron emission tomography following
matter acutely following human traumatic brain injury. J Neurotrauma head injury. Ann Nucl Med 2011;25(6):396–405. Epub 2011, Apr 2.
2004;21(2):149–61. 89. Koizumi H, Fujisawa H, Kurokawa T, et al. Recovered neuronal viability
65. Vespa P, Bergsneider M, Hattori N, et al. Metabolic crisis without brain revealed by Iodine-123-iomazenil SPECT following traumatic brain injury.
ischemia is common after traumatic brain injury: a combined microdialysis J Cereb Blood Flow Metab 2010;30(10):1673–81. Epub 2010, Aug 4.
and positron emission tomography study. J Cereb Blood Flow Metab 90. Heiss WD, Grond M, Thiel A, et al. Permanent cortical damage detected by
2005;25(6):763–74. flumazenil positron emission tomography in acute stroke. Stroke 1998;
66. Bergsneider M, Hovda DA, McArthur DL, et al. Metabolic recovery 29(2):454–61.
following human traumatic brain injury based on FDG-PET: time course 91. Heiss WD, Grond M, Thiel A, et al. Tissue at risk of infarction rescued by
and relationship to neurological disability. J Head Trauma Rehabil 2001; early reperfusion: a positron emission tomography study in systemic
16(2):135–48. recombinant tissue plasminogen activator thrombolysis of acute stroke.
67. García-Panach J, Lull N, Lull JJ, et al. A voxel-based analysis of FDG-PET in J Cereb Blood Flow Metab 1998;18(12):1298–307.
traumatic brain injury: regional metabolism and relationship between the 92. Savic I, Ingvar M, Stone-Elander S. Comparison of [11C]flumazenil and
thalamus and cortical areas. J Neurotrauma 2011;28(9):1707–17. [18F]FDG as PET markers of epileptic foci. J Neurol Neurosurg Psychiatry
68. Provenzano FA, Jordan B, Tikofsky RS, et al. F-18 FDG PET imaging of 1993;56(6):615–21.
chronic traumatic brain injury in boxers: a statistical parametric analysis. 93. Richardson MP, Koepp MJ, Brooks DJ, et al. Benzodiazepine receptors in
Nucl Med Commun 2010;31(11):952–7. focal epilepsy with cortical dysgenesis: an 11C-flumazenil PET study. Ann
69. Kremer S, Nicolas-Ong C, Schunck T, et al. Usefulness of functional MRI Neurol 1996;40(2):188–98.
associated with PET scan and evoked potentials in the evaluation of brain 94. Tai YF, Piccini P. Applications of positron emission tomography (PET) in
functions after severe brain injury: preliminary results. J Neuroradiol 2010; neurology. J Neurol Neurosurg Psychiatry 2004;75(5):669–76.
37(3):159–66. Epub 2009, Sep 24. 95. Rojas S, Martin A, Arranz MJ, et al. Imaging brain inflammation with [(11)
70. Peskind ER, Petrie EC, Cross DJ, et al. Cerebrocerebellar hypometabolism C]PK11195 by PET and induction of the peripheral-type benzodiazepine
associated with repetitive blast exposure mild traumatic brain injury in 12 receptor after transient focal ischemia in rats. J Cereb Blood Flow Metab
Iraq war veterans with persistent post-concussive symptoms. Neuroimage 2007;27(12):1975–86.
2011;54 Suppl 1:S76–82. Epub 2010, Apr 10. 96. Banati RB, Newcombe J, Gunn RN, et al. The peripheral benzodiazepine
71. Bernat JL. Ethical issues in the treatment of severe brain injury: the impact binding site in the brain in multiple sclerosis: quantitative in vivo imaging
of new technologies. Ann N Y Acad Sci 2009;1157:117–30. of microglia as a measure of disease activity. Brain 2000;123(Pt 11):2321–37.
Section IV—Radiology 298.e3

97. Cagnin A, Brooks DJ, Kennedy AM, et al. In-vivo measurement of activated 104. Yu EY, Mankoff DA. Positron emission tomography imaging as a cancer
microglia in dementia. Lancet 2001;358(9280):461–7. biomarker. Expert Rev Mol Diagn 2007;7(5):659–72.
98. Cagnin A, Myers R, Gunn RN, et al. In vivo visualization of activated glia 105. Harris RS, Schuster DP. Visualizing lung function with positron emission
by [11C] (R)-PK11195-PET following herpes encephalitis reveals projected tomography. J Appl Physiol 2007;102(1):448–58. Epub 2006, Oct 12.
neuronal damage beyond the primary focal lesion. Brain 2001;124 106. Chen Q, Liu W, Li H, et al. Molecular imaging in patients with mood
(Pt 10):2014–27. disorders: a review of PET findings. Eur J Nucl Med Mol Imaging
99. Ramlackhansingh AF, Brooks DJ, Greenwood RJ, et al. Inflammation after 2011;38(7):1367–80. Epub 2011, Apr 12.
trauma: microglial activation and traumatic brain injury. Ann Neurol 107. Brooks DJ, Pavese N. Imaging biomarkers in Parkinson’s disease. Prog
2011;70(3):374–83. Neurobiol 2011;Aug 30. Epub ahead of print.
100. Landt J, D’Abrera JC, Holland AJ, et al. Using positron emission 108. Sacher J, Neumann J, Fünfstück T, et al. Mapping the depressed brain: a
tomography and carbon 11-labeled Pittsburgh compound B to image brain meta-analysis of structural and functional alterations in major depressive
fibrillar β-amyloid in adults with down syndrome: safety, acceptability, and disorder. J Affect Disord 2011;Sep 2. Epub ahead of print.
feasibility. Arch Neurol 2011;68(7):890–6. Epub 2011, Mar 14. 109. Melrose RJ, Ettenhofer ML, Harwood D, et al. Cerebral metabolism,
101. Vallabhajosula S. Positron emission tomography radiopharmaceuticals for cognition, and functional abilities in Alzheimer disease. J Geriatr Psychiatry
imaging brain beta-amyloid. Semin Nucl Med 2011;41(4):283–99. Neurol 2011;24(3):127–34.
doi:10.1053/j.semnuclmed.2011.02.005. 110. Yakushev I, Schreckenberger M, Müller MJ, et al. Functional implications of
102. Barthel H, Gertz HJ, Dresel S, et al. Cerebral amyloid-β PET with hippocampal degeneration in early Alzheimer’s disease: a combined DTI
florbetaben (18F) in patients with Alzheimer’s disease and healthy controls: and PET study. Eur J Nucl Med Mol Imaging 2011;Jul 27. Epub ahead of
a multicentre phase 2 diagnostic study. Lancet Neurol 2011;10(5):424–35. print.
Epub 2011, Apr 8. 111. Kawai N, Maeda Y, Kudomi N, et al. Correlation of biological aggressiveness
103. Koivunen J, Scheinin N, Virta JR, et al. Amyloid PET imaging in patients assessed by 11C-methionine PET and hypoxic burden assessed by
with mild cognitive impairment: a 2-year follow-up study. Neurology 18F-fluoromisonidazole PET in newly diagnosed glioblastoma. Eur J Nucl
2011;76(12):1085–90. Epub 2011, Feb 16. Med Mol Imaging 2011;38(3):441–50. Epub 2010, Nov 12.
V
Chapter
30  

Neurosonology: Transcranial
Doppler and Transcranial
Color-Coded Duplex
Sonography
Jaroslaw Krejza and Michal Arkuszewski

Introduction Transcranial Doppler and TCCS Techniques


Doppler ultrasonography is a technique to measure the veloc- Structures imaged by B-mode are displayed proportionally to
ity and pulsatility of blood flow within the intracranial and the intensity of returning echoes, whereas flow velocity is
the extracranial arteries. Transcranial Doppler (TCD) ultraso- detected in Doppler mode if there is a difference in frequency
nography is frequently used for the clinical evaluation of cere- between emitted and returning echoes.3 The magnitude of the
bral vasospasm following subarachnoid hemorrhage (SAH), frequency shift (ΔF) depends on the ultrasound transmission
to evaluate cerebral autoregulation (CA), detect major arterial velocity in the insonated tissue (C), the relative velocities of
occlusion, monitor recanalization in acute stroke, monitor the reflector (blood, V), and the frequency of the source (Fo).
cerebral circulation during major cardiovascular procedures, The observed frequency shift (ΔF) is ΔF = 2 VFo/C.3,5 The shift
and assess hemodynamics in patients with intracranial hyper- is measured only for that component of motion occurring
tension among other indications.1,2 Technologic advances along the axis of the ultrasound beam. Therefore absolute
such as transcranial color-coded duplex sonography (TCCS) velocity measurements require that a correction be made for
have made TCD more reliable and accurate in its detection of the angle (θ) between the vessel and the beam as follows: V =
hemodynamic abnormalities. Newer applications of this tech- ΔFC/(2 Fo cos θ). In pulsed wave Doppler, a transducer gener-
nology include therapeutic use of ultrasound in the acute ates ultrasound pulses and detects returning echoes. Assuming
stroke setting. that the speed of transmission of ultrasound in human tissues
is a constant, the time delay between the emitted pulse and
the returning echo enables the sampled structure’s depth to
Technical Aspects of Transcranial be determined. The pulse’s duration and repetition frequency
Doppler impose limits on the maximum velocity that can be measured.
Anatomy is not displayed, however. Identification of intracra-
Basics of Ultrasound nial arteries in conventional TCD relies therefore on the a
Ultrasonic waves that enter human tissue are transmitted, priori defined depths of the sample volume and the direction
absorbed, reflected, and scattered.3,4 Transmission properties of flow.
of a tissue depend on its density and elasticity. Density and Duplex Doppler imaging combines pulsed-wave Doppler
speed of propagation of ultrasound (US) waves determine a with two-dimensional, real-time B-mode imaging, which
tissue’s acoustic impedance. The larger the difference in acous- allows precise placement of the sample volume in the vessel.
tic impedance between tissues, the more US waves are reflected. Optimal angle correction for velocity calculations can be per-
Reflection further depends on the angle of insonation, and formed, as the course of the vessel in relation to the US beam
stronger echoes are received when the angle of insonation is is visually depicted. Color-coded duplex sonography is the
zero. Structures imaged by US B-mode, the two-dimensional most commonly used technology today for carotid imaging
visualization of the internal organs and structures on the gray- and TCCS (Fig. 30.1). Red indicates blood flow toward the
scale image, are displayed proportionally to the intensity of transducer, whereas blue represents flow away from the trans-
returning echoes.3,4 ducer. High-flow velocities are depicted with increasing
300 © Copyright 2013 Elsevier Inc. All rights reserved.
Section V—Cerebral Blood Flow 301

Sonographic
M2 MCA A1 ACA M1 MCA probe Temporal
M2 MCA window
M1 MCA P2 PCA
A2 ACA

P2 PCA
A B
Fig. 30.1  A, A typical semiaxial image of M1 and M2 segments of the right MCA superimposed on a sector-shaped conventional B-mode image.
The sample volume is precisely placed on a green color spot related to an aliasing artifact, which indicates the site of highest flow acceleration in the
M1 segment. The angle between the course of the vessel in relation to the ultrasound beam is measured by an electronic cursor. This approach allows
the angle-corrected flow velocity measurements from the waveform displayed below the B-mode image to be obtained. B, The complex spatial
relationships between ultrasound beam, courses of M1 and M2 MCA, A2 ACA and P2 PCA, and the site of temporal window. ACA, Anterior cerebral
artery; MCA, middle cerebral artery; PCA, posterior cerebral artery.

brightness, thus the presence, direction, and flow disturbances therapeutic applications. The thermal index (TI), provided in
can be quickly identified. US machines estimates the risk from heat. When the TI is
The insonation angle is important in flow velocity mea- greater than 1, the risks of US should be weighed against the
surements with TCCS because trajectory of a particular vessel benefits.10 The exposure has to be minimized despite reports
in the intracranial space is unpredictable.6,7 Thus if the veloc- that short-term monitoring does not increase temperature at
ity measurements are adjusted for the angle, the velocity the temporal window in vivo.13,14
values are closer to the true values because the vessel’s geo-
metric factor is accounted for. If a measured velocity is
170 cm/second and the angle is 20 degrees, the true velocity Nonthermal Effects
is 181 cm/second; however, if the angle is 40 degrees, the true US can also produce various mechanical effects such as cavita-
velocity is 222 cm/second. Such difference, caused by errone- tion, pressure amplitude, force, torque, and acoustic stream-
ous flow velocity measurement, can change management of a ing.9,15 Cavitation occurs when US passes through an area that
patient. TCD/TCCS cannot be performed in about 11% of contains a cavity, such as a gas bubble. US can cause the bubble
patients because the acoustic window in the temporal bone is to expand and contract rhythmically. When bubbles pulsate,
weak or absent.8 Also following a pterional craniotomy the they send secondary US waves in all directions. These second-
acoustic window may be degraded because of subtemporal ary waves can actually improve US imaging. If the bubbles
bone work or the presence of Gelfoam in the epidural space. contract toward the point of collapsing, they can build up very
Frequencies used for neurosonology range between 1 and high temperatures and pressures for a few tens of nanosec-
3.5 MHz for TCD/TCCS and 7.5 and 16 MHz for carotid onds. These high temperatures and pressures can produce free
imaging. radicals and other toxic compounds that, although unlikely,
could theoretically cause genetic damage.16 The rapid con­
traction of bubbles can also cause microjets of liquid that
Bioeffects and Safety of Ultrasound can damage cells. The safety guidelines for diagnostic US
Diagnostic US can produce heat that may be hazardous to are designed to prevent cavitations to occur.9,10 US-induced
sensitive organs.9 Nonthermal effects, such as pressure changes changes in pressure, force, torque, and streaming, in turn, can
and mechanical disturbances, in tissue have not been demon- cause audible sounds, electrical changes in cell membranes
strated in humans.10 US used in therapy, however, can cause that make them more permeable to large molecules, move-
both substantial temperature increase and mechanical damage ment and redistribution of cells in liquid, and cell damage.17
in the tissue.10-12 In the experimental setting using promyelocytic leukemia
cells, sonoporation may lead to antiproliferation effects
including cell-cycle arrest and apoptosis through disrupting
Thermal Effects various cell signaling pathways.18 In liquids, US causes a type
Denser tissue absorbs more heat from US, thus fluid does not of stirring action called acoustic streaming. As the acoustic
heat very much, whereas bone heats the most, and on bone pressure of US increases, the flow of liquid speeds up. When
surface the heat accumulation can be up to 50 times faster the streaming liquid comes near a solid object, shearing may
than in soft tissues. This heating effect can be of importance, occur, which can damage platelets and lead to abnormal blood
particularly with use of long-term TCD monitoring and its clotting.
302 Section V—Cerebral Blood Flow

resulting decrease in extracellular pH causes vasodilation;


Effects of Ultrasound Contrast Agents hypocapnia leads to vasoconstriction and CBF decrease. The
These agents usually take the form of stable gas-filled micro- importance of neurogenic regulation in the control of CBF is
bubbles (MBs) that can produce cavitations or microstream- a matter of debate. The cerebral vessels are innervated by an
ing, the risk of which increases with the mechanical index extrinsic system such as nerve fibers that originate in ganglia
value. Additionally, a radiation force produced by the acoustic belonging to sympathetic, parasympathetic, and sensory
wave, considered as noncavitational and nonthermal mecha- ganglia, and an intrinsic system that is composed from nerves
nisms of ultrasonic bioeffects, may act on particles and originating in different parts of brain such as the nucleus
microbubbles.19 basalis, locus coeruleus, and raphe nucleus.29,30 Sympathetic
stimulation constricts large cerebral arteries, but CBF does not
decrease because of immediately compensated vasodilation
Cerebral Vascular Reactivity and of resistance arterioles.31 Sympathetic innervation seems to
Transcranial Ultrasound Testing of protect the brain against MAP increases because sympathetic
Cerebral Blood Flow Regulation activation shifts the upper and lower limits of CA toward a
higher MAP. Activation of the parasympathetic vascular nerve
Cerebral Autoregulation fibers causes vasodilation.32 Vascular fibers belonging to the
CA is the ability of brain microvasculature to maintain cere- intrinsic system33-35 may modulate vascular tone directly or
bral blood flow (CBF) relatively constant despite wide varia- through stimulation of perivascular interneurons and glial
tions in cerebral perfusion pressure (CPP). In the healthy cells.36,37 However, the significance of this system is being
state CA keeps CBF relatively constant within the range of elucidated.34,35
mean arterial blood pressure (MAP) between 60 and 150 mm
Hg.20 However, the upper and lower limits of CA can be
shifted up or down by endogenous as well as exogenous Assessment of Cerebral Blood Flow
factors. Sympathetic nervous system stimulation and angio- Regulation with Ultrasound
tensin II can shift the upper and lower limits of CA up toward The CA can be evaluated by measuring relative blood flow
higher pressures, whereas antihypertensive medications have changes in response to the change in the MAP using “static”
opposite effects.21,22 Patients who have untreated hypertension or “dynamic” approaches.2,38-41 In the static method, CA effi-
have limits of regulation set at a higher level compared with ciency (i.e., the overall change in cerebral arterial resistance
healthy people21; thus overzealous antihypertensive treatment [CAR] induced by changes in MAP) is assessed under steady-
may lead to a dangerous CBF reduction at a relatively high state conditions. In this method the first cerebral blood flow
MAP. The mechanisms of CA are complex and not fully velocity (CBFV) measurement obtained at a constant baseline
understood and appear to be maintained by three different MAP is followed by another steady-state measurement that is
control pathways: vasogenic, metabolic, and neurogenic.23 taken after the CAR to a change in MAP. CA functions prop-
The vasogenic mechanism of CA is based on the intrinsic erly if the MAP change within the lower and upper limits of
ability of cerebral vessels to respond to changes in the wall’s CA, estimated for healthy humans, does not affect CBFV.
shear stress and transluminal pressure.24 An increase in Dynamic methods use rapid MAP changes and analyze the
transluminal pressure activates vascular smooth muscle instantaneous CBFV in the process.38 Systemic hemodynamic
cells, leading to vasoconstriction,25,26 whereas increased shear changes may be induced using rapid leg cuff deflation, pro-
stress27 triggers vasodilation.24 In metabolic regulation, cere- gressive lower body negative pressure, the Valsalva maneuver,
brovascular resistance (CVR) is modified by PaCO2, PaO2, and deep breathing, ergometric exercise, or head-down tilting
release of adenosine and potassium ions from neurons in (Fig. 30.2). Dynamic methods are able to assess both efficiency
response to insufficient blood supply.28 Hypercapnia and the and latency of the CA response, that is, the overall change in

A B

Fig. 30.2  Transcranial Doppler (TCD) tracings that illustrate dynamic autoregulation testing based on flow velocity measurements in the
middle cerebral artery during head-down tilting in a healthy volunteer. A, In the supine position mean blood flow velocity is 70 cm/second;
pulsatility index is 0.80; systolic and diastolic blood pressure are 120 mm Hg and 78 mm Hg, respectively; and heart rate is 68 beats per minute. B, While
the patient is in an upright position, mean blood flow velocity decreased to 62 cm/second; the pulsatility index increased to 1.05; blood pressure
changed to 125 mm Hg and 70 mm Hg; and heart rate increased to 75 beats per minute. (Courtesy the Department of Neurology, Ernst Moritz Arndt University
of Greifswald, Germany.)
Section V—Cerebral Blood Flow 303

CAR and the time in which the CAR change is achieved. The define an abnormal response to acetazolamide, including: (1)
use of systemic hypotension to challenge the CA response is greater than 5% decrement in absolute CBF, (2) a less than
limited, however, because pharmacologically induced hypo- 10% increment in absolute CBF, (3) an absolute change of less
tension could result in ischemic injury in patients with already than 10 mL/100 g/minute, and (4) a value greater than two
insufficient CBF. standard deviations below control values. Acetazolamide is
Two commonly used, but somewhat less dynamic methods generally well tolerated in a dose less than 1000 mg. The most
of physiologic challenge of CA that carry much less risk of commonly reported side effects include transient circumoral
ischemia include administration of acetazolamide and sys- numbness, paresthesias, and headaches.47
temic PaCO2 manipulations. Acetazolamide is a carbonic Manipulation of systemic partial CO2 pressure (PaCO2)
anhydrase inhibitor that acts as a potent vasodilator when also can be used to test CVR. Low-level hypercarbia results
administered intravenously in a dose of 15 to 18 mg/kg in a reproducible CBF increase between 0.01 and 0.02 mL/g/
of body weight (standard dose 1000 mg).42 Acetazolamide min for each 1 mm Hg PaCO2 increase. The effect is rapid
slowly penetrates the blood-brain barrier, where it reversibly and quickly reversible.51 CO2 manipulation is performed
inhibits carbonic anhydrase that catalyzes the conversion of using several techniques, including: (1) breath holding or
bicarbonate and hydrogen ion to water and carbon dioxide hyperventilation, (2) re-breathing, or (3) inhalation of 3% to
(CO2). Acetazolamide decreases the production of bicarbon- 5% CO2. The effect of altered CO2 tension on CBF is quanti-
ate and results in a decrease in the extracellular pH in the fied either as a fixed effect for a particular CO2 manipulation
brain.43,44 This induced acidosis results in vasodilation,45,46 and (similar to the quantification of acetazolamide effect) or as
CBF increase,46 whereas MAP, heart rate (HR), and respiratory the slope of the CO2-CBF relationship according to the fol-
rate are unaffected.47 The effect on CBF may be seen within lowing formula52:
the first 5 minutes after administration.48 In healthy subjects, CVR = [(CBFpost-CO2 − CBFpre-CO2 )/(CBFpre-CO2 )] ×
peak CBF augmentation of about 30% to 60% occurs at
[100/(Pa pre-CO2 – Pa post-CO2 )]
approximately 10 minutes after bolus intravenous administra-
tion and changes little over the next 20 minutes (Fig. 30.3).49 The combined use of breath holding and hyperventilation
This response can be blunted in an older population.50 The tests allows assessment of the full range of vasomotor reactivity
percentage of increase in CBF after acetazolamide administra- (VMRr), both during hypercarbia (vasodilation) and hypo­
tion may be used to define cerebrovascular reserve as follows: carbia (vasoconstriction). The relative difference between
minimal and maximal CBF defines VMRr as follows (Fig. 30.4):
CVR = (CBFpost − CBFpre )/CBFpreacetazolamide × 100%
Although a global, symmetric increase in CBF by about VMRr = [(breath holding CBF − hyperventilation CBF)/
30% is accepted as normal, a variety of criteria are used to rest CBF] × 100%

A B
Fig. 30.3  Blood flow velocity measurements taken from the right middle cerebral artery with transcranial color-coded duplex sonography before
(A) and after (B) administration of 1000 mg acetazolamide in a healthy volunteer. The studies show healthy autoregulation expressed by a significant
(71%) increase in the mean flow velocity.

MCA velocity at rest MCA velocity after MCA velocity after


A B hyperventilation C breath holding

Fig. 30.4  Transcranial Doppler (TCD) tracings that show vasomotor reactivity range (78%) in a healthy volunteer determined by TCD velocity
measurements in the middle cerebral artery (MCA) (A) at rest mean blood flow velocity (BFV) is 60 cm/second, (B) after hyperventilation mean BFV
is 40 cm/second, and (C) after breath holding, mean BFV is 86 cm/second. (Courtesy Dr. G. Kozyra, the Department of Neurology, Medical University of Gdańsk,
Poland.)
304 Section V—Cerebral Blood Flow

The CBF increase in hypercarbia also can by described as a between the MCA velocity after release of compression, and
breath holding index (BHI): the velocity before onset of compression and SA, which is
the strength of CA calculated by normalizing the THRR for
BHI = [(breath holding CBF − rest CBF)/rest CBF)]/
changes in the MCA CBFV at the onset of compression. In
time of breath holding CBF] ×100%
theory, the THR test assesses both the gradient and the limits
In clinical practice, the magnitude of CBF changes with CO2 of the CA plateau without differentiating between the two.
manipulation is smaller than that achieved with acetazol- The test has been used to study anesthetic agents and in crit-
amide. The average increase in CVR is 1.1% to 2.9%. With 5% ical care. The coefficient of variation less than 10% is much
CO2 inhalation, the CBF response observed usually is approxi- lower than other CA tests, making it suitable for compari-
mately half of the response observed with acetazolamide. sons. The main advantages are reproducibility, simplicity,
However, the correlation between CBF changes produced by and lack of pharmacologic intervention. There is a very small
each technique varies from poor to moderate.53-55 CO2 manip- risk of detaching carotid artery atheroma, but various studies
ulation also may itself alter systemic MAP; an increase of about have demonstrated a good safety record even in at-risk
10 mm Hg that may be induced with 5% CO2 then may obfus- patients.
cate evaluation of the CO2-mediated CBF response.53 However,
CO2 manipulation is easily performed, generally well toler-
ated, and has no long-term effects. The rapid response to CO2 Influence of Anesthetic Agents
is particularly suitable for TCD or TCCS measurements. on Intracranial Blood Velocity
An important phenomenon that can affect the results of CA
testing is that of “steal,”49 which is created by the interaction Induction Agents
between regions of different cerebrovascular response and the Intravenous (IV) induction agents depress cerebral metabolic
systemic effects of the agents used for physiologic or pharma- rate of oxygen (CMRO2) without affecting coupling between
cologic challenge. Areas that have normal CA will show vaso- flow and metabolism, thus CBF and subsequently flow veloc-
dilation, lowering the perfusion pressure in the larger blood ity in the MCA decreases in a dose-dependent fashion.61,62
vessels supplying both normal and abnormal areas. This lower Propofol has little effect on CA or cerebrovascular reactivity
perfusion pressure results in a decrease in blood flow in the to carbon dioxide (CRCO2), although hypercapnia may impair
regions that are already maximally dilated. CA at around 8 kPa.63-65 Ketamine tends to increase MCA
velocity because of an “excitatory effect”66; however, when the
effect is blunted by preexisting anesthesia, ketamine appears
Assessment of CA Functionality with TCD to decrease MCA velocity. Volatile anesthetic agents can
and TCCS decrease MCA velocity by reducing CMRO2, but also can
Implementation of TCD/TCCS is based on the premise that increase velocity by direct vasodilation.67-69 The net effect
CBF changes will be reflected only by changes in CBFV in depends largely on the agent and the dose used. When equi-
major brain arteries. If CA is intact, changes in MAP do not potent clinical doses are used, sevoflurane has little effect on
affect CBFV, because of compensatory change in peripheral MCA velocity and CA; desflurane has the greatest effect, and
arterial resistance. Static measures of CA are the correlation isoflurane an intermediate effect.70-74 It should be noted that
coefficient (r) between CBFV and mean MAP as well as the it is a dose-related phenomenon.
“index of static autoregulation”(sARi), which is a ratio of Nitrous oxide at concentrations of 30% to 60% increases
percentage of change in CVR to percentage of change in flow velocity in MCA and CBF.64,75 It may also disrupt CA
MAP (sARi = % CVR/% MAP).56,57 For perfect CA the r when added to volatile anesthetic agents or propofol.76,77 This
would equal 0 and sARi would be 1, whereas complete lack of has been thought to be a result of excitation associated with
CA would yield an r and sARi of 1 and 0, respectively. The low dose anesthesia. However Matta et al64 observed increased
most commonly proposed threshold between normal and MCA velocity when nitrous was added to isoelectric EEG
impaired CA for both r and sARi is 0.5.56,58 Evaluation of CA doses of propofol. Muscle relaxants appear not to change flow
in dynamic methods is based on a “dynamic autoregulation velocity in MCA in patients with neurologic injury.78,79
index” (dARi). This is the dynamic equivalent of sARi and is Opioids at moderate doses have no significant effect on MCA
defined as dARi equals (ΔCVR/ΔT)/ ΔMAP, where ΔT is the velocity; however, at high doses they reduce flow velocity by a
time when the CA response occurs.57,59 Thus dARi not only presumed central mechanism80,81 unless they cause seizures,
evaluates the overall change in CBFV induced by a change in which would presumably be associated with hyperemia. Vaso-
MAP, but also quantifies the time dependence of a CA active agents such as norepinephrine,82 dobutamine, and
response. A value of dARi that equals 0 represents the absence dopexamine83 have been shown to have no effect on CA and
of CA, whereas a value of 9 corresponds to an efficient CA CRCO2 in volunteers and in patients anaesthetized with iso-
response. The proposed threshold between normal and flurane and propofol.84 Glyceryl trinitrate appears not to
impaired CA is 5.57,60 Static and dynamic methods of evalua- affect CA.85
tion of CA yield similar results.57
In the transient hyperemic response (THR) test, a brief
compression (3-10 seconds) of the ipsilateral common
Limitations of TCD and TCCS
carotid artery (CCA) results in a sudden reduction in the TCD measures velocities in large arteries, thus mapping of
middle cerebral artery (MCA) velocity due to distal vasodila- regional impairment within the brain is not possible. Regional
tion, if CA is intact. Subsequently a transient increase in the areas of impaired CA in an artery distribution may be missed
MCA velocity is seen on release of the compression. Two when averaged with areas that are better perfused. Further-
CA indices have been described: THRR, which is the ratio more, correlation between flow velocities and CBF is weak in
Section V—Cerebral Blood Flow 305

some patients, mostly because of collateral circulation.86,87 SAH patients, such as recurrent hemorrhage, hydrocephalus,
Assumptions about the changes in flow only hold if the arte- metabolic disorders, and seizures can produce similar neuro-
rial diameter and angle of insonation remains constant. logic symptoms.88
Digital subtraction angiography (DSA) is the gold standard
Applications of Transcranial test to diagnose cerebral VSP, but this method is an impractical
monitoring tool because it is invasive and carries a small risk
Doppler Ultrasonography in of stroke, renal injury, and other complications.90 Other
Neurocritical Care Monitoring methods, such as computed tomography (CT) and magnetic
resonance angiography, xenon-enhanced computed tomogra-
Diagnosis and Monitoring phy (Xe-CT), and CT perfusion are discussed in Chapters 26
of Cerebral Vasospasm through 28. These techniques may provide useful information
Cerebral vasospasm (VSP) contributes to morbidity and mor- but there are practical limitations to their daily use as a moni-
tality among patients after SAH.88 Medical treatment of VSP toring tool.91,92
with induced hypertension, hemodilution, and iso- or hyper- The knowledge that blood flow velocity is increased in a
volemia (3-H) can help improve CBF. However, use of these constricted vessel has led to the use of TCD to detect and
therapies can be associated with increased cerebral edema or monitor cerebral VSP at bedside.2, 93 However, TCD studies are
hemorrhagic transformation in areas of infarction, congestive operator dependent, and the accuracy of TCD may differ from
heart failure, or pulmonary edema and so are best used when one study to another. These variations have raised concerns
the diagnosis of VSP is made rather than prophylactically.88,89 about the utility of TCD to reliably detect VSP, particularly in
When medical treatment is unsuccessful, endovascular meth- vessels other than MCA.94,95 Generally mild or severe vaso-
ods (e.g., intra-arterial infusion of vasodilative agents, or bal- spasm in the MCA can be consistently diagnosed, and the
loon angioplasty) may restore flow. The outcome of patients diagnostic accuracy can be improved using TCCS rather than
who develop VSP and in particular delayed cerebral ischemia TCD.95-99 When using TCCS, 80% of DSA-diagnosed cases of
(DCI) associated with VSP is improved with early treatment. mild narrowing and 92% of cases of severe MCA narrowing
However, when to initiate various interventions often may be can be properly classified. Age and sex correction of flow
uncertain because the diagnosis and monitoring of VSP may velocity further increases TCCS performance, especially for
be difficult and unreliable when based solely on the neurologic the diagnosis of less severe MCA spasm.96 The improved accu-
examination. Furthermore, other complications common in racy of TCCS is associated with its ability to image arteries,

A B

C D
Fig. 30.5  Transcranial color-coded duplex sonography (TCCS) and transcranial Doppler (TCD) tracings that show the importance of the
angle of insonation, effect of proper placement of the sample volume on flow velocity measurements, and diagnostic value of the ratio
of intracranial middle cerebral artery (MCA) to the ipsilateral extracranial internal carotid artery in a subarachnoid hemorrhage (SAH)
patient. A, The sample volume is placed in the M2 segment of the right MCA at a distance of 34 mm from the probe and an angle of 6 degrees;
the mean velocity is 217 cm/second. B, The sample is placed at the distance of 36 mm in the same artery segment, and the angle is 43 degrees; the
velocity obtained is 393 cm/second, an increase of 181%. In both images the sample is placed on the site of aliasing artifact indicated by blue. With
conventional TCD the change in blood flow velocity may be missed if the sample is placed at the distance of 45 to 55 mm, usually used in MCA
interrogation. C, The mean flow velocity in the extracranial internal carotid artery is 27 cm/second; thus the velocity ratios are 8.04, and 14.56,
respectively for A and B measurements. These findings are consistent with severe vasospasm identified in the M2 segment of the MCA during
angiography (D, solid arrow). There also is severe vasospasm of A1 segment of the ACA (dashed arrow).
306 Section V—Cerebral Blood Flow

to position the sample volume in the specific site of the resistance, may be low when there is symptomatic large vessel
investigated artery, and to determine the angle between the vasospasm (i.e., there is compensatory distal vasodilation for
artery and the US beam (Fig. 30.5).99,100 Unlike the MCA, the hypoperfusion).107
the accuracy of TCD in the anterior cerebral artery (ACA),
posterior cerebral artery (PCA), internal carotid artery (ICA),
basilar artery (BA), and vertebral artery (VA) is not as well Middle Cerebral Artery Vasospasm
studied and accuracy appears low. The accuracy of TCD and TCCS to detect VSP is best deter-
TCD or TCCS evaluations are valuable in the day-to-day mined for MCA; TCCS perhaps shows better agreement with
care of SAH patients to detect VSP and evaluate therapeutic DSA than TCD.95-99,108 A mean blood flow velocity (BFV)
effects.101-103 A baseline study should be obtained at admission greater than 200 cm/second, a rapid rise in flow velocities
when the probability of large artery spasm is still relatively low, (>50 cm/second/day), or Lindegaard ratio (VMCA : VICA) greater
to establish reference values to which subsequent measure- than 6 indicate severe arterial narrowing.94,109,110 VSP is unlikely
ments can be compared and then be repeated daily, par­ if the mean BFV is less than 120 cm/second. However, about
ticularly during the period of high risk for VSP. Serial 60% of patients have velocities that fall between these two
measurements are useful because a rapid increase in flow values. In addition, negative results do not exclude the pres-
velocity (>65 cm/second over 24 hours) is associated with a ence of VSP. The best Doppler parameter is peak-systolic
poorer outcome, and is suggested as an indication to consider velocity, and a threshold of 182 cm/second corresponds to
induced hypertension (i.e., symptomatic vasospasm or DCI is maximal diagnostic accuracy. TCCS efficiency, sensitivity,
likely). In these patients the velocity ratios (VMCA : VICA, specificity, positive predictive value (PPV), and negative pre-
VtICA : VICA), or Lindegaard ratio that compares intra- to extra- dictive value (NPV) are reported to be 92%, 86%, 93%, 73%,
cranial blood flow velocities, should be calculated because the and 97%, respectively.95 The VMCA : VICA ratio (Lindegaard
velocity ratios are not altered by the therapy and so reflect index104) can help differentiate patients with high intracranial
arterial narrowing. A ratio of less than 3 is rarely found in flow velocities associated with hyperemia from those with a
patients with VSP, and ratios of greater than 6 may distinguish BFV increase from VSP (see Fig. 30.5). Studies suggest that the
moderate from severe MCA VSP.104 Several factors including overall accuracy of the VMCA : VICA ratio in the diagnosis of mild
age and sex, course of the artery, increased ICP and CPP, MAP, and moderate to severe MCA narrowing is better than the
hematocrit (Hct), PaCO2, temperature, presence and pattern respective accuracy of velocity measurements alone.98 A value
of collateral flow, therapeutic interventions (e.g., hypervol- 3.6 of the ratio appears to be threshold to diagnose mild (up
emia or use of vasopressors with impaired CA), presence of to 25% narrowing) spasm in the M1 segment of the MCA,
impaired CA, and multiple segment narrowing influence flow whereas a threshold of 4.4 indicates moderate to severe spasm
velocities and should be considered. For example, correction (>25% artery narrowing).98 The thresholds are higher than the
for PaCO2 and Hct may be required if artificial ventilation or upper normal reference limits of the VMCA : VICA ratio, calcu-
hemodilution is in progress, and when interpreting Doppler lated on the basis of the mean velocity.111 Neural networks can
results, global and local ICP and CA state should be taken into be used to improve TCCS diagnostic performance; classifica-
account. High impedance indices may suggest the presence of tion accuracy is 92% in moderate to severe spasm detection,
localized or generalized increased ICP, and usually are associ- and 87% in spasm of other grades.112
ated with poorer outcome.98 CA also may be impaired in many
patients with VSP (Fig. 30.6).105,106 Finally, the pulsatility index
(PI), which provides information about distal vascular Vasospasm of Other Arteries
TCD-based diagnosis for vessels other than the MCA is less
well defined and generally less accurate.108 Visualization of the
Breath ACA with TCD or TCCS is more difficult than that of the
holding MCA.6 TCD studies based on relatively small sample sizes
30 sec show a sensitivity of 42% and specificity of 76% when 120 cm/
second mean velocity is used as a threshold to detect ACA
Vasospasm spasm. A TCCS threshold of 75 cm/second mean BFV is
associated with 71% sensitivity and 85% specificity.97 False-
negative results for the ACA may be explained by collateral
flow through the anterior communicating artery (ACoA) and
difficulties with angiographic differentiation of ACA hypopla-
sia from vasospasm. The VACA : VICA ratio can be helpful to
Control differentiate spasm from the normal artery; the ratio VACA : VICA
varies between 0.54 and 2.55.104,111 In practice, however, a diag-
Fig. 30.6  Transcranial Doppler (TCD) waveforms from the middle nosis of unilateral ACA spasm is not always a necessity because
cerebral artery obtained from a patient with vasospasm after subarachnoid the hemodynamic consequences may be reduced if the ACoA
hemorrhage (SAH) (upper panel), and a healthy volunteer (lower panel) is patent. However, bilateral ACA spasm may reduce flow to
before and after breath holding. In the SAH patient mean blood flow
the distal ACA segments. In these patients TCCS can detect
velocity (BFV) increased by 5 cm/second to 175 cm/second after 30
seconds of breath holding, which is consistent with impaired increases in velocity that involve at least one artery.
autoregulation (breath holding index = 0.01). In the healthy subject There are few published data on TCD diagnosis of ICA,
BFV increased by 30 cm/second to 88 cm/second (breath holding index PCA, VA, and BA spasm.94, 113 TCD sensitivity is reported to
= 1.72). (Courtesy the Department of Neurology, Ernst Moritz Arndt University of be 25%, 48%, 44%, and 77%, respectively, whereas specificity
Greifswald, Germany.) is 91%, 69%, 88%, and 79%, respectively.94 The sensitivity
Section V—Cerebral Blood Flow 307

(100%) and specificity (95%) for BA spasm are greater when one or more COGIF and complete recanalization when flow
the mean BFV in the BA is compared with the extracranial VA within a previously occluded vessel is now grade 3B or 4;
greater than 2 is considered a threshold for spasm.113 Normal (2) no change—the grade observed at baseline persists; and
reference ranges of the velocity ratio VPCA : VVA (0.76-2.90) also (3) worsening when the measured flow decreases one or more
can help interpret abnormal velocity results in the posterior grades.
circulation.111 TCD is not useful to detect VSP in more distal In acute stroke TCCS also can be used to examine the posi-
branches of intracranial arteries.114,115 Data on TCCS in this tion of the third ventricle and a potential midline shift.117,132-134
context are lacking. Intracerebral hemorrhage, including hemorrhagic transfor-
mation of an infarct, aneurysms, and arteriovenous malfor-
Ischemic Cerebrovascular Disease mations occasionally may be detected by TCCS,117,135-141
although it is not the preferred imaging method for these
Acute Stroke conditions. TCD or TCCS can be used to assess CA in the
TCD and TCCS can be used in acute stroke and to evaluate ischemic hemisphere; it often is impaired even in minor
recanalization therapies as a tool to assess occlusive disease in stroke.142 When CA is impaired, less favorable outcome is
the main segments of the intracranial arteries, to evaluate early observed after recombinant tissue-plasminogen activator
recanalization, and to guide therapeutic decisions.2,116-122 The (rtPA) administration.143,144 Similarly, impaired CA ipsilateral
Thrombolyis in Brain Ischemia (TIBI) scores based on TCD to an acute ischemic stroke is associated with larger
findings can be used to assess initial hemodynamics in acute infarction.145
stroke and early recanalization.123-125 This scale has six levels
of hemodynamic abnormalities; grades 0 to 3 are associated
with worse outcome,126 whereas grades 4 and 5 (i.e., normal Chronic Hypoperfusion
flow and hyperperfusion) are associated with favorable Focal arterial stenosis disrupts the laminar flow pattern in
outcome. Because TIBI grades are frequently dichotomized as diastole. This is associated with flow instability and turbulence
grades 0-3 or grades 4-5, a Consensus on Grading Intracranial with an increase in the range of flow velocities known as
Flow (COGIF) obstruction score is suggested to better reflect spectral broadening.3 As the degree of stenosis progresses, flow
hemodynamic changes based on Doppler findings. velocities increase at the point of maximum narrowing. In
very severe stenosis the reduced residual lumen causes flow
 Grade 1. This indicates no flow and corresponds to an
velocities to decrease and leads to flow cessation with com-
occlusion of the M1 segment of the MCA or the carotid
plete lumen obliteration. Doppler insonation proximal to an
bifurcation. The main diagnostic criterion is the absence
occluded vessel shows a stump flow pattern on spectral wave-
of a color Doppler flow signal and Doppler spectrum in
form. The downstream cerebrovascular impedance can be
the location of the proximal MCA segment.127-129 To make
estimated with the pulsatility and resistance indices.28,146 In
a reliable diagnosis requires sufficient visibility of other
patients with stenosis in proximal cerebral vessels and impaired
arteries (e.g., the ACA A1-segment, C1-segment of intra-
CA, even slight MAP fluctuations may result in ischemia
cranial ICA, ipsilateral A2 or of the contralateral anterior
because of global or local hypoperfusion, especially when col-
circulation) or veins (deep middle cerebral vein) of the
lateral circulation is insufficient.147 Hence where there is slowly
anterior circulation. When insonation conditions are
developing stenosis, ischemia can result from chronic and
unsatisfactory and a reliable diagnosis is not possible, use
acute factors. Typically slowly progressive stenosis causes a
of an US contrast agent is required.
decrease in local perfusion, mostly at the regions farthest from
 Grade 2. This represents low flow in the M1 segment and
the main supplying arteries—the arterial border zones.147 The
may be found in (1) an upstream carotid obstruction that
perfusion decrease makes these areas susceptible to fluctua-
reduces downstream flow in the MCA, (2) intracranial
tions in systemic MAP, oxygen supply, and local vascular resis-
ICA disease with compensatory flow diversion to the con-
tance. When a CPP decrease is short-lived, transient neurologic
tralateral hemisphere and an increase in the asymmetry
deficits may occur, but when the CPP reduction lasts longer,
index,130,131 (3) partial recanalization of the M1 segment
infarction can occur. Thus it is important to know in advance
of the MCA or the carotid bifurcation, and (4) down-
whether a patient who has arterial stenosis is at risk of brain
stream obstruction (i.e., MCA branch occlusion), when
infarct caused by hypoperfusion alone.148 TCD studies suggest
the asymmetry index is high. Upstream and downstream
that the risk of cerebral infarction in the territory of a severely
obstruction have to be differentiated from partial M1
stenotic or occluded ICA or MCA is greater (relative risk = 8)
recanalization.
in subjects with a diminished CA response (Fig. 30.7). The
 Grade 3. This represents High-flow velocities. A focal
annual risk of ipsilateral stroke in these patients with impaired
increase in velocity indicates intracranial stenosis (grade
CA is 23.7%.149 However, an association between impaired
3A), whereas increased flow velocities along the complete
CVR and recurrent ischemic events in asymptomatic carotid
arterial segment indicate hyperperfusion (grade 3B) and
stenosis patients is yet to be determined.150
may be found in the initial stages of recanalization or with
collateral flow across the involved artery. The degree of
intracranial stenosis may be incorrectly estimated when Selection of Patients for Extracranial
there is hyperperfusion. to Intracranial Bypass Surgery
 Grade 4. Normal flow (i.e., flow velocities) is within the
It was expected that some patients with severe stenosis or
normal reference range.
occlusion in a major cerebral artery may benefit from
Follow-up studies can be classified as (1) reflow—partial extracranial-to-intracranial (EC-IC) bypass. Although the
recanalization is assumed when there is an improvement by EC-IC bypass trial performed in the 1980s found no benefit,151
308 Section V—Cerebral Blood Flow

P1 rest

a b c
M1

P1 P1 bh

P1 contr
A
P1 rest

PoCoA

P1 P1 bh
Fig. 30.7  Line graph that illustrates the vasomotor reactivity (VMRr)
determined with bilateral transcranial Doppler (TCD) blood flow velocity
(BFV) measurements in the right middle cerebral artery (MCA) (green
curve, VMRr = 91%) and left MCA (yellow curve, VMRr = 58%) in a B
patient with occlusion of the left internal carotid artery and 70% to 80%
stenosis of the right internal carotid artery, at rest (a), after hyperventilation Fig. 30.8  Transcranial color-coded duplex sonography (TCCS) examina-
(b), after breath holding (c). (White curve illustrates patient’s heart rate.) tion and transcranial Doppler (TCD) assessment of vasomotor reactivity
First time measure 10:18:00 and last time measure 10:23:00. (Courtesy in the P1 segment of posterior cerebral artery in (A) a healthy volunteer:
Dr. G. Kozyra, the Department of Neurology, Medical University of Gdańsk, Poland). mean blood flow velocity (BFV) at rest is 46 cm/second (P1 rest), mean
BFV during hypercarbia after 30 seconds of breath holding (BH) is 69 cm/
second (P1), and breath holding index (BHI) is 1.36; and (B) in a patient
with moyamoya disease: mean BFV at rest is 103 cm/second (P1 rest),
mean BFV during hypercarbia is 109 cm/second (P1 bh) and after 30
it is very likely that this trial included many patients whose seconds of breath holding (BHI = 0.2). In this patient the middle cerebral
strokes were thromboembolic and so did not have impaired artery is occluded and there is collateral flow via posterior communicat-
local perfusion. TCD has been proposed for the selection of ing artery (PoCoA) and compensatory high velocities in the P1. (Courtesy
Department of Neurology, Ernst Moritz Arndt University of Greifswald, Germany.)
patients for bypass—patients with impaired CA and in whom
thromboembolic infarction is unlikely.152 Although in these
patients the CA response improved after surgery153,154 and
selected studies showed clinical benefits,155,156 most current
guidelines do not recommend the use of EC-IC arterial bypass hypoperfusion. However, these same studies also show that
for the treatment of acute ischemic stroke157,158 or in primary159 CA function does not improve in many patients, so surgery
or secondary160 stroke prevention. then may be the preferred treatment option.
TCD testing of CA in patients with moyamoya disease also
may be useful because these patients often have multiple
severe proximal intracranial stenosis and extensive collateral Hyperperfusion Syndrome
development (Fig. 30.8). Surgical revascularization proce- After Carotid Endarterectomy
dures when performed are associated with an improved CA Hyperperfusion syndrome is a rare but serious complication
response. In addition, it seems that new vessels develop better of carotid endarterectomy (CEA) that causes cerebral edema
from grafts to areas where the CA is impaired, independent of and intracerebral hemorrhage.165,166 It may also occur after
local CBF, suggesting that impaired CA may promote a local angioplasty or stenting of the extracrainal and intracranial
angiogenic drive.161,162 arteries.167,168 TCD assessment of CA can help predict which
patients are at high risk for this phenomenon.165,169-171 For
example, Hosoda et al. observed postoperative hyperperfu-
Assessment of Cerebral Autoregulation sion after CEA only in patients following CEA with a less than
to Guide Conservative Management 10% CBF response after acetazolamide.169 In these high-risk
of Occlusive Cerebrovascular Disease patients, strict MAP control should be instituted in the early
In the setting of impaired CA the primary goal is to maintain postoperative period.167,170
adequate CPP, such as by optimizing cardiac function, limiting
orthostatic MAP fluctuations, and carefully using antihyper-
tensive medication. By contrast, when CA is intact, manage- Balloon Occlusion Test
ment may be better centered on control of hypertension and Balloon occlusion test (BOT) of a major vessel (e.g., the ICA
thromboembolism. The relative merits of surgical or medical or VA) is used to assess the adequacy of collateral circulation
treatment for impaired CA still have to be elucidated. In addi- before permanent vessel occlusion or to help guide decisions
tion, CA function can improve over time without surgical about aneurysm surgery or need for a bypass. Patients who
revascularization163,164 because adequate medical therapy for tolerate up to 30 minutes of vessel occlusion without clinical
months to several years may allow sufficient collateral circula- deficits are considered able to tolerate permanent occlusion.172
tion to form, thus reducing the risk of stroke from However, approximately 10% of these patients still may
Section V—Cerebral Blood Flow 309

ICA R

Fig. 30.9  Computed tomography (CT) image (left) shows extensive hemispheric hypodensity, effacement of the sulci, midline shift, and hemorrhage
(arrows) in a traumatic brain injury (TBI) patient. Transcranial color-coded duplex sonography (TCCS) shows no blood flow in the major basal arteries
and oscillating waveform pattern in the terminal internal carotid artery, consistent with brain death. ICA, Internal carotid artery.

develop an infarct. Several techniques including pharmaco- brain death prognosis before organ donation.179,180 Transor-
logically induced hypotension, stump pressure measurements, bital imaging may be used when transtemporal imaging is
and perfusion imaging are used to increase the sensitivity of not feasible.181 Typical flow patterns are reduced or absent
BOT, although there is no consensus on the optimal protocol. diastolic flow, reverberant flow, and short systolic spikes
TCD testing of CA with acetazolamide has been performed (Fig. 30.9). In addition, PI and RI are high because of mark-
during BOT with promising results.172,173 edly reduced systolic flows. However, these patterns also may
be seen temporarily following bolus administration of seda-
tives. In these patients TCD can be used to help make deci-
Increased Intracranial Pressure, Cerebral sions about brain death when sedative drugs preclude formal
Circulatory Arrest, and Brain Death testing.182 The Doppler findings are perhaps best used to
The cranium acts as a near rigid container of virtually incom- exclude brain death rather than make the diagnosis, which still
pressible brain (80%), blood (12%), and cerebrospinal fluid has a clinical foundation (see Chapter 13). TCCS may be more
(CSF) (8%)174 that are in a state of dynamic equilibrium. reliable than TCD to demonstrate cerebral circulatory arrest
When CPP is normal (80-100 mm Hg), ICP is usually less because the suitability of a temporal acoustic window can be
than 15 mm Hg. During every cardiac systole some CSF and easily determined. This can be important because lack of a
venous blood is pushed out of the cranium to prevent an ICP Doppler signal from arteries cannot be discriminated from
increase. When this compensatory mechanism is reduced or lack of a window when TCD is used.183 In cases of difficult
absent, ICP can increase logarithmically as pathologic volume acoustic window, the use of contrast enhancement with TCCS
further increases.175 This in turn can reduce CBF, and it then may help in detecting cerebral circulatory arrest.184
may be necessary to focus management to maintain CBF even
if this causes a further increase in ICP, because neural tissue
generally is more sensitive to ischemia than to increased ICP.176 Traumatic Brain Injury
However, a decrease in CPP causes vasodilation that may be TCD has several applications after TBI: noninvasive evalua-
further exacerbated by increased CO2, potassium ions, and tion of ICP, assessment of need for invasive monitoring, blood
adenosine associated with hypoxia from insufficient CBF. At flow evaluation, assessment of autoregulation and vasoreac-
this point, significant vasodilation does not help improve CBF. tivity, help in calculating ICP indices such as cerebrovascular
Increased ICP also constricts cerebral veins and sinuses, and reactivity and pressure reactivity, and outcome predic-
the pressure is transmitted through its thin, compliant walls tion.41,185,186 Three phases associated with changes in CBF have
inside so the point of maximal resistance of the cerebral vas- been described during the first 2 weeks after TBI.187,188 Phase
cular bed shifts from the arterial to the venous system.177 In 1 occurs on the day of injury and is associated with a low CBF,
other words, CBF decreases because of lower and lower CPP, normal MCA BFV on TCD, and normal arteriovenous differ-
and dilated arteries contain more blood and increased cerebral ence in oxygen (AVDO2). Phase 2 occurs 1 to 2 days postinjury,
blood volume further increases ICP. When the ICP equals when CBF and MCA BFV on TCD are increased, and AVDO2
systemic MAP, CPP decreases to zero, leading to complete is decreased (i.e., relative hyperemia). Phase 3 occurs 4 to 15
CBF cessation. A sudden increase of Pulsatility Index (PI = days postinjury, when CBF is decreased and MCA BFV on
[peak-systolic velocity − end-diastolic velocity]/time-averaged TCD is increased. This suggests a vasospastic phase with the
maximum mean velocity) or Resistivity Index (RI = [peak- peak BFVs generally measured at days 9 to 11. Elevated BFVs
systolic velocity − end-diastolic velocity]/peak-systolic veloc- and increased PI and RI are independent factors associated
ity) values may suggest a critical increase of ICP. with poor outcome from TBI (Figs. 30.10 and 30.11).135,189,190
When there is cerebral circulatory arrest, brain death TCD can be used to assess CA after TBI191 and noninvasively
follows. Although TCD is not a formal test to determine brain assess cerebral perfusion pressure.192 For example, Czosnyka
death, it has very high specificity and sensitivity—100% and et al. used semi-continuous monitoring of MCA velocity with
96%, respectively in one study178—and can be used to help in invasive intracranial and arterial pressure measurement to cal-
brain death assessment, for example, to shorten the process of culate the ability of severe TBI patients to autoregulate in
310 Section V—Cerebral Blood Flow

Fig. 30.10  Traumatic brain injury (TBI) patients who have very high or very low impedance indices and in particular on side of injury have worse
outcomes than patients with indices within normal range (left). Graph that illustrates transcranial color-coded duplex sonography (TCCS) data of
Doppler impedance indices (right) in a patient who had surgery for an intracerebral hematoma. TCCS performed after the first surgery showed high
indices that prompted repeat surgery. The second surgery led to a decrease of the indices. In contrast, intracranial pressure (ICP) measurements were
not as sensitive to changes in the patient’s hemodynamics. RI, Resistivity index ([peak-systolic velocity − end-diastolic velocity]/peak-systolic velocity).

Ved - ipsilateral hyperventilation and mannitol effectively lower ICP only


Ved - contralateral when CA is intact. Minor head injuries also can disturb CA,
ICP4 [mm Hg]
RI - ipsilateral
RI - contralateral
although the effect on outcome is unclear.198
ICP3
indexes and mean velocities in MCA’s
Standardized values of impedance

300%
ICP5
ICP2
Liver Failure
Cerebral edema, increased ICP, and impaired CA may com-
200% plicate acute and chronic liver failure because of disturbed
flow-metabolism coupling.199-201 However, it can be difficult to
use invasive ICP monitors because of liver-induced coagula-
100%
tion abnormalities. Instead TCD and TCCS can be used to
MCA spasm - ipsilateral noninvasively assess intracranial hemodynamics in patients
ICP1 D1 <2.5 mm

Hyperemia in MCA
who develop hepatic encephalopathy. Changes in CA in ful-
MCA spasm
- ipsilateral
D1 >5 mm
- contralateral
D1 <2.5 mm
minant hepatic failure may develop similar to that seen in
0%
4 10 15 20 TBI.202 Changes in MCA blood flow velocity generally are
y
jur

ry

ry
ge
f in

ge

Days after injury


proportional to changes in CBF because the MCA diameter
ur

ur
yo

Is

II s
Da

does not change significantly. TCD findings can help predict


Fig. 30-11  Transcranial color-coded duplex sonography (TCCS)
brainstem death in fulminant hepatic failure203; however, it is
study that illustrates the utility of TCCS monitoring in a traumatic
brain injury (TBI) patient with an intracranial hematoma and
still to be elucidated whether TCD monitoring can help make
subarachnoid hemorrhage. The intracranial hematoma was operated a difference to outcome. Where TCD may be particularly
on twice. After the first surgery, impedance indices in both middle useful is to guide induced hypothermia for patients with
cerebral arteries (MCAs) remained, which suggested that local intracranial hepatic encephalopathy through measurement of the PI to
pressure (ICP) was still increased. A postoperative computed tomography assess the ICP response to therapy. The PI is considered a
(CT) scan showed that the intracranial hematoma was not completely measure of distal CVR, although mathematical models suggest
evacuated. After the second surgery, the impedance indices decreased a complex relationship between PI and multiple hemody-
to normal values. At 5 days, blood flow velocities increased (hyperemia), namic variables.204
because the diameters of both MCAs did not change. Ten days after
injury, blood flow velocities increased due to vasospasm. RI, Resistivity
index ([peak-systolic velocity − end-diastolic velocity]/peak-systolic
velocity).
Microembolic Signals and
Monitoring During Vascular
response to spontaneous fluctuations in arterial pressure
Procedures
based on the regression of CPP on mean and systolic BFVs.41 Microembolic signals (MESs) can be detected using TCD fre-
They found that loss of CA within 2 days of TBI was associated quency downstream from vascular atherothrombotic or car-
with poor outcome,193 although other studies did not confirm diothrombotic lesions. The nature of MESs is heterogeneous,
this observation.194 Systolic flow velocities may provide a and their prevalence is highly variable; however, MESs are
stronger association with outcome than mean flow veloci- thought to represent the cerebral embolic load. For this reason
ties.195 Disruption of CA also is associated with poor condition TCD monitoring for MESs can be useful during a variety of
at presentation.196 Moderate hyperventilation (PaCO2 3.7 kPa) vascular or cardiac procedures including CEA, carotid angio-
can help improve CA after TBI,194 although an effect on plasty and stenting (CAS), intracranial endovascular proce-
outcome is unclear.197 These same studies suggest that dures, or cardiac surgery; predict stroke risk in patients with
Section V—Cerebral Blood Flow 311

asymptomatic carotid stenosis205; and to guide antiplatelet or embolism, hypotension with a significant reduction of MCA
anticoagulation therapy in select patients at risk for embolic BFVs, and angioplasty-induced asystole are associated with
stroke (e.g., after carotid or vertebral artery dissection or adverse outcomes.225,226 However, the frequency of procedure-
symptomatic carotid stenosis).206 related silent cerebral lesions (i.e., radiographic and not clini-
Stroke is the most common major periprocedural compli- cal) appears to be independent of the number of MESs during
cation of CEA or CAS (2%-11% of patients).207,208 TCD mon- the procedure.227 TCD monitoring for MESs also is useful
itoring can help guide management to reduce the perioperative during and after transluminal angioplasty in the posterior
stroke rate.209,210 During CEA hemodynamic changes mea- circulation to guide appropriate use of anticoagulant or anti-
sured by TCD insonation of the MCA manifest as ipsilateral platelet therapies.228 Thromboembolic events that result in
reductions in BFV consistent with a flow volume decline.211 A neurologic deficits occur in 2.4% to 5.2% of patients who
slight decrease first may be observed during administration of undergo endovascular aneurysm occlusion.229 TCD monitor-
anesthetic agents. At cross-clamping, a more severe reduction ing during and immediately after coiling can help identify
(60%-90% of preclamp values) may be detected. When mean patients at high risk of thromboembolic complications.230,231
BFV is less than 15 cm/second (10%-20% of cases),212,213 cere- Acute stroke or cognitive decline associated with cerebral
bral ischemia will develop if not immediately corrected212,214 hypoperfusion or emboli can complicate cardiac surgery.232-
239
(e.g., through placement of a shunt to restore blood flow back Both stroke and cognitive dysfunction are associated with
to 50%-120% of preclamp values). A decrease in MCA BFV the number of intraoperative MESs240 Theses MESs are het-
of more than 70% at cross-clamping is considered by some as erogeneous and may represent gaseous particles, atheroscle-
threshold for a shunt,209,215 although others propose higher rotic plaque components, platelet-fibrin aggregates, and
cutoff values.210,212 High-dose thiopental sodium or propofol lipid-laden or fatty particles.241,242 The introduction of mem-
anesthesia during this phase of the operation is another brane rather than bubble oxygenators and in-line filtration
option to attenuate the effects of brain ischemia.216 A moder- can help decrease the number of MESs during cardiopulmo-
ate increase in TCD velocities is not uncommon during the nary bypass.243 Just as in other invasive procedures, MESs do
days after the operation. However, a persistent and severe not appear at random during the course of cardiac surgery;
increase is seen in patients who develop the hyperperfusion clamping and unclamping of the aorta are associated with
syndrome and can be used to guide blood pressure more than 60% of the total number of MESs.244-247 In partic-
management. ular, the release of an aortic clamp placed over an atheroscle-
During CEA both particulate microemboli and air micro- rotic plaque is associated with an increased risk of cerebral
bubbles occur (Fig. 30.12); these can be detected as MESs by embolism.248,249 The number of MESs is especially high
TCD. Microembolism is particularly prevalent at cross-clamp during cardiac ejection after the release of aortic cross-clamps
release, but only MESs detected during dissection, wound and immediately after bypass. These signals correspond to
closure, shunting, and in the immediate postoperative phase atheromatous debris arising from the aorta. Better selection
of surgery have been associated with brain infarction.210,217,218 of the site of clamp placement by epiaortic US and the use of
In particular more than 10 MESs during initial exposure219 filters distal to the clamp may help reduce perioperative
and more than 20 MESs per hour during the immediate post- stroke.250
operative phase are associated with brain infarcts.217,218 MES
detection therefore can prompt the surgeon to change the
operative technique or to use antithrombotic agents.220 Therapeutic Use of Ultrasound
Carotid angioplasty and stenting is associated with a greater
risk of microemboli than CEA.221,222 Consequently antiem-
in Acute Stroke
bolic devices are increasingly used to reduce stroke risk during Successful thrombolysis depends on the delivery of tissue plas-
CAS. In these patients TCD monitoring can help assess the minogen activator (tPA) to the thrombus through residual
efficacy of cerebral protection provided by these devices blood flow around the arterial obstruction.251,252 Experimental
during stenting, and through assessment of TCD spectral pat- and limited clinical studies suggest that low-frequency253,254 or
terns evaluate the hemodynamic responses to the proce- diagnostic255 US also can have thrombolytic effects and so may
dure.223,224 TCD-identified microemboli during poststent augment the effects of tPA.256-258 This has encouraged assess-
dilation (>5 showers), particulate macroembolism, massive air ments of US for thrombolysis even with standard US equip-
ment.259 In addition, US can increase the uptake of tPA into a
clot,252 increase the binding of tPA to fibrin,260 and reversibly
increase fluid permeation through fibrin.261 These effects may
CTVII occur through acoustic cavitations because degassing reduces
the US effect on flow through fibrin. If bubbles are induced
50 to grow by US to diameters larger than the pore size of the
fibrin lattice surrounding it, stretching of clot fibers may
0 occur. If bubbles collapse sufficiently violently to produce
–50 broadband acoustic emissions, additional inertial cavitations
may produce localized stresses, hot spots, or microjets
–100 that may further alter the structure of clot fibers.262 It also
is possible that the heat generated by US accelerates throm-
Fig. 30.12  Microembolic signals (arrows) in the transcranial Doppler bolysis.263 US-accelerated thrombolysis may be further
(TCD) velocity waveforms from the middle cerebral artery. (Courtesy enhanced by administration of US contrast MBs.264-270 Human
Dr. G. Kozyra, the Department of Neurology, Medical University of Gdańsk, Poland.) CLOTBUST studies271,272 involved MBs with dedicated
312 Section V—Cerebral Blood Flow

machines to make US delivery easier and more reliable for 7. Krejza J, Mariak Z, Babikian VL. Importance of angle correction in the
measurement of blood flow velocity with transcranial Doppler sonography.
thrombolysis, in which complete MCA recanalization with
AJNR Am J Neuroradiol 2001;22(9):1743–7.
dramatic clinical recovery occurred in 49% of patients receiv- 8. Krejza J, Swiat M, Pawlak MA, et al. Suitability of temporal bone acoustic
ing intravenous rtPA combined with transcranial ultrasound window: conventional TCD versus transcranial color-coded duplex
versus 30% in patients receiving intravenous rtPA only.123 sonography. J Neuroimaging 2007;17(4):311–14.
Transcranial US can be attenuated by bones of the cranium; 9. Nyborg WL. Biological effects of ultrasound: development of safety
guidelines. Part II. General review. Ultrasound Med Biol 2001;27(3):301–33.
consequently, its use may not be possible in 10% to 15% of 10. Barnett SB, Ter Haar GR, Ziskin MC, et al. International recommendations
patients.8 There are several strategies to overcome this problem: and guidelines for the safe use of diagnostic ultrasound in medicine.
(1) use of endovascular wires and transducers to deliver US Ultrasound Med Biol 2000;26(3):355–66.
locally; (2) use of lower-frequency and subsequently higher- 11. Wu J, Winkler AJ, O’Neill TP. Effect of acoustic streaming on ultrasonic
heating. Ultrasound Med Biol 1994;20(2):195–201.
power US for transcutaneous US-enhanced thrombolysis; or
12. Daffertshofer M, Gass A, Ringleb P, et al. Transcranial low-frequency
(3) use of US contrast to induce and increase the number of ultrasound-mediated thrombolysis in brain ischemia: increased risk of
cavitations at the site where the US beam of a high mechanical hemorrhage with combined ultrasound and tissue plasminogen activator:
index is targeted.273-278 Frequencies used include 20 kHz, 40 results of a phase II clinical trial. Stroke 2005;36(7):1441–6.
kHz, 170 kHz, 300 kHz, 1 MHz, and 2 MHz, at intensities 13. Mariak Z, Krejza J, Swiercz M, et al. Human brain temperature in vivo: lack
of heating during color transcranial Doppler ultrasonography.
from 0.25 to 10 W/cm2.264,279-283 J Neuroimaging 2001;11(3):308–12.
14. Karagoz I, Kartal MK. A new safety parameter for diagnostic ultrasound
Conclusion thermal bioeffects: safe use time. J Acoust Soc Am 2009;125(6):3601–10
15. Krasovitski B, Frenkel V, Shoham S, et al. Intramembrane cavitation as a
unifying mechanism for ultrasound-induced bioeffects. Proc Natl Acad Sci U
Optimal management of patients in the NCCU depends in
S A 2011;108(8):3258–63.
part on understanding the physiology for the individual 16. Kondo T, Kodaira T, Kano E. Free radical formation induced by ultrasound
patient. TCD ultrasonography first described in the 1980s is a and its effects on strand breaks in DNA of cultured FM3A cells. Free Radic
noninvasive test that can be used to help understand patient Res Commun 1993;19(Suppl 1):S193–200.
pathophysiology and monitor for responses to management. 17. Hynynen K, McDannold N, Sheikov NA, et al. Local and reversible
blood-brain barrier disruption by noninvasive focused ultrasound at
Technologic advances such as TCCS now make TCD more frequencies suitable for trans-skull sonications. Neuroimage 2005;24(1):
reliable and accurate. This is a well-known test that records 12–20.
the velocity (in cm/sec) and direction of blood flow in the 18. Zhong W, Sit WH, Wan JM, et al. Sonoporation induces apoptosis and cell
basal vessels of the brain and is most frequently used to evalu- cycle arrest in human promyelocytic leukemia cells. Ultrasound Med Biol
2011;37(12):2149–59.
ate for vasospasm, particularly in the MCA, in SAH patients.
19. Sarvazyan AP, Rudenko OV, Nyborg WL. Biomedical applications of
In addition, TCD and TCCS can be used to noninvasively radiation force of ultrasound: historical roots and physical basis. Ultrasound
evaluate: 1) intracranial pressure, 2) other causes of vessel Med Biol 2010;36(9):1379–94.
stenosis, e.g., atherosclerosis, 3) TBI patients, 4) hemodynamic 20. Lassen NA. Cerebral blood flow and oxygen consumption in man. Physiol
alterations observed in intracranial and extracranial occlusive Rev 1959;39(2):183–238.
21. Strandgaard S, Olesen J, Skinhoj E, et al. Autoregulation of brain circulation
disease, and 5) emboli among other disease states. However, in severe arterial hypertension. BMJ 1973;1(5852):507–10.
TCD does not measure flow rate (mL/min) or perfusion (mL 22. Waldemar G, Schmidt JF, Andersen AR, et al. Angiotensin converting enzyme
per 100/g/min). Instead, changes in TCD velocities reflect rela- inhibition and cerebral blood flow autoregulation in normotensive and
tive CBF changes provided that the recorded vessel diameter hypertensive man. J Hypertens 1989;7(3):229–35.
23. Rudzinski W, Swiat M, Tomaszewski M, et al. Cerebral hemodynamics and
remains constant. Therefore TCD also can be used to examine
investigations of cerebral blood flow regulation. Nucl Med Rev Cent East Eur
CBF reactivity in response to CO2, blood pressure, acetazol- 2007;10(1):29–42.
amide, or head position and thus help assess cerebral auto- 24. Wallis SJ, Firth J, Dunn WR. Pressure-induced myogenic responses in human
regulation. While noninvasive, TCD is not a technique that is isolated cerebral resistance arteries. Stroke 1996;27(12):2287–90.
easy to use for continuous monitoring, although continued 25. Garcia-Roldan JL, Bevan JA. Flow-induced constriction and dilation of
cerebral resistance arteries. Circ Res 1990;66(5):1445–8.
technologic improvements now make this possible. In addi- 26. Thorin-Trescases N, Bevan JA. High levels of myogenic tone antagonize the
tion, TCD ultrasound examination requires training and prac- dilator response to flow of small rabbit cerebral arteries. Stroke
tice, and so the accuracy of the examination often depends on 1998;29(6):1194–200.
the operator’s skill and experience. 27. McDonald DA. Arterial impedance. In: McDonald DA, editor. Blood flow in
arteries. Baltimore: Williams & Wilkins; 1977. p. 351–89.
28. Kontos HA, Raper AJ, Patterson JL. Analysis of vasoactivity of local pH,
References PaCO2 and bicarbonate on pial vessels. Stroke 1977;8(3):358–60.
29. Baffi J, Gorcs T, Slowik F, et al. Neuropeptides in the human superior
1. Alexandrov AV, Sloan MA, Tegeler CH, et al. Practice standards for cervical ganglion. Brain Res 1992;570(1-2):272–8.
transcranial Doppler (TCD) ultrasound. Part II. Clinical indications and 30. Bleys RL, Cowen T. Innervation of cerebral blood vessels: morphology,
expected outcomes. J Neuroimaging 2010;Oct 26. Epub ahead of print. plasticity, age-related, and Alzheimer’s disease-related neurodegeneration.
2. Edmonds Jr HL, Isley MR, Sloan TB, et al. American Society of Microsc Res Tech 2001;53(2):106–18.
Neurophysiologic Monitoring and American Society of Neuroimaging joint 31. Baumbach GL, Heistad DD. Effects of sympathetic stimulation and changes
guidelines for transcranial Doppler ultrasonic monitoring. J Neuroimaging in arterial pressure on segmental resistance of cerebral vessels in rabbits and
2011;21(2):177–83. cats. Circ Res 1983;52(5):527–33.
3. Nelson TR, Pretorius DH. The Doppler signal: where does it come from and 32. Morita-Tsuzuki Y, Hardebo JE, Bouskela E. Inhibition of nitric oxide
what does it mean? AJR Am J Roentgenol 1988;151(3):439–47. synthase attenuates the cerebral blood flow response to stimulation of
4. Taylor KJW, Burns PN, Wells PNT. Clinical applications of Doppler postganglionic parasympathetic nerves in the rat. J Cereb Blood Flow Metab
ultrasound. New York: Raven Press; 1995. 1993;13(6):993–7.
5. Hoeks AP, Reneman RS. Biophysical principles of vascular diagnosis. J Clin 33. Kalaria RN, Stockmeier CA, Harik SI. Brain microvessels are innervated by
Ultrasound 1995;23(2):71–9. locus ceruleus noradrenergic neurons. Neurosci Lett 1989;97(1-2):203–8.
6. Krejza J, Mariak Z, Melhem ER, et al. A guide to the identification of major 34. Moreno MJ, Lopez DP, Conde MV, et al. Cat cerebral arteries are
cerebral arteries with transcranial color Doppler sonography. AJR Am J functionally innervated by serotoninergic fibers from central and peripheral
Roentgenol 2000;174(5):1297–303. origins. Stroke 1995;26(2):271–5.
Section V—Cerebral Blood Flow 313

35. Sato A, Sato Y, Uchida S. Activation of the intracerebral cholinergic nerve 45. Okazawa H, Yamauchi H, Sugimoto K, et al. Effects of acetazolamide on
fibers originating in the basal forebrain increases regional cerebral blood cerebral blood flow, blood volume, and oxygen metabolism: a positron
flow in the rat’s cortex and hippocampus. Neurosci Lett 2004;361(1-3):90–3. emission tomography study with healthy volunteers. J Cereb Blood Flow
36. Cauli B, Tong XK, Rancillac A, et al. Cortical GABA interneurons in Metab 2001;21(12):1472–9.
neurovascular coupling: relays for subcortical vasoactive pathways. 46. Sorteberg W, Lindegaard KF, Rootwelt K, et al. Effect of acetazolamide on
J Neurosci 2004;24(41):8940–9. cerebral artery blood velocity and regional cerebral blood flow in normal
37. Mulligan SJ, MacVicar BA. Calcium transients in astrocyte endfeet cause subjects. Acta Neurochir (Wien) 1989;97(3-4):139–45.
cerebrovascular constrictions. Nature 2004;431(7005):195–9. 47. Sullivan HG, Kingsbury TB, Morgan ME, et al. The rCBF response to
38. Aaslid R, Lash SR, Bardy GH, et al. Dynamic pressure–flow velocity Diamox in normal subjects and cerebrovascular disease patients. J Neurosurg
relationships in the human cerebral circulation. Stroke 2003;34(7):1645–9. 1987;67(4):525–34.
39. Paulson OB, Strandgaard S, Edvinsson L. Cerebral autoregulation. 48. Kuwabara Y, Ichiya Y, Sasaki M, et al. Time dependency of the acetazolamide
Cerebrovasc Brain Metab Rev 1990;2(2):161–92. effect on cerebral hemodynamics in patients with chronic occlusive cerebral
40. Tiecks FP, Lam AM, Aaslid R, et al. Comparison of static and dynamic arteries. Early steal phenomenon demonstrated by [15O]H2O positron
cerebral autoregulation measurements. Stroke 1995;26(6):1014–19. emission tomography. Stroke 1995;26(10):1825–9.
41. Czosnyka M, Brady K, Reinhard M, et al. Monitoring of cerebrovascular 49. Yonas H, Pindzola RR. Physiological determination of cerebrovascular
autoregulation: facts, myths, and missing links. Neurocrit Care 2009;10(3): reserves and its use in clinical management. Cerebrovasc Brain Metab Rev
373–86. 1994;6(4):325–40.
42. Grossmann WM, Koeberle B. The dose-response relationship of 50. Petrella JR, DeCarli C, Dagli M, et al. Age-related vasodilatory response to
acetazolamide on the cerebral blood flow in normal subjects. Cerebrovasc acetazolamide challenge in healthy adults: a dynamic contrast-enhanced MR
Dis 2000;10(1):65–9. study. AJNR Am J Neuroradiol 1998;19(1):39–44.
43. Heuser D, Astrup J, Lassen NA, et al. Brain carbonic acid acidosis after
acetazolamide. Acta Physiol Scand 1975;93(3):385–90. A complete list of references for this chapter can be found online at
44. Vorstrup S, Henriksen L, Paulson OB. Effect of acetazolamide on cerebral www.expertconsult.com.
blood flow and cerebral metabolic rate for oxygen. J Clin Invest 1984;74(5):
1634–9.
Section V—Cerebral Blood Flow 313.e1

References 28. Kontos HA, Raper AJ, Patterson JL. Analysis of vasoactivity of local pH,
PaCO2 and bicarbonate on pial vessels. Stroke 1977;8(3):358–60.
1. Alexandrov AV, Sloan MA, Tegeler CH, et al. Practice standards for 29. Baffi J, Gorcs T, Slowik F, et al. Neuropeptides in the human superior
transcranial Doppler (TCD) ultrasound. Part II. Clinical indications and cervical ganglion. Brain Res 1992;570(1-2):272–8.
expected outcomes. J Neuroimaging 2010;Oct 26. Epub ahead of print. 30. Bleys RL, Cowen T. Innervation of cerebral blood vessels: morphology,
2. Edmonds Jr HL, Isley MR, Sloan TB, et al. American Society of plasticity, age-related, and Alzheimer’s disease-related neurodegeneration.
Neurophysiologic Monitoring and American Society of Neuroimaging joint Microsc Res Tech 2001;53(2):106–18.
guidelines for transcranial Doppler ultrasonic monitoring. J Neuroimaging 31. Baumbach GL, Heistad DD. Effects of sympathetic stimulation and changes
2011;21(2):177–83. in arterial pressure on segmental resistance of cerebral vessels in rabbits
3. Nelson TR, Pretorius DH. The Doppler signal: where does it come from and cats. Circ Res 1983;52(5):527–33.
and what does it mean? AJR Am J Roentgenol 1988;151(3):439–47. 32. Morita-Tsuzuki Y, Hardebo JE, Bouskela E. Inhibition of nitric oxide
4. Taylor KJW, Burns PN, Wells PNT. Clinical applications of Doppler synthase attenuates the cerebral blood flow response to stimulation of
ultrasound. New York: Raven Press; 1995. postganglionic parasympathetic nerves in the rat. J Cereb Blood Flow
5. Hoeks AP, Reneman RS. Biophysical principles of vascular diagnosis. J Clin Metab 1993;13(6):993–7.
Ultrasound 1995;23(2):71–9. 33. Kalaria RN, Stockmeier CA, Harik SI. Brain microvessels are innervated by
6. Krejza J, Mariak Z, Melhem ER, et al. A guide to the identification of major locus ceruleus noradrenergic neurons. Neurosci Lett 1989;97(1-2):203–8.
cerebral arteries with transcranial color Doppler sonography. AJR Am J 34. Moreno MJ, Lopez DP, Conde MV, et al. Cat cerebral arteries are
Roentgenol 2000;174(5):1297–303. functionally innervated by serotoninergic fibers from central and peripheral
7. Krejza J, Mariak Z, Babikian VL. Importance of angle correction in the origins. Stroke 1995;26(2):271–5.
measurement of blood flow velocity with transcranial Doppler sonography. 35. Sato A, Sato Y, Uchida S. Activation of the intracerebral cholinergic nerve
AJNR Am J Neuroradiol 2001;22(9):1743–7. fibers originating in the basal forebrain increases regional cerebral blood
8. Krejza J, Swiat M, Pawlak MA, et al. Suitability of temporal bone acoustic flow in the rat’s cortex and hippocampus. Neurosci Lett 2004;361(1-3):
window: conventional TCD versus transcranial color-coded duplex 90–3.
sonography. J Neuroimaging 2007;17(4):311–14. 36. Cauli B, Tong XK, Rancillac A, et al. Cortical GABA interneurons in
9. Nyborg WL. Biological effects of ultrasound: development of safety neurovascular coupling: relays for subcortical vasoactive pathways.
guidelines. Part II. General review. Ultrasound Med Biol 2001;27(3): J Neurosci 2004;24(41):8940–9.
301–33. 37. Mulligan SJ, MacVicar BA. Calcium transients in astrocyte endfeet cause
10. Barnett SB, Ter Haar GR, Ziskin MC, et al. International recommendations cerebrovascular constrictions. Nature 2004;431(7005):195–9.
and guidelines for the safe use of diagnostic ultrasound in medicine. 38. Aaslid R, Lash SR, Bardy GH, et al. Dynamic pressure–flow velocity
Ultrasound Med Biol 2000;26(3):355–66. relationships in the human cerebral circulation. Stroke 2003;34(7):1645–9.
11. Wu J, Winkler AJ, O’Neill TP. Effect of acoustic streaming on ultrasonic 39. Paulson OB, Strandgaard S, Edvinsson L. Cerebral autoregulation.
heating. Ultrasound Med Biol 1994;20(2):195–201. Cerebrovasc Brain Metab Rev 1990;2(2):161–92.
12. Daffertshofer M, Gass A, Ringleb P, et al. Transcranial low-frequency 40. Tiecks FP, Lam AM, Aaslid R, et al. Comparison of static and dynamic
ultrasound-mediated thrombolysis in brain ischemia: increased risk of cerebral autoregulation measurements. Stroke 1995;26(6):1014–19.
hemorrhage with combined ultrasound and tissue plasminogen activator: 41. Czosnyka M, Brady K, Reinhard M, et al. Monitoring of cerebrovascular
results of a phase II clinical trial. Stroke 2005;36(7):1441–6. autoregulation: facts, myths, and missing links. Neurocrit Care 2009;10(3):
13. Mariak Z, Krejza J, Swiercz M, et al. Human brain temperature in vivo: lack 373–86.
of heating during color transcranial Doppler ultrasonography. 42. Grossmann WM, Koeberle B. The dose-response relationship of
J Neuroimaging 2001;11(3):308–12. acetazolamide on the cerebral blood flow in normal subjects. Cerebrovasc
14. Karagoz I, Kartal MK. A new safety parameter for diagnostic ultrasound Dis 2000;10(1):65–9.
thermal bioeffects: safe use time. J Acoust Soc Am 2009;125(6):3601–10 43. Heuser D, Astrup J, Lassen NA, et al. Brain carbonic acid acidosis after
15. Krasovitski B, Frenkel V, Shoham S, et al. Intramembrane cavitation as a acetazolamide. Acta Physiol Scand 1975;93(3):385–90.
unifying mechanism for ultrasound-induced bioeffects. Proc Natl Acad Sci 44. Vorstrup S, Henriksen L, Paulson OB. Effect of acetazolamide on cerebral
U S A 2011;108(8):3258–63. blood flow and cerebral metabolic rate for oxygen. J Clin Invest 1984;74(5):
16. Kondo T, Kodaira T, Kano E. Free radical formation induced by ultrasound 1634–9.
and its effects on strand breaks in DNA of cultured FM3A cells. Free Radic 45. Okazawa H, Yamauchi H, Sugimoto K, et al. Effects of acetazolamide on
Res Commun 1993;19(Suppl 1):S193–200. cerebral blood flow, blood volume, and oxygen metabolism: a positron
17. Hynynen K, McDannold N, Sheikov NA, et al. Local and reversible emission tomography study with healthy volunteers. J Cereb Blood Flow
blood-brain barrier disruption by noninvasive focused ultrasound at Metab 2001;21(12):1472–9.
frequencies suitable for trans-skull sonications. Neuroimage 46. Sorteberg W, Lindegaard KF, Rootwelt K, et al. Effect of acetazolamide on
2005;24(1):12–20. cerebral artery blood velocity and regional cerebral blood flow in normal
18. Zhong W, Sit WH, Wan JM, et al. Sonoporation induces apoptosis and cell subjects. Acta Neurochir (Wien) 1989;97(3-4):139–45.
cycle arrest in human promyelocytic leukemia cells. Ultrasound Med Biol 47. Sullivan HG, Kingsbury TB, Morgan ME, et al. The rCBF response to
2011;37(12):2149–59. Diamox in normal subjects and cerebrovascular disease patients.
19. Sarvazyan AP, Rudenko OV, Nyborg WL. Biomedical applications of J Neurosurg 1987;67(4):525–34.
radiation force of ultrasound: historical roots and physical basis. 48. Kuwabara Y, Ichiya Y, Sasaki M, et al. Time dependency of the
Ultrasound Med Biol 2010;36(9):1379–94. acetazolamide effect on cerebral hemodynamics in patients with chronic
20. Lassen NA. Cerebral blood flow and oxygen consumption in man. Physiol occlusive cerebral arteries. Early steal phenomenon demonstrated by [15O]
Rev 1959;39(2):183–238. H2O positron emission tomography. Stroke 1995;26(10):1825–9.
21. Strandgaard S, Olesen J, Skinhoj E, et al. Autoregulation of brain circulation 49. Yonas H, Pindzola RR. Physiological determination of cerebrovascular
in severe arterial hypertension. BMJ 1973;1(5852):507–10. reserves and its use in clinical management. Cerebrovasc Brain Metab Rev
22. Waldemar G, Schmidt JF, Andersen AR, et al. Angiotensin converting 1994;6(4):325–40.
enzyme inhibition and cerebral blood flow autoregulation in normotensive 50. Petrella JR, DeCarli C, Dagli M, et al. Age-related vasodilatory response to
and hypertensive man. J Hypertens 1989;7(3):229–35. acetazolamide challenge in healthy adults: a dynamic contrast-enhanced
23. Rudzinski W, Swiat M, Tomaszewski M, et al. Cerebral hemodynamics and MR study. AJNR Am J Neuroradiol 1998;19(1):39–44.
investigations of cerebral blood flow regulation. Nucl Med Rev Cent East 51. Brian JE. Carbon dioxide and the cerebral circulation. Anesthesiology
Eur 2007;10(1):29–42. 1998;88(5):1365–86.
24. Wallis SJ, Firth J, Dunn WR. Pressure-induced myogenic responses in 52. Kanno I, Uemura K, Higano S, et al. Oxygen extraction fraction at
human isolated cerebral resistance arteries. Stroke 1996;27(12):2287–90. maximally vasodilated tissue in the ischemic brain estimated from the
25. Garcia-Roldan JL, Bevan JA. Flow-induced constriction and dilation of regional CO2 responsiveness measured by positron emission tomography.
cerebral resistance arteries. Circ Res 1990;66(5):1445–8. J Cereb Blood Flow Metab 1988;8(2):227–35.
26. Thorin-Trescases N, Bevan JA. High levels of myogenic tone antagonize the 53. Kazumata K, Tanaka N, Ishikawa T, et al. Dissociation of vasoreactivity to
dilator response to flow of small rabbit cerebral arteries. Stroke 1998;29(6): acetazolamide and hypercapnia. Comparative study in patients with chronic
1194–200. occlusive major cerebral artery disease. Stroke 1996;27(11):2052–8.
27. McDonald DA. Arterial impedance. In: McDonald DA, editor. Blood flow in 54. Gambhir S, Inao S, Tadokoro M, et al. Comparison of vasodilatory effect of
arteries. Baltimore: Williams & Wilkins; 1977. p. 351–89. carbon dioxide inhalation and intravenous acetazolamide on brain
313.e2 Section V—Cerebral Blood Flow

vasculature using positron emission tomography. Neurol Res 1997;19(2): 81. Paris A, Scholz J, von Knobelsdorff G, et al. The effect of remifentanil on
139–44. cerebral blood flow velocity. Anesth Analg 1998;87(3):569–73.
55. Ringelstein EB, Van Eyck S, Mertens I. Evaluation of cerebral vasomotor 82. Moppett IK, Sherman RW, Wild MJ, et al. Effects of norepinephrine and
reactivity by various vasodilating stimuli: comparison of CO2 to glyceryl trinitrate on cerebral haemodynamics: transcranial Doppler study
acetazolamide. J Cereb Blood Flow Metab 1992;12(1):162–8. in healthy volunteers. Br J Anaesth 2008;100(2):240–4.
56. Lam JM, Hsiang JN, Poon WS. Monitoring of autoregulation using laser 83. Moppett IK, Wild MJ, Sherman RW, et al. Effects of ephedrine, dobutamine
Doppler flowmetry in patients with head injury. J Neurosurg and dopexamine on cerebral haemodynamics: transcranial Doppler studies
1997;86(3):438–45. in healthy volunteers. Br J Anaesth 2004;92(1):39–44.
57. Tiecks FP, Lam AM, Aaslid R, et al. Comparison of static and dynamic 84. Strebel SP, Kindler C, Bissonnette B, et al. The impact of systemic
cerebral autoregulation measurements. Stroke 1995;26(6):1014–19. vasoconstrictors on the cerebral circulation of anesthetized patients.
58. Bouma GJ, Muizelaar JP. Relationship between cardiac output and cerebral Anesthesiology 1998;89(1):67–72.
blood flow in patients with intact and with impaired autoregulation. 85. Endoh H, Honda T, Ohashi S, et al. The influence of nitroglycerin and
J Neurosurg 1990;73(3):368–74. prostaglandin E1 on dynamic cerebral autoregulation in adult patients
59. Aaslid R, Lindegaard KF, Sorteberg W, et al. Cerebral autoregulation during propofol and fentanyl anaesthesia. Anaesthesia 2001;56(10):947–52.
dynamics in humans. Stroke 1989;20(1):45–52. 86. Brauer P, Kochs E, Werner C, et al. Correlation of transcranial Doppler
60. Tiecks FP, Douville C, Byrd S, et al. Evaluation of impaired cerebral sonography mean flow velocity with cerebral blood flow in patients with
autoregulation by the Valsalva maneuver. Stroke 1996;27(7):1177–82. intracranial pathology. J Neurosurg Anesthesiol 1998;10(2):80–5.
61. Kofke WA, Dong ML, Bloom M, et al. Transcranial Doppler 87. Demolis P, Tran Dinh YR, Giudicelli JF. Relationships between cerebral
ultrasonography with induction of anesthesia for neurosurgery. regional blood flow velocities and volumetric blood flows and their
J Neurosurg Anesthesiol 1994;6(2):89–97. respective reactivities to acetazolamide. Stroke 1996;27(10):1835–9.
62. Renou AM, Vernhiet J, Macrez P, et al. Cerebral blood flow and metabolism 88. van Gijn J, Rinkel GJE. Subarachnoid haemorrhage: diagnosis, causes and
during etomidate anaesthesia in man. Br J Anaesth 1978;50(10):1047–51. management. Brain 2001;124:249–78.
63. Harrison JM, Girling KJ, Mahajan RP. Effects of target-controlled infusion 89. Shimoda M, Oda S, Tsugane R, et al. Intracranial complications of
of propofol on the transient hyperaemic response and carbon dioxide hypervolemic therapy in patients with a delayed ischemic deficit attributed
reactivity in the middle cerebral artery. Br J Anaesth 1999;83(6):839–44. to vasospasm. J Neurosurg 1993;78(3):423–9.
64. Matta BF, Lam AM. Nitrous oxide increases cerebral blood flow velocity 90. Willinsky RA, Taylor SM, terBrugge K, et al. Neurologic complications of
during pharmacologically induced EEG silence in humans. J Neurosurg cerebral angiography: prospective analysis of 2,899 procedures and review
Anesthesiol 1995;7(2):89–93. of the literature. Radiology 2003;227(2):522–8.
65. McCulloch TJ, Visco E, Lam AM. Graded hypercapnia and cerebral 91. Anderson GB, Ashforth R, Steinke DE, et al. CT angiography for the
autoregulation during sevoflurane or propofol anesthesia. Anesthesiology detection of cerebral vasospasm in patients with acute subarachnoid
2000;93(5):1205–9. hemorrhage. AJNR Am J Neuroradiol 2000;21(6):1011–15.
66. Mayberg TS, Lam AM, Matta BF, et al. Ketamine does not increase cerebral 92. Heiserman JE. MR angiography for the diagnosis of vasospasm after
blood flow velocity or intracranial pressure during isoflurane/nitrous oxide subarachnoid hemorrhage. Is it accurate? Is it safe? AJNR Am J Neuroradiol
anesthesia in patients undergoing craniotomy. Anesth Analg 1995;81(1): 2000;21(9):1571–2.
84–9. 93. Aaslid R, Huber P, Nornes H. Evaluation of cerebrovascular spasm with
67. Kuroda Y, Murakami M, Tsuruta J, et al. Blood flow velocity of middle transcranial Doppler ultrasound. J Neurosurg 1984;60(1):37–41.
cerebral artery during prolonged anesthesia with halothane, isoflurane, and 94. Lysakowski C, Walder B, Costanza MC, et al. Transcranial Doppler versus
sevoflurane in humans. Anesthesiology 1997;87(3):527–32. angiography in patients with vasospasm due to a ruptured cerebral
68. Matta BF, Heath KJ, Tipping K, et al. Direct cerebral vasodilatory effects of aneurysm: a systematic review. Stroke 2001;32(10):2292–8.
sevoflurane and isoflurane. Anesthesiology 1999;91(3):677–80. 95. Mariak Z, Krejza J, Swiercz M, et al. Accuracy of transcranial color Doppler
69. Mielck F, Stephan H, Buhre W, et al. Effects of 1 MAC desflurane on ultrasonography in the diagnosis of middle cerebral artery spasm
cerebral metabolism, blood flow and carbon dioxide reactivity in humans. determined by receiver operating characteristic analysis. J Neurosurg 2002;
Br J Anaesth 1998;81(2):155–60. 96(2):323–30.
70. Gupta S, Heath K, Matta BF. Effect of incremental doses of sevoflurane on 96. Krejza J, Mariak Z, Lewko J. Standardization of flow velocities with respect
cerebral pressure autoregulation in humans. Br J Anaesth to age and sex improves the accuracy of transcranial color Doppler
1997;79(4):469–72. sonography of middle cerebral artery spasm. Am J Roentgenol 2003;181(1):
71. Heath KJ, Gupta S, Matta BF. The effects of sevoflurane on cerebral 245–52.
hemodynamics during propofol anesthesia. Anesth Analg 1997;85(6):1284–7. 97. Proust F, Callonec F, Clavier E, et al. Usefulness of transcranial color-coded
72. Matta BF, Lam AM, Mayberg TS, et al. Cerebrovascular response to carbon sonography in the diagnosis of cerebral vasospasm. Stroke 1999;30(5):
dioxide during sodium nitroprusside- and isoflurane-induced hypotension. 1091–8.
Br J Anaesth 1995;74(3):296–300. 98. Krejza J, Kochanowicz J, Mariak Z, et al. Middle cerebral artery spasm after
73. Strebel S, Lam AM, Matta B, et al. Dynamic and static cerebral subarachnoid hemorrhage: detection with transcranial color-coded duplex
autoregulation during isoflurane, desflurane, and propofol anesthesia. US. Radiology 2005;236(2):621–9.
Anesthesiology 1995;83(1):66–76. 99. Swiat M, Weigele J, Hurst RW, et al. Middle cerebral artery vasospasm:
74. Summors AC, Gupta AK, Matta BF. Dynamic cerebral autoregulation transcranial color-coded duplex sonography versus conventional
during sevoflurane anesthesia: a comparison with isoflurane. Anesth Analg non-imaging TCD. Crit Care Med 2009;37(3):963–8.
1999;88(2):341–5. 100. Krejza J, Rudzinski W, Pawlak MA, et al. Angle-corrected imaging
75. Girling KJ, Cavill G, Mahajan RP. The effects of nitrous oxide and oxygen transcranial Doppler sonography versus imaging and nonimaging
on transient hyperemic response in human volunteers. Anesth Analg 1999; transcranial Doppler sonography in children with sickle cell disease. AJNR
89(1):175–80. Am J Neuroradiol 2007;28(8):1613–18.
76. Bedforth NM, Girling KJ, Harrison JM, et al. The effects of sevoflurane and 101. Muizelaar JP, Zwienenberg M, Rudisill NA, et al. The prophylactic use of
nitrous oxide on middle cerebral artery blood flow velocity and transient transluminal balloon angioplasty in patients with Fisher grade 3
hyperemic response. Anesth Analg 1999;89(1):170–4. subarachnoid hemorrhage: a pilot study. J Neurosurg 1999;91(1):51–8.
77. Matta BF, Lam AM. Nitrous oxide increases cerebral blood flow velocity 102. Murayama Y, Malisch T, Guglielmi G, et al. Incidence of cerebral vasospasm
during pharmacologically induced EEG silence in humans. J Neurosurg after endovascular treatment of acutely ruptured aneurysms: report on 69
Anesthesiol 1995;7(2):89–93. cases. J Neurosurg 1997;87(6):830–5.
78. Kovarik WD, Mayberg TS, Lam AM, et al. Succinylcholine does not change 103. Wardlaw JM, Offin R, Teasdale GM, et al. Is routine transcranial Doppler
intracranial pressure, cerebral blood flow velocity, or the ultrasound monitoring useful in the management of subarachnoid
electroencephalogram in patients with neurologic injury. Anesth Analg hemorrhage? J Neurosurg 1998;88(2):272–6.
1994;78(3):469–73. 104. Lindegaard KF, Nornes H, Bakke SJ, et al. Cerebral vasospasm diagnosis by
79. Kofke WA, Shaheen N, McWhorter J, et al. Transcranial Doppler means of angiography and blood velocity-measurements. Acta
ultrasonography with induction of anesthesia and neuromuscular blockade Neurochirurgica 1989;100(1-2):12–24.
in surgical patients. J Clin Anesth 2001;13(5):335–8. 105. Lang EW, Diehl RR, Mehdorn HM. Cerebral autoregulation testing after
80. Hanel F, Werner C, von Knobelsdorff G, et al. The effects of fentanyl and aneurysmal subarachnoid hemorrhage: the phase relationship between
sufentanil on cerebral hemodynamics. J Neurosurg Anesthesiol 1997;9(3): arterial blood pressure and cerebral blood flow velocity. Crit Care Med
223–7. 2001;29(1):158–63.
Section V—Cerebral Blood Flow 313.e3

106. Tseng MY, Czosnyka M, Richards H, et al. Effects of acute treatment with 129. Kenton AR, Martin PJ, Abbott RJ, et al. Comparison of transcranial
pravastatin on cerebral vasospasm, autoregulation, and delayed ischemic color-coded sonography and magnetic resonance angiography in acute
deficits after aneurysmal subarachnoid hemorrhage: a phase II randomized stroke. Stroke 1997;28(8):1601–6.
placebo-controlled trial. Stroke 2005;36(8):1627–32. 130. Baumgartner RW, Mattle HP, Schroth G. Assessment of >/=50% and <50%
107. Rajajee V, Fletcher JJ, Pandey AS, et al. Low pulsatility index on transcranial intracranial stenoses by transcranial color-coded duplex sonography. Stroke
doppler predicts symptomatic large vessel vasospasm after aneurysmal 1999;30(1):87–92.
sub-arachnoid hemorrhage. Neurosurgery 2011;Nov 14. Epub ahead of print. 131. Gortler M, Niethammer R, Widder B. Differentiating subtotal carotid artery
108. Washington CW, Zipfel GJ. Participants in the International Multi- stenoses from occlusions by colour-coded duplex sonography. J Neurol
disciplinary Consensus Conference on the critical care management of 1994;241(5):301–5.
subarachnoid hemorrhage. Detection and monitoring of vasospasm and 132. Seidel G, Gerriets T, Kaps M, et al. Dislocation of the third ventricle due to
delayed cerebral ischemia: a review and assessment of the literature. space-occupying stroke evaluated by transcranial duplex sonography.
Neurocrit Care 2011;15(2):312–7. J Neuroimaging 1996;6(4):227–30.
109. Grosset DG, Straiton J, McDonald I, et al. Use of transcranial Doppler 133. Stolz E, Gerriets T, Fiss I, et al. Comparison of transcranial color-coded
sonography to predict development of a delayed ischemic deficit after duplex sonography and cranial CT measurements for determining third
subarachnoid hemorrhage. J Neurosurg 1993;78(2):183–7. ventricle midline shift in space-occupying stroke. AJNR Am J Neuroradiol
110. Vora YY, Suarez-Almazor M, Steinke DE, et al. Role of transcranial Doppler 1999;20(8):1567–71.
monitoring in the diagnosis of cerebral vasospasm after subarachnoid 134. Gerriets T, Stolz E, Modrau B, et al. Sonographic monitoring of midline
hemorrhage. Neurosurgery 1999;44(6):1237–47. shift in hemispheric infarctions. Neurology 1999;52(1):45–9.
111. Krejza J, Szydlik P, Liebeskind DS, et al. Age and sex variability and normal 135. Krejza J, Mariak Z, Bert RJ. Transcranial colour Doppler sonography in
reference values for the V(MCA)/V(ICA) index. AJNR Am J Neuroradiol emergency management of intracerebral haemorrhage caused by an
2005;26(4):730–5. arteriovenous malformation: case report. Neuroradiology
112. Swiercz M, Kochanowicz J, Weigele J, et al. Learning vector quantization 2000;42(12):900–4.
neural networks improve accuracy of transcranial color-coded duplex 136. Becker G, Winkler J, Hofmann E, et al. Differentiation between ischemic
sonography in detection of middle cerebral artery spasm-preliminary and hemorrhagic stroke by transcranial color-coded real-time sonography.
report. Neuroinformatics 2008;6(4):279–90. J Neuroimaging 1993;3(1):41–7.
113. Soustiel JF, Bruk B, Shik B, et al. Transcranial Doppler in vertebrobasilar 137. Seidel G, Kaps M, Dorndorf W. Transcranial color-coded duplex
vasospasm after subarachnoid hemorrhage. Neurosurgery 1998;43(2): sonography of intracerebral hematomas in adults. Stroke
282–91. 1993;24(10):1519–27.
114. Newell DW, Grady MS, Eskridge JM, et al. Distribution of angiographic 138. Maurer M, Shambal S, Berg D, et al. Differentiation between intracerebral
vasospasm after subarachnoid hemorrhage: implications for diagnosis by hemorrhage and ischemic stroke by transcranial color-coded duplex-
transcranial Doppler ultrasonography. Neurosurgery 1990;27(4):574–7. sonography. Stroke 1998;29(12):2563–7.
115. Sloan MA, Haley Jr EC, Kassell NF, et al. Sensitivity and specificity of 139. Baumgartner RW, Mattle HP, Schroth G. Transcranial colour-coded duplex
transcranial Doppler ultrasonography in the diagnosis of vasospasm sonography of cerebral arteriovenous malformations. Neuroradiology 1996;
following subarachnoid hemorrhage. Neurology 1989;39(11):1514–18. 38(8):734–7.
116. Allendoerfer J, Goertler M, von Reutern GM. Prognostic relevance of 140. Griewing B, Motsch L, Piek J, et al. Transcranial power mode Doppler
ultra-early Doppler sonography in acute ischaemic stroke: a prospective duplex sonography of intracranial aneurysms. J Neuroimaging 1998;8(3):
multicentre study. Lancet Neurol 2006;5(10):835–40. 155–8.
117. Gerriets T, Goertler M, Stolz E, et al. Feasibility and validity of transcranial 141. Klotzsch C, Bozzato A, Lammers G, et al. Three-dimensional transcranial
duplex sonography in patients with acute stroke. J Neurol Neurosurg color-coded sonography of cerebral aneurysms. Stroke 1999;30(11):
Psychiatry 2002;73(1):17–20. 2285–90.
118. Malferrari G, Bertolino C, Casoni F, et al. The eligible study: ultrasound 142. Aries MJ, Elting JW, De Keyser J, et al. Cerebral autoregulation in stroke: a
assessment in acute ischemic stroke within 3 hours. Cerebrovasc Dis review of transcranial Doppler studies. Stroke 2010;41(11):2697–704. Epub
2007;24(5):469–76. 2010, Oct 7.
119. Sekoranja L, Loulidi J, Yilmaz H, et al. Intravenous versus combined 143. Eames PJ, Blake MJ, Dawson SL, et al. Dynamic cerebral autoregulation and
(intravenous and intra-arterial) thrombolysis in acute ischemic stroke: a beat to beat blood pressure control are impaired in acute ischaemic stroke.
transcranial color-coded duplex sonography–guided pilot study. Stroke J Neurol Neurosurg Psychiatry 2002;72(4):467–72.
2006;37(7):1805–9. 144. Reinhard M, Wihler C, Roth M, et al. Cerebral autoregulation dynamics in
120. Wong KS, Li H, Chan YL, et al. Use of transcranial Doppler ultrasound to acute ischemic stroke after rtPA thrombolysis. Cerebrovasc Dis 2008;26(2):
predict outcome in patients with intracranial large-artery occlusive disease. 147–55.
Stroke 2000;31(11):2641–7. 145. Reinhard M, Rutsch S, Lambeck J, et al. Dynamic cerebral autoregulation
121. Wunderlich MT, Stolz E, Seidel G, et al. Conservative medical treatment associates with infarct size and outcome after ischemic stroke. Acta Neurol
and intravenous thrombolysis in acute stroke from carotid T occlusion. Scand 2012;125(3):156–62. Epub 2011, Apr 6.
Cerebrovasc Dis 2005;20(5):355–61. 146. Gosling RG, King DH. Arterial assessment by Doppler-shift ultrasound.
122. Wunderlich MT, Goertler M, Postert T, et al. Recanalization after Proc R Soc Med 1974;67(6 Pt 1):447–9.
intravenous thrombolysis: does a recanalization time window exist? 147. Derdeyn CP, Grubb Jr RL, Powers WJ. Cerebral hemodynamic impairment:
Neurology 2007;68(17):1364–8. methods of measurement and association with stroke risk. Neurology 1999;
123. Alexandrov AV, Molina CA, Grotta JC, et al. Ultrasound-enhanced systemic 53(2):251–9.
thrombolysis for acute ischemic stroke. N Engl J Med 2004;351(21):2170–8. 148. Powers WJ. Cerebral hemodynamics in ischemic cerebrovascular disease.
124. Demchuk AM, Burgin WS, Christou I, et al. Thrombolysis in brain Ann Neurol 1991;29(3):231–40.
ischemia (TIBI) transcranial Doppler flow grades predict clinical severity, 149. Kuroda S, Houkin K, Kamiyama H, et al. Long-term prognosis of medically
early recovery, and mortality in patients treated with intravenous tissue treated patients with internal carotid or middle cerebral artery occlusion:
plasminogen activator. Stroke 2001;32(1):89–93. can acetazolamide test predict it? Stroke 2001;32(9):2110–16.
125. Molina CA, Ribo M, Rubiera M, et al. Microbubble administration 150. King A, Serena J, Bornstein NM, et al. Does impaired cerebrovascular
accelerates clot lysis during continuous 2-MHz ultrasound monitoring in reactivity predict stroke risk in asymptomatic carotid stenosis? A
stroke patients treated with intravenous tissue plasminogen activator. prospective substudy of the asymptomatic carotid emboli study. Stroke
Stroke 2006;37(2):425–9. 2011;42(6):1550–5
126. Hackworthy RA, Sorensen SG, Fitzpatrick PG, et al. Dependence of 151. Failure of extracranial-intracranial arterial bypass to reduce the risk of
assessment of coronary artery reperfusion during acute myocardial ischemic stroke. Results of an international randomized trial. The EC/IC
infarction on angiographic criteria and interobserver variability. Am J Bypass Study Group. N Engl J Med 1985;313(19):1191–200.
Cardiol 1988;62(9):538–42. 152. Adams HP, Jr., Powers WJ, Grubb RL, Jr., et al. Preview of a new trial of
127. Gahn G, Gerber J, Hallmeyer S, et al. Contrast-enhanced transcranial extracranial-to-intracranial arterial anastomosis: the carotid occlusion
color-coded duplex sonography in stroke patients with limited bone surgery study. Neurosurg Clin N Am 2001;12(3):613–61x.
windows. AJNR Am J Neuroradiol 2000;21(3):509–14. 153. Karnik R, Valentin A, Ammerer HP, et al. Evaluation of vasomotor
128. Gerriets T, Postert T, Goertler M, et al. DIAS I: duplex-sonographic reactivity by transcranial Doppler and acetazolamide test before and after
assessment of the cerebrovascular status in acute stroke—a useful tool for extracranial-intracranial bypass in patients with internal carotid artery
future stroke trials. Stroke 2000;31(10):2342–5. occlusion. Stroke 1992;23(6):812–17.
313.e4 Section V—Cerebral Blood Flow

154. Vorstrup S, Brun B, Lassen NA. Evaluation of the cerebral vasodilatory experimental intracranial hypertension. J Neurol Neurosurg Psychiatry
capacity by the acetazolamide test before EC-IC bypass surgery in patients 1974;37(4):392–402.
with occlusion of the internal carotid artery. Stroke 1986;17(6):1291–8. 178. Hadani M, Bruk B, Ram Z, et al. Application of transcranial doppler
155. Kuroda S, Kamiyama H, Abe H, et al. Acetazolamide test in detecting ultrasonography for the diagnosis of brain death. Intensive Care Med 1999;
reduced cerebral perfusion reserve and predicting long-term prognosis in 25(8):822–8.
patients with internal carotid artery occlusion. Neurosurgery 1993;32(6): 179. Marinoni M, Alari F, Mastronardi V, et al. The relevance of early TCD
912–18. monitoring in the intensive care units for the confirming of brain death
156. Schmiedek P, Piepgras A, Leinsinger G, et al. Improvement of diagnosis. Neurol Sci 2011;32(1):73–7. Epub 2010, Sep 25.
cerebrovascular reserve capacity by EC-IC arterial bypass surgery in 180. Sharma D, Souter MJ, Moore AE, et al. Clinical experience with transcranial
patients with ICA occlusion and hemodynamic cerebral ischemia. Doppler ultrasonography as a confirmatory test for brain death: a
J Neurosurg 1994;81(2):236–44. retrospective analysis. Neurocrit Care 2011;14(3):370–6.
157. European Stroke Organisation (ESO) Executive Committee; ESO Writing 181. Soldatos T, Karakitsos D, Wachtel M, et al. The value of transcranial
Committee. Guidelines for management of ischaemic stroke and transient Doppler sonography with a transorbital approach in the confirmation of
ischaemic attack 2008. Cerebrovasc Dis 2008;25(5):457–507. cerebral circulatory arrest. Transplant Proc 2010;42(5):1502–6.
158. Adams HP, del Zoppo G, Alberts MJ, et al. Guidelines for the early 182. Ducrocq X, Hassler W, Moritake K, et al. Consensus opinion on diagnosis
management of adults with ischemic stroke: a guideline from the American of cerebral circulatory arrest using Doppler-sonography: Task Force Group
Heart Association/American Stroke Association Stroke Council, Clinical on cerebral death of the Neurosonology Research Group of the World
Cardiology Council, Cardiovascular Radiology and Intervention Council, Federation of Neurology. J Neurol Sci 1998;159(2):145–50.
and the Atherosclerotic Peripheral Vascular Disease and Quality of Care 183. Krejza J, Fryc J, Owlasiuk M, et al. Transcranial color Doppler sonography
Outcomes in Research Interdisciplinary Working Groups. Stroke 2007; in the emergency diagnosis of middle cerebral artery occlusion in a patient
38(5):1655–711. after head injury. Clin Imaging 2001;25(2):90–4.
159. Goldstein LB, Bushnell CD, Adams RJ, et al. Guidelines for the primary 184. Llompart-Pou JA, Abadal JM, Velasco J, et al. Contrast-enhanced
prevention of stroke: a guideline for healthcare professionals from the transcranial color sonography in the diagnosis of cerebral circulatory arrest.
American Heart Association/American Stroke Association. Stroke 2011; Transplant Proc 2009;41(5):1466–8
42(2):517–84. 185. Sorrentino E, Budohoski KP, Kasprowicz M, et al. Critical thresholds for
160. Furie KL, Kasner SE, Adams RJ, et al. Guidelines for the prevention of transcranial Doppler indices of cerebral autoregulation in traumatic brain
stroke in patients with stroke or transient ischemic attack: a guideline for injury. Neurocrit Care 2011;14(2):188–93.
healthcare professionals from the American heart association/American 186. Melo JR, Di Rocco F, Blanot S, et al. Transcranial Doppler can predict
stroke association. Stroke 2011;42(1):227–76. intracranial hypertension in children with severe traumatic brain injuries.
161. Iwama T, Hashimoto N, Takagi Y, et al. Predictability of extracranial/ Childs Nerv Syst 2011;27(6):979–84. Epub 2011, Jan 5.
intracranial bypass function: a retrospective study of patients with occlusive 187. Martin NA, Doberstein C, Zane C, et al. Posttraumatic cerebral arterial
cerebrovascular disease. Neurosurgery 1997;40(1):53–9. spasm: transcranial Doppler ultrasound, cerebral blood flow, and
162. Nariai T, Suzuki R, Matsushima Y, et al. Surgically induced angiogenesis to angiographic findings. J Neurosurg 1992;77(4):575–83.
compensate for hemodynamic cerebral ischemia. Stroke 1994;25(5):1014–21. 188. Steiger HJ, Aaslid R, Stooss R, et al. Transcranial Doppler monitoring in
163. Ogasawara K, Ogawa A, Terasaki K, et al. Use of cerebrovascular reactivity head injury: relations between type of injury, flow velocities, vasoreactivity,
in patients with symptomatic major cerebral artery occlusion to predict and outcome. Neurosurgery 1994;34(1):79–85.
5-year outcome: comparison of xenon-133 and iodine-123-IMP single- 189. Zurynski YA, Dorsch NW, Fearnside MR. Incidence and effects of increased
photon emission computed tomography. J Cereb Blood Flow Metab 2002; cerebral blood flow velocity after severe head injury: a transcranial Doppler
22(9):1142–8. ultrasound study. II. Effect of vasospasm and hyperemia on outcome.
164. Widder B, Kleiser B, Krapf H. Course of cerebrovascular reactivity in J Neurol Sci 1995;134(1-2):41–6.
patients with carotid artery occlusions. Stroke 1994;25(10):1963–7. 190. Kochanowicz J, Krejza J, Mariak Z, et al. Detection and monitoring of
165. Henderson RD, Phan TG, Piepgras DG, et al. Mechanisms of intracerebral cerebral hemodynamic disturbances with transcranial color-coded duplex
hemorrhage after carotid endarterectomy. J Neurosurg 2001;95(6):964–69. sonography in patients after head injury. Neuroradiology 2006;48(1):
166. Piepgras DG, Morgan MK, Sundt TM, Jr., et al. Intracerebral hemorrhage 31–6.
after carotid endarterectomy. J Neurosurg 1988;68(4):532–6. 191. Len TK, Neary JP. Cerebrovascular pathophysiology following mild
167. Zhang R, Zhou G, Xu G, et al. Posterior circulation hyperperfusion traumatic brain injury. Clin Physiol Funct Imaging 2011;31(2):85–93.
syndrome after bilateral vertebral artery intracranial stenting. Ann Vasc 192. Brandi G, Béchir M, Sailer S, et al. Transcranial color-coded duplex
Surg 2009;23(5):686.e1–5. sonography allows assessing cerebral perfusion pressure noninvasively
168. Sfyroeras GS, Karkos CD, Arsos G, et al. Cerebral hyperperfusion after following severe traumatic brain injury. Acta Neurochir (Wien)
carotid stenting: a transcranial Doppler and SPECT study. Vasc 2010;152(6):965–72.
Endovascular Surg 2009;43(2):150–6 193. Czosnyka M, Smielewski P, Kirkpatrick P, et al. Continuous assessment of
169. Hosoda K, Kawaguchi T, Ishii K, et al. Prediction of hyperperfusion after the cerebral vasomotor reactivity in head injury. Neurosurgery
carotid endarterectomy by brain SPECT analysis with semiquantitative 1997;41(1):11–7.
statistical mapping method. Stroke 2003;34(5):1187–93. 194. Newell DW, Weber JP, Watson R, et al. Effect of transient moderate
170. Reigel MM, Hollier LH, Kazmier FJ, et al. Late survival in abdominal aortic hyperventilation on dynamic cerebral autoregulation after severe head
aneurysm patients: the role of selective myocardial revascularization on the injury. Neurosurgery 1996;39(1):35–43.
basis of clinical symptoms. J Vasc Surg 1987;5(2):222–7. 195. Budohoski KP, Reinhard M, Aries MJ, et al. Monitoring cerebral
171. Pennekamp CW, Tromp SC, Ackerstaff RG, et al. Prediction of cerebral autoregulation after head injury. Which component of transcranial Doppler
hyperperfusion after carotid endarterectomy with transcranial Doppler. Eur flow velocity is optimal? Neurocrit Care 2011;Jun 21. Epub ahead of print.
J Vasc Endovasc Surg 2012;Jan 18. Epub ahead of print. 196. Smielewski P, Czosnyka M, Kirkpatrick P, et al. Evaluation of the transient
172. Jain R, Hoeffner EG, Deveikis JP, et al. Carotid perfusion CT with balloon hyperemic response test in head-injured patients. J Neurosurg 1997;86(5):
occlusion and acetazolamide challenge test: feasibility. Radiology 2004; 773–8.
231(3):906–13. 197. Muizelaar JP, Lutz HA, III, Becker DP. Effect of mannitol on ICP and CBF
173. Okudaira Y, Arai H, Sato K. Cerebral blood flow alteration by and correlation with pressure autoregulation in severely head-injured
acetazolamide during carotid balloon occlusion: parameters reflecting patients. J Neurosurg 1984;61(4):700–6.
cerebral perfusion pressure in the acetazolamide test. Stroke 1996;27(4): 198. Strebel S, Lam AM, Matta BF, et al. Impaired cerebral autoregulation after
617–21. mild brain injury. Surg Neurol 1997;47(2):128–31.
174. Mokri B. The Monro-Kellie hypothesis: applications in CSF volume 199. Larsen FS, Ejlersen E, Hansen BA, et al. Functional loss of cerebral blood
depletion. Neurology 2001;56(12):1746–8. flow autoregulation in patients with fulminant hepatic failure. J Hepatol
175. Kingman TA, Mendelow AD, Graham DI, et al. Experimental intracerebral 1995;23(2):212–17.
mass: time-related effects on local cerebral blood flow. J Neurosurg 1987; 200. Larsen FS, Olsen KS, Ejlersen E, et al. Cerebral blood flow autoregulation
67(5):732–8. and transcranial Doppler sonography in patients with cirrhosis. Hepatology
176. Sahay KB, Mehrotra R, Sachdeva U, et al. Elastomechanical characterization 1995;22(3):730–6.
of brain tissues. J Biomech 1992;25(3):319–26. 201. Strauss GI, Hansen BA, Herzog T, et al. Cerebral autoregulation in patients
177. Johnston IH, Rowan JO. Raised intracranial pressure and cerebral blood with end-stage liver disease. Eur J Gastroenterol Hepatol
flow. 3. Venous outflow tract pressures and vascular resistances in 2000;12(7):767–71.
Section V—Cerebral Blood Flow 313.e5

202. Larsen FS, Ejlersen E, Strauss G, et al. Cerebrovascular metabolic 226. Ackerstaff RG, Suttorp MJ, van den Berg JC, et al. Prediction of early
autoregulation is impaired during liver transplantation. Transplantation cerebral outcome by transcranial Doppler monitoring in carotid bifurcation
1999;68(10):1472–6. angioplasty and stenting. J Vasc Surg 2005;41(4):618–24.
203. Larsen FS, Pott F, Hansen BA, et al. Transcranial Doppler sonography may 227. Rosenkranz M, Fiehler J, Niesen W, et al. The amount of solid cerebral
predict brain death in patients with fulminant hepatic failure. Transplant microemboli during carotid stenting does not relate to the frequency of
Proc 1995;27(6):3510–11. silent ischemic lesions. AJNR Am J Neuroradiol 2006;27(1):157–61.
204. de Riva N, Budohoski KP, Smielewski P, et al. Transcranial Doppler 228. Sawada M, Hashimoto N, Nishi S, et al. Detection of embolic signals during
pulsatility index: what it is and what it isn’t. Neurocrit Care 2012;Feb 4. and after percutaneous transluminal angioplasty of subclavian and
Epub ahead of print. vertebral arteries using transcranial Doppler ultrasonography. Neurosurgery
205. King A, Shipley M, Markus H, et al. Optimizing protocols for risk 1997;41(3):535–40.
prediction in asymptomatic carotid stenosis using embolic signal detection: 229. Pelz DM, Lownie SP, Fox AJ. Thromboembolic events associated with the
the Asymptomatic Carotid Emboli Study. Stroke 2011;42(10):2819–24. treatment of cerebral aneurysms with Guglielmi detachable coils. AJNR Am
Epub 2011, Aug 18. J Neuroradiol 1998;19(8):1541–7.
206. King A, Bath PM, Markus HS. Clopidogrel versus dipyridamole in addition 230. Song JK, Niimi Y, Fernandez PM, et al. Thrombus formation during
to aspirin in reducing embolization detected with ambulatory transcranial intracranial aneurysm coil placement: treatment with intra-arterial
Doppler: a randomized trial. Stroke 2011 Mar;42(3):650–5. Epub 2011, abciximab. AJNR Am J Neuroradiol 2004;25(7):1147–53.
Jan 21. 231. Ries T, Buhk JH, Kucinski T, et al. Intravenous administration of
207. Executive Committee for the Asymptomatic Carotid Atherosclerosis Study. acetylsalicylic acid during endovascular treatment of cerebral aneurysms
Endarterectomy for asymptomatic carotid artery stenosis. JAMA 1995; reduces the rate of thromboembolic events. Stroke 2006;37(7):1816–21.
273(18):1421–8. 232. Gilman S. Cerebral disorders after open-heart operations. N Engl J Med
208. Hartmann A, Hupp T, Koch HC, et al. Prospective study on the 1965;272:489–98.
complication rate of carotid surgery. Cerebrovasc Dis 1999;9(3):152–6. 233. McKhann GM, Goldsborough MA, Borowicz Jr LM, et al. Cognitive
209. Spencer MP. Transcranial Doppler monitoring and causes of stroke from outcome after coronary artery bypass: a one-year prospective study. Ann
carotid endarterectomy. Stroke 1997;28(4):685–91. Thorac Surg 1997;63(2):510–15.
210. Ackerstaff RG, Moons KG, van de Vlasakker CJ, et al. Association of 234. Blauth CI, Smith PL, Arnold JV, et al. Influence of oxygenator type on the
intraoperative transcranial doppler monitoring variables with stroke from prevalence and extent of microembolic retinal ischemia during
carotid endarterectomy. Stroke 2000;31(8):1817–23. cardiopulmonary bypass. Assessment by digital image analysis. J Thorac
211. Kofke WA, Brauer P, Policare R, et al. Middle cerebral artery blood flow Cardiovasc Surg 1990;99(1):61–9.
velocity and stable xenon-enhanced computed tomographic blood flow 235. Gravlee GP, Hudspeth AS, Toole JF. Bilateral brachial paralysis from
during balloon test occlusion of the internal carotid artery. Stroke 1995; watershed infarction after coronary artery bypass. A report of two cases and
26(9):1603–6. review of the predisposing anatomic and physiological mechanisms.
212. Halsey Jr JH. Risks and benefits of shunting in carotid endarterectomy. The J Thorac Cardiovasc Surg 1984;88(5 Pt 1):742–7.
International Transcranial Doppler Collaborators. Stroke 1992;23(11): 236. Padayachee TS, Parsons S, Theobold R, et al. The detection of microemboli
1583–7. in the middle cerebral artery during cardiopulmonary bypass: a
213. Hayes PD, Vainas T, Hartley S, et al. The Pruitt-Inahara shunt transcranial Doppler ultrasound investigation using membrane and bubble
maintains mean middle cerebral artery velocities within 10% of oxygenators. Ann Thorac Surg 1987;44(3):298–302.
preoperative values during carotid endarterectomy. J Vasc Surg 2000; 237. Pugsley W, Klinger L, Paschalis C, et al. The impact of microemboli during
32(2):299–306. cardiopulmonary bypass on neuropsychological functioning. Stroke 1994;
214. Babikian VL, Cantelmo NL, Wijman CAC. Neurovascular monitoring 25(7):1393–9.
during carotid endarterectomy. In: Babikian VL, Wechsler LR, editors. 238. Vanninen R, Aikia M, Kononen M, et al. Subclinical cerebral complications
Transcranial Doppler ultrasonography. 2nd ed. Boston: Butterworth- after coronary artery bypass grafting: prospective analysis with magnetic
Heinemann; 1999. p. 231–45. resonance imaging, quantitative electroencephalography, and
215. Cao P, Giordano G, Zannetti S, et al. Transcranial Doppler monitoring neuropsychological assessment. Arch Neurol 1998;55(5):618–27.
during carotid endarterectomy: is it appropriate for selecting patients in 239. Wityk RJ, Goldsborough MA, Hillis A, et al. Diffusion- and perfusion-
need of a shunt? J Vasc Surg 1997;26(6):973–9. weighted brain magnetic resonance imaging in patients with neurologic
216. Frawley JE, Hicks RG, Gray LJ, et al. Carotid endarterectomy without a complications after cardiac surgery. Arch Neurol 2001;58(4):571–6.
shunt for symptomatic lesions associated with contralateral severe stenosis 240. Clark RE, Brillman J, Davis DA, et al. Microemboli during coronary artery
or occlusion. J Vasc Surg 1996;23(3):421–7. bypass grafting: genesis and effect on outcome. J Thorac Cardiovasc Surg
217. Levi CR, O’Malley HM, Fell G, et al. Transcranial Doppler detected cerebral 1995;109(2):249–57.
microembolism following carotid endarterectomy. High microembolic 241. Challa VR, Moody DM, Bell MA. Small capillary and arteriolar dilatations
signal loads predict postoperative cerebral ischaemia. Brain 1997; (SCADS) in brain after cardiopulmonary bypass. In: McKhan GM, Gibbs
120 (Pt 4):621–9. CJ, editors. Neurological outcomes after coronary bypass surgery. New
218. Cantelmo NL, Babikian VL, Samaraweera RN, et al. Cerebral York: Thieme; 1993. p. 330–4.
microembolism and ischemic changes associated with carotid 242. Moody DM, Bell MA, Challa VR, et al. Brain microemboli during cardiac
endarterectomy. J Vasc Surg 1998;27(6):1024–30. surgery or aortography. Ann Neurol 1990;28(4):477–86.
219. Jansen C, Ramos LM, van Heesewijk JP, et al. Impact of microembolism 243. Deverall PB, Padayachee TS, Parsons S, et al. Ultrasound detection of
and hemodynamic changes in the brain during carotid endarterectomy. micro-emboli in the middle cerebral artery during cardiopulmonary bypass
Stroke 1994;25(5):992–7. surgery. Eur J Cardiothorac Surg 1988;2(4):256–60.
220. Lennard N, Smith J, Dumville J, et al. Prevention of postoperative 244. Barbut D, Hinton RB, Szatrowski TP, et al. Cerebral emboli detected during
thrombotic stroke after carotid endarterectomy: the role of transcranial bypass surgery are associated with clamp removal. Stroke
Doppler ultrasound. J Vasc Surg 1997;26(4):579–84. 1994;25(12):2398–402.
221. Jordan WD, Jr., Voellinger DC, Doblar DD, et al. Microemboli detected by 245. Barbut D, Yao FS, Hager DN, et al. Comparison of transcranial Doppler
transcranial Doppler monitoring in patients during carotid angioplasty ultrasonography and transesophageal echocardiography to monitor emboli
versus carotid endarterectomy. Cardiovasc Surg 1999;7(1):33–8. during coronary artery bypass surgery. Stroke 1996;27(1):87–90.
222. Gupta N, Corriere MA, Dodson TF, et al. The incidence of microemboli to 246. Yao FS, Barbut D, Hager DN, et al. Detection of aortic emboli by
the brain is less with endarterectomy than with percutaneous transesophageal echocardiography during coronary artery bypass surgery.
revascularization with distal filters or flow reversal. J Vasc Surg 2011;53(2): J Cardiothorac Vasc Anesth 1996;10(3):314–17.
316–22. Epub 2010, Dec 3. 247. Barbut D, Caplan LR. Brain complications of cardiac surgery. Curr Probl
223. Reimers B, Corvaja N, Moshiri S, et al. Cerebral protection with filter Cardiol 1997;22(9):449–80.
devices during carotid artery stenting. Circulation 2001;104(1):12–5. 248. Gardner TJ, Horneffer PJ, Manolio TA, et al. Stroke following coronary
224. Jeong HS, Song HJ, Lee JH, et al. Interpretation of TCD spectral patterns artery bypass grafting: a ten-year study. Ann Thorac Surg
detected during carotid artery stent interventions. J Endovasc Ther 2011; 1985;40(6):574–81.
18(4):518–26. 249. Roach GW, Kanchuger M, Mangano CM, et al. Adverse cerebral outcomes
225. Antonius Carotid Endaterectomy, Angioplasty, and Stenting Study Group. after coronary bypass surgery. Multicenter Study of Perioperative Ischemia
Transcranial Doppler monitoring in angioplasty and stenting of the carotid Research Group and the Ischemia Research and Education Foundation
bifurcation. J Endovasc Ther 2003;10(4):702–10. Investigators. N Engl J Med 1996;335(25):1857–63.
313.e6 Section V—Cerebral Blood Flow

250. Caplan LR. Protecting the brains of patients after heart surgery. Arch 267. Dijkmans PA, Knaapen P, Sieswerda GT, et al. Quantification of myocardial
Neurol 2001;58(4):549–50. perfusion using intravenous myocardial contrast echocardiography in
251. Blinc A, Kennedy SD, Bryant RG, et al. Flow through clots determines the healthy volunteers: comparison with positron emission tomography. J Am
rate and pattern of fibrinolysis. Thromb Haemost 1994;71(2):230–5. Soc Echocardiogr 2006;19(3):285–93.
252. Francis CW, Blinc A, Lee S, et al. Ultrasound accelerates transport of 268. Cintas P, Nguyen F, Boneu B, et al. Enhancement of enzymatic fibrinolysis
recombinant tissue plasminogen activator into clots. Ultrasound Med Biol with 2-MHz ultrasound and microbubbles. J Thromb Haemost 2004;2(7):
1995;21(3):419–24. 1163–6.
253. Behrens S, Daffertshofer M, Spiegel D, et al. Low-frequency, low-intensity 269. Culp WC, Porter TR, Lowery J, et al. Intracranial clot lysis with intravenous
ultrasound accelerates thrombolysis through the skull. Ultrasound Med microbubbles and transcranial ultrasound in swine. Stroke 2004;35(10):
Biol 1999;25(2):269–73. 2407–11.
254. Akiyama M, Ishibashi T, Yamada T, et al. Low-frequency ultrasound 270. Mizushige K, Kondo I, Ohmori K, et al. Enhancement of ultrasound-
penetrates the cranium and enhances thrombolysis in vitro. Neurosurgery accelerated thrombolysis by echo contrast agents: dependence on
1998;43(4):828–32. microbubble structure. Ultrasound Med Biol 1999;25(9):1431–7.
255. Spengos K, Behrens S, Daffertshofer M, et al. Acceleration of thrombolysis 271. Saqqur M, Tsivgoulis G, Molina CA, et al. Design of a PROspective
with ultrasound through the cranium in a flow model. Ultrasound Med multi-national CLOTBUST collaboration on reperfusion therapies for
Biol 2000;26(5):889–95. stroke (CLOTBUST-PRO). Int J Stroke 2008;3(1):66–72.
256. Kramer C, Aguilar MI, Hoffman-Snyder C, et al. Safety and efficacy of 272. Alexandrov AV, Demchuk AM, Burgin WS, et al. Ultrasound-enhanced
ultrasound-enhanced thrombolysis in the treatment of acute middle thrombolysis for acute ischemic stroke. Phase I. Findings of the
cerebral artery infarction: a critically appraised topic. Neurologist 2011; CLOTBUST trial. J Neuroimaging 2004;14(2):113–17.
17(6):346–51. 273. Rosenschein U, Rozenszajn LA, Kraus L, et al. Ultrasonic angioplasty in
257. Rubiera M, Alexandrov AV. Sonothrombolysis in the management of acute totally occluded peripheral arteries: initial clinical, histological, and
ischemic stroke. Am J Cardiovasc Drugs 2010;10(1):5–10. angiographic results. Circulation 1991;83(6):1976–86.
258. Tsivgoulis G, Eggers J, Ribo M, et al. Safety and efficacy of ultrasound- 274. Siegel RJ, Cumberland DC, Crew JR. Ultrasound recanalization of diseased
enhanced thrombolysis: a comprehensive review and meta-analysis of arteries: from experimental studies to clinical application. Surg Clin North
randomized and nonrandomized studies. Stroke 2010;41(2):280–7. Epub Am 1992;72(4):879–97.
2009, Dec 31. 275. Luo H, Steffen W, Cercek B, et al. Enhancement of thrombolysis by external
259. Alexandrov AV, Demchuk AM, Felberg RA, et al. High rate of complete ultrasound. Am Heart J 1993;125(6):1564–9.
recanalization and dramatic clinical recovery during tPA infusion when 276. Steffen W, Cumberland D, Gaines P, et al. Catheter-delivered high intensity,
continuously monitored with 2-MHz transcranial Doppler monitoring. low frequency ultrasound induces vasodilation in vivo. Eur Heart J 1994;
Stroke 2000;31(3):610–14. 15(3):369–76.
260. Siddiqi F, Odrljin TM, Fay PJ, et al. Binding of tissue-plasminogen activator 277. Rosenschein U, Bernstein JJ, DiSegni E, et al. Experimental ultrasonic
to fibrin: effect of ultrasound. Blood 1998;91(6):2019–25. angioplasty: disruption of atherosclerotic plaques and thrombi in vitro and
261. Siddiqi F, Blinc A, Braaten J, et al. Ultrasound increases flow through fibrin arterial recanalization in vivo. J Am Coll Cardiol 1990;15(3):711–17.
gels. Thromb Haemost 1995;73(3):495–8. 278. Hong AS, Chae JS, Dubin SB, et al. Ultrasonic clot disruption: an in vitro
262. Polak JF. Ultrasound energy and the dissolution of thrombus. N Engl J Med study. Am Heart J 1990;120(2):418–22.
2004;351(21):2154–5. 279. Sehgal CM, Leveen RF, Shlansky-Goldberg RD. Ultrasound-assisted
263. Krejza J, Weigele JB, Alokaili R, et al. Sonothrombolysis in acute ischemic thrombolysis. Invest Radiol 1993;28(10):939–43.
stroke for patients ineligible for rt-PA. Neurology 2006;66(1):154–5. 280. Suchkova V, Siddiqi FN, Carstensen EL, et al. Enhancement of fibrinolysis
264. Tachibana K, Tachibana S. Albumin microbubble echo-contrast material as with 40-kHz ultrasound. Circulation 1998;98(10):1030–5.
an enhancer for ultrasound accelerated thrombolysis. Circulation 1995; 281. Olsson SB, Johansson B, Nilsson AM, et al. Enhancement of thrombolysis
92(5):1148–50. by ultrasound. Ultrasound Med Biol 1994;20(4):375–82.
265. Molina CA, Ribo M, Rubiera M, et al. Microbubble administration 282. Blinc A, Francis CW, Trudnowski JL, et al. Characterization of ultrasound-
accelerates clot lysis during continuous 2-MHz ultrasound monitoring in potentiated fibrinolysis in vitro. Blood 1993;81(10):2636–43.
stroke patients treated with intravenous tissue plasminogen activator. 283. Lauer CG, Burge R, Tang DB, et al. Effect of ultrasound on tissue-type
Stroke 2006;37(2):425–9. plasminogen activator-induced thrombolysis. Circulation
266. Xie F, Tsutsui JM, Lof J, et al. Effectiveness of lipid microbubbles and 1992;86(4):1257–64.
ultrasound in declotting thrombosis. Ultrasound Med Biol 2005;31(7):
979–85.
V
Chapter
31  

Laser Doppler Flowmetry,


Thermal Diffusion
Flowmetry, and Orthogonal
Polarizing Spectral Imaging
Frederik A. Pennings

metabolism are recognized. Local microvascular CBF can be


Introduction measured continuously and invasively in the neurocritical
Brain functional and structural integrity depends on a con- care unit (NCCU) with laser Doppler flowmetry (LDF) and
stant supply of oxygen and glucose through cerebral blood thermal diffusion flowmetry (TDF). Many years of clinical
flow (CBF) to the cerebral microcirculation. Accordingly, experience have been obtained with these monitoring modali-
adequate function of the cerebral microcirculation is a prereq- ties. This chapter gives an overview of LDF and TDF: history,
uisite for sufficient oxygen (O2) delivery to brain cells. Under technology, validation, clinical applications, and develop-
normal conditions, the brain makes adjustments in local CBF ments are described. In addition, orthogonal polarizing spec-
to support metabolic demands and prevent energy failure. tral (OPS) imaging and its successor sidestream dark field
However, if an imbalance occurs between supply (CBF) and (SDF) imaging are discussed as examples of optical techniques
consumption (cerebral metabolic rate of oxygen [CMRO2]), that allow direct observation of the human microcirculatory
cerebral ischemia may develop. If CBF falls below a critical morphology and function.
level and oxygen extraction has reached its limit, electrical and
certain metabolic functions are lost, and irreversible brain
damage (infarction) may occur.1 The development of tissue
Laser Doppler Flowmetry
infarction is related to both the degree and duration of CBF Bedside evaluation of CBF has been a long-standing goal in
reduction.2 This important concept implicates that certain neurocritical care. Potentially, it could assess flow alterations,
thresholds exist below which infarction occurs and that there measure the effect of therapy, and evaluate the integrity of
is a limited period of time that ischemia may still be reversible cerebral autoregulation. LDF was first introduced in 1977 to
(“therapeutic window”). Therefore, it may be helpful to have measure cutaneous blood flow, and after commercialization
monitoring instruments that can detect cerebral ischemia in the early 1980s, a stable and steadily growing clinical appli-
before these thresholds are crossed and allow the clinician to cation has occurred.4,5 LDF provides continuous, qualitative
take measures in a timely manner to prevent the development measurements of microvascular perfusion intraoperatively or
of brain infarction. In addition instruments are needed that in the intensive care unit as a bedside monitoring device; this
provide information on morphology and function of the is attractive to clinicians working with critical neurologic dis-
cerebral microcirculation to better understand the pathophys- eases in which cerebral ischemia is frequently encountered.
iology of cerebral ischemia. In particular, continuous assess- However, the routine clinical applicability of this optical tech-
ment of CBF helps to assess cerebral autoregulation and nique remains logistically complex and is hindered by various
pressure-reactivity. This is important because both can be artifacts and is limited by the absence of quantitative regional
independent factors associated with outcome after acute brain cerebral blow flow (rCBF) values. Consequently diagnostic
injury.3 and prognostic values have not been reported and instead LDF
Current treatment modalities to prevent or reduce ischemic is best used as a trend monitor in individual patients.
brain damage are mostly guided by parameters such as intra- In LDF, a fiber-optic laser probe with a diameter of 0.5 to
cranial pressure (ICP), cerebral perfusion pressure (CPP), 1 mm is applied to the surface of the brain or within the brain
brain tissue oxygen tension (PbtO2), and cerebral metabolism to illuminate a tissue volume of approximately 1 mm3 with
and not by direct CBF measurements. Direct monitoring of monochromatic laser light of various wavelengths from 670
CBF could provide the opportunity to diagnose and to correct to 810 nm. As light impinges on the brain tissue, photons are
insufficient CBF before deficits in tissue oxygenation and scattered and Doppler shifted in a random fashion via dynamic
314 © Copyright 2013 Elsevier Inc. All rights reserved.
Section V—Cerebral Blood Flow 315

Non-Doppler shifted light ICP in the early and the acute stages of subarachnoid hemor-
rhage (SAH).19
Scattering In the intensive care unit LDF can be used to help predict
tissue target outcome. For example, Lam et al. continually assessed auto-
regulation with LDF in 31 comatose head-injured patients. A
Photodetector linear relationship between cortical LDF and CPP was used as
Doppler shifted light
an indicator for loss of autoregulation. Intact autoregulation
correlated with a good outcome and persistent loss of auto-
regulation with a poor outcome.20 However, loss of contact
with the cerebral surface was responsible for unreliable data
in 16% of patients. Smielewski et al. assessed the transient
hyperemic response after traumatic brain injury using LDF
Laser diode and found that patients with an unfavorable outcome had
Fig. 31.1  Optical schematic of laser Doppler flowmetry. significantly lower LDF readings than patients with a favorable
outcome.21 The concomitant use of LDF with transcranial
Doppler (TCD) to help detect delayed cerebral ischemia con-
firmed that high-flow velocities and ischemia are not often
red blood cells and stationary tissue cells. A fraction of scat- related.22 LDF also can be used intraoperatively to assess
tered shifted and nonshifted photons is detected by photore- microvascular flow alterations in a region of interest during
ceptors, which generates an electric signal. This electric signal aneurysm surgery, arteriovenous malformation (AVM) resec-
contains information about power and frequency propor- tions, and bypass procedures and to interpret the physiologic
tional to the volume or concentration (fraction of total changes associated with the surgery, understand the cortical
photons that is frequency shifted) and velocity of red blood microcirculation, and help predict postoperative hyper- or
cells (signal frequency broadening) (Fig. 31.1). The commer- hypoperfusion syndromes.23-25 In addition LDF studies have
cially available laser Doppler signal processors use Bonner and been used to assess blood flow in pericranial flaps during
Nossal’s algorithm to analyze the signal and to produce a flow reconstructive surgery.26
output. The product of volume and velocity scales linearly Despite LDF’s excellent temporal and dynamic resolution
with the perfusion (CBF) and is expressed in arbitrary units with ultra short time responses to fluctuations in CBF there
(AUs).6,7 are several limiting factors that may compromise data
The principles of LDF measurements are implemented in quality.27 Different inter- and intraindividual LDF signals can
surface (cortical) probes and intraparenchymal (subcortical) be generated by device-related factors such as calibration pro-
probes to obtain real-time continuous local microvascular cedure, monochromatic wavelength, and probe configura-
arbitrary flow values from capillaries at the cortical surface tion. The penetration depth of LDF is approximately 1 mm,
and within the brain. The cortical probe consists of a thin depending on wavelength distance between transmitting and
contoured disk with a long optical fiber that is connected receiving fibers (fiber separation). A shorter wavelength or a
to a monitor. The probe is positioned subdurally and so can smaller fiber separation will give lesser tissue penetration.
be prone to motion artifacts caused by brain pulsations or Artifacts can be induced by a large number of external
subsidence of brain swelling. To avoid disturbances of the derangements such as the heterogeneity of microvascular
flow values, the probe should be placed in a relatively architecture, changes in hematocrit, tissue or probe motion,
normal brain region without large vessels. The intraparen- room temperature, strong external light, and sound. LDF
chymal probe is introduced through a fixed bolt in the skull, only measures a very small area of brain tissue that may not
which eliminates motion artifacts, and advanced to the necessarily represent the global CBF due to the heterogeneity
white matter. The benefit is that measurements are obtained of the cerebral microcirculation. Multichannel instruments
from the same local brain region as other intracranial moni- and laser Doppler perfusion imaging were introduced to
toring devices such as ICP and microdialysis, which may overcome potential limitations associated with temporal and
improve interpretations of those parameters. The main spatial resolution.28,29 These factors make inter- and intraindi-
manufacturers of laser Doppler instruments are Perimed AB vidual comparison of data difficult. Therefore a detailed stan-
(Stockholm, Sweden), Moor Instruments Ltd. (Axminster, dardized measurement protocol is a prerequisite to obtain
UK), Vasamedics Inc. (St. Paul, MN), Transonic Systems Inc. reproducible and comparable LDF data. This protocol
(Ithaca, NY), Oxford Optronix Ltd (Oxford, UK) and LEA includes at least device-related technical data, anatomic site
Medizintechnik (Giessen, Germany). selection, and frequent calibrations (every 15 minutes) to cir-
LDF provides a reliable measurement of microvascular cumvent improper data collection due to changes in hemato-
(local) rCBF as demonstrated by multiple validation studies. crit, motion, room temperature, strong external light, and
A high correlation is found between LDF and several other sound. Finally, the inability of LDF to produce absolute CBF
blood flow monitoring techniques such as xenon (Xe) clear- values hinders the definition of a threshold value that indi-
ance method, radioactive microspheres, hydrogen clearance cates cerebral ischemia; that is to say, LDF is a trend monitor.
technique, iodoantipyrine method, and thermal diffusion.8-14
Furthermore, it has been successfully applied in the brain-
injured patient to assess autoregulation, carbon dioxide (CO2)
Thermal Diffusion Flowmetry
reactivity, and response to therapeutic interventions and to TDF is based on thermal conductivity of brain tissue to
detect ischemic insults.15-18 For example, Schubert et al. have measure quantitative blood flow. In 1933, Gibbs demonstrated
used LDF studies to detect hypoperfusion independent from that changes in CBF could be detected with a heated
316 Section V—Cerebral Blood Flow

1 mm

Passive
thermistor

Temperature

Measurement diameter = 8 mm
Release of
Active cross clamp
thermistor 8 mm

Release of Hyperemia New CBF


bypass clamp
A B
Fig. 31.2  A, Schematic of the Hemedex probe illustrating thermal diffusion technology. B, An annotated view of the Hemedex monitor shows 30
minutes of intraoperative cerebral blood flow (CBF) measurements made in the middle cerebral artery (MCA) vascular territory during an EC/IC bypass.
Note the hyperemia after the release of the bypass clamp and the subsequent establishment of a new and higher CBF level. (Courtesy Frank Bowman, PhD.)

thermocouple and Carter and Atkinson developed a mathe- technique can be applied intraoperatively and at the bedside
matical model to calculate absolute rCBF values.30,31 In 1973 to prevent, detect, or treat abnormalities in cerebral perfusion
TDF was introduced and validation studies showed that rCBF and so alter clinical outcome. Superficial (cortical) rCBF
values obtained by TDF are in good agreement with rCBF values between 40 to 70 mL/100 g/min are normal and values
values obtained in xenon-enhanced computed tomography less than 20 mL/100 g/min or greater than 70 mL/100 g/min
(Xe-CT) studies and the hydrogen clearance method.32,33 represent respectively ischemia and hyperemia.35 The TDF
Currently two technical systems are commercially available microprobe measures subcortical white matter perfusion, and
that use the temperature difference measured between a a mean TDF value of 18 to 25 mL/100 g/min is considered
thermistor and temperature sensor to calculate CBF. A small normal.36
Silastic probe(s) (Flowtronics, Inc., Phoenix, AZ) with two In critically ill brain-injured patients, rCBF monitoring with
gold disks can be placed subdurally on the cortex to measure Silastic TDF sensors can help predict outcome. Specifically,
superficial rCBF. During placement large vessels and patho- patients with a significant increase in rCBF from baseline
logic tissue need to be avoided to prevent artifacts. A confi- values, have a good outcome, whereas patients with initial very
dence factor can be calculated by turning off the heater to low values and those without an increase in flow from baseline
determine proper contact with the cerebral cortex. A confi- have a poor outcome.35 In this study, however, reliable data
dence factor of 0.8 or higher is considered to represent reliable could only be obtained in 37 of 56 patients because the routine
data. When the correct position is confirmed the dura can be application of this technique is not always easy. rCBF can
closed. Recently, an intraparenchymal thermal diffusion complement parameters such as CCP, ICP and PbtO2, and so
microprobe (Hemedex Inc., Cambridge, MA) has been intro- facilitate the interpretation of these parameters. In closed head
duced and validated that eliminates artifacts caused by loss of injury, monitoring of CPP and treatment of low CPP is con-
surface contact and obtains rCBF measurements from the sidered necessary to prevent cerebral ischemia. However, with
same vascular territory as other intraparenchymal monitoring a disrupted autoregulation, a CPP greater than 70 mm Hg may
devices (i.e., ICP, PbtO2).33 This novel TDF microprobe consists still be associated with ischemic events, and therefore restora-
of a thermistor in the tip of the probe and a temperature tion and maintenance of adequate perfusion could be per-
sensor located a few millimeters proximal that provides sensi- formed under guidance of continuous rCBF monitoring.37
tive and continuous absolute rCBF values even in the case of Likewise use of a Hemedex probe can help evaluate cerebral
minor flow changes (Fig. 31.2, A). It is inserted through a bolt autoregulation and CO2 vasoreactivity by allowing for calcula-
fixed in the skull with the tip approximately 25 mm below the tion of local cerebral vascular resistance (CPP/rCBF) in
dura in normal brain tissue, which can be verified with a CT response to a blood pressure or hyperventilation challenge.38
scan. Automatic recalibration occurs approximately every 30 Alternatively continuous monitoring of TDF-CBF, CPP, and
minutes; this results in a loss of data for 2 to 5 minutes. In brain oxygen allows calculation of flow- and oxygen-related
addition, significant baseline shifts of CBF that occur after autoregulation indices.39 TDF provides absolute values (unlike
each recalibration independent of hemodynamic stability are LDF) and therefore has the ability to distinguish between
a concern.34 Both systems have a built-in safety mechanism elevations of ICP associated with ischemia or hyperemia. A
that prevents CBF measurements when temperature is greater study of SAH and traumatic brain injury patients found a
than 39o C. This safety mechanism thus interrupts monitoring correlation between CBF measured by the TDF microprobe
of the cerebral perfusion during periods of high fever that are and PbtO2, indicating that tissue oxygen levels were determined
frequent in patients with severe closed head injury or sub- in part by regional CBF. This same study showed that loss of
arachnoid hemorrhage. data due to recalibrations or periods of fever is a concern. The
TDF provides in real time the dynamic changes in actual TDF microprobe allowed CBF measurement 64% of the time
cerebral perfusion, that is, quantitative data. Therefore this when PbtO2 monitoring was possible.34
Section V—Cerebral Blood Flow 317

In SAH, TDF can be used to help detect vasospasm and Orthogonal Polarizing
monitor for delayed cerebral ischemia.36,40 Vajkoczy et al.
found a threshold of 15 mL/100 g/min to diagnose symptom-
Spectral Imaging
atic vasospasm had a sensitivity of 90% and specificity of Disturbances of microvascular function seem to play a role
75%.36 In addition, the effectiveness of treatment can be deter- in the development of cerebral ischemia. Various cerebral
mined with continuous rCBF monitoring. Therefore TDF pathologies such as SAH or traumatic brain injury may impair
represents a useful and effective technique to help detect microcirculatory function by dysregulation of the vascular
delayed cerebral ischemia in SAH. Whether the use of this tone or intravascular microthrombosis.47,48 Furthermore, the
technique in the NCCU can lead to improved clinical outcome development of cerebral ischemia itself due to a mismatch in
will need further study. supply and demand may alter microcirculatory hemodynam-
TDF can be used intraoperatively to study hemodynamic ics and intensify ischemia. These assumptions are based on
changes that occur in the surrounding normal brain tissue direct visualization of the cerebral microcirculation in animals
during aneurysm clipping, bypass surgery (Fig. 31.2, B), or with a cranial window technique or conventional intravital
resections of AVM’s and brain tumors.41-44 Acute changes in microscopy and on indirect monitoring techniques in
cortical CBF with vascular manipulations during aneurysm humans.49 Therefore the ability to observe the morphology
clipping have been demonstrated with TDF. Furthermore, and function of the human cerebral microcirculation may aid
increased perinidal cortical CBF values were found with in the understanding of microvascular disturbances contrib-
TDF and LDF after AVM resection. Detection of hyperemic uting to ischemia.
values may indicate pending normal perfusion pressure The authors introduced a novel optical technique in the late
breakthrough, and therapeutic interventions (i.e., barbiturate 1990s, OPS imaging, that allows for the direct visualization of
coma) can be instituted promptly to avoid further brain tissue the microcirculation and qualitative evaluation of changes in
damage. A decrease in peritumoral CBF was found before flow, vessel diameter, capillary density, and vasoreactivity.50-52
tumor removal, which recovered after completion of the A thorough evaluation of the diagnostic value and limitations
resection, indicating that compression of the peritumoral of OPS imaging is useful to define its role in clinical research.
microvasculature was responsible for a decrease in blood flow. A first step in this direction was the validation of this optical
During epilepsy surgery, it was demonstrated that normaliza- technique. Several animal studies demonstrated that OPS
tion of peri-ictal bihemispheric cortical CBF occurs in tem- imaging produced similar or better images of the micro­
poral lobe epilepsy from a reduction of cortical CBF circulation compared to conventional intravital microscopy,
interictally.45,46 which allows quantification of microvascular changes.53-55 It
Limitations associated with TDF include “nonmonitroing” was demonstrated that this also is true for the functional
during recalibration and fever, sensor displacement or loss of assessment of the human cerebral microcirculation. Moreover,
brain surface contact, and that only a small area of brain tissue with this technique changes in CO2 reactivity and autoregula-
is monitored (20 to 30 mm3). Therefore global ischemia or tion of microvascular flow could be assessed in patients
focal ischemia in other unmeasured flow regions will be undergoing aneurysm surgery and in a focal contusion model
missed. This limitation may be overcome partially by the use (Fig. 31.3).51,56 This renders OPS imaging particularly useful
of multiple sensors on a strip or placement of multiple intra- to assess changes in microvascular flow during physiologic
parenchymal microprobes. Complications associated with challenges in which perfusion is changed.
TDF microprobes are very rare and similar to other implanted To further define the clinical value of OPS imaging it is
intraparenchymal probes (i.e., an approximate 1% risk of sig- necessary to investigate whether this imaging technique can
nificant bleeding or infection). provide new insights on underlying microcirculatory changes

*
*
A
A V

A 100 µm B

Fig. 31.3  Images of the cerebral microcirculation before (A) and during (B) hyperventilation. Besides a large amount of subarachnoid blood (*),
arterioles (A) and venules (V) can be distinguished from each other. After hyperventilation a decrease in arteriolar diameter is observed with a bead
string–like constriction pattern (B). Note that the caliber of the venules is unaffected.
318 Section V—Cerebral Blood Flow

that may play a role in the pathophysiology of cerebral isch- microcirculatory hemodynamic parameters as well as changes
emia. In addition, the question needs to be answered whether in mechanisms that regulate flow. These qualities may hold
OPS imaging data can be used to help improve outcome in promise to evaluate underlying microcirculatory mechanisms
patients in whom microcirculatory disturbances contribute to responsible for ischemia.
ischemia. Uhl et al. compared the cerebral microcirculation of
patients with incidental aneurysms (n = 3) to the cerebral
microcirculation of 10 SAH patients, before and after clip- References
ping.57 They reported three major findings in patients with 1. Astrup J, Siesjo BK, Symon L. Thresholds in cerebral ischemia—the ischemic
SAH: the detection of microvasospasm in SAH patients in penumbra. Stroke 1981;12:723–5.
2. Jones TH, Morawetz RB, Crowell RM, et al. Thresholds of focal cerebral
combination with a reduced functional capillary density, ischemia in awake monkeys. J Neurosurg 1981;54:773–82.
which increased significantly during the operation. A relation- 3. Czosnyka M, Brady K, Reinhard M, et al. Monitoring of cerebrovascular
ship between intraoperative microvasospasm and clinical autoregulation: facts, myths, and missing links. Neurocrit Care
outcome could not be observed and further evaluation is 2009;10(3):373–86. Epub 2009 Jan 6.
needed. It has been observed that the human cerebral micro- 4. Bonner RF, Nossal R. Continuous measurement of tissue blood flow by
laser-Dopplerspectroscopy. Am J Physiol 1977;2:H441–8.
circulation is compromised in the course of neurovascular 5. Rajan V, Varghese B, van Leeuwen TG, et al. Review of methodological
diseases, which may contribute to cerebral ischemia.51,52,58 developments in laser Doppler flowmetry. Lasers Med Sci (online first)
Clinical studies, however, also failed to demonstrate a correla- 2008;60.
tion between the severity of microcirculatory impairment and 6. Bonner RF, Nossal R. Principles of laser-Doppler flowmetry. In: Shepherd
AP, Oberg PA, editors. Laser Doppler flowmetry. Boston: Kluwer Academic;
clinical outcome. The small number of studied patients may 1990. p.17–45.
explain this. In patients with septic shock, sublingual micro- 7. Bolognese P, Miller JI, Heger IM, et al. Laser Doppler flowmetry in
circulatory function is severely altered, and these alterations neurosurgery. J Neurosurg Anesthesiol 1993;5:151–8.
are more pronounced in nonsurvivors. In addition, improve- 8. Neufeld GR, Galante SR, Whang JM, et al. Skin blood flow from gas
ment in microcirculatory function after resuscitation can be a transport: helium xenon and laser Doppler compared. Microvasc Res 1988;
35:143–52.
better outcome predictor than changes in cardiac index, blood 9. Olsson P. A comparison between the 133Xe washout and laser Doppler
pressure, or lactate.59,60 Thus OPS imaging may enable the techniques for estimation of nasal mucosal blood flow in humans. Acta
correlation of microvascular hemodynamic changes in the Otolaryngol 1986;102:106–12.
human brain with metabolic and clinical parameters. 10. Eyre JA, Essex TJH, Flecknell PA, et al. A comparison of measurements of
cerebral blood flow in the rabbit using laser Doppler spectroscopy and
Several limitations of OPS imaging need to be addressed. radionuclide labelled microspheres. Clin Phys Physiol Meas 1988;9:65–74.
First, a craniotomy is required to obtain information on the 11. Lindsberg PJ, O’Neill JT, Paakari IA, et al. Laser-Doppler flowmetry in
cerebral microcirculation. This limits its use only to the monitoring regulation of rapid microcirculatory changes in spinal cord. Am
intraoperative period. To overcome this limitation, extracra- J Physiol 1992;263:H285–92.
nial compartments that may represent the cerebral micro­ 12. Fakuda O, Endo S, Kuwayama N, et al. The characteristics of laser-Doppler
flowmetry for the measurement of regional cerebral blood flow.
circulation could be studied. In this regard, the retina or Neurosurgery 1995;36:358–64.
sublingual microcirculation may serve as potential targets. 13. Rundquist I, Smith QR, Michel ME, et al. Sciatic nerve blood flow measured
Second, the superficial penetration depth of 500 µm and the by laser Doppler flowmetry and [14C]iodoantipyrine. Am J Physiol 1985;
image quality limits the visualization of the framework of the 248:H311–17.
14. Nitzan M, Fairs S, Roberts V. Simultaneous measurement of skin blood flow
capillary network and density, which represents the meta- by the transient thermal-clearance method and laser Doppler flowmetry.
bolic activity of the surrounding brain tissue. To resolve these Med Biol Eng Comput 1988;26:407–10.
drawbacks SDF imaging (MicroVision Medical Inc., Amster- 15. Kirkpatrick PJ, Smielweski P, Piechnik S, et al. Early effects of mannitol in
dam, the Netherlands) has been introduced into the field of patients with head injuries assessed using bedside multimodality monitoring.
neurosurgery.61 In this modality, a light guide imaging the Neurosurgery 1996;39:714–20.
16. Kirkpatrick PJ, Smielweski P, Czosnyka M, et al. Continuous monitoring of
microcirculation is surrounded by light-emitting diodes of a cortical perfusion by laser Doppler flowmetry in ventilated patients with
wavelength (530 nm) absorbed by the hemoglobin of eryth- head injury. J Neurol Neurosurg Psychiatry 1994;57:1382–8.
rocytes so that they can be clearly observed as flowing cells. 17. Le Bihan D, Turner R. The capillary network: a link between IVIM and
The concentrically placed light-emitting diodes at the tip of classical perfusion. Magn Reson Med 1992;27:171–8.
18. Zweifel C, Czosnyka M, Lavinio A, et al. A comparison study of cerebral
the probe directly penetrate deep into the tissue that illumi- autoregulation assessed with transcranial Doppler and cortical laser Doppler
nates the microcirculation. This way of observing the cere- flowmetry. Neurol Res 2010;32(4):425–8.
bral microcirculation provides clear images of the capillaries 19. Schubert GA, Seiz M, Hegewald AA, et al. Hypoperfusion in the acute phase
and leukocytes without blurring. The improved resolution of subarachnoid hemorrhage. Acta Neurochir Suppl 2011;110(Pt 1):35–8.
allows for the determination of leukocyte-endothelium adhe- 20. Lam JMK, Hsiang JNK, Poon WS. Monitoring of autoregulation using laser
Doppler flowmetry in patients with head injury. J Neurosurg 1997;86:438–45.
sion and quantification of vascular leakage. Third, the image 21. Smielewski P, Czosnyka M, Kirkpatrick P, et al. Evaluation of the transient
processing software used in previous studies prohibited ac- hyperemic response test in head injured patients. J Neurosurg 1997;86:
curate erythrocyte velocity measurement above 1 mm/sec. 773–8.
Thus essential information on arteriolar velocity could not 22. Johnson WD, Bolognese P, Miller JI, et al. Continuous postoperative lCBF
monitoring in aneurysmal SAH patients using a combined ICP-laser
be calculated. Therefore a semiquantitative method, the mi- Doppler fiberoptic probe. J Neurosurg Anesthesiol 1996;8:199–207.
crocirculatory flow index (MFI), was introduced to describe 23. Rosenblum BR, Bonner RF, Oldfield EH. Intraoperative measurement of
the changes in flow velocities and flow patterns.58 However, cortical blood flow adjacent to cerebral AVM using laser Doppler
the software has been improved to eliminate most of these velocimetry. J Neurosurg 1987;66:396–9.
problems and allow velocity measurements of ve­nules and 24. Leahy MJ, de Mul FFM, Nilsson GE, et al. Principles and practice of the
laser-Doppler perfusion technique. Technol Health Care 1999;7:143–62.
arterioles as well as an MFI for the area of interest.62 25. Kawamata T, Kawashima A, Yamaguchi K, et al. Usefulness of intraoperative
In conclusion, OPS imaging and its successor laser Doppler flowmetry and thermography to predict a risk of postoperative
SDF can provide continuous information on important hyperperfusion after superficial temporal artery-middle cerebral artery
Section V—Cerebral Blood Flow 319

bypass for moyamoya disease. Neurosurg Rev 2011;34(3):355–62; discussion 39. Hecht N, Fiss I, Wolf S, et al. Modified flow- and oxygen-related
362. Epub 2011 Jun 4. autoregulation indices for continuous monitoring of cerebral autoregulation.
26. Klein KU, Stadie A, Fukui K, et al. Measurement of cortical microcirculation J Neurosci Methods 2011;15:201(2):399–403. Epub 2011 Aug 17.
during intracranial aneurysm surgery by combined laser-Doppler flowmetry 40. Hänggi D. Participants in the International Multi-Disciplinary Consensus
and photospectrometry. Neurosurgery 2011;69(2):391–8. Conference on the Critical Care Management of Subarachnoid Hemorrhage.
27. Miles B, Davis S, Crandall C, et al. Laser-Doppler examination of the blood Monitoring and detection of vasospasm. II: EEG and invasive monitoring.
supply in pericranial flaps. J Oral Maxillofac Surg 2010;68(8):1740–5. Epub Neurocrit Care 2011;15(2):318–23
2010 May 20. 41. Nagao S, Veta K, Mino S, et al. Monitoring of cortical blood flow during
28. Hill SA, Pigott MI, Saunders MI, et al. Microregional blood flow in murine excision of arteriovenous malformation by thermal diffusion method. Surg
and human tumours assessed using laser-Doppler microprobes. Br J Cancer Neurol 1989;32:137–43.
1996;S260–3. 42. Ohmoto T, Nagao S, Mino S, et al. Monitoring of cortical blood flow during
29. Warden K, Jakobsson A, Nilsson GE. Laser-Doppler perfusion imaging by temporary arterial occlusion in aneurysm surgery by the thermal diffusion
dynamic light scattering. IEEE Trans Biomed Eng 1993;40:309–16. method. Neurosurgery 1991;28:49–54.
30. Gibbs FA. A thermoelectric blood flow recorder in the form of a needle. In: 43. Thome C, Vajkoczy P, Horn P, et al. Continuous monitoring of regional
Proceedings of the Society for Experimental Biology and Medicine, San blood flow during temporary arterial occlusion in aneurysm surgery.
Francisco, 1933;141–6. J Neurosurg 2001;95:402–11.
31. Carter LP, Atkinson JR. Cortical blood flow in controlled hypotension as 44. Carter LP, Weinand ME, Oommen KJ. Cerebral blood flow monitoring in
measured by thermal diffusion. J Neurol Neurosurg Psychiatry 1973;36: intensive care by thermal diffusion. Acta Neurochir Suppl 1993;59:43–6.
906–13. 45. Gonzalez G, Rivero S, Ahern GL, et al. Normalization of periictal
32. Gaines C, Carter LP, Crowell RM. Comparison of local cerebral blood flow bihemispheric cerebral perfusion in temporal lobe epilepsy. Pathophysiology
determined by thermal and hydrogen clearance. Stroke 1983;14:66–9. 2004;11:31–4.
33. Vajkoczy P, Roth H, Horn P, et al. Continuous monitoring of regional 46. Weinand ME, Carter LP, Oommen KJ, et al. Response of human epileptic
cerebral blood flow: experimental and clinical validation of a novel thermal temporal lobe cortical blood flow to hyperventilation. Epilepsy Res 1995;21:
diffusion microprobe. J Neurosurg 2000;93:265–74. 221–6.
34. Jaeger M, Soehle M, Schuhmann MU, et al. Correlation of continuously 47. Cook DA. Mechanisms of cerebral vasospasm in subarachnoid haemorrhage.
monitored regional cerebral blood flow and brain tissue oxygen. Acta Pharmacol Ther 1995;66:259–84.
Neurochir 2005;147:51–6. 48. Stein SC, Graham DI, Chen XH, et al. Association between intravascular
35. Sioutos PJ, Orozco JA, Carter LP, et al. Continuous regional cerebral cortical microthrombosis and cerebral ischemia in traumatic brain injury.
blood flow monitoring in head injured patients. Neurosurgery 1995;36: Neurosurgery 2004; 54:687–91.
943–50. 49. Caro CG, Pedley TJ, Schroter RC, et al. The mechanics of the circulation.
36. Vajkoczy P, Horn P, Thome C, et al. Regional cerebral blood flow monitoring Oxford, UK. Medical Publications, 2006.
in the diagnosis of delayed ischemia following aneurismal subarachnoid 50. Groner W, Winkelman JW, Harris AG, et al. Orthogonal polarization spectral
hemorrhage. J Neurosurg 2003;98:1227–34. imaging: a new method for study of the microcirculation. Nat Med
37. Miller JI, Chou MW, Capocelli A, et al. Continuous intracranial 1999;5:1209–12.
multimodality monitoring comparing local cerebral blood flow, cerebral
perfusion pressure, and microvascular resistance. Acta Neurochir Suppl 1998; A complete list of references for this chapter can be found online at
71:82–4. www.expertconsult.com.
38. Rosenthal G, Sanchez-Mejia RO, Phan N, Hemphill 3rd JC, et al.
Incorporating a parenchymal thermal diffusion cerebral blood flow probe in
bedside assessment of cerebral autoregulation and vasoreactivity in patients
with severe traumatic brain injury. J Neurosurg 2011;114(1):62–70. Epub
2010 Aug 13.
Section V—Cerebral Blood Flow 319.e1

References 27. Miles B, Davis S, Crandall C, et al. Laser-Doppler examination of the blood
supply in pericranial flaps. J Oral Maxillofac Surg 2010;68(8):1740–5. Epub
1. Astrup J, Siesjo BK, Symon L. Thresholds in cerebral ischemia—the ischemic 2010 May 20.
penumbra. Stroke 1981;12:723–5. 28. Hill SA, Pigott MI, Saunders MI, et al. Microregional blood flow in murine
2. Jones TH, Morawetz RB, Crowell RM, et al. Thresholds of focal cerebral and human tumours assessed using laser-Doppler microprobes. Br J Cancer
ischemia in awake monkeys. J Neurosurg 1981;54:773–82. 1996;S260–3.
3. Czosnyka M, Brady K, Reinhard M, et al. Monitoring of cerebrovascular 29. Warden K, Jakobsson A, Nilsson GE. Laser-Doppler perfusion imaging by
autoregulation: facts, myths, and missing links. Neurocrit Care 2009;10(3): dynamic light scattering. IEEE Trans Biomed Eng 1993;40:309–16.
373–86. Epub 2009 Jan 6. 30. Gibbs FA. A thermoelectric blood flow recorder in the form of a needle. In:
4. Bonner RF, Nossal R. Continuous measurement of tissue blood flow by Proceedings of the Society for Experimental Biology and Medicine, San
laser-Doppler spectroscopy. Am J Physiol 1977;2:H441–8. Francisco, 1933;141–6.
5. Rajan V, Varghese B, van Leeuwen TG, et al. Review of methodological 31. Carter LP, Atkinson JR. Cortical blood flow in controlled hypotension as
developments in laser Doppler flowmetry. Lasers Med Sci (online first) measured by thermal diffusion. J Neurol Neurosurg Psychiatry 1973;36:
2008:60. 906–13.
6. Bonner RF, Nossal R. Principles of laser-Doppler flowmetry. In: Shepherd 32. Gaines C, Carter LP, Crowell RM. Comparison of local cerebral blood flow
AP, Oberg PA, editors. Laser Doppler flowmetry. Boston: Kluwer Academic; determined by thermal and hydrogen clearance. Stroke 1983;14:66–9.
1990. p.17–45. 33. Vajkoczy P, Roth H, Horn P, et al. Continuous monitoring of regional
7. Bolognese P, Miller JI, Heger IM, et al. Laser Doppler flowmetry in cerebral blood flow: experimental and clinical validation of a novel thermal
neurosurgery. J Neurosurg Anesthesiol 1993;5:151–8. diffusion microprobe. J Neurosurg 2000;93:265–74.
8. Neufeld GR, Galante SR, Whang JM, et al. Skin blood flow from gas 34. Jaeger M, Soehle M, Schuhmann MU, et al. Correlation of continuously
transport: helium xenon and laser Doppler compared. Microvasc Res 1988; monitored regional cerebral blood flow and brain tissue oxygen. Acta
35:143–52. Neurochir 2005;147:51–6.
9. Olsson P. A comparison between the 133Xe washout and laser Doppler 35. Sioutos PJ, Orozco JA, Carter LP, et al. Continuous regional cerebral cortical
techniques for estimation of nasal mucosal blood flow in humans. Acta blood flow monitoring in head injured patients. Neurosurgery 1995;36:
Otolaryngol 1986;102:106–12. 943–50.
10. Eyre JA, Essex TJH, Flecknell PA, et al. A comparison of measurements of 36. Vajkoczy P, Horn P, Thome C, et al. Regional cerebral blood flow monitoring
cerebral blood flow in the rabbit using laser Doppler spectroscopy and in the diagnosis of delayed ischemia following aneurismal subarachnoid
radionuclide labelled microspheres. Clin Phys Physiol Meas 1988;9:65–74. hemorrhage. J Neurosurg 2003;98:1227–34.
11. Lindsberg PJ, O’Neill JT, Paakari IA, et al. Laser-Doppler flowmetry in 37. Miller JI, Chou MW, Capocelli A, et al. Continuous intracranial
monitoring regulation of rapid microcirculatory changes in spinal cord. Am multimodality monitoring comparing local cerebral blood flow, cerebral
J Physiol 1992;263:H285–92. perfusion pressure, and microvascular resistance. Acta Neurochir Suppl 1998;
12. Fakuda O, Endo S, Kuwayama N, et al. The characteristics of laser-Doppler 71:82–4.
flowmetry for the measurement of regional cerebral blood flow. 38. Rosenthal G, Sanchez-Mejia RO, Phan N, Hemphill 3rd JC, et al.
Neurosurgery 1995;36:358–64. Incorporating a parenchymal thermal diffusion cerebral blood flow probe in
13. Rundquist I, Smith QR, Michel ME, et al. Sciatic nerve blood flow measured bedside assessment of cerebral autoregulation and vasoreactivity in patients
by laser Doppler flowmetry and [14C]iodoantipyrine. Am J Physiol 1985; with severe traumatic brain injury. J Neurosurg 2011;114(1):62–70. Epub
248:H311–17. 2010 Aug 13.
14. Nitzan M, Fairs S, Roberts V. Simultaneous measurement of skin blood flow 39. Hecht N, Fiss I, Wolf S, et al. Modified flow- and oxygen-related
by the transient thermal-clearance method and laser Doppler flowmetry. autoregulation indices for continuous monitoring of cerebral autoregulation.
Med Biol Eng Comput 1988;26:407–10. J Neurosci Methods 2011;15:201(2):399–403. Epub 2011 Aug 17.
15. Kirkpatrick PJ, Smielweski P, Piechnik S, et al. Early effects of mannitol in 40. Hänggi D. Participants in the International Multi-Disciplinary Consensus
patients with head injuries assessed using bedside multimodality monitoring. Conference on the Critical Care Management of Subarachnoid Hemorrhage.
Neurosurgery 1996;39:714–20. Monitoring and detection of vasospasm. II: EEG and invasive monitoring.
16. Kirkpatrick PJ, Smielweski P, Czosnyka M, et al. Continuous monitoring of Neurocrit Care 2011;15(2):318–23
cortical perfusion by laser Doppler flowmetry in ventilated patients with 41. Nagao S, Veta K, Mino S, et al. Monitoring of cortical blood flow during
head injury. J Neurol Neurosurg Psychiatry 1994;57:1382–8. excision of arteriovenous malformation by thermal diffusion method. Surg
17. Le Bihan D, Turner R. The capillary network: a link between IVIM and Neurol 1989;32:137–43.
classical perfusion. Magn Reson Med 1992;27:171–8. 42. Ohmoto T, Nagao S, Mino S, et al. Monitoring of cortical blood flow during
18. Zweifel C, Czosnyka M, Lavinio A, et al. A comparison study of cerebral temporary arterial occlusion in aneurysm surgery by the thermal diffusion
autoregulation assessed with transcranial Doppler and cortical laser Doppler method. Neurosurgery 1991;28:49–54.
flowmetry. Neurol Res 2010;32(4):425–8. 43. Thome C, Vajkoczy P, Horn P, et al. Continuous monitoring of regional
19. Schubert GA, Seiz M, Hegewald AA, et al. Hypoperfusion in the acute phase blood flow during temporary arterial occlusion in aneurysm surgery.
of subarachnoid hemorrhage. Acta Neurochir Suppl 2011;110(Pt 1):35–8. J Neurosurg 2001;95:402–11.
20. Lam JMK, Hsiang JNK, Poon WS. Monitoring of autoregulation using laser 44. Carter LP, Weinand ME, Oommen KJ. Cerebral blood flow monitoring
Doppler flowmetry in patients with head injury. J Neurosurg 1997;86: in intensive care by thermal diffusion. Acta Neurochir Suppl 1993;59:
438–45. 43–6.
21. 21. Smielewski P, Czosnyka M, Kirkpatrick P, et al. Evaluation of the 45. Gonzalez G, Rivero S, Ahern GL, et al. Normalization of periictal
transient hyperemic response test in head injured patients. J Neurosurg bihemispheric cerebral perfusion in temporal lobe epilepsy. Pathophysiology
1997;86:773–8. 2004;11:31–4.
22. Johnson WD, Bolognese P, Miller JI, et al. Continuous postoperative lCBF 46. Weinand ME, Carter LP, Oommen KJ, et al. Response of human epileptic
monitoring in aneurysmal SAH patients using a combined ICP-laser temporal lobe cortical blood flow to hyperventilation. Epilepsy Res 1995;21:
Doppler fiberoptic probe. J Neurosurg Anesthesiol 1996;8:199–207. 221–6.
23. Rosenblum BR, Bonner RF, Oldfield EH. Intraoperative measurement of 47. Cook DA. Mechanisms of cerebral vasospasm in subarachnoid haemorrhage.
cortical blood flow adjacent to cerebral AVM using laser Doppler Pharmacol Ther 1995;66:259–84.
velocimetry. J Neurosurg 1987;66:396–9. 48. Stein SC, Graham DI, Chen XH, et al. Association between intravascular
24. Leahy MJ, de Mul FFM, Nilsson GE, et al. Principles and practice of the microthrombosis and cerebral ischemia in traumatic brain injury.
laser-Doppler perfusion technique. Technol Health Care 1999;7:143–62. Neurosurgery 2004; 54:687–91.
25. Kawamata T, Kawashima A, Yamaguchi K, et al. Usefulness of intraoperative 49. Caro CG, Pedley TJ, Schroter RC, et al. The mechanics of the circulation.
laser Doppler flowmetry and thermography to predict a risk of postoperative Oxford, UK. Medical Publications 2006.
hyperperfusion after superficial temporal artery-middle cerebral artery 50. Groner W, Winkelman JW, Harris AG, et al. Orthogonal polarization spectral
bypass for moyamoya disease. Neurosurg Rev 2011;34(3):355–62; discussion imaging: a new method for study of the microcirculation. Nat Med
362. Epub 2011 Jun 4. 1999;5:1209–12.
26. Klein KU, Stadie A, Fukui K, et al. Measurement of cortical microcirculation 51. Pennings FA, Bouma GJ, Ince C. Direct observation of the human
during intracranial aneurysm surgery by combined laser-Doppler flowmetry microcirculation during aneurysm surgery reveals increased arteriolar
and photospectrometry. Neurosurgery 2011;69(2):391–8. contractility. Stroke 2004;35(6):1284–88.
319.e2 Section V—Cerebral Blood Flow

52. Pennings FA, Albrecht KW, Muizelaar JP, et al. Abnormal responses of the 58. Pennings FA, Ince C, Bouma GJ. Continuous real-time visualization of the
human cerebral microcirculation to papaverin during aneurysm surgery. human cerebral microcirculation during arteriovenous malformation
Stroke 2009;40(1):317–20. Epub 2008 Oct 9. surgery using orthogonal polarization spectral imaging. Neurosurgery 2006;
53. Harris AG, Sinitsina I, Messmer K. The Cytoscan model E-II, a new 59:167–71.
reflectance microscope for intravital microscopy: comparison with the 59. De BD, Creteur J, Preiser JC, et al. Microvascular blood flow is altered in
standard fluorescence method. J Vasc Res 2000;37:469–76. patients with sepsis. Am J Respir Crit Care Med 2002;166:98–104.
54. Langer S, Harris AG, Biberthaler P, et al. Orthogonal polarization spectral 60. Sakr Y, Dubois MJ, De BD, et al. Persistent microcirculatory alterations are
imaging as a tool for the assessment of hepatic microcirculation: a validation associated with organ failure and death in patients with septic shock. Crit
study. Transplantation 2001;71:1249–56. Care Med 2004;32:1825–31.
55. Mathura KR, Vollebregt KC, Boer K, et al. Comparison of OPS imaging and 61. Ince C. The microcirculation is the motor of sepsis. Crit Care Suppl 2005;4:
conventional capillary microscopy to study the human microcirculation. S13–9.
J Appl Physiol 2001;91:74–8. 62. Dobbe JG, Streekstra GJ, Atasever B, et al. Measurement of functional
56. Kroppenstedt SN, Thomale UW, Griebenow M, et al. Effects of early and late microcirculatory geometry and velocity distributions using automated image
intravenous norepinephrine infusion on cerebral perfusion, microcirculation, analysis. Med Biol Eng Comput 2008;46:659–70.
brain-tissue oxygenation, and edema formation in brain-injured rats. Crit
Care Med 2003;31:2211–21.
57. Uhl E, Lehmberg J, Steiger HJ, et al. Intraoperative detection of early
microvasospasm in patients with subarachnoid hemorrhage by using
orthogonal polarization spectral imaging. Neurosurgery 2003;52:1307–15.
V
Chapter
32  

Jugular Bulb Oximetry


Amit Prakash and Basil F. Matta

Introduction Physiologic Basis of


Jugular bulb venous monitoring provides information about
SjvO2 Interpretation
global cerebral hemodynamics and metabolism and has Jugular bulb oximetry has evolved from a combination of old
become an important tool in the clinical management of and new technologies. The first measurements taken from the
neurologically injured patients. Other techniques to measure jugular bulb were used to calculate cerebral blood flow (CBF)
cerebral oxygenation include near infrared spectroscopy using the Fick principle.1
(NIRS), which noninvasively detects changes in blood hemo- Cerebral metabolic rate of oxygen (CMRO2) is calculated
globin concentrations associated with neural activity and from the CBF and the difference between the arteriovenous
multiparameter intraparenchymal probes that are used to difference in oxygen (AVDO2) using the equation:
measure brain tissue and cerebrospinal fluid PO2, PaCO2, and
pH. This chapter focuses on jugular bulb venous oximetry. CMRO2 = CBF × (arterial oxygen content minus jugular
venous bulb oxygen content)
The oxygen content can be determined using co-oximetry,
Anatomy or calculated using the equations:
The venous drainage from the brain forms six major cranial
sinuses (superior and inferior sagittal, occipital, right and left Arterial oxygen content =
transverse, and the straight sinus). These venous channels, [(Hgb × 1.34 × SaO2 ) + (PaO2 × 0.0031)]
located between the dura mater and the periosteum lining the Jugular venous oxygen content =
cranium, are devoid of valves or muscle in their walls. The [(Hgb × 1.34 × SjvO2 ) + (PvO2 × 0.0031)]
final common pathway for the majority of blood returning
from the brain is through the right and left sigmoid sinus, Where Hgb = hemoglobin in g/dL, SaO2 = arterial satura-
which curves down the posterior fossa, over the mastoid tion, SjvO2 = jugular venous oxygen saturation, PaO2 = oxygen
portion of the temporal bone, and then runs forward to form partial pressure in arterial blood in mm Hg, and PvO2 =
the right and left internal jugular veins in the posterior part oxygen partial pressure in venous blood in mm Hg.
of the jugular foramen. Because the hemoglobin concentration is the same in arte-
The internal jugular vein (IJV) begins as a small dilation rial and venous blood, and the amount of dissolved oxygen is
called the jugular bulb at the jugular foramen, and runs down minimal, CMRO2 can be estimated from CBF and the differ-
the neck in the carotid sheath to form the brachiocephalic vein ence in the arteriojugular bulb oxygen saturation.
by joining the subclavian vein behind the medial end of the
CMRO2 = CBF × (SaO2 − SjvO2 )
clavicle. Within the carotid sheath, the IJV is lateral to the
vagus nerve and the carotid artery. Therefore the arteriovenous oxygen content (or saturation)
Posterior to the vein lie the transverse processes of the cervi- difference can be used to relate for changes in metabolism
cal vertebra, the cervical plexus, phrenic nerve, vertebral vein, with alterations in CBF. The equation may be written as
the first part of the subclavian artery, and the thoracic duct follows:
toward the left. Anterolateral to the vein are superficial cervical
AVDO2 = Hgb × 1.34 (SaO2 − SvO2 ) + 0.003 (PaO2 − PvO2 )
fascia, platysma, transverse cutaneous nerve, deep cervical
fascia, and the sternocleidomastoid muscle. The lower end of In the absence of infarction or necrosis, the cerebral AVDO2
IJV drains into the right atrium. and CBF appear to be inversely related.2 Hypoperfusion and
The jugular bulb is the final common pathway for venous ischemia result in an increased AVDO2, whereas hyperemia
blood that drains from the cerebral hemispheres, cerebel- will lead to a reduced AVDO2. The same principle can be
lum, and brainstem. Therefore the jugular venous oxygen applied to other metabolites such as glucose and lactate.
saturation (SjvO2) reflects the balance between supply and
consumption of oxygen by the brain. Contribution of the
extracranial circulation is estimated to range from zero to
Insertion Technique
6.6%. This is exacerbated if the blood is contaminated with The use of ultrasound (US) in accordance with National Insti-
blood from the facial vein that joins the IJV a few centime- tute for Clinical Excellence (NICE) guidelines3 for anatomic
ters below the jugular bulb. localization and IJV puncture has facilitated procedure
320 © Copyright 2013 Elsevier Inc. All rights reserved.
Section V—Cerebral Blood Flow 321

performance. Besides, SjvO2 monitoring is commonly used to


manage head injured patients. Because severe head injury fre-
quently has been associated with cervical injury, US-guided
insertion reduces the need for head manipulation, thus making
it a safer technique. Overall, SjvO2 catheters have a complica-
tion rate similar to that seen with the placement of antegrade
jugular venous lines.4 Jugular bulb catheters are best avoided
IJV
in those with coagulation abnormalities, sepsis, or local neck Carotid A
trauma. SCM

Optimal Side for Monitoring


Jugular Bulb Oxygenation
SjvO2 monitoring accurately reflects global and hemispheric
cerebral oxygenation, only when the dominant jugular bulb is
cannulated.5 However, selecting the optimal side for a jugular
bulb catheter remains controversial. Cortical blood from the
sagittal sinus flows into the right lateral sinus and subcortical
blood from the straight sinus usually goes to the left lateral
sinus. Overall, flow is greater to the right jugular venous bulb.6,7
Up to 15% difference between right and left SjvO2 has been
demonstrated in patients with head trauma.8 If an intracranial Fig. 32.1  The authors’ technique for insertion of jugular bulb
pressure (ICP) monitor is in situ, it is possible to determine catheters. The junction between the sternal and clavicular heads of the
sternocleidomastoid muscle (SCM) is localized and the skin is punctured
the dominant side of venous drainage by sequential temporary
under ultrasound guidance with a 16-gauge, 5.25-inch Angiocath
manual occlusion of the right and then left internal jugular catheter (Becton, Dickinson, Franklin Lakes, NJ) mounted with a 5-mL
vein and observing the resultant ICP change.9 Compressing syringe. During gentle aspiration the needle is passed in a cranial
the dominant jugular vein will lead to venous obstruction and direction for 1 to 2 cm, at an angle of 15 to 20 degrees in the sagittal
a greater increase in ICP, thus providing a clue for the best plane. Once the vein is entered, the catheter is advanced over the needle
side for SjvO2 monitoring. If the ICP increase is similar during until a slight elastic resistance is felt, or when the tip of the catheter is
right and left IJV compression, the side with the greatest degree estimated to be just behind the mastoid process. Carotid A, Carotid
of pathology as determined by computed tomography (CT) artery; IJV, internal jugular vein.
scan is used to insert a jugular bulb catheter. In the case of
diffuse injury, the right side is used.10 In the authors’ unit, right-
sided jugular venous oximetry is used almost exclusively.
continuously aspirating, gently to maintain intraluminal posi-
tion of the needle. The catheter then is advanced over the
Site of Cannulation needle until a slight elastic resistance is felt, or when the tip of
US-guided visualization insertion reduces the need for scru- the catheter is estimated to be just behind the mastoid process.
pulous landmark identification. However, there are two main If difficulty in localizing the vein is encountered, the patient
sites for retrograde cannulation of the internal jugular vein: is tilted 15 degrees head down until the vein is located, after
(1) lateral to the carotid artery at the level of the inferior which the head is elevated. A guidewire (Seldinger technique)
border of the thyroid cartilage6 and (2) at the junction of the may be used in a similar manner.
sternal and clavicular heads of the sternocleidomastoid muscle If needed, central venous access also can be obtained as a
(Fig. 32.1). The cannulation can be approached either by the part of the same procedure. Once the wire for the SjvO2 cath-
Seldinger method or cannula over needle method. One eter is in situ, a guidewire is passed in the antegrade direction
method is to approach the vessel at the junction of the sternal through a separate puncture site, using the Seldinger tech-
and cla­vicular heads of the sternocleidomastoid muscle under nique under US guidance. After placement of the guidewires
US guidance. for both lines, the retrograde catheter for SjvO2 followed by
Once the site is selected, it is prepared with antiseptic solu- the central venous catheter is threaded over the guidewire.
tion, draped with sterile towels, and infiltrated with local anes- This sequence prevents the accidental shearing of the central
thetic. The patient is then placed in the horizontal position, venous catheter when the SjvO2 catheter is inserted. The cath-
with the neck in the neutral position or slightly turned to the eter is connected to a slow continuous infusion of 0.9% saline,
contralateral side. The IJV is then localized with an US probe, to prevent blockage.
between the junction of the sternal and clavicular heads of the For continuous monitoring, a fiber-optic catheter is passed
sternocleidomastoid. Under US guidance, the skin is punc- through a 16-gauge cannula placed in a similar manner as
tured with a 16-gauge 5.25-inch Angiocath catheter (Becton, described previously. A pressurized flush system is used to
Dickinson, Franklin Lakes, NJ) mounted on a 5-mL syringe. maintain catheter patency and reduce the incidence of wall
With continuous gentle aspiration the IJV is punctured at artefacts.11 A lateral cervical spine x-ray or an anteroposterior
almost 90 degrees under direct vision, which is confirmed by chest x-ray that includes a view of the neck is obtained to
the flashback in the syringe. Once the vein is entered, the confirm the catheter position.12 The tip of the catheter should
operating hand with the needle and the syringe is dropped lie at the level of and just medial to the mastoid bone above
down to about 15 to 20 degrees in a cranial direction, while the lower border of C1.
322 Section V—Cerebral Blood Flow

80
Measurement Obtained
from Jugular Bulb Catheters
SjvO2 can be measured by sampling blood intermittently or
continuously by using a fiber-optic oximetric catheter. Serial
or intermittent sampling is cheaper and allows calculation of

SjvO2 %
AVDO2, glucose, and lactate. The interpretation of jugular 40
oxygen saturation is based on several assumptions. However,
there is a good correlation between fiber-optic and catheter
SjvO2 and with direct brain oxygen measurements.13 Some of
the early problems encountered with the use of fiber-optic
oximetric catheters seem to have been reduced by the develop-
ment of new “stiffer” catheters less prone to kinking and 0
curling back, and by careful positioning and calibration. This PaCO2 PaCO2
has been confirmed by Gunn et al.11 in intracranial surgery 25 mm Hg 30 mm Hg
and by Souter and Andrews,14 and Coplin et al.15 in an inten-
sive care setting. Nonetheless, suspected jugular bulb desatu- 2 mL/min
ration should be verified by co-oximetry before therapeutic 4 mL/min
interventions. 10 mL/min
Two fiber-optic catheters are available for jugular bulb Fig. 32.2  The speed of blood withdrawal from jugular bulb
oximetry: (1) the Oximetrix (Abbott Laboratories, North catheters affects accuracy of reading. Jugular venous oxygen
Chicago) and 2) the Edslab II (Baxter Healthcare Corpora- saturation (SjvO2) values are higher with faster rates of blood withdrawal
tion, Irvine, CA).16 Both are 4F-gauge double-lumen cathe- because of contamination with extracranial blood. Optimal rate appears
ters. After insertion the distal lumen should be aspirated for to be 2 mL/min.
in vivo calibration. The other lumen contains two optical
fibers, one to transmit and the other to receive light trans-
mitted to and then returned from the jugular bulb. The
Oximetrix catheter has three light-emitting diodes; the is an increased risk of local and systemic infection with long-
Edslab II has two. These send red and near infrared light at term placement.
1-ms intervals to blood passing through the jugular bulb,
where it is absorbed, reflected, and refracted. The reflected
light from hemoglobin is detected by a photoelectric sensor
Interpretation of the Measurements
and averaged for the previous 5 seconds to calculate hemo- Normal SjvO2 values range between 55% and 75%. In simple
globin oxygen saturation that is then updated every second. terms, SjvO2 levels less than 55% suggest cerebral oxygen
If the light source at the catheter tip abuts the vessel wall, a demand exceeding supply, for example, as a consequence of
low light intensity or signal quality alarm is displayed. The hypoperfusion (ischemia), whereas levels greater than 75%
Edslab II system requires the insertion of the patient’s hemo- indicate relative hyperemia (Tables 32.1 and 32.2). Microdi-
globin concentration for in vivo calibration; this may affect alysis studies in traumatic brain injury (TBI) patients demon-
its accuracy when the hemoglobin concentration is unstable strate that jugular oxygen desaturation is associated with
such as with rapid massive blood loss. However, presently elevated glutamate.19 Evidence for cellular dysfunction
there is no clinical data that compare the two catheters in detected by microdialysis of metabolites such as glutamate,
such circumstances. glycerol, lactate, and pyruvate consistently occurs when SjvO2
Continuous fiber-optic Oximetrix catheters can be cali- is less than 45%.20 However, SjvO2 is a global hemispheric
brated before (in vitro), or after insertion (in vivo). In vivo measurement and hence regional ischemia cannot be detected.
calibration is more accurate and subject to less drift.9,14,17 Drift For example, Chieregato et al.21 suggest that jugular bulb
can be further reduced by recalibration at 12-hour intervals. oximetry, without ICP monitoring, is suboptimal to manage
The sampling port of the catheter should be continuously a patient with subarachnoid haemorrhage (SAH) and raised
flushed with heparinized saline (1 IU/mL) at a rate of 2 to intracranial pressure.22 Therefore although a normal SjvO2
4 mL/hr to maintain its patency. Contamination of jugular does not guarantee absence of regional ischemia, a low SjvO2
bulb blood with extracranial blood depends on the speed of indicates either an increase in oxygen extraction or a reduction
blood withdrawal from the catheter, with up to 25% higher in oxygen delivery, which may be an early warning of cerebral
SjvO2 values with faster rates of blood withdrawal (>2 mL/ ischemia.23 The volume of ischemic brain to cause a change in
min)18 (Fig. 32.2). SjvO2 is large, however. For example, Coles et al.24 examined
15 TBI patients with positron emission tomography (PET)
within 24 hours of head injury to map CBF, CMRO2, and
Complications on Insertion oxygen extraction fraction (OEF). The SjvO2 correlated with
The commonest complications are carotid artery puncture the ischemic brain volume (measured by PET OEF) but SjvO2
and hematoma formation, which occur in about 1% to 4% of values of 50% only occurred when ischemic brain volume was
insertions and are usually self-limiting. Pneumothorax, venous 170 plus or minus 63 mL (mean ± 95% coagulation index
air embolism, and venous thrombosis are infrequent compli- [CI]), that is, 13 plus or minus 5% of the brain. Finally in some
cations, as is damage to adjacent structures such as the carotid patients, changes in SjvO2 associated with intracranial hyper-
artery, vagus and phrenic nerves, and the thoracic duct. There tension may only be observed after herniation.
Section V—Cerebral Blood Flow 323

outcome in TBI.19,30,31 For example, Robertson et al.19 observed


Table 32.1  A Summary of the Main that mortality was 21% in TBI patients with no desaturation,
Causes and Treatment of Low and High 37% with a single episode, and 69% after multiple episodes.
Jugular Saturations Although reduced SjvO2 is associated with a wide AVDO2,
High SjvO2 Abnormal Hyperemia outcome also may be poor with a reduced AVDO2 because
autoregulation cerebral metabolism is reduced once there is cerebral infarc-
Increased oxygen Polycythemia tion.31 Together these data suggest a potential benefit to detect
supply and treat episodes of jugular destauration.27
Deceased oxygen Hypothermia
consumption Sedative drugs
Many episodes of SjvO2 desaturation appear to be associ-
Anesthetic drugs ated with suboptimal cerebral perfusion pressure (CPP), with
Cerebral Infarction cerebral arterial vasospasm, or from hyperventilation.
Extracerebral blood However, abnormalities in jugular oxygenation may persist
Low SjvO2 Abnormal even when ICP and CPP are corrected. For example, Le Roux
autoregulation et al.32 studied 32 patients with severe TBI who had elevated
Deceased oxygen Hypoxia ICP and were treated with surgery or mannitol. Patients whose
supply Hypotension
Intracranial hypertension
AVDO2 failed to improve were more likely to develop a cere-
Hyperventilation bral infarct or have a poor outcome than when AVDO2
Low cardiac output improved (i.e., decrease >1 vol%) after correction of the ele-
Anemia vated ICP or decreased CPP. In addition, when very early
Increased oxygen Hyperthermia jugular bulb oximetry data from patients with severe TBI are
consumption Seizures
Sepsis
analyzed, there is evidence that altered and inadequate CPP is
common in the first few hours after injury. These findings
SjvO2, Jugular venous oxygen saturation. have important implications for the emergency management
of such patients.33
The role of lactate after TBI has been evaluated using ret-
rograde jugular catheters. Some studies suggest that an increase
Table 32.2  Arteriovenous Difference in CSF lactate is a useful marker of cerebral ischemia, and in
in Oxygen particular knowledge of cerebral lactate (or cerebral anaerobic
metabolism) can help define infarction.34 However Poca
AVDO2 = CMRO2 / CBF (calculation − 1.39 (SaO2 − 
SjvO2) × Hgb/100 et al.35 found that arteriojugular venous differences of lactate
Normal = 5-7.5 vol% (5.1 - 8.3 vol%) do not reliably reflect increased cerebral lactate production
Narrow AVDO2 − <5 vol% = hyperemia (CBF > CMRO2) when compared with microdialysis measurements of lactate.
Wide AVDO2 − >7.5 vol% = low flow (CBF < CMRO2) This difference, however, may be explained, one method
AVDO2, Arteriovenous difference in oxygen; CBF, cerebral blood flow; being a global measure and the other a local measure. Other
CMRO2, cerebral metabolic rate of oxygen; Hgb, hemoglobin; markers such as endothelin-1,36-38 cytokines,39 arteriovenous
SaO2, arterial saturation; SjvO2, jugular venous oxygen saturation. difference of PaCO2,21 markers of coagulation and inflamma-
tion thrombin-antithrombin complex, fibrin D-dimer, and
prothrombin fragment,40,41 markers of oxidative damage viz
Care must be taken when using a jugular bulb catheter malonyaldehyde,42 and type B natriuretic peptide43 have all
because incorrect placement can result in extracerebral con- been assessed using jugular bulb catheters (i.e., not with a
tamination or artifacts associated with catheter movement fiber-optic oximeter), in research studies. In addition after
may occur. A slow continuous infusion of 0.9% saline and injections of ice-cold indocyanine green, global CBF values
flush system to maintain patency can help reduce the inci- can be assessed using the mean transit time principle by exam-
dence of wall artifacts.11 Anemia also may narrow the AVDO2 ining temperature and dye concentration changes in the tho-
and affect measurements. Similarly the rate of sedative infu- racic aorta and the jugular bulb.44
sion (e.g., propofol) can affect readings.25 In addition, many
retrograde jugular catheters require constant recalibration
and unless done, accuracy suffers.13,26 Long-term use is associ- Intraoperative Management of Patients
ated with a small risk of infection and thrombosis. Undergoing Intracranial Aneurysm Surgery
Several studies have described the use of jugular bulb cathe-
ters during neurosurgical procedures and in particular intra-
Clinical Applications cranial aneurysm surgery. For example, Matta et al. observed
a 50% incidence of jugular venous desaturation in patients
Traumatic Brain Injury and during neurosurgical procedures.4 However, the incidence of
Subarachnoid Hemorrhage severe desaturation (defined as SjvO2 <45%) was 17%. The
Despite intensive care management, with invasive hemody- episodes of desaturation could not have been predicted by
namic and ICP monitoring, episodes of cerebral venous desat- the PaCO2 level alone—without SjvO2 monitoring many of
uration are common in comatose patients with TBI and the episodes would have gone unnoticed. Moss et al. studied
SAH.27-29 Patients with episodes of cerebral venous oxygen the effects of changing mean arterial pressure (MAP) on
desaturation (SjvO2 <50% for more than 15 minutes) have a SjvO2 and lactate oxygen index (LOI) in 26 patients during
higher mortality than those without. There is a well-described cerebral aneurysm surgery.45 An SjvO2 value less than 54%
association between episodes of SjvO2 desaturation and poor was considered to indicate cerebral hypoperfusion. They were
324 Section V—Cerebral Blood Flow

able to identify a critical MAP of between 80 and 110 mm Hg prevented by achieving normal values of mixed SvO2, partial
in 9 patients. Patients with an LOI less than 0.08 at any time pressure of PaCO2 and central venous pressure (CVP); this
during the procedure had a worse outcome immediately after provides insight to potential neurocritical care unit (NCCU)
surgery. Although an increase in MAP resulted in a similar management strategies. Others have also found a benefit to
increase in SjvO2 in 19 of the patients, this was not always PaCO2 control. For example, during one lung ventilation for
accompanied by an improved LOI. Together these studies lung surgery, SjvO2 has been used to examine effects on cere-
highlight the potential benefit of jugular bulb cannulation to bral oxygen. In these patients Iwata et al.53 observed that
assess cerebral hypoperfusion during the intra- and postop- hypercapnia, not hypertension, significantly improved cere-
erative management of SAH patients. In addition, significant bral oxygen balance.
desaturation has been observed with intraoperative aneurysm
rupture.46
Cardiac Arrest
The ability to diagnose evidence for brain injury after cardiac
Carotid Endarterectomy arrest is important in some patients. Takasu et al. continuously
Detection of cerebral ischemia throughout cross clamping monitored systemic and jugular venous oxygenation during
during carotid endarterectomy (CEA) is critical. Several the 24 hours after resuscitation in cardiac arrest in eight
studies have demonstrated the potential value of SjvO2 mon- patients.54 The three patients that survived had significantly
itoring to detect cerebral ischemia during carotid endarter- lower SjvO2 (67%) than nonsurvivors (80%), whereas mixed
ectomy. For example, Crossman et al.46 studied 37 patients venous saturations were higher in the survivors than in the
who underwent awake CEA to correlate neurologic deficits nonsurvivors (74% and 64%, respectively). The authors
with SjvO2 desaturation. A 25% change in SjvO2 was associ- suggest that damaged neurons are unable to utilize oxygen
ated with the development of clinically apparent cerebral adequately, and this resulted in the high SjvO2 value. However,
ischemia. Moritz et al. examined 48 patients during awake normal SjvO2 values vary between 55% and 85%, and so it is
CEA. Twelve patients developed intraoperative neurologic unlikely that this information will be of benefit in the immedi-
deficit, which correlated with SjvO2 desaturations. They ate management of these patients.
found that arteriojugular venous lactate content difference Hypothermia is indicated for cardiac arrest. Although
(≥0.16 mmol/L) and lactate oxygen index was associated hypothermia improves outcome in patients after cardiac
with intraoperative ischemia.47 arrest, there remains a question whether the cardiodepressive
effects of hypothermia may lead to secondary brain damage.
Studies by Bisschops et al.55 using a jugular bulb catheter
Diagnosis of Brain Death among other monitors suggest that jugular bulb oxygenation
Although the diagnosis of brain death remains a clinical one, gradually increases, whereas cerebral oxygen extraction
confirmatory or complementary tests are useful. Extreme remains normal, and transcranial Doppler (TCD) measure-
hyperoxia of the jugular venous blood has been suggested as ments of flow decrease, suggesting decreased cerebral meta-
a possible sign of brain death.48 Díaz-Regañón et al.49 assessed bolic activity.
the usefulness of SjvO2 monitoring as a complementary test
to diagnose brain death. They studied 118 patients and mea-
sured the ratio of central venous oxygen (ScO2) to SjvO2 at the As a Guide to Clinical Management
time of diagnosis of brain death. A ratio of less than 1 was in the Intensive Care Unit
found in 114 of the 118 patients. In a survey of the intensive care management of severe TBI in
the United Kingdom, published in 1996, 12% of the units
questioned used a jugular bulb catheter for routine monitor-
Cardiac and Thoracic Surgery ing, in more than 50% of their TBI patients.56 Since 2001 the
Jugular bulb oxygen desaturation is associated with poor use of jugular bulb oximetry has progressed from being a
outcome in patients undergoing cardiac surgery.50 For research tool to a common clinical tool used in many hospi-
example, Croughwell et al. used jugular bulb catheters to cal- tals. In some patients it is used alone to assess cerebral oxy-
culate AVDO2 in 255 patients during cardiopulmonary genation but in many it is used with other measures such as
bypass.51 Desaturation (SjvO2 <50%) occurred in 23% of the near infrared spectroscopy (NIRS) or direct brain oxygen.
patients and was associated with worse postoperative cogni- These various tools can supplement and complement
tive function. The desaturations observed were the result of each other.
inadequate CBF in proportion to oxygen consumption, as Prolonged hyperventilation can aggravate neurologic
indicated by a decreased CBF to AVDO2 ratio. Other potential outcome. However, acute hyperventilation (HV) may be life-
causes include microembolic events and disturbances in cere- saving in many patients provided the potential deleterious
bral autoregulation. The episodes of desaturation were consequences of HV are identified.57 Hypocapnia from HV
observed during both normothermic and hypothermic car- induces cerebral vasoconstriction, and the resultant decrease
diopulmonary bypass. In a follow-up study, the same group in cerebral blood volume and ICP may improve CPP. However,
highlighted the importance of continuously monitoring cere- excessive cerebral vasoconstriction causes cerebral ischemia.
bral oxygenation during cardiac surgery as neither mixed In addition, hyperventilation shifts the oxygen-hemoglobin
venous oximetry nor systemic pump venous saturation was dissociation curve to the left, and reduces the amount of
able to detect the adequacy of cerebral perfusion in these oxygen released from haemoglobin to brain tissue. Therefore
patients.52 During off-pump coronary artery bypass grafting HV should not be used to lower ICP without some measure
surgery jugular desaturations are common but can be of cerebral oxygenation (CBF).58,59 Hyperoxia can be used as
Section V—Cerebral Blood Flow 325

8 to changes in perfusion pressure. For example, Lewis et al.


Focal pathology demonstrated that a critically low SjvO2 is a late indicator of
None failed autoregulation, and when used with TCD may be useful
6 to determine the level of “optimal” CPP in the early resuscita-
∆PbO2 (mm Hg)

tion of TBI62 (Fig. 32.4).

3
Conclusion
By providing an indication of inadequate cerebral oxygen-
0 ation and by inference inadequate CBF, cerebral oximetry
may allow appropriate therapy to be given before neurologic
damage becomes permanent. There is no gold standard to
–3 measure cerebral oxygenation. Jugular venous bulb oximetry
22–26 26–30 30–34 34–38 is a global hemispheric measure with low sensitivity for
PaCO2 (mm Hg) regional ischemia and so it is best used with other monitors
of brain function. Regional brain oxygenation can be exam-
Fig. 32.3  Graph that illustrates the effect of hyperventilation on brain ined by using NIRS or direct brain oxygen monitors (see
tissue oxygenation in areas of focal pathology and with no focal
Chapters 33 and 35), and these techniques can be used in a
pathology. (Reproduced with permission from Gupta AK, Hutchinson PJ, Al-Rawi
P, et al. Measuring brain tissue oxygenation compared with jugular venous oxygen
complementary fashion in select patients. At present no one
saturation for monitoring cerebral oxygenation after traumatic brain injury. Anesth
method is sufficiently reliable to facilitate the clinical man-
Analg 1999;88:549–53.) agement of neurologically injured patient with absolute cer-
tainty when used alone. However, further research and
improvement in technology, and the ability to incorporate
SjvO2 <50% these methods into multimodal monitoring can permit
early interventions with a possible consequence of improved
No outcome.

SpO2 <50% Yes Correct hypoxemia References


1. Ketty SS, Schmidt CF. The nitrous oxide method for the quantitative
No determination of cerebral blood flow in man: theory, procedure and normal
values. J Clin Invest 1948;27:476–83.
2. Obrist W, Langfitt T, Jaggi J, et al. Cerebral blood flow and metabolism in
PaCO2 <4.0 kPa Yes Raise PaCO2 comatose patients with acute head injury: relationship to intracranial
hypertension. J Neurosurg 1984;61:241–53.
No 3. National Institute for Clinical Excellence. NICE technology appraisal
guidance No 49: guidance on the use of ultrasound locating devices for
placing central venous catheters. 2002. London NICE Available from
Hemoglobin <10g/dL Yes Blood transfusion www.nice.org.uk/pdf/ultrasound_49_GUIDANCE.pdf
4. Matta BF, Lam AM, Mayberg TS, et al. A critique of the intraoperative use of
jugular venous bulb catheters during neurosurgical procedures. Anesth Analg
No 1994;79:745–50.
5. Lam JM, Chan MS, Poon WS. Cerebral venous oxygen saturation
monitoring: is dominant jugular bulb cannulation good enough? Br J
Mean arterial Support blood Neurosurg 1996;10(4):357–64.
Yes
pressure flow pressure 6. Gibbs EL, Gibbs FA. The cross sectional areas of the vessels that form the
torcular and the manner in which blood is distributed to the right and to
No the left lateral sinus. Anat Rec 1934;54:419.
7. Gibbs EL, Lennox WG, Giggs FA. Bilateral internal jugular blood,
comparison of A-V differences, oxygen-dextrose ratios and respiratory
Mannitol, furosemide, quotients. Am J Psychiatry 1945;102:184–90.
Intracranial propofol, thiopentone,
hypertension Yes 8. Stocchetti N, Paparella A, Brindelli F, et al. Cerebral venous oxygen
hypothermia saturation studied with bilateral samples in the internal jugular veins.
Neurosurgery 1994;34:38–44.
Fig. 32.4  Suggested algorithm to treat jugular venous desaturation. 9. Andrews PJD, Dearden NM, Miller JD. Jugular bulb cannulation: description
g/dL, Grams per deciliter (conventional unit); kPa, kilo pascals (1 kpa = of a cannulation technique and a validation of a new continuous monitor.
7.5 mm Hg); PaCO2 , arterial carbon dioxide tension; SjvO2 , jugular Br J Anaesth 1991;67:553–8.
venous oxygen saturation; SpO2 , arterial saturation. 10. Matta BF, Lam AM, Mayberg TS, et al. A critique of the intraoperative use of
jugular venous bulb catheters during neurosurgical procedures. Anesth Analg
1994;79:745–50.
11. Gunn HC, Matta BF, Lam AM, et al. Accuracy of continuous jugular bulb
a temporary measure to improve cerebral oxygen delivery venous oximetry during intracranial surgery. J Neurosurg Anesthesiol
during hyperventilation.60,61 SjvO2 monitoring in such sce- 1995;7:174–7.
narios, allows fine-tuning of ventilation so that optimal brain 12. Bankier AA, Fleischmann D, Windiscch A, et al. Position of jugular oxygen
oxygenation is achieved (Fig. 32.3). saturation catheter in patients with head trauma: assessment by use of plain
films. Am J Radiol 1995;164:437–41.
Brain injury often is associated with impaired cerebral pres- 13. Gopinath SP, Valadka AB, Uzura M, et al. Comparison of jugular venous
sure autoregulation. The critical perfusion threshold can be oxygen saturation and brain tissue PO2 as monitors of cerebral ischemia
established by close examination of SjvO2 changes in response after head injury. Crit Care Med 27:2337–45, 1999
326 Section V—Cerebral Blood Flow

14. Souter MJ, Andrews PJD. Validation of the Edslab dual lumen oximetry 34. Robertson CS, Grossman RG, Goodman C, et al. The predictive value of
catheter for continuous monitoring of jugular bulb oxygen saturation after cerebral anerobic metabolism with infarction after head injury. J Neurosurg
severe head injury. Br J Anaesth 1996;76:744–6. 1987;67:361–8.
15. Coplin WM, O’Keefe GE, Grady MS, et al. Accuracy of continuous jugular 35. Poca MA, Sahuquillo J, Vilalta A, et al. Lack of utility of arteriojugular
bulb oximetry in the intensive care unit. Neurosurgery 1998;42(3):533–9. venous differences of lactate as a reliable indicator of increased brain
16. De Deyne C, Decruyenaere J, Colardyn F. How to interpret jugular bulb anaerobic metabolism in traumatic brain injury. J Neurosurg 2007;106(4):
oximetry? In: Vincent JL, editor. Yearbook of intensive care and emergency 530–7.
medicine. Berlin: Springer-Verlag, 1996. p. 731–41. 36. Menon DK, Day D, Kuc RE, et al. Arteriojugular endothelin-1 gradients in
17. Lewis SB, Myburgh JA, Reilly PL. Detection of cerebral venous desaturation aneurysmal subarachnoid haemorrhage. Clin Sci (Lond) 2002;103(Suppl
by continuous jugular bulb oximetry following acute neurotrauma. Anaesth 48):399S–403S.
Intensive Care 1995;23:307–14. 37. Chatfield DA, Brahmbhatt DH, Sharp T, et al. Juguloarterial endothelin-1
18. Matta BF, Lam AM. The speed of blood withdrawal affects the accuracy of gradients after severe traumatic brain injury. Neurocrit Care 2011;14(1):
jugular venous bulb oxygen saturation measurements. Anesthesiology 1996; 55–60.
86:806–8. 38. Helmy A, Carpenter KL, Menon DK, et al. The cytokine response to human
19. Robertson CS, Gopinath SP, Goodman C, et al. SjvO2 monitoring in head traumatic brain injury: temporal profiles and evidence for cerebral
injured patients. J Neurotrauma 1995;12:891–6. parenchymal production. J Cereb Blood Flow Metab 2011;31(2):658–70.
20. Chan MT, Ng SC, Lam JM, et al. Re-defining the ischemic threshold for Epub 2010 Aug 18.
jugular venous oxygen saturation—a microdialysis study in patients with 39. Muroi C, Mink S, Seule M, et al. Monitoring of the inflammatory response
severe head injury. Acta Neurochir 2005 Suppl;95:63–6. after aneurysmal subarachnoid haemorrhage in the clinical setting: review of
21. Chieregato A, Marchi M, Fainardi E, et al. Cerebral arterio-venous PaCO2 literature and report of preliminary clinical experience. Acta Neurochir
difference, estimated respiratory quotient, and early posttraumatic outcome: Suppl 2011;110(Pt 1):191–6.
comparison with arterio-venous lactate and oxygen differences. J Neurosurg 40. Nekludov M, Antovic J, Bredbacka S, et al. Coagulation abnormalities
Anesthesiol 2007;19(4):222–8. associated with severe isolated traumatic brain injury: cerebral arterio-
22. Chieregato A, Targa L, Zatelli R. Limitations of jugular bulb oxyhemoglobin venous differences in coagulation and inflammatory markers. J Neurotrauma
saturation without intracranial pressure monitoring in subarachnoid 2007;24(1):174–80.
hemorrhage. J Neurosurg Anesthesiol 1996;8:21–5. 41. Paolin A, Nardin L, Gaetani P, et al. Oxidative damage after severe head
23. Gupta AK, Hutchinson PJ, Al-Rawi P, et al. Measuring brain tissue injury and its relationship to neurological outcome. Neurosurgery 2002;
oxygenation compared with jugular venous oxygen saturation for 51(4):949–54; discussion 954–5.
monitoring cerebral oxygenation after traumatic brain injury. Anesth Analg 42. Powner DJ, Hergenroeder GW, Awili M, et al. Hyponatremia and
1999;88:549–53. comparison of NT-pro-BNP concentrations in blood samples from jugular
24. Coles JP, Fryer TD, Smielewski P, et al. Incidence and mechanisms of cerebral bulb and arterial sites after traumatic brain injury in adults: a pilot study.
ischemia in early clinical head injury. J Cereb Blood Flow Metab 2004;24(2): Neurocrit Care 2007;7(2):119–23.
202–11. 43. Jägersberg M, Schaller C, Boström J, et al. Simultaneous bedside
25. Haranishi Y, Kuroda Y, Matayoshi Y, et al. Infusion rate of propofol and assessment of global cerebral blood flow and effective cerebral perfusion
jugular venous oxygen saturation. J Anesth 2007;21(4):516–8. Epub 2007 pressure in patients with intracranial hypertension. Neurocrit Care 2010;
Nov 1. 12(2):225–33.
26. Goetting MG, Preston G. Jugular bulb catheterization: experience with 123 44. Moss E, Dearden NM, Berridge JC. Effects of changes in mean arterial
patients. Crit Care Med 1990;18(11):1220–3. pressure on SjvO2 during cerebral aneurysm surgery. Br J Anaesth 1995;75:
27. Schneider GH, Helden AV, Lanksch WR, et al. Continuous monitoring of 527–30.
jugular bulb oxygen saturation in comatose patients—therapeutic 45. Rozet I, Newell DW, Lam AM. Intraoperative jugular bulb desaturation
implications. Acta Neurochir (Wein) 1995;134:71–5. during acute aneurysmal rupture. Can J Anaesth 2006;53(1):97–100.
28. Sheinberg GM, Kanter MJ, Robertson CS, et al. Continuous monitoring of 46. Crossman J, Banister K, Bythell V, et al. Predicting clinical ischaemia during
jugular venous oxygen saturation in head-injured patients. J Neurosurg awake carotid endarterectomy: use of the SjvO2 probe as a guide to selective
1992;76:212–7. shunting. Physiol Meas 2003;24:347–35.
29. Gopinath SP, Rogertson CS, Constant CF, et al. Jugular venous desaturation 47. Moritz S, Kasprzak P, Woertgen C, et al. The accuracy of jugular bulb
and outcome after head injury. J Neurol Neurosurg Psychiatry 1994;57: venous monitoring in detecting cerebral ischemia in awake patients
717–23. undergoing carotid endarterectomy. J Neurosurg Anesthesiol 2008;20:
30. Robertson CS, Contant CF, Narayan RK, Grossman RG. Cerebral blood flow, 8–14.
AVDO2, and neurologic outcome in head-injured patients. J Neurotrauma 48. Ogawa M, Sugimoto T, Katsurada T. Hyperoxia of internal jugular venous
1992;9(Suppl 1):S349–58. blood in brain death. J Neurosurg 1973;39:442–7.
31. Jaggi JL, Obrist WD, Gennarelli TA, et al. Relationship of early cerebral 49. Díaz-Regañón G, Miñambres E, Holanda M, et al. Usefulness of venous
blood flow and metabolism to outcome in acute head injury. J Neurosurg oxygen saturation in the jugular bulb for the diagnosis of brain death: report
1990;72:176–82. of 118 patients. Intensive Care Med 2002;28:1724–8.
32. Le Roux P, Lam AM, Newell DW, et al. Cerebral arteriovenous difference of 50. Miura N, Yoshitani K, Kawaguchi M, et al. Jugular bulb desaturation during
oxygen: a predictor of cerebral infarction and outcome in severe head injury. off-pump coronary artery bypass surgery. J Anesth 2009;23(4):477–82. Epub
J Neurosurg 1997;87:1–8. 2009 Nov 18.
33. De Deyne C, Decruyenaere J, Calle P, et al. Analysis of very early jugular bulb
oximetry data after severe head injury: implications for the emergency A complete list of references for this chapter can be found online at
management. Eur J Emerg Med 1996;3(2):69–72. www.expertconsult.com.
Section V—Cerebral Blood Flow 326.e1

References 25. Haranishi Y, Kuroda Y, Matayoshi Y, et al. Infusion rate of propofol and
jugular venous oxygen saturation. J Anesth 2007;21(4):516–8. Epub 2007
1. Ketty SS, Schmidt CF. The nitrous oxide method for the quantitative Nov 1.
determination of cerebral blood flow in man: theory, procedure and normal 26. Goetting MG, Preston G. Jugular bulb catheterization: experience with 123
values. J Clin Invest 1948;27:476–83. patients. Crit Care Med 1990;18(11):1220–3.
2. Obrist W, Langfitt T, Jaggi J, et al. Cerebral blood flow and metabolism in 27. Schneider GH, Helden AV, Lanksch WR, et al. Continuous monitoring of
comatose patients with acute head injury: relationship to intracranial jugular bulb oxygen saturation in comatose patients—therapeutic
hypertension. J Neurosurg 1984;61:241–53. implications. Acta Neurochir (Wein) 1995;134:71–5.
3. National Institute for Clinical Excellence. NICE technology appraisal 28. Sheinberg GM, Kanter MJ, Robertson CS, et al. Continuous monitoring of
guidance No 49: guidance on the use of ultrasound locating devices for jugular venous oxygen saturation in head-injured patients. J Neurosurg
placing central venous catheters. 2002. London NICE Available from 1992;76:212–7.
www.nice.org.uk/pdf/ultrasound_49_GUIDANCE.pdf 29. Gopinath SP, Rogertson CS, Constant CF, et al. Jugular venous desaturation
4. Matta BF, Lam AM, Mayberg TS, et al. A critique of the intraoperative use of and outcome after head injury. J Neurol Neurosurg Psychiatry 1994;57:
jugular venous bulb catheters during neurosurgical procedures. Anesth Analg 717–23.
1994;79:745–50. 30. Robertson CS, Contant CF, Narayan RK, Grossman RG. Cerebral blood flow,
5. Lam JM, Chan MS, Poon WS. Cerebral venous oxygen saturation AVDO2, and neurologic outcome in head-injured patients. J Neurotrauma
monitoring: is dominant jugular bulb cannulation good enough? Br J 1992;9(Suppl 1):S349–58.
Neurosurg 1996;10(4):357–64. 31. Jaggi JL, Obrist WD, Gennarelli TA, et al. Relationship of early cerebral
6. Gibbs EL, Gibbs FA. The cross sectional areas of the vessels that form the blood flow and metabolism to outcome in acute head injury. J Neurosurg
torcular and the manner in which blood is distributed to the right and to 1990;72:176–82.
the left lateral sinus. Anat Rec 1934;54:419. 32. Le Roux P, Lam AM, Newell DW, et al. Cerebral arteriovenous difference of
7. Gibbs EL, Lennox WG, Giggs FA. Bilateral internal jugular blood, oxygen: a predictor of cerebral infarction and outcome in severe head injury.
comparison of A-V differences, oxygen-dextrose ratios and respiratory J Neurosurg 1997;87:1–8.
quotients. Am J Psychiatry 1945;102:184–90. 33. De Deyne C, Decruyenaere J, Calle P, et al. Analysis of very early jugular bulb
8. Stocchetti N, Paparella A, Brindelli F, et al. Cerebral venous oxygen oximetry data after severe head injury: implications for the emergency
saturation studied with bilateral samples in the internal jugular veins. management. Eur J Emerg Med 1996;3(2):69–72.
Neurosurgery 1994;34:38–44. 34. Robertson CS, Grossman RG, Goodman C, et al. The predictive value of
9. Andrews PJD, Dearden NM, Miller JD. Jugular bulb cannulation: description cerebral anerobic metabolism with infarction after head injury. J Neurosurg
of a cannulation technique and a validation of a new continuous monitor. 1987;67:361–8.
Br J Anaesth 1991;67:553–8. 35. Poca MA, Sahuquillo J, Vilalta A, et al. Lack of utility of arteriojugular
10. Matta BF, Lam AM, Mayberg TS, et al. A critique of the intraoperative use of venous differences of lactate as a reliable indicator of increased brain
jugular venous bulb catheters during neurosurgical procedures. Anesth Analg anaerobic metabolism in traumatic brain injury. J Neurosurg 2007;106(4):
1994;79:745–50. 530–7.
11. Gunn HC, Matta BF, Lam AM, et al. Accuracy of continuous jugular bulb 36. Menon DK, Day D, Kuc RE, et al. Arteriojugular endothelin-1 gradients in
venous oximetry during intracranial surgery. J Neurosurg Anesthesiol aneurysmal subarachnoid haemorrhage. Clin Sci (Lond) 2002;103(Suppl
1995;7:174–7. 48):399S–403S.
12. Bankier AA, Fleischmann D, Windiscch A, et al. Position of jugular oxygen 37. Chatfield DA, Brahmbhatt DH, Sharp T, et al. Juguloarterial endothelin-1
saturation catheter in patients with head trauma: assessment by use of plain gradients after severe traumatic brain injury. Neurocrit Care
films. Am J Radiol 1995;164:437–41. 2011;14(1):55–60.
13. Gopinath SP, Valadka AB, Uzura M, et al. Comparison of jugular venous 38. Helmy A, Carpenter KL, Menon DK, et al. The cytokine response to human
oxygen saturation and brain tissue PO2 as monitors of cerebral ischemia traumatic brain injury: temporal profiles and evidence for cerebral
after head injury. Crit Care Med 27:2337–45, 1999 parenchymal production. J Cereb Blood Flow Metab 2011;31(2):658–70.
14. Souter MJ, Andrews PJD. Validation of the Edslab dual lumen oximetry Epub 2010 Aug 18.
catheter for continuous monitoring of jugular bulb oxygen saturation after 39. Muroi C, Mink S, Seule M, et al. Monitoring of the inflammatory response
severe head injury. Br J Anaesth 1996;76:744–6. after aneurysmal subarachnoid haemorrhage in the clinical setting: review of
15. Coplin WM, O’Keefe GE, Grady MS, et al. Accuracy of continuous jugular literature and report of preliminary clinical experience. Acta Neurochir
bulb oximetry in the intensive care unit. Neurosurgery 1998;42(3):533–9. Suppl 2011;110(Pt 1):191–6.
16. De Deyne C, Decruyenaere J, Colardyn F. How to interpret jugular bulb 40. Nekludov M, Antovic J, Bredbacka S, et al. Coagulation abnormalities
oximetry? In: Vincent JL, editor. Yearbook of intensive care and emergency associated with severe isolated traumatic brain injury: cerebral arterio-
medicine. Berlin: Springer-Verlag; 1996. p. 731–41. venous differences in coagulation and inflammatory markers. J Neurotrauma
17. Lewis SB, Myburgh JA, Reilly PL. Detection of cerebral venous desaturation 2007;24(1):174–80.
by continuous jugular bulb oximetry following acute neurotrauma. Anaesth 41. Paolin A, Nardin L, Gaetani P, et al. Oxidative damage after severe head
Intensive Care 1995;23:307–14. injury and its relationship to neurological outcome. Neurosurgery 2002;
18. Matta BF, Lam AM. The speed of blood withdrawal affects the accuracy of 51(4):949–54; discussion 954–5.
jugular venous bulb oxygen saturation measurements. Anesthesiology 1996; 42. Powner DJ, Hergenroeder GW, Awili M, et al. Hyponatremia and
86:806–8. comparison of NT-pro-BNP concentrations in blood samples from jugular
19. Robertson CS, Gopinath SP, Goodman C, et al. SjvO2 monitoring in head bulb and arterial sites after traumatic brain injury in adults: a pilot study.
injured patients. J Neurotrauma 1995;12:891–6. Neurocrit Care 2007;7(2):119–23.
20. Chan MT, Ng SC, Lam JM, et al. Re-defining the ischemic threshold for 43. Jägersberg M, Schaller C, Boström J, et al. Simultaneous bedside assessment
jugular venous oxygen saturation—a microdialysis study in patients with of global cerebral blood flow and effective cerebral perfusion pressure in
severe head injury. Acta Neurochir 2005 Suppl;95:63–6. patients with intracranial hypertension. Neurocrit Care 2010;12(2):
21. Chieregato A, Marchi M, Fainardi E, et al. Cerebral arterio-venous PaCO2 225–33.
difference, estimated respiratory quotient, and early posttraumatic outcome: 44. Moss E, Dearden NM, Berridge JC. Effects of changes in mean arterial
comparison with arterio-venous lactate and oxygen differences. J Neurosurg pressure on SjvO2 during cerebral aneurysm surgery. Br J Anaesth
Anesthesiol 2007;19(4):222–8. 1995;75:527–30.
22. Chieregato A, Targa L, Zatelli R. Limitations of jugular bulb oxyhemoglobin 45. Rozet I, Newell DW, Lam AM. Intraoperative jugular bulb desaturation
saturation without intracranial pressure monitoring in subarachnoid during acute aneurysmal rupture. Can J Anaesth 2006;53(1):97–100.
hemorrhage. J Neurosurg Anesthesiol 1996;8:21–5. 46. Crossman J, Banister K, Bythell V, et al. Predicting clinical ischaemia during
23. Gupta AK, Hutchinson PJ, Al-Rawi P, et al. Measuring brain tissue awake carotid endarterectomy: use of the SjvO2 probe as a guide to selective
oxygenation compared with jugular venous oxygen saturation for shunting. Physiol. Meas 2003;24:347–35.
monitoring cerebral oxygenation after traumatic brain injury. Anesth Analg 47. Moritz S, Kasprzak P, Woertgen C, et al. The accuracy of jugular bulb venous
1999;88:549–53. monitoring in detecting cerebral ischemia in awake patients undergoing
24. Coles JP, Fryer TD, Smielewski P, et al. Incidence and mechanisms of cerebral carotid endarterectomy. J Neurosurg Anesthesiol 2008;20(1):8–14.
ischemia in early clinical head injury. J Cereb Blood Flow Metab 2004;24(2): 48. Ogawa M, Sugimoto T, Katsurada T. Hyperoxia of internal jugular venous
202–11. blood in brain death. J Neurosurg 1973;39:442–7.
326.e2 Section V—Cerebral Blood Flow

49. Díaz-Regañón G, Miñambres E, Holanda M, et al. Usefulness of venous 56. Matta B, Menon D. Severe head injury in the United Kingdom and Ireland: a
oxygen saturation in the jugular bulb for the diagnosis of brain death: report survey of practice and implications for management. Crit Care Med 1996;
of 118 patients. Intensive Care Med 2002;28:1724–8. 24:1743–8.
50. Miura N, Yoshitani K, Kawaguchi M, et al. Jugular bulb desaturation during 57. Muizelaar JP, Marmarou A, Ward JD, et al. Adverse effects of prolonged
off-pump coronary artery bypass surgery. J Anesth 2009;23(4):477–82. Epub hyperventilation in patients with severe head injury: a randomised clinical
2009 Nov 18. trial. J Neurosurg 1991;75:731–9.
51. Croughwell ND, Newman MF, Blumenthal JA, et al. Jugular bulb saturation 58. Schneider GH, Helden AV, Lanksch WR, et al. Continuous monitoring of
and cognitive dysfunction after cardiopulmonary bypass. Ann Thorac Surg jugular bulb oxygen saturation in comatose patients—therapeutic
1994;58:1702–8. implications. Acta Neurochir 1995;134:71–5.
52. Croughwell ND, White WD, Smith LR, et al. Jugular bulb saturation and 59. Sheinbrg M, Kanter MJ, Rogertson CS. Continuous monitoring of jugular
mixed venous saturation during cardiopulmonary bypass. J Card Surg 1995; venous oxygen saturation in head-injured patients. J Neurosurg
10:503–8. 1992;76:212–7.
53. Iwata M, Inoue S, Kawaguchi M, et al. The effect of hypercapnia and 60. Matta BF, Lam AM, Mayberg TS. The influence of arterial hyperoxygenation
hypertension on cerebral oxygen balance during one-lung ventilation for on cerebral venous oxygen content during hyperventilation. Can J Anaesth
lung surgery during propofol anesthesia. J Clin Anesth 2010;22(8):608–13. 1994;41:1041–6.
54. Takasu A, Yagi K, Ishihara S, Okada Y. Combined continuous monitoring of 61. Thiagarajan A, Goverdhan P, Chari P, et al. The effect of hyperventilation
systemic and cerebral oxygen metabolism after cardiac arrest. Resuscitation and hyperoxia on cerebral venous oxygen saturation in patients with
1995;29:189–94. traumatic brain injury. Anesth Analg 1998;87:850–3.
55. Bisschops LL, Hoedemaekers CW, Simons KS, et al. Preserved metabolic 62. Lewis S, Wong M, Myburgh J, et al. Determining cerebral perfusion pressure
coupling and cerebrovascular reactivity during mild hypothermia after thresholds in severe head trauma. Acta Neurochir Suppl (Wien) 1998;
cardiac arrest. Crit Care Med 2010;38(7):1542–7. 71:174–6.
Chapter
33  
V

Near Infrared Spectroscopy


Pippa G. Al-Rawi and Peter J. Kirkpatrick

measure tissue oxygenation in the adult brain. In addition,


Introduction NIRS use has expanded to examine oxygenation in other
Frans Jöbsis1 first described the use of in vivo tissue near tissues and to estimate the adequacy of systemic circulation14;
infrared spectroscopy (NIRS) in the human brain in 1977. this can be particularly useful in resuscitation from shock or
The technique is based on the concept that light of wave- sepsis.15-17 Technologic advances have led to the development
lengths 680 to 1000 nm is able to penetrate human tissue of compact, portable instruments that detect changes in
and is absorbed by the chromophores oxyhemoglobin (HbO2) optical attenuation of several wavelengths of light, with the
and deoxyhemoglobin (HHb) and the cytochrome oxidase. potential to derive a tissue oxygen saturation from spontane-
Changes in the detected light levels therefore can represent ous HHb and HbO2 signal changes.13,18-20 NIRS shows promise
changes in concentrations of these chromophores. as a clinical tool in the context of bedside cerebral blood flow
The noninvasive nature of the technique, which uses high- measurements, and to image the brain.21
intensity light emitted from sources placed on the surface of This chapter looks at the clinical application of NIRS but
the scalp, led to its first clinical application in monitoring the stresses the importance of both the assumptions on which
cerebral oxygenation status of premature infants. Initially, NIRS is based, and the limitations of the technology, to inter-
near infrared spectroscopy NIRS was not quantified and only pret the results correctly. It describes the authors’ use of this
provided a trend of increased or decreased oxygenation. The technique and how it is integrated with other monitoring
early machines were large and cumbersome, and the detected modalities.
signals showed considerable drift and were prone to move-
ment artifact.
The clinical use of NIRS for monitoring the brain is well
Basic Principles
established in neonates, in whom transillumination is possible The theory behind NIRS is described in detail elsewhere,22-24
because of the thin skull and small dimensions.2 However, the but the basic principle is based on the fact that light in the
clinical application of NIRS to monitor the adult brain has near infrared range (680-1000 nm) can pass through skin,
been hampered because it must be applied in reflectance bone, and other tissues with relative ease. The near infrared
mode. This has resulted in concerns about quantification, the light is delivered via fiber-optic bundles, or optodes, placed
volume and type of tissue being illuminated, and most signifi- either on opposite sides of the head, or close together (3.5-6 cm
cantly the issue of signal contamination by the extracranial distance) on the same side (Fig. 33.1). Light penetrates the
tissue layers.3 A number of algorithms have been applied to scalp and brain tissue and is subjected to scatter and absorp-
try to overcome these issues, and techniques such as time- tion within the various tissues. A fraction of the photons is
resolved, phase-resolved, and spatially resolved spectroscopy captured by the second optode and conveyed to a measuring
have been developed. device.
The use of NIRS to evaluate cerebral hemodynamic changes The concentration of some light-absorbing compounds
has been reported in a variety of medical and neurosurgical such as melanin, bilirubin, and water remains virtually con-
conditions, including those associated with disturbed cerebral stant over time. However, the concentrations of others, such
circulation.4-12 The authors’ experience has shown that in as HbO2, HHb, and cytochrome oxidase, vary according to
patients with head injury, changes in oxyhemoglobin correlate tissue oxygenation and metabolism. Cytochrome oxidase, the
well with changes in jugular venous oxygen saturation (SjvO2), terminal enzyme of the respiratory chain, also contributes to
transcranial Doppler (TCD), and laser Doppler.9 The authors the near infrared spectrum of cortical tissue. To calculate the
also have seen that NIRS can provide a more sensitive indica- concentration of the chromophobes requires knowledge of
tor of desaturation events than SjvO2, and a threshold for the absorption spectra for HbO2 and HHb, and assumes
cerebral ischemia has been defined.13 constant scattering properties. The modified Beer-Lambert
NIRS is an evolving technology, and there has been renewed law that includes a differential path length factor to account
interest in NIRS as an easy-to-use, noninvasive technique to for scattering within the tissue can be used to convert
© Copyright 2013 Elsevier Inc. All rights reserved. 327
328 Section V—Cerebral Blood Flow

m Detector
Emitter 6.0 c
4.5-

Skin

Bone

CSF

Fig. 33.1  Schematic representation of near infrared


spectroscopy on a patient’s head. CSF, Cerebrospinal Brain
fluid. tissue

measurements of light attenuation into concentrations of both of the receiving probes. The difference in absorption
these chromophores. The Beer-Lambert law is defined as: coefficients between the two probes should then reflect
∆Α = L ⋅ ∆µa absorption and scatter within the cerebral tissue alone.
However, this is an oversimplification that has resulted in
where A = light attenuation, L = differential path length, and measurement inaccuracies.35
µa = absorption scattering coefficient. The concepts about light distribution in the adult head are
This calculation is very reliable when applied to an infant’s complex and are still being investigated. Mathematical in vitro
head because the skull is thin enough to allow transillumina- modeling has attempted to predict the distribution of light
tion of light from one side to the other. In an adult, however, through the extracerebral layers.36 Multilayer studies have
the relative thickness of the scalp, skull, and brain prevents highlighted the influence that superficial tissue layers can have
transmission spectroscopy, and NIRS must be used in reflec- on light distribution in tissues and provide further evidence
tance mode, with the emitting and receiving optodes placed that NIRS measurements may underestimate changes in con-
on the same side of the head. Reflectance mode oximetry centration. Mathematical simulations using Monte Carlo
depends on a proportion of the light that passes through brain techniques and finite element modeling have demonstrated
tissue. The human head is comprised of multiple tissue layers, that the cerebrospinal fluid (CSF) layer that surrounds the
all of which have different scattering properties and contain brain can act as an optical short-cut for light as it passes
varying concentrations of light-absorbing compounds.25 This through the tissues. It has been suggested that this phenom-
results in the introduction of unknown, nonlinear variables enon can prevent the near infrared light from penetrating
for light absorption and scattering coefficients.26 Various algo- any deeper than the cerebral grey matter, or even cause it to
rithms have been applied to attempt quantification, but doubts bypass the cerebral compartment altogether.37,38
are expressed over their accuracy.27-30 Modern near infrared spectrometers detect oxygenation
changes in cerebral tissues. However, many machines still
remain sensitive, by varying degrees, to changes in extracere-
Extracranial Contamination bral oxygenation. These changes can influence estimates of
Extracranial contamination is a major issue during the use cerebral oxygenation. Conditions in which changes in extra-
of NIRS in the adult brain, although extracranial contribu- cerebral oxygenation may seem unlikely should not be taken
tions to NIRS measurements initially were thought to be for granted. For example, changes in extracerebral oxygen-
insignificant. Bone was considered to be transparent to light, ation in the scalp have been detected during the performance
whereas the skin contribution was estimated to be approxi- of functional activation tasks. Even in the absence of extrace-
mately 5%. Over time, however, numerous studies have rebral oxygenation changes, the extracerebral tissues may con-
demonstrated that extracranial contamination is a significant found attempts to calibrate and quantify changes in cerebral
problem.29,31-33 oxygenation. In any NIRS study it is essential to consider
In reflectance mode, the near infrared light emitter and whether the changes detected could be due, even in part, to
detectors are placed within a few centimeters of each other extracerebral contamination. To reliably measure cerebral
on the surface of the scalp. The larger the optode distance oxygenation with NIRS, extracerebral changes must be shown
used, the greater the proportion of near infrared light that to be insignificant, or their effect eliminated.
should pass through the brain tissue. However, even when a
large inter-optode distance is used there is still a major con-
tribution from the extracranial tissue to the NIRS signal.34
Instrumentation
Other methods are used to correct for the effects of the There has been rapid growth and development of NIRS tech-
extracranial tissue layers; one is to subtract measurements nology, and novel optical instruments continue to be devel-
from two receiving optodes placed in line with the transmit- oped and evaluated for diagnostic and functional imaging in
ting optode. The underlying assumption is that the path the clinical environment. Although the early machines
length of light through the extracranial tissues is the same for were useful only as trend monitors, current commercial
Section V—Cerebral Blood Flow 329

machines detect changes in optical attenuation of a number changes are measured by the middle photodiode, whereas TOI
of wavelengths of light, are compact, are portable, and enable is measured by using all three. TOI is the ratio of oxygenated
noninvasive measurements of cerebral oxygenation at the to total tissue hemoglobin and can be expressed as:
bedside.13,18-20 HbO2
The INVOS Cerebral Oximeter (Somanetics, Troy, MI) and × 100
the Hamamatsu 100, 200, and 300 (Hamamatsu Photonics HbO2 + HHb
KK, Hamamatsu, Japan) are among popular commercially The NIRO 200 and 100 are the latest commercial updates to
available devices for brain monitoring. In addition, multi- the NIRO 300 and use only three wavelengths of delivered
probe devices (e.g., INVOS 5100) are used in newborns or light (775, 810, and 850 nm) for the measurement calcula-
pediatric intensive care units (ICUs) to examine regional cere- tions. The NIRO 200 is a two-channel system that allows
bral (rSO2C), splanchnic (rSO2S), and renal (rSO2R) tissue bihemispheric data to be collected, whereas the NIRO 100 is
oxygenation. Several research groups also have produced their a single-channel machine.
own specialized in-house NIRS equipment, which may be
available locally. When published values from different instru-
ments are compared, it should be remembered that different Clinical Neuromonitoring
instruments may provide different values for cerebral tissue
oxygen saturations because the data are derived from different Clinical Application
volumes of tissue.39 Among current techniques, spatially NIRS has many applications in the ICU and the operating
resolved spectroscopy appears to be the most promising, and room, and it can be used to detect real-time changes in
reliably shows changes in chromophores in brain tissue.13,40,41 regional oxygen saturation of cerebral and somatic tissues.14
Its use is well described in cardiac surgery46 and neonatal and
pediatric critical care,2,47-49and most recently several studies
Spatially Resolved Spectroscopy have described use of NIRS to evaluate tissue perfusion and
Spatially resolved spectroscopy is described in a number of the dynamic response of the microcirculation to a stress test
publications.8,42,43 Spatial resolution relies on the measure- in patients who are dehydrated, septic, or in shock.15-17,48,50
ment of the attenuation gradient as a function of source- These studies in critically ill adults suggest NIRS can be used
detector separation. Using a modified diffusion equation, a to guide goal-directed therapy and that blood pressure is not
product of the absorption and scattering coefficients is calcu- always a reliable surrogate for peripheral blood flow and
lated. When the tissue is treated as homogeneous, the scatter- oxygen delivery. In addition, greater mortality is observed in
ing coefficient can be assumed to be a constant (k) in the near septic patients in whom forearm skeletal muscle rSO2 is less
infrared wavelength. To increase the accuracy of the calcula- than or equal to 60% throughout the first 24 hours of ICU
tion, however, a wavelength dependency for the scattering care and low tissue oxygen saturation (StO2) persists in sicker
coefficient is derived in the form of: patients (e.g., greater Acute Physiology and Chronic Health
Evaluation [APACHE] or Sequential Organ Failure Assess-
k(1− hλ)
ment [SOFA] scores). This information about systemic
where λ = wavelength and h is the normalized slope of the oxygenation can be very useful in many patients in the neuro-
scattering coefficient along λ. From here, the relative absorp- critical care unit (NCCU) who may develop sepsis or systemic
tion coefficients and thus the relative concentrations of HbO2 inflammatory response syndrome (SIRS) among other sys-
and HHb can be obtained. temic disorders, because many shock states have been shown
to be associated with a decrease in perfused capillary density
and an increase in the heterogeneity of microcirculatory
Hamamatsu NIRO 300 perfusion.15
The NIRO 300 is a noninvasive bedside monitor that provides The ability to monitor cerebral oxygenation noninvasively
continuous online measurements of hemoglobin and cyto- and at the bedside in patients at risk of cerebral ischemia is a
chrome oxidase concentrations, and a calculated tissue oxygen potential advantage of NIRS and its use to evaluate changes
index (TOI). The optodes are held in a black custom-made in regional cerebral oxygenation, cerebral blood flow (CBF),
holder that allows them to be set at either a 4.5- or 5-cm dis- and oxygen utilization in the brain is reported in a variety of
tance. By using two sets of optodes, two channel simultaneous medical and neurosurgical conditions,8,13,14,51-54 in particular
measurements are possible. those associated with disturbed cerebral circulation such as
Four wavelengths of light (775, 810, 850, and 910 nm) are head injury and intracranial hemorrhage, and in patients
delivered by four pulsed laser diodes, and the scattered light undergoing carotid endarterectomy.9,55-57 In head-injured
is detected by three closely placed photodiodes. The concen- patients changes in HbO2 detected by NIRS appear to correlate
tration changes of the chromophores HbO2, HHb, total hemo- well with changes in SjvO2, TCD, and laser Doppler flowmetry
globin, and cytochrome oxidase are measured by conventional (LDF).9 The information for NIRS also appears to have some
differential spectroscopy using a modified Beer-Lambert prognostic information (e.g., cerebral oxygen desaturation is
law,44,45 whereas the basic principle behind calculation of the associated with prolonged intensive care and total hospital
tissue oxygenation index is spatially resolved reflectance spec- lengths of stay).58 Zweifel et al.4 compared an NIRS-based
troscopy. The basic measurement made is the rate of increase index of cerebrovascular pressure reactivity (PRx), called total
of light attenuation with respect to source or detector spacing, hemoglobin reactivity (THx), against standard measurements
and TOI is calculated from this using photon diffusion of PRx that requires invasive intracranial pressure (ICP) mon-
theory.43 The hemoglobin and cytochrome concentration itoring. PRx and THx had a significant association across
330 Section V—Cerebral Blood Flow

averaged individual recordings and across patients. Optimal Direct monitoring of skin flow is possible using LDF to assess
cerebral perfusion pressure (CPP) could also be assessed with relative changes in scalp blood flow. If the LDF probe is placed
THx in about half of patients. These data suggest that NIRS on the skin in the immediate vicinity of the NIRS probes, skin
may be useful in some traumatic brain injury (TBI) patients flow changes that may affect NIRS measurements can be
to optimize and target therapy particularly those who do not crudely monitored.
have an ICP monitor. Others have described the use of NIRS Second, most NIRS systems are placed in the frontotempo-
to help guide therapy for vasospasm after subarachnoid hem- ral region for adult brain tissue monitoring. For optimal per-
orrhage (SAH)9 and observed that NIRS may distinguish dif- formance the skin surface should be clean and dry and the
ferent cerebral oxygenation patterns associated with different optodes placed high on the forehead and in a position to avoid
seizure types.59 the sagittal sinus, muscle (temporalis muscle), and midline.
Intraoperative use of NIRS during cardiac surgery7,8 or Hair also should be avoided whenever possible because it can
carotid endarterectomy (CEA) is well described.10,13,55,60,61 reduce return of the near infrared signal because of light
These studies have helped to define a potential NIRS-based absorption by the hair follicles. When hair is present it also is
“threshold” for ischemia particularly when using spatially difficult to adequately seal out light. Third, ambient light
resolved spectroscopy (e.g., NIRO 300, Hamamatsu Photon- should be eliminated. The use of a bandage to eliminate light
ics).13 However, it is difficult to determine how useful NIRS is that covers the optode holder is beneficial. It also will help to
during cardiac surgery without fully understanding how reduce scalp movement during the study. Care is needed to
extracorporeal circulation affects the assumptions on which avoid tissue ischemia and necrosis especially with long-term
NIRS technology is based. The information from CEA studies monitoring. Fourth, it is useful to support the optical fiber
may be more relevant to the NCCU. For example, the NIRO cables to prevent both damage and the weight of the fibers
500 can be used successfully to monitor patients who undergo dislodging the optodes. Finally, using NIRS as part of a mul-
CEA under general anesthesia.10 In some studies once the timodal monitoring system helps to confirm true clinical
extracranial component is subtracted, a threshold for severe changes versus artifact. When interpreting NIRS values, signal
critical ischemia can be defined.62,63 This threshold then can change or percentage change from baseline may be more
be used to guide the selective use of an intraoperative carotid useful than absolute values. This is particularly true if com-
artery shunt. Other studies using bilateral cerebral oximetry parisons between individual patients, clinical centers, or NIRS
in patients undergoing CEA under regional anesthesia62 or machines are required.
when INVOS is compared with somatosensory evoked poten-
tials51; however, they have not found it possible to determine
the critical cerebral oxygen saturation or change in saturation Limitations
that requires shunt insertion because of marked individual A number of limitations should be considered before NIRS
variability of rSO2 and the derived changes. In addition, cere- can contribute to clinical decision making. First, the major
bral oxygen saturation may decrease without neurologic limitation of NIRS is extracranial contamination. The hetero-
dysfunction. geneity of the scalp and skull affect the transmission of near
infrared light through the head and may cause significant
individual variability. Second, NIRS monitors cerebral oxygen
Troubleshooting saturation in an uncertain mixture of arteries, capillaries, and
Troubleshooting advice provided by the manufacturer comes veins. The sampling volume of gray and white matter is
with all near infrared spectrometers and in the first instance unknown. Third, current NIRS probes are not magnetic reso-
the user should consult the appropriate operating guide. nance imaging (MRI) compatible; this limits its use in poten-
However, some issues may be considered common to all NIRS tial MRI validation studies. Fourth, NIRS does not resolve
devices and studies. focal abnormalities in CBF but may be useful as a monitor of
First, the light source and detector optodes should be spaced general changes that indicate secondary brain injury. Fifth,
according to the study requirements, although increasing the some early studies suggest elevated intracranial pressure may
optode distance may help improve the sensitivity of NIRS to limit the reliability of NIRS.65 Finally, the quantitative accu-
brain tissue changes.30 An optode distance greater than 3 to racy of calculated cerebral oxygen saturation still needs to be
4 cm should reduce the extracerebral contribution because as established, and a range of normal values defined.
the optode distance is increased, a greater proportion of light
passes through the brain. Germon et al.34 in a study in which
optode distance was increased during NIRS studies suggest Evaluation of the NIRO 300 as a Monitor
that a separation of 5.5 cm may increase sensitivity to cerebral of Cerebral Ischemia
changes, whereas extracranial effects may plateau at 4.8 to Patients who undergo CEA often may experience brief periods
5.5 cm. By contrast when placed close together most of the of cerebral ischemia during cross-clamping of the internal
transmitted light likely passes through the scalp and superfi- carotid artery (ICA). During CEA, segregation of the NIRS
cial tissue. For example, in normal volunteers during verbal signal between intra- and extracranial vascular territories is
fluency tasks, optodes placed 5 mm apart may only measure possible by staged application of vascular clamps to the exter-
skin changes rather than reflect any cerebral function.64 The nal and internal carotid arteries. In this way, the authors have
distance between optodes and emitter also may depend on previously identified NIRS thresholds for cerebral ischemia by
practical constraints such as other monitors, surgical sites, extracting the extracranial signal.62
dressings, and the signal-to-noise ratio. Measurement of both At the authors’ institution routine multimodal monitoring
arterial blood pressure and peripheral saturation can address during CEA includes frontal cutaneous LDF, TCD mean flow
in part how extracranial contamination affects the signal. velocity (FV) measurements of the ipsilateral middle cerebral
Section V—Cerebral Blood Flow 331

LDF

NIRS

TCD

cfm

Fig. 33.2  Intraoperative multimodal monitoring setup, including cerebral function monitoring (cfm), transcranial Doppler (TCD), laser Doppler flow
(LDF), and near infrared spectroscopy (NIRS).

artery, and mean arterial blood pressure (ABP).10 The LDF Typical data obtained during CEA for an individual patient
(Moor Instruments Ltd, Axminster, UK) is modified to use are illustrated in Figure 33.3. The authors found that changes
light in the visible spectrum (wavelength 650 nm) to avoid any in TOI correlated with changes in intracranial blood flow
interference with the NIRS signal. The authors incorporated (i.e., FV), but not with cutaneous skin flow (LDF). Notably,
the NIRO 300 into this multimodal monitoring system to ΔTOI was not seen without an associated ΔFV. These obser-
detect cerebral hemodynamic changes during highly con- vations demonstrate that TOI is derived largely from the
trolled conditions and determine the anatomic source of the intracranial (ICA) vascular bed. The sensitivity and specificity
calculated TOI (%) (Fig. 33.2).13 of TOI change to intracranial changes (as detected by TCD)
The authors studied 167 patients who had elective CEA. was 87.5% and 100%, respectively.67 Most importantly, the
Monitoring was applied after induction of anesthesia. NIRS authors defined a %ΔTOI threshold of −13%, above which no
optodes were placed high on the ipsilateral forehead to avoid patients showed any evidence of ischemia (Fig. 33.4). This
the temporalis muscle and hair and sufficiently lateral from threshold provided 100% sensitivity and 93.2% specificity to
the midline to avoid the superior sagittal sinus. Optode spacing identify patients who met preset criteria for cerebral ischemia
was 5 cm (see Fig. 33.2). Sequential clamping of the external during CEA.13
carotid artery (ECA) followed by the ICA was performed It is important to realize that both relative changes in chro-
intraoperatively with a sufficient time interval (≈ 2 minutes) mophore concentration (using a modified Beer-Lambert Law)
to assess the extracranial contribution to the NIRS signal. and a quantified TOI (using spatially resolved spectroscopy)
Simultaneous measurements of hemoglobin concentration are provided with the NIRO 300. Biphasic changes were seen
and TOI changes (ΔTOI) were assessed by observing changes in HbO2 and HHb on sequential clamping, whereas ΔTOI was
in LDF (ΔLDF) and FV (ΔFV) to identify the extracerebral only associated with ICA-related chromophore changes (see
and intracerebral contribution to the NIRS signal. The time Fig. 33.3). The HbO2 and HHb values derived by the NIRO
of intraoperative events and in particular the time of vascular 300 therefore cannot reliably be taken to represent the intra-
clamp applications and data signals from all monitored cranial compartment in the same way as TOI and should be
parameters were digitized and collected on computers using interpreted with caution.
specialized software.66 Maximum physiologic stability gener-
ally is observed at the time of clamp application rather than
removal; therefore data from this period were used for analy- Cerebral Blood Volume
sis. Multivariable linear regression was used to calculate and Cerebral Blood Flow
the correlation coefficient between: (1) ΔTOI and ΔFV and There is no ideal method to measure cerebral blood volume
(2) ΔTOI and ΔLDF. To account for baseline variation in TOI, (CBV) or CBF safely, rapidly, repeatedly, and noninvasively at
interrupted time series analysis was applied. From these data the bedside. Established methods to measure CBF directly
a baseline threshold of 2% was derived, and therefore a ΔTOI tend to be technically challenging, are time consuming, use
greater than 2% was considered significant. radioactive substances, or require patient transport to an
332 Section V—Cerebral Blood Flow

ECA on ICA on Shunt inserted imaging site away from the NCCU.68-71 Because NIRS can
measure HHb and HbO2, it follows that it also can measure
1000 total hemoglobin (THgb). Assuming the whole blood HHb
800 concentration and the large to small cerebral vessel-to-hema-
LDF (au)

600 tocrit ratio remains constant for the duration of the measure-
400 ment, changes in THgb may be assumed to imply a change in
200
CBV, according to the following equation:
0 ∆CBV = ∆[THgb] × (0.89 / Hgb)
80
60 Calculation of absolute quantified CBV and CBF also is pos-
FVm (cm/s)

sible, based on the generalized application of the Fick prin-


40
ciple by using HbO2 and HHb as endogenous intravascular
20 tracer molecules, and assuming no venous outflow of the
0 chromophores during the measurement, quantified CBF and
4 CBV can be obtained by using transient graded hypoxia.72-75
HHb (µmol/L)

2.75 Despite a good agreement between NIRS CBF and 133Xe


washout,73,76 this technique has not been applied clinically
1.5
because of concerns over the induced hypoxemia and ques-
0.25 tions over the accuracy of the calculated CBV. Studies have
–1 shown significantly different CBV readings and a large inter-
4 subject variability.77
HbO2 (µmol/L)

2.39 However, it also is possible to measure blood flow by using


0.76 NIRS and a light-absorbing tracer. Broadly speaking, the rate
of accumulation of a tracer in a given tissue is equal to the
–0.87 rate of inflow minus the rate of outflow. If a tracer is intro-
–2.5 duced rapidly and the rate of accumulation is measured over
70 time, blood flow can be measured as a ratio of the tracer
62.5 accumulated to the quantity of tracer introduced, over a given
TOI (%)

55 time.
47.5
40
Indocyanine Green
0 1 2 3 4 5 6 7 8 9 Indocyanine green (ICG) is used as a diagnostic tool in
Time several medical applications. It is considered safe, and serious
side effects are uncommon. ICG shows a strong absorption in
Fig. 33.3  Graphic display of data obtained from a patient during the near infrared range, and changes in the attenuation of
elective carotid endarterectomy. The vertical lines demonstrate time
near infrared light, and therefore the optical density of bio-
of application of vascular clamps. ECA, External carotid artery; FVm,
mean ipsilateral middle cerebral artery flow velocity; HbO2, oxyhemoglobin
logic tissue, have been recorded after the administration of
concentration; HHb, deoxyhemoglobin concentration; ICA, internal ICG.78-81 Using ICG as the intravascular tracer, the arterial
carotid artery; LDF, frontal cutaneous laser Doppler flow; TOI, tissue concentration can be measured either by repeated blood
oxygen index.
10

5
0
–87 –77 –67 –57 –47 –37 –27 –17 7 3 13 23 33 43 53
% TOI diff (baseline, ICA clamp on)

–5

–10

–15

–20

–25

–30
Fig. 33.4  Scatterplot that compares percentage
change in middle cerebral artery (MCA) flow –35
velocity (%ΔFV) with percentage change in tissue
oxygen index (%ΔTOI) from baseline to –40
postapplication when the internal carotid artery –45
(ICA) was clamped in 167 patients during carotid
endarterectomy. Patients with a sustained fall in –50
ipsilateral cerebral function monitoring are
–55
marked in red. Dotted line indicates a level of
−13% ΔTOI. % FV diff (baseline, ICA clamp on)
Section V—Cerebral Blood Flow 333

sampling or with a peripheral dye densitometer, whereas the stimulation in the human brain frontal region by a cognitive
concentration of ICG in the brain is measured by NIRS. The process, were published in 1993.110 Since then, optical topog-
theory behind ICG use during NIRS is described in detail raphy using multichannel NIRS instruments has been success-
elsewhere.82 After baseline measurements rapid bolus injec- fully used to assess infant and adult brain function in a wide
tion of ICG (0.3-0.5 mg/kg) is given through a central vein variety of tasks,98,108,111-114 and strong correlation, particularly
and the arterial concentration measured either by blood sam- HbO2, has been found between functional magnetic resonance
pling or with a peripheral dye densitometer. The rate of imaging (fMRI) changes and NIR optical measures.114,115 The
arrival of ICG in the brain is detected by NIRS, and ICG con- current resolution of available instruments is similar to posi-
centrations can be calculated from the changes in optical tron emission tomography (PET); however, changes in skin
density.83 Using a modified Fick principle,84 the amount of blood flow still need to be accounted for.64
ICG dye delivered to the brain can be calculated from the NIR optical imaging, in which a transverse slice or three-
area under the curve of the arterial ICG concentration dimensional image is obtained, is technically more difficult.
change. CBF, CBV, and mean transit time (MTT) then can be Measurements are made using large source-detector spacings,
calculated.82-88 and consequently light must be integrated over time intervals
ICG NIRS has high spatial and temporal resolution, is safe, of several seconds or longer to obtain an adequate signal.
and can be performed easily at the bedside. Most studies have Although this prevents monitoring of fast hemodynamic
concluded that it shows reproducibility and merits further changes associated with functional activation, it does obtain
development as a clinical tool. In animal studies a good cor- information on oxygenation deeper in the brain, and has been
relation is found with 133Xe washout88,89 and ICG used with used to reveal variations in blood volume and tissue oxygen-
NIRS to measure CBF. The technique has been validated in ation in newborn babies suffering from hypoxic-ischemic
infants when compared with other methods for resting CBF brain injury.100,102,106,107 At publication, no standardized
measurements.87,90 However, attempts to quantify NIRS ICG approach for NIR optical imaging data analysis and image
in the adult brain have given erroneously low values.85,91-94 reconstruction has been established.
Further validation and modification of this technique as a
bedside measurement of CBF in adults are needed.
Future Developments
NIRS is an evolving technology that holds significant potential
Near Infrared Optical Imaging for technical advancement. Improvements in the methodol-
NIR optical imaging is an emerging technology with great ogy, quantitative accuracy, and regional specificity will increase
potential as an affordable noninvasive functional imaging its clinical applicability. In particular, NIRS shows promise as
modality for use in clinical practice. It provides the opportu- a low-cost noninvasive clinical tool for bedside CBF measure-
nity to study brain function through hemodynamic, meta- ments and as a cerebral imaging modality to map structure
bolic, and neuronal measures. van Houten et al.95 described and function. For these techniques to improve, more sophis-
the first imaging spectrometer capable of providing a two- ticated imaging algorithms must be developed to account for
dimensional image of the neonatal brain. Since then, a variety the heterogeneity of the tissues and to enable image recon-
of imaging methods have been applied,96-99 and continuous struction with higher spatial resolution and sensitivity. Inte-
real-time optical imaging of the brain has been shown to be gration of NIRS into multimodality monitoring and imaging
both feasible and achievable. systems, such as computed tomography (CT), MRI, and ultra-
The major requirement for bedside optical imaging is an sound, will allow for co-registration.
NIRS device that can accurately measure photon path length There are considerable research efforts to further develop
from multiple detectors and process this information into NIRS instrumentation and image reconstruction algorithms.
tomographic images. The use of near infrared light for imaging Portable NIRS instruments combined with wireless telemetry
presents a special challenge because of the multiple scattering are being tested. Soon it is likely that more multichannel
that takes place within the tissue, which complicates the image imaging near infrared spectrometers will be developed to
reconstruction. This has necessitated development of highly enable detailed spatial information on cerebral oxygenation,
sophisticated image reconstruction algorithms. The detailed perfusion, and blood flow to be obtained at the bedside. In
principles of these techniques are beyond the scope of this addition, combined ICP and NIRS monitors have been devel-
chapter, and can be found elsewhere.100-102 Novel optical oped, and with these devices new algorithms can be developed
instruments are being developed and evaluated by several to subtract the extracerebral contamination from the NIRS
groups for diagnostic and functional imaging in the clinical signal.116
environment.103-109
NIR optical imaging involves acquiring multiple reflectance
measurements using small source-detector spacings placed
Conclusion
over a large area of the head, either simultaneously or in rapid All neuromonitoring methods, including NIRS, require tech-
succession. The theory is based on a diffusion approximation, nical expertise and experience to efficiently interpret the
and two-dimensional images of hemodynamic and oxygen- results. However, NIRS has several potential advantages over
ation changes are reconstructed, based on the chromophore other monitors. It allows for continuous noninvasive moni-
concentrations in the tissues. Brain activity with responses as toring that is safe and relatively easy to use and can register
fast as 100 ms can be studied and the resolution obtained is complex mechanisms of oxygen balance in the brain. Having
in the order of 10 mm. However, only the cortical surface is a continuous, real-time measure of cerebral oxygenation can
imaged. potentially identify critical ischemic events before they mani-
The first reported observations of NIRS absorbance changes fest clinically and, in some cases, before other monitors may
attributable to repetitive total Hb changes in response to document changes. The major limitations associated with
334 Section V—Cerebral Blood Flow

NIRS, however, are changes that occur in the extracerebral 17. Nanas S, Gerovasili V, Renieris P, et al. Non-invasive assessment of the
microcirculation in critically ill patients. Anaesth Intensive Care 2009;37(5):
tissues that can significantly degrade the results. Among
733–9.
current commercially available NIRS instruments, the NIRO 18. Dullenkopf A, Frey B, Baenziger O, et al. Measurement of cerebral
300 appears to most reliably reflect changes in cerebral tissue oxygenation state in anaesthetized children using the INVOS 5100 cerebral
oxygenation to a high degree of sensitivity and specificity, and oximeter. Paediatr Anaesth 2003;13:384–91.
a threshold for cerebral ischemia during carotid endarterec- 19. McKeating EG, Monjardino JR, Signorini DF, et al. A comparison of the
INVOS 3100 and the Critikon 2020 near-infrared spectrophotometer as
tomy has been defined. monitors of cerebral oxygenation. Anaesthesia 1997;52:136–40.
NIRS technology has developed rapidly. A number of 20. Yoshitani K, Kawaguchi M, Tatsumi K, et al. A comparison of the INVOS
instruments for CBF, regional imaging, and functional activa- 4100 and the NIRO 300 near-infrared spectrometers. Anesth Analg 2002;94:
tion are being developed. NIRS could be of enormous clinical 586–90.
21. Ferrari M, Mottola L, Quaresima V. Principles, techniques and limitations of
value as a portable bedside monitoring tool that can be
near infrared spectroscopy. Can J Appl Physiol 2004;29(4):463–87.
coupled with other cerebral imaging modalities such as MRI 22. Cope M, Delpy DT. System for long-term measurement of cerebral blood
and PET. However, before routine use for monitoring the and tissue oxygenation on newborn infants by near infra-red
adult brain, it is recommended that several aspects of NIRS transillumination. Med Biol Eng Comp 1988;26:289–94.
technology need refinement. Further validation studies to 23. Piantadosi CA. Near infrared spectroscopy: principles and application to
non-invasive assessment of tissue oxygenation. J Crit Care 1989;4:308–18.
investigate NIRS reliability in different clinical scenarios are 24. Wyatt JS, Cope M, Delpy DT, et al. Measurement of optical path length for
required. The accuracy of the regional cerebral oxygen satura- cerebral near-infrared spectroscopy in newborn infants. Dev Neurosci 1990;
tion as an absolute value may be deceiving because no reliable 12:140–4.
data about normal or critical threshold values exist, and so the 25. Dehghani H, Delpy DT. Near infrared spectroscopy of the adult head: effect
of scattering and absorbing obstructions in the CSF layer on light
clinician is unable to check the accuracy of the given values.
distribution in the tissue. Appl Opt 2000;39:4721–9.
For this reason, only the trend information should be included 26. Okada E, Firbank M, Schweiger M, et al. Theoretical and experimental
in the evaluation of cerebral oximetry. investigation of near-infrared light propagation in a model of the adult head.
Appl Opt 1997;36(1):21–31.
27. Harris DN, Bailey SM. Near infrared spectroscopy in adults: does the
References INVOS 3100 really measure intracerebral oxygenation? Anaesthesia 1993;48:
694–6.
1. Jöbsis FF. Non-invasive infrared monitoring of cerebral and myocardial 28. Komiyama T, Quaresima V, Shigematsu H, et al. Comparison of two spatially
oxygen sufficiency and circulatory parameters. Science 1977;198:1264–7. resolved near-infrared photometers in the detection of tissue oxygen
2. Toet MC, Lemmers PM. Brain monitoring in neonates. Early Hum Dev saturation: poor reliability at very low oxygen saturation. Clin Sci 2001;101:
2009;85(2):77–84. Epub 2009, Jan 17. 715–8.
3. Schwarz G, Litscher G, Kleinert R, et al. Cerebral oximetry in dead subjects. 29. Kytta J, Ohman J, Tanskanen P, et al. Extracranial contribution to cerebral
J Neurosurg Anesthesiol 1996;8(3):189–93. oximetry in brain dead patients: a report of six cases. J Neurosurg
4. Zweifel C, Castellani G, Czosnyka M, et al. Noninvasive monitoring of Anaesthesiol 1999;11(4):252–4.
cerebrovascular reactivity with near infrared spectroscopy in head-injured 30. Litscher G, Schwarz G. Transcranial cerebral oximetry: is it clinically useless
patients. J Neurotrauma 2010;27(11):1951–8. Epub 2010, Oct 28. at this moment to interpret absolute values obtained by the INVOS 3100
5. Kim MN, Durduran T, Frangos S, et al. Noninvasive measurement of cerebral oximeter? Biomed Tech (Berl) 1997;42(4):74–7.
cerebral blood flow and blood oxygenation using near-infrared and diffuse 31. Young AER, Germon TJ, Barnett NJ, et al. Behaviour of near-infrared light
correlation spectroscopies in critically brain-injured adults. Neurocrit Care in the adult human head: implications for clinical near-infrared
2010;12(2):173–80. spectroscopy. Br J Anaesth 2000;84(1):38–42.
6. Mutoh T, Ishikawa T, Suzuki A, et al. Continuous cardiac output and 32. Germon TJ, Evans P, Barnett N, et al. Optode separation determines
near-infrared spectroscopy monitoring to assist in management of sensitivity of near infrared spectroscopy to intra- and extracranial
symptomatic cerebral vasospasm after subarachnoid hemorrhage. oxygenation changes. J Cereb Blood Flow Metab 1996;15:617.
Neurocrit Care 2010;13(3):331–8. 33. Samra SK, Stanley JC, Zelenock GB, et al. An assessment of contributions
7. du Plessis AJ, Newburger J, Jonas RA, et al. Cerebral oxygenation supply and made by extracranial tissues during cerebral oximetry. J Neurosurg
utilisation during infact cardiac surgery. Ann Neurol 1995;37:488–97. Anaesthesiol 1999;11(1):1–5.
8. Al-Rawi PG, Smielewski P, Hobbiger H, et al. Assessment of spatially 34. Germon TJ, Evans DH, Barnett N, et al. Cerebral near infrared spectroscopy:
resolved spectroscopy during cardiopulmonary bypass. J Biomed Opt emitter-detector separation must be increased. Br J Anaesth 1999;82(6):
1999;4(2):208–16. 831–7.
9. Kirkpatrick PJ, Smielewski P, Czosnyka M, et al. Near infrared spectroscopy 35. Yoshitani K, Kawaguchi M, Okuno T, et al. Measurement of optical path
use in patients with head injury. J Neurosurg 1995;83:963–70. length using phase-resolved spectroscopy in patients undergoing
10. Kirkpatrick PJ, Smielewski P, Whitfield P, et al. An observational study of cardiopulmonary bypass. Anesth Analg 2007;104:341–6.
near infrared spectroscopy during carotid endarterectomy. J Neurosurg 36. Hiraoka M, Firbank M, Essenpries M. A Monte Carlo investigation of optical
1995;82:756–63. path length in inhomogeneous tissue and its application to near infrared
11. Kurth CD, Steven JM, Nicholson SC. Cerebral oxygenation during pediatric spectroscopy. Phys Med Biol 1993;38:1859–76.
surgery using deep hypothermic circulatory arrest. Anaesthesiology 37. Firbank M, Schweiger M, Delpy DT. Investigation of light piping through
1995;82:74–82. clear regions of scattering objects. SPIE 1995;2389:167–73.
12. Liem KD, Hopman JCW, Oeseburg B, et al. Cerebral oxygenation and 38. Okada E, Firbank M, Schweiger M, et al. A theoretical and experimental
haemodynamics during induction of extracorporeal membrane oxygenation investigation of the effect of sulci on light propagation in brain tissue. SPIE
as investigated by near infrared spectroscopy. Paediatrics 1995;95:555–61. 1995;2626:2–8.
13. Al-Rawi PG, Kirkpatrick PJ. Tissue oxygen index (TOI): thresholds for 39. Delpy DT, Cope M. Quantification in tissue near-infrared spectroscopy. Phil
cerebral ischaemia using near infrared spectroscopy. Stroke 2006;37: Trans R Soc Lond B 1997;352:649–59.
2720–5. 40. Fantini S, Franceschini MA, Maier JS, et al. Frequency-domain multichannel
14. Moerman A, Wouters P. Near-infrared spectroscopy (NIRS) monitoring in optical detector for non-invasive tissue spectroscopy and oximetry. Opt Eng
contemporary anesthesia and critical care. Acta Anaesthesiol Belg 2010;61(4): 1995;34:32–42.
185–94. 41. Hazeki O, Tamura M. Quantitative analysis of haemoglobin oxygenation
15. De Backer D, Ospina-Tascon G, Salgado D, et al. Monitoring the state of brain in situ by near-infrared spectrophotometry. J App Physiol
microcirculation in the critically ill patient: current methods and future 1988;64:796–802.
approaches. Intensive Care Med 2010;36(11):1813–25. Epub 2010, Aug 6. 42. Matcher SJ, Kirkpatrick PJ, Nahid K, et al. Absolute quantification methods
16. Rodriguez A, Lisboa T, Martín-Loeches I, et al. Mortality and regional in tissue near infrared spectroscopy. Proc SPIE 1993;2389:486–95.
oxygen saturation index in septic shock patients: a pilot study. J Trauma 43. Suzuki S, Takasaki S, Ozaki T, et al. A tissue oxygenation monitor using NIR
2011;70(5):1145–52. spatially resolved spectroscopy. Proc SPIE 1999;3597:582–92.
Section V—Cerebral Blood Flow 335

44. Delpy DT, Cope M, van der Zee P, et al. Estimation of optical pathlength 48. Hanson SJ, Berens RJ, Havens PL, et al. Effect of volume resuscitation
through tissue from direct time of flight measurement. Phys Med Biol 1988; on regional perfusion in dehydrated pediatric patients as measured by
33(12):1433–42. two-site near-infrared spectroscopy. Pediatr Emerg Care 2009;25(3):
45. Patterson MS, Chance B, Wilson BC. Time resolved reflectance and 150–3.
transmittance for the noninvasive measurement of tissue optical properties. 49. Petrova A, Mehta R. Near-infrared spectroscopy in the detection of regional
Appl Opt 1989;28:2331–6. tissue oxygenation during hypoxic events in preterm infants undergoing
46. Brady K, Joshi B, Zweifel C, et al. Real-time continuous monitoring of critical care. Pediatr Crit Care Med 2006;7(5):449–54.
cerebral blood flow autoregulation using near-infrared spectroscopy 50. Lima A, van Bommel J, Jansen TC, et al. Low tissue oxygen saturation at the
in patients undergoing cardiopulmonary bypass. Stroke 2010;41(9): end of early goal-directed therapy is associated with worse outcome in
1951–6. critically ill patients. Crit Care 2009;13(Suppl 5):S13. Epub 2009, Nov 30.
47. Dani C, Pratesi S, Fontanelli G, et al. Blood transfusions increase cerebral,
splanchnic, and renal oxygenation in anemic preterm infants. Transfusion A complete list of references for this chapter can be found online at
2010;50(6):1220–6. Epub 2010, Jan 22. www.expertconsult.com.
Section V—Cerebral Blood Flow 335.e1

References 27. Harris DN, Bailey SM. Near infrared spectroscopy in adults: does the
INVOS 3100 really measure intracerebral oxygenation? Anaesthesia
1. Jöbsis FF. Non-invasive infrared monitoring of cerebral and myocardial 1993;48:694–6.
oxygen sufficiency and circulatory parameters. Science 1977;198:1264–7. 28. Komiyama T, Quaresima V, Shigematsu H, et al. Comparison of two
2. Toet MC, Lemmers PM. Brain monitoring in neonates. Early Hum Dev spatially resolved near-infrared photometers in the detection of tissue
2009;85(2):77–84. Epub 2009, Jan 17. oxygen saturation: poor reliability at very low oxygen saturation. Clin Sci
3. Schwarz G, Litscher G, Kleinert R, et al. Cerebral oximetry in dead subjects. 2001;101:715–8.
J Neurosurg Anesthesiol 1996;8(3):189–93. 29. Kytta J, Ohman J, Tanskanen P, et al. Extracranial contribution to cerebral
4. Zweifel C, Castellani G, Czosnyka M, et al. Noninvasive monitoring of oximetry in brain dead patients: a report of six cases. J Neurosurg
cerebrovascular reactivity with near infrared spectroscopy in head-injured Anaesthesiol 1999;11(4):252–4.
patients. J Neurotrauma 2010;27(11):1951–8. Epub 2010, Oct 28. 30. Litscher G, Schwarz G. Transcranial cerebral oximetry: is it clinically useless
5. Kim MN, Durduran T, Frangos S, et al. Noninvasive measurement of at this moment to interpret absolute values obtained by the INVOS 3100
cerebral blood flow and blood oxygenation using near-infrared and diffuse cerebral oximeter? Biomed Tech (Berl) 1997;42(4):74–7.
correlation spectroscopies in critically brain-injured adults. Neurocrit Care 31. Young AER, Germon TJ, Barnett NJ, et al. Behaviour of near-infrared light
2010;12(2):173–80. in the adult human head: implications for clinical near-infrared
6. Mutoh T, Ishikawa T, Suzuki A, et al. Continuous cardiac output and spectroscopy. Br J Anaesth 2000;84(1):38–42.
near-infrared spectroscopy monitoring to assist in management of 32. Germon TJ, Evans P, Barnett N, et al. Optode separation determines
symptomatic cerebral vasospasm after subarachnoid hemorrhage. Neurocrit sensitivity of near infrared spectroscopy to intra- and extracranial
Care 2010;13(3):331–8. oxygenation changes. J Cereb Blood Flow Metab 1996;15:617.
7. du Plessis AJ, Newburger J, Jonas RA, et al. Cerebral oxygenation supply 33. Samra SK, Stanley JC, Zelenock GB, et al. An assessment of contributions
and utilisation during infact cardiac surgery. Ann Neurol 1995;37:488–97. made by extracranial tissues during cerebral oximetry. J Neurosurg
8. Al-Rawi PG, Smielewski P, Hobbiger H, et al. Assessment of spatially Anaesthesiol 1999;11(1):1–5.
resolved spectroscopy during cardiopulmonary bypass. J Biomed Opt 34. Germon TJ, Evans DH, Barnett N, et al. Cerebral near infrared
1999;4(2):208–16. spectroscopy: emitter-detector separation must be increased. Br J Anaesth
9. Kirkpatrick PJ, Smielewski P, Czosnyka M, et al. Near infrared spectroscopy 1999;82(6):831–7.
use in patients with head injury. J Neurosurg 1995;83:963–70. 35. Yoshitani K, Kawaguchi M, Okuno T, et al. Measurement of optical path
10. Kirkpatrick PJ, Smielewski P, Whitfield P, et al. An observational study of length using phase-resolved spectroscopy in patients undergoing
near infrared spectroscopy during carotid endarterectomy. J Neurosurg cardiopulmonary bypass. Anesth Analg 2007;104:341–6.
1995;82:756–63. 36. Hiraoka M, Firbank M, Essenpries M. A Monte Carlo investigation of
11. Kurth CD, Steven JM, Nicholson SC. Cerebral oxygenation during pediatric optical path length in inhomogeneous tissue and its application to near
surgery using deep hypothermic circulatory arrest. Anaesthesiology 1995; infrared spectroscopy. Phys Med Biol 1993;38:1859–76.
82:74–82. 37. Firbank M, Schweiger M, Delpy DT. Investigation of light piping through
12. Liem KD, Hopman JCW, Oeseburg B, et al. Cerebral oxygenation and clear regions of scattering objects. SPIE 1995;2389:167–73.
haemodynamics during induction of extracorporeal membrane 38. Okada E, Firbank M, Schweiger M, et al. A theoretical and experimental
oxygenation as investigated by near infrared spectroscopy. Paediatrics investigation of the effect of sulci on light propagation in brain tissue. SPIE
1995;95:555–61. 1995;2626:2–8.
13. Al-Rawi PG, Kirkpatrick PJ. Tissue oxygen index (TOI): thresholds for 39. Delpy DT, Cope M. Quantification in tissue near-infrared spectroscopy.
cerebral ischaemia using near infrared spectroscopy. Stroke 2006;37: Phil Trans R Soc Lond B 1997;352:649–59.
2720–5. 40. Fantini S, Franceschini MA, Maier JS, et al. Frequency-domain
14. Moerman A, Wouters P. Near-infrared spectroscopy (NIRS) monitoring in multichannel optical detector for non-invasive tissue spectroscopy and
contemporary anesthesia and critical care. Acta Anaesthesiol Belg 2010; oximetry. Opt Eng 1995;34:32–42.
61(4):185–94. 41. Hazeki O, Tamura M. Quantitative analysis of haemoglobin oxygenation
15. De Backer D, Ospina-Tascon G, Salgado D, et al. Monitoring the state of brain in situ by near-infrared spectrophotometry. J App Physiol
microcirculation in the critically ill patient: current methods and future 1988;64:796–802.
approaches. Intensive Care Med 2010;36(11):1813–25. Epub 2010, Aug 6. 42. Matcher SJ, Kirkpatrick PJ, Nahid K, et al. Absolute quantification methods
16. Rodriguez A, Lisboa T, Martín-Loeches I, et al. Mortality and regional in tissue near infrared spectroscopy. Proc SPIE 1993;2389:486–95.
oxygen saturation index in septic shock patients: a pilot study. J Trauma 43. Suzuki S, Takasaki S, Ozaki T, et al. A tissue oxygenation monitor using
2011;70(5):1145–52. NIR spatially resolved spectroscopy. Proc SPIE 1999;3597:582–92.
17. Nanas S, Gerovasili V, Renieris P, et al. Non-invasive assessment of the 44. Delpy DT, Cope M, van der Zee P, et al. Estimation of optical pathlength
microcirculation in critically ill patients. Anaesth Intensive Care 2009;37(5): through tissue from direct time of flight measurement. Phys Med Biol
733–9. 1988;33(12):1433–42.
18. Dullenkopf A, Frey B, Baenziger O, et al. Measurement of cerebral 45. Patterson MS, Chance B, Wilson BC. Time resolved reflectance and
oxygenation state in anaesthetized children using the INVOS 5100 cerebral transmittance for the noninvasive measurement of tissue optical properties.
oximeter. Paediatr Anaesth 2003;13:384–91. Appl Opt 1989;28:2331–6.
19. McKeating EG, Monjardino JR, Signorini DF, et al. A comparison of the 46. Brady K, Joshi B, Zweifel C, et al. Real-time continuous monitoring of
INVOS 3100 and the Critikon 2020 near-infrared spectrophotometer as cerebral blood flow autoregulation using near-infrared spectroscopy in
monitors of cerebral oxygenation. Anaesthesia 1997;52:136–40. patients undergoing cardiopulmonary bypass. Stroke 2010;41(9):
20. Yoshitani K, Kawaguchi M, Tatsumi K, et al. A comparison of the INVOS 1951–6.
4100 and the NIRO 300 near-infrared spectrometers. Anesth Analg 2002; 47. Dani C, Pratesi S, Fontanelli G, et al. Blood transfusions increase cerebral,
94:586–90. splanchnic, and renal oxygenation in anemic preterm infants. Transfusion
21. Ferrari M, Mottola L, Quaresima V. Principles, techniques and limitations 2010;50(6):1220–6. Epub 2010, Jan 22.
of near infrared spectroscopy. Can J Appl Physiol 2004;29(4):463–87. 48. Hanson SJ, Berens RJ, Havens PL, et al. Effect of volume resuscitation on
22. Cope M, Delpy DT. System for long-term measurement of cerebral blood regional perfusion in dehydrated pediatric patients as measured by two-site
and tissue oxygenation on newborn infants by near infra-red near-infrared spectroscopy. Pediatr Emerg Care 2009;25(3):150–3.
transillumination. Med Biol Eng Comp 1988;26:289–94. 49. Petrova A, Mehta R. Near-infrared spectroscopy in the detection of regional
23. Piantadosi CA. Near infrared spectroscopy: principles and application to tissue oxygenation during hypoxic events in preterm infants undergoing
non-invasive assessment of tissue oxygenation. J Crit Care 1989;4:308–18. critical care. Pediatr Crit Care Med 2006;7(5):449–54.
24. Wyatt JS, Cope M, Delpy DT, et al. Measurement of optical path length for 50. Lima A, van Bommel J, Jansen TC, et al. Low tissue oxygen saturation at
cerebral near-infrared spectroscopy in newborn infants. Dev Neurosci the end of early goal-directed therapy is associated with worse outcome in
1990;12:140–4. critically ill patients. Crit Care 2009;13(Suppl 5):S13. Epub 2009, Nov 30.
25. Dehghani H, Delpy DT. Near infrared spectroscopy of the adult head: effect 51. Kirkpatrick PJ, Smielewski P, Lam JMK, et al. Use of near infrared
of scattering and absorbing obstructions in the CSF layer on light spectroscopy for the clinical monitoring of adult brain. J Biomed Opt
distribution in the tissue. Appl Opt 2000;39:4721–9. 1996;1(4):363–72.
26. Okada E, Firbank M, Schweiger M, et al. Theoretical and experimental 52. McGown AD, Makker H, Elwell C, et al. Measurement of changes in
investigation of near-infrared light propagation in a model of the adult cytochrome oxidase redox state during obstructive sleep apnea using
head. Appl Opt 1997;36(1):21–31. near-infrared spectroscopy. Sleep 2003;26(6):710–6.
335.e2 Section V—Cerebral Blood Flow

53. Duncan A, Meek JH, Clemence M, et al. Optical pathlength measurement 79. Hongo K, Kobayashi S, Okudera H, et al. Noninvasive cerebral optical
on adult head, calf, forearm and the head of the newborn infant using spectroscopy: depth resolved measurements of creebral haemodynamics
phase resolved optical spectroscopy. Physiol Med Biol 1995;40:295–304. using indocyanine green. Neurol Res 1995;17:89–93.
54. Nollert G, Jonas RA, Reichart B. Optimising cerebral oxygenation during 80. Roberts I, Fallon P, Kirkham FJ, et al. Estimation of cerebral blood flow
cardiac surgery: a review of experimental and clinical investigations with with near infrared spectroscopy and indocyanine green. Lancet 1993;342:
near infrared spectrophotometry. Thorac Cardiovasc Surg 2000;48:247–53. 1425.
55. Beese U, Langer H, Lang W, et al. Comparison of near-infrared 81. Shinohara H, Tanaka A, Kital T, et al. Direct measurement of hepatic
spectroscopy and somatosensory evoked potentials for the detection of indocyanine green clearance with near infrared spectroscopy: separate
cerebral ischaemia during carotid endarterectomy. Stroke 1998;29:2032–7. evaluation of uptake and removal. Hepatology 1996;23:137–44.
56. Germon TJ, Kane NM, Manara AR, et al. Near-infrared spectroscopy in 82. Hopton P, Walsh TS, Lee A. Measurement of cerebral blood volume using
adults: effects of extracranial ischaemia and intracranial hypoxia on near-infrared spectroscopy and indocyanine green elimination. J Appl
estimation of cerebral oxygenation. Br J Anaesth 1994;73:503–6. Physiol 1999;87(5):1981–7.
57. Gopinath SP, Robertson CS, Contant CF, et al. Early detection of delayed 83. Keller E, Nadler A, Alkhadi H, et al. Noninvasive measurement of regional
traumatic intracranial hematomas using near-infrared spectroscopy. cerebral blood flow and regional cerebral blood volume by near infrared
J Neurosurg 2001;83:438–44. spectroscopy and indocyanine green dye dilution. NeuroImage 2003;22(3):
58. Yao F-S, Tseng C-C, Braverman JM, et al. Cerebral oxygen desaturation is 828−39.
associated with prolonged lengths of stay in the intensive care unit and 84. Springett R, Sakata Y, Delpy DT. Precise measurement of cerebral blood
hospital. Anesthesiology 1999;91:123. flow in newborn piglets from the bolus passage of indocyanine green. Phys
59. Sokol DK, Markand ON, Daly EC, et al. Near infrared spectroscopy (NIRS) Med Biol 2001;46:2209–25.
distinguishes seizure types. Seizure 2000;9(5):323–7. 85. Gora F, Shinde S, Elwell CE, et al. Noninvasive measurement of cerebral
60. Williams IM, Mead G, Picton AJ, et al. The influence of contralateral blood flow in adults using near-infrared spectroscopy and indocyanine
carotid stenosis and occlusion on cerebral oxygen saturation during carotid green: a pilot study. J Neurosurg Anaesthesiol 2002;14(3):218–22.
artery surgery. Eur J Vasc Endovasc Surg 1995;10:198–206. 86. Boushel R, Langberg H, Olesen J, et al. Regional blood flow during exercise
61. Samra SK, Dorje P, Zelenock GB, et al. Cerebral oximetry in patients in humans measured by near infrared spectroscopy and indocyanine green.
undergoing carotid endarterectomy under regional anaesthesia. Stroke J Appl Physiol 2000;89:1868–78.
1996;27:49–55. 87. Patel J, Marks K, Roberts I, et al. Measurement of cerebral blood flow in
62. Kirkpatrick PJ, Lam JMK, Al-Rawi PG, et al. Defining thresholds for critical newborn infants using near infrared spectroscopy with indocyanine green.
ischaemia by using near-infrared spectroscopy in the adult brain. Pediatr Res 1998;43:34–9.
J Neurosurg 1998;89(3):389–94. 88. Kuebler WM, Sckell A, Habler O, et al. Noninvasive measurement of
63. Lam JMK, Smielewski P, Al-Rawi P, et al. Internal and external carotid regional cerebral blood flow by near-infrared spectroscopy and indocyanine
contributions to near-infrared spectroscopy during carotid endarterectomy. green. J Cereb Blood Flow Metab 1998;18(4):445–56.
Stroke 1997;28(5):906–11. 89. Colacino JM, Grubb B, Jöbsis FF. Infra-red technique for cerebral blood
64. Takahashi T, Takikawa Y, Kawagoe R, et al. Influence of skin blood flow on flow: comparison with 133xenon clearance. Neurol Res 1981;3:17–31.
near-infrared spectroscopy signals measured on the forehead during a 90. Roberts IG, Fallon P, Kirkham FJ, et al. Measurement of cerebral blood flow
verbal fluency task. Neuroimage 2011;May 10. Epub ahead of print. during cardiopulmonary bypass with near infrared spectroscopy. J Thorac
65. Muellner T, Schramm W, Kwasny O, et al. Patients with increased Cardiovasc Surg 1998;115:94–102.
intracranial pressure cannot be monitored using near infrared spectroscopy. 91. Keller E, Nadler A, Alkadhi H, et al. Noninvasive measurement of regional
Br J Neurosurg 1998;12(2):136–9. cerebral blood flow and regional cerebral blood volume by near-infrared
66. Smielewski P, Czosnyka M, Steiner L, et al. ICM+: software for on-line spectroscopy and indocyanine green dye dilution. Neuroimage 2003;22(3):
analysis of bedside monitoring data after severe head trauma. Acta 828–39.
Neurochir Suppl 2005;95:43–9. 92. Rothoerl RD, Schebesch KM, Faltermeier R, et al. Lack of correlation
67. Al-Rawi PG, Smielewski P, Kirkpatrick PJ. Evaluation of a near infrared between Xenon133 and near infrared spectroscopy/indocyanine green rCBF
spectrometer (NIRO 300) for the detection of intracranial oxygenation measurements. Neurol Res 2003;25(5):528–32.
changes in the adult head. Stroke 2001;32(11):2492–500. 93. Al-Rawi PG, Leung TS, Smielewski P, et al. Quantification of cerebral blood
68. Gallagher CN, Hutchinson PJ, Pickard JD. Neuroimaging in trauma. Curr flow using near infrared spectroscopy and indocyanine green in patients
Opin Neurol 2007;20(4):403–9. with severe subarachnoid haemorrhage. Br J Neurosurg 2006;20(5):333–58.
69. Obrist WD, Thompson HK, Wang HS, et al. Regional cerebral blood flow 94. Kohl-Bareis M, Obrig H, Steinbrink J, et al. Noninvasive monitoring of
estimated by 133-xenon inhalation. Stroke 1975;6:245–56. cerebral blood flow by a dye bolus method: separation of brain from skin
70. Gideon P, Sperling B, Arlien-Soborg P, et al. Long-term follow-up of and skull signals. J Biomed Opt 2002;7(3):464–70.
cerebral infarction patients with proton magnetic resonance spectroscopy. 95. van Houten JP, Benaron DA, Spilman S, et al. Imaging brain injury using
Stroke 1994;25:967–73. time-resolved near infrared light scanning. Pediat Res 1996;39:470–6.
71. Saunders DE, Howe FA, van den Boogaart A, et al. Continuing ischaemic 96. Wolf M, Ferrari M, Quaresima V. Progress of near-infrared spectroscopy
damage after acute middle cerebral artery infarction in humans and topography for brain and muscle clinical applications. J Biomed Opt
demonstrated by short-echo proton spectroscopy. Stroke 1995;26: 2007;12(6):1–14.
1007–13. 97. Hoshi Y. Functional near-infrared spectroscopy: current status and future
72. Edwards AD, Wyatt JS, Richardson CE, et al. Cotside measurements of prospects. J Biomed Opt 2007;12(6):062106-1–062106-9.
cerebral blood flow in ill newborn infants by near infrared spectroscopy. 98. Obrig H, Villringer A. Beyond the visible; Imaging the human brain with
Lancet 1988;ii:770–1. light. J Cereb Blood Flow Metab 2003;23(1):1–18.
73. Skov L, Pryds O, Griesen G. Estimating cerebral blood flow in newborn 99. Strangman G, Boas DA, Sutton JP. Non-invasive neuroimaging using
infants: comparison of near infrared spectroscopy and 133Xe clearance. near-infrared light. Biol Psychiatry 2002;52:679–93.
Pediatr Res 1991;30:570–3. 100. Austin T, Gibson AP, Branco G, et al. Three dimensional optical imaging of
74. Wyatt JS, Cope M, Delpy DT, et al. Quantification of cerebral blood volume blood volume and oxygenation in the neonatal brain. NeuroImage
in newborn infants by near infrared spectroscopy. J Appl Physiol 2006;31:1426–33.
1990;68:1086–91. 101. Arridge SR, Schweiger M. Image reconstruction in optical tomography. Phil
75. Elwell CE, Cope M, Edwards AD, et al. Quantification of adult cerebral Trans R Soc Lond B 1997;352:717–26.
haemodynamics by near-infrared spectroscopy. App Physiol 102. Hebden JC. Advances in optical imaging of the newborn infant brain.
1994;77:2753–60. Psychophysiology 2003;40(4):501–10.
76. Bucher HU, Edwards AD, Lipp AE, et al. Comparison between near infrared 103. Benaron DA, Hintz SR, Villringer A, et al. Noninvasive functional
spectroscopy and 133xenon clearance for estimation of cerebral blood flow imaging of human brain using light. J Cereb Blood Flow Metab 2000;20:
in critically ill preterm infants. Pediatr Res 1992;33:56–60. 469–77.
77. Wolf M, von Siebenthal K, Keel M, et al. Comparison of three methods to 104. Danen RM, Wang Y, Li XD, et al. Regional imager for low resolution
measure absolute cerebral hemoglobin concentration in neonates by functional imaging of the brain with diffusing near-infrared light.
near-infrared spectrophotometry. J Biomed Opt 2002;7:221–7. Photochem Photobiol 1985;67:33–40.
78. Landsman MLJ, Kwant G, Mook GA, et al. Light-absorbing properties, 105. Franceschini MA, Toronov V, Filiaci ME, et al. On line optical imaging of
stability, and spectral stabilization of indocyanine green. J Appl Physiol the human brain with 160ms temporal resolution. Opt Express
1976;40:575–83. 2000;6:49–57.
Section V—Cerebral Blood Flow 335.e3

106. Hebden JC, Gibson A, Austin T, et al. Imaging changes in blood volume 112. Hoshi Y, Tamura M. Dynamic multichannel near-infrared optical imaging
and oxygenation in the newborn infant brain using three-dimensional of human brain activity. J App Physiol 1993;75(4):1842–6.
optical tomography. Phys Med Biol 2004;49:1117–30. 113. Kato T, Kamei A, Takashima S, et al. Human visual cortical function during
107. Hintz SR, Cheong WF, van Houten JP, et al. Bedside imaging of intracranial photic stimulation monitoring by means of near-infrared spectroscopy.
hemorrhage in the neonate using light: comparison with ultrasound, J Cereb Blood Flow Metab 1993;13(3):516–20.
computed tomography, and magnetic resonance imaging. Pediat Res 114. Mehagnoul-Schipper DJ, van der Kallen BF, Colier WN, et al.
1999;45(1):54–9. Simultaneous measurements of cerebral oxygenation changes during brain
108. Kusaka T, Kawada K, Okubo K, et al. Noninvasive optical imaging in the activation by near-infrared spectroscopy and functional magnetic resonance
visual cortex in young infants. Hum Brain Mapp 2004;22:122–32. imaging in healthy young and elderly subjects. Hum Brain Mapp 2002;16:
109. Schroeter ML, Bucheler MM, Muller K, et al. Towards a standard analysis 14–23.
for functional near-infrared imaging. Neuroimage 2004;21:283–90. 115. Strangman G, Culver JP, Thompson JH, et al. A quantitative comparison of
110. Chance B, Zhuang Z, UnAh C, et al. Cognition-activated low-frequency simultaneous BOLD fMRI and NIRS recordings during functional brain
modulation of light absorption in human brain. Proc Natl Acad Sci USA activation. Neuroimage 2002;17:719–31.
1993;90:3370–4. 116. Keller E, Froehlich J, Muroi C, et al. Neuromonitoring in intensive care: a
111. Villringer A, Planck J, Hock C, et al. Near infrared spectroscopy (NIRS): new brain tissue probe for combined monitoring of intracranial pressure
a new tool to study hemodynamic changes during activation of brain (ICP) cerebral blood flow (CBF) and oxygenation. Acta Neurochir Suppl
function in human adults. Neurosci Lett 1993;154:101–4. 2011;110(Pt 2):217–20.
VI
Chapter
34  

Intracranial Pressure
Randall M. Chesnut

This chapter is dedicated to professor Douglas Miller for his sinus (Starling resistor),9 pressure-related venous compres-
personal influence on my understanding of brain injury and sion, or thrombosis. The volume of the combined arteriove-
his genius in originating the concept of targeted therapy, the nous system can increase actively, due to increased flow
implications of which most of us are only now coming to recruitment from elevated metabolic activity, or passively,
understand. such as from hypercapnia or blood pressure elevation in the
face of abnormal pressure autoregulation.10 CSF production is
fixed, so volume increases within this compartment generally
Introduction increase as a result of outflow obstruction or abnormal
An intracranial pressure (ICP) monitor is considered the gold resorption.
standard intracranial monitor. In particular, the most recent Increases in the volume of one compartment (or the addi-
guidelines for the management of severe traumatic brain tion of a compartment such as a hematoma) may be buffered
injury (TBI) contain level 2 recommendations (moderate by compensatory decreases in the volume of other compart-
degree of clinical certainty) for the use of an ICP monitor in ments. When the increase is sufficiently slow, these compensa-
comatose TBI patients who have an abnormal admission com- tory changes can maintain a stable ICP up to the point of their
puted tomography (CT) scan.1 Whether to use an ICP monitor exhaustion (Fig. 34.2). At that point, the ICP rises rapidly with
in comatose patients who have a normal CT scan after TBI or any further increase in compartmental volume. If the rate of
in other disorders is less certain (level 3; clinical certainty not initial volume increase is rapid, the compensatory mecha-
established), although neurosurgeons often will consider an nisms are likely to fail earlier.
ICP monitor in conditions as diverse as subarachnoid hemor- One consequence of these compensatory changes is that the
rhage (SAH), meningitis, and liver failure.2,3 In addition, CT brain compliance changes as its volume buffering capacity is
scan findings may not always predict whether ICP is increased.4 exhausted. This means that for a given volume challenge, the
Consequently, there is variation across centers on how fre- resultant ICP elevation will increase as the compliance
quently an ICP monitor is used. There are very strong physi- decreases. Therefore even when volume buffering keeps the
ologic arguments for ICP-based management for many acute ICP relatively stable, knowing the compliance of the brain is
neurologic conditions that require admission to a neurocriti- a very useful index of the status of that buffering system (vide
cal care unit (NCCU) and a well-described association between infra).
elevated ICP and mortality. Having the ability to continuously
monitor ICP avoids “empirical” ICP treatment or blind pro-
phylactic treatment. This is important because many treat- The Monro-Kellie Doctrine and
ments for ICP, although effective in lowering ICP, may have
other deleterious side effects.
Intracranial Pressure Management
The Monro-Kellie doctrine helps explain ICP and is consis-
tent with the framework that supports current therapeutic
The Monro-Kellie Doctrine interventions, all of which aim to lower or prevent an
The basic concept of ICP is couched in the Monro-Kellie increase in the volume of one or more of the intracranial
doctrine.5-7 This doctrine is founded on the fixed volume compartments.
of the skull, such that the interior pressure is a function
of the “volumes” of the individual physiologic intracranial
compartments—brain, blood, and cerebrospinal fluid (CSF)— Mass Lesion
plus mass lesions, when present (Fig. 34.1). Following trauma, The simplest compartmental correction is for mass lesions.
brain volume increases due to edema, either vasogenic or Removal of a mass (e.g., a hematoma) is the oldest approach
cytotoxic, the latter being predominant.8 The vascular com- to control intracranial hypertension and acts directly to
partment is primarily composed of venous blood, the volume improve the intracranial volume balance. This is one of the
of which can increase due to outflow obstruction. Such few areas in TBI or other acute neurologic disorders where a
obstruction can be extracranial (e.g., increased intrathoracic pathophysiologic entity can be dealt with directly and defini-
pressure or compression of the jugular outflow system) or tively and reinforces the value of a CT scan to avoid medical
internal, from kinking of vessels draining into the sagittal treatment of a surgical lesion.
338 © Copyright 2013 Elsevier Inc. All rights reserved.
Section VI—Intracranial Monitoring 339

MONRO KELLIE DOCTRINE - NORMAL STATE MONRO KELLIE DOCTRINE - COMPENSATED STATE

Pressure
Volume

Blood Blood
Blood compliance Blood compliance
CSF CSF
CSF compliance CSF compliance
Mass Mass
Brain Brain

A
Fig. 34.1  The Monro-Kellie doctrine. The fixed volume of the skull
contains brain, blood, and cerebrospinal fluid (CSF) compartments, plus
mass lesions when present. The combined volumes of these compart­
ments determine the intracranial pressure (ICP). The blood and CSF
MONRO KELLIE DOCTRINE - STRESSED COMPENSATION
compartments can shift some of their volume outside of the skull 
through natural means; this allows some degree of compensation for
increases in other compartments.

Pressure
Volume

Blood
The Cerebrospinal Fluid Compartment Blood compliance
CSF
The natural intracranial CSF buffering system attempts to CSF compliance
shunt a larger volume into the spinal subarachnoid space (i.e., Mass
it is subject to outflow tract patency). If interventricular pas- Brain
sages or egress from the fourth ventricle is compromised (e.g.,
a posterior fossa hematoma), the system will fail. An external
ventricular drain can be inserted in such instances, or when
caregivers want to facilitate CSF drainage by controlling it.
This can be useful if there is a sufficient CSF volume and if
the ventricular system does not collapse from drainage. An B
open ventriculostomy, however, renders ICP measurement
unreliable, so these drains should be clamped before recording
ICP.
MONRO KELLIE DOCTRINE - EXAUSTED COMPENSATION
Cerebral Blood Volume
Management of the cerebral blood volume (CBV) can address
Pressure

the venous or arterial compartments. Optimized drainage of Volume

intracranial venous blood by (1) elevating the head of the bed, Blood
(2) avoiding compression of the venous blood in the neck (e.g., Blood compliance
CSF
by a tight cervical orthosis or a tracheostomy tie), or (3) reduc- CSF compliance
ing intrathoracic pressures through careful ventilator manage- Mass
ment can help prevent elevation of the venous compartment Brain
or interference with its volume buffering capacity. CBV can
be decreased through vasoconstriction. Commonly this is
achieved through hyperventilation (HV) to induce hypocap-
nea. Because HV influences the resistance vessels that are on
the inflow side, it also decreases cerebral blood flow (CBF) and
so may cause ischemia.11,12 Prophylactic HV therefore has a C
limited if any role to play in ICP management because of this
Fig. 34.2  Volume compensation. An increase in the volume of one or
potential toxicity.12 If used, for example, when hyperemia con-
more intracranial compartments or the addition of a mass lesion initially
tributes to increased ICP, HV should be used carefully and an is compensated by shifting cerebrospinal fluid (CSF) into the spinal
additional monitor to look for ischemia should be considered subarachnoid space and venous blood out of the intracranial space (A).
if it is used aggressively. HV-induced vasoconstriction is a There is little effect on intracranial pressure (ICP). As volumes increase,
relatively transient effect, and is thought to last less than the ability to compensate decreases and ICP begins to rise (B). When
24 hours, with a risk of rebound vasodilation on discontinu- compensation is exhausted, any further volume challenge results in large
ation.13 Normobaric hyperoxia or hyperbaric oxygen therapy increases in ICP (C).
340 Section VI—Intracranial Monitoring

also may induce vasoconstriction and so alter ICP14-16 with underlying pathology. Therapeutic hypothermia, when deliv-
fewer deleterious effects on cerebral metabolism than HV. ered early and not as a treatment for intracranial hyperten-
However, the role of hyperbaric oxygen is still to be elucidated. sion, was an attempt to address some of the pathophysiologic
Finally, mannitol may induce transient vasoconstriction abnormalities initiated at the time of trauma, but unfortu-
through altered blood viscosity, but the effects on CBF and nately this treatment does not improve outcome from adult
volume and hence on ICP depend on autoregulation.17,18 or pediatric TBI.29,30 By contrast, induced hypothermia is
useful in cardiac arrest31; however, in this condition it is not
used as specific ICP treatment.
The Brain The importance of this skepticism is to remind caregivers
The natural volume buffering capacity of brain tissue is that because treatment modalities are not targeted at specific
limited and has a very long time constant. In general, the brain molecular or biochemical processes that must be reversed,
volume increases after injury, due primarily to edema. Both they need to avoid harming the patient in the choice of or
cytotoxic and vasogenic mechanisms occur in post-traumatic persistence with individual treatments. Obviously, ICP and
edema. However, several lines of evidence support a cellular cerebral perfusion can be manipulated in many ways. When a
origin that involves aquaporin-mediated transmembrane given approach appears ineffective or begins to exert systemic
water passage.8,19,20 The role of vasogenic mechanisms remains toxicity, the therapeutic approach or tactics should immedi-
unclear. This is important because current osmotic agents ately be changed. This presumes that an ICP monitor is in
(mannitol, hypertonic saline) work through the extracellular place; in the modern NCCU ICP treatment should be guided
compartment. The only medical treatment to decrease extra- by such technology and not given empirically. There is no one
cellular edema in the brain compartment is hyperosmolar best way to treat increased ICP and ideally, a pragmatic
therapy (osmotherapy). Alternatively, increased brain volume approach, guided by multimodality monitoring and coupled
can be reduced through the surgical removal of tissue, either with a view toward balanced critical care of the entire patient,
contused or pulped parenchyma or normal, “silent” tissue.21 must form the guiding principles.
The surgical removal of contusions remains controversial and
has largely been abandoned.
The Role of ICP Monitoring and
Decompressive Craniectomy Treatment in TBI Management
The final Monro-Kellie–related approach is to invalidate the The use of an ICP monitor and ICP treatment is best studied
premise that underlies the doctrine through decompressive in TBI and the techniques and expectations used to treat ele-
craniectomy (DC); this renders the intracranial space no vated ICP in other conditions such as subarachnoid hemor-
longer a constant. A generous skull opening and a large durot- rhage, intracerebral hemorrhage, ischemic stroke, encephalitis,
omy or nonconstraining duraplasty compensates for increased and metabolic disorders among others are based in large part
volumes of one or more intracranial compartments by increas- on the experience from TBI.32,33 The focus of this chapter
ing the volume of containment and adding a compliance therefore is on ICP and TBI. ICP data can be used to predict
buffer (because the bone-free area is not rigid). DC effectively outcome and evolution of intracranial pathology, calculate
reduces increased ICP in conditions such as stroke, TBI, and and manage cerebral perfusion pressure (CPP), direct man-
SAH.22-24 However, the exact timing of DC and its effect on agement strategies, and limit the use of potentially deleterious
outcome particularly in TBI remains debated.25-28 therapies. An ICP monitor is considered be a conditio sine qua
non of severe TBI care. Controversy about ICP monitoring,
however, is reflected in the variability of its clinical use.34-38 In
Implications of Monro-Kellie–Driven addition are various philosophies about how to integrate ICP
data into treatment—ICP-based therapy, CPP-based therapy,
Therapy Lund therapy, and optimized HV among others.39-41 Insertion
The Monro-Kellie doctrine is a useful framework to organize of an ICP monitor is the first step from “observational,” “clini-
and understand medical and surgical approaches to the cal,” or “empirical” management of severe TBI (or pathologies
injured brain. However, it is focused on the clinical manage- in which increased ICP occurs) to monitor-based, physiologic
ment of ICP, and the invoked therapeutic modalities are care. Despite experience dating back to the 1960s, considerable
phenomenologic and based on their perceived ability to lower controversy surrounds ICP-monitor–based care in TBI.
ICP. None of these treatments, however, specifically addresses
the pathophysiology of TBI and other acute brain injuries.
Osmotherapy probably works to decrease the water volume of Is High Intracranial Pressure
the least expanded compartment. Reduction of brain blood
volume is phenomenologic because primary venous or arte-
Associated with Poor Outcome?
rial hypervolemia usually is not an underlying pathologic Since the seminal work of Lundberg and Guillaume and Janny
event in adults, and induced vasoconstriction may decrease in the 1950s and 1960s, a large body of observational data
oxygen delivery and be harmful. CSF drainage can address a demonstrates that ICP elevation is associated with poor
specific pathophysiologic abnormality if outflow obstruction outcome and that ICP cannot be estimated accurately or reli-
occurs but in the absence of hydrocephalus remains phenom- ably on clinical grounds.39,42-50 The association between intra-
enologic. Management such as sedation, analgesia, neuromus- cranial hypertension and poor outcome appears to be
cular blockade, and second-tier interventions such a high-dose independent from other confounding variables45,46 and pro-
barbiturates and DC also are focused on ICP and not the portional to the amount of time that the ICP is greater than
Section VI—Intracranial Monitoring 341

20 to 25 mm Hg.45,51 In SAH, elevated ICP is associated with emphasize two important issues about ICP monitors: (1) con-
poor outcome but on multivariate analysis is not an indepen- tinued equipoise about the role of ICP monitors in TBI man-
dent factor.32 Outcome also may be poor because treatment agement, and (2) ICP monitoring exists only in the context of
for high ICP can be harmful in some circumstances. For an associated treatment philosophy, which confounds the
example, mannitol administration can produce renal damage analyses of the value of ICP monitoring versus the efficacy of
or hypotension52 and HV may induce cerebral ischemia,12 a specific treatment strategy. In the two studies that reported
whereas barbiturates and hypothermia both increase the risk no benefit to an ICP monitor,34,35 all the patients were treated
of infection and hypotension.29,30,53-55 Therapies to augment for suspected intracranial hypertension, whether monitored
CPP are associated with increased systemic complications,56 or not. Because insertion of an intraparenchymal ICP monitor
and even the most basic treatments, sedation and mechanical is of very low risk, it is unlikely that display of the ICP will
ventilatory control, may increase ICU length of stay and expo- adversely influence outcome or improve recovery per se.
sure to iatrogenic injuries such as ventilator-associated pneu- Implicit in any discussion about the role of an ICP monitor
monia or skin breakdown.57,58 In particular when the proper in TBI management is that the interventions generated by the
ICP threshold is unclear (vide infra) or there is mild ICP ICP data improve outcome. Embedded within such a monitor-
elevation without a perfusion deficit, a patient may be at more driven therapy question are several issues, including (1) the
risk of a treatment complication than of harm from ICP. In validity of the treatment threshold and whether that threshold
these circumstances other monitors or information may help applies to all patients, (2) whether a given treatment regimen
guide ICP management. is appropriate in all monitored patients, (3) the choice of
therapy, (4) the efficacy and risk-to-benefit ratio of various
treatments alone or in combination, and (5) an understanding
Does Treating Intracranial of the interaction between therapy and management interven-
tions made in a given patient for other reasons. Consequently,
Hypertension Improve Outcome? although use of an ICP monitor may make sound physiologic
Whether treatment of intracranial hypertension is associated sense, the ability to demonstrate that its use makes a difference
with improved outcome is unclear. Certainly in individual to outcome may be confounded by several reasons: (1) wrong
patients an effect can be seen, but there is no Class I evidence treatment threshold, (2) wrong individual therapeutic agent,
that demonstrates ICP management is associated with better (3) wrong therapeutic approach, (4) toxic effects of ICP treat-
outcome.59 Such a trial may be difficult to perform because ment, (5) misinterpretation of ICP data, (6) wrong patient
ICP monitoring and management have become standard of population, (7) lack of utility of ICP monitoring, or (8) a
practice. Early retrospective observational data not adjusted combination of all. An example of how such confounding may
for other variables suggest that ICP control may be beneficial. affect whether a monitor is useful are the RCTs that demon-
For example, Marshall60 observed better outcome in patients strated that routine use of right heart catheterization in the
whose ICP was less than 15 mm Hg than those whose ICP was ICU is not associated with better outcome64-66 (i.e., use of a
greater than 40 mm Hg for more than 15 minutes. Saul and monitor does always mean better outcome unless effective
Ducker61 also observed that patients with severe TBI whose management strategies exist).
ICP was treated when it was greater than 15 mm Hg did better
than those treated when ICP was greater than 25 mm Hg.
Eisenberg et al.53 conducted a randomized clinical trial (RCT)
An Intracranial Pressure Threshold
that examined high-dose barbiturate therapy for refractory Among several areas on how best to interpret and treat ICP is
intracranial hypertension in severe TBI; ICP control rather the threshold value used for treatment and in particular
than clinical outcome was the primary objective. An associa- whether a single threshold applies to all patients or in one
tion between ICP control and improved outcome for all patient at all times. Too high a value may allow unrecognized
patient groups (barbiturate group, control group, and control neural injury, whereas too low a value may result in overtreat-
group that crossed over to barbiturate treatment) was observed. ment and iatrogenic complications. The most frequently used
Whether these results apply to patients with mild elevated ICP threshold of 20 mm Hg was adopted from the accepted upper
or ICP that is responsive to management is uncertain. In SAH, limit of normal for lumbar CSF pressure measurements and
Heuer et al.32 in a retrospective analysis of 433 patients from early work by Lundberg.43 Schreiber et al. used multiple
observed that among 21 patients whose raised ICP did not regression modeling and found that ICP greater than or equal
respond to mannitol, all had a poor outcome. By contrast, to 15 mm Hg was associated independently with mortality
among 145 patients whose elevated ICP responded to man- after severe TBI. However, they did not analyze treatment
nitol, 66.9% had a favorable outcome. Most recently Stein thresholds per se.67 Marmarou et al.45 analyzed 428 monitored
et al.62 performed an extensive literature review of TBI trials patients from the Traumatic Coma Data Bank and found that
and case series reported after 1970 and divided patients into the proportion of total monitoring time that the ICP was
those with and without ICP monitoring and intensive therapy. greater than 20 mm Hg was an independent predictor of
Meta-analysis showed that mortality was 12% lower and favor- 6-month outcome. However, ICP treatment thresholds of
able outcome 6% greater in the aggressively treated group. 20 or 25 mm Hg were used, which confounds separation
There also are several reports that use of an ICP monitor is of the effects of ICP of greater than or equal to 20 mm Hg
not associated with improved outcome after TBI or that “ICP from treatment toxicity. Nevertheless, an ICP threshold of
monitoring in accordance with the guidelines for the manage- more than 20 mm Hg is now central to ICP management
ment of severe traumatic brain injury63 is associated with based largely on post hoc analysis of studies in which 20 mm
worsening of survival in TBI patients.”34,35 These studies have Hg was the treatment trigger (i.e., there are no controlled
several flaws that limit meaningful conclusions but they studies on of the “best” ICP treatment threshold). This is
342 Section VI—Intracranial Monitoring

important for several reasons. First, observational data for gradient) empirically (i.e., the neurologic examination) when
which other monitors are used—jugular bulb catheters, brain brain herniation will occur although it is clear once it has
oxygen, or microdialysis—suggest that brain metabolism may occurred.76 It is preferable to avoid herniation than to treat it
be abnormal even when ICP and CPP are normal.68,69 Second, post hoc.77,78 Third, information from an ICP monitor may
an ICP threshold of 20 mm Hg was established during a time provide useful acuity information and so guide patient care
period when systemic hypertension was thought to be a risk and perhaps ICU resources. For example, a patient with a
factor for intracranial hypertension, patients were routinely worrisome-appearing CT scan79 who does not have intracra-
kept “dry,” and CPP was not a monitored variable. Conse- nial hypertension may not require the same degree of treat-
quently, many TBI patients in the intensive care unit (ICU) ment as a patient with a similar scan but elevated ICP.80
had relatively low blood pressures and the only real limit was Similarly, a patient with elevated ICP that is refractory to
to avoid systolic pressures less than 90 mm Hg. This represents escalating management becomes an early candidate for
a mean arterial pressure of about 70 mm Hg. Because normal “second tier” treatments or if very high, even withdrawal of
autoregulation becomes impaired at approximately 50 mm care. Fourth, ICP trends can be an early warning of mass lesion
Hg, an ICP value of 20 mm Hg therefore may represent bor- expansion, the appearance of new lesions, or evolution of
derline ischemia; that is, the threshold value may be a edema, ischemia, or hydrocephalus and allow these conditions
by-product of the treatment regimens of that period. At the to be effectively managed before clinical findings change or
time of this publication, the management focus is on not only they are detected on periodic imaging studies.81 Finally, because
ICP but also CPP and in some centers brain oxygen or other ICP values have prognostic value it can guide management
parameters. This combined with sedatives that lower the cere- and prognosis discussions with the family.
bral metabolic rate mean that the flow-to-perfusion conse-
quences of an ICP that is 20 mm Hg or just elevated may be
different today compared with the time period when this Intracranial Pressure
threshold was decided upon.
Perhaps more important than a single ICP threshold may
Monitoring Technology
be a trend over time, ICP waveform analysis, or whether the Technology evaluation is not an ideal topic for evidence-based
ICP value is associated with other detrimental effects. The medicine. However, the various ICP monitor systems are well
concept that the ICP threshold may vary among patients and described in the ICP monitoring technology section of the
within patients over time (i.e., targeted) is not new. In the guidelines for the management of severe traumatic brain
1970s and 1980s, professor Douglas Miller often used an ICP injury.75 This report concluded that at the time of its writing,
threshold of 25 to 30 mm Hg in TBI patients with acceptable three technologies are sufficiently accurate to be interchange-
indicators of cerebral oxygenation and compliance. This able in use: (1) a ventricular catheter connected to an external
concept of permissive intracranial hypertension is practiced strain gauge; (2) catheter tip strain gauge devices, or (3) cath-
at several institutions including the author’s own but is best eter tip fiber-optic technology. Each of these systems can be
guided by information from multimodality monitoring. For placed into the ventricle. Catheter tip strain gauges or fiber-
example, a brain oxygen monitor may complement the infor- optics also can be placed into the parenchyma. The most
mation from an ICP monitor. Some but not all observational commonly used devices include the Camino or the Ventrix
studies suggest that when both ICP and brain oxygen are (Integra Neurosciences, Plainsboro, NJ), Codman microsen-
treated, the outcome may be better than if just ICP is treated sor (Codman, Raynham, MA), the Spiegelberg ICP sensor and
after TBI.70 This effect may occur in part because the deleteri- compliance device (Spiegelberg KG, Hamburg, Germany), and
ous consequences of ICP overtreatment may be avoided. Con- the Raumedic ICP sensor and multiparameter probe (Rau-
sistent with this Chambers et al. calculated receiver-operating medic AG, Munchberg, Germany). The pneumatic Spiegel-
characteristic curves for ICP and CPP in 227 patients71 and berg ICP monitor unlike the other parenchymal monitors also
found that the sensitivity of ICP for outcome increased at an allows in vivo calibration and intracranial compliance moni-
ICP of 10 mm Hg, but by 30 mm Hg it had only reached 61%, toring. The evidence report also concluded that fluid-coupled
with a cutoff value of 35 mm Hg that varied with the CT or pneumatic devices placed in the subarachnoid, subdural,
classification. and epidural compartments are less accurate.75 Noninvasive
technologies for ICP monitoring based on a variety of tech-
niques such as ultrasound, transcranial Doppler, acoustic
The Value of Intracranial properties of cranial bones, tympanic membrane displace-
ment, heart rate variability and cardiac coupling, ophthalmo-
Pressure Monitoring dynamometry, and measurement of optic nerve diameter are
Although understanding of ICP is incomplete and treatment under investigation. Although more than 30 noninvasive ICP
approaches still need to be refined, there are many other poten- monitors have been patented since the 1990s, most are still too
tial benefits from ICP monitoring. First, without an ICP cumbersome or not sufficiently accurate to be used in clinical
monitor, CPP is not known. Even transient episodes of isch- practice.
emia can be devastating to the traumatized brain,72-74 making The ventricular catheter connected to a fluid-coupled exter-
it critical to accurately and continuously monitor CPP.75 nal strain gauge is considered the gold standard ICP monitor
Because insertion of intraparenchymal ICP monitors is safe, in part because it can be used also to treat ICP through CSF
the ability to monitor CPP per se is a supportable argument drainage. Control of elevated ICP is observed in about 50%
for widespread ICP monitoring. Second, cerebral herniation of patients in whom an external ventricular drain (EVD) is
is a pressure issue and an ICP monitor may allow early detec- inserted after other initial measures fail.82 Traditional ventricu-
tion. It is impossible to determine the pressure (pressure lar catheter external transducers only permit intermittent ICP
Section VI—Intracranial Monitoring 343

monitoring when the drain is closed (i.e., not draining), duration of monitoring, presence of intraventricular hemor-
although some catheters have internal transducers that permit rhage or SAH, open skull fracture (with or without CSF leak),
CSF drainage at the same time ICP is monitored. An EVD may trauma, flushing of the catheter, and CSF leakage at the inser-
miss episodes of increased ICP when it is draining.83 This gold tion site.93,96-100 Because most data are from case series, it
standard status, however, may take precedence because it was remains uncertain how best to prevent or manage infections.
the first monitoring system available. Furthermore, there are However, strategies such as use of closed drainage systems, a
no clinical outcome studies that show that one monitoring long subcutaneous tunnel, reduced duration of ventricular
technology is superior to others. Because a ventricular catheter cannulation (i.e., prompt removal when not needed), avoiding
is placed into the ventricle, it is “logically” felt to optimally catheter irrigation or flushing or if needed done using aseptic
represent the ICP. However, even though gradients have techniques, and avoiding CSF leakage from any site are recom-
been demonstrated between parenchymal and ventricular mended. There does not appear to be a role for continuous
systems,84-87 none of these has ever been found to be of clinical antibiotic prophylaxis.75 Silver- or antibiotic-impregnated
significance. Parenchymal devices are easier to place, particu- catheters may decrease the incidence of catheter-related CSF
larly when altered ventricular anatomy may limit ventricular infections, although the exact role for these catheters is still
catheter placement. However, intraparenchymal fiber-optic debated.101-103 Because CSF sampling may predispose to con-
and electronic strain gauge systems are more expensive and tamination or infection, sampling should be indicated by spe-
cannot be recalibrated once in situ. Consequently, outside the cific clinical criteria rather than be a routine practice. Even a
ability of ventricular-catheter monitors to drain CSF as a reduction of CSF sampling from daily to every 3 days can
potentially therapeutic maneuver, the choice of an ICP monitor reduce the rate of ventriculitis from 10% to 3%.94 Use of stan-
is probably best decided based on factors such as accuracy, dard management protocols particularly with a bundled
reliability, complication rates, ease of insertion, and cost. approach also can help reduce the infection rate.104,105 There
does not appear to be a role for routine replacement, because
it appears that the risk of replacement outweighs any potential
Potential Complications of benefit.98,100,106-108 The use of parenchymal ICP monitors
should be considered when there is systemic infection or an
Intracranial Pressure Monitors open skull fracture because there are no reports in the litera-
Ventricular catheters and ICP bolts traditionally have been ture of infections associated with these devices in children or
placed by neurosurgeons. However, neurointensivists now are adults.109-111 There is, however, one report of an abscess that
inserting these monitors on a more frequent basis.88 The com- developed several weeks after an ICP monitor was removed
plications rate appears to depend more on the technique used from a 35-week-old child who required both prolonged ICP
and the device (ventricular catheter vs. parenchymal monitor) monitoring and steroids.112
than on who places the monitor once he or she has been
adequately trained.89,90
Hemorrhage
The precise incidence of hemorrhagic complications follow-
Infection ing ICP monitor insertion depends on what device is used,
The definition of infection of an ICP monitor is somewhat insertion technique, and how hemorrhages were identified.
controversial. It is easier to detect bacterial involvement of Ideally, imaging studies should be obtained before and after
a ventricular catheter rather than a parenchymal monitor ICP monitor insertion, but few studies include such imaging.
because CSF sampling can be periodically performed. In the Retrospective case series suggest that the risk of intracranial
absence of signs of ventriculitis, positive cultures may be hemorrhage after placement of an EVD in adults is between
better termed colonization.63 When this definition (coloniza- 2% and 10%.113-116 The risk may be greater in children
tion rather than infection) is used, positive cultures are (17.6%).117 Most hemorrhages are less than 15 mL in volume
observed in 8% of ventriculostomy CSF cultures and 14% of and therefore “clinically insignificant.” The incidence of clini-
the cultures obtained from the tip of an intraparenchymal cally significant hemorrhages associated with ventriculostomy
device once it is removed. In clinical practice, however, routine placement is about 1%.116 After insertion of an intraparenchy-
surveillance of intraparenchymal monitors is not performed. mal ICP monitor in adults the incidence of intracranial hem-
By contrast CSF sampling is common with ventricular cath- orrhage is estimated to be between zero and 11%85,110,118,119 in
eters. Positive CSF cultures may prompt removal of the device adults. The incidence is similar in children.111,117,118 Gelabert-
and antibiotics that may lengthen hospital stay, expose patients Gonzalez et al. described 1000 consecutive patients who had
to the potential hazards of parenteral antibiotics, and expose intraparenchymal ICP monitors; 922 had postinsertion CT
them to the risk of catheter replacement. Therefore although imaging.110 The overall hemorrhage rate was 2.5%, and 0.66%
both types of monitors may be colonized, the consequences required surgery for an intraparenchymal or epidural hemor-
are markedly different. This must be balanced against the rhage. Two studies have examined intraparenchymal and
value of CSF drainage. intraventricular devices concurrently. The hemorrhage inci-
Iatrogenic catheter-related ventriculitis and meningitis dence is significantly greater for ventriculostomies.114,117
may occur in 5% to 20% of EVDs through direct catheter
contamination at insertion, through retrograde bacterial colo-
nization, or if introduced when the system is accessed or Technical Issues
flushed.91-95 The overall device infection rate is between 6 Ventricular catheter displacement, accidental removal, or
and 8 per 1000 drainage days.95 Risk factors for infection blockage may occur. When blockage occurs either by debris
include the presence of concurrent systemic infections, longer or blood that obliterates holes in the catheter or catheter
344 Section VI—Intracranial Monitoring

displacement into the parenchyma, CSF drainage can generate


a large pressure gradient between the catheter lumen and the Table 34.1  Advantages and Disadvantages
ventricle. In this circumstance ICP is underestimated when it of Ventricular and Parenchymal ICP Monitors
is monitored at the same time as CSF is drained and the CSF Ventriculostomy Intraparenchymal Monitor
waveform becomes flattened. When this occurs the drainage
Advantages Advantages
system should be closed to see if the waveform returns. If not, • Reference standard (by • Ease of placement
the system may need to be gently flushed with 1 to 2 mL of default) • Accurately reproduces the
normal saline. The role of thrombolytic agents to clear blood • Can be rezeroed CSF pulse waveform
clots with intraventricular hemorrhage is unclear. Technical • Cheap without mechanical
complications such as catheter breakage or dislodgement • Therapeutic drainage of damping
CSF
occur in about 4.5% of intraparenchymal devices. Most of
these occur during transport, nursing maneuvers, or patient Disadvantages Disadvantages
• Plugging • Can’t be rezeroed without
activities.110 None appear to influence the patient’s course or • Disconnection removal-potential for drift
outcome. Different parenchymal devices including fiber-optic, • Can’t measure ICP • More prone to technical
strain gauge, and pneumatic technologies are available. Only accurately when open to failure (catheter fracture,
the pneumatic Spiegelberg ICP monitor allows in vivo calibra- drainage pull-out, etc.)
tion. Bench testing shows that parenchymal monitors have • Ventriculitis • Incompatible with MRI
• Brain must be • More expensive
excellent accuracy; however, studies suggest suspicion that penetrated
zero-drift rates can be clinically important in strain gauge • Difficult to place with
monitors.120,121 Drift is very rare in fiber-optic catheters.122 In displaced, compressed,
addition, there are descriptions of disturbed baseline pressures or small ventricles
• Complications of
of the Codman and Raumedic ICP sensors by electrostatic overdrainage
discharges.123 • Requires rezeroing of
the system with any
change in the height of
the head of the bed
Optimal Placement of potential for error
Intraparenchymal Devices • Dampens the waveform
due to the mechanical
Intraparenchymal ICP monitors are placed into the brain properties of the tubing
parenchyma through a small burr hole and a cranial access and if air is present
device or bolt that may allow placement of one to three moni- CSF, Cerebrospinal fluid; ICP, intracranial pressure; MRI, magnetic resonance
tors. Usually these monitors are placed into the nondominant imaging.
frontal lobe when the injury or pathology is diffuse. However,
in focal injury these monitors may be better placed on the side
of the pathology or in pericontusional parenchyma because,
unlike ventricular catheters, intraparenchymal devices have or protocols to correct coagulopathies before an EVD or ICP
the potential to represent local, “compartmental” pressures, monitor is placed. Factor administration, however, may delay
should supratentorial gradients exist. In particular, intra­ the procedure. Studies suggest an international normalized
parenchymal devices placed in the hemisphere contralateral ratio (INR) less than or equal to 1.6 is acceptable to place a
to a mass lesion may underestimate ICP even with brain ventricular catheter following TBI.128,129 Many physicians
herniation.124 regard a platelet count of 100,000 necessary to safely place an
Some but not all clinical studies in TBI with older technol- ICP monitor; however, there is little literature on this topic.
ogy such as subarachnoid bolts (not recommended in the Davis et al. addressed the need to normalize coagulation
TBI evidence report75) often demonstrated interhemispheric parameters before inserting a fiberoptic intraparenchymal
gradients.125-127 Sahuquillo et al. using fiber-optic intraparen- ICP monitor.130 They retrospectively reviewed 157 patients
chymal monitors also found interhemispheric gradients in including laboratory values and postinsertion CT images.
patients with focal lesions that could be greater than 10 mm They included 10 patients with borderline INR values (1.3-
Hg.87 One quarter of such gradients produced a difference in 1.6) and 12 patients with INR values greater than or equal to
calculated CPP of greater than or equal to 5 mm Hg, which the 1.7 at the time of placement. Eleven of their patients had
authors felt had the potential to alter treatment. The authors platelet counts between 50,000 and 100,000 at the time of
therefore concluded that “to optimize ICP monitoring in insertion. Three patients (1.9%) had insertion-related pete-
patients with a mass lesion larger than 25 mL or midline shift, chial hemorrhages without clinical significance. They con-
the measuring device should preferably be implanted in both cluded that intraparenchymal monitor insertion with an INR
the brain parenchyma and the side of the mass” (Table 34.1). less than or equal to 1.6 is safe. They reported that the use of
fresh frozen plasma (FFP) to normalize the INR when less
than 1.6 delayed monitoring and was not necessary. These
Insertion of ICP Monitors data and other studies suggest that delaying ICP monitor
in Patients with Abnormal insertion may not be necessary in patients with mild coagu-
lopathy if an intraparenchymal device is used.122 In patients
Coagulation Studies with fulminant hepatic failure recombinant activated factor
Patients who require an ICP monitor may have abnormal VII (rFVIIa) has been recommended if an ICP monitor is
coagulation parameters. Many centers have routine practices required.131
Section VI—Intracranial Monitoring 345

How to Monitor Intracranial significant when compliance is very poor. The magnitude of
how a volume change influences ICP is illustrated by studies
Pressure and for How Long on the amount of blood volume change necessary to produce
Continuous digital recording is the best method to acquire a 1 mm Hg ICP elevation. Studies on TBI patients suggest that
ICP data because this method can be linked to other monitors such volumes are as small as 0.42 to 0.5 cc, and less with
to calculate derived indexes and should not miss transient decreased compliance.137,138 Hypercapnea can increase the sen-
changes in ICP.132 This is not feasible in all ICUs. Instead sitivity of the ICP to volume challenges.
NCCU staff chart ICP values on a regular basis. End-hour Compensation for a volume change is time dependent.
recording generally correlates well with continuous record- Compensatory responses in the CSF or CBV compartments
ing133; however, to identify episodic events, recording every can partially buffer volume increases (e.g., evolving cerebral
10 minutes may be needed. Even episodes of increased ICP as edema). By contrast, rapid volume challenges are less buffered
brief as 5 minutes can aggravate outcome134; use of alarms may and have a greater influence on ICP. This may include naso-
help identify this. Following TBI patients who develop tracheal suctioning, patient turning, or physiologic insults
increased ICP generally do so in the first week135 and although such as hypercarbia (increased CBV) that all may increase ICP
there are several patterns to the timing of ICP elevation, about when cerebral compliance is poor.
20% first show ICP elevations after 72 hours.136 The ICP To quantify cerebral compliance requires the ability to
monitor should be kept in place while clinically indicated but deliver a reproducible volume challenge and measure the ICP
usually can be removed once the patient starts to follow com- change. The volume-pressure response (VPR) can be mea-
mands. If the patient remains comatose, removal of the sured by rapidly injecting a small (1 cc) volume of fluid into
monitor can be considered if ICP is normal during the first a patient’s ventricle through a ventriculostomy and measuring
72 hours. If ICP is elevated, the monitor may be removed once the immediate ICP response (VPR = ICP change/volume
ICP is normal for at least 24 hours without any specific treat- injected).139,140 This provides a snapshot of the compliance at
ment other than sedation for the ventilator. A follow-up CT that moment but does not describe the shape of the volume-
scan that demonstrates resolving mass effect is useful to guide pressure curve, which may vary among patients and within
this decision, and other monitors such as brain oxygen or an patients over time.141 Assuming a mono-exponential relation-
electroencephalogram (EEG) may help corroborate this. ship, logarithmic conversion of this data provides the pressure-
volume index (PVI), which defines the volume change that
would produce a 10-fold increase in ICP (PVI = V/loge[Po/
Cerebral Compliance and Pm]).142 The PVI describes the shape of the compliance curve
but it does not represent the patient’s position along that
Compensatory Reserve curve. This renders the PVR and PVI of complementary value.
Cerebral compliance is the “stiffness” of the intracranial com- The injection of fluid into the ventricular system, however,
partment and is physically represented by the ICP response to requires “opening” the system. This appears to be associated
a change in intracranial volume. In keeping with the Monro- with an increased risk of infection.143 The ICP also may not
Kellie doctrine, a small increase in intracranial volume when always readily return to baseline after fluid injection and
the compliance is good produces only a small increase in the therefore cause intracranial hypertension. Although the same
ICP (Fig. 34.3). When compliance is compromised, the ICP calculations may be done with fluid withdrawal, this is more
response to the same volume change increases and becomes difficult to perform and is rarely used. Hence measurement of
PVR and PVI (compliance) through a ventriculostomy is not
routine in clinical practice. Simple but very indirect methods
such as pressure on the patient’s abdomen and watching the
Decreasing cerebral compliance
ICP response may provide qualitative estimates about the state
of compliance.
The Spiegelberg ICP sensor and compliance device allows
compliance testing. A ventriculostomy is inserted and at the
catheter tip is a small (0.1 cc) balloon that can be automati-
cally inflated and deflated in a cyclic fashion. Although the
Pressure

∆P
response to such a small volume challenge can be difficult to
detect among the noise inherent in clinical ICP monitoring,
signal-triggered averaging is used to extract that signal and
∆V analyze it to provide compliance data. Experimental and clini-
∆P cal (in hydrocephalus) validation testing of this system against
∆V
VPR/PVI values provides support for its reliability.144,145
However, it is not in widespread clinical use.
Volume The clinical evaluation of cerebral compliance is qualitative,
based on observation of the ICP waveform.146,147 The first three
Fig. 34.3  Cerebral compliance represents the ICP response (ΔP) to
wavelets prominent in the ICP pulse waveform are labeled
a given volume challenge (ΔV). As compliance is progressively
compromised (see Fig. 34.1), the pressure response increases. When P1, P2, and P3 (Fig. 34.4). The first component (P1) is generated
compliance is mildly to moderately stressed, the intracranial pressure by the systolic pulse wave. The second (P2—the “tidal wave”)
(ICP) may remain acceptable despite marginal remaining buffering is a harmonic wave and reflects cerebral compliance. The
capacity. When compliance is exhausted, a small volume challenge may third (P3—the “dicrotic wave”) represents the reflection of the
result in severe intracranial hypertension or acute herniation. dicrotic venous wave that results from closure of the aortic
346 Section VI—Intracranial Monitoring

P1 reserve of the intracranial compartment and cerebrovascular


P2 autoregulatory reserve.155,156 When the RAP index is near zero,
P3
there is a good compensatory reserve, whereas a negative PRx
(–1 to 0) suggests there is preserved autoregulation. By con-
trast a high RAP index (+1), or a high PRx (>0.3) infers there
Fig. 34.4  A normal intracranial pressure (ICP) waveform, in which the
is little compensatory reserve in the intracranial compartment
first peak (P1) is higher than subsequent peaks (P2 and P3).
and therefore even small changes in volume may lead to a
rapid ICP increase. These indices are primarily used in a
research environment and have not yet entered routine prac-
tice, although there is a described association between abnor-
mal RAP, PRx ,and poor outcome.157 Dynamic autoregulation
also may be assessed by analysis of the ICP response to induced
or physiologic challenges.80,155 This may help guide choice of
CPP thresholds.155 Finally, insight into the brain’s compensa-
tory reserve may be estimated from the Therapeutic Intensity
Level (TIL) score. This is a quantitative measure of what man-
agement is needed to manage ICP.158 A greater TIL implies that
multiple therapies or more complex therapies are required to
control ICP.
A

Conclusion
Elevated ICP is common after most forms of acute brain
injury, and its prevention and management are central to the
care of many patients admitted to the NCCU. There is well-
described association between significant ICP elevation and
poor outcome, and its avoidance or rapid treatment appears
to be associated with reduced morbidity and mortality when
part of protocol-driven therapy. There is, however, ongoing
debate about the precise treatment threshold, the optimal
B
approach to lower ICP, and whether or how mild elevation in
Fig. 34.5  Intracranial pressure (ICP) waveform response to ICP should be managed, in both the adult and pediatric pop-
treatment. ICP recording A represents abnormal compliance, with P2 ulations.159 Other parameters such as CPP may be every bit
and P3 being higher than P1. Treatment with osmotherapy and improving as important as ICP, and the interactions between these
the blood pressure resulted in a decrease of the ICP accompanied with variables and their management protocols require further
improvement in the waveform such that P1 is again greater than P2 and
clarification. In addition with the advent of multimodality
P3 (recording B).
monitoring, it is apparent that ICP values should be inter-
preted carefully and with other clinical and radiologic param-
valve. Other, smaller wavelets may follow these three. Under eters. However, because ICP plays a fundamental role in
normal conditions (i.e., good compliance) P1 is the highest almost all aspects of TBI management, and in the care of
component of the pulse waveform (Fig. 34.5, A). As compli- most patients admitted to the NCCU, knowing its precise
ance worsens, often with intracranial hypertension, the ampli- value is integral to direct therapy, allocate resources, estimate
tude of P2 may increase until it equals or exceeds P1 (see Fig. prognosis, and prevent (rather than just treat) secondary
34.5, B), and comes to resemble the arterial pulse waveform. brain insults. Because there are many safe and effective ICP
These changes may be constant or may vary over time (i.e., monitoring technologies, there is little if any role for empiri-
with the respiratory cycle). The elevation of P2 over P1 is a cal ICP management. Furthermore, accurate monitoring of
sensitive (99%) but not specific (1%-17%) predictor of subse- the physiologic state and the response to treatment can help
quent ICP increases associated with nursing maneuvers.148 optimize treatment protocols.
Patients with abnormal waveforms tend to have worse out-
comes. Waveform indices such as high-frequency centroid or
waveform transmission are associated with mortality. However, References
quantitative ICP waveform analysis has had limited success in 1. Bratton SL, Chesnut RM, Ghajar J, et al. Guidelines for the management of
clinical practice149-152 because the indices do not appear to be severe traumatic brain injury. VI. Indications for intracranial pressure
independent of other parameters. Exactly how waveform anal- monitoring. J Neurotrauma 2007;24(Suppl. 1):S37–44.
2. Odetola FO, Clark SJ, Lamarand KE, et al. Intracranial pressure monitoring
ysis can help guide patient management or help predict intra- in childhood meningitis with coma: a national survey of neurosurgeons in
cranial hypertension is still being elucidated.153,154 the United States. Pediatr Crit Care Med 2011;12(6):e350–6.
Advances in computerized monitoring and data processing 3. Kamat P, Kunde S, Vos M, et al. Invasive intracranial pressure monitoring is
now permit online, real-time analysis of the interdependence a useful adjunct in the management of severe hepatic encephalopathy
between the regulatory processes of ICP. This has led to the associated with pediatric acute liver failure. Pediatr Crit Care Med 2011.
Epub ahead of print.
development of derived indices of cerebrovascular pressure 4. Kouvarellis AJ, Rohlwink UK, Sood V, et al. The relationship between basal
reactivity (PRx) and cerebrospinal compensatory reserve cisterns on CT and time-linked intracranial pressure in paediatric head
(RAP). These indices provide insight into the compensatory injury. Childs Nerv Syst 2011;27(7):1139–44.
Section VI—Intracranial Monitoring 347

5. Kellie G. An account of the appearances observed in the dissection of two 28. Hutchinson PJ, Timofeev I, Kolias AG, et al. Decompressive craniectomy for
of the three individuals presumed to have perished in the storm of the 3rd, traumatic brain injury: the jury is still out. Br J Neurosurg 2011;25(3):441–2.
and whose bodies were discovered in the vicinity of Leith on the morning of 29. Clifton GL, Miller ER, Choi SC, et al. Lack of effect of induction of
the 4th November 1821 with some reflections on the pathology of the brain. hypothermia after acute brain injury. N Engl J Med 2001;344(8):556–63.
Transmissions of Medical and Chirurgical Science, Edinburgh 1824;1:84–169. 30. Hutchison JS, Ward RE, Lacroix J, et al. Hypothermia therapy after traumatic
6. Monro A. Observations on the structure and function of the nervous system. brain injury in children. N Engl J Med 2008;358(23):2447–56.
Edinburgh: Creech and Johnson; 1783. 31. Bernard SA, Gray TW, Buist MD, et al. Treatment of comatose survivors of
7. Cushing H. The third circulation in studies in intracranial physiology and out-of-hospital cardiac arrest with induced hypothermia. N Engl J Med
surgery. London: Oxford University Press; 1926. 2002;346:557–63.
8. Marmarou A. A review of progress in understanding the pathophysiology 32. Heuer G, Smith MJ, Elliott JP, et al. The relationship between intracranial
and treatment of brain edema. Neurosurg Focus 2007;22(5):E1. pressure and other clinical variables in patients with aneurysmal
9. Luce JM, Huseby JS, Kirk W, et al. Starling resistor regulates cerebral venous subarachnoid hemorrhage. J Neurosurg 2004;101:408–16.
outflow in dogs. J Appl Physiol 1982;53(6):1496–503. 33. Wendell LC, Khan A, Raser J, et al. Successful management of refractory
10. Rosner MJ. Pathophysiology and management of increased intracranial intracranial hypertension from acute hyperammonemic encephalopathy in a
pressure. In: Andrews BT, editor. Neurosurgical intensive care. New York: woman with ornithine transcarbamylase deficiency. Neurocrit Care 2010;13;
McGraw Hill; 1993. p. 57–112. 113–7.
11. Gopinath SP, Robertson CS, Contant CF, et al. Jugular venous desaturation 34. Cremer OL, van Dijk GW, van Wensen E, et al. Effect of intracranial pressure
and outcome after head injury. J Neurol Neurosurg Psychiatry 1994;57(6): monitoring and targeted intensive care on functional outcome after severe
717–23. head injury. Crit Care Med 2005;33(10):2207–13.
12. Muizelaar JP, Marmarou A, Ward JD, et al. Adverse effects of prolonged 35. Shafi S, Diaz-Arrastia R, Madden C, et al. Intracranial pressure monitoring
hyperventilation in patients with severe head injury: a randomized clinical in brain-injured patients is associated with worsening of survival. J Trauma
trial. J Neurosurg 1991;75(5):731–9. 2008;64(2):335–40.
13. Muizelaar JP, van der Poel HG, Li ZC, et al. Pial arteriolar vessel diameter 36. Bulger EM, Nathens AB, Rivara FP, et al. Management of severe head injury:
and CO2 reactivity during prolonged hyperventilation in the rabbit. institutional variations in care and effect on outcome. Crit Care Med
J Neurosurg 1988;69(6):923–7. 2002;30(8):1870–6.
14. Rockswold SB, Rockswold GL, Vargo JM, et al. Effects of hyperbaric 37. Ghajar J, Hariri RJ, Narayan RK, et al. Survey of critical care management of
oxygenation therapy on cerebral metabolism and intracranial pressure in comatose, head-injured patients in the United States. Crit Care Med
severely brain injured patients. J Neurosurg 2001;94(3):403–11. 1995;23(3):560–7.
15. Rockswold SB, Rockswold GL, Zaun DA, et al. A prospective, randomized 38. Hesdorffer DC, Ghajar J, Iacono L. Predictors of compliance with the
clinical trial to compare the effect of hyperbaric to normobaric hyperoxia on evidence-based guidelines for traumatic brain injury care: a survey of United
cerebral metabolism, intracranial pressure, and oxygen toxicity in severe States trauma centers. J Trauma 2002;52(6):1202–9.
traumatic brain injury. J Neurosurg 2010;112(5):1080–94. 39. Becker DP, Miller JD, Ward JD, et al. The outcome from severe head injury
16. Tolias CM, Reinert M, Seiler R, et al. Normobaric hyperoxia–induced with early diagnosis and intensive management. J Neurosurg 1977;47(4):
improvement in cerebral metabolism and reduction in intracranial pressure 491–502.
in patients with severe head injury: a prospective historical cohort-matched 40. Rosner MJ, Rosner SD, Johnson AH. Cerebral perfusion pressure:
study. J Neurosurg 2004;101(3):435–44. management protocol and clinical results. J Neurosurg 1995;83(6):949–62.
17. Muizelaar JP, Wei EP, Kontos HA, et al. Mannitol causes compensatory 41. Grande PO. The “Lund concept” for the treatment of severe head trauma–
cerebral vasoconstriction and vasodilation in response to blood viscosity physiological principles and clinical application. Intensive Care Med
changes. J Neurosurg 1983;59(5):822–8. 2006;32(10):1475–84. Epub 2006, Aug 2.
18. Muizelaar JP, Lutz 3rd HA, Becker DP. Effect of mannitol on ICP and CBF 42. Guillaume J, Janny P. Monometrie intracranienne continué; Intérêt physio-
and correlation with pressure autoregulation in severely head-injured pathologique et clinique de la methode. La Presse Medicale 1951;59:953–5.
patients. J Neurosurg 1984;61(4):700–6. 43. Lundberg N. Continuous recording and control of ventricular fluid pressure
19. Stiefel MF, Tomita Y, Marmarou A. Secondary ischemia impairing the in neurosurgical practice. Acta Psychiatr Scand Suppl 1960;149:1–193.
restoration of ion homeostasis following traumatic brain injury. J Neurosurg 44. Haar FL, Sadhu VK, Sampson J, et al. Can CT scan findings predict ICP in
2005;103(4):707–14. closed head injury patients? The Thirtieth Annual Meeting of the Congress
20. Marmarou A, Signoretti S, Fatouros PP, et al. Predominance of cellular of Neurological Surgeons. Houston, Texas; 1980. p. 122–4.
edema in traumatic brain swelling in patients with severe head injuries. 45. Marmarou A, Anderson RL, Ward JD, et al. Impact of ICP instability and
J Neurosurg 2006;104(5):720–30. hypotension on outcome in patients with severe head trauma. J Neurosurg
21. Bullock MR, Chesnut R, Ghajar J, et al. Guidelines for the surgical 1991;75(Suppl):S159–66.
management of traumatic brain injury. Neurosurgery 2006;58(Suppl. 3):Si–60. 46. Marshall LF, Gautille T, Klauber MR, et al. The outcome of severe head
22. Hutchinson P, Timofeev I, Kirkpatrick P. Surgery for brain edema. injury. J Neurosurg 1991;75(Suppl):S28–36.
Neurosurg Focus 2007;22(5):E14. 47. Marshall LF, Smith RW, Shapiro HM. The outcome with aggressive
23. Timofeev I, Czosnyka M, Nortje J, et al. Effect of decompressive craniectomy treatment in severe head injuries. Part I. The significance of intracranial
on intracranial pressure and cerebrospinal compensation following pressure monitoring. J Neurosurg 1979;50(1):20–5.
traumatic brain injury. J Neurosurg 2008;108(1):66–73. 48. Miller JD, Becker DP, Ward JD, et al. Significance of intracranial
24. Li LM, Timofeev I, Czosnyka M, et al. Review article: the surgical approach hypertension in severe head injury. J Neurosurg 1977;47(4):503–16.
to the management of increased intracranial pressure after traumatic brain 49. Pitts E, Katkis J. Effects of megadose steroids on ICP in traumatic coma. In:
injury. Anesth Analg 2010;111(3):736–48. Epub 2010, Aug 4. Shulman K, Marmarou A, Miller A, editors. Intracranial pressure IV. New
25. Vahedi K, Hofmeijer J, Juettler E, et al. Early decompressive surgery in York: Springer-Verlag; 1972. p. 638–42.
malignant infarction of the middle cerebral artery: a pooled analysis of three 50. Schoon P, Benito Mori L, Orlandi G, et al. Incidence of intracranial
randomised controlled trials. Lancet Neurol 2007;6(3):215–22. hypertension related to jugular bulb oxygen saturation disturbances in severe
26. Hofmeijer J, Kappelle LJ, Algra A, et al. Surgical decompression for traumatic brain injury patients. Acta Neurochir Suppl 2002;81:285–7.
space-occupying cerebral infarction (the Hemicraniectomy After Middle
cerebral artery infarction with Life-threatening Edema Trial [HAMLET]): A complete list of references for this chapter can be found online at
a multicenter, open, randomized trial. Lancet Neurol 2009;8(4):326–33. www.expertconsult.com.
27. Cooper DJ, Rosenfeld JV, Murray L, et al. Decompressive craniectomy in
diffuse traumatic brain injury. N Engl J Med 2011;364(16):1493–502. Epub
2011, Mar 25.
Section VI—Intracranial Monitoring 347.e1

References 26. Hofmeijer J, Kappelle LJ, Algra A, et al. Surgical decompression for
space-occupying cerebral infarction (the Hemicraniectomy After Middle
1. Bratton SL, Chesnut RM, Ghajar J, et al. Guidelines for the management of cerebral artery infarction with Life-threatening Edema Trial [HAMLET]):
severe traumatic brain injury. VI. Indications for intracranial pressure a multicenter, open, randomized trial. Lancet Neurol 2009;8(4):326–33.
monitoring. J Neurotrauma 2007;24(Suppl. 1):S37–44. 27. Cooper DJ, Rosenfeld JV, Murray L, et al. Decompressive craniectomy in
2. Odetola FO, Clark SJ, Lamarand KE, et al. Intracranial pressure monitoring diffuse traumatic brain injury. N Engl J Med 2011;364(16):1493–502. Epub
in childhood meningitis with coma: a national survey of neurosurgeons in 2011, Mar 25.
the United States. Pediatr Crit Care Med 2011;12(6):e350–6. 28. Hutchinson PJ, Timofeev I, Kolias AG, et al. Decompressive craniectomy for
3. Kamat P, Kunde S, Vos M, et al. Invasive intracranial pressure monitoring is traumatic brain injury: the jury is still out. Br J Neurosurg 2011;25(3):
a useful adjunct in the management of severe hepatic encephalopathy 441–2.
associated with pediatric acute liver failure. Pediatr Crit Care Med 2011. 29. Clifton GL, Miller ER, Choi SC, et al. Lack of effect of induction of
Epub ahead of print. hypothermia after acute brain injury. N Engl J Med 2001;344(8):556–63.
4. Kouvarellis AJ, Rohlwink UK, Sood V, et al. The relationship between basal 30. Hutchison JS, Ward RE, Lacroix J, et al. Hypothermia therapy after
cisterns on CT and time-linked intracranial pressure in paediatric head traumatic brain injury in children. N Engl J Med 2008;358(23):2447–56.
injury. Childs Nerv Syst 2011;27(7):1139–44. 31. Bernard SA, Gray TW, Buist MD, et al. Treatment of comatose survivors of
5. Kellie G. An account of the appearances observed in the dissection of two of out-of-hospital cardiac arrest with induced hypothermia. N Engl J Med
the three individuals presumed to have perished in the storm of the 3rd, and 2002;346:557–63.
whose bodies were discovered in the vicinity of Leith on the morning of the 32. Heuer G, Smith MJ, Elliott JP, et al. The relationship between intracranial
4th November 1821 with some reflections on the pathology of the brain. pressure and other clinical variables in patients with aneurysmal
Transmissions of Medical and Chirurgical Science, Edinburgh 1824;1:84–169. subarachnoid hemorrhage. J Neurosurg 2004;101:408–16.
6. Monro A. Observations on the structure and function of the nervous 33. Wendell LC, Khan A, Raser J, et al. Successful management of refractory
system. Edinburgh: Creech and Johnson; 1783. intracranial hypertension from acute hyperammonemic encephalopathy in
7. Cushing H. The third circulation in studies in intracranial physiology and a woman with ornithine transcarbamylase deficiency. Neurocrit Care
surgery. London: Oxford University Press; 1926. 2010;13;113–7.
8. Marmarou A. A review of progress in understanding the pathophysiology 34. Cremer OL, van Dijk GW, van Wensen E, et al. Effect of intracranial
and treatment of brain edema. Neurosurg Focus 2007;22(5):E1. pressure monitoring and targeted intensive care on functional outcome
9. Luce JM, Huseby JS, Kirk W, et al. Starling resistor regulates cerebral venous after severe head injury. Crit Care Med 2005;33(10):2207–13.
outflow in dogs. J Appl Physiol 1982;53(6):1496–503. 35. Shafi S, Diaz-Arrastia R, Madden C, et al. Intracranial pressure monitoring
10. Rosner MJ. Pathophysiology and management of increased intracranial in brain-injured patients is associated with worsening of survival. J Trauma
pressure. In: Andrews BT, editor. Neurosurgical intensive care. New York: 2008;64(2):335–40.
McGraw Hill; 1993. p. 57–112. 36. Bulger EM, Nathens AB, Rivara FP, et al. Management of severe head
11. Gopinath SP, Robertson CS, Contant CF, et al. Jugular venous desaturation injury: institutional variations in care and effect on outcome. Crit Care
and outcome after head injury. J Neurol Neurosurg Psychiatry 1994;57(6): Med 2002;30(8):1870–6.
717–23. 37. Ghajar J, Hariri RJ, Narayan RK, et al. Survey of critical care management
12. Muizelaar JP, Marmarou A, Ward JD, et al. Adverse effects of prolonged of comatose, head-injured patients in the United States. Crit Care Med
hyperventilation in patients with severe head injury: a randomized clinical 1995;23(3):560–7.
trial. J Neurosurg 1991;75(5):731–9. 38. Hesdorffer DC, Ghajar J, Iacono L. Predictors of compliance with the
13. Muizelaar JP, van der Poel HG, Li ZC, et al. Pial arteriolar vessel diameter evidence-based guidelines for traumatic brain injury care: a survey of
and CO2 reactivity during prolonged hyperventilation in the rabbit. United States trauma centers. J Trauma 2002;52(6):1202–9.
J Neurosurg 1988;69(6):923–7. 39. Becker DP, Miller JD, Ward JD, et al. The outcome from severe head injury
14. Rockswold SB, Rockswold GL, Vargo JM, et al. Effects of hyperbaric with early diagnosis and intensive management. J Neurosurg 1977;47(4):
oxygenation therapy on cerebral metabolism and intracranial pressure in 491–502.
severely brain injured patients. J Neurosurg 2001;94(3):403–11. 40. Rosner MJ, Rosner SD, Johnson AH. Cerebral perfusion pressure:
15. Rockswold SB, Rockswold GL, Zaun DA, et al. A prospective, randomized management protocol and clinical results. J Neurosurg 1995;83(6):
clinical trial to compare the effect of hyperbaric to normobaric hyperoxia 949–62.
on cerebral metabolism, intracranial pressure, and oxygen toxicity in severe 41. Grande PO. The “Lund concept” for the treatment of severe head trauma–
traumatic brain injury. J Neurosurg 2010;112(5):1080–94. physiological principles and clinical application. Intensive Care Med 2006;
16. Tolias CM, Reinert M, Seiler R, et al. Normobaric hyperoxia–induced 32(10):1475–84. Epub 2006, Aug 2.
improvement in cerebral metabolism and reduction in intracranial pressure 42. Guillaume J, Janny P. Monometrie intracranienne continué; Intérêt
in patients with severe head injury: a prospective historical cohort-matched physio-pathologique et clinique de la methode. La Presse Medicale 1951;59:
study. J Neurosurg 2004;101(3):435–44. 953–5.
17. Muizelaar JP, Wei EP, Kontos HA, et al. Mannitol causes compensatory 43. Lundberg N. Continuous recording and control of ventricular fluid
cerebral vasoconstriction and vasodilation in response to blood viscosity pressure in neurosurgical practice. Acta Psychiatr Scand Suppl 1960;149:
changes. J Neurosurg 1983;59(5):822–8. 1–193.
18. Muizelaar JP, Lutz HA, 3rd, Becker DP. Effect of mannitol on ICP and CBF 44. Haar FL, Sadhu VK, Sampson J, et al. Can CT scan findings predict ICP in
and correlation with pressure autoregulation in severely head-injured closed head injury patients? The Thirtieth Annual Meeting of the Congress
patients. J Neurosurg 1984;61(4):700–6. of Neurological Surgeons. Houston, Texas; 1980. p. 122–4.
19. Stiefel MF, Tomita Y, Marmarou A. Secondary ischemia impairing the 45. Marmarou A, Anderson RL, Ward JD, et al. Impact of ICP instability and
restoration of ion homeostasis following traumatic brain injury. hypotension on outcome in patients with severe head trauma. J Neurosurg
J Neurosurg 2005;103(4):707–14. 1991;75(Suppl):S159–66.
20. Marmarou A, Signoretti S, Fatouros PP, et al. Predominance of cellular 46. Marshall LF, Gautille T, Klauber MR, et al. The outcome of severe head
edema in traumatic brain swelling in patients with severe head injuries. injury. J Neurosurg 1991;75(Suppl):S28–36.
J Neurosurg 2006;104(5):720–30. 47. Marshall LF, Smith RW, Shapiro HM. The outcome with aggressive
21. Bullock MR, Chesnut R, Ghajar J, et al. Guidelines for the surgical treatment in severe head injuries. Part I. The significance of intracranial
management of traumatic brain injury. Neurosurgery 2006;58(Suppl. 3): pressure monitoring. J Neurosurg 1979;50(1):20–5.
Si–60. 48. Miller JD, Becker DP, Ward JD, et al. Significance of intracranial
22. Hutchinson P, Timofeev I, Kirkpatrick P. Surgery for brain edema. hypertension in severe head injury. J Neurosurg 1977;47(4):503–16.
Neurosurg Focus 2007;22(5):E14. 49. Pitts E, Katkis J. Effects of megadose steroids on ICP in traumatic coma. In:
23. Timofeev I, Czosnyka M, Nortje J, et al. Effect of decompressive Shulman K, Marmarou A, Miller A, editors. Intracranial pressure IV. New
craniectomy on intracranial pressure and cerebrospinal compensation York: Springer-Verlag; 1972. p. 638–42.
following traumatic brain injury. J Neurosurg 2008;108(1):66–73. 50. Schoon P, Benito Mori L, Orlandi G, et al. Incidence of intracranial
24. Li LM, Timofeev I, Czosnyka M, et al. Review article: the surgical approach hypertension related to jugular bulb oxygen saturation disturbances in
to the management of increased intracranial pressure after traumatic brain severe traumatic brain injury patients. Acta Neurochir Suppl 2002;81:
injury. Anesth Analg 2010;111(3):736–48. Epub 2010, Aug 4. 285–7.
25. Vahedi K, Hofmeijer J, Juettler E, et al. Early decompressive surgery in 51. Vik A, Nag T, Fredriksli OA, et al. Relationship of “dose” of intracranial
malignant infarction of the middle cerebral artery: a pooled analysis of hypertension to outcome in severe traumatic brain injury.
three randomised controlled trials. Lancet Neurol 2007;6(3):215–22. J Neurosurg 2008;109(4):678–84.
347.e2 Section VI—Intracranial Monitoring

52. Chesnut RM, Gautille T, Blunt BA, et al. Neurogenic hypotension in 78. Morris GF, Juul N, Marshall SB, et al. Neurological deterioration as a
patients with severe head injuries. J Trauma 1998;44(6):958–63; discussion potential alternative endpoint in human clinical trials of experimental
63–4. pharmacological agents for treatment of severe traumatic brain injuries.
53. Eisenberg H, Frankowski R, Contant C, et al. High-dose barbiturate control Executive Committee of the International Selfotel Trial. Neurosurgery
of elevated intracranial pressure in patients with severe head injury. 1998;43(6):1369–72; discussion 72–4.
J Neurosurg 1988;69:15–23. 79. Marshall LF, Bowers-Marshall S, Klauber MR, et al. A new classification of
54. Schwartz ML, Tator CH, Rowed DW, et al. The University of Toronto head head injury based on computerized tomography. J Neurosurg 1991;
injury treatment study: a prospective, randomized comparison of 75(Suppl):S14–20.
pentobarbital and mannitol. Can J Neurological Sci 1984;11(4):434–40. 80. Hiler M, Czosnyka M, Hutchinson P, et al. Predictive value of initial
55. Ward JD, Becker DP, Miller JD, et al. Failure of prophylactic barbiturate computerized tomography scan, intracranial pressure, and state of
coma in the treatment of severe head injury. J Neurosurg 1985;62(3):383–8. autoregulation in patients with traumatic brain injury. J Neurosurg 2006;
56. Robertson CS, Valadka AB, Hannay HJ, et al. Prevention of secondary 104(5):731–7.
ischemic insults after severe head injury. Crit Care Med 1999;27(10): 81. Servadei F, Antonelli V, Giuliani G, et al. Evolving lesions in traumatic
2086–95. subarachnoid hemorrhage: prospective study of 110 patients with emphasis
57. Girard TD, Kress JP, Fuchs BD, et al. Efficacy and safety of a paired sedation on the role of ICP monitoring. Acta Neurochir Suppl 2002;81:81–2.
and ventilator weaning protocol for mechanically ventilated patients in 82. Timofeev I, Dahyot-Fizelier C, Keong N, et al. Ventriculostomy for control
intensive care (Awakening and Breathing Controlled trial): a randomised of raised ICP in acute traumatic brain injury. Acta Neurochir Suppl
controlled trial. Lancet 2008;371(9607):126–34. 2008;102:99–104.
58. Kress JP, Pohlman AS, O’Connor MF, et al. Daily interruption of sedative 83. Exo J, Kochanek PM, Adelson PD, et al. Intracranial pressure-monitoring
infusions in critically ill patients undergoing mechanical ventilation. N Engl systems in children with traumatic brain injury: combining therapeutic and
J Med 2000;342(20):1471–7. diagnostic tools. Pediatr Crit Care Med 2010. Epub ahead of print.
59. Forsyth RJ, Wolny S, Rodrigues B. Routine intracranial pressure monitoring 84. Mollman HD, Rockswold GL, Ford SE. A clinical comparison of
in acute coma. Cochrane Database Syst Rev 2010; CD002043. subarachnoid catheters to ventriculostomy and subarachnoid bolts: a
60. Marshall LF. Treatment of brain swelling and brain edema in man. Adv prospective study. J Neurosurg 1988;68(5):737–41.
Neurol 1980;28:459–69. 85. Ostrup RC, Luerssen TG, Marshall LF, et al. Continuous monitoring of
61. Saul TG, Ducker TB. Effect of intracranial pressure monitoring and intracranial pressure with a miniaturized fiberoptic device. J Neurosurg
aggressive treatment on mortality in severe head injury. J Neurosurg 1987;67(2):206–9.
1982;56(4):498–503. 86. Piek J, Bock WJ. Continuous monitoring of cerebral tissue pressure in
62. Stein SC, Georgoff P, Meghan S, et al. 150 years of treating severe traumatic neurosurgical practice–experiences with 100 patients. Intensive Care Med
brain injury: a systematic review of progress in mortality. J Neurotrauma 1990;16(3):184–8.
2010;27(7):1343–53. 87. Sahuquillo J, Poca MA, Arribas M, et al. Interhemispheric supratentorial
63. Bullock R, Chesnut R, Clifton G, et al. Guidelines for the management of intracranial pressure gradients in head-injured patients: are they clinically
severe head injury. J Neurotrauma 1996;13(11):639–734. important? J Neurosurg 1999;90(1):16–26.
64. Harvey S, Harrison DA, Singer M, et al. Assessment of the clinical 88. Ehtisham A, Taylor S, Bayless L, et al. Placement of external ventricular
effectiveness of pulmonary artery catheters in management of patients in drains and intracranial pressure monitors by neurointensivists. Neurocrit
intensive care (PAC-Man): a randomised controlled trial. Lancet 2005; Care 2009; 10:241–7.
366(9484):472–7. 89. Harris CH, Smith RS, Helmer SD, et al: Placement of intracranial pressure
65. Richard C, Warszawski J, Anguel N, et al. Early use of the pulmonary artery monitors by non-neurosurgeons. Am Surg 2002;68(9):787–90.
catheter and outcomes in patients with shock and acute respiratory distress 90. Kaups KL, Parks SN, Morris CL: Intracranial pressure monitor placement
syndrome: a randomized controlled trial. JAMA 2003;290(20):2713–20. by midlevel practitioners. J Trauma 1998;45(5):884–6.
66. Sandham JD, Hull RD, Brant RF, et al. A randomized, controlled trial of the 91. Beer R, Lackner P, Pfausler B, et al. Nosocomial ventriculitis and meningitis
use of pulmonary-artery catheters in high-risk surgical patients. N Engl J in neurocritical care patients. J Neurol 2008;255:1617–24.
Med 2003;348(1):5–14. 92. Lo CH, Spelman D, Bailey M, et al. External ventricular drain infections are
67. Schreiber MA, Aoki N, Scott BG, et al. Determinants of mortality in independent of drain duration: an argument against elective revision.
patients with severe blunt head injury. Arch Surg 2002;137(3):285–90. J Neurosurg 2007;106:378–83.
68. Stiefel MF, Udoetek J, Spiotta A, et al. Conventional neurocritical care and 93. Camacho EF, Boszczowski I, Basso M, et al. Infection rate and risk factors
cerebral oxygenation after traumatic brain injury. J Neurosurg 2006;105: associated with infections related to external ventricular drain. Infection.
568–75. 2011;39(1):47–51. Epub 2011, Jan 25.
69. Le Roux P, Lam AM, Newell DW, et al. Cerebral arteriovenous difference of 94. Williams TA, Leslie GD, Dobb GJ, et al. Decrease in proven ventriculitis by
oxygen: a predictor of cerebral infarction and outcome in severe head reducing the frequency of cerebrospinal fluid sampling from
injury. J Neurosurg 1997;87:1–8. extraventricular drains. J Neurosurg 2011. Epub ahead of print.
70. Nangunoori R, Maloney-Wilensky E, Stiefel MD, et al. Brain tissue oxygen 95. Scheithauer S, Bürgel U, Ryang YM, et al. Prospective surveillance of drain
based therapy and outcome after severe traumatic brain injury: a systematic associated meningitis/ventriculitis in a neurosurgery and neurological
literature review. Neurocritical Care 2012;17:131−8. intensive care unit. J Neurol Neurosurg Psychiatry 2009;80(12):1381–5.
71. Chambers IR, Treadwell L, Mendelow AD. Determination of threshold Epub 2009, Mar 16.
levels of cerebral perfusion pressure and intracranial pressure in severe 96. Mayhall CG, Archer NH, Lamb VA, et al. Ventriculostomy-related
head injury by using receiver-operating characteristic curves: an infections: a prospective epidemiologic study. N Engl J Med 1984;310(9):
observational study in 291 patients. J Neurosurg 2001;94(3):412–6. 553–9.
72. Chesnut RM, Marshall LF, Klauber MR, et al. The role of secondary brain 97. Aucoin P, Kotilainen H, Gantz N, et al. Intracranial pressure monitors:
injury in determining outcome from severe head injury. J Trauma 1993; epidemiologic study of risk factors and infections. Am J Med
34(2):216–22. 1986;80:369–76.
73. Fearnside MR, Cook RJ, McDougall P, et al. The Westmead Head Injury 98. Holloway KL, Barnes T, Choi S, et al. Ventriculostomy infections: the effect
Project outcome in severe head injury. A comparative analysis of of monitoring duration and catheter exchange in 584 patients. J Neurosurg
pre-hospital, clinical and CT variables. Br J Neurosurg 1993;7(3):267–79. 1996;85(3):419–24.
74. Jones PA, Andrews PJ, Midgley S, et al. Measuring the burden of secondary 99. Lozier AP, Sciacca RR, Romagnoli MF, et al. Ventriculostomy-related
insults in head-injured patients during intensive care. J Neurosurg infections: a critical review of the literature. Neurosurgery 2002;51(1):170–
Anesthesiol 1994;6(1):4–14. 81; discussion 181–2.
75. Bratton S, Bullock R, Carney N, et al. Guidelines for the management of 100. Winfield JA, Rosenthal P, Kanter RK, et al. Duration of intracranial pressure
severe brain injury: 2007 Revision. J Neurotrauma 2007;24(Suppl .1): monitoring does not predict daily risk of infectious complications.
S1–106. Neurosurgery 1993;33(3):424–30; discussion 430–1.
76. Marshall LF, Barba D, Toole BM, et al. The oval pupil: clinical significance 101. Zabramski JM, Whiting D, Darouiche RO, et al. Efficacy of antimicrobial-
and relationship to intracranial hypertension. J Neurosurg 1983;58:566–8. impregnated external ventricular drain catheters: a prospective,
77. Juul N, Morris GF, Marshall SB, et al. Intracranial hypertension and randomized, controlled trial. J Neurosurg 2003;98(4):725–30.
cerebral perfusion pressure: influence on neurological deterioration and 102. Fichtner J, Guresir E, Seifert V, et al. Efficacy of silver-bearing external
outcome in severe head injury. The Executive Committee of the ventricular drainage catheters: a retrospective analysis. J Neurosurg
International Selfotel Trial. J Neurosurg 2000;92(1):1–6. 2010;112:840–6.
Section VI—Intracranial Monitoring 347.e3

103. Muttaiyah S, Ritchie S, John S, et al. Efficacy of antibiotic- 129. Matevosyan K, Madden C, Barnett SL, et al. Coagulation factor levels in
impregnated external ventricular drain catheters. J Clin Neurosci 2010;17: neurosurgical patients with mild prolongation of prothrombin time: effect
296–8. on plasma transfusion therapy. J Neurosurg 2010;114:3–7.
104. Honda H, Jones JC, Craighead MC, et al. Reducing the incidence of 130. Davis JW, Davis IC, Bennink LD, et al. Placement of intracranial pressure
intraventricular catheter-related ventriculitis in the neurology-neurosurgical monitors: are “normal” coagulation parameters necessary? J Trauma
intensive care unit at a tertiary care center in St Louis, Missouri: an 8-year 2004;57(6):1173–7.
follow-up study. Infect Control Hosp Epidemiol 2010;31(10):1078–81. 131. Le TV, Rumbak MJ, Liu SS, et al. Insertion of intracranial pressure
105. Leverstein-van Hall MA, Hopmans TE, van der Sprenkel JW, et al. A bundle monitors in fulminant hepatic failure patients: early experience using
approach to reduce the incidence of external ventricular and lumbar recombinant factor VII. Neurosurgery 2010;66(3):455–8; discussion 458.
drain-related infections. J Neurosurg. 2010;112(2):345–53. 132. Smielewski P, Lavinio A, Timofeev I, et al. ICM? a flexible platform for
106. Park P, Garton HJ, Kocan MJ, et al. Risk of infection with prolonged investigations of cerebrospinal dynamics in clinical practice. Acta Neurochir
ventricular catheterization. Neurosurgery 2004;55(3):594–9; discussion Suppl 2008;102:145–51.
599–601. 133. Venkatesh B, Garrett P, Fraenkel DJ, et al. Indices to quantify changes in
107. Wong GK, Poon WS, Wai S, et al. Failure of regular external ventricular intracranial and cerebral perfusion pressure by assessing agreement
drain exchange to reduce cerebrospinal fluid infection: result of a between hourly and semi-continuous recordings. Intensive Care Med 2004;
randomised controlled trial. J Neurol Neurosurg Psychiatry 2002;73(6): 30:510–13.
759–61. 134. Stein DM, Hu PF, Brenner M, et al. Brief episodes of intracranial
108. Paramore CG, Turner DA. Relative risks of ventriculostomy infection and hypertension and cerebral hypoperfusion are associated with poor
morbidity. Acta Neurochirurgica 1994;127(1–2):79–84. functional outcome after severe traumatic brain injury. J Trauma 2011;
109. Flibotte JJ, Lee KE, Koroshetz WJ, Rosand J, et al. Continuous antibiotic 71(2):364–74.
prophylaxis and cerebral spinal fluid infection in patients with intracranial 135. Bremmer R, de Jong BM, Wagemakers M, et al. The course of intracranial
pressure monitors. Neurocrit Care 2004;1(1):61–8. pressure in traumatic brain injury: relation with outcome and
110. Gelabert-Gonzalez M, Ginesta-Galan V, Sernamito-Garcia R, et al. The CT-characteristics. Neurocrit Care 2010;12:362–8.
Camino intracranial pressure device in clinical practice: assessment in 1000 136. O’Phelan KH, Park D, Efird JT, et al. Patterns of increased intracranial
cases. Acta Neurochir (Wien) 2006;148(4):435–41. Epub 2005, Dec 27. pressure after severe traumatic brain injury. Neurocrit Care 2009;10(3):
111. Jensen RL, Hahn YS, Ciro E. Risk factors of intracranial pressure 280–6.
monitoring in children with fiberoptic devices: a critical review. Surg 137. Stocchetti N, Mattioli C, Paparella A, et al. Bedside assessment of CO2
Neurol 1997;47(1):16–22. reactivity in head injury: changes in CBF estimated by changes in ICP and
112. Morton R, Lucas 2nd TH, Ko A, et al. Intracerebral abscess associated with cerebral extraction of oxygen. J Neurotrauma 1993;10:187.
the Camino intracranial pressure monitor: case report and review of the 138. Yoshihara M, Bandoh K, Marmarou A. Cerebrovascular carbon dioxide
literature. Neurosurgery 2011. Epub ahead of print. reactivity assessed by intracranial pressure dynamics in severely head
113. Narayan RK, Kishore PR, Becker DP, et al. Intracranial pressure: to monitor injured patients. J Neurosurg 1995;82(3):386–93.
or not to monitor? A review of our experience with severe head injury. 139. Miller JD, Garibi J, Pickard JD. A clinical study of intracranial volume
J Neurosurg 1982;56(5):650–9. pressure relationships. Br J Surg 1973;60(4):316.
114. Guyot LL, Dowling C, Diaz FG, et al. Cerebral monitoring devices: analysis 140. Miller JD, Garibi J, Pickard JD. Induced changes of cerebrospinal fluid
of complications. Acta Neurochir Suppl 1998;71:47–9. volume: effects during continuous monitoring of ventricular fluid pressure.
115. North B, Reilly P. Comparison among three methods of intracranial Arch Neurol 1973;28(4):265–9.
pressure recording. Neurosurgery 1986;18(6):730–2. 141. Miller JD. Volume and pressure in the craniospinal axis. Clin Neurosurg
116. Wiesmann M, Mayer TE. Intracranial bleeding rates associated with two 1975;22:76–105.
methods of external ventricular drainage. J Clin Neurosci 2001;8(2):126–8. 142. Maset AL, Marmarou A, Ward JD, et al. Pressure-volume index in head
117. Anderson RC, Kan P, Klimo P, et al. Complications of intracranial pressure injury. J Neurosurg 1987;67(6):832–40.
monitoring in children with head trauma. J Neurosurg 2004;101(Suppl. 1): 143. Luerssen TG, Chesnut RM, Van Berkum-Clark M, et al. Post traumatic
53–8. cerebrospinal fluid infections in the Traumatic Coma Data Bank: the
118. Blaha M, Lazar D. Traumatic brain injury and haemorrhagic complications influence of type and management of ICP monitors. In: Avezaat C, van
after intracranial pressure monitoring. J Neurol Neurosurg Psychiatry Eijndhoven JHM, Maas AJR, et al, editors. Intracranial pressure VIII. Berlin:
2005;76(1):147. Springer-Verlag; 1993. p. 157–63.
119. Koskinen LO, Olivecrona M. Clinical experience with the intraparenchymal 144. Piper I, Spiegelberg A, Whittle I, et al. A comparative study of the
intracranial pressure monitoring Codman MicroSensor system. Spiegelberg compliance device with a manual volume-injection method: a
Neurosurgery 2005;56(4):693–8; discussion 698. clinical evaluation in patients with hydrocephalus. Br J Neurosurg 1999;
120. Citerio G, Piper I, Chambers IR, et al. Multicenter clinical assessment of the 13(6):581–6.
Raumedic Neurovent-P intracranial pressure sensor: a report by the 145. Yau YH, Piper IR, Clutton RE, et al. An experimental evaluation of a new
BrainIT group. Neurosurgery 2008;63:1152–8. intraparenchymal continuous compliance probe: preliminary studies. Acta
121. Al-Tamimi YZ, Helmy A, Bavetta S, et al. Assessment of zero drift in the Neurochir 2002;81:181–2.
Codman intracranial pressure monitor: a study from 2 neurointensive care 146. Adolph RJ, Fukusumi H, Fowler NO. Origin of cerebrospinal fluid
units. Neurosurgery 2009;64:94–8; discussion 98–9. pulsations. Am J Physiol 1967;212(4):840–6.
122. Martinez-Manas RM, Santamarta D, de Campos JM, et al. Camino 147. Hamer J, Alberti E, Hoyer S, et al. Influence of systemic and cerebral
intracranial pressure monitor: prospective study of accuracy and vascular factors on the cerebrospinal fluid pulse waves. J Neurosurg 1977;
complications. J Neurol Neurosurg Psychiatry 2000;69:82–6. 46(1):36–45.
123. Eide PK, Bakken A. The baseline pressure of intracranial pressure (ICP) 148. Mitchell P, Kirkness C, Burr R, et al. Intracranial pressure and transcranial
sensors can be altered by electrostatic discharges. Biomed Eng Online Doppler waveform analysis. In: Marmarou A, Bullock MR, Avezaat CJ,
2011;10(1):75. Epub ahead of print. editors. Intracranial pressure and neuromonitoring in brain injury. New
124. Dahlqvist MB, Andres RH, Raabe A, et al. Brain herniation in a patient York: Springer; 1997. p. 420.
with apparently normal intracranial pressure: a case report. J Med Case Rep 149. Contant Jr CF, Robertson CS, Crouch J, et al. Intracranial pressure
2010;4:297. waveform indices in transient and refractory intracranial hypertension.
125. Marshall LE, Zovickian J, Ostrup R, et al. Multiple simultaneous recordings J Neurosci Methods 1995;57(1):15–25.
of ICP in patients with acute mass lesions. In: Miller JD, Teasdale GM, 150. Panerai RB. Assessment of cerebral pressure autoregulation in humans:
Rowan JO, et al, editors. Intracranial pressure VI. Berlin: Springer-Verlag; a review of measurement methods. Physiol Meas 1998;19(3):305–38.
1986. p. 184–6. 151. Robertson CS, Narayan RK, Contant CF, et al. Clinical experience with a
126. Weaver DD, Winn HR, Jane JA. Differential intracranial pressure in patients continuous monitor of intracranial compliance. J Neurosurg 1989;71
with unilateral mass lesions. J Neurosurg 1982;56(5):660–5. (5 Pt 1):673–80.
127. Yano M, Ikeda Y, Kobayashi S, et al. Intracranial pressure in head-injured 152. Wachi A, Marmarou A, Sato K. [Test of non-invasive PVI measure with a
patients with various intracranial lesions is identical throughout the fiber-optic pressure monitoring device in head injured patients]. No To
supratentorial intracranial compartment. Neurosurgery 1987;21(5):688–92. Shinkei 1990;42(6):561–8.
128. Bauer DF, McGwin Jr G, Melton SM, et al. The relationship between INR 153. Hu X, Xu P, Asgari S, et al. Forecasting ICP elevation based on prescient
and development of hemorrhage with placement of ventriculostomy. J changes of intracranial pressure waveform morphology. IEEE Trans Biomed
Trauma 2010. Epub ahead of print. Eng 2010;57(5):1070–8.
347.e4 Section VI—Intracranial Monitoring

154. Hamilton R, Xu P, Asgari S, et al. Forecasting intracranial pressure elevation 157. Balestreri M, Czosnyka M, Steiner LA, et al. Association between outcome,
using pulse waveform morphology. Conf Proc IEEE Eng Med Biol Soc cerebral pressure reactivity and slow ICP waves following head injury. Acta
2009;2009:4331–4. Neurochir Suppl 2005;95:25–8.
155. Steiner LA, Czosnyka M, Piechnik SK, et al. Continuous monitoring of 158. Maset AL, Marmarou A, Ward JD, et al. Pressure-volume index in head
cerebrovascular pressure reactivity allows determination of optimal cerebral injury. J Neurosurg 1987;67:832–40.
perfusion pressure in patients with traumatic brain injury. Crit Care Med 159. Padayachy LC, Figaji AA, Bullock MR. Intracranial pressure monitoring for
2002;30(4):733–8. traumatic brain injury in the modern era. Childs Nerv Syst
156. Czosnyka M, Guazzo E, Whitehouse M, et al. Significance of intracranial 2010;26(4):441–52.
pressure waveform analysis after head injury. Acta Neurochir 1996;138:531–
41; discussion 41–2.
VI
Chapter
35  

Brain Oxygen
Mauro Oddo and Peter D. le Roux

Introduction Definition
Maintenance of an adequate tissue oxygenation is a funda­ Brain tissue oxygen is defined as the partial pressure of oxygen
mental objective in the field of critical care medicine in in the interstitial space of the brain and reflects the availability
general, and the assessment of tissue oxygenation is an essen­ of oxygen for oxidative energy production. There has been
tial part of the management of the critically ill patient. This some debate as to whether the monitor systems measure tissue
provides information about oxygen supply and use in specific oxygen pressure or tension, and, accordingly, several abbrevia­
tissue beds.1 Hypoxia is defined as a reduction of tissue oxy­ tions have been used for brain tissue oxygen, including PbO2,
genation to levels that are insufficient to maintain cellular PbrO2, PbtO2, PtiO2, and BTO2. A consensus conference at the
function and metabolism. The pathophysiology of tissue 13th International Symposium on Intracranial Pressure and
hypoxia can be classified as follows: Brain Monitoring held July 2007 in San Francisco, California,
Ischemic proposed that PbtO2 be used as the standard abbreviation.
Cytopathic Accordingly, this chapter uses the PbtO2 when referring to
Anemic brain tissue oxygen pressure. Consistent with this abbreviation
Hypoxic clinical studies suggest that PbtO2 may be best defined by the
In the brain tissue, ischemic hypoxia may result from macro­ equation:
vascular ischemia (i.e., reduced or absent cerebral blood flow
PbtO2 = CBF × AVTO2
[CBF], e.g., cerebral thrombosis, vasospasm, low arterial
carbon dioxide tension [PaCO2], alkalosis) or from micro­ where AVTO2 is arterial oxygen tension (PaO2) minus oxygen
vascular ischemia (e.g., blood-brain barrier disruption and partial pressure in venous blood (PvO2)—that is, PbtO2
brain capillary endothelial dysfunction).2 Cytopathic hypoxia represents the interaction between plasma oxygen tension
in the brain includes a complex cascade of events after and CBF.5
the initial brain insult (e.g., glutamate excitotoxicity, intracel­
lular calcium influx, free radical and inflammatory cytokine
release) that lead to cell energy and mitochondrial dysfunc­
Technology
tion,3,4 diffusion limited oxygen delivery to the cell,5,6 and There are four methods to measure brain oxygen: jugular
reduced oxygen extraction.7 Anemia8,9 and impaired systemic venous bulb oximetry (Chapter 32), direct brain tissue oxygen
oxygenation10 are additional causes of brain tissue hypoxia tension measurement, near infrared spectroscopy (Chapter
(Table 35.1). 33), and oxygen-15 positron emission tomography (PET)
Brain hypoxia can cause secondary brain damage. Thus (Chapter 29). This chapter discusses direct brain tissue oxygen
monitoring of brain tissue oxygen partial pressure to dis­ tension measurement, which is now the most common tech­
criminate between normal and critically impaired tissue nique used in the neurocritical care unit (NCCU) to assess
oxygenation has been integrated into the management of cerebral oxygenation. The technique involves the insertion of
patients with acute brain injury, and several lines of evidence a fine catheter (approximately 0.5 mm in diameter) into the
suggest that it is a clinically useful modality in neurocritical brain parenchyma, specifically the white matter, to continu­
care.11-22 In addition, several observational studies show that ously measure PbtO2. The PbtO2 catheters or probes can be
cerebral perfusion pressure (CPP) and intracranial pressure inserted through a single- or multiple-lumen bolt secured in
(ICP) are not surrogates for brain oxygen and that brain a single burr hole or they may be tunneled under the scalp.
oxygenation varies independently of cerebral hemodynamics The procedure to insert the monitor may be performed in the
and ICP.19,23,24 Like other monitors, a brain oxygen monitor operating room or at the bedside in the NCCU. PbtO2 probes
is not intended to be used alone; instead it should be used generally are placed adjacent to an ICP monitor and through
as a supplement or complement to other monitors, for the same triple-lumen bolt. Ideally the position and function
example, an ICP monitor (see Chapter 34) or microdialysis of the monitor should be confirmed with a head computed
(see Chapter 36). tomography (CT) scan or oxygen challenge test particularly if
348 © Copyright 2013 Elsevier Inc. All rights reserved.
Section VI—Intracranial Monitoring 349

e−
Table 35.1  Causes of Brain Tissue Hypoxia
Etiology Pathophysiology
Current measurement
Ischemic
Macrovascular ↓ O2 delivery, ↓ CBF (thrombosis,
vasospasm, low PaCO2) + −
Microvascular BBB disruption, vasogenic edema, 700 mV polarization
endothelial dysfunction
Cytopathic ↓ O2 extraction (cytotoxic edema,
diffusion limited O2 delivery,
mitochondrial dysfunction) 1
3
Anemic ↓ Hemoglobin concentration
Hypoxic ↓ PaO2, SaO2

BBB, Blood-brain barrier; CBF, cerebral blood flow; PaCO2, arterial carbon
dioxide tension; PaO2, arterial oxygen tension; SaO2, arterial oxygen 5
saturation. 2 4

O2

the initial PbtO2 reading after 30 to 60 minutes of stabilization


is abnormal. A
Two primary technologies underlie PbtO2 measurement:
one is based on the Clark principle, the other on an optical
technique. The Clark principle uses the electrochemical prop­
erties of noble metals to measure the oxygen content of tissue.1
The Clark electrode consists of a membrane that covers a layer
of electrolyte and two metallic electrodes. Oxygen diffuses
through the membrane and is electrochemically reduced at
the cathode. The greater the oxygen partial pressure, the more
oxygen diffuses through the membrane. The change in voltage
between the reference electrode and the measuring electrode
is proportional to the amount of oxygen molecules being
reduced on the cathode. The Licox PbtO2 probe (Integra Life­
sciences, Plainsboro, NJ) uses such a closed polarographic
Clark-type cell with reversible electrochemical electrodes (Fig.
35.1). This oxygen-consuming process is temperature depen­
dent, and so requires constant calibration of the system to
patient temperature. The Licox system includes a brain tem­
perature probe (thermocouple) that is inserted through a
triple-lumen bolt with the PbtO2 and ICP monitor (e.g.,
Camino, Integra Lifesciences). This temperature probe mea­
sures brain temperature and allows for automatic calibration.
The new Licox PMO probe can measure PbtO2 and brain tem­
perature using a single probe.25 The Neurovent-P Temp (Rau­ B
medic AG, Munchberg, Germany), which uses the same
polarographic technique as the Licox, also can measure PbtO2 Fig. 35.1  A, Schematic diagram of the Licox polarographic oxygenation
and brain temperature in one catheter. There are important probe. The numbered components of the diagram are (1) polyethylene
tube diffusion membrane; (2) polarographic gold cathode; (3) polaro-
differences in values obtained by the Neurovent and Licox
graphic silver anode; (4) cell filled with electrolyte; and (5) cerebral tissue.
devices and so the values obtained by these devices cannot be B, Schematic diagram of the Licox probe that illustrates placement
used interchangeably.26-28 Purpose-designed triple-lumen through a cranial bolt into the cerebral tissues. Placement is similar to
cranial access devices29 allow simultaneous PbtO2, ICP, and an intracranial pressure (ICP) monitor and is frequently used through 
cerebral microdialysis monitoring. However, if brain tempera­ the same bolt. (Adapted from Mulvey JM, Dorsch M, Mudaliar Y, et al. Multimo-
ture is not measured (e.g., a microdialysis catheter is inserted dality monitoring in severe traumatic brain injury: the role of brain tissue oxygen-
through the lumen instead of the temperature probe), the ation monitoring. Neurocrit Care 2004;1(3):391–402.)
monitor should be calibrated manually on a regular basis, for
example, every 30 minutes by using core body temperature.
The Licox system averages oxygen over a probe area of 14 to are used to measure concentrations of substances by photo­
18 mm2 and has excellent long-term stability, even after 7 days. chemical reactions that create changes in optical properties of
The Neurovent-P Temp has a greater surface area of 24 mm2. indicator compounds.1 OxyLabPO2 (Oxford Optronix Ltd.,
The second technique used to measure PbtO2 is based on a Oxford, UK) allows PbtO2 to be measured using optical fluo­
fluorescence quenching technique in which a marker changes rescence technology. Unlike the Licox, this process does not
color according to the ambient amount of gas. Optode sensors consume oxygen and does not affect the measured oxygen
350 Section VI—Intracranial Monitoring

level. However, the probe measures a smaller area. The SjvO2.32 Postinsertion noncontrast head CT confirmation of
Neurotrend (Codman, Johnson & Johnson, Raynham, MA) probe position in the brain parenchyma (Fig. 35.2)
uses this technique, but it is no longer commercially available is important to interpret readings.33,34 In some patients a
for clinical use. The accuracy and clinical stability of the CT-perfusion study also may be useful. For example, if PbtO2
Neurotrend sensor also appears to be less than the Licox readings are consistently low or poorly responsive to therapy,
system.30 There are several other important differences it is useful to know the monitor’s proximity to a focal abnor­
between techniques based on the Clark principle (e.g., the mality or whether the region it is in is hypoperfused. In these
Licox) and on an optical technique (e.g., Neurotrend). First, cases the PbtO2 threshold for treatment may be lower than
Licox catheters are precalibrated and can be inserted without normally used. A “run-in” or equilibration time of up to 1
any pre-use calibration. However, postinsertion stabilization hour is required before readings are stable. Adjustment of
(about 1 hour) is required before readings are reliable. By insertion depth (when used through an access device) is not
contrast, the Neurotrend monitor needs bedside calibration to possible with the Licox system, and if the monitor is removed,
a defined oxygen concentration. Second, the catheters are of the insertion bolt then should be replaced if a new monitor is
different lengths; the Neurotrend is inserted at a greater depth inserted to avoid potential contamination.
than the Licox catheter. Third, the critical PbtO2 threshold for Transiently increasing the inspiratory oxygen fraction
hypoxia is different, so it is difficult to compare studies that (FiO2) and observing the corresponding PbtO2 increase can
use the different techniques. In addition, the Neurotrend help confirm function or exclude the presence of surrounding
changed design in 1998, making it difficult to compare between microhemorrhages or sensor damage at insertion. An oxygen
old and more recent studies that describe this technology.31 challenge is therefore performed when monitoring starts and,
in some institutions, daily thereafter as part of regular clinical
care to evaluate the function and responsiveness of the PbtO2
Catheter Placement probe or to determine “oxygen reactivity.” The oxygen chal­
PbtO2 catheters are placed in the right frontal lobe white matter lenge involves increasing the FiO2 from baseline to 1.0 for
in diffuse brain injury or on the side of maximal pathology approximately 5 minutes. This typically leads to a several-fold
when there is focal injury. In subarachnoid hemorrhage (SAH) increase in the amount of oxygen dissolved in the plasma,
patients, the authors place the PbtO2 monitor where maximal translating into a mean threefold increase in PbtO2.5,35 However,
vasospasm is likely to develop based on the distribution of the response to hyperoxia usually is less robust when the
SAH and aneurysm location. Recommendations for the pre­ monitor is in an underperfused (CBF is <20 mL/100 g/min)
ferred brain area to be monitored with a PbtO2 probe are sum­ region.36
marized in Table 35.2.
In general, monitors are placed when the Glasgow Coma
Scale (GCS) score is equal to or less than 8 (i.e., consistent Regional Versus Global
with the indications for an ICP monitor) (see Chapter 34).
There are no specific guidelines on how long PbtO2 should be
Measurement
monitored. In traumatic brain injury (TBI) patients the PbtO2 probes sample approximately 15 mm2 of tissue around
authors leave the monitor in place until ICP is normal for 24 the tip, and the PbtO2 value depends on O2 diffusion from the
hours without any specific treatment other than sedation for vasculature to a small amount of tissue.5,6 Therefore it is a
ventilation. On average, in patients with severe TBI this is 4 to
5 days. In SAH patients the monitor is kept in place during
the risk period for vasospasm. In addition, continuous PbtO2
can be monitored twice as long as jugular venous oxygen satu­
ration (SjvO2) and without needing recalibration. In studies
that compare PbtO2 and SjvO2, good quality data are acquired
95% of the time with PbtO2, but only 43% of the time for

Table 35.2  Recommended Areas for Brain


Oxygen Probe Insertion
Intracranial Pathology Catheter Location
Traumatic brain injury
Diffuse injury Right frontal lobe
Focal injury (subdural Pericontusional tissue
hematoma, contusion)
Subarachnoid hemorrhage Expected distribution area of the
parent artery of the aneurysm,
at highest risk for developing
symptomatic vasospasm and
delayed ischemia
Fig. 35.2  A computed tomography (CT) head scan that demonstrates
Cerebral infarction Area of lesion, at distance from
the infarcted tissue the position of a Licox probe in the white matter of the right frontal
cortex.
Section VI—Intracranial Monitoring 351

regional monitor with values that depend on probe location.


This has led to debate about probe location and whether the Table 35.3  Brain Oxygen Values in Humans*
value can be used to make decisions about global oxygenation. Condition PbtO2 Values
In most patients PbtO2 is measured in “normal appearing”
Normal 25-50 mm Hg
frontal subcortical white matter; there is evidence that this
local measurement can be regarded as an indicator of global Hypoxic thresholds
oxygenation.31,32,37-39 Consistent with this, two clinical studies Moderate brain hypoxia 15-25 mm Hg
have shown good correlation between PbtO2 and SjvO2, used Critical brain hypoxia <15 mm Hg*
to assess global brain oxygenation, in areas without focal
pathology after TBI.32,37 In areas with focal pathology, this Severe brain hypoxia <10 mm Hg
correlation between PbtO2 and SjvO2 was absent, and the brain Elevated >50 mm Hg
tissue oximetry values reflected regional brain oxygenation PbtO2, Brain tissue oxygen pressure.
better than jugular bulb oximetry.37 Other studies also show *PbtO2 thresholds according to Brain Trauma Foundation guidelines. In a
that when the probe is in tissue immediately adjacent to a National Institutes of Health–sponsored phase II trial that is expected to finish
contusion or other pathology (e.g., a subdural hematoma), patient recrurtment in 2013 to examine PbtO2, treatment is initiated when
PbtO2 is <20 mm Hg.
values are often lower, even if CPP is higher.33,34,40 In addition,
when the monitor is placed adjacent to abnormal brain,
regional hypoxia lasts longer than in normal-appearing tissue33
and the relationship with outcome appears to be more robust.34 compromised brain tissue oxygen and often is taken as a
threshold at which to consider or initiate therapy to correct
brain oxygen.9,33 This value is the threshold at which therapy
Values in Normal is initiated in an National Institutes of Health (NIH)–
and Pathologic Conditions sponsored phase II trial that is expected to finish patient
recruitment in late 2013. The 2007 guidelines for severe TBI
Normal Values recommend that PbtO2 values less than 15 mm Hg be consid­
The Licox monitor provides a measure of PbtO2 in units ered as the critical threshold for “ischemia.”57 Microdialysis
of tension (mm Hg). Usually oxygen content is expressed in studies show that at this level other markers of ischemia are
units of concentration (mL O2/100 mL) and oxygen delivery elevated.13 Normal mitochondria require an oxygen level of
to the brain and cerebral metabolic rate of oxygen (CMRO2) about 1.5 mm Hg to function; this corresponds with a PbtO2
are expressed in mL O2/100 g brain/minute. The conversion level between 15 and 20 mm Hg in normal white matter.31 A
factor: 1 mm Hg equals 0.003 mL O2/100 g brain can be used PbtO2 less than 10 mm Hg is a marker of severe brain tissue
to compare PbtO2 with other oxygen concentration measure­ hypoxia and is an independent factor associated with both
ments. Normal PbtO2 measurements have been acquired mortality and unfavorable outcome.14,17,49 Values of 0 mm Hg
experimentally, but human measurements have been restricted that persist longer than 30 minutes and show no response to
to “normal” values obtained during neurosurgical procedures an oxygen challenge are consistent with brain death.58,59 Table
and in normal-appearing brain after TBI. In animals, normal 35.3 summarizes normal and hypoxic thresholds of PbtO2.
PbtO2 varies from 30 to 40 mm Hg.41,42 In humans without
severe brain injury, PbtO2 ranges from 37 to 48 mm Hg.39,43-45
Mean values of 23 mm Hg have been measured in awake
Observational Data
subjects undergoing functional neurosurgery.46 PbtO2 values in A large body of observational clinical data on PbtO2, primarily
patients with normal ICP are between 25 and 30 mm Hg in in patients with severe TBI and less frequently SAH, has
TBI.32,47 accrued since the 1980s. These findings may be summarized
as follows. First, PbtO2 may be abnormal, despite a normal ICP
and CPP both in adults and children19,21,23,24,46,60 and when
Hypoxic Thresholds SjvO2 is normal.37 This suggests that PbtO2 may be a novel
The identification of hypoxic brain tissue permits interven­ target for resuscitation following brain injury and can supple­
tion potentially before irreversible injury occurs; that is, the ment the information provided by an ICP monitor or
therapeutic window can be widened. In addition, threshold retrograde jugular catheter.46 Second, compromised PbtO2 is
values provide therapeutic endpoints. However, in managing common after acute brain injury even after adequate resusci­
a patient, trends over time and how the PbtO2 value relates to tation, defined as normal ICP and CPP, and an episode of
other variables such as ICP and CPP, also should be consid­ reduced PbtO2 may complicate the ICU course of up to 70%
ered. In addition, threshold is not the only factor that is of patients, again often independently of ICP and CPP.19,23,24,61
important in terms of outcome; the duration spent below Third, imaging studies obtained at admission after TBI do
threshold and the depth or severity of brain tissue hypoxia not reliably predict whether brain hypoxia will develop.62
also are important.48-50 Indeed the duration of PbtO2 less than Fourth, reduced PbtO2 is associated with other markers of
10 mm Hg is an independent factor associated with poor cellular distress such as an elevated lactate-to-pyruvate
outcome after severe TBI, and this association also is indepen­ ratio on microdialysis that can be corrected with a variety
dent of ICP.17 Threshold values can vary with probe type and of therapies.12,63 Common therapies include head position,
probe site, and so values obtained with different makes of venti­lator manipulation, CPP augmentation, and sedation
monitors (e.g., Licox vs. Neurovent vs. Neurotrend) are not that overall are successful in correcting the abnormality in
interchangeable.27,31,34 Various PbtO2 thresholds have been pro­ about 70% of episodes of reduced PbtO2.10,54,61,64-71 Fifth, PbtO2
posed.6,33,48,49,51-56 A PbtO2 less than 20 mm Hg indicates values may help guide therapeutic interventions (e.g., blood
352 Section VI—Intracranial Monitoring

transfusion),9,72-75 osmotherapy,76-79 and decompressive crani­ autoregulation curve that regulates CBF across a wide range
ectomy (DC),80-84 or identify deleterious effects of other treat­ of mean arterial pressures (MAPs).115 CPP and MAP augmen­
ments such as vasoconstriction with hyperventilation or tation can increase CBF and therefore PbtO2,76,54,66,114 whereas
shivering with induced hypothermia.69,70,85,86 In addition, the PbtO2 response to an oxygen challenge is reduced in areas
PbtO2 monitors may identify patients at risk for cerebral com­ with low CBF.116 Together this suggests that PbtO2 can provide
promise associated with hospital transport.87 Sixth, PbtO2- information about CBF and impending cerebral ischemia in
pressure reactivity may be used to assess cerebral autoregulation certain conditions. However, PbtO2 is more than a marker
and help target CPP in individual patients.88-91 Seventh, PbtO2 of ischemia and PET, and microdialysis studies suggest that
has been used for early detection of delayed cerebral ischemia hypoxia in the brain can be independent of CPP and more
(DCI) in SAH patients and to evaluate the effects of various related to diffusion rather than perfusion abnormalities.4-6,117
therapies for DCI, temporary artery occlusion during surgery, The total amount of oxygen that diffuses across the blood-
angiography, or pharmacologic angioplasty.76,92-96 In addition, brain barrier per unit time is defined as follows:
studies in SAH have shown that in some patients nimodipine
CBF × (CaO2 − CVO2 )[1] or CBF × [% SaO2 × (1.34) ×
or intra-arterial papaverine can have unexpected adverse
hemoglobin + 0.003 × (PaO2 )] − [% SvO2 ×
effects on PbtO2.97,98 Finally, several observational studies dem­
(1.34) × hemoglobin + 0.003 × (PvO2 )] 5,118
onstrate an independent association between low PbtO2 and
poor outcome in adults as well as children with TBI and in The very close association between the product of CBF and
SAH.14,15,17,21,48,49,56,99-103 Together these various observations arteriovenous oxygen tension difference and PbtO2 implies a
suggest that care based on information provided by a PbtO2 relationship between the amount of dissolved plasma oxygen
monitor may be a reasonable strategy in severe acute brain that passes through a given volume of brain per unit time and
injury. Consistent with this several observational studies or the steady-state oxygen concentration in brain tissue. Rosen­
based on historical controls, but not all, suggest that the com­ thal et al., who used regional CBF and PbtO2 monitoring,
bination of PbtO2- and ICP-based care is associated with better showed that PbtO2 may more appropriately reflect the product
outcome than just ICP-based care alone.18,51,104-107 In part this of CBF and arteriovenous oxygen tension difference (CBF ×
potential outcome benefit may be associated with better- AVTO2). This suggests that besides CBF, PaO2 is also an impor­
targeted care in each patient, determination of an “optimal” tant determinant of PbtO210,63,119 and that PbtO2 is not an isch­
CPP, and avoidance of inappropriate ICP treatment and emia monitor per se.
potential side effects if ICP is slightly elevated (≈25 mm Hg) Unlike a SjvO2 monitor, which reflects the venous oxygen
but PbtO2 is normal, a concept known as permissive intracra- content in blood that exits the brain and so indicates the
nial hypertension. At the time this book was published a mul­ balance between oxygen delivery and oxygen use, PbtO2 is
ticenter phase II trial was underway to further evaluate the more a measurement of the oxygen that accumulates in brain
role of PbtO2-based care in severe TBI. tissue. Based on the formula PbtO2 = CBF × AVTO2, reduced
PbtO2 can occur when CBF is reduced (ischemic hypoxia) and
also when there is normal CBF, for example, with impaired O2
Interpretation of Measured Values extraction due to increased gradients for oxygen diffusion,6
Information from a PbtO2 monitor, like other monitors, should cell energy crisis,4,120 or mitochondrial dysfunction121 (cyto­
be interpreted with other data such as clinical examination, pathic hypoxia). Hence PbtO2 may be considered a marker of
CT scan findings, ICP, CPP, pulmonary status, and hemoglo­ “cellular function” rather than just simply an “ischemia
bin among other variables. In addition, the PbtO2 value needs monitor,” which makes it an appropriate target for therapy in
to be temperature corrected. This is performed automatically NCCU patients.
when a brain temperature probe is placed with a Licox; if a
temperature probe is not inserted, systemic temperature can
be manually entered into the device. It also is essential to know
Safety
exactly where the monitor is placed (see earlier text). In the The Licox system is safe. Overall the incidence of device-
brain the sensor reads an integration of all small vessels in the associated contusion is less than 2%. Most of these are of no
area of the probe, and so the PbtO2 value depends on the rela­ clinical consequence and instead are only identified on
tive predominance of veins or arteries, and the number, diam­ follow-up CT.48,49,56,122-128 This incidence is similar to that
eter, and spatial distribution of the local microvasculature and observed with intraparenchymal ICP monitors and is less than
so can be influenced by changes in cerebral capillary perfu­ that associated with external ventricular drains. Catheter-
sion.108 Because the venous fraction within cortical microvas­ related infections have not been reported.48,49,56,123-127 Technical
culature is 70%, it has been suggested that PbtO2 predominantly complications such as device displacement or malfunction are
reflects venous PO2.109 However, exactly what PbtO2 measures more common and may complicate up to 10% of device inser­
is still to be fully elucidated but it likely represents (1) the tions. In addition, device removal (all three monitors and
balance between regional oxygen delivery and cellular oxygen bolt) is suggested when a magnetic resonance imaging (MRI)
consumption, (2) oxygen diffusion rather than total oxygen scan is obtained. Over time gliosis around the probe may
delivery or cerebral oxygen metabolism, or (3) oxygen that affect the reading. However, most studies suggest the measure­
accumulates in brain tissue because PET studies suggest it may ments remain accurate with little if any drift. Display errors
correlate inversely with oxygen extraction fraction can occur, but the maximal probe display error is reported to
(OEF).5,54,109,110 be 1.07 plus or minus 2.14%, tested at temperatures between
Many factors can affect PbtO2. Among these the effect of 22° C and 37° C and at oxygen pressures of 0, 44, and 150 mm
CPP and CBF has been the most studied.52,54,66 PbtO2 is associ­ Hg.122,125 This small error is unlikely to be relevant to daily
ated with regional CBF.109,111-114 This relationship follows the clinical practice.
Section VI—Intracranial Monitoring 353

Clinical Applications patients with severe TBI, SAH, and stroke (e.g., autoregula­
tion)88-90 that may help identify an individual CPP target, and
Indications for Use potentially guide several therapies including (1) CPP54,67,68,92,96;
PbtO2 monitoring has been used most frequently in severe TBI (2) induced hypertension66; (3) osmotherapy76-79,129; (4)
and poor-grade SAH. There are reports, however, of monitor­ DC80-84; (5) hyperventilation69,70,86,115,130; (6) normobaric hyper­
ing use in a variety of other conditions including in brain oxia (although its role in resuscitation remains controver­
tumors, intracerebral hemorrhage stroke, cerebral edema sial)5,36,63,71,119,131-134; (7) blood transfusion, particularly in
associated with metabolic abnormalities, and meningitis, patients with impaired cerebrovascular reserve8,135-138 since the
among others. Like ICP monitoring there is no Class I evi­ transfusion threshold used in general critical care may not
dence on which to base recommendation about use, and the apply to these patients,9,72-74 and so rather than use a hemo­
evidence is derived mainly from retrospective case-control globin (Hgb) threshold for transfusion, blood is administered
and prospective observational studies (Class III evidence). when oxygen delivery is compromised139-142; (8) fluid balance143;
Based on these studies, PbtO2 monitoring is recommended in (9) titration of sedatives including use of propofol or barbi­
patients with TBI and a GCS score less than 9, abnormal head turates for burst suppression or ICP control144-146; and
CT scan, polytrauma, and hemodynamic instability (i.e., when (10) induced normothermia.147
ICP monitoring is indicated). In patients with SAH, PbtO2
monitoring is recommended in those with a GCS score less
than 9 and in those at high risk for DCI (e.g., a patient with Subarachnoid Hemorrhage
thick SAH and intraventricular hemorrhage on admission Continuous PbtO2 monitoring can help detect DCI in SAH.
head CT scan). PbtO2 monitoring also may be considered in This can supplement information obtained by once daily tran­
patients with large middle cerebral artery stroke who are at scranial Doppler.90,148 In addition PbtO2 data can be used to
high risk for malignant brain edema. Table 35.4 summarizes guide management of DCI and evaluate autoregulation. In
some of the important applications of PbtO2 monitoring in particular, observational clinical studies suggest that induced
neurocritical care. hypertension alone, but not hypervolemia and hemodilution
(which have the opposite effect) improves PbtO2,92,93 suggesting
there is a limited if any role for “triple H” prophylaxis after
Other Monitors SAH. PbtO2 monitoring also has been used during cerebral
PbtO2 monitoring is best used with other monitors, in par­ angiography and pharmacologic angioplasty.98,124
ticular ICP and in some patients SjvO2 and a postinsertion
head CT scan. In addition, PbtO2 data supplement that derived
from cerebral microdialysis, CBF velocity measured by tran­ Use During Surgery
scranial Doppler ultrasound or regional cerebral blood flow Use of a PbtO2 monitor during aneurysm surgery is well
measurements, near infrared spectroscopy, and intracranial described.94,95,149-151 A correctly positioned PbtO2 monitor
temperature.31,50,126 allows the effects of temporary arterial occlusion to be exam­
ined; reduced PbtO2 and especially brain hypoxia indicate low
CBF and are associated with cerebral infarction.149,152 PbtO2
Intensive Care Unit Management measurements also have been used during surgery to examine
Observational data suggest PbtO2 monitoring can provide oxygenation of cerebral tissue during arteriovenous malfor­
valuable insight into the intensive care unit (ICU) care of mation (AVM)150,153 or tumor surgery.154,155 Reduced PbtO2
before AVM resection suggests low perfusion and chronic
hypoxia, whereas a marked PbtO2 increase after AVM removal
indicates hyperperfusion. The effects of inhalational agents156,157
Table 35.4  Some Clinical Applications of and propofol144,158 on cerebral autoregulation and oxygenation
PbtO2 Monitoring in Neurocritical Care during anesthesia have been examined using PbtO2 monitor­
ing. These studies show a dose-dependent loss of autoregula­
1. CPP management tion but a corresponding increase in PbtO2 provided CPP is
a. MAP target
b. Cerebral autoregulation and individual CPP target
maintained with inhalational agents, but not with propofol.
c. Induced hypertension and triple H therapy
2. ICP control PbtO2 Reactivity
a. Choice of osmotic agent (mannitol vs. hypertonic saline)
b. Timing for decompressive craniectomy The increase in PbtO2 relative to an increase in arterial PO2 is
3. Hemoglobin threshold for blood transfusion
termed brain tissue oxygen reactivity. It is believed that this
a. Treatment of anemia in patients with impaired reactivity is controlled by an oxygen regulatory mechanism,
cerebrovascular reserve (poor-grade SAH) and that this mechanism may be disturbed after brain injury.
4. Management of mechanical ventilation van Santbrink et al. examined the brain tissue oxygen response,
a. PaO2/FiO2 ratio, PEEP that is, the change in PbtO2 in response to changes in PaO2 and
b. Optimal PaCO2 target showed that a greater response in the first 24-hours post injury
was independently associated with an unfavorable outcome.159
CPP, Cerebral perfusion pressure; FiO2, inspiratory oxygen fraction;
ICP, intracranial pressure; MAP, mean arterial pressure; SAH, subarachnoid The effect of PaCO2 on this mechanism has been studied in
hemorrhage; PaCO2, arterial carbon dioxide tension; PaO2, arterial oxygen dogs160 and in pigs115; PbtO2 shows a linear correlation with
tension; PbtO2, brain tissue oxygen pressure; PEEP, positive end-expiratory
pressure.
carbon dioxide (CO2) and MAP. A linear correlation with CBF
during CO2 reactivity testing also was found. This suggests
354 Section VI—Intracranial Monitoring

that PbtO2 is influenced by factors that regulate CBF, namely therapy for NCCU patients. Future studies are needed to
CO2 and MAP. examine whether PbtO2-targeted therapy may improve the
outcome of patients with severe brain injury.
Oxygen Reactivity Index
Soehle et al.161 introduced the concept of PbtO2 autoregulation, References
defined as the ability of the brain to maintain PbtO2 despite 1. Siegemund M, van Bommel J, Ince C. Assessment of regional tissue
CPP changes. This may facilitate identification of appropriate oxygenation. Intensive Care Med 1999;25:1044–60.
individual CPP targets. Additional studies by Lang et al. 2. Coles JP. Regional ischemia after head injury. Curr Opin Crit Care 2004;10:
120–5.
showed a significant correlation between static cerebral auto­ 3. Marmarou A, Signoretti S, Fatouros P, et al. Mitochondrial injury measured
regulation (determined using blood flow velocity in relation by proton magnetic resonance spectroscopy in severe head trauma patients.
to changing CPP) and cerebral tissue oxygen reactivity (the Acta Neurochir Suppl 2005;95:149–51.
rate of PbtO2 change relative to changing CPP).91,102 This sug­ 4. Vespa P, Bergsneider M, Hattori N, et al. Metabolic crisis without brain
gests a close link between regulation of CBF and oxygenation. ischemia is common after traumatic brain injury: a combined microdialysis
and positron emission tomography study. J Cereb Blood Flow Metab 2005;
Impaired brain tissue oxygen pressure reactivity index is asso­ 25:763–74.
ciated with worse outcome after TBI,89 SAH,90 and stroke.88 5. Rosenthal G, Hemphill JC, 3rd, Sorani M, et al. Brain tissue oxygen tension
These findings suggest that manipulation of PbtO2 by altering is more indicative of oxygen diffusion than oxygen delivery and metabolism
PaO2 or CPP can be used to optimize CPP management. In in patients with traumatic brain injury. Crit Care Med 2008;36:1917–24.
6. Menon DK, Coles JP, Gupta AK, et al. Diffusion limited oxygen delivery
patients with acute ischemic stroke, impaired CPP-brain tissue following head injury. Crit Care Med 2004;32:1384–90.
oxygen reactivity index predicts the development of malig­ 7. Coles JP, Cunningham AS, Salvador R, et al. Early metabolic characteristics
nant edema after middle cerebral artery infarction.88 of lesion and nonlesion tissue after head injury. J Cereb Blood Flow Metab
2009;29(5):965–75.
8. Hare GM. Anaemia and the brain. Curr Opin Anaesthesiol 2004;17:363–9.
Outcome Prediction 9. Oddo M, Milby A, Chen I, et al. Hemoglobin concentration and cerebral
metabolism in patients with aneurysmal subarachnoid hemorrhage. Stroke
The association between reduced PbtO2 or brain hypoxia, 2009;40(4):1275–81.
including the duration and the depth or severity of brain 10. Rosenthal G, Hemphill JC, Sorani M, et al. The role of lung function in
tissue hypoxia, and poor outcome in TBI has been well brain tissue oxygenation following traumatic brain injury. J Neurosurg 2008;
108:59–65.
described in several observational studies, although the 11. Hoffman WE, Charbel FT, Edelman G. Brain tissue oxygen, carbon dioxide,
strength of this relationship may depend in part on probe and pH in neurosurgical patients at risk for ischemia. Anesth Analg 1996;
location relative to other pathology.* This includes an associa­ 82(3):582–6.
tion between low PbtO2 and long-term neuropsychological 12. Hlatky R, Valadka AB, Goodman JC, et al. Patterns of energy substrates
performance.162 Importantly, episodes of brain tissue hypoxia during ischemia measured in the brain by microdialysis. J Neurotrauma
2004;21(7):894–906.
may occur when CPP and ICP are normal,23,50,60,103 thus 13. Hlatky R, Valadka AB, Goodman JC, et al. Evolution of brain tissue injury
emphasizing the value of using both monitors. Reduced PbtO2 after evacuation of acute traumatic subdural hematomas. Neurosurgery
also is associated with outcome in SAH, although the relation­ 2007;61(Suppl 1):249–54; discussion 254–5.
ship is less robust than in TBI.148,163 This association between 14. Maloney-Wilensky E, Gracias V, Itkin A, et al. Brain tissue oxygen and
outcome after severe traumatic brain injury: a systematic review. Crit Care
outcome and PbtO2 has led to the concept of PbtO2-based care. Med 2009;37(6):2057–63. Review.
Some but not all observational series suggest that the addition 15. Chang JJ, Youn TS, Benson D, et al. Physiologic and functional outcome
of PbtO2-based care to conventional ICP- and CPP-based care correlates of brain tissue hypoxia in traumatic brain injury. Crit Care Med
is associated with improved outcome after severe TBI.18,51,104- 2009;37(1):283–90.
107,164,165
At publication, this question was being evaluated in a 16. Friess SH, Ralston J, Eucker SA, et al. Neurocritical care monitoring
correlates with neuropathology in a swine model of pediatric traumatic
multicenter phase II clinical trial. brain injury. Neurosurgery 2011;69(5):1139–47; discussion 1147.
17. Oddo M, Levine JM, Mackenzie L, et al. Brain hypoxia is associated with
Conclusion short-term outcome after severe traumatic brain injury independently of
intracranial hypertension and low cerebral perfusion pressure. Neurosurgery
2011;69(5):1037–45; discussion 1045.
Monitoring of brain tissue oxygen partial pressure is a safe and 18. Nangunoori R, Maloney-Wilensky E, Stiefel MD, et al. Brain tissue oxygen
reliable technique that allows bedside evaluation of cellular based therapy and outcome after severe traumatic brain injury: a systematic
function in patients with severe brain injury. PbtO2 depends literature review. Neurocrit Care 2011;Aug 16. Epub ahead of print.
on CBF, arteriovenous oxygen tension difference, Hgb concen­ 19. Eriksson EA, Barletta JF, Figueroa BE, et al. Cerebral perfusion pressure and
tration, and systemic oxygenation among other variables. Low intracranial pressure are not surrogates for brain tissue oxygenation in
traumatic brain injury. Clin Neurophysiol 2011;Nov 18. Epub ahead of print.
PbtO2 is frequently observed after acute brain injury and can 20. Feng JF, Zhao X, Gurkoff GG, et al. Post-traumatic hypoxia exacerbates
result from several pathologic mechanisms including isch­ neuronal cell death in the hippocampus. J Neurotrauma 2012;Jan 30. Epub
emia, impaired cell oxygen extraction, anemia, and reduced ahead of print.
systemic oxygenation, among others. PbtO2 monitoring helps 21. Rohlwink UK, Zwane E, Graham Fieggen A, et al. The relationship between
intracranial pressure and brain oxygenation in children with severe
optimize CPP, PaCO2, PaO2, and hemoglobin targets, and traumatic brain injury. Neurosurgery 2012;70(5):1220–31.
guide management of elevated ICP. This can help avoid delete­ 22. Bhatia A, Gupta AK. Neuromonitoring in the intensive care unit. II. Cerebral
rious side effects of treatments and reduce the extent of brain oxygenation monitoring and microdialysis. Intensive Care Med 2007;33:
tissue hypoxia, a variable that is associated with poor outcome. 1322–8.
As such, PbtO2 monitoring can complement the use of existing 23. Stiefel MF, Udoetuk JD, Spiotta AM, et al. Conventional neurocritical care
and cerebral oxygenation after traumatic brain injury. J Neurosurg 2006;105:
intracranial monitors and help target and individualize 568–75.
24. Gracias VH, Guillamondegui OD, Stiefel, MF, et al. Cerebral cortical
*References 14, 17, 21, 34, 48, 49, 56, 99, 101, 103, 130. oxygenation: a pilot study. J Trauma 2004;56: 469–74.
Section VI—Intracranial Monitoring 355

25. Stewart C, Haitsma I, Zador Z, et al. The new Licox combined brain tissue 38. Gopinath SP, Valadka A, Contant CF, et al. Relationship between global and
oxygen and brain temperature monitor: assessment of in vitro accuracy and cortical cerebral blood flow in patients with head injuries. Neurosurgery
clinical experience in severe traumatic brain injury. Neurosurgery 2008;63: 1999;44:1273–8; discussion 1278–9.
1159–64; discussion 1164–5. 39. Hoffman WE, Charbel FT, Edelman G, et al. Brain tissue oxygen pressure,
26. Orakcioglu B, Sakowitz OW, Neumann JO, et al. Evaluation of a novel brain carbon dioxide pressure and pH during ischemia. Neurol Res 1996;18:
tissue oxygenation probe in an experimental swine model. Neurosurgery 54–6.
2010;67(6):1716–22; discussion 1722–3. 40. Sarrafzadeh AS, Kiening KL, Bardt TF, et al. Cerebral oxygenation in
27. Dengler J, Frenzel C, Vajkoczy P, et al. Cerebral tissue oxygenation measured contusioned vs. nonlesioned brain tissue: monitoring of PtiO2 with Licox
by two different probes: challenges and interpretation. Intensive Care Med and Paratrend. Acta Neurochir Suppl 1998;71:186–9.
2011;37:1809–15. 41. Critchley GR, Bell BA. Acute cerebral tissue oxygenation changes following
28. Dengler J, Frenzel C, Vajkoczy P, et al. The oxygen reactivity index and its experimental subarachnoid hemorrhage. Neurol Res 2003;25:451–6.
relation to sensor technology in patients with severe brain lesions. Neurocrit 42. Zauner A, Bullock R, Di X, et al. Brain oxygen, CO2, pH, and temperature
Care 2012;Mar 7. Epub ahead of print. monitoring: evaluation in the feline brain. Neurosurgery 1995;37:1168–76;
29. Hutchinson PJ, Hutchinson DB, Barr RH, et al. A new cranial access device discussion 1176–7.
for cerebral monitoring. Br J Neurosurg 2000;14:46–8. 43. Hoffman WE, Charbel FT, Edelman G. Brain tissue oxygen, carbon dioxide,
30. Jaeger M, Soehle M, Meixensberger J. Brain tissue oxygen (PtiO2): a clinical and pH in neurosurgical patients at risk for ischemia. Anesth Analg
comparison of two monitoring devices. Acta Neurochir Suppl 2005;95: 1996;82:582–6.
79–81. 44. Hoffman WE, Charbel FT, Edelman G, et al. Brain tissue oxygen pressure,
31. Maloney-Wilensky E, Le Roux P. The physiology behind direct brain oxygen carbon dioxide pressure, and pH during hypothermic circulatory arrest. Surg
monitors and practical aspects of their use. Childs Nerv Syst Neurol 1996;46:75–9.
2010;26(4):419–30. 45. Meixensberger J, Dings J, Kuhnigk H, et al. Studies of tissue PO2 in normal
32. Kiening KL, Unterberg AW, Bardt TF, et al. Monitoring of cerebral and pathological human brain cortex. Acta Neurochir Suppl (Wien) 1993;59:
oxygenation in patients with severe head injuries: brain tissue PO2 versus 58–63.
jugular vein oxygen saturation. J Neurosurg 1996;85:751–7. 46. Pennings FA, Schuurman PR, van den Munckhof P, et al. Brain tissue oxygen
33. Longhi L, Pagan F, Valeriani V, et al. Monitoring brain tissue oxygen tension pressure monitoring in awake patients during functional neurosurgery: the
in brain-injured patients reveals hypoxic episodes in normal-appearing and assessment of normal values. J Neurotrauma 2008;25:1173–7.
in peri-focal tissue. Intensive Care Med 2007;33:2136–42. 47. Dings J, Meixensberger J, Amschler J, et al. Brain tissue pO2 in relation to
34. Ponce LL, Pillai S, Cruz J, et al. Position of probe determines prognostic cerebral perfusion pressure, TCD findings and TCD-CO2-reactivity after
information of brain tissue pO2 in severe traumatic brain injury. severe head injury. Acta Neurochir (Wien) 1996;138:425–34.
Neurosurgery 2012;Jan 27. Epub ahead of print. 48. Valadka AB, Gopinath SP, Contant CF, et al. Relationship of brain tissue PO2
35. Scheufler KM. Tissue oxygenation and capacity to deliver O2: do the two go to outcome after severe head injury. Crit Care Med 1998;26:1576–81.
together? Transfus Apher Sci 2004;31:45–54. 49. van den Brink WA, van Santbrink H, Steyerberg EW, et al. Brain oxygen
36. Hlatky R, Valadka AB, Gopinath SP, et al. Brain tissue oxygen tension tension in severe head injury. Neurosurgery 2000;46:868–76; discussion
response to induced hyperoxia reduced in hypoperfused brain. J Neurosurg 876–8.
2008;108(1):53–8. 50. Oddo M, Levine JM, Frangos S, et al. Brain lactate metabolism in humans
37. Gupta AK, Hutchinson PJ, Al-Rawi P, et al. Measuring brain tissue with subarachnoid hemorrhage. Stroke 2012;43(5):1418–21.
oxygenation compared with jugular venous oxygen saturation for
monitoring cerebral oxygenation after traumatic brain injury. Anesth Analg A complete list of references for this chapter can be found online at
1999;88:549–53. www.expertconsult.com.
Section VI—Intracranial Monitoring 355.e1

References 26. Orakcioglu B, Sakowitz OW, Neumann JO, et al. Evaluation of a novel
brain tissue oxygenation probe in an experimental swine model.
1. Siegemund M, van Bommel J, Ince C. Assessment of regional tissue Neurosurgery 2010;67(6):1716–22; discussion 1722–3.
oxygenation. Intensive Care Med 1999;25:1044–60. 27. Dengler J, Frenzel C, Vajkoczy P, et al. Cerebral tissue oxygenation
2. Coles JP. Regional ischemia after head injury. Curr Opin Crit Care 2004;10: measured by two different probes: challenges and interpretation. Intensive
120–5. Care Med 2011;37:1809–15.
3. Marmarou A, Signoretti S, Fatouros P, et al. Mitochondrial injury measured 28. Dengler J, Frenzel C, Vajkoczy P, et al. The oxygen reactivity index and its
by proton magnetic resonance spectroscopy in severe head trauma patients. relation to sensor technology in patients with severe brain lesions.
Acta Neurochir Suppl 2005;95:149–51. Neurocrit Care 2012;Mar 7. Epub ahead of print.
4. Vespa P, Bergsneider M, Hattori N, et al. Metabolic crisis without brain 29. Hutchinson PJ, Hutchinson DB, Barr RH, et al. A new cranial access device
ischemia is common after traumatic brain injury: a combined microdialysis for cerebral monitoring. Br J Neurosurg 2000;14:46–8.
and positron emission tomography study. J Cereb Blood Flow Metab 2005; 30. Jaeger M, Soehle M, Meixensberger J. Brain tissue oxygen (PtiO2): a clinical
25:763–74. comparison of two monitoring devices. Acta Neurochir Suppl 2005;95:
5. Rosenthal G, Hemphill JC, 3rd, Sorani M, et al. Brain tissue oxygen 79–81.
tension is more indicative of oxygen diffusion than oxygen delivery and 31. Maloney-Wilensky E, Le Roux P. The physiology behind direct brain
metabolism in patients with traumatic brain injury. Crit Care Med 2008;36: oxygen monitors and practical aspects of their use. Childs Nerv Syst 2010;
1917–24. 26(4):419–30.
6. Menon DK, Coles JP, Gupta AK, et al. Diffusion limited oxygen delivery 32. Kiening KL, Unterberg AW, Bardt TF, et al. Monitoring of cerebral
following head injury. Crit Care Med 2004;32:1384–90. oxygenation in patients with severe head injuries: brain tissue PO2 versus
7. Coles JP, Cunningham AS, Salvador R, et al. Early metabolic characteristics jugular vein oxygen saturation. J Neurosurg 1996;85:751–7.
of lesion and nonlesion tissue after head injury. J Cereb Blood Flow Metab 33. Longhi L, Pagan F, Valeriani V, et al. Monitoring brain tissue oxygen tension
2009;29(5):965–75. in brain-injured patients reveals hypoxic episodes in normal-appearing and
8. Hare GM. Anaemia and the brain. Curr Opin Anaesthesiol 2004;17:363–9. in peri-focal tissue. Intensive Care Med 2007;33:2136–42.
9. Oddo M, Milby A, Chen I, et al. Hemoglobin concentration and cerebral 34. Ponce LL, Pillai S, Cruz J, et al. Position of probe determines prognostic
metabolism in patients with aneurysmal subarachnoid hemorrhage. Stroke information of brain tissue pO2 in severe traumatic brain injury.
2009;40(4):1275–81. Neurosurgery 2012;Jan 27. Epub ahead of print.
10. Rosenthal G, Hemphill JC, Sorani M, et al. The role of lung function in 35. Scheufler KM. Tissue oxygenation and capacity to deliver O2: do the two go
brain tissue oxygenation following traumatic brain injury. J Neurosurg together? Transfus Apher Sci 2004;31:45–54.
2008;108:59–65. 36. Hlatky R, Valadka AB, Gopinath SP, et al. Brain tissue oxygen tension
11. Hoffman WE, Charbel FT, Edelman G. Brain tissue oxygen, carbon dioxide, response to induced hyperoxia reduced in hypoperfused brain. J Neurosurg
and pH in neurosurgical patients at risk for ischemia. Anesth Analg 1996; 2008;108(1):53–8.
82(3):582–6. 37. Gupta AK, Hutchinson PJ, Al-Rawi P, et al. Measuring brain tissue
12. Hlatky R, Valadka AB, Goodman JC, et al. Patterns of energy substrates oxygenation compared with jugular venous oxygen saturation for
during ischemia measured in the brain by microdialysis. J Neurotrauma monitoring cerebral oxygenation after traumatic brain injury. Anesth Analg
2004;21(7):894–906. 1999;88:549–53.
13. Hlatky R, Valadka AB, Goodman JC, et al. Evolution of brain tissue injury 38. Gopinath SP, Valadka A, Contant CF, et al. Relationship between global and
after evacuation of acute traumatic subdural hematomas. Neurosurgery cortical cerebral blood flow in patients with head injuries. Neurosurgery
2007;61(Suppl 1):249–54; discussion 254–5. 1999;44:1273–8; discussion 1278–9.
14. Maloney-Wilensky E, Gracias V, Itkin A, et al. Brain tissue oxygen and 39. Hoffman WE, Charbel FT, Edelman G, et al. Brain tissue oxygen pressure,
outcome after severe traumatic brain injury: a systematic review. Crit Care carbon dioxide pressure and pH during ischemia. Neurol Res 1996;18:
Med 2009;37(6):2057–63. Review. 54–6.
15. Chang JJ, Youn TS, Benson D, et al. Physiologic and functional outcome 40. Sarrafzadeh AS, Kiening KL, Bardt TF, et al. Cerebral oxygenation in
correlates of brain tissue hypoxia in traumatic brain injury. Crit Care Med contusioned vs. nonlesioned brain tissue: monitoring of PtiO2 with Licox
2009;37(1):283–90. and Paratrend. Acta Neurochir Suppl 1998;71:186–9.
16. Friess SH, Ralston J, Eucker SA, et al. Neurocritical care monitoring 41. Critchley GR, Bell BA. Acute cerebral tissue oxygenation changes
correlates with neuropathology in a swine model of pediatric traumatic following experimental subarachnoid hemorrhage. Neurol Res 2003;25:
brain injury. Neurosurgery 2011;69(5):1139–47; discussion 1147. 451–6.
17. Oddo M, Levine JM, Mackenzie L, et al. Brain hypoxia is associated with 42. Zauner A, Bullock R, Di X, et al. Brain oxygen, CO2, pH, and temperature
short-term outcome after severe traumatic brain injury independently of monitoring: evaluation in the feline brain. Neurosurgery 1995;37:1168–76;
intracranial hypertension and low cerebral perfusion pressure. discussion 1176–7.
Neurosurgery 2011;69(5):1037–45; discussion 1045. 43. Hoffman WE, Charbel FT, Edelman G. Brain tissue oxygen, carbon dioxide,
18. Nangunoori R, Maloney-Wilensky E, Stiefel MD, et al. Brain tissue oxygen and pH in neurosurgical patients at risk for ischemia. Anesth Analg
based therapy and outcome after severe traumatic brain injury: a systematic 1996;82:582–6.
literature review. Neurocrit Care 2011;Aug 16. Epub ahead of print. 44. Hoffman WE, Charbel FT, Edelman G, et al. Brain tissue oxygen pressure,
19. Eriksson EA, Barletta JF, Figueroa BE, et al. Cerebral perfusion pressure and carbon dioxide pressure, and pH during hypothermic circulatory arrest.
intracranial pressure are not surrogates for brain tissue oxygenation in Surg Neurol 1996;46:75–9.
traumatic brain injury. Clin Neurophysiol 2011;Nov 18. Epub ahead of 45. Meixensberger J, Dings J, Kuhnigk H, et al. Studies of tissue PO2 in normal
print. and pathological human brain cortex. Acta Neurochir Suppl (Wien) 1993;
20. Feng JF, Zhao X, Gurkoff GG, et al. Post-traumatic hypoxia exacerbates 59:58–63.
neuronal cell death in the hippocampus. J Neurotrauma 2012;Jan 30. Epub 46. Pennings FA, Schuurman PR, van den Munckhof P, et al. Brain tissue
ahead of print. oxygen pressure monitoring in awake patients during functional
21. Rohlwink UK, Zwane E, Graham Fieggen A, et al. The relationship between neurosurgery: the assessment of normal values. J Neurotrauma 2008;25:
intracranial pressure and brain oxygenation in children with severe 1173–7.
traumatic brain injury. Neurosurgery 2012;70(5):1220–31. 47. Dings J, Meixensberger J, Amschler J, et al. Brain tissue pO2 in relation to
22. Bhatia A, Gupta AK. Neuromonitoring in the intensive care unit. II. cerebral perfusion pressure, TCD findings and TCD-CO2-reactivity after
Cerebral oxygenation monitoring and microdialysis. Intensive Care Med severe head injury. Acta Neurochir (Wien) 1996;138:425–34.
2007;33:1322–8. 48. Valadka AB, Gopinath SP, Contant CF, et al. Relationship of brain tissue
23. Stiefel MF, Udoetuk JD, Spiotta AM, et al. Conventional neurocritical care PO2 to outcome after severe head injury. Crit Care Med 1998;26:1576–81.
and cerebral oxygenation after traumatic brain injury. J Neurosurg 2006; 49. van den Brink WA, van Santbrink H, Steyerberg EW, et al. Brain oxygen
105:568–75. tension in severe head injury. Neurosurgery 2000;46:868–76; discussion
24. Gracias VH, Guillamondegui OD, Stiefel, MF, et al. Cerebral cortical 876–8.
oxygenation: a pilot study. J Trauma 2004;56: 469–74. 50. Oddo M, Levine JM, Frangos S, et al. Brain lactate metabolism in humans
25. Stewart C, Haitsma I, Zador Z, et al. The new Licox combined brain tissue with subarachnoid hemorrhage. Stroke 2012;43(5):1418–21.
oxygen and brain temperature monitor: assessment of in vitro accuracy and 51. Stiefel MF, Spiotta A, Gracias VH, et al. Reduced mortality rate in patients
clinical experience in severe traumatic brain injury. Neurosurgery with severe traumatic brain injury treated with brain tissue oxygen
2008;63:1159–64; discussion 1164–5. monitoring. J Neurosurg 2005;103:805–11.
355.e2 Section VI—Intracranial Monitoring

52. Doppenberg EM, Zauner A, Bullock R, et al. Correlations between brain 76. Al-Rawi PG, Zygun D, Tseng MY, et al. Cerebral blood flow augmentation
tissue oxygen tension, carbon dioxide tension, pH, and cerebral blood in patients with severe subarachnoid haemorrhage. Acta Neurochir Suppl
flow: a better way of monitoring the severely injured brain? Surg Neurol 2005;95:123–7.
1998;49:650–4. 77. Francony G, Fauvage B, Falcon D, et al. Equimolar doses of mannitol and
53. Doppenberg EM, Zauner A, Watson JC, et al. Determination of the hypertonic saline in the treatment of increased intracranial pressure. Crit
ischemic threshold for brain oxygen tension. Acta Neurochir Suppl Care Med 2008;36(3):795–800.
1998;71:166–9. 78. Oddo M, Levine JM, Frangos S, et al. Effect of mannitol and hypertonic
54. Johnston AJ, Steiner LA, Coles JP, et al. Effect of cerebral perfusion pressure saline on cerebral oxygenation in patients with severe traumatic brain
augmentation on regional oxygenation and metabolism after head injury. injury and refractory intracranial hypertension. J Neurol Neurosurg
Crit Care Med 2005;33:189–95; discussion 255–7. Psychiatry 2009;80(8):916–20.
55. Zauner A, Doppenberg EM, Woodward JJ, et al. Continuous monitoring of 79. Sakowitz OW, Stover JF, Sarrafzadeh AS, et al. Effects of mannitol bolus
cerebral substrate delivery and clearance: initial experience in 24 patients administration on intracranial pressure, cerebral extracellular metabolites,
with severe acute brain injuries. Neurosurgery 1997;41:1082–91; discussion and tissue oxygenation in severely head-injured patients. J Trauma 2007;
1091–3. 62:292–8.
56. van Santbrink H, Maas AI, Avezaat CJ. Continuous monitoring of partial 80. Weiner GM, Lacey MR, Mackenzie L, et al. Decompressive craniectomy for
pressure of brain tissue oxygen in patients with severe head injury. elevated intracranial pressure and its effect on the cumulative ischemic
Neurosurgery 1996;38:21–31. burden and therapeutic intensity levels after severe traumatic brain injury.
57. Bratton SL, Chestnut RM, Ghajar J, et al. Guidelines for the management of Neurosurgery 2010;66:1111–19.
severe traumatic brain injury. X. Brain oxygen monitoring and thresholds. 81. Strege RJ, Lang EW, Stark AM, et al. Cerebral edema leading to
J Neurotrauma 2007;24(Suppl 1):S65–70. decompressive craniectomy: an assessment of the preceding clinical and
58. Smith ML, Counelis GJ, Maloney-Wilensky E, et al. Brain tissue oxygen neuromonitoring trends. Neurol Res 2003;25:510–5.
tension in clinical brain death: a case series. Neurol Res 2007;29:755–9. 82. Stiefel MF, Heuer GG, Smith MJ, et al. Cerebral oxygenation following
59. Figaji AA, Kent SJ. Brain tissue oxygenation in children diagnosed with decompressive hemicraniectomy for the treatment of refractory intracranial
brain death. Neurocrit Care 2010;12(1):56–61. hypertension. J Neurosurg 2004;101:241–7.
60. Chen HI, Stiefel MF, Oddo M, et al. Detection of cerebral compromise with 83. Jaeger M, Soehle M, Meixensberger J. Effects of decompressive craniectomy
multimodality monitoring in patients with subarachnoid hemorrhage. on brain tissue oxygen in patients with intracranial hypertension. J Neurol
Neurosurgery 2011;69(1):53–63; discussion 63. Neurosurg Psychiatry 2003;74:513–5.
61. Bohman LE, Heuer GG, Macyszyn L, et al. Medical management of 84. Ho CL, Wang CM, Lee KK, et al. Cerebral oxygenation, vascular reactivity,
compromised brain oxygen in patients with severe traumatic brain injury. and neurochemistry following decompressive craniectomy for severe
Neurocrit Care 2011;14(3):361–9. traumatic brain injury. J Neurosurg 2008;108:943–9.
62. Katsnelson M, Mackenzie L, Frangos S, et al. Are initial radiographic and 85. Oddo M, Frangos S, Maloney-Wilensky E, et al. Effect of shivering on brain
clinical scales associated with subsequent intracranial pressure and brain tissue oxygenation during induced normothermia in patients with severe
oxygen levels after severe traumatic brain injury? Neurosurgery 2012;70(5): brain injury. Neurocrit Care 2010;12(1):10–6.
1095–105. 86. Coles JP, Minhas PS, Fryer TD, et al. Effect of hyperventilation on cerebral
63. Tolias CM, Reinert M, Seiler R, et al. Normobaric hyperoxia—induced blood flow in traumatic head injury: clinical relevance and monitoring
improvement in cerebral metabolism and reduction in intracranial pressure correlates. Crit Care Med 2002;30:1950–9.
in patients with severe head injury: a prospective historical cohort-matched 87. Swanson E, Mascitelli J, Stiefel M, et al. The effect of patient
study. J Neurosurg 2004;101:435–44. transport on brain oxygen in comatose patients. Neurosurgery 66:
64. Ledwith MB, Bloom S, Maloney-Wilensky E, et al. Effect of body position 925–32, 2010.
on cerebral oxygenation and physiologic parameters in patients with acute 88. Dohmen C, Bosche B, Graf R, et al. Identification and clinical impact of
neurological conditions. J Neurosci Nurs 2010;42(5):280–7. impaired cerebrovascular autoregulation in patients with malignant middle
65. Pascual JL, Georgoff P, Maloney-Wilensky E, et al. Reduced brain tissue cerebral artery infarction. Stroke 2007;38:56–61.
oxygen in traumatic brain injury: are most commonly used interventions 89. Jaeger M, Schuhmann MU, Soehle M, et al. Continuous assessment of
successful? J Trauma 2011;70(3):535–46. cerebrovascular autoregulation after traumatic brain injury using brain
66. Johnston AJ, Steiner LA, Chatfield DA, et al. Effect of cerebral perfusion tissue oxygen pressure reactivity. Crit Care Med 2006;34:1783–8.
pressure augmentation with dopamine and norepinephrine on global and 90. Jaeger M, Schuhmann MU, Soehle M, et al. Continuous monitoring of
focal brain oxygenation after traumatic brain injury. Intensive Care Med cerebrovascular autoregulation after subarachnoid hemorrhage by brain
2004;30:791–7. tissue oxygen pressure reactivity and its relation to delayed cerebral
67. Marin-Caballos AJ, Murillo-Cabezas F, Cayuela-Dominguez A, et al. infarction. Stroke 2007;38:981–6.
Cerebral perfusion pressure and risk of brain hypoxia in severe head injury: 91. Lang EW, Czosnyka M, Mehdorn HM. Tissue oxygen reactivity and cerebral
a prospective observational study. Crit Care 2005;9:R670–6. autoregulation after severe traumatic brain injury. Crit Care Med 2003;31:
68. Stocchetti N, Chieregato A, De Marchi M, et al. High cerebral perfusion 267–71.
pressure improves low values of local brain tissue O2 tension (PtiO2) in 92. Muench E, Horn P, Bauhuf C, et al. Effects of hypervolemia and
focal lesions. Acta Neurochir Suppl 1998;71:162–5. hypertension on regional cerebral blood flow, intracranial pressure, and
69. Clausen T, Scharf A, Menzel M, et al. Influence of moderate and profound brain tissue oxygenation after subarachnoid hemorrhage. Crit Care Med
hyperventilation on cerebral blood flow, oxygenation and metabolism. 2007;35:1844–51; quiz 52.
Brain Res 2004;1019:113–23. 93. Ekelund A, Reinstrup P, Ryding E, et al. Effects of iso- and hypervolemic
70. Carmona Suazo JA, Maas AI, van den Brink WA, et al. CO2 reactivity and hemodilution on regional cerebral blood flow and oxygen delivery for
brain oxygen pressure monitoring in severe head injury. Crit Care Med patients with vasospasm after aneurysmal subarachnoid hemorrhage. Acta
2000;28:3268–74. Neurochir (Wien) 2002;144:703–12; discussion 712–3.
71. Menzel M, Doppenberg EM, Zauner A, et al. Increased inspired oxygen 94. Kett-White R, Hutchinson PJ, Al-Rawi PG, et al. Cerebral oxygen and
concentration as a factor in improved brain tissue oxygenation and microdialysis monitoring during aneurysm surgery: effects of blood
tissue lactate levels after severe human head injury. J Neurosurg 1999;91: pressure, cerebrospinal fluid drainage, and temporary clipping on
1–10. infarction. J Neurosurg 2002;96:1013–9.
72. Leal-Noval SR, Rincon-Ferrari MD, Marin-Niebla A, et al. Transfusion of 95. Kett-White R, Hutchinson PJ, Czosnyka M, et al. Effects of variation in
erythrocyte concentrates produces a variable increment on cerebral cerebral haemodynamics during aneurysm surgery on brain tissue oxygen
oxygenation in patients with severe traumatic brain injury: a preliminary and metabolism. Acta Neurochir Suppl 2002;81:327–9.
study. Intensive Care Med 2006;32:1733–40. 96. Dudkiewicz M, Proctor KG. Tissue oxygenation during management of
73. Smith MJ, Stiefel MF, Magge S, et al. Packed red blood cell transfusion cerebral perfusion pressure with phenylephrine or vasopressin. Crit Care
increases local cerebral oxygenation. Crit Care Med 2005;33:1104–8. Med 2008;36:2641–50.
74. Zygun DA, Nortje J, Hutchinson PJ, et al. The effect of red blood cell 97. Stiefel MF, Heuer GG, Abrahams JM, et al. The effect of nimodipine on
transfusion on cerebral oxygenation and metabolism after severe traumatic cerebral oxygenation in patients with poor-grade subarachnoid
brain injury. Crit Care Med 2009;37:1074–8. hemorrhage. J Neurosurg 2004;101:594–9.
75. Figaji AA, Zwane E, Kogels M, et al. The effect of blood transfusion on 98. Stiefel MF, Spiotta A, Udoetek J, et al. Intra-arterial papaverine used to
brain oxygenation in children with severe traumatic brain injury. Pediatr treat cerebral vasospasm reduces brain oxygen. Neurocritical Care 2006;4:
Crit Care Med 2010;11(3):325–31. 113–8.
Section VI—Intracranial Monitoring 355.e3

99. Figaji AA, Zwane E, Thompson C, et al. Brain tissue oxygen tension 124. Carvi y Nievas M, Toktamis S, Hollerhage HG, et al. Hyperacute
monitoring in pediatric severe traumatic brain injury. part 1: relationship measurement of brain-tissue oxygen, carbon dioxide, pH, and intracranial
with outcome. Childs Nerv Syst 2009;25(10):1325–33. pressure before, during, and after cerebral angiography in patients with
100. Ramakrishna R, Stiefel M, Udoteuk J, et al. Brain oxygen and outcome in aneurysmatic subarachnoid hemorrhage in poor condition. Surg Neurol
patients with aneurysmal subarachnoid hemorrhage. J Neurosurgery 2005;64:362–7; discussion 367.
2008;109(6):1075–82. 125. Dings J, Meixensberger J, Roosen K. Brain tissue pO2-monitoring: catheter
101. Dings J, Jager A, Meixensberger J, et al. Brain tissue pO2 and outcome after stability and complications. Neurol Res 1997;19:241–5.
severe head injury. Neurol Res 1998;20(Suppl 1):S71–5. 126. Lang EW, Mulvey JM, Mudaliar Y, et al. Direct cerebral oxygenation
102. Ang BT, Wong J, Lee KK, et al. Temporal changes in cerebral tissue monitoring—a systematic review of recent publications. Neurosurg Rev
oxygenation with cerebrovascular pressure reactivity in severe traumatic 2007;30:99–106; discussion 107.
brain injury. J Neurol Neurosurg Psychiatry 2007;78:298–302. 127. Nortje J, Gupta AK. The role of tissue oxygen monitoring in patients with
103. Chang JJ, Youn TS, Benson D, et al. Physiologic and functional outcome acute brain injury. Br J Anaesth 2006;97:95–106.
correlates of brain tissue hypoxia in traumatic brain injury. Crit Care Med 128. Bailey RL, Quattrone F, Curtain C, et al. The safety of multimodal
2009;37:283–90. monitoring in severe brain injury. Montreal, Canada, Neurocritical Care
104. Spiotta AM, Stiefel MF, Gracias VH, et al. Brain tissue oxygen-directed Society Meeting, 2011.
management and outcome in patients with severe traumatic brain injury. 129. Hartl R, Bardt TF, Kiening KL, et al. Mannitol decreases ICP but does not
J Neurosurg 2010;113(3):571–80. improve brain-tissue pO2 in severely head-injured patients with
105. Narotam PK, Morrison JF, Nathoo N. Brain tissue oxygen monitoring in intracranial hypertension. Acta Neurochir Suppl 1997;70:40–2.
traumatic brain injury and major trauma: outcome analysis of a brain 130. Sarrafzadeh AS, Kiening KL, Callsen TA, et al. Metabolic changes during
tissue oxygen-directed therapy. J Neurosurg 2009;111(4):672–82. impending and manifest cerebral hypoxia in traumatic brain injury. Br J
106. Martini RP, Deem S, Yanez ND, et al. Management guided by brain tissue Neurosurg 2003;17:340–6.
oxygen monitoring and outcome following severe traumatic brain injury. 131. Bullock MR. Hyperoxia: good or bad? J Neurosurg 2003;98:943–4;
J Neurosurg 2009;111(4):644–9. discussion 944.
107. Meixensberger J, Jaeger M, Vath A, et al. Brain tissue oxygen guided 132. Diringer MN. Hyperoxia: good or bad for the injured brain? Curr Opin
treatment supplementing ICP/CPP therapy after traumatic brain injury. Crit Care 2008;14:167–71.
J Neurol Neurosurg Psychiatry 2003;74:760–4. 133. Diringer MN, Aiyagari V, Zazulia AR, et al. Effect of hyperoxia on cerebral
108. Haitsma IK, Maas AI. Advanced monitoring in the intensive care unit: metabolic rate for oxygen measured using positron emission tomography
brain tissue oxygen tension. Curr Opin Crit Care 2002;8:115-20. in patients with acute severe head injury. J Neurosurg 2007;106:526–9.
109. Scheufler KM, Rohrborn HJ, Zentner J. Does tissue oxygen-tension reliably 134. Longhi L, Stocchetti N. Hyperoxia in head injury: therapeutic tool? Curr
reflect cerebral oxygen delivery and consumption? Anesth Analg 2002;95: Opin Crit Care 2004;10:105–9.
1042–8. 135. Borgstrom L, Johannsson H, Siesjo BK. The influence of acute
110. Scheufler K-M, Lehnert A, Rohrborn H-J, et al: Individual values of brain normovolemic anemia on cerebral blood flow and oxygen consumption of
tissue oxygen pressure, microvascular oxygen saturation, cytochrome redox anesthetized rats. Acta Physiol Scand 1975;93:505–14.
level and energy metabolites in detecting critically reduced cerebral energy 136. Dexter F, Hindman BJ. Effect of haemoglobin concentration on brain
state during acute changes in global cerebral perfusion. J Neurosurg oxygenation in focal stroke: a mathematical modelling study. Br J Anaesth
Anesthesiol 2004; 16:210–19. 1997;79:346–51.
111. Hemphill JC, 3rd, Smith WS, Sonne DC, et al. Relationship between brain 137. Hare GM, Mazer CD, Hutchison JS, et al. Severe hemodilutional anemia
tissue oxygen tension and CT perfusion: feasibility and initial results. AJNR increases cerebral tissue injury following acute neurotrauma. J Appl Physiol
Am J Neuroradiol 2005;26:1095–100. 2007;103:1021–9.
112. Jaeger M, Soehle M, Schuhmann MU, et al. Correlation of continuously 138. Hare GM, Mazer CD, Mak W, et al. Hemodilutional anemia is associated
monitored regional cerebral blood flow and brain tissue oxygen. Acta with increased cerebral neuronal nitric oxide synthase gene expression.
Neurochir (Wien) 2005;147:51–6; discussion 56. J Appl Physiol 2003;94:2058–67.
113. Menzel M, Doppenberg EM, Zauner A, et al. Cerebral oxygenation in 139. Kramer A, Le Roux P. Red blood cell transfusion and transfusion
patients after severe head injury: monitoring and effects of arterial alternatives in traumatic brain injury. Curr Treat Options Neurol 2012;14:
hyperoxia on cerebral blood flow, metabolism and intracranial pressure. 150–63.
J Neurosurg Anesthesiol 1999;11:240–51. 140. Le Roux PD. Participants in the International Multi-disciplinary Consensus
114. Reinert M, Barth A, Rothen HU, et al. Effects of cerebral perfusion pressure Conference on the Critical Care Management of Subarachnoid
and increased fraction of inspired oxygen on brain tissue oxygen, lactate Hemorrhage. Anemia and transfusion after subarachnoid hemorrhage.
and glucose in patients with severe head injury. Acta Neurochir (Wien) Neurocrit Care 2011;15(2):342–53.
2003;145:341–9; discussion 349–50. 141. Oddo M, Levine JM, Kumar M, et al Anemia and brain oxygen after severe
115. Hemphill JC, 3rd, Knudson MM, Derugin N, et al. Carbon dioxide traumatic brain injury. Intensive Care Med 2012;38(9):1497–504.
reactivity and pressure autoregulation of brain tissue oxygen. Neurosurgery 142. Naidech AM, Jovanovic B, Wartenberg KE, et al. Higher hemoglobin is
2001;48:377–83; discussion 383–4. associated with improved outcome after subarachnoid hemorrhage. Crit
116. Hlatky R, Valadka AB, Gopinath SP, et al. Brain tissue oxygen tension Care Med 2007;35:2383–9.
response to induced hyperoxia reduced in hypoperfused brain. J Neurosurg 143. Fletcher JJ, Bergman K, Blostein PA, et al. Fluid balance, complications, and
2008;108:53–8. brain tissue oxygen tension monitoring following severe traumatic brain
117. Oddo M, Le Roux P. Brain tissue oxygen monitors: more than an ischemia injury. Neurocrit Care 2010;13(1):47–56.
monitor. Crit Care Med 2008;36:1984–5. 144. Johnston AJ, Steiner LA, Chatfield DA, et al. Effects of propofol on cerebral
118. Kety SS, Schmidt CF. The nitrous oxide method for the quantitative oxygenation and metabolism after head injury. Br J Anaesth 2003;91:781–6.
determination of cerebral blood flow in man: theory, procedure and 145. Chen HI, Malhotra NR, Oddo M, et al. Barbiturate infusion for intractable
normal values. J Clin Invest 1948;27:476–83. intracranial hypertension and its effect on brain oxygenation. Neurosurgery
119. Nortje J, Coles JP, Timofeev I, et al. Effect of hyperoxia on regional 2008;63:880–6; discussion 886–7.
oxygenation and metabolism after severe traumatic brain injury: 146. Thorat JD, Wang EC, Lee KK, et al. Barbiturate therapy for patients with
preliminary findings. Crit Care Med 2008;36:273–81. refractory intracranial hypertension following severe traumatic brain
120. Abate MG, Trivedi M, Fryer TD, et al. Early derangements in oxygen and injury: its effects on tissue oxygenation, brain temperature and
glucose metabolism following head injury: the ischemic penumbra and autoregulation. J Clin Neurosci 2008;15:143–8.
pathophysiological heterogeneity. Neurocrit Care 2008;9:319–25. 147. Oddo M, Frangos S, Milby A, et al. Induced normothermia attenuates
121. Merenda A, Bullock R. Clinical treatments for mitochondrial dysfunctions cerebral metabolic distress in patients with aneurysmal subarachnoid
after brain injury. Curr Opin Crit Care 2006;12:90–6. hemorrhage and refractory fever. Stroke 2009;40(5):1913–6. Epub 2009,
122. Dings J, Meixensberger J, Jager A, et al. Clinical experience with 118 brain Feb 26.
tissue oxygen partial pressure catheter probes. Neurosurgery 1998;43: 148. Kett-White R, Hutchinson PJ, Al-Rawi PG, et al. Adverse cerebral events
1082–95. detected after subarachnoid hemorrhage using brain oxygen and
123. Brawanski A, Faltermeier R, Rothoerl RD, et al. Comparison of near- microdialysis probes. Neurosurgery 2002;50:1213–21; discussion 21–2.
infrared spectroscopy and tissue p(O2) time series in patients after severe 149. Gelabert-Gonzalez M, Fernandez-Villa JM, Ginesta-Galan V. Intra-operative
head injury and aneurysmal subarachnoid hemorrhage. J Cereb Blood Flow monitoring of brain tissue O2 (PtiO2) during aneurysm surgery. Acta
Metab 2002;22:605–11. Neurochir (Wien) 2002;144:863–6; discussion 866–7.
355.e4 Section VI—Intracranial Monitoring

150. Hoffman WE, Charbel FT, Edelman G, et al. Brain tissue oxygenation in 159. van Santbrink H, van den Brink WA, Steyerberg EW, et al. Brain tissue
patients with cerebral occlusive disease and arteriovenous malformations. oxygen response in severe traumatic brain injury. Acta Neurochir (Wien)
Br J Anaesth 1997;78:169–71. 2003;145:429–38; discussion 438.
151. Hoffman WE, Wheeler P, Edelman G, et al. Hypoxic brain tissue following 160. Hoffman WE, Edelman G, Wheeler P. Cerebral oxygen reactivity in the dog.
subarachnoid hemorrhage. Anesthesiology 2000;92:442–6. Neurol Res 2000;22:620–2.
152. Szelenyi A, Jung CS, Schon H, Seifert V. Brain tissue oxygenation 161. Soehle M, Jaeger M, Meixensberger J. Online assessment of brain tissue
monitoring supplementary to somatosensory evoked potential monitoring oxygen autoregulation in traumatic brain injury and subarachnoid
for aneurysm surgery. Initial clinical experience. Neurol Res 2002;24:555–62. hemorrhage. Neurol Res 2003;25:411–7.
153. Hoffman WE, Charbel FT, Edelman G, et al. Brain tissue gases and pH 162. Meixensberger J, Renner C, Simanowski R, et al. Influence of cerebral
during arteriovenous malformation resection. Neurosurgery 1997;40:294– oxygenation following severe head injury on neuropsychological testing.
300; discussion 301. Neurol Res 2004;26:414–7.
154. Pennings FA, Bouma GJ, Kedaria M, et al. Intraoperative monitoring of 163. Meixensberger J, Vath A, Jaeger M, et al. Monitoring of brain tissue
brain tissue oxygen and carbon dioxide pressures reveals low oxygenation oxygenation following severe subarachnoid hemorrhage. Neurol Res
in peritumoral brain edema. J Neurosurg Anesthesiol 2003;15:1–5. 2003;25:445–50.
155. Tijero T, Ingelmo I, Garcia-Trapero J, et al. Usefulness of monitoring brain 164. Adamides AA, Cooper DJ, Rosenfeldt FL, et al. Focal cerebral oxygenation
tissue oxygen pressure during awake craniotomy for tumor resection: a case and neurological outcome with or without brain tissue oxygen-guided
report. J Neurosurg Anesthesiol 2002;14:149–52. therapy in patients with traumatic brain injury. Acta Neurochir (Wien)
156. Hoffman WE, Edelman G. Isoflurane increases brain oxygen reactivity in 2009;151(11):1399–409.
dogs. Anesth Analg 2000;91:637–41. 165. Martini RP, Deem S, Yanez ND, et al. Management guided by brain tissue
157. Hoffman WE, Edelman G. Enhancement of brain tissue oxygenation during oxygen monitoring and outcome following severe traumatic brain injury.
high dose isoflurane anesthesia in the dog. J Neurosurg Anesthesiol J Neurosurg 2009;111(4):644–9.
2000;12:95–8.
158. Steiner LA, Johnston AJ, Chatfield DA, et al. The effects of large-dose
propofol on cerebrovascular pressure autoregulation in head-injured
patients. Anesth Analg 2003;97:572–6.
VI
Chapter
36  

Cerebral Microdialysis
Martin Smith

injured brain. MD should therefore be seen as a tool to explore


Introduction the neurochemical features of secondary brain injury pro-
Cerebral microdialysis (MD) is a well-established laboratory cesses with other monitors as part of a multimodal monitor-
tool that is being increasingly used as a bedside monitor to ing technique.9
provide online analysis of brain tissue biochemistry during
neurointensive care. MD samples substances present in brain
extracellular fluid (ECF) and has opened new avenues to
Principles of Microdialysis
monitor the injured brain. Because MD measures changes at The principles of the MD technique have been reviewed in
the cellular level, it is an attractive technique to detect and detail elsewhere.1,2,10-14 In brief, a MD catheter consists of a fine
monitor cerebral hypoxia, ischemia, and other causes of cel- double-lumen probe, lined at its tip with a semipermeable
lular dysfunction. To date MD has been predominantly applied dialysis membrane. The probe tip is placed into biologic
to patients with traumatic brain injury (TBI) and subarach- tissue, in this case the brain, and perfused via an inlet tube
noid hemorrhage (SAH) and has increased understanding with fluid isotonic to the tissue interstitium. The perfusate
of the pathophysiology of these conditions. Studies also have passes along the membrane before exiting through outlet
addressed its potential role as a clinical monitoring technique tubing into a microvial designed to collect minute amounts of
that can guide individualized and targeted therapy after brain fluid (Figs. 36.1 and 36.2). The microvials are removed regu-
injury.1,2 Future studies must identify the effect of therapeutic larly, usually every hour, and placed in a bedside analyzer,
maneuvers on brain tissue biochemistry and establish the rela- which measures and records the changing brain tissue chem-
tionship between MD variables and outcome. istry. Subsequently the samples can be analyzed off-line for
other substances.
MD is based on the principle of diffusion of water-soluble
History substances across the semipermeable MD membrane driven
The first use of the MD technique was in 1966 when Bito by their concentration gradient. Molecules at high concentra-
inserted membrane-lined sacks containing 6% dextran into tion in the brain ECF pass into the perfusate with minimum
the cerebral hemispheres of dogs and analyzed the fluid that passage of water, and because the perfusate flows along the
they contained for amino acid levels when they were removed membrane and is removed at a constant rate, the concentra-
10 weeks later.3 During the 1970s Delgado developed the tion gradient across the membrane is maintained. In this way
“dialytrode” device,4 which was refined later and simplified by the MD catheter acts as an artificial blood capillary with the
Tossman and Ungerstedt into the “microdialysis” method.5 perfusate gradually equilibrating to the composition of ECF.
This initially was used to study neurochemical processes in The concentration of substrate in the collected fluid (the
animals, but in 1990, the first application in the human brain microdialysate) depends in part on the balance between
was described.6 The use of cerebral MD was popularized substrate delivery to, and uptake or excretion from, the ECF
following the introduction of commercially available micro- (Fig. 36.3). This simple concept provides a powerful technique
dialysis catheters and a bedside analyzer (CMA 600, CMA in which any molecule small enough to pass across the
Microdialysis, Solna, Sweden) and was first used as a neuro- membrane can be sampled.
chemical monitoring tool in neurocritical care in 1992.7 Unless there is total equilibration across the dialysis mem-
Devices for microdialysis in the human brain were CE-marked brane, the concentration of a given molecule in the microdi-
according to the European Medical Device Directive in 1995 alysate will be lower than its concentration in the brain ECF.
and approved by the U.S. Food and Drug Administration The proportion of the true ECF concentration collected in the
in 2002. microdialysate is termed the relative recovery and is dependent
Today MD is a well-established research tool but it has not on many factors, including dialysis membrane pore size,
yet become widely established as a routine clinical monitor.8 membrane area, perfusate flow rate, and diffusion properties
However, some centers that have extensive experience of its of the substance. Because of the many variables that affect the
use as a research tool are beginning to incorporate MD moni- relative recovery, it is essential to consider the sampling
toring into clinical practice.2 When considering the clinical methods and materials used when comparing measured MD
role of MD, it is important to appreciate that it contributes a values. In clinical practice the most commonly used system
single, albeit vital, component of the whole picture of the comprises a catheter that is 10 mm in length with a 20-kDa
356 © Copyright 2013 Elsevier Inc. All rights reserved.
Section VI—Intracranial Monitoring 357

(CMA 70, CMA Microdialysis, Solna, Sweden) or 100-kDa 20-kDa molecular weight cutoff catheter, whereas 100-kDa
(CMA 71, CMA Microdialysis, Solna, Sweden) molecular cutoff catheters allow investigation of higher molecular weight
weight cutoff, perfused with commercially available physio- biomarkers. For the more clinically relevant MD variables,
logic perfusion fluid (Perfusion Fluid CNS, CMA Microdialy- 100-kDa catheters have equivalent recovery to 20-kDa cutoff
sis, Solna, Sweden) at a rate of 0.3 µL/min. Commonly catheters,15 and many centers now use the higher cutoff cath-
measured bedside MD variables are reliably captured using a eters as a routine.16 A perfusate flow rate of 0.3 µL/min
allows hourly sampling at the bedside, striking a balance
between adequate microdialysate volume and acceptable
recovery rates. Higher flow rates permit more frequent
sampling but at the cost of lower concentration of measured
2
substances in the microdialysate. A standard flow rate of
1 0.3 µL/min and a 10-cm catheter achieves a recovery rate in
excess of 70% for routinely measured metabolites.17 By using
lower perfusate flow rates it is possible to approach 100%
5
recovery and measurement of the absolute concentration of a
substance in brain ECF.18
Although hourly MD sampling rates are adequate for most
3 clinical purposes, continuous sampling would be advanta-
6 4 geous when rapid and transient changes in brain metabolite
levels need to be determined. A continuous MD method has
7
been described for research purposes but it is not yet
sufficiently developed for clinical application.19 This system
incorporates continuous flow of dialysate into a glucose and
lactate analyzer that uses flow-injection dual-assay enzyme-
Fig. 36.1  Photograph of the components of a clinical microdialysis
based biosensors able to determine values of metabolites
catheter (CMA Microdialysis, Solna, Sweden). (1), Pump connector;
(2), inlet tube; (3), microdialysis catheter; (4), microdialysis membrane; every 30 seconds. The analyzer can detect changes 9 minutes
(5), outlet tube; (6), microvial holder; (7), microvial for collection of after an event has occurred, and the temporal resolution is
microdialysate. (Reproduced with permission from Tisdall MM, Smith M. Cerebral limited only by probe-to-sensor tubing length and perfusate
microdialysis: research technique or clinical tool. Br J Anaesth 2006;97(1):18–25.) flow rate.

Microdialysate collected Isotonic fluid perfused


via outlet tube via inlet tube

Microdialysis
catheter
Molecules in ECF
equilibrate across MD
membrane

Semi-permeable
Brain tissue membrane
interstitium

Fig. 36.2  Schematic representation of a microdialysis catheter in brain tissue. Fluid isotonic to the brain extracellular fluid is pumped through
the microdialysis catheter at a rate of 0.3 µL/min. Molecules at high concentration in the brain extracellular fluid equilibrate across the semipermeable
microdialysis membrane and can be analyzed in the collected perfusate (the microdialysate). ECF, Extracellular fluid; MD, microdialysis. (Modified with
permission from Tisdall MM, Smith M. Cerebral microdialysis: research technique or clinical tool. Br J Anaesth 2006;97(1):18–25.)
358 Section VI—Intracranial Monitoring

Decreased
glucose and oxygen

Glucose
Ischemia lactate:
markers virgule
pyruvate
ratio
Capillary

Degeneration
marker Glycerol

Microdialysis catheter

Fig. 36.3  Schematic representation of the relationship between blood capillary and microdialysis catheter in brain tissue. The microdialysis
catheter acts as an artificial blood capillary and the concentration of substrate in the collected fluid (microdialysate) is related to the balance between
substrate delivery to, and uptake or excretion from, the extracellular fluid. (Reproduced with permission from Tisdall MM, Smith M. Cerebral microdialysis:
research technique or clinical tool. Br J Anaesth 2006;97(1):18–25.)

Clinical Relevance of Monitoring Microdialysis Markers


Tissue Biochemistry in the Many substances can be measured using MD, but the key
variables that are most relevant to neurocritical care can be
Injured Brain categorized as follows11:
Acute brain injury frequently is exacerbated by secondary
• Energy-related metabolites, for example, glucose, lactate,
events that lead to secondary brain injury. This occurs follow-
and pyruvate
ing activation by the primary injury of an autodestructive
• Neurotransmitters, for example, glutamate, aspartate,
cascade of metabolic, immunologic, biochemical, and inflam-
gamma-aminobutyric acid (GABA)
matory changes that render the brain more susceptible to
• Markers of tissue damage and inflammation, for example,
systemic physiologic insults that can result in irreversible cell
glycerol, potassium, cytokines
damage or death.20 Although these pathologic processes are
• Exogenous substances, for example, drugs
poorly understood, they include calcium overload, increased
production of free radicals, release of neurotoxic levels of The different types of markers that can be recovered by MD
excitatory amino acids (EAAs), and failing cellular metabo- highlights the utility of the technique to provide surrogate
lism. Ultimately these changes can cause cellular swelling, biochemical outcomes that reflect underlying pathologic
increase in intracranial pressure (ICP), further neuronal loss, mechanisms of injury.26 These highlighted markers represent
and, if unchecked, can result in increased mortality and wors- a small fraction of those that have been recovered by MD, and
ened outcome in survivors. a comprehensive review lists those that are less well evalu-
Secondary brain injury is a potentially modifiable cause of ated.10 Several studies have addressed the recovery of protein
mortality and morbidity, and the primary aim of neurocritical biomarkers such as S100β, tau, beta amyloid proteins, and
care treatment of brain injury is to prevent or reduce the neurofilament.27-29 What these various biomarkers mean is still
burden of secondary injury.21 Conventional intracranial mon- being elucidated, and the role of biomarkers in neurocritical
itoring techniques, such as measurement of ICP, are often care is discussed further in Chapter 18.
“reactive” and may indicate changes only when irreversible Commercially available assays for bedside use are those for
tissue damage has already occurred. Monitoring brain tissue glucose, lactate, pyruvate, glycerol, and glutamate, and tenta-
biochemistry through microdialysis has the potential to guide tive normal values for these variables in adults are described
individualized therapy after brain injury,2 to identify impend- (Table 36.1). These normal values were derived from a study
ing or early onset secondary injury in some cases before there by Reinstrup et al., who inserted MD catheters into the frontal
is a change in ICP,22 to detect cerebral compromise when ICP cortex of patients undergoing surgery for benign posterior
or cerebral perfusion pressure (CPP) is normal,23 and allow fossa lesions and collected microdialysate samples in the
timely implementation of neuroprotective strategies.24,25 postoperative course to demonstrate baseline metabolite
Section VI—Intracranial Monitoring 359

Table 36.1  Suggested Normal Concentrations Table 36.2  Biochemical Markers of


of Commonly Measured Biochemical Markers Secondary Brain Injury
in Microdialysate Samples from the
Microdialysis
Uninjured Human Brain Variable Biomarker for: Comments
Normal Value Normal Value Low glucose • Hypoxia/ • Should be interpreted
Microdialysate ± SD ± SD ischemia in association with
Concentration Reinstrup et al.30 Schulz et al.31 • Reduced serum glucose
cerebral concentration
Glucose (mmol/L) 1.7 ± 0.9 2.1 ± 0.2
glucose supply
Lactate (mmol/L) 2.9 ± 0.9 3.1 ± 0.3 • Cerebral
hyperglycolysis
Pyruvate (µmol/L) 166 ± 47 151 ± 12
Increased LPR • Hypoxia/ • Most reliable
Lactate/pyruvate ratio 23 ± 4 19 ± 2
ischemia biomarker of ischemia
Glycerol (µmol/L) 82 ± 44 82 ± 12 • Cellular redox • Independent of
state catheter recovery
Glutamate (µmol/L) 16 ± 16 14 ± 3.3 • Reduced • Tissue hypoxic
From Reinstrup P, Stahl N, Mellergard P, et al. Intracerebral microdialysis in
cerebral threshold for raised
clinical practice: baseline values for chemical markers during wakefulness, glucose supply LPR not established
anesthesia, and neurosurgery. Neurosurgery 2000;47(3):701–9; and • Impairment of
Schulz MK, Wang LP, Tange M, et al. Cerebral microdialysis monitoring: glycolytic
determination of normal and ischemic cerebral metabolisms in patients with pathway
aneurysmal subarachnoid hemorrhage. J Neurosurg 2000;93(5):808–14.
Increased • Hypoxia/ • Increased glycerol
glycerol ischemia may also occur due to
• Cell spillover from
membrane systemic glycerol or
degradation from the formation of
concentrations from the uninjured human brain.30 Concen- glycerol from glucose
trations of metabolites in microdialysate samples from patients
Increased • Hypoxia/ • Wide variability in
with SAH but no clinical or radiologic evidence of cerebral glutamate ischemia glutamate levels
ischemia also have been considered to represent “normal” • Excitotoxicty within and between
values.31 A summary of the pathophysiologic changes moni- patients
tored by cerebral MD biomarkers is shown in Table 36.2.
LPR, Lactate/pyruvate ratio.
There have been far fewer MD studies in children; these
suggest there may be differences in MD findings in children
compared with adults.32 Brain ischemia and hypoxia underlie
many of the secondary cerebral insults that can cause brain
damage; this results in a typical pattern of change in MD cell integrity. Microdialysate glucose levels are reduced in
variables including an increase in glycerol and glutamate con- patients following TBI, and a concentration consistently less
centrations, a reduction in glucose, and an increase in the than 0.66 mmol/L in the first 50 hours post injury is associ-
lactate/pyruvate ratio (LPR) and lactate/glucose ratio (LGR). ated with poor outcome.35 The etiology of this low glucose
Although an elevated LPR frequently is interpreted as a sign concentration is likely to be multifactorial. In the acute period
of cerebral hypoxia or ischemia there also are many other after TBI there is typically a reduction in oxidative metabo-
causes of an elevated LPR that are not related to hypoxia or lism36 and increase in glucose metabolism.37 Extremely low
ischemia.33 Consistent with this Nelson et al.34 observed that brain ECF glucose is observed during periods of severe hypoxia
altered local biochemistry detected with MD in severe TBI or ischemia after TBI2,38 and SAH,39 and associated with a
patients represented long-term metabolic patterns and was brain tissue oxygen (PbtO2) of less than 1.3 kPa (10 mm Hg).40,41
weakly correlated with ICP and CPP; that is, metabolic changes However, there is a poor correlation between positron emis-
other than those related to pressure or flow can influence MD sion tomography (PET)–defined ischemia and low MD
findings. glucose concentration42 suggesting that in some cases at least,
low MD glucose concentration may be associated with hyper-
glycolysis rather than decreased supply of glucose and oxygen
Markers of Glucose Metabolism because of reduced cerebral perfusion.
Tissue hypoxia often is the final common pathway for cellular Glucose is taken up into neurons and glia and initially
damage after acute brain injury, and the most commonly metabolized to pyruvate by glycolysis. When there is adequate
investigated substances assayed by MD relate to the aerobic oxygen delivery and tissue oxygenation, pyruvate enters the
and anaerobic metabolism of glucose. The determinants of citric acid cycle and ultimately is metabolized to carbon
cerebral extracellular glucose concentration are complex and dioxide and water with the generation of adenosine triphos-
depend on peripheral blood glucose concentration, local cap- phate (ATP). During ischemia, pyruvate is diverted into the
illary blood flow, and brain cell uptake of glucose. A particular anaerobic pathway and is metabolized to lactate. The mea-
advantage of cerebral MD monitoring after brain injury is its surement of ECF lactate and pyruvate concentrations there-
ability to assess not only the cerebral delivery of glucose but fore provides information about the extent of anaerobic
also its use. glycolysis. However, the absolute value of lactate within the
The energy demands of the brain are met by the metabo- brain ECF is not in itself indicative of the degree of anaerobic
lism of glucose, and a continued supply is vital to maintain metabolism for several reasons. The production of lactate
360 Section VI—Intracranial Monitoring

depends on a continued supply of glucose, which may fail MD glycerol concentration typically is elevated in the first 24
during complete ischemia, and the increased levels of gluta- hours after severe TBI, presumably as a result of the primary
mate and potassium that are associated with ischemia drive injury, and then exponentially declines during the ensuring 3
astrocyte lactate production and hyperglycolysis.37 An increase days. Clausen et al. observed that PbtO2 less than 1.3 kPa
in ECF glutamine may reflect this accelerated astrocyte (10 mm Hg) and CPP less than 70 mm Hg were associated
metabolism and be found in vulnerable rather than already with elevated mean cerebral MD glycerol levels, although indi-
ischemic tissue.43 In addition, lactate can act as a metabolic vidual episodes below the same thresholds also occurred
substrate to sustain increased energy needs through the without an increase in MD glycerol concentration.63 In this
astrocyte-neuron lactate shuttle.44-46 To correct for the vari- study mean MD glycerol concentration was similar in patients
able sources of ECF lactate and the dynamic variation in with favorable and unfavorable outcomes. The interpretation
glucose delivery, the LPR47-49 and LGR31,38 have been used as of elevated glycerol levels after TBI therefore requires further
more accurate markers of anaerobic metabolism. Because validation. Glycerol also is able to leak through a damaged
lactate and pyruvate have very similar molecular weights, the blood brain barrier (BBB) and cause spuriously high brain
LPR is independent of catheter recovery in vivo.50 The LPR is ECF glycerol concentration because of the high levels of
therefore a robust and reliable marker of cell energy dysfunc- plasma glycerol that occur due to stress-induced triglyceride
tion and is the most widely monitored MD variable after degradation or the administration of exogenous glycerol.66 To
brain injury. help distinguish a compromised BBB effect from a true intra-
In the human brain, worsening hypoxia, ischemia, or edema cerebral event, systemic glycerol concentration can be simul-
can lead to an increase in the LPR,51-53 which is associated with taneously measured via an MD catheter placed in the
severe reductions in PbtO254 and increases in PET-measured abdominal subcutaneous adipose tissue.64
oxygen extraction fraction.55 An increase in LPR above estab-
lished thresholds (20-25) is associated with poor outcome
after TBI56-58 and SAH,49,59,60 and traditionally is assumed to Excitotoxicity
indicate tissue hypoxia or ischemia. However, it has proved Ischemia, TBI, SAH, and other pathologies can lead to cell
difficult to establish the tissue hypoxic threshold for a raised depolarization and release of EAAs such as glutamate and
LPR,61 and it is increasingly apparent that anaerobic glycolysis aspartate. Excitotoxicity is one of several mechanisms impli-
may occur from ineffective utilization of delivered oxygen cated in neuronal injury, and there was early enthusiasm
because of mitochondrial failure, among other causes.33,42 For about using MD to measure glutamate because animal studies
example, Vespa et al. compared MD variables with PET- demonstrated raised glutamate concentration in global cere-
derived measurement of glucose and oxygen metabolism in bral ischaemia67 and TBI.68 Subsequent studies in humans
19 patients with severe TBI.42 There was a 25% incidence of also demonstrated an association between increased MD glu-
metabolic crisis, defined in this study as LPR greater than 40, tamate concentration and poor outcome after TBI18 and
but only a 2.4% incidence of PET-defined ischemia. It was SAH.49,59 For example, Staub et al. observed 30-fold increases
concluded that these data indicate that the LPR is a reliable in ECF glutamate, aspartate, and GABA in SAH patients with
marker of cellular dysfunction and of inadequate substrate unfavorable outcome at 3 months compared with those with
delivery. An elevated LPR therefore may be classified as type I favorable outcome.69 The increased concentration of EAA in
(ischemic), in which the pyruvate is decreased and there is a patients with poor outcome followed a biphasic course, with
marked increase in lactate, or type II (nonischemic hypergly- maximal concentrations on the first to second, and seventh,
colysis), in which the pyruvate is normal or increased. In a days after the insult. It is likely that the early increase repre-
type I LPR elevation there is oxygen and glucose lack, whereas sents the degree of initial injury and the later increase the
in type II LPR elevation there is mitochondrial failure or development of vasospasm-related ischemic injury. Prolonged
failure of effective use of delivered oxygen and glucose.42 In elevation of MD glutamate also is observed after severe TBI
animal models of TBI, average LPR has a strong correlation and is associated with poor outcome.70 High MD-glutamate
with total injury volume on neuropathologic examination,62 levels have been correlated with clinical events involving
confirming the value of LPR monitoring in the neurocritical hypoxia, ischemia, reduced PbtO2,40 and low CPP.71 The role of
care unit (NCCU). glutamate in excitotoxicity after TBI has been challenged,72
and there are multiple causes of increased ECF glutamate
concentration after brain injury.10 Nevertheless, recent studies
Markers of Tissue Damage suggest glutamate levels may provide useful information in
Failure of cellular metabolism results in disruption of cell some patients. For example, Timofeev et al. found that MD
membrane function that then leads to intracellular influx of glutamate was associated weakly with mortality in 223 severe
calcium, induction of phospholipase, and ultimately to deg- TBI patients but that this association disappeared after mul-
radation of cell membranes. This results in release of phos- tivariable analysis.56 Chamoun et al. observed two patterns of
pholipids and, after enzymatic breakdown, free fatty acids and glutamate elevation after TBI: (1) glutamate levels tended to
glycerol into the brain ECF. Glycerol is reliably recovered from normalize over the monitoring period (120 hours); and (2)
the ECF and is therefore a useful MD marker of tissue hypoxia glutamate levels tended to increase with time or remain
and cell damage.63,64 The degree of the MD glycerol elevation abnormally elevated.73 Pattern 1 patients had a lower mortal-
associated with pathology (e.g., hypoxia or ischemia) may be ity rate (17.1% versus 39.6%) and a better 6-month func-
dramatic, with four- or eightfold increases recorded in severe tional outcome among survivors (41.2% versus 20.7%)
or complete ischemia respectively.31 High levels of interstitial compared to pattern 2. In addition glutamate levels greater
glycerol are associated with an unfavorable outcome after TBI than 20 mmol/L were associated with a nearly twofold
and indicate the severity of parenchymal damage.65 Cerebral increase in mortality.
Section VI—Intracranial Monitoring 361

to be tunneled under the skin and fixed with sutures. Although


Variability of Measured Metabolites the catheter can be inserted to varying depths at any orienta-
After brain injury there are wide variations in MD variables tion, it is susceptible to migration or displacement depending
over time between different subjects and within individu- on the security of the fixation.
als.7,24,49,70 These are likely to represent the changing levels of There always is potential for tissue trauma at the time of
metabolic activity within the injured brain55 but make it dif- catheter insertion. Animal and human studies demonstrate
ficult to interpret single MD measurements or measurement that artificially high levels of substrate that result from trauma
obtained in isolation. Although “normal” and threshold MD or inflammation at the time of insertion usually abate within
values are published, cerebral MD must be seen as a trend the first hour of placement.77 A “run-in” period of at least
monitor, and the information provided by MD should be 1 hour should therefore be allowed before using MD data.
interpreted with other measured variables, clinical informa-
tion, or radiologic findings.2
Clinical Applications of
Catheter Placement Microdialysis Monitoring
MD monitors local tissue biochemistry and reflects meta- Several lines of clinical evidence suggest that MD may assist
bolic disturbances and neurochemical changes only in the clinical decision making, such as management of cerebral per-
region of the brain where the catheter is located. Differences fusion pressure,80 hyperventilation,81 and the appropriateness
between MD measured variables are observed in areas close of surgical procedures.82 This includes deciding when none-
to, and far away from, focal traumatic lesions. For example, mergent surgery may be performed after severe TBI. For
Engstrom et al. found in 22 severe TBI patients that MD example, Yokobori et al.83 examined 25 patients with MD and
markers of ischemia were significantly different in tissue several other monitors. They observed that the extracellular
adjacent to a parenchymal lesion when compared with glucose concentration improved, and the value of LPR
normal tissue in the contralateral hemisphere.74 Vespa et al. decreased 4 days after injury, whereas the average PRx
also observed regional differences in brain tissue chemistry decreased daily and became negative on the fifth day after
after TBI and confirmed the importance of catheter loca- injury. These results suggested that the injured brain may
tion.75 In this study, there was persistent metabolic dysfunc- become “less vulnerable” to secondary insults 4 to 5 days after
tion in pericontusional tissue, both in baseline values and injury. Microdialysis also may be used to guide therapies such
trends across time, compared with normal tissue. However, as hyperoxia,84 induced hypothermia,85 or normothermia;86,87
despite biochemical differences between “perilesional” and help establish optimal hemoglobin88,89, and glucose90,91 levels
less-injured brain, Timofeev et al.76 observed that both areas for transfusion and glycemic control; and provide information
showed a relationship between other physiologic variables to help determine prognosis. Finally, MD findings may provide
(e.g., ICP, CPP, and PbtO2) and biochemistry. In particular, early warning of bacterial meningitis in conditions such as
reduced CPP or brain oxygen was associated with worse neu- poor grade SAH, in which clinical signs may be masked by the
rochemistry (e.g., increased LPR), although these effects were effect of the hemorrhage.92 However, to offer added value over
greater in perilesional tissue and when cerebrovascular reac- and above that provided by other intracranial monitors, cere-
tivity was abnormal. bral MD must not only guide treatment, but also do so in a
There are consensus recommendations on where to place way that reduces the burden of secondary brain injury and
MD catheters to monitor patients after TBI and SAH.77 The thereby offers the potential to improve functional outcome in
MD catheter should be placed in “at-risk” tissue (e.g., the area survivors.
surrounding a mass lesion after TBI), or the vascular territory
most likely to be affected by vasospasm after SAH. These loca-
tions should allow biochemical changes to be measured in the Monitoring Evolution of Brain Injury
area of brain most vulnerable to secondary injury. In diffuse Monitoring of biochemical changes in at-risk brain tissue
axonal injury catheter placement in the nondominant frontal using MD may provide clinically useful indications of evolv-
lobe is recommended. However, others suggest that the cath- ing brain injury at the bedside. Hlatky et al. compared focal
eter should always be placed in a normal brain region so that measurements of MD and PbtO2 from brain tissue under an
it can be used to indicate global cerebral metabolism.78 evacuated subdural hematoma to global measures of ICP, CPP,
Whether white or gray matter should be monitored is another and jugular venous oxygen saturation (SjvO2) in patients with
variable to consider to interpret MD results. White matter has severe TBI.93 Seventeen of the 33 (52%) patients developed
a lower metabolic demand than gray matter and receives a delayed focal secondary injury (contusion or infarction) that
smaller proportion of the cerebral blood flow. Most studies was associated with reduced PbtO2 and LPR elevation in the
allude to white matter placement. Commercial MD catheters absence of global changes in ICP, CPP, or SjvO2. There were
have a gold tip so that the catheter position can be confirmed no changes in MD variables and PbtO2 in the 11 patients with
by computed tomography scan.77 an uneventful clinical course. The remaining 5 patients devel-
The MD catheter is a parenchymal probe that usually is oped fatal refractory intracranial hypertension and demon-
inserted though a cranial access device, including single- strated markedly elevated LPR and glutamate, and reduced
lumen bolts designed only for MD catheters or ones that glucose concentration. Other studiers have suggested that
contain multiple-angled lumens to transmit other parenchy- changes in MD markers may precede changes in ICP. For
mal probes such as PbtO2 or ICP sensors.79 The catheter also example, Adamides et al., who studied 14 patients with severe
can be placed through a burr hole or under direct vision at TBI, observed that elevations in brain lactate, LPR and glyc-
the time of craniotomy. These techniques require the catheter erol often preceded an episode of intracranial hypertension by
362 Section VI—Intracranial Monitoring

more than 2 hours.22 In particular the absolute elevation of risk of imminent intracranial hypertension, with odds ratios
brain lactate was the earliest and strongest predictor of ele- of 9.8 (95% CI 5.8-16.1) and 2.2 (95% CI 1.6-3.8), respec-
vated ICP. tively. An abnormal LPR was predictive of an increase in ICP
above normal levels in 89% of cases. Not all studies have found
evidence for the predictive value of MD. For example, Peerde-
Prognostication man et al. observed no association between MD glycerol
Information obtained from MD may help determine progno- concentration and secondary events, such as intracranial
sis in the unconscious patient when clinical examination is not hypertension and low CPP, after TBI.65 Glutamate appears to
possible. Several studies in both TBI and SAH have demon- be a sensitive indicator of impending cerebral ischemia after
strated an association with disturbed MD values and worse SAH95 but appears to have less predictive value after TBI.24
clinical condition and outcome. In SAH, disturbed cerebral It remains to be seen whether the early warnings provided
metabolism is related to the severity of SAH. Sarrafzadeh et al. by MD measured biochemical changes after TBI and SAH in
studied 149 SAH patients who underwent surgery; cerebral some studies can be exploited to expand the window for thera-
metabolism was deranged in poor (IV and V) compared to peutic intervention and whether this will improve outcome.
good (I-III) grade patients.60 Noteworthy in particular were
significant elevations of LPR, LGR, and glycerol in poor grade
patients. Elevated LPR and glutamate concentration also were Monitoring the Effects of Treatment
markers of poor outcome at 12 months. After severe TBI, on the Injured Brain
metabolic changes and alterations in CBF may be more Clinical use of MD in the NCCU usually is applied to TBI and
complex, but consistently low MD glucose levels early after SAH patients, and this has improved understanding of the
injury have been associated with poor outcome35 as have many pathophysiologic processes that occur in the injured
increases in LPR.57,58 These extracellular metabolic markers brain, particularly when used with other monitors or imaging
may be independently associated with outcome after severe techniques (e.g., PET96). In addition, cerebral MD has increased
TBI. For example, Timofeev et al. collected prospective obser- understanding of how current and potential neurointensive
vational neuromonitoring data from 223 severe TBI patients.56 care treatment strategies affect the injured brain. This can be
In a multivariate logistic regression model that used data aver- of great importance since each therapy can have both benefits
aged over the whole monitoring period (median duration 4 and deleterious side effects and most are best used in a tar-
days), significant independent factors associated with mortal- geted fashion. For example, Oddo et al. have observed that
ity included brain glucose, LPR, ICP, cerebrovascular pressure induced normothermia can improve brain metabolism after
reactivity index, and age, whereas pyruvate was a significant acute brain injury but that when shivering occurs, tissue
independent negative predictor of mortality. Levels of gluta- hypoxia may occur.86,87 MD therefore is being applied increas-
mate and glycerol were associated with mortality in univariate ingly as a surrogate endpoint to evaluate therapeutic interven-
but not multivariate analysis. tional strategies or as a means of choosing an appropriate
management.
Prediction of Secondary Injury
Because cerebral MD measures change at the cellular level, it Assessment of Adequacy of Cerebral Perfusion
is a useful tool to describe molecular events triggered by brain There is a wide range recommended for the “optimal” CPP
injury. Brain ECF glutamate and glycerol concentrations cor- after TBI and other brain injuries. Rather than there being a
relate with regional cerebral blood flow after SAH, and LPR single CPP level that applies to all patients, the optimal CPP
shows high sensitivity and specificity for brain energy dys- varies among patients and over time in individual patients;
function that often is associated with symptoms of ischemia.94 MD can help identify this level. For example, Nordstrom et al.
In addition, abnormal MD findings may precede changes in studied 50 severe TBI patients using an MD catheter placed in
ICP.22 MD therefore has the potential to detect secondary the at-risk tissue surrounding a focal lesion and a second in
brain insults including hypoxia or ischemia before changes the contralateral frontal lobe (normal tissue).80 The lactate
can be identified in the patient’s neurologic status or by more concentration was higher in the at-risk compared with normal
conventional monitoring techniques such as ICP measure- brain and LPR increased in the at-risk tissue when CPP fell
ment. For example, Skjoth-Rasmussen et al. observed an isch- below 50 mm Hg (see also the example at Fig. 36.4). These
emic MD pattern, defined as an increase in LPR and LGR authors concluded that cerebral MD can be used to assess the
followed by an increase in glycerol concentration in 17 of 18 safe lower limit of CPP and that CPP management might be
patients who experienced a delayed ischemic neurologic individualized by MD rather than delivered to generic target
deficit (DIND) after SAH but in only 3 of 24 who did not.25 values. However, these conclusions have been challenged by
The mean delay from the peak of the LPR and LGR to the data from a later study.75 Although frequent and sustained
occurrence of the DIND was 23 hours (range 4 to 50 hours) increases in LPR and glutamate were observed in pericontu-
and the mean delay from the onset of a full ischemic pattern sional but not normal brain tissue of TBI patients, these
was 11 hours (range –11 hours to + 48 hours). The ability of abnormalities were not related to CPP alterations. In particu-
MD to predict the occurrence of DIND with high sensitivity lar CPP that decreased to less than 60 mm Hg was not associ-
and specificity warrants further investigation. In a recent pro- ated with signs of increased metabolic distress, and it also was
spective observational study Belli et al. analyzed the correla- not possible to identify an optimal CPP range using MD.
tion between MD markers metabolic impairment and changes However, MD markers of ischemia were improved by removal
in ICP after TBI.24 An LPR greater than 25 and glycerol greater of mass lesions, suggesting that although dynamic changes in
than 100 µmol/L were associated with a significantly greater MD values can occur with changes in CPP, there are persistent
Section VI—Intracranial Monitoring 363

40 Low CPP

30
LPR

20

B
10

Increased blood pressure

0
00:00 12:00 00:00 12:00
Time (hours)
Fig. 36.4  Graph illustrating the changes in the lactate/pyruvate ratio (LPR) in “at-risk” (A) and normal (B) brain during a period of low and normal
cerebral perfusion pressure. The normal range for the LPR is shown by the shaded area. Note the rise in LPR in the at-risk tissue during a period of
cerebral hypoperfusion with normal values measured by the catheter in normal brain tissue. CPP, cerebral perfusion pressure. (Modified with permission
from Tisdall MM, Smith M. Cerebral microdialysis: research technique or clinical tool. Br J Anaesth 2006;97(1):18–25.)

differences in regional brain chemistry that far outweigh the [>140 mg/dL]) occurs more frequently in patients with DIND
impact of global measures of perfusion. Although many than in asymptomatic patients but is unrelated to cerebral
studies confirm the presence of a biochemical “penumbral” glucose levels. In this study, both cerebral low-glucose epi-
zone surrounding focal traumatic brain lesions,75,93 it remains sodes (<0.6 mmol/L [<10.8 mg/dL]) and high-glucose epi-
to be proven whether therapeutic intervention, such as CPP- sodes (>2.6 mmol/L [>47 mg/dL]) occurred independently of
directed therapy, can protect this penumbral zone from further blood glucose concentration. However, low cerebral glucose
damage.74 occurred more frequently in symptomatic patients and was
associated with other signs of cellular distress such as increase
in LPR, glutamate, and glycerol concentrations. The combina-
Glycemic Control tion of low cerebral glucose and systemic hyperglycemia also
The association between hyperglycemia and outcome after was associated with unfavorable outcome. These findings
acute brain injury is well described. However, the optimal merit further investigation and suggest that established targets
management of blood glucose and its relationship to brain for glycemic control after brain injury might not be univer-
glucose is still to be fully elucidated. Manipulation of blood sally applicable and that cerebral glucose concentration may
glucose concentration can alter ECF glucose in both normal97 present a target for individualized glucose management.
and injured98 brain. Poor outcome in the acute phase after Further studies are required to identify which patients might
severe TBI is associated with high systemic glucose concentra- benefit from such intervention. Studies also suggest that more
tions99 but also with low cerebral MD glucose concentra- “moderate” systemic glucose control may be reasonable
tions.35 Vespa et al. observed that tight systemic glycemic because brain glucose increases once systemic glucose is
control (defined in this study as blood glucose concentration 7.8 mmol/L (140 mg/dL), whereas “tight” glucose control may
of 5.0-6.7 mmol/L [90-120 mg/dL]) did not improve func- increase the risk of cell energy dysfunction in the brain.90,91
tional outcome after TBI and was associated with an increased
incidence of MD markers of cellular distress.98 Similar find-
ings are observed in SAH patients.90 Schlenk et al. observed Hyperventilation
that insulin infusion was associated with stable blood glucose Hyperventilation (HV) can be used to control increased ICP
concentration but also with a significant decrease in cerebral after TBI, but to do so requires an additional monitor to
glucose.100 The brain glucose decrease is likely associated with ensure that it does not contribute to brain ischemia. Although
high brain glucose utilization because the LPR and glutamate there appears to be no role for prophylactic HV, brief periods
did not increase, effectively excluding ischemia as the cause of of HV therapy do not appear to worsen brain ischemia or
the low brain glucose concentration. Other investigators101 neurologic outcome, although this may depend on how or
have observed that systemic hyperglycemia (>7.8 mmol/L when HV is used. Marion et al. examined the effect of 30
364 Section VI—Intracranial Monitoring

minutes of HV (mean PaCO2 24.6 mm Hg) on the extracel- collected in the microdialysate. This opens the door to inves-
lular metabolites associated with ischemia after severe TBI.81 tigate novel biomarkers of brain injury; the possible applica-
HV trials were performed 24 to 36 hours after injury and again tions are immense and as yet unexplored.
at 3 to 4 days. MD lactate, pyruvate, and glutamate were mea- Brain ECF S100β has been successfully measured in vivo
sured before and for 4 hours after HV. At 24 to 36 hours, HV using MD, and increases in ECF S100β are related to second-
resulted in 13.7% to 395% increases in MD glutamate in 14 ary events, including intracranial hypertension after TBI and
of 20 patients, and increases in LPR of 10.8% to 227% in all vasospasm after SAH.107 The 100-kDa MD cutoff catheter
20 patients. Interestingly, these changes were not associated allows for the improved recovery of S100β.28 MD also has been
with a significant decrease in local cerebral blood flow (CBF). used to measure brain extracellular N-acetyl aspartate (eNAA)
At 3 to 4 days after injury, 10 of 13 patients had an increase after severe TBI.108 Levels of eNAA were 34% less in nonsur-
in glutamate, whereas only 3 of 13 had an increase in LPR. vivors compared with survivors, with a nonrecoverable
These data indicate that even brief periods of HV during the decrease in eNAA concentration from day 4 after injury
first 4 days after TBI can increase markers of cellular ischemia onward. This was associated with a rise in LPR and glycerol.
in selectively vulnerable regions of brain and that HV-induced Because NAA is synthesized in the mitochondria of neurons,
changes are much more common early after injury. It remains these results confirm the importance of mitochondrial dys-
to be seen whether these findings can translate into effective function as a contributor to poor outcome following TBI. The
titration of HV guided by changes in MD variables. measurement of eNAA also may have potential as a prognostic
marker after TBI and could be used to determine the efficacy
of therapeutic interventions aimed at mitochondrial function.
Testing New Treatment Interventions Severe TBI often is associated with axonal injury and loss.
MD also has been used to assess novel therapeutic interven- Neurofilament heavy chains (NF-H) are cytoskeletal proteins
tions. There is interest in normobaric hyperoxia (NBH) or found in axons, and studies show that with a 100-kDa MD
hyperbaric oxygen as a potential treatment strategy after cutoff membrane that two proteolytic breakdown products of
severe TBI. Some investigators102 but not all103 have observed NF-H, NfH(476-986) and NfH(476-1026), can be measured
that NBH is associated with improved markers of cerebral using cerebral MD with a relative recovery of 20%. In severe
metabolism after severe TBI. Two studies have used MD with TBI, Petzold et al. observed that the MD NF-H levels were
other monitoring modalities to investigate the underlying associated with the mechanism of injury and other physio-
mechanisms of action of NBH. Nortje et al. compared MD logic parameters and were of prognostic value.27 Other studies
and PbtO2 to 15O-PET variables within a region of interest suggest that MD monitoring of brain ECF total tau and
around MD catheters placed in at-risk tissue in 11 patients β-amyloid 1-42 proteins that are important biomarkers for
with severe TBI.84 MD lactate and pyruvate were unchanged axonal injury and for Alzheimer’s disease may help evaluate
during NBH and, although the LPR showed a statistically axonal injury after moderate to severe TBI.29
significant reduction, the magnitude of the reduction was The temporal profile of nitric oxide (NO) metabolite con-
small. Global 15O-PET variables were unchanged during NBH, centrations (i.e., nitrite and nitrate ([NOx]), also has been
but a universal increase in cerebral metabolic rate of oxygen investigated using MD.109 NOx concentrations demonstrated
was seen in at-risk tissue. These data suggest that NBH results a typical temporal profile after SAH of an initial peak followed
in significant enhancement of oxidative metabolism in tissue by an exponential decay. The decrease in NO metabolites over
that is physiologically at risk of nonsurvival. In another study, time was associated with altered MD-derived energy- or
MD variables were combined with near infrared spectroscopy damage-related compounds and could be related to reduced
measurement of changes in oxidized cytochrome c oxidase NO availability, potentially leading to an imbalance of vaso-
concentration.104 Cytochrome c oxidase is the terminal elec- dilatory and vasoconstrictive factors. In preliminary TBI
tron acceptor of the mitochondrial transfer chain and there- studies, higher concentrations of NO measured with cerebral
fore plays a crucial role in the dynamics of cellular oxygen MD are associated with more favorable metabolism.110 Because
utilization and energy production.105 Changes in cytochrome NO is a vasodilator, this effect may be associated with increased
c oxidase concentration have been validated as a marker of blood flow and delivery of oxygen and glucose.
cellular energy status.106 During short periods of hyperoxia MD catheters with high cutoff membranes allow macro-
(FiO2 = 1), MD lactate concentration and LPR were signifi- molecules to be sampled off-line.16 Changes in extracellular
cantly decreased, and this was associated with an increase in concentrations of cytokines, chemokines, and neurotrophic
the oxidation state of cytochrome c oxidase. This study con- factors have been described after brain injury.111 Multiple
firms oxidation in brain cellular and mitochondrial redox compounds, including interleukin (IL)-1β, IL-6, and IL-8;
state after TBI. The findings of both these studies are consis- macrophage inflammatory protein-1β; vascular endothelial
tent with increased aerobic metabolism and imply a preferen- growth factor; and fibroblast growth factor-2, have been mea-
tial metabolic benefit from hyperoxia. The clinical significance sured. This illustrates the opportunity to monitor many bio-
of these findings needs to be confirmed in future studies. chemical events simultaneously in the human brain and may
provide insight into events ranging from aneurysm size to
delayed ischemia,112 although the prognostic significance of
Research Applications MD-measured inflammatory markers has still to be eluci-
of Microdialysis dated.113 In addition MD allows multiple body fluids to be
examined (e.g., plasma, cerebrospinal fluid [CSF], and brain
Novel Biomarkers ECF). For example, Sarrafzadeh et al. observed that in SAH,
Any molecule present in the brain ECF that is small enough cerebral, but not plasma IL-6, levels were associated with
to cross the semipermeable dialysis membrane will be delayed cerebral ischemia (DCI) after SAH.114 Catheters with
Section VI—Intracranial Monitoring 365

high cutoff membranes have the potential to expand MD to local tissue biochemistry, and accurate placement of the cath-
the study of protein chemistry as a routine bedside method eter is crucial. Furthermore, because there are wide variations
during neurointensive care.115 in measured variables, trend data are more important than
absolute values. MD is used routinely in a few centers but it
has not yet been introduced into widespread clinical practice.
Drug Delivery Although clinical experience is rapidly increasing, carefully
The pharmacokinetics of drugs in plasma has been extensively designed prospective studies are required to determine the
studied; however, drug pharmacokinetics and pharmacody- value of cerebral MD in the management of brain-injured
namics in the human brain has received much less attention patients. However, because of its unique ability to contribute
because of the obvious difficulty in sampling. MD can be used important information about the processes of secondary
to investigate drug penetration in the human brain and brain injury, MD has the potential to become established as a
measure concentration at, or as close as possible to, the site of key component of multimodal monitoring during neuroin-
action. Many drugs have been studied using MD, and these tensive care. In addition, MD values may be used as surrogate
are reviewed in detail elsewhere.26 In one study of particular endpoints in the early phase clinical trials.
relevance to neurointensive care, Tisdall et al. investigated the
relationship between brain ECF free phenytoin concentration
measured with MD and plasma free phenytoin concentra-
References
tion.116 There was no relationship between microdialysate and 1. Hutchinson PJ. Microdialysis in traumatic brain injury: methodology and
pathophysiology. Acta Neurochir Suppl 2005;95:441–5.
plasma free phenytoin concentration, suggesting that in the 2. Tisdall MM, Smith M. Cerebral microdialysis: research technique or clinical
context of brain injury, measurement of plasma free phenyt- tool. Br J Anaesth 2006;97(1):18–25.
oin concentration may not provide an accurate reflection of 3. Bito L, Davson H, Levin E, et al. The concentrations of free amino acids and
regional brain ECF drug concentration. This has implications other electrolytes in cerebrospinal fluid, in vivo dialysate of brain, and blood
for dosing regimens that rely on measurement of plasma plasma of the dog. J Neurochem 1966;13(11):1057–67.
4. Delgado JM, DeFeudis FV, Roth RH, et al. Dialytrode for long term
levels. MD also may be used to measure muscle levels of drugs intracerebral perfusion in awake monkeys. Arch Int Pharmacodyn Ther
such as muscle relaxants to help examine the drug use and 1972;198(1):9–21.
effects in patients in the NCCU.117 Finally, it is possible to 5. Tossman U, Ungerstedt U. Microdialysis in the study of extracellular levels of
engineer the delivery of substances to the brain using MD, and amino acids in the rat brain. Acta Physiol Scand 1986;128(1):9–14.
6. Hillered L, Persson L, Ponten U, et al. Neurometabolic monitoring of the
this has many potential applications in the NCCU and in the ischaemic human brain using microdialysis. Acta Neurochir (Wien) 1990;
treatment of other disorders such as brain tumors.118-121 102(3-4):91–7.
7. Persson L, Hillered L. Chemical monitoring of neurosurgical intensive
care patients using intracerebral microdialysis. J Neurosurg 1992;76(1):
Proteomics 72–80.
8. Goodman JC, Robertson CS. Microdialysis: is it ready for prime time? Curr
A potentially exciting new field is the application of pro- Opin Crit Care 2009;15(2):110–17.
teomics to MD. Using a proteomic approach based on two- 9. Tisdall MM, Smith M. Multimodal monitoring in traumatic brain injury:
dimensional gel-electrophoresis and subsequent mass current status and future directions. Br J Anaesth 2007;99(1):61–7.
spectroscopy, Maurer et al. created a protein map for the 10. Hillered L, Vespa PM, Hovda DA. Translational neurochemical research in
acute human brain injury: the current status and potential future for
global protein expression pattern of the microdialysate in cerebral microdialysis. J Neurotrauma 2005;22(1):3–41.
three patients with thromboembolic stroke.122 Ten proteins 11. Johnston AJ, Gupta AK. Advanced monitoring in the neurology intensive
were found exclusively in the microdialysate (but not in CSF) care unit: microdialysis. Curr Opin Crit Care 2002;8(2):121–7.
and indicate the possibility of monitoring disease progression. 12. Landolt H, Langemann H. Cerebral microdialysis as a diagnostic tool in
In another study, protein expression profiles in microdialysate acute brain injury. Eur J Anaesthesiol 1996;13(3):269–78.
13. Lee GJ, Park JH, Park HK. Microdialysis applications in neuroscience. Neurol
samples were measured in SAH patients.123 Differences in Res 2008;30(7):661–8.
protein expression were detected in those with vasospasm 14. Peerdeman SM, van Tulder MW, Vandertop WP. Cerebral microdialysis as a
compared with those without, and the changes in protein monitoring method in subarachnoid hemorrhage patients, and correlation
concentrations were detected 3.8 days before the onset of with clinical events: a systematic review. J Neurol 2003;250(7):797–805.
15. Hutchinson PJ, O’Connell MT, Nortje J, et al. Cerebral microdialysis
symptoms. It therefore may become possible to use MD pro- methodology: evaluation of 20 kDa and 100 kDa catheters. Physiol Meas
teomic techniques to identify early markers for vasospasm and 2005;26(4):423–8.
enable selective early therapeutic intervention in this high-risk 16. Hillman J, Aneman O, Anderson C, et al. A microdialysis technique for
group of patients. routine measurement of macromolecules in the injured human brain.
Neurosurgery 2005;56(6):1264–8.
17. Hutchinson PJ, O’Connell MT, Al-Rawi PG, et al. Clinical cerebral
Conclusion microdialysis: a methodological study. J Neurosurg 2000;93(1):37–43.
18. Langemann H, Alessandri B, Mendelowitsch A, et al. Extracellular levels of
Cerebral MD presently is the only method available to measure glucose and lactate measured by quantitative microdialysis in the human
brain tissue biochemistry at the bedside, and the introduction brain. Neurol Res 2001;23(5):531–6.
19. Bhatia R, Hashemi P, Razzaq A, et al. Application of rapid-sampling, online
of a commercially available analyzer has made online cerebral microdialysis to the monitoring of brain metabolism during aneurysm
MD monitoring a reality in the NCCU. Its use has provided surgery. Neurosurgery 2006;58(4 Suppl 2):ONS–20.
insight into the pathophysiology after acute brain injury, and 20. Teasdale GM, Graham DI. Craniocerebral trauma: protection and retrieval
MD abnormalities are associated with worse clinical condition of the neuronal population after injury. Neurosurgery 1998;43(4):723–37.
and outcome. There also is accumulating evidence that MD 21. Smith MI. Neurocritical care: has it come of age? Br J Anaesth
2004;93(6):753–5.
may have the potential to provide early warning of impending 22. Adamides AA, Rosenfeldt FL, Winter CD, et al. Brain tissue lactate elevations
neurologic deterioration, thus allowing timely implementa- predict episodes of intracranial hypertension in patients with traumatic
tion of neuroprotective strategies. However, MD reflects only brain injury. J Am Coll Surg 2009;209(4):531–9.
366 Section VI—Intracranial Monitoring

23. Chen HI, Stiefel MF, Oddo M, et al. Detection of cerebral compromise with 37. Bergsneider M, Hovda DA, Shalmon E, et al. Cerebral hyperglycolysis
multimodality monitoring in patients with subarachnoid hemorrhage. following severe traumatic brain injury in humans: a positron emission
Neurosurgery 2011;69:53–63. tomography study. J Neurosurg 1997;86(2):241–51.
24. Belli A, Sen J, Petzold A, Russo S, et al. Metabolic failure precedes 38. Goodman JC, Valadka AB, Gopinath SP, et al. Extracellular lactate and
intracranial pressure rises in traumatic brain injury: a microdialysis study. glucose alterations in the brain after head injury measured by microdialysis.
Acta Neurochir (Wien) 2008;150(5):461–9. Crit Care Med 1999;27(9):1965–73.
25. Skjoth-Rasmussen J, Schulz M, Kristensen SR, et al. Delayed neurological 39. Unterberg AW, Sakowitz OW, Sarrafzadeh AS, et al. Role of bedside
deficits detected by an ischemic pattern in the extracellular cerebral microdialysis in the diagnosis of cerebral vasospasm following aneurysmal
metabolites in patients with aneurysmal subarachnoid hemorrhage. subarachnoid hemorrhage. J Neurosurg 2001;94(5):740–9.
J Neurosurg 2004;100(1):8–15. 40. Valadka AB, Goodman JC, Gopinath SP, et al. Comparison of brain tissue
26. Helmy A, Carpenter KL, Hutchinson PJ. Microdialysis in the human brain oxygen tension to microdialysis-based measures of cerebral ischemia in
and its potential role in the development and clinical assessment of drugs. fatally head-injured humans. J Neurotrauma 1998;15(7):509–19.
Curr Med Chem 2007;14(14):1525–37. 41. Robertson CS, Gopinath SP, Uzura M, et al. Metabolic changes in the brain
27. Petzold A, Tisdall MM, Girbes AR, et al. In vivo monitoring of neuronal loss during transient ischemia measured with microdialysis. Neurol Res 1998;20
in traumatic brain injury: a microdialysis study. Brain 2011;134(Pt 2): Suppl 1:S91–4.
464–83. 42. Vespa P, Bergsneider M, Hattori N, et al. Metabolic crisis without brain
28. Afinowi R, Tisdall M, Keir G, et al. Improving the recovery of S100β protein ischemia is common after traumatic brain injury: a combined microdialysis
in cerebral microdialysis: implications for multimodal monitoring in and positron emission tomography study. J Cereb Blood Flow Metab
neurocritical care. J Neurosci Methods 2009;181(1):95–9. 2005;25(6):763–74.
29. Marklund N, Blennow K, Zetterberg H, et al. Monitoring of brain interstitial 43. Samuelsson C, Hillered L, Enblad P, et al. Microdialysis patterns in
total tau and beta amyloid proteins by microdialysis in patients with subarachnoid hemorrhage patients with focus on ischemic events and brain
traumatic brain injury. J Neurosurg 2009;110(6):1227–37. interstitial glutamine levels. Acta Neurochir (Wien) 2009;151(5):437–46;
30. Reinstrup P, Stahl N, Mellergard P, et al. Intracerebral microdialysis in discussion 446. Epub 2009, Mar 19.
clinical practice: baseline values for chemical markers during wakefulness, 44. Boumezbeur F, Petersen KF, Cline GW, et al. The contribution of blood
anesthesia, and neurosurgery. Neurosurgery 2000;47(3):701–9. lactate to brain energy metabolism in humans measured by dynamic 13C
31. Schulz MK, Wang LP, Tange M, et al. Cerebral microdialysis monitoring: nuclear magnetic resonance spectroscopy. J Neurosci 2010;30(42):13983–91.
determination of normal and ischemic cerebral metabolisms in patients with 45. Pellerin L, Magistretti PJ. Food for thought: challenging the dogmas. J Cereb
aneurysmal subarachnoid hemorrhage. J Neurosurg 2000;93(5):808–14. Blood Flow Metab 2003;23(11):1282–6.
32. Charalambides C, Sgouros S, Sakas D. Intracerebral microdialysis in children. 46. Gallagher CN, Carpenter KL, Grice P, et al. The human brain utilizes lactate
Childs Nerv Syst 2010;26(2):215–20. via the tricarboxylic acid cycle: a 13C-labelled microdialysis and high-
33 Larach DB, Kofke WA, Le Roux P. Potential non-hypoxic/ischemic causes resolution nuclear magnetic resonance study. Brain 2009;132(10):2839–49.
of increased cerebral interstitial fluid lactate/pyruvate ratio: a review of 47. Hutchinson PJ, Al-Rawi PG, O’Connell MT, et al. Monitoring of brain
available literature. Neurocrit Care 2011;Feb 19. Epub ahead of print. metabolism during aneurysm surgery using microdialysis and brain
34. Nelson DW, Thornquist B, MacCallum RM, et al. Analyses of cerebral multiparameter sensors. Neurol Res 1999;21(4):352–8.
microdialysis in patients with traumatic brain injury: relations to 48. Nilsson OG, Polito A, Saveland H, et al. Are primary supratentorial
intracranial pressure, cerebral perfusion pressure and catheter placement. intracerebral hemorrhages surrounded by a biochemical penumbra? A
BMC Med 2011 2;9:21. microdialysis study. Neurosurgery 2006;59(3):521–8.
35. Vespa PM, McArthur D, O’Phelan K, et al. Persistently low extracellular 49. Persson L, Valtysson J, Enblad P, et al. Neurochemical monitoring using
glucose correlates with poor outcome 6 months after human traumatic brain intracerebral microdialysis in patients with subarachnoid hemorrhage.
injury despite a lack of increased lactate: a microdialysis study. J Cereb Blood J Neurosurg 1996;84(4):606–16.
Flow Metab 2003;23(7):865–77. 50. Persson L, Hillered L. Intracerebal microdialysis. J Neurosurg 2008;85:984–5.
36. Hovda DA, Yoshino A, Kawamata T, et al. Diffuse prolonged depression of
cerebral oxidative metabolism following concussive brain injury in the rat: A complete list of references for this chapter can be found online at
a cytochrome oxidase histochemistry study. Brain Res 1991;567(1):1–10. www.expertconsult.com.
Section VI—Intracranial Monitoring 366.e1

References 29. Marklund N, Blennow K, Zetterberg H, et al. Monitoring of brain


interstitial total tau and beta amyloid proteins by microdialysis in patients
1. Hutchinson PJ. Microdialysis in traumatic brain injury: methodology and with traumatic brain injury. J Neurosurg 2009;110(6):1227–37.
pathophysiology. Acta Neurochir Suppl 2005;95:441–5. 30. Reinstrup P, Stahl N, Mellergard P, et al. Intracerebral microdialysis in
2. Tisdall MM, Smith M. Cerebral microdialysis: research technique or clinical clinical practice: baseline values for chemical markers during wakefulness,
tool. Br J Anaesth 2006;97(1):18–25. anesthesia, and neurosurgery. Neurosurgery 2000;47(3):701–9.
3. Bito L, Davson H, Levin E, et al. The concentrations of free amino acids 31. Schulz MK, Wang LP, Tange M, et al. Cerebral microdialysis monitoring:
and other electrolytes in cerebrospinal fluid, in vivo dialysate of brain, and determination of normal and ischemic cerebral metabolisms in patients with
blood plasma of the dog. J Neurochem 1966;13(11):1057–67. aneurysmal subarachnoid hemorrhage. J Neurosurg 2000;93(5):808–14.
4. Delgado JM, DeFeudis FV, Roth RH, et al. Dialytrode for long term 32. Charalambides C, Sgouros S, Sakas D. Intracerebral microdialysis in
intracerebral perfusion in awake monkeys. Arch Int Pharmacodyn Ther children. Childs Nerv Syst 2010;26(2):215–20.
1972;198(1):9–21. 33. Larach DB, Kofke WA, Le Roux P. Potential non-hypoxic/ischemic causes
5. Tossman U, Ungerstedt U. Microdialysis in the study of extracellular levels of increased cerebral interstitial fluid lactate/pyruvate ratio: a review of
of amino acids in the rat brain. Acta Physiol Scand 1986;128(1):9–14. available literature. Neurocrit Care 2011;Feb 19. Epub ahead of print.
6. Hillered L, Persson L, Ponten U, et al. Neurometabolic monitoring of the 34. Nelson DW, Thornquist B, MacCallum RM, et al. Analyses of cerebral
ischaemic human brain using microdialysis. Acta Neurochir (Wien) microdialysis in patients with traumatic brain injury: relations to
1990;102(3-4):91–7. intracranial pressure, cerebral perfusion pressure and catheter placement.
7. Persson L, Hillered L. Chemical monitoring of neurosurgical intensive BMC Med 2011 2;9:21.
care patients using intracerebral microdialysis. J Neurosurg 1992;76(1): 35. Vespa PM, McArthur D, O’Phelan K, et al. Persistently low extracellular
72–80. glucose correlates with poor outcome 6 months after human traumatic
8. Goodman JC, Robertson CS. Microdialysis: is it ready for prime time? Curr brain injury despite a lack of increased lactate: a microdialysis study. J
Opin Crit Care 2009;15(2):110–17. Cereb Blood Flow Metab 2003;23(7):865–77.
9. Tisdall MM, Smith M. Multimodal monitoring in traumatic brain injury: 36. Hovda DA, Yoshino A, Kawamata T, et al. Diffuse prolonged depression of
current status and future directions. Br J Anaesth 2007;99(1):61–7. cerebral oxidative metabolism following concussive brain injury in the rat:
10. Hillered L, Vespa PM, Hovda DA. Translational neurochemical research in a cytochrome oxidase histochemistry study. Brain Res 1991;567(1):1–10.
acute human brain injury: the current status and potential future for 37. Bergsneider M, Hovda DA, Shalmon E, et al. Cerebral hyperglycolysis
cerebral microdialysis. J Neurotrauma 2005;22(1):3–41. following severe traumatic brain injury in humans: a positron emission
11. Johnston AJ, Gupta AK. Advanced monitoring in the neurology intensive tomography study. J Neurosurg 1997;86(2):241–51.
care unit: microdialysis. Curr Opin Crit Care 2002;8(2):121–7. 38. Goodman JC, Valadka AB, Gopinath SP, et al. Extracellular lactate and
12. Landolt H, Langemann H. Cerebral microdialysis as a diagnostic tool in glucose alterations in the brain after head injury measured by microdialysis.
acute brain injury. Eur J Anaesthesiol 1996;13(3):269–78. Crit Care Med 1999;27(9):1965–73.
13. Lee GJ, Park JH, Park HK. Microdialysis applications in neuroscience. 39. Unterberg AW, Sakowitz OW, Sarrafzadeh AS, et al. Role of bedside
Neurol Res 2008;30(7):661–8. microdialysis in the diagnosis of cerebral vasospasm following aneurysmal
14. Peerdeman SM, van Tulder MW, Vandertop WP. Cerebral microdialysis as a subarachnoid hemorrhage. J Neurosurg 2001;94(5):740–9.
monitoring method in subarachnoid hemorrhage patients, and correlation 40. Valadka AB, Goodman JC, Gopinath SP, et al. Comparison of brain tissue
with clinical events: a systematic review. J Neurol 2003;250(7):797–805. oxygen tension to microdialysis-based measures of cerebral ischemia in
15. Hutchinson PJ, O’Connell MT, Nortje J, et al. Cerebral microdialysis fatally head-injured humans. J Neurotrauma 1998;15(7):509–19.
methodology: evaluation of 20 kDa and 100 kDa catheters. Physiol Meas 41. Robertson CS, Gopinath SP, Uzura M, et al. Metabolic changes in the brain
2005;26(4):423–8. during transient ischemia measured with microdialysis. Neurol Res 1998;
16. Hillman J, Aneman O, Anderson C, et al. A microdialysis technique for 20(Suppl 1):S91–4.
routine measurement of macromolecules in the injured human brain. 42. Vespa P, Bergsneider M, Hattori N, et al. Metabolic crisis without brain
Neurosurgery 2005;56(6):1264–8. ischemia is common after traumatic brain injury: a combined microdialysis
17. Hutchinson PJ, O’Connell MT, Al-Rawi PG, et al. Clinical cerebral and positron emission tomography study. J Cereb Blood Flow Metab 2005;
microdialysis: a methodological study. J Neurosurg 2000;93(1):37–43. 25(6):763–74.
18. Langemann H, Alessandri B, Mendelowitsch A, et al. Extracellular levels of 43. Samuelsson C, Hillered L, Enblad P, et al. Microdialysis patterns in
glucose and lactate measured by quantitative microdialysis in the human subarachnoid hemorrhage patients with focus on ischemic events and brain
brain. Neurol Res 2001;23(5):531–6. interstitial glutamine levels. Acta Neurochir (Wien) 2009;151(5):437–46;
19. Bhatia R, Hashemi P, Razzaq A, et al. Application of rapid-sampling, online discussion 446. Epub 2009, Mar 19.
microdialysis to the monitoring of brain metabolism during aneurysm 44. Boumezbeur F, Petersen KF, Cline GW, et al. The contribution of blood
surgery. Neurosurgery 2006;58(4 Suppl 2):ONS–20. lactate to brain energy metabolism in humans measured by dynamic 13C
20. Teasdale GM, Graham DI. Craniocerebral trauma: protection and retrieval nuclear magnetic resonance spectroscopy. J Neurosci
of the neuronal population after injury. Neurosurgery 1998;43(4):723–37. 2010;30(42):13983–91.
21. Smith MI. Neurocritical care: has it come of age? Br J Anaesth 2004;93(6): 45. Pellerin L, Magistretti PJ. Food for thought: challenging the dogmas. J
753–5. Cereb Blood Flow Metab 2003;23(11):1282–6.
22. Adamides AA, Rosenfeldt FL, Winter CD, et al. Brain tissue lactate 46. Gallagher CN, Carpenter KL, Grice P, et al. The human brain utilizes lactate
elevations predict episodes of intracranial hypertension in patients with via the tricarboxylic acid cycle: a 13C-labelled microdialysis and high-
traumatic brain injury. J Am Coll Surg 2009;209(4):531–9. resolution nuclear magnetic resonance study. Brain 2009;132(10):2839–49.
23. Chen HI, Stiefel MF, Oddo M, et al. Detection of cerebral compromise with 47. Hutchinson PJ, Al-Rawi PG, O’Connell MT, et al. Monitoring of brain
multimodality monitoring in patients with subarachnoid hemorrhage. metabolism during aneurysm surgery using microdialysis and brain
Neurosurgery 2011;69:53–63. multiparameter sensors. Neurol Res 1999;21(4):352–8.
24. Belli A, Sen J, Petzold A, Russo S, et al. Metabolic failure precedes 48. Nilsson OG, Polito A, Saveland H, et al. Are primary supratentorial
intracranial pressure rises in traumatic brain injury: a microdialysis study. intracerebral hemorrhages surrounded by a biochemical penumbra? A
Acta Neurochir (Wien) 2008;150(5):461–9. microdialysis study. Neurosurgery 2006;59(3):521–8.
25. Skjoth-Rasmussen J, Schulz M, Kristensen SR, et al. Delayed neurological 49. Persson L, Valtysson J, Enblad P, et al. Neurochemical monitoring using
deficits detected by an ischemic pattern in the extracellular cerebral intracerebral microdialysis in patients with subarachnoid hemorrhage. J
metabolites in patients with aneurysmal subarachnoid hemorrhage. Neurosurg 1996;84(4):606–16.
J Neurosurg 2004;100(1):8–15. 50. Persson L, Hillered L. Intracerebal microdialysis. J Neurosurg
26. Helmy A, Carpenter KL, Hutchinson PJ. Microdialysis in the human brain 2008;85:984–5.
and its potential role in the development and clinical assessment of drugs. 51. Enblad P, Valtysson J, Andersson J, et al. Simultaneous intracerebral
Curr Med Chem 2007;14(14):1525–37. microdialysis and positron emission tomography in the detection of
27. Petzold A, Tisdall MM, Girbes AR, et al. In vivo monitoring of neuronal ischemia in patients with subarachnoid hemorrhage. J Cereb Blood Flow
loss in traumatic brain injury: a microdialysis study. Brain 2011; Metab 1996;6(4):637–44.
134(Pt 2):464–83. 52. Stahl N, Mellergard P, Hallstrom A, et al. Intracerebral microdialysis and
28. Afinowi R, Tisdall M, Keir G, et al. Improving the recovery of S100β bedside biochemical analysis in patients with fatal traumatic brain lesions.
protein in cerebral microdialysis: implications for multimodal monitoring Acta Anaesthesiol Scand 2001;45(8):977–85.
in neurocritical care. J Neurosci Methods 2009;181(1):95–9.
366.e2 Section VI—Intracranial Monitoring

53. Zetterling M, Hallberg L, Hillered L, et al. Brain energy metabolism in 78. Vespa PM. The implications of cerebral ischemia and metabolic
patients with spontaneous subarachnoid hemorrhage and global cerebral dysfunction for treatment strategies in neurointensive care. Curr Opin Crit
edema. Neurosurgery 2010;66(6):1102–10. Care 2006;12(2):119–23.
54. Benveniste H. Brain microdialysis. J Neurochem 1989;52(6):1667–79. 79. Hutchinson PJ, Hutchinson DB, Barr RH, et al. A new cranial access device
55. Hutchinson PJ, Gupta AK, Fryer TF, et al. Correlation between cerebral for cerebral monitoring. Br J Neurosurg 2000;14(1):46–8.
blood flow, substrate delivery, and metabolism in head injury: a combined 80. Nordstrom CH, Reinstrup P, Xu W, et al. Assessment of the lower
microdialysis and triple oxygen positron emission tomography study. J limit for cerebral perfusion pressure in severe head injuries by bedside
Cereb Blood Flow Metab 2002;22(6):735–45. monitoring of regional energy metabolism. Anesthesiology 2003;98(4):
56. Timofeev I, Carpenter KL, Nortje J, et al. Cerebral extracellular chemistry 809–14.
and outcome following traumatic brain injury: a microdialysis study of 223 81. Marion DW, Puccio A, Wisniewski SR, et al. Effect of hyperventilation on
patients. Brain 2011;134(Pt 2):484–94. extracellular concentrations of glutamate, lactate, pyruvate, and local
57. Hutchinson PJ, Al-Rawi PG, O’Connell MT, et al. Head injury monitoring cerebral blood flow in patients with severe traumatic brain injury. Crit Care
using cerebral microdialysis and Paratrend multiparameter sensors. Med 2002;30(12):2619–25.
Zentralbl Neurochir 2000;61(2):88–94. 82. Boret H, Fesselet J, Meaudre E, et al. Cerebral microdialysis and P(ti)O2 for
58. Zauner A, Doppenberg EM, Woodward JJ, et al. Continuous monitoring of neuro-monitoring before decompressive craniectomy. Acta Anaesthesiol
cerebral substrate delivery and clearance: initial experience in 24 patients Scand 2006;50(2):252–4.
with severe acute brain injuries. Neurosurgery 1997;41(5):1082–91. 83. Yokobori S, Watanabe A, Matsumoto G, et al. Time course of recovery from
59. Kett-White R, Hutchinson PJ, Al-Rawi PG, et al. Adverse cerebral events cerebral vulnerability after severe traumatic brain injury: a microdialysis
detected after subarachnoid hemorrhage using brain oxygen and study. J Trauma 2011;Apr 15. Epub ahead of print.
microdialysis probes. Neurosurgery 2002;50(6):1213–21. 84. Nortje J, Coles JP, Timofeev I, et al. Effect of hyperoxia on regional
60. Sarrafzadeh A, Haux D, Kuchler I, et al. Poor-grade aneurysmal oxygenation and metabolism after severe traumatic brain injury:
subarachnoid hemorrhage: relationship of cerebral metabolism to outcome. preliminary findings. Crit Care Med 2008;36(1):273–81.
J Neurosurg 2004;100(3):400–6. 85. Schneweis S, Grond M, Staub F, et al. Predictive value of neurochemical
61. Johnston AJ, Steiner LA, Coles JP, et al. Effect of cerebral perfusion pressure monitoring in large middle cerebral artery infarction. Stroke 2001;32:
augmentation on regional oxygenation and metabolism after head injury. 1863–7.
Crit Care Med 2005;33(1):189–95. 86. Oddo M, Frangos S, Milby A, et al. Induced normothermia attenuates
62. Friess SH, Ralston J, Eucker SA, et al. Neurocritical care monitoring cerebral metabolic distress in patients with aneurysmal subarachnoid
correlates with neuropathology in a swine model of pediatric traumatic hemorrhage and refractory fever. Stroke 2009;40(5):1913–6.
brain injury. Neurosurgery 2011;Jun 10. Epub ahead of print. 87. Oddo M, Frangos S, Maloney-Wilensky E, et al. Effect of shivering on brain
63. Clausen T, Alves OL, Reinert M, et al. Association between elevated brain tissue oxygenation during induced normothermia in patients with severe
tissue glycerol levels and poor outcome following severe traumatic brain brain injury. Neurocrit Care 2010;12(1):10–6.
injury. J Neurosurg 2005;103(2):233–8. 88. Oddo M, Milby A, Chen I, et al. Hemoglobin concentration and cerebral
64. Hillered L, Valtysson J, Enblad P, et al. Interstitial glycerol as a marker for metabolism in patients with aneurysmal subarachnoid hemorrhage: a
membrane phospholipid degradation in the acutely injured human brain. microdialysis study. Stroke 2009;40(4):1275–81.
J Neurol Neurosurg Psychiatry 1998;64(4):486–91. 89. Kurtz P, Schmidt JM, Claassen J, et al. Anemia is associated with metabolic
65. Peerdeman SM, Girbes AR, Polderman KH, et al. Changes in cerebral distress and brain tissue hypoxia after subarachnoid hemorrhage. Neurocrit
interstitial glycerol concentration in head-injured patients: correlation with Care 2010;13:10–6.
secondary events. Intensive Care Med 2003;29(10):1825–8. 90. Oddo M, Schmidt JM, Carrera C, et al. Impact of tight glycemic control on
66. Konig K, Rickels E, Heissler HE, et al. Artificial elevation of brain tissue cerebral glucose metabolism after severe brain injury: a microdialysis study.
glycerol by administration of a glycerol-containing agent: case report. Crit Care Med 2008;36:3233–8.
J Neurosurg 2001;94(4):621–3. 91. Schlenk F, Vajkoczy P, Sarrafzadeh A. Inpatient hyperglycemia following
67. Benveniste H, Drejer J, Schousboe A, et al. Elevation of the extracellular aneurysmal subarachnoid hemorrhage: relation to cerebral metabolism and
concentrations of glutamate and aspartate in rat hippocampus during outcome. Neurocrit Care 2009;11(1):56–63. Epub 2009, May 6.
transient cerebral ischemia monitored by intracerebral microdialysis. 92. Schlenk F, Frieler K, Nagel A, et al. Cerebral microdialysis for detection of
J Neurochem 1984;43(5):1369–74. bacterial meningitis in aneurysmal subarachnoid hemorrhage patients: a
68. Nilsson P, Hillered L, Ponten U, et al. Changes in cortical extracellular levels cohort study. Crit Care 2009;13(1):R2. Epub 2009, Jan 20.
of energy-related metabolites and amino acids following concussive brain 93. Hlatky R, Valadka AB, Goodman JC, et al. Evolution of brain tissue injury
injury in rats. J Cereb Blood Flow Metab 1990;10(5):631–7. after evacuation of acute traumatic subdural hematomas. Neurosurgery
69. Staub F, Graf R, Gabel P, et al. Multiple interstitial substances measured by 2004;55(6):1318–23.
microdialysis in patients with subarachnoid hemorrhage. Neurosurgery 94. Sarrafzadeh AS, Haux D, Ludemann L, et al. Cerebral ischemia in
2000;47(5):1106–15. aneurysmal subarachnoid hemorrhage: a correlative microdialysis-PET
70. Hutchinson PJ, Al-Rawi PG, O’Connell MT, et al. On-line monitoring of study. Stroke 2004;35(3):638–43.
substrate delivery and brain metabolism in head injury. Acta Neurochir 95. Nilsson OG, Brandt L, Ungerstedt U, et al. Bedside detection of brain
Suppl 2000;76:431–5. ischemia using intracerebral microdialysis: subarachnoid hemorrhage and
71. Vespa P, Prins M, Ronne-Engstrom E, et al. Increase in extracellular delayed ischemic deterioration. Neurosurgery 1999;45(5):1176–84.
glutamate caused by reduced cerebral perfusion pressure and seizures after 96. Gallagher CN, Carpenter KL, Grice P, et al. The human brain utilizes lactate
human traumatic brain injury: a microdialysis study. J Neurosurg 1998; via the tricarboxylic acid cycle: a 13C-labelled microdialysis and high-
89(6):971–82. resolution nuclear magnetic resonance study. Brain 2009;132(Pt 10):
72. Obrenovitch TP, Urenjak J. Is high extracellular glutamate the key to 2839–49.
excitotoxicity in traumatic brain injury? J Neurotrauma 1997;14(10):677–98. 97. Abi-Saab WM, Maggs DG, Jones T, et al. Striking differences in glucose and
73. Chamoun R, Suki D, Gopinath SP, et al. Role of extracellular glutamate lactate levels between brain extracellular fluid and plasma in conscious
measured by cerebral microdialysis in severe traumatic brain injury. J human subjects: effects of hyperglycemia and hypoglycemia. J Cereb Blood
Neurosurg 2010;113(3):564–70. Flow Metab 2002;22(3):271–9.
74. Engstrom M, Polito A, Reinstrup P, et al. Intracerebral microdialysis in 98. Vespa P, Boonyaputthikul R, McArthur DL, et al. Intensive insulin therapy
severe brain trauma: the importance of catheter location. J Neurosurg 2005; reduces microdialysis glucose values without altering glucose utilization or
102(3):460–9. improving the lactate/pyruvate ratio after traumatic brain injury. Crit Care
75. Vespa PM, O’Phelan K, McArthur D, et al. Pericontusional brain tissue Med 2006;34(3):850–6.
exhibits persistent elevation of lactate/pyruvate ratio independent of 99. Jeremitsky E, Omert LA, Dunham CM, et al. The impact of hyperglycemia
cerebral perfusion pressure. Crit Care Med 2007;35(4):1153–60. on patients with severe brain injury. J Trauma 2005;58(1):47–50.
76. Timofeev I, Czosnyka M, Carpenter K, et al. Interaction between brain 100. Schlenk F, Graetz D, Nagel A, et al. Insulin-related decrease in cerebral
chemistry and physiology after traumatic brain injury: impact of glucose despite normoglycemia in aneurysmal subarachnoid hemorrhage.
autoregulation and microdialysis catheter location. J Neurotrauma Crit Care 2008;12(1):R9.
2011;Apr 13. Epub ahead of print. 101. Schlenk F, Nagel A, Graetz D, et al Hyperglycemia and cerebral glucose in
77. Bellander BM, Cantais E, Enblad P, et al. Consensus meeting on aneurysmal subarachnoid hemorrhage. Intensive Care Med
microdialysis in neurointensive care. Intensive Care Med 2008;34(7):1200–7.
2004;30(12):2166–69.
Section VI—Intracranial Monitoring 366.e3

102. Tolias CM, Reinert M, Seiler R, et al. Normobaric hyperoxia–induced 112. Hanafy KA, Grobelny B, Fernandez L, et al. Brain interstitial fluid
improvement in cerebral metabolism and reduction in intracranial pressure TNF-alpha after subarachnoid hemorrhage. J Neurol Sci
in patients with severe head injury: a prospective historical cohort-matched 2010;291(1-2):69–73.
study. J Neurosurg 2004;101(3):435–44. 113. Cederberg D, Siesjö P. What has inflammation to do with traumatic brain
103. Magnoni S, Ghisoni L, Locatelli M, et al. Lack of improvement in cerebral injury? Childs Nerv Syst 2010;26(2):221–6.
metabolism after hyperoxia in severe head injury: a microdialysis study. J 114. Sarrafzadeh A, Schlenk F, Gericke C, et al. Relevance of cerebral
Neurosurg 2003;98(5):952–8. interleukin-6 after aneurysmal subarachnoid hemorrhage. Neurocrit Care
104. Tisdall MM, Tachtsidis I, Leung TS, et al. Increase in cerebral aerobic 2010;13(3):339–46.
metabolism by normobaric hyperoxia after traumatic brain injury. J 115. Hillman J, Aneman O, Persson M, et al. Variations in the response of
Neurosurg 2008;109:424–32. interleukins in neurosurgical intensive care patients monitored using
105. Richter OM, Ludwig B. Cytochrome c oxidase—structure, function, and intracerebral microdialysis. J Neurosurg 2007;106(5):820–5.
physiology of a redox-driven molecular machine. Rev Physiol Biochem 116. Tisdall M, Russo S, Sen J, et al. Free phenytoin concentration measurement
Pharmacol 2003;147:47–74. in brain extracellular fluid: a pilot study. Br J Neurosurg 2006;20(5):285–9.
106. Springett R, Wylezinska M, Cady EB, et al. Oxygen dependency of cerebral 117. Fernández-Candil J, Gambús PL, Trocóniz IF, et al. Feasibility of
oxidative phosphorylation in newborn piglets. J Cereb Blood Flow Metab microdialysis to determine interstitial rocuronium concentration in the
2000;20(2):280–9. muscle tissue of anesthetized neurosurgical patients. J Neurosurg
107. Sen J, Belli A, Petzold A, Russo S, et al. Extracellular fluid S100β in the Anesthesiol 2010;22(2):163–9.
injured brain: a future surrogate marker of acute brain injury? Acta 118. Redgate ES, Boggs S, Grudziak A, et al. Polyamines in brain tumor therapy.
Neurochir (Wien) 2005;147(8):897–900. J Neurooncol 1995;25(2):167–79.
108. Belli A, Sen J, Petzold A, et al. Extracellular N-acetylaspartate depletion in 119. Ronquist G, Hugosson R, Sjolander U, et al. Treatment of malignant glioma
traumatic brain injury. J Neurochem 2006;96(3):861–9. by a new therapeutic principle. Acta Neurochir (Wien) 1992;114(1-2):8–11.
109. Sakowitz OW, Wolfrum S, Sarrafzadeh AS, et al. Temporal profiles of 120. Marcus HJ, Carpenter KL, Price SJ, et al. In vivo assessment of high-grade
extracellular nitric oxide metabolites following aneurysmal subarachnoid glioma biochemistry using microdialysis: a study of energy-related
hemorrhage. Acta Neurochir Suppl 2002;81:351–4. molecules, growth factors and cytokines. J Neurooncol 2010;97(1):11–23.
110. Carpenter KL, Timofeev I, Al-Rawi PG, et al. Nitric oxide in acute brain 121. Blakeley JO, Olson J, Grossman SA, et al. New Approaches to Brain Tumor
injury: a pilot study of NO(x) concentrations in human brain Therapy (NABTT) Consortium. Effect of blood brain barrier permeability
microdialysates and their relationship with energy metabolism. Acta in recurrent high grade gliomas on the intratumoral pharmacokinetics of
Neurochir Suppl 2008;102:207–13. methotrexate: a microdialysis study. J Neurooncol 2009;91(1):51–8. Epub
111. Mellergard P, Aneman O, Sjogren F, et al. Changes in extracellular 2008, Sep 12.
concentrations of some cytokines, chemokines, and neurotrophic factors 122. Maurer MH, Berger C, Wolf M, et al. The proteome of human brain
after insertion of intracerebral microdialysis catheters in neurosurgical microdialysate. Proteome Sci 2003;1(1):7.
patients. Neurosurgery 2008;62(1):151–7. 123. Maurer MH, Haux D, Sakowitz OW, et al. Identification of early markers
for symptomatic vasospasm in human cerebral microdialysate after
Chapter
37  
VI

Brain Temperature
Nino Stocchetti and Elisa R. Zanier

Maintaining a constant brain temperature depends on a


Introduction “tight” balance between heat production and heat dissipa­
Mammals have evolved sophisticated thermoregulatory sys­ tion. The brain dissipates heat primarily to the systemic cir­
tems to maintain body temperature within an optimal range culation. Blood entering the brain is at core temperature,
for cellular biochemical reactions. Under normal conditions, which is lower than brain temperature. Heat is transferred
excluding hibernating mammals, core temperature is main­ from the brain to the circulating blood. Increased cerebral
tained around 37° C. However, body temperature fluctuates blood flow (CBF) increases heat transfer and therefore brain
over the day with an early morning nadir and peak in the cooling. As a result of this heat transfer, the temperature of
evening and with activity. Temperature also is lower in older venous blood leaving the brain in the internal jugular vein is
individuals and males and will vary according to the site at greater than core blood temperature.3 On average, the gradi­
which it is measured. Mean rectal temperature is between ent between brain and body core temperature is modest,
36.7° C and 37.5° C, whereas mean axillary temperatures are ranging from 0.3° C to 1.1° C,4-8 and usually is higher in the
between 35.5° C and 37° C. Minor fluctuations around the brain. There is a reasonable correlation between BT and sys­
set-point of 37.5° C are well tolerated, but variation that temic measures of temperature; however, this relationship
exceeds a few degrees may be deleterious. Body temperature changes at the extremes of the physiologic temperature range
results from a balance between mechanisms for heat produc­ such that the brain-body gradient may be reversed with
tion and mechanisms for heat dispersion. These mechanisms brain hypothermia or hyperthermia.9 Some consider a rever­
are coordinated by the central nervous system, specifically, by sal of this gradient a poor prognostic sign in traumatic brain
the preoptic and anterior hypothalamic nuclei.1 The brain injury (TBI).10
itself is particularly sensitive to temperature. In general, In some species, anatomic variations in brain vasculature
hyperthermia is associated with exacerbation of brain injury, improve cooling efficiency. In dogs, goats, and sheep the inter­
whereas induced hypothermia, or even prevention of fever nal carotid artery may be small or absent, and blood circulates
may be neuroprotective.2 Brain temperature (BT) may there­ to the skull base via the external carotid artery. Before entering
fore be an important therapeutic target; however, in clinical the circle of Willis, the external carotid artery divides into a
practice, core body temperature is commonly used as a series of small arteries, “the carotid rete,” or “rete mirabilis,”
surrogate. which flow through the cavernous sinus. During panting,
This chapter reviews the physiology of brain temperature evaporation in the oral and nasal cavities lowers venous blood
regulation, literature regarding the clinical impact of tempera­ temperature in the cavernous sinus. Heat then is transferred
ture abnormalities on brain injury, and methods to monitor from arterial blood entering the brain in the carotid rete to
brain temperature. In addition, methods to measure body venous blood in the cavernous sinus, thus facilitating heat
temperature are briefly reviewed. removal from the brain.
Heat also may be dispersed outward by direct conduction
through the calvaria,11 but the skull acts as an insulator,
Brain Temperature Under impeding efficient heat exchange. However, when the skull
Physiologic Conditions is opened, for example, after decompressive craniectomy, a
temperature gradient forms between exposed (noninsulated)
The Brain as a Heater brain and ambient temperature; this may result in a BT that
All metabolic processes produce heat. For example, glucose is lower than core temperature.4,12,13
and oxygen are converted via the Krebs cycle into adenosine
triphosphate (ATP), water, and heat. The brain consumes 25%
of total body glucose and 20% of total body oxygen for ATP Brain Temperature Is Heterogeneous
production. About 43% of the energy contained in glucose is There is spatial variation in brain temperature. Structures that
converted to ATP, and 67% is dissipated as heat. Given its high are more metabolically active or deeper have higher tempera­
metabolic activity, the brain generates a considerable amount tures.3 A thermal gradient exists within the brain that allows
of heat. heat transfer from deep gray structures to white matter to
© Copyright 2013 Elsevier Inc. All rights reserved. 367
368 Section VI—Intracranial Monitoring

cortex. This has been confirmed in human volunteers by increase in ICP may be observed with fever in patients with
proton magnetic resonance spectroscopy.14 Furthermore, acute brain damage.4,21 This ICP increase may be a surrogate
when temperature probes are withdrawn from the lateral ven­ for increased fuel delivery to the brain in the setting of
tricle to the brain surface, slight (nonstatistically significant) increased metabolic demand. It has been demonstrated that
temperature differences are detected.15 In patients with hydro­ during fever there may be preservation of a normal cerebral
cephalus, BT has been measured at various depths beneath lactate/pyruvate ratio (measured by microdialysis) indicating
the pial surface. Temperature increases gradually as a function adequate substrate delivery.21 However, once maximal cerebral
of depth, and the highest temperature is measured in the vasodilation occurs, further increases in CMRO2 may not be
ventricle.16 compensated by further increases in CBF and there may be
cellular energy crisis. On the other hand microdialysis studies
show that induced normothermia can attenuate the fever-
Brain Temperature Is Dynamic associated metabolic distress.22
Because the determinants of brain temperature are dynamic, The relationship between BT and ICP is not simple. A clear
brain temperature is dynamic. Heat production is determined relationship does not exist when cumulative BT and ICP data
by cerebral metabolic rate, and heat removal depends on are pooled.8,23 In a population, ICP depends on several factors
regional CBF and arterial blood temperature. Each of these and cannot be predicted solely by BT. However, in certain
variables fluctuates. The first BT measurements were obtained individual cases an association between ICP and BT may be
in animals in which large BT fluctuations—up to 2° C to detected.21
3° C —were observed on exposure to various environmental
challenges and upon performance of different behaviors and
tasks.3 Hyperthermia Is Deleterious in
When systemic temperature increases abruptly, e.g., in the Experimental Models of Brain Injury
heat shock, high fever, or strenuous physical exercise, systemic In the experimental setting it has been demonstrated that
arterial temperature may increase to a point where the normal hyperthermia aggravates neuronal injury after traumatic24 or
temperature gradient between blood and brain disappears or ischemic insults to the brain.25-28 Even mild hyperthermia
is even reversed. Conversely, BT may decrease below core tem­ may exacerbate outcome.29 In animal models of focal cerebral
perature when brain metabolism is inhibited, for example, ischemia, infarct volume varies with temperature30,31 and any
profound general anesthesia.3 Another “special case” is the deleterious effect is proportional to the degree of hyperther­
intentional induction of systemic hypothermia, for example, mia. The impact of hyperthermia (and also hypothermia)
after cardiac arrest. External or internal cooling reduces core on intracerebral hemorrage (ICH) is less clear.32 In an ICH
and blood temperature, leading to a sharp gradient between model in rats, mild to moderate temperature elevations do
the hot brain and the cooled blood. Specific studies docu­ not worsen outcome,33 although induced hypothermia can
menting the effect of systemic hypothermia on BT, such as improve outcome.34 Hyperthermia may damage uninjured
after cardiac arrest, are limited only to a few cases.17 Variations brain cells; cell membranes and mitochondria are selectively
in metabolic rate or in CBF in the single patient can change vulnerable.3 Damage is not limited to neurons but also
or reverse the gradient between BT and core temperature. This involves glia and endothelial cells. Consequences of hyper­
may in part explain variable findings on the gradients between thermia include increased glutamate release, disruption of
core and brain temperature reported in the literature.1,7,8 the blood-brain barrier, increased proinflammatory cytokines
and exacerbation of inflammatory cascades, up-regulation
of various enzymes, and expression of heat shock proteins,
Clinical Relevance of among others.3,25
Brain Temperature
Brain Temperature and Fever Is Associated with Worse Outcomes
Intracranial Pressure After Severe Brain Injury
Because the cellular machinery for energy production is sensi­ Fever is common after severe brain injury. However, the
tive to temperature, changes in BT may have profound conse­ precise mechanism by which neurologic injury results in
quences. In particular BT, brain metabolic rate, oxygen, and fever is still to be fully elucidated.35 This may include direct
glucose consumption are linked. Furthermore temperature injury to the thermoregulatory centers in the hypothalamus.
elevations have been reported to increase inflammatory pro­ In addition, fever is common when there is intracranial hem­
cesses, neuronal excitotoxicity, neurotransmitter release, free orrhage and, in particular, intraventricular hemorrhage.36
radical production, and intracellular glutamate concentra­ Other risk factors include the severity of injury, depressed
tions, and potentiate the sensitivity of neurons to excitotoxic consciousness, and use of antiepileptic drugs, especially phe­
injury in the brain.18-20 These may influence cerebral metabolic nytoin.37 Although a causal role has not been established in
rate of oxygen (CMRO2) that is physiologically coupled to humans, an association between hyperthermia and worse
CBF through modulation of vessel diameter. Therefore an outcome has been demonstrated in patients admitted to
increase CMRO2 results in vasodilation and increased CBF, neurocritical care for various pathologies.38-40 For example,
which in turn, leads to increased cerebral blood volume. This observational clinical studies demonstrate that elevated body
increase in blood volume is compensated by egress of cerebro­ temperature is associated in a dose-dependent way with
spinal fluid from the cranial vault and collapse of low-pressure longer intensive care unit (ICU) and hospital length of stay,
cerebral veins. Once these compensatory mechanisms are higher mortality, and worse hospital disposition.38,39 An asso­
exhausted, intracranial pressure (ICP) increases. Clinically, an ciation between fever burden and worse outcome has been
Section VI—Intracranial Monitoring 369

found in subarachnoid hemorrhage,41 and in acute ischemic 34° C) after cardiac arrest and observed brain temperatures as
stroke there is a strong relationship between admission tem­ low as 32° C. There was a decrease in the lactate/pyruvate ratio
perature and mortality.42 Higher temperature also is associ­ that paralleled induction of hypothermia.17 Yoo et al. com­
ated with an increased risk of hemorrhagic transformation pared brain and axillary temperature in 54 patients submitted
of an ischemic stroke.43 In patients with ICH who survive the to normothermia and 9 to hypothermia: brain temperature
first 72 hours after hospital admission, duration of fever is was lowered below normal but remained consistently higher
associated with poor outcome, and fever seems to be an than axillary temperature by approximately 1° C.59
independent prognostic factor in these patients.44 Fever also
is common after TBI, especially in the more severe cases.45,46
In a meta-analysis of 39 studies that included more than Fever Control and Induced Normothermia
14,000 patients Greer et al. found a consistent association Fever in the neurocritical care unit (NCCU) is very common
between higher body temperature and worse outcomes and its adverse effect on brain injury is well described. Hence
across multiple outcome measures.40 fever control rather than induced hypothermia may be a rea­
Some recreational drugs cause metabolic neural activation sonable treatment option in the NCCU. Consistent with this
and often increase oxygen consumption and heat production. premise it appears that hypothermia does not provide any
This is typical of cocaine, heroin, amphetamine-like sub­ added benefit to cerebral protection compared with normo­
stances, and ketamine. Brain temperature has been studied in thermia among patients undergoing brain surgery.60 Clinical
rats during cocaine administration, which confirms a dose- studies show that prophylactic normothermia using intravas­
dependent temperature increase.3 Heat stroke causes hyper­ cular cooling can reduce significant increases in BT in patients
thermia without an elevation in the hypothalamic set-point. with severe TBI61 and in a 2010 case control study, Badjatia
It is characterized by a core body temperature greater than et al.62 observed that fever control may be associated with
40° C due to exposure and is accompanied by hot and dry skin, improved outcome after subarachnoid hemorrhage (SAH).
nervous system failure (including delirium, convulsions, and Several recent advances in temperature management mean
coma), and a systemic inflammatory response. It may culmi­ that fever control and maintaining normothermia now are
nate in multiorgan system failure and death.47 feasible on a consistent basis in the NCCU. However, there are
still many unanswered questions about its use, such as in
which patients, indications, and when to start fever control.
Therapeutic Hypothermia
The neuroprotective effects of hypothermia have been dem­
onstrated in the experimental setting in models of ischemia Brain Temperature Monitoring
and trauma. In the clinical setting therapeutic hypothermia
is associated with decreased mortality and improved neuro­ Systemic and Core Temperature
logic outcome after cardiac arrest.48-51 There also may be a Systemic or core temperature is routinely measured in every
benefit to prehospital hypothermia for cardiac arrest.52 Con­ patient in the NCCU. From this, brain temperature is inferred.
sequently the European Resuscitation Council and American However, several lines of evidence show that brain tempera­
Heart Association guidelines recommend the use of hypo­ ture differs from core temperature and cannot be estimated
thermia in patients after cardiac arrest. Hypothermia also is reliably by monitors placed outside the brain, including the
thought to be associated with better neurologic outcome tympanic membrane.1
after neonatal birth asphyxia. Prophylactic hypothermia as It is rare that temperature is monitored using a thermom­
a method for neuroprotection after TBI in humans has eter. Instead, thermocouple or thermistor devices, infrared
received considerable interest. Multiple single-center studies sensors, or liquid crystal devices are used. Thermocouples are
have suggested benefit; however, two large multicenter trials based on the phenomenon that a conductor subject to a tem­
failed to show efficacy.53,53a A meta-analysis suggested that perature gradient will generate a voltage that can be measured.
prophylactic hypothermia was associated with a nonsignifi­ A thermistor is a semiconductor that varies its resistance
cant reduction in mortality and a significant improvement according to temperature. Infrared sensors (e.g., tympanic
in functional outcome.54,55 Virtually every single center study membrane thermometers) detect heat energy given off by
has suggested that hypothermia can help control intracranial radiation; that is, they do not need to be in direct contact with
hypertension after TBI. There is a consensus, however, that the object measured. However, this may affect accuracy
hypothermia is not necessary if ICP is controlled with other because the device picks up temperature from the external ear
therapies.56 Two multicenter randomized trials to examine rather than the tympanic membrane. Newer tympanic mem­
hypothermia in TBI are in progress in Europe (Eurotherm; brane thermometer models are more accurate.63 Thermo­
www.eurotherm3235trial.eu/home/index.phtml) and in Aus­ tropic liquid crystals usually are incorporated into disposable
tralia and New Zealand (POLAR; www.anzicrc.monash.org/ plastic strips and have a range between 34° C and 40° C. They
polar-rct.html). In addition, it does not appear that routine are not as accurate as other monitors.
pharmacologic or physical cooling provides a benefit in There are several sites from which temperature can be mon­
stroke patients.57 One promising new area for induced hypo­ itored. Broadly speaking the sites can be divided into: (1) core
thermia (core temperature of 32° C to 34° C) is for patients or central sites that represent the temperature of highly per­
with acute liver failure and encephalopathy.58 fused organs, for example, esophageal, pulmonary artery,
Most studies of therapeutic hypothermia have measured nasopharynx, or tympanic membrane, the last because of its
core body temperature, and data on BT during hypothermia proximity to the internal carotid artery; (2) intermediate
are scarce. Nordmark et al. recorded brain temperature from sites (e.g., oral [sublingual], rectal, or bladder); or (3) periph­
four patients undergoing therapeutic hypothermia (32° C to eral (e.g., forehead skin or axilla). Core or central sites are
370 Section VI—Intracranial Monitoring

preferable because they are less influenced by vasomotor or a temperature correction tool in its monitor, whereas the new
thermoregulatory mechanisms. Intermediate sites tend to lag Licox probe incorporates BT in its measurements.64 Oxygen
behind central monitors in a response to temperature changes tension may vary with changes of BT for several reasons, such
and may be influenced by urine flow (bladder) or bacteria in as CBF changes, shift of the oxyhemoglobin curve, and pos­
rectum (rectal), whereas peripheral sensors are affected by sible changes in metabolic rate, among others. Interpretation
many environmental influences. of brain oxygen tension during hyperthermia or hypothermia
therefore requires consideration of these factors.8 Similar
changes in oxygen associated with temperature are observed
Direct Brain Temperature in normal individuals, for example, during exercise.71
Monitoring Methods
Brain temperature can be measured directly using probes
placed into cerebral tissue or the lateral ventricles. The first Brain Temperature and Local Cerebral
thermal recording from the brain of an awake animal dates Blood Flow Measurement
back to 1860, when the relationship between temperature and BT can be used to determine local CBF (see Chapter 31). This
brain activation was noted.3 Intraventricular monitors provide CBF monitor (Hemedex) is based on the principle that
a global (average) measure of brain temperature, whereas thermal conductivity of cerebral cortical tissues varies propor­
intraparenchymal temperature monitors (which are often tionally with CBF. Therefore measurement of thermal diffu­
coupled with brain tissue oxygen sensors) measure regional sion at the cortical surface may be used to determine CBF. This
temperature. Various commercially available probes exist for monitoring system consists of two small metal plates (therm­
clinical use (http://raumedic.com; www.integra-ls.com). An istors) that are inserted into brain tissue. One of the plates is
earlier version of the Licox (Integra Lifesciences, New Bruns­ heated, thereby establishing a temperature gradient between
wick, NJ) system included separated sensors for brain oxygen the two thermistors; this allows CBF to be calculated from the
and brain temperature. Newer Licox probes incorporate both temperature difference between the plates.72 However, the
sensors in a single probe. In vitro analysis of the new Licox system is not able to measure CBF when temperature is greater
probe for simultaneous reading of brain oxygen tension and than 39° C.
BT reveals a tendency to underestimate BT, measuring a mean
of 0.67 plus or minus 0.22° C below the reference experimen­
tal temperature.64 There is ongoing work to be able to incor­ Selective Brain Cooling
porate BT into a single probe that can monitor multiple How to cool is beyond the scope of this chapter, but a brief
parameters.65 mention is made of selective brain cooling because systemic
Brain temperature also has been studied noninvasively hypothermia, the method used in major clinical trials, is
using magnetic resonance spectroscopy (MRS)66 and diffusion limited by the time to target temperature, the depth of hypo­
magnetic resonance imaging (MRI).67 Focal readings, in thermia, and complications. Selective brain cooling may solve
volumes as small as 4 cm3, have been performed with limited these issues. A variety of devices, for example, intraventricular
accuracy (approximately 1° C).11 In addition, BT is higher cooling catheters,73 neck pads,74 or cool packs applied to the
than that of the extracranial structures with wide variations dura during a craniotomy75 among many others, have been
observed depending on the regions studied. In contrast, Childs used to selectively cool the brain and generally they demon­
et al. found that the average BT was 0.5° C lower than tem­ strate localized cooling of the brain while maintaining relative
perature measured by MRS at three extracerebral sites, includ­ systemic normothermia. The most promise may lie in the use
ing oral cavity, tympanum, and temporal artery.14 In MR-based of the paranasal sinuses76 or be derived from an understanding
studies the mean brain-body (rectal) temperature difference of how seals selectively cool their brains to reduce oxygen
is about 1.3 plus or minus 0.4° C.68 consumption in the brain during a dive.77 In addition, implant­
able cooling devices in the field of neuromodulation may
become clinically feasible in the near future.78
Brain Temperature
Monitoring Complications
Complications associated with the Licox monitor including
Conclusion
the BT probe are very rare and generally are associated with Brain temperature is an important and dynamic variable in
insertion of the bolt to hold the probes rather than the monitor patients with acute brain damage. However, BT cannot be
itself.69 Similar to other invasive intracranial monitors, the reliably estimated from extracerebral measurements, and it
main risks include cerebral hemorrhage and infection. varies according to brain region. The main determinants of
BT include cerebral metabolic rate and CBF. It is well under­
stood that fever is deleterious to the brain in a variety of
Brain Temperature and disease states. Induced hypothermia confers neuroprotection
Brain Oxygen Tension after cardiac arrest, but its role is less certain for other disor­
Temperature has an effect on brain tissue oxygen tension. ders such as TBI, SAH, or stroke. Instead, fever control and
Given a constant tissue oxygen content, pressure is propor­ normothermia may be reasonable alternatives.
tional to temperature, according to the fundamental gas laws. Brain temperature may be measured invasively, usually in
Therefore oxygen tension increases during hyperthermia and combination with other intracranial monitoring devices such
decreases during hypothermia because of this biophysical as ICP and brain tissue oxygen probes. Brain temperature
property. Hypoxia itself may induce hypothermia.70 For this monitoring may be of particular value when using therapeutic
reason the Licox probe used to measure brain oxygen includes temperature modulation in neurocritical care, such as during
Section VI—Intracranial Monitoring 371

controlled normothermia or induced hypothermia, or to 25. Takagi K, Ginsberg MD, Globus MY, et al. Effect of hyperthermia on
glutamate release in ischemic penumbra after middle cerebral artery
guide the interpretation of other important intracranial
occlusion in rats. Am J Physiol 1994;267(5 Pt 2): H1770–6.
parameters, such as brain tissue oxygenation. 26. Busto R, Dietrich WD, Globus MY, et al. The importance of brain
temperature in cerebral ischemic injury. Stroke 1989;20(8):1113–4.
27. Corbett D, Thornhill J. Temperature modulation (hypothermic and
References hyperthermic conditions) and its influence on histological and behavioral
outcomes following cerebral ischemia. Brain Pathol 2000;10(1):145–52.
1. Childs C. Human brain temperature: regulation, measurement and 28. Reglodi D, Somogyvari-Vigh A, Maderdrut JL, et al. Postischemic
relationship with cerebral trauma. Part 1. Br J Neurosurg 2008;4:486–96. spontaneous hyperthermia and its effects in middle cerebral artery occlusion
2. Badjatia N. Hyperthermia and fever control in brain injury. Crit Care Med in the rat. Exper Neurol 2000;163(2):399–407.
2009;37(7 Suppl):S250–7. 29. Sakurai A, Atkins CM, Alonso OF, et al. Mild hyperthermia worsens the
3. Kiyatkin EA. Brain hyperthermia as physiological and pathological neuropathological damage associated with mild traumatic brain injury in
phenomena. Brain Res Rev 2005;50(1):27–56. rats. J Neurotrauma 2011;25. Epub ahead of print.
4. Rossi S, Roncati Zanier E, et al. Brain temperature, body core temperature, 30. Kim Y, Busto R, Dietrich WD, et al. Delayed postischemic hyperthermia in
and intracranial pressure in acute cerebral damage. J Neurol Neurosurg awake rats worsens the histopathological outcome of transient focal cerebral
Psychiatry 2001;71(4):448–54. ischemia. Stroke 1996;27(12):2274–81.
5. Mellergård P. Intracerebral temperature in neurosurgical patients: 31. Noor R, Wang CX, Shuaib A. Effects of hyperthermia on infarct volume
intracerebral temperature gradients and relationships to consciousness level. in focal embolic model of cerebral ischemia in rats. Neurosci Lett 2003;
Surg Neurol 1995;43(1):91–5. 349(2):130–2.
6. Soukup J, Zauner A, Doppenberg EM, et al. The importance of brain 32. MacLellan CL, Clark DL, Silasi G, et al. Use of prolonged hypothermia to
temperature in patients after severe head injury: relationship to intracranial treat ischemic and hemorrhagic stroke. J Neurotrauma 2009;26(3):313–23.
pressure, cerebral perfusion pressure, cerebral blood flow, and outcome. 33. MacLellan CL, Colbourne F. Mild to moderate hyperthermia does not
J Neurotrauma 2002;19(5):559–71. worsen outcome after severe intracerebral hemorrhage in rats. J Cereb Blood
7. Rumana CS, Gopinath SP, Uzura M, et al. Brain temperature exceeds Flow Metab 2005;25:1020–9.
systemic temperature in head-injured patients. Crit Care Med 1988;23: 34. MacLellan CL, Davies LM, Fingas MS, et al. The influence of hypothermia
562–7. on outcome after intracerebral hemorrhage in rats. Stroke 2006;37(5):1266–
8. Spiotta AM, Stiefel MF, Heuer GG, et al. Brain hyperthermia after traumatic 70. Epub 2006, Mar 30.
brain injury does not reduce brain oxygen. Neurosurgery 2008;62(4):864–72. 35. Thompson HJ. Elevated body temperature in the neuroscience intensive care
9. Smith CM, Adelson PD, Chang YF, et al. Brain-systemic temperature unit. Crit Care Med 2005;33:1672.
gradient is temperature-dependent in children with severe traumatic brain 36. Commichau C, Scarmeas N, Mayer SA. Risk factors for fever in the
injury. Pediatr Crit Care Med 2011;12(4):449–54. neurologic intensive care unit. Neurology 2003;60:837–41.
10. Kuo JR, Lo CJ, Wang CC, et al. Measuring brain temperature while 37. Rabinstein AA, Sandhu K. Non-infectious fever in the neurological intensive
maintaining brain normothermia in patients with severe traumatic brain care unit: incidence, causes and predictors. J Neurol Neurosurg Psychiatry
injury. J Clin Neurosci 2011;18(8):1059–63. Epub 2011, Jul 1. 2007;78:1278–80.
11. Corbett R, Laptook A, Weatherall P. Noninvasive measurements of human 38. Kilpatrick MM, Lowry DW, Firlik AD, et al. Hyperthermia in the
brain temperature using volume-localized proton magnetic resonance neurosurgical intensive care unit. Neurosurgery 2000;47(4):850–6.
spectroscopy. J Cereb Blood Flow Metab 1997;17(4):363–9. 39. Diringer MN, Reaven NL, Funk SE, et al. Elevated body temperature
12. Nakagawa K, Hills NK, Kamel H, et al. The effect of decompressive independently contributes to increased length of stay in neurologic intensive
hemicraniectomy on brain temperature after severe brain injury. Neurocrit care unit patients. Crit Care Med 2004;32(7):1489–95.
Care 2011;15(1):101–6. 40. Greer DM, Funk SE, Reaven NL, et al. Impact of fever on outcome in
13. Suehiro E, Fujisawa H, Koizumi H, et al. Significance of differences between patients with stroke and neurologic injury: a comprehensive meta-analysis.
brain temperature and core temperature (delta T) during mild hypothermia Stroke 2008;39(11):3029–35.
in patients with diffuse axonal injury. Neurol Med Chir (Tokyo) 2011;51(8): 41. Naidech AM, Bendok BR, Bernstein RA, et al. Fever burden and functional
551–5. recovery after subarachnoid hemorrhage. Neurosurgery 2008;63(2):212–8.
14. Childs C, Hiltunen Y, Vidyasagar R, et al. Determination of regional brain 42. Kammersgaard LP, Jorgensen HS, Rungby JA, et al. Admission body
temperature using proton magnetic resonance spectroscopy to assess temperature predicts long-term mortality after acute stroke: the Copenhagen
brain-body temperature differences in healthy human subjects. Magn Reson Stroke Study. Stroke 2002;33(7):1759–62.
Med 2007;57(1):59–66. 43. Leira R, Sobrino T, Blanco M, et al. A higher body temperature is associated
15. Fountas KN, Kapsalaki EZ, Feltes CH, et al. Intracranial temperature: is it with haemorrhagic transformation in patients with acute stroke untreated
different throughout the brain? Neurocrit Care 2004;1(2):195–9. with recombinant tissue-type plasminogen activator (rtPA). Clin Sci (Lond)
16. Hirashima Y, Takaba M, Endo S, et al. Intracerebral temperature in patients 2012;122(3):13–9.
with hydrocephalus of varying aetiology. J Neurol Neurosurg Psychiatry 44. Schwarz S, Hafner K, Aschoff A, et al. Incidence and prognostic significance
1998;64(6):792–4. of fever following intracerebral hemorrhage. Neurology 2001;54(2):354–61.
17. Nordmark J, Rubertsson S, Mörtberg E, et al. Intracerebral monitoring in 45. Soukup J, Zauner A, Doppenberg EM, et al. The importance of brain
comatose patients treated with hypothermia after a cardiac arrest. Acta temperature in patients after severe head injury: relationship to intracranial
Anaesthesiol Scand 2009;53(3):289–98. pressure, cerebral perfusion pressure, cerebral blood flow, and outcome.
18. Suehiro E, Fujisawa H, Ito H, et al. Brain temperature modifies glutamate J Neurotrauma 2002;19(5):559–71.
neurotoxicity in vivo. J Neurotrauma 1999;16:285–97. 46. Stocchetti N, Rossi S, Roncati Zanier E, et al. Pyrexia in head-injured
19. Dietrich WD, Chatzipanteli K, Vitarbo E, et al. The role of inflammatory patients admitted to intensive care. Intensive Care Med 2002;28(11):1555–62.
processes in the pathophysiology and treatment of brain and spinal cord 47. Bouchama A, Knochel JP. Heat stroke. N Engl J Med 2002;346:1978–88.
trauma. Acta Neurochir Suppl 2004; 89:69–74. 48. Hypothermia after Cardiac Arrest Study Group. Mild therapeutic
20. Takagi K, Ginsberg MD, Globus MY, et al. Effect of hyperthermia on hypothermia to improve the neurologic outcome after cardiac arrest. N Engl
glutamate release in ischemic penumbra after middle cerebral artery J Med 2002;346(8): 49–56.
occlusion in rats. Am J Physiol 1994;267:H1770–6. 49. Bernard SA, Gray TW, Buist MD, et al. Treatment of comatose survivors of
21. Stocchetti N, Protti A, Lattuada M, et al. Impact of pyrexia on out-of-hospital cardiac arrest with induced hypothermia. N Engl J Med
neurochemistry and cerebral oxygenation after acute brain injury. J Neurol 2002;346(8):557–63.
Neurosurg Psychiatry 2005;76(8):1135–9. 50. Holzer M, Bernard S, Hachimi-Idrissi S, et al. Hypothermia for
22. Oddo M, Frangos S, Milby A, et al. Induced normothermia attenuates neuroprotection after cardiac arrest: systematic review and individual patient
cerebral metabolic distress in patients with aneurysmal subarachnoid data meta-analysis. Crit Care Med 2005;33(2):414–8.
hemorrhage and refractory fever. Stroke 2009;40(5):1913–6.
23. Huschak G, Hoell T, Wiegel M, et al. Does brain temperature correlate with A complete list of references for this chapter can be found online at
intracranial pressure? J Neurosurg Anesthesiol 2008;20(2):105–9. www.expertconsult.com.
24. Dietrich WD, Alonso O, Halley M, et al. Delayed posttraumatic brain
hyperthermia worsens outcome after fluid percussion brain injury: a light
and electron microscopic study in rats. Neurosurgery 1996;38(3):533–41.
Section VI—Intracranial Monitoring 371.e1

References 27. Corbett D, Thornhill J. Temperature modulation (hypothermic and


hyperthermic conditions) and its influence on histological and behavioral
1. Childs C. Human brain temperature: regulation, measurement and outcomes following cerebral ischemia. Brain Pathol 2000;10(1):145–52.
relationship with cerebral trauma. Part 1. Br J Neurosurg 2008;4:486–96. 28. Reglodi D, Somogyvari-Vigh A, Maderdrut JL, et al. Postischemic
2. Badjatia N. Hyperthermia and fever control in brain injury. Crit Care Med spontaneous hyperthermia and its effects in middle cerebral artery occlusion
2009;37(7 Suppl):S250–7. in the rat. Exper Neurol 2000;163(2):399–407.
3. Kiyatkin EA. Brain hyperthermia as physiological and pathological 29. Sakurai A, Atkins CM, Alonso OF, et al. Mild hyperthermia worsens the
phenomena. Brain Res Rev 2005;50(1):27–56. neuropathological damage associated with mild traumatic brain injury in
4. Rossi S, Roncati Zanier E, et al. Brain temperature, body core temperature, rats. J Neurotrauma 2011;25. Epub ahead of print.
and intracranial pressure in acute cerebral damage. J Neurol Neurosurg 30. Kim Y, Busto R, Dietrich WD, et al. Delayed postischemic hyperthermia in
Psychiatry 2001;71(4):448–54. awake rats worsens the histopathological outcome of transient focal cerebral
5. Mellergård P. Intracerebral temperature in neurosurgical patients: ischemia. Stroke 1996;27(12):2274–81.
intracerebral temperature gradients and relationships to consciousness level. 31. Noor R, Wang CX, Shuaib A. Effects of hyperthermia on infarct volume in
Surg Neurol 1995;43(1):91–5. focal embolic model of cerebral ischemia in rats. Neurosci Lett 2003;349(2):
6. Soukup J, Zauner A, Doppenberg EM, et al. The importance of brain 130–2.
temperature in patients after severe head injury: relationship to intracranial 32. MacLellan CL, Clark DL, Silasi G, et al. Use of prolonged hypothermia to
pressure, cerebral perfusion pressure, cerebral blood flow, and outcome. treat ischemic and hemorrhagic stroke. J Neurotrauma 2009;26(3):313–23.
J Neurotrauma 2002;19(5):559–71. 33. MacLellan CL, Colbourne F. Mild to moderate hyperthermia does not
7. Rumana CS, Gopinath SP, Uzura M, et al. Brain temperature exceeds worsen outcome after severe intracerebral hemorrhage in rats. J Cereb Blood
systemic temperature in head-injured patients. Crit Care Med 1988;23: Flow Metab 2005;25:1020–9.
562–7. 34. MacLellan CL, Davies LM, Fingas MS, et al. The influence of hypothermia
8. Spiotta AM, Stiefel MF, Heuer GG, et al. Brain hyperthermia after traumatic on outcome after intracerebral hemorrhage in rats. Stroke 2006;37(5):1266–
brain injury does not reduce brain oxygen. Neurosurgery 2008;62(4): 70. Epub 2006, Mar 30.
864–72. 35. Thompson HJ. Elevated body temperature in the neuroscience intensive care
9. Smith CM, Adelson PD, Chang YF, et al. Brain-systemic temperature unit. Crit Care Med 2005;33:1672.
gradient is temperature-dependent in children with severe traumatic brain 36. Commichau C, Scarmeas N, Mayer SA. Risk factors for fever in the
injury. Pediatr Crit Care Med 2011;12(4):449–54. neurologic intensive care unit. Neurology 2003;60:837–41.
10. Kuo JR, Lo CJ, Wang CC, et al. Measuring brain temperature while 37. Rabinstein AA, Sandhu K. Non-infectious fever in the neurological intensive
maintaining brain normothermia in patients with severe traumatic brain care unit: incidence, causes and predictors. J Neurol Neurosurg Psychiatry
injury. J Clin Neurosci 2011;18(8):1059–63. Epub 2011, Jul 1. 2007;78:1278–80.
11. Corbett R, Laptook A, Weatherall P. Noninvasive measurements of human 38. Kilpatrick MM, Lowry DW, Firlik AD, et al. Hyperthermia in the
brain temperature using volume-localized proton magnetic resonance neurosurgical intensive care unit. Neurosurgery 2000;47(4):850–6.
spectroscopy. J Cereb Blood Flow Metab 1997;17(4):363–9. 39. Diringer MN, Reaven NL, Funk SE, et al. Elevated body temperature
12. Nakagawa K, Hills NK, Kamel H, et al. The effect of decompressive independently contributes to increased length of stay in neurologic intensive
hemicraniectomy on brain temperature after severe brain injury. Neurocrit care unit patients. Crit Care Med 2004;32(7):1489–95.
Care 2011;15(1):101–6. 40. Greer DM, Funk SE, Reaven NL, et al. Impact of fever on outcome in
13. Suehiro E, Fujisawa H, Koizumi H, et al. Significance of differences between patients with stroke and neurologic injury: a comprehensive meta-analysis.
brain temperature and core temperature (delta T) during mild hypothermia Stroke 2008;39(11):3029–35.
in patients with diffuse axonal injury. Neurol Med Chir (Tokyo) 2011;51(8): 41. Naidech AM, Bendok BR, Bernstein RA, et al. Fever burden and functional
551–5. recovery after subarachnoid hemorrhage. Neurosurgery 2008;63(2):212–8.
14. Childs C, Hiltunen Y, Vidyasagar R, et al. Determination of regional brain 42. Kammersgaard LP, Jorgensen HS, Rungby JA, et al. Admission body
temperature using proton magnetic resonance spectroscopy to assess temperature predicts long-term mortality after acute stroke: the Copenhagen
brain-body temperature differences in healthy human subjects. Magn Reson Stroke Study. Stroke 2002;33(7):1759–62.
Med 2007;57(1):59–66. 43. Leira R, Sobrino T, Blanco M, et al. A higher body temperature is associated
15. Fountas KN, Kapsalaki EZ, Feltes CH, et al. Intracranial temperature: is it with haemorrhagic transformation in patients with acute stroke untreated
different throughout the brain? Neurocrit Care 2004;1(2):195–9. with recombinant tissue-type plasminogen activator (rtPA). Clin Sci (Lond)
16. Hirashima Y, Takaba M, Endo S, et al. Intracerebral temperature in patients 2012;122(3):13–9.
with hydrocephalus of varying aetiology. J Neurol Neurosurg Psychiatry 44. Schwarz S, Hafner K, Aschoff A, et al. Incidence and prognostic significance
1998;64(6):792–4. of fever following intracerebral hemorrhage. Neurology 2001;54(2):354–61.
17. Nordmark J, Rubertsson S, Mörtberg E, et al. Intracerebral monitoring in 45. Soukup J, Zauner A, Doppenberg EM, et al. The importance of brain
comatose patients treated with hypothermia after a cardiac arrest. Acta temperature in patients after severe head injury: relationship to intracranial
Anaesthesiol Scand 2009;53(3):289–98. pressure, cerebral perfusion pressure, cerebral blood flow, and outcome.
18. Suehiro E, Fujisawa H, Ito H, et al. Brain temperature modifies glutamate J Neurotrauma 2002;19(5):559–71.
neurotoxicity in vivo. J Neurotrauma 1999;16:285–97. 46. Stocchetti N, Rossi S, Roncati Zanier E, et al. Pyrexia in head-injured
19. Dietrich WD, Chatzipanteli K, Vitarbo E, et al. The role of inflammatory patients admitted to intensive care. Intensive Care Med 2002;28(11):1555–62.
processes in the pathophysiology and treatment of brain and spinal cord 47. Bouchama A, Knochel JP. Heat stroke. N Engl J Med 2002;346:1978–88.
trauma. Acta Neurochir Suppl 2004; 89:69–74. 48. Hypothermia after Cardiac Arrest Study Group. Mild therapeutic
20. Takagi K, Ginsberg MD, Globus MY, et al. Effect of hyperthermia on hypothermia to improve the neurologic outcome after cardiac arrest. N Engl
glutamate release in ischemic penumbra after middle cerebral artery J Med 2002;346(8): 49–56.
occlusion in rats. Am J Physiol 1994;267:H1770–6. 49. Bernard SA, Gray TW, Buist MD, et al. Treatment of comatose survivors of
21. Stocchetti N, Protti A, Lattuada M, et al. Impact of pyrexia on out-of-hospital cardiac arrest with induced hypothermia. N Engl J Med
neurochemistry and cerebral oxygenation after acute brain injury. J Neurol 2002;346(8):557–63.
Neurosurg Psychiatry 2005;76(8):1135–9. 50. Holzer M, Bernard S, Hachimi-Idrissi S, et al. Hypothermia for
22. Oddo M, Frangos S, Milby A, et al. Induced normothermia attenuates neuroprotection after cardiac arrest: systematic review and individual patient
cerebral metabolic distress in patients with aneurysmal subarachnoid data meta-analysis. Crit Care Med 2005;33(2):414–8.
hemorrhage and refractory fever. Stroke 2009;40(5):1913–6. 51. Sagalyn E, Band RA, Gaieski DF, et al. Therapeutic hypothermia after cardiac
23. Huschak G, Hoell T, Wiegel M, et al. Does brain temperature correlate with arrest in clinical practice: review and compilation of recent experiences. Crit
intracranial pressure? J Neurosurg Anesthesiol 2008;20(2):105–9. Care Med 2009;37(7 Suppl):S223–6.
24. Dietrich WD, Alonso O, Halley M, et al. Delayed posttraumatic brain 52. Cullen D, Augenstine D, Kaper L, et al. Therapeutic hypothermia initiated in
hyperthermia worsens outcome after fluid percussion brain injury: a light the pre-hospital setting: a meta-analysis. Adv Emerg Nurs J 2011;33(4):
and electron microscopic study in rats. Neurosurgery 1996;38(3):533–41. 314–21.
25. Takagi K, Ginsberg MD, Globus MY, et al. Effect of hyperthermia on 53. Clifton GL, Miller ER, Choi SC, et al. Lack of effect of induction of
glutamate release in ischemic penumbra after middle cerebral artery hypothermia after acute brain injury. N Engl J Med 2001;344(8):556–63.
occlusion in rats. Am J Physiol 1994;267(5 Pt 2): H1770–6. 53a.  Clifton GL, Valadka A, Zygun D, et al. Very early hypothermia induction in
26. Busto R, Dietrich WD, Globus MY, et al. The importance of brain patients with severe brain injury (the National Acute Brain Injury Study:
temperature in cerebral ischemic injury. Stroke 1989;20(8):1113–4. Hypothermia II): a randomised trial. Lancet Neurol 2011;10(2):131–9.
371.e2 Section VI—Intracranial Monitoring

54. Mcilvoy LH. The effect of hypothermia and hyperthermia on acute brain 67. Kozak LR, Bango M, Szabo M, et al. Using diffusion MRI for measuring the
injury. AACN Clin Issues 2005;16(4): 488–500. temperature of cerebrospinal fluid within the lateral ventricles. Acta Paediatr
55. Polderman KH. Induced hypothermia and fever control for prevention and 2010;99(2):237–43. Epub 2009, Oct 20.
treatment of neurological injuries. Lancet 2008;371(9628):1955–69. 68. Covaciu L, Rubertsson S, Ortiz-Nieto F, et al. Human brain MR spectroscopy
56. Childs C, Wieloch T, Lecky F, et al. Report of a consensus meeting on human thermometry using metabolite aqueous-solution calibrations. J Magn Reson
brain temperature after severe traumatic brain injury: its measurement and Imaging 2010;31(4):807–14.
management during pyrexia. Front Neurol 2010;1:146. 69. Maloney-Wilensky E, Gracias V, Itkin A, et al. Brain tissue oxygen and
57. Den Hertog HM, van der Worp HB, Tseng MC, et al. Cooling therapy for outcome after severe traumatic brain injury: a systematic review. Crit Care
acute stroke. Cochrane Database Syst Rev 2009;(1):CD001247. Med 2009;37(6):2057–63.
58. Rabinstein AA. Treatment of brain edema in acute liver failure. Curr Treat 70. Osaka T. Hypoxia-induced hypothermia mediated by noradrenaline and
Options Neurol 2010;12(2):129–41. nitric oxide in the rostromedial preoptic area. Neuroscience 2011;179:170–8.
59. Yoo DS, Kim DS, Park CK, et al. Significance of temperature difference Epub 2011, Jan 28.
between cerebral cortex and axilla in patients under hypothermia 71. Rasmussen P, Nybo L, Volianitis S, et al. Cerebral oxygenation is reduced
management. Acta Neurochir Suppl 2002;81:85–7. during hyperthermic exercise in humans. Acta Physiol (Oxf)
60. Milani WR, Antibas PL, Prado GF. Cooling for cerebral protection during 2010;199(1):63–70. Epub 2010, Jan 25.
brain surgery. Cochrane Database Syst Rev 2011;(10):CD006638. 72. Bhatia A, Gupta AK. Neuromonitoring in the intensive care unit. I.
61. Fischer M, Lackner P, Beer R, et al. Keep the brain cool—endovascular Intracranial pressure and cerebral blood flow monitoring. Intensive Care
cooling in patients with severe traumatic brain injury: a case series study. Med 2007;33(7):1263–71.
Neurosurgery 2011;68(4):867–73; discussion 873. 73. Moomiaie RM, Gould G, Solomon D, et al. Novel intracranial brain cooling
62. Badjatia N, Fernandez L, Schmidt JM, et al. Impact of induced catheter to mitigate brain injuries. J Neurointerv Surg 2011;Jun 14. Epub
normothermia on outcome after subarachnoid hemorrhage: a case-control ahead of print.
study. Neurosurgery 2010;66(4):696–700; discussion 700–1. 74. Mink S, Schwarz U, Mudra R, et al. Treatment of resistant fever: new method
63. Bock M, Hohlfeld U, von Engeln K, et al. The accuracy of a new infrared ear of local cerebral cooling. Neurocrit Care 2011;15(1):107–12.
thermometer in patients undergoing cardiac surgery. Can J Anaesth 2005; 75. King C, Robinson T, Dixon CE, et al. Brain temperature profiles during
52(10):1083–7. epidural cooling with the ChillerPad in a monkey model of traumatic brain
64. Stewart C, Haitsma I, Zador Z, et al. The new Licox combined brain tissue injury. J Neurotrauma 2010;27(10):1895–903. Epub 2010, Sep 16.
oxygen and brain temperature monitor: assessment of in vitro accuracy and 76. Gallup AC, Hack GD. Human paranasal sinuses and selective brain cooling:
clinical experience in severe traumatic brain injury. Neurosurgery 2008; a ventilation system activated by yawning? Med Hypotheses 2011;77(6):970–
63(6):1159–65. 3. Epub 2011, Sep 8.
65. Li C, Wu PM, Wu Z, et al. Brain temperature measurement: a study of in 77. Blix AS, Walløe L, Messelt EB, et al. Selective brain cooling and its vascular
vitro accuracy and stability of smart catheter temperature sensors. Biomed basis in diving seals. J Exp Biol 2010;213(Pt 15):2610–6.
Microdevices 2011;Sep 28. Epub ahead of print. 78. Fujii M, Fujioka H, Oku T, et al. Application of focal cerebral cooling for the
66. Murakami T, Ogasawara K, Yoshioka Y, et al. Brain temperature measured by treatment of intractable epilepsy. Neurol Med Chir (Tokyo)
using proton MR spectroscopy predicts cerebral hyperperfusion after carotid 2010;50(9):839–44.
endarterectomy. Radiology 2010;256(3):924–31.
VII
Chapter
38  

Device Development
Gerald P. Roston and Brandon von Tobel

Introduction medical device development focus on the technical aspects of


In times past, tribal medicine men jealously guarded their the process.5-8 The more business-oriented focus of this
secret knowledge, typically passing it from father to son, chapter complements these other sources.
thereby protecting the family’s status and importance. Over
the centuries mankind has come to realize that broadly sharing
this critical knowledge benefits society. As a result, in today’s
Developing Medical Products
society more than 1 million people worldwide study medicine. For new or established companies, the decision to pursue the
Interestingly, the learning process is not radically different development of a new product is never taken lightly because
today than in years past: students apprentice under those with a significant commitment of the company’s resources is
greater experience and learn by reading and practicing the required. To bring a new drug to market, for example, may
taught skills. require the better part of a decade and cost more than $500
Although this process is quite adequate to teach skills, it falls million. Therefore one of the most important aspects of the
short with the ability to produce devices or medicines. Con- product development process is to determine if there is a need
sider the carnage that may ensue if people were provided a for the product. The need assessment is revisited frequently
recipe to make aspirin, or a pacemaker, or a monitor rather during the development process, whose steps are (1) idea gen-
than being able to buy one whose efficacy, or functionality, is eration, (2) conceptual design, (3) detail design, (4) clinical
well understood.1 The key word is “buy,” which implies the trials, and (5) manufacturing. This section outlines the activi-
existence of an entity willing to sell, possibly on a large scale. ties that occur during each step of this process.9
This is the focus of this chapter, namely, how to translate an
idea for a medical product from a notion to a salable product.
Because this textbook is about monitoring, the focus of this Idea Generation
chapter is on medical devices rather than pharmaceutical All products available for purchase start as an idea in some-
compounds. Although much of the material presented applies one’s head. The original idea may have sourced from an engi-
equally to both, the time and expense associated with drug neer who realized that some technology could be of value to
development are typically considerably greater than that for a medical practitioner or the idea may have sourced from a
medical devices. In addition, the business models may be dif- clinician who identified a need during the course of practice.
ferent. It is not uncommon for a pharmaceutical start-up to These two camps are referred to as “technology push” and
be acquired before market introduction but after certain mile- “market pull.” Technology push typically requires more time
stones are achieved, whereas medical device companies typi- and effort to fully develop, but often may lead to a broad range
cally are not acquired until the product has successfully of products, often more than imagined by the original inves-
launched. tor. Market pull ideas typically are accepted more quickly into
Much of the material in this chapter discusses the business the marketplace, but usually do not lead to a broad suite of
and financial aspects of bringing a product to market, because follow-on products.
most medical practitioners do not have a business back- Once a product idea has been formed, the concept must be
ground. By contrast, many medical practitioners have a scien- tested. For medical devices the first tests performed are risk
tific or engineering background and so only a general overview analyses—does the use of the product pose greater risk to the
of the product development life cycle and some key develop- user than the problem it is intended to address? Assuming that
ment activities are discussed. However, the medical device the product does not pose unreasonable risk, the next task is
development process has become more complex over the years to determine if the product meets customer needs. There are
in large part because of regulatory requirements, business several methods to perform this assessment.10
decisions (reimbursement), and technology advances. This The easiest method, but the one that provides the least valu-
development process has recently been detailed2 (Table 38.1) able information, is to perform secondary research, that is, to
and practical ways to address specific challenges have been analyze published documents to understand the market.
detailed in workshops of academia, government, and industry Although secondary research is important, it cannot truly
representatives.3 Several universities around the world also answer the question, “If I make it, will they buy it?” To learn
have developed dedicated educational programs in life science the answer to this question, the best approach is to ask poten-
technology innovation.4 In addition, most books that address tial customers, in particular, those with purchase authority. To
374 © Copyright 2013 Elsevier Inc. All rights reserved.
Section VII—Computers, Engineering, and the Future 375

Table 38.1  The Five Major Phases Record Keeping


and Decision Gates for Medical The Safe Medical Devices Act of 1990 added design valida-
Device Development tion requirements to the Good Manufacturing Practices
This process is applicable to a broad range of medical
(GMP) requirements.18 Fundamentally, the entity that designs
technologies and may be used to develop sophisticated a medical device must document the procedures used to
premarket approval and premarket notification (510[k]) ensure that the specified requirements are met. Procedures
devices and less sophisticated devices that may be exempt should be generic rather than detailed. That is, they should
from most regulatory requirements. include major activities and assignments for subsystems, as
• Phase I/gate 1: Initiation, opportunity, and risk analysis
• Phase II/gate 2: Formulation, concept, and feasibility opposed to detailing responsibility for component selection.
• Phase III/gate 3: Design, development, verification, and It also is understood that design procedures change, thus the
validation standard requires that an entity’s procedure be regularly
• Phase IV/gate 4: Final validation and product launch reviewed and updated.
preparation
• Phase V: Product launch and post-launch assessment

From Pietzsch JB, Shluzas LA, Pate-Cornell ME, et al. Stage-gate process for Device Class
the development of medical devices. J Med Dev 2009;3:021004–15.
All medical devices are categorized into one of three device
classes:
 Class I: These are non–life-sustaining devices whose failure
poses no risk to life. Examples of class I devices include
bandages, stethoscopes, and examination gloves. Most
maximize the value of the information obtained from poten-
class I devices are exempt from the premarket notification
tial customers, a structured approach, such as quality func-
or GMP regulation.
tional deployment (QFD)11 is recommended. The reason that  Class II: These devices are non–life sustaining; however,
this process is so powerful is that it is a well-defined process
the controls in place for class I devices are insufficient to
to translate customer needs to product features and keep them
ensure safety and effectiveness. Examples of class II devices
evident throughout the design process.
include catheters, powered wheelchairs, and many clinical
The final step in this phase of the design process is to thor-
chemistry test systems. Although some class II devices are
oughly document the product requirements.12 Though there
exempt from premarket notification, others may require
is some disagreement, requirements should state only what the
clinical studies as rigorous as an investigational device
product should do and not how it does it. This approach
exemption (IDE) for premarket approval (PMA).
provides the broadest palette for conceptual designs.  Class III: These devices are typically those that support or
sustain human life and whose failure is life threatening.
Conceptual Design Examples of class III devices include heart valves, implanted
electrical devices, and joint implants. The vast majority of
Conceptual design is the process of structuring a solution to
class III devices require PMA, a process that includes labo-
a design problem. During this stage the process is mostly
ratory testing, animal testing, failure mode analysis, manu-
qualitative, not quantitative. For instance, if one were design-
facturing standards, and safety and efficacy testing as
ing a robotic surgical assistant, a choice between electric and
determined in a human clinical trial. Needless to say, to
hydraulic actuators would be decided during this phase. This
obtain PMA can be a long, costly process.
decision may require some quantitative analysis. For example,
“Can a motor fit within the space allotted and still provide When setting out to develop a medical device, the design team
sufficient motive force without requiring the user to specify should determine the device class, as early as possible so that
part numbers, bolt patterns, and so on.”13 the planning process can incorporate the appropriate activities
For engineers, the conceptual design phase is usually the into the overall project plan. For new devices for which prec-
most enjoyable. Conceptual design requires great creativity, as edent is unclear, the design team should submit a Request for
a number of widely disparate approaches should be developed Determination to the FDA.
to ensure that the best ones are considered for further devel-
opment. Methods used to generate ideas include (1) literature
searches, (2) biomimicry, (3) reverse engineering of related Extent of Business Activities
products, (4) study of analogous systems, and (5) brainstorm- In the grossest sense, the activities needed to bring a device to
ing, among others. Once a number of concepts have been market are conception, design, production, distribution, and
developed, they are assessed against the requirements previ- support. A key decision for a new business entity is to deter-
ously developed to determine those that best meet the speci- mine which of these activities are to be performed in-house
fied needs. and which are outsourced. This decision has two major
At this juncture it is important to note several issues that impacts: first, the amount of investment needed, and second,
are relevant to design of medical devices rather than other the FDA regulations that must be addressed. For many com-
product types. In the United States, the Food and Drug panies, outsourcing manufacturing offers a good blend of
Administration (FDA) regulates medical devices. Although activities, allowing the company to minimize capital costs,
entire books are written on the subject of complying with FDA earn a reasonable rate of return, and reduce FDA regulatory
requirements,14-16 the following two (and potentially three) issues (though the manufacturing partner must comply with
issues greatly affect the development process.17 FDA requirements).
376 Section VII—Computers, Engineering, and the Future

The conceptual design phase ends with a design review that factors are known, details of the trail design, such as study
presents the requirements, the concepts developed, and the size, candidate pool, and controls, among others can be
concept(s) selected for further development. formulated. In addition, trials must be carefully designed
to minimize errors and biases. From a business perspective,
trial design is critical because the claims that the business
Detail Design wishes to make about its device must be supported by clini-
Most novel designs includes one, and sometimes more, ele- cal outcomes.
ments that have not been previously realized. For example,
lab-on-chip devices replace manual fluid introduction and
mixing with microfluidic components. Before the design for Manufacturing
the final product is completed, the development team typically Once the design is completed, a pilot run is performed to
performs a series of experiments to validate the functionality validate manufacturing processes. This is the first build of the
of the novel component.8 Staying with the example, do the device in its final form using the manufacturing documenta-
microfluidic devices deliver sufficient head and flow to provide tion. This part of the process also includes assembly workforce
sufficient agitation? training, validation of supplier plans, and checking that man-
Once all of the subsystems are known to work, it is common ufacturing objectives have been achieved.23
to couple all of them together in a bench-top prototype to A critical aspect of this phase is product packaging and
assess overall system functionality. This prototype may bear labeling. Packaging must be designed to ensure that the
little physical resemblance to the final device. However, the product reaches the end-user in an appropriate condition
manner in which it operates is analogous. Depending on the (which typically means that the product maintains sterility)
nature of the product, the bench-top prototype may be used and that the package can be easily opened. Labeling includes
to perform in vitro or in vivo testing, again providing further all printed or graphic material that accompanies a product
evidence of the device’s safety and efficacy. and advertising. Depending on the type of product, labeling
Once this testing is completed, the team should have suf- requirements may include the manufacturer’s contact infor-
ficient information to design the final device. This detailed mation, intended uses, and directions for use, to name a few.
design consists of all of the information necessary to reliably Once all of this information is available, it is submitted to
and repeatedly produce the device. Often, methods such as the FDA, either as a 510(k) premarket notification submission
Design for Six Sigma19,20 are used to help ensure that changes or a PMA submission. Approval from the FDA is required to
in performance due to manufacturing variations do not result legally offer the product for sale in the United States. Other
in performance falling outside the specified bounds. This regulations apply in other countries. However, FDA approval
design phase ends with a review that presents the results of is not necessarily sufficient for the product to gain acceptance.
the experiments performed, the detail design, the bill of mate- In the United States, much of the cost for medical products
rials, and how these perform together to meet the design and procedures is paid for by third-party insurers. Underlying
specifications. these payments is a list of codes that source from the Centers
for Medicare & Medicaid Services (CMS). Because codes are
defined in terms of services provided, and not specific devices,
Clinical Trials new products may be covered by existing codes. However,
Certain classes of medical device must undergo clinical inves- novel devices may require the development of new CMS
tigation21 before the FDA provides marketing approval. Animal codes, and without these codes, many practitioners may be
model testing often is performed before human trials, espe- hesitant to adopt a new device.
cially in those cases for which safety and efficacy are unknown.
Animal use to test a device is a privilege and numerous con-
trols exist to ensure that no more than the minimum number
Financing Product Development
of animals are used, that they are properly housed and cared A corollary to Murphy’s law states, “The first 90% of the task
for, and that upon the conclusion of the experiment, they are takes 90% of the money, and the last 10% takes the other
humanely euthanized. 90%.” Based on the previous discussion about medical device
The institutional review board of the facility that oversees design and development, this statement may, in fact, be overly
the trial must approve all devices that undergo human clinical optimistic. Depending on the type of device, the development
investigation. In addition, the FDA must approve trials for process can require years and tens of millions of dollars.
significant risk devices through submission of an IDE. The Because most medical device inventors are not independently
IDE exempts the manufacturer from certain portions of the wealthy, outside investment often is required to bring a new
Food, Drug, and Cosmetic (FD&C) Act during the trial phase. medical device to market.
However, the manufacturer is still required to comply with all The world of outside investment can be broadly divided
design controls and all necessary manufacturing controls to into two categories: those who invest primarily to obtain a
ensure patient safety and compliance with the manufacturer’s (hopefully very high) return on their investment and all
quality claims. others. The former are typified by angel investors and venture
The goal of a clinical trial is to determine the safety of a capitalists; the latter by universities, federal and state govern-
device and its effectiveness in terms of the intended claims.22 ments, family, and others. Angel investors and venture capital-
This requires concise and specific definition of the study’s ists play such an important role in medical device development
objectives. The objectives must consider factors such as that the entire next section is devoted to them. This section
comparison to approved modalities of treatment or whether starts with some general guidance about business practices
it treats a symptom or an underlying cause. Once these and then discusses non-equity investment opportunities.
Section VII—Computers, Engineering, and the Future 377

The founder of a business focused on developing a new for faculty-founded start-ups, the university will provide
medical device is convinced of the value and importance of favorable terms, such as defraying patent prosecution costs
the invention. However, Ralph Waldo Emerson’s adage that if until the company has revenue. Third, the office of technology
one “build[s] a better mousetrap and the world will beat a transfer may have specialists whose role is to help with
path to your door” is not always true. It takes considerable company formation. These individuals can provide guidance
effort to convince others that the potential product will both with business plan writing, recommendations for local service
address an important medical need and produce a profit.24 providers, and identification of funding opportunities.
Simply providing experimental data is insufficient because the The passage of the Bayh-Dole Act also has motivated uni-
inventor will be asking potential investors to trust him or her versities to commercialize technology. Universities with active
with millions of dollars. technology transfer offices can generate tens of millions of
To access outside funding of most any sort, a company must dollars annually in license fees, royalty, and equity. To capital-
have a business plan.25,26 There are innumerable books about ize on this opportunity, a number of states and universities
writing business plans27,28 (see further suggestions later in this have created funding sources for university inventors to assist
chapter). Two cautionary notes: first, most business plan soft- them to bring their inventions to market. Examples of such
ware programs should be avoided, because they typically do programs include the following:
not add much value. Second, the writing of the plan should  Boston University Ignition Award and Launch Award30:
not be outsourced because the entrepreneur must know the
The former helps bridge the gap between basic science and
entire plan, the sources used to develop it, and so on. Con-
the product development and the latter is designed to help
tained within the plan is a financial model,29 arguably the
faculty members start new companies based on technolo-
single most important aspect of the plan. Attention should be
gies that they invented at the university.
focused on building a rational model, because potential inves-  Michigan Universities Commercialization Initiative31: A
tors will be more interested in the assumptions that underlie
collaboration designed to complement and enhance the
the model rather than the actual number presented.
technology transfer at Michigan academic and research
As a final cautionary note, inventors who intend to seek
institutions by supporting commercialization of IP.
outside investment should engage accountants and attorneys.  University of Utah Technology Commercialization Pro­
Although it is true that forming a business requires little more
ject32: Focused on further developing novel techno­
than completing a simple form, the business’s rules of opera-
logies that are near commercialization in all areas of
tion (referred to as an operating agreement for limited liability
technology.
companies [LLCs] and bylaws for corporations) dictate how
the business is run. If these rules of operation are not profes- For the university inventor, such initiatives offer great value.
sionally written, they may impede potential investments. Sim- First, the competition for funds is limited to a small pool of
ilarly, an accounting system that does not adhere to generally applicants, thus the odds of funding are relatively high. Second,
accepted accounting principles (GAAPs) will make it very dif- the peer-review process typically used provides some valida-
ficult for a proper financial analysis to be performed, which tion of the commercial potential of the technology. Third, the
again may impede potential investments. As a technology- funds often can be used for non–technology-focused activi-
focused business, the most valuable assets are the inventor’s ties, such as engaging business and marketing professionals
intellectual property. Although it is certainly possible to apply from outside the university, which helps to accelerate the
for patents and trademarks independently, engaging an intel- growth of the business. The only downside to these programs
lectual property (IP) attorney is strongly recommended to is that the funding available is typically limited.
ensure that the protection sought is as broad as possible.
Finally, an attorney should review all dealings that can materi-
ally affect the business, such as grants of corporate ownership, Government Research Funds
licensing agreements, and partnering deals, among others, Growing a technology-focused start-up from inception to
before being executed. profitability is a very high-risk proposition. Though exact
figures are not available, it is estimated that less than one
quarter of all businesses are still operational after 10 years—
University Research for technology-focused start-ups, the number is smaller. On
If the developer is a university employee when the medical the other hand, new products and companies account for a
device idea is first invented, the first stop is the school’s office disproportionate fraction of a country’s gross domestic
of technology transfer. Since the passage of the University and product (GDP).33 As such, all levels of government are incen-
Small Business Patent Procedures Act (also known as the Bayh- tivized to support early-stage companies because of the poten-
Dole Act) in 1980, most universities have created offices that tial return on investment. In the United States, one federally
deal exclusively with licensing and IP. For a university inventor, mandated program that supports small businesses dominates
working with the office of technology transfer provides several all others: the Small Business Innovation Research (SBIR)
important benefits. First, as dictated by the Bayh-Dole Act, if program (and the very closely related Small Business Technol-
the research was funded by the federal government, the univer- ogy Transfer [STTR] program).34,35
sity has the right to retain ownership of the IP created. As such, Originally authorized by Congress in 1982, key objectives
should the technology be commercialized, the inventor will of the programs are to stimulate U.S. technologic innovation,
typically receive financial remuneration as a percentage of the create new opportunities for small businesses to participate in
licensing revenue earned by the university. Second, the inven- federally sponsored research and development, and increase
tor of the IP is often granted first right-of-refusal with regard private-sector commercialization of innovations derived from
to licensing the technology for commercial purposes. Often, federal research and development (R&D). As legislated, all
378 Section VII—Computers, Engineering, and the Future

SBIR STTR others are contracting agencies, whereas the NIH and NSF and
Phase I 6 months 12 months
others are granting agencies. With contracting agencies the
Feasibility $100k $100k agency establishes the needs (which are typically highly
focused), there are more fiscal requirements, and the project
Phase II 2 years 2 years
Prototype $750k $750k
initiator (who works for the agency) is the primary proposal
reviewer. With granting agencies, the investigator identifies
Phase III As long as it takes the problem and specifies the approach (as long as it falls
Commercialization non-SBIR/STTR funding within the agency’s purview), there is more fiscal flexibility,
and proposals are peer reviewed. From a business perspective,
Fig. 38.1  General time and funding outline for Small Business Innovation
Research (SBIR) and Small Business Technology Transfer (STTR) programs.
winning SBIR contracts from contracting agencies is benefi-
cial because on successful completion of phase II, the agency
may wish to purchase the product that has just been devel-
oped. Winning SBIR contracts from granting agencies is ben-
federal agencies whose extramural research and development eficial because this indicates that the technology was deemed
budgets exceed $100 million have a mandated set-aside of meritorious to a panel of experts, and so provides great cred-
2.5% of their budget to fund SBIR projects. In addition, all ibility to the company.
federal agencies whose extramural research and development As valuable as the SBIR/STTR programs are, all potential
budgets exceed $1 billion have a mandated set aside of 0.3% applicants for SBIR/STTR funding should be aware of several
of their budget to fund STTR projects. In fiscal year (FY) 2007, facts:
12 agencies participated in the SBIR/STTR program, with  The programs are highly competitive. On average, fewer
total funding of $2.315 billion.
than one in eight phase I proposals is funded and typically
Of the 12 participating agencies, the Department of Defense
half of all phase II proposals are funded.
(DOD) accounted for 54.9%, the National Institutes of Health  Federal funding has restrictions. Project money may not
(NIH) accounted for 28.1%, and the National Aeronautics and
be used for certain necessary business activities such as
Space Administration (NASA), Department of Energy (DOE),
patent prosecution, thus other sources of financing are
and National Science Foundation (NSF) combined accounted
required.
for 14.2% of the of the total funding, respectively. Developers  The SBIR/STTR cycle is slow. It is not atypical for a project
of medical devices would typically focus on applications to the
(defined as the first writing of a phase I proposal to the
NIH; however, the DOD and NSF occasionally seek medical
end of the phase II project) to exceed 3.5 years. For certain
device applications as well.
types of products, where time-to-market is important, this
SBIR/STTR programs are three-phased programs, as out-
long time frame may not be acceptable.
lined in Figure 38.1. The durations and dollar figures are  The federal government has rights to IP developed.
statutory guidelines—each agency sets its own rules.
Quoting a recent DOD SBIR solicitation: “The govern-
The key requirements to allow access to the source of
ment receives a royalty-free license for its use, reserves the
funding are that the firm be a U.S. for-profit business with 500
right to require the patent holder to license others in
or fewer employees and that work be performed in the United
certain limited circumstances, and requires that anyone
States. Other requirements about corporate ownership, prin-
exclusively licensed to sell the invention in the United
cipal investigator employment, and so on can be found online.
States must normally manufacture it domestically.”
Major changes to the SBIR program are being proposed in
Congress during the production of this book, and so the
information provided herein is likely to change. (The reader
may refer to information about these programs online at State Commercialization Funds
www.sbir.gov.) State governments realize that having a vibrant start-up com-
Participating in the program is straightforward. A small munity is valuable for the state. Companies that become suc-
business must first obtain a data universal numbering system cessful will hire more people, spend more money, and pay
(DUNS) number (fedgov.dnb.com), federal tax identification more taxes. To help build such communities, many states
number (www.irs.gov), and a bank account that accepts elec- have created programs that support technology-focused
tronic funds transfers, and then register with the central con- start-up businesses. Such programs span the gamut from
tractor registration (www.ccr.gov). Next, the small business business plan competitions that provide ten of thousands of
reads the solicitations published by the 12 agencies and applies dollars to SBIR matching programs that offer hundreds of
for phase I projects for which it is qualified. The phase I appli- thousands of dollars to other competitive programs that
cation takes the form of a proposal along with a project provide millions of dollars. The following list highlights
budget, brief resumes of key personnel, and letters of support several such programs.
from partners or customers. Small businesses that successfully
complete a phase I project will almost always be invited to  Texas Emerging Technology Fund (ETF): Established in
submit a phase II proposal. The phase II proposal is very much 2005, the Texas ETF was intended to “expedite innovation
like the phase I, but with two additions: a section that describes and commercialization of research, promote a substantial
the outcome of the phase I project, and a commercialization increase in high-quality jobs, and increase higher educa-
plan that describes how the technology being developed will tion applied technology research capabilities.”36 Compa-
be brought to market. nies with disruptive technologies were encouraged to
There are some important differences between the SBIR/ apply for funding up to $5 million. The decision process
STTR programs offered by the agencies. The DOD and several included a thorough vetting process (technology, IP, and
Section VII—Computers, Engineering, and the Future 379

commercial potential), which added significant credibility revenue, they can provide easy access to cash and other useful
when seeking future investments. The funding mecha- expertise.
nisms were very favorable from the companies’ perspective
and typically took the form of equity. (It is no longer
available.) Friends, Family, and Fools
 Kentucky SBIR/STTR Matching Funds Award37: The pur­ Entrepreneurs often turn to their friends, families, other
pose of the program is to foster job creation and economic people they know, and credit cards to help fund the early
development in Kentucky by increasing the competitive stages of a business. These individuals typically invest because
position of small businesses to attract SBIR/STTR funding. they personally know the entrepreneur and trust that he or
This is accomplished by providing matching funds to com- she will provide a return on their investment. Unlike angel
panies that have been granted a federal SBIR/STTR investors, these individuals are not required to be high–net
program, phase I or phase II, in one of the state’s identified worth individuals. However, it is critically important that such
focus areas. Companies can receive up to 100% of the transactions be managed properly, with appropriate legal
amount of the SBIR/STTR award (with some limitation) documentation and signatures.
and may receive up to a total of five such awards. The
application is not subject to vetting (other than ensuring
that certain criteria are met), and the funds are provided Foundations, Not-for-Profit Organizations
in the form of a grant. Organizations whose mission is aligned with the company’s
product development efforts can potentially provide assis-
Although these programs vary widely, they typically have tance. In some cases, the assistance may be other than cash,
several attributes in common: because some not-for-profits are restricted in their ability to
finance for-profit businesses. However, these organizations
 The majority of the funding provided is to be used within
typically have extensive networks of like-minded people
the state that makes the award. In fact, companies that
with whom they can share information about the entrepre-
move out of the awarding state may be obligated to repay
neur’s efforts, which can lead to investments and other
the monies upon so doing.

opportunities.
The programs typically focus on commercialization, not
basic research. The states are investing in the company
with the hope that jobs, and taxes, will be created in the Commercial Banks
relatively near future.

Commercial banks rarely provide loans to start-up businesses
The funding may be provided as a grant, loan, or convert-
because collateral is required to secure the loan. Unless the
ible debt. The entities that run these programs typically
entrepreneur is willing to collateralize his or her own assets,
structure the deals so that they do not hinder or preclude
such as a house, there is rarely anything of bank-accepted
future investments.

value within the business to offer as collateral.
Funds often are provided on a first-come, first-served basis,
Figure 38.2 overlies a product’s life cycle with the typically
with a fixed amount of funding allocated to the program.
available sources of funding for the particular stage of
As such, meritorious applications to these programs can
development. The dollar values shown provide guidance
go unfunded if the program exhausts its prescribed dollar
about the typical order of magnitude of the funding. The light
allocation (e.g., toward the end of the fiscal year).

blue shaded section, that period of time during which cumu-
There is one potential problem with state-run programs.
lative profits are negative, is often referred to as the “valley
In certain states, the legislation for the program may not
of death.”
provide sufficient privacy protection for documents sub-
mitted for consideration. Before applying for one of these
programs, and before submitting any written reports, a Angel and Venture Funding
device developer must fully understand what access to
Angel investors and venture capital (VC) firms are potential
documents is available to interested parties using the
sources of capital available to entrepreneurs trying to start a
Freedom of Information Act (FOIA).
company. Angel investors (also known simply as angels) are
Other than potential FOIA issues, these state-run programs high–net worth individuals who invest their own money into
are valuable. Companies need unrestricted money to find cus- companies. Often angels have domain expertise or a personal
tomers, market their products, and seek potential investors; interest in the technology being developed. Although the mag-
other sources of funding, such as SBIR/STTR, are limited in nitude of angel investing in the United States is large, esti-
their ability to support such activities. Thus for companies mated to exceed $23.3 billion in 2010, individual angels, or
that seek to commercialize a product that results from R&D angel groups, typically do not invest more than several
activities, these funds can be valuable. However, the award hundred thousand dollars into a single firm. Due to the high
amounts are typically insufficient to bring a product, espe- costs associated with bringing a medical product to market,
cially a medical device, to market on their own. angels are not typically well equipped to provide substantial
assistance to entrepreneurs in this field. Thus the remainder
of this section focuses on venture capital.
Other Sources of Funds A venture fund is a private equity investment entity that
There are several other possible sources of funding for start-up invests capital in companies on behalf of third-party investors.
businesses. Although these sources typically do not have the Usually structured as an LLC or a general partnership, VC
financial resources to fund a company from inception to first firms receive investment capital from limited partners, be they
380 Section VII—Computers, Engineering, and the Future

Investigation Feasibility Development Introduction Growth Maturity

Equity markets,
banks ($10M)

Profit Venture capital ($1M)

Pre-seed funds, angels ($100k)

SBIR/STTR, other gov.


support ($100k)

Friends, family,
founders ($10k)

Time
Fig. 38.2  The life cycle of a start-up company, showing stages of development and typically available funding sources. SBIR, Small Business Innovation
Research; STTR, Small Business Technology Transfer.

Pre-Deal Capitalization Structure Class B Preferred Units


Common Common Pro forma Pro forma Preferred Pro forma Pro forma
shares options units ownership issuance units ownership
Entrepreneur 10,000,000 – 10,000,000 100.0% – 10,000,000 66.7%
Venture capitalist – – – 0.0% 5,000,000 5,000,000 33.3%
Total 10,000,000 – 10,000,000 100.0% 5,000,000 15,000,000 100.0%

Fig. 38.3  Simplified capitalization table showing the change in percentage ownership upon accepting an investment.

high–net worth individuals or large financial institutions or go-to-market strategy to name just a few of the “bonuses” that
some combination of the two. VC firms typically make invest- the ideal VC firm can offer to the entrepreneur.39
ments in exchange for company shares, that is, equity in the The timeless VC adage in the medical device space goes
company is exchanged for capital financing from the venture something like this: every entrepreneur has impenetrable IP
capitalist. and every venture capitalist promises to aid the entrepreneur
The exact number of shares that the VC firm receives for to surmount every hurdle during the process of taking a
the cash investment is determined by what is called the pre- product to market. The truth of the matter usually lies some-
money valuation of the given company. The pre-money valu- where in between the two.
ation is a negotiated value of the company’s intrinsic value
before taking money from the VC firm. If the VC firm and the
company agree that the company is worth $10 million before What Equity Investors Are Seeking
the investment (i.e., the pre-money valuation is $10 million), One of the first hindrances to receiving VC is getting the atten-
and the amount of invested capital is $5 million, then the VC tion of a venture capitalist. The best means to find a venture
firm is effectively buying 33% of the company. By putting $5 capitalist is through personal contacts who can then recom-
million into a company that is worth $10 million, the post- mend the inventor and thus personalize the introduction. A
money valuation of the company is $15 million. More simply trusted contact can break down even the strongest of barriers.
put, $5 million added to $10 million equals $15 million; $5 Another means to meet a venture capitalist is at a conference.
million divided by $15 million equals 33% of the company The last method is the “cold call” or “cold email,” in which the
(Fig. 38.3).38 venture capitalist is contacted without a formal introduction.
Accompanying the capital that entrepreneurs can receive The venture capitalist is thereby introduced to the company
from VC firms is the experience that venture capitalists can or idea for a company in an impromptu and extemporaneous
offer to start-up companies. For immature companies, VC can way. (A list of venture capitalists that focus on medical devices
provide the added benefit of the managerial, technical, and can be located at the following website: www.devicelink.com.)
industry-based expertise of the venture capitalists. For medical David Lawee, the founder of Mosaic Venture Partners offers
device companies specifically, the venture capitalist may these tips for getting in the door:
help the entrepreneur navigate through the complex regula-
tory approval process, set up clinical trials, understand  It dramatically improves your chances to come in through
the reimbursement coding system, establish a manufacturing a trusted reference.
relationship, ensure timely and complete filing of IP protect-  You have to be savvy about who those people [are]…not
ing patents, negotiate distribution contracts, or develop a to listen to everybody and say, “Oh, that guy’s a trusted
Section VII—Computers, Engineering, and the Future 381

referral.” If you associate with someone who is not consid- The clinical and regulatory process is one of the many
ered to be credible, then you get tainted by that. hurdles that entrepreneurs must leap over on the path to
 There is so much available on the Internet. Go out, read taking a medical device to market. This process, and indeed it
up, and learn about the market. Figure out how you’re is a process, requires a deliberate and well-executed strategy
going to get to the influencers. We do that all the time for when dealing with the FDA. Whether the company is filing a
our companies. You need to do that…it’s just a normal 510(k) or a PMA, the clinical trials that will support the FDA
business skill.40 filing require thoughtful planning, careful selection of sites
and principal investigators, and clearly defined endpoints and
During the initial contact phase, it is customary to provide the protocols. The choice of the PMA or 10(k) mechanism can be
venture capitalist with an executive summary or a “one-pager” important because most recalls are of medical devices origi-
that summarizes the company. The one-pager is ideal because nally cleared through the 510(k) process or were considered
it forces the entrepreneur to include only the most pertinent of low risk and so exempt from review.43
information that relates to the device in a “one page only” When determining the economics of a medical device, the
format. After sending the one-pager and arranging a confer- venture capitalist considers the micro- and macro-effects that
ence call or an in-person meeting, it is imperative to have a the product will have on the system. Devices often are used
solid investor PowerPoint presentation. This presentation not directly by doctors or other medical practitioners. When ana-
only helps guide the company during the story-telling process, lyzing a device, the cost to the doctor, namely the physician
but also shows the VC firm how detail-oriented and stream- economics or micro-effect, must be considered in the grand
lined the company truly is. economic picture. With respect to the macro-effect, the
Most venture capitalists target medical device technology venture capitalist considers the costs to the system as a whole,
companies at a specific stage, be it early stage (i.e., pre-FDA for example, does the device increase office throughput or
approval) or late stage and post-FDA approval (i.e., market reduce operating room time? Because of the American third-
launch). Some venture capitalists are stage agnostic and will party payer system, venture capitalists must consider the reim-
entertain entrepreneurs at any stage in the development cycle. bursement rate of a medical device. Devices are paid for as
The goal of VC investing is to make money on a calculated part of a Diagnosis-Related Group (DRG) hospital code,
risk investment. The goal, as novel as it may sound, is to invest through a Current Procedural Terminology (CPT) code, or by
as little as possible and to generate as high a rate of return as the patient him- or herself (an out-of-pocket expense). The
possible. Buy low, sell high. All venture capitalists have their process to establish a new CPT code is a lengthy (greater than
own investment thesis, but certain investment metrics do 1 year) and exacting process that relies on careful filings and
exist. Following is a general list of the basic investment criteria ultimately on sales generated from the device. The venture
of a venture capitalist: capitalist will consider the true “purchaser” of the device when
evaluating a device.
 Unmet clinical need
When it comes to distributing and selling a device, the
 Large uncrowded market

venture capitalist will consider the marketplace and the big
Strong intellectual property

players in the given clinical space. Because of established dis-
Clinical and regulatory process

tribution channels, large medical device companies can easily
System and physician economics

add a new product to their sales representatives’ “bags” and
Established reimbursement protocol

effectively blanket the country with the device in question.
Distribution and sales strategies

Unfortunately, a fledgling company does not have the where-
Experienced management team

withal or the resources to emulate this national or global
Multiple on invested capital

approach. The venture capitalist will consider the competitive
Overall cash return on investment
environment during his or her analysis and weigh the different
This list, if thoughtfully addressed by the medical device sales strategies.
company in question, will help guide the venture capitalist’s How intimately venture capitalists work with their portfolio
decision-making process. The goal of the start-up company is companies will determine how important the vetting of the
to put together a clear presentation that shows that the device management team’s biographies is to them. In the end, the
is novel and meets an unmet need for a specific patient popu- shepherding of portfolio companies through the entire process
lation. With that in mind, the entrepreneur must show how will require intense interaction. The better the communica-
this device is a major advancement over existing technology tion process is, the more successful the experience will prove
and that it fulfills a truly unmet clinical need in a market that to be. Hardworking and assiduous people often generate suc-
will generate significant returns.41 cessful outcomes. The returns that a venture capitalist looks
When the market is considered, the saying is “the bigger, for in its investment process are based largely on the method-
market, the better.” Unfortunately orphan diseases often are ology of the specific VC firm. In the end, a patent-protected
excluded after a market-based analysis. Some big markets device that addresses a disease state with a large market and
include the most common disease pathologies such as diabe- that can be produced with high gross margins and low operat-
tes, congestive heart failure, obesity, or sleep apnea. ing costs is the venture capitalist’s four-leaf clover.
Strong intellectual property is the sine qua non of VC invest-
ing. Patents protect an investment and in essence are the true
assets of a start-up company. Because patent assessment Pros and Cons of an Equity Investment
requires real dollars and cents in terms of costs for legal review, The pros and cons of taking on an equity investment lie in the
this part of the due diligence process is often the last step after locus of control. Control comes in two forms: financial and
a letter of intent, or financial term sheet, is extended to the administrative. Before taking money from a venture capitalist,
company.42 the entrepreneur has 100% control over his or her company.
382 Section VII—Computers, Engineering, and the Future

The decision making is not diluted by a board of directors and 6. Management biographies
is solely in the hands of the founders. Once the company 7. Organization chart
decides to take on capital from a venture capitalist, the power 8. Patent family tree
structure of the company changes significantly. In exchange 9. Summary of preclinical studies
for the risk of their investment, the venture capitalists gain 10. Summary of clinical trial protocols
significant control over company decisions, in addition to a 11. Clinical trial contacts
significant portion of the company’s ownership (and conse- 12. Explanation or overview of device
quently value). 13. Capitalization table
The key to ensuring a safe journey lies in shrewd negotia- 14. Market model
tion of the investment documents. Every line in every one of 15. Indications for gold standard competitor
the investment documents, from the purchase agreement 16. Competitive grid
to the shareholders’ agreement, is written for a reason.44 To 17. Clinical literature review
ensure that no surprises arise, seasoned legal representatives 18. Clinical literature articles
should scour the documents and negotiate the most favorable
terms possible. The list of problems that can arise from poorly At this stage in the investment algorithm, a venture capitalist
negotiated documents is limitless, so the enterpreneur must will decide whether to extend a letter of intent, often referred
be keenly aware of all the pitfalls and windfalls that exist in to as a term sheet. This letter of intent will clearly define the
the legal contracts. investment parameters of the venture capitalist, namely, the
The pros of taking on VC money depend upon the venture pre-money valuation, the amount of invested capital, whether
capitalist in question. Before taking money from a given or not warrants are to be a part of the transaction, the tranch-
venture capitalist, the venture capitalist should be asked for ing of the investment, and any milestones that the company
permission to contact some of their portfolio company chief must meet to receive the different investment tranches.
executive officers (CEOs) or general managers (GMs). A (“Tranche” is French for a slice, so if a venture capitalist
transparent venture capitalist will not find anything amiss decides to invest $6 million, it may provide the investment in
with this request and should facilitate this process. A good three $2 million tranches.) The letter of intent also usually
firm has nothing to hide. If red flags emerge from these con- defines the capitalization structure of the company and the
versations, the answer to the question of whether or not this terms of the stock investment—conversion, voting rights,
venture capitalist will add value on top of his or her equity redemption rights, antidilution protection, liquidation prefer-
investment should be apparent. ence, and first-offer rights. The rest of the letter of intent
If the venture capitalist receives positive reviews, the defines the closing conditions, the transactions fees, and the
ideal venture capitalist will buttress the management team legal and confidential nature of the agreement.
with expertise in any number of medical device-related After the so-called “doctor calls” and the negotiation of the
areas. Suffice it to say, a seasoned VC firm will offer its experi- letter of intent, the legal evaluation of the company’s IP is
ence in the field of medical devices, its contact lists, and usually the last step in a VC firm’s due diligence process
the overall expertise of its team, and so ensure that capital because of the high cost of IP attorneys’ fees.
will be coupled not with a loss of control but a gain of a If he or she decides to pursue an investment, the venture
valued resource. capitalist will send a letter of intent to the company with a
monetary valuation of the company. From this starting point,
the negotiation between the company and the venture capital-
The Equity Investment Process ist begins. Ideally, the terms on either end meet somewhere in
After a phone call or an in-person investor PowerPoint pre- the middle about the valuation and the acceptable terms for
sentation, the venture capitalist will perform due diligence on the transaction to occur.
the company based on the interest level generated by these
interactions. The key to getting the attention of the venture
capitalist is a clear presentation that hits on the key parameters
Conclusion
that venture capitalists consider important to make an equity To bring a novel medical device to market is a challenging
investment. Initial due diligence can either precede or come proposition. The range of activities that need to be successfully
after another conference call or another in-office meeting. The managed range from understanding the clinical issues to
number of the calls and meetings varies from firm to firm, but developing an engineered device to raising capital to comply-
the average inclusive number probably exceeds three (calls and ing with federal regulations to eventually selling the product.
meetings). The venture capitalist also may supplement infor- Despite the challenges, this is a process undertaken by people
mation requests and in-person demonstrations by the on a daily basis because of the potential rewards. The single
company with calls to physicians and other trusted experts in most important fact to remember is that others who have
the given field. A typical due diligence binder that the venture done this before are available, sometimes at little to no cost,
capitalist will request may include any of the following to assist new inventors in their endeavors. Therefore, one of
documents: the most important activities in which a new entrepreneur
should participate is networking. Networking provides the
1. Financial plan and top line assumptions for the plan opportunity to learn about resources available and lets others
2. Milestone chart reflecting key milestones of the plan learn about the new project. Through networking it is possible
3. Term sheet proposal to obtain recommendations for service providers, find em­
4. Investor presentation ployees, and identify people who can serve on a board of
5. Regulatory process advisors.
Section VII—Computers, Engineering, and the Future 383

Regardless of where an enterpreneur lives, there are local 17. Fries RC. Handbook of medical device design. New York: Marcel Dekker;
2001.
resources that can provide assistance. Most every city has a
18. US FDA/CDRH: Good manufacturing practice/quality systems. at
chamber of commerce,45 an organization whose focus is on http://www.fda.gov/medicaldevices/deviceregulationandguidance
the local business community. Chambers typically have regu- /postmarketrequirements/qualitysystemsregulations/default.htm.
larly scheduled meetings, which are usually great places for Accessed Sept 4, 2012.
networking. They also offer a range of services, such as provid- 19. El-Haik B. Medical device design for six sigma: a road map for safety and
effectiveness. Hoboken, NJ: Wiley-Interscience; 2008.
ing group insurance, that may be of value to small businesses. 20. Justiniano JM. Six sigma for medical device design. Boca Raton, FL: CRC
In addition, many chambers have SCORE offices,46 business Press; 2005.
counseling and mentoring organizations, another invaluable 21. Becker KM, Whyte JJ. Clinical evaluation of medical devices: principles and
resource for startups. case studies. Totowa, NJ: Humana Press; 2006.
22. Chiacchlerini RP. Medical device clinical trial design, conduct, and analysis.
Many cities, or regions, have economic development
Health care technology policy I: the role of technology in the cost of health
corporations, typically public-private partnerships focused on care; 1994. p. 278–85.
growing businesses. Services offered vary widely, but some 23. DeVor RE. Statistical quality design and control: contemporary concepts and
offer business incubator space, accelerator programs, and/or methods. Upper Saddle River, NJ: Pearson/Prentice-Hall; 2007.
consulting services for entrepreneurs. All states have state- 24. Dorf RC. Technology ventures: from idea to enterprise. New York:
McGraw-Hill; 2007.
wide economic development corporations that establish 25. Mullins J. The new business road test: what entrepreneurs and executives
programs (such as those discussed previously), provide should do before writing a business plan. 2nd ed. Upper Saddle River, NJ: FT
tax incentives, and a broad range of programs to assist Press; 2008.
businesses. 26. Kawasaki G. The art of the start: the time-tested, battle-hardened guide for
anyone starting anything. New York: Portfolio Hardcover; 2004.
All states also have small business (and technology) devel-
27. Rule RC. The rule book of business plans for startups. Irvine, CA:
opment centers.47 These organizations provide counseling, Entrepreneur Press; 2000.
training, research, and advocacy for small businesses. The 28. DeThomas A, Derammelaere S. Writing a convincing business plan.
program, run jointly with the federal Small Business Admin- Hauppage, NY: Barron’s Educational Series; 2008.
istration, typically provides everything from classroom learn- 29. Benninga S. Financial modeling. 3rd ed. Cambridge, MA: MIT Press; 2008.
30. BU—Office of Technology Development—awards/funding overview.
ing to one-on-one coaching. Available at http: www.bu.edu/otd/awards. Accessed Sept 4, 2012.
The bottom line is this: starting a business and working to 31. Michigan Universities Commercialization Initiative (MUCI) Home Page.
make it successful is an extremely rewarding process. In so Available at http://www.muci.org. Accessed Sept 4, 2012.
doing, the entrepreneur enriches himself or herself, his or her 32. The University of Utah. Available at http://www.research.utah.edu/funding/
tcp.html. Accessed Sept 4, 2012.
community, and in the case of medical devices, society as a
33. Lord Robert McCredie, M.O.A.K.F.F.F. Innovation: from new knowledge to
whole. Although challenging, the resources available to entre- new products. (2002).Available at http://www.atse.org.au/
preneurs are greater than ever, and make now the best time to index.php?sectionid=344. Accessed Sept 4, 2012.
start a business. 34. SBIR—Small business innovation research. Available at www.sbir.gov.
Accessed Sept 4, 2012.
References 35. SBIR/STTR Gateway—news and search engine. Available at www.zyn.com/
sbir. Accessed Sept 4, 2012.
1. Harman RJ. Development and control of medicines and medical devices. 36. Texas Life Science Center. Available at http://www.texaslifesciences.com/.
London: Pharmaceutical Press; 2004. Accessed Sept 4, 2012.
2. Pietzsch JB, Shluzas LA, Pate-Cornell ME, et al. Stage-gate process for the 37. Technology & innovation—funding| The Kentucky Cabinet for Economic
development of medical devices. J Med Dev 2009;3:021004–15. Development. Available at www.thinkkentucky.com/DCI/SBIR. Accessed
3. Linehan JH, Chaney A. Academic/industry challenges for medical device Sept 4, 2012.
development. Sci Transl Med 2010;2:63mr6. 38. Wilmerding A. Deal terms. Boston: Aspatore; 2003.
4. Yock PG, Brinton TJ, Zenios SA. Teaching biomedical technology innovation 39. Marks, K.H. 0.14.2.b: The handbook of financing growth: equity
as a discipline. Sci Transl Med 2011;3:92cm18. investment documentation—encyclopedia—library—VC experts.
5. Kutz M. Biomedical engineering & design handbook. Vols. I and II. New Available at http://vcexperts.com/vce/encyclopedia-of-private-equity.
York: McGraw-Hill Professional; 2009. Accessed Sept 4, 2012.
6. Fries RC. Reliable design of medical devices. 2nd ed. Boca Raton, FL: CRC 40. Knight J. Equity financing: understanding venture capital. Available
Press; 2005. atwww.canadaone.com/ezine/aug00/venture_capital.html. Accessed Sept 4,
7. Prutchi D, Norris M. Design and development of medical electronic 2012.
instrumentation: a practical perspective of the design, construction, and test 41. Knight HJ. Patent strategy for researchers and research managers. Chichester,
of medical devices. Hoboken, NJ: Wiley-Interscience; 2004). UK: John Wiley & Sons; 2001.
8. Kucklick TR. The medical device R&D handbook. 2nd ed. Boca Raton, FL: 42. Elka N. Medical device firms can boost their chances of success. Available
CRC Press; 2005. atwww.altassets.com/casefor/sectors/2007/nz11478.php. Accessed Sept 4,
9. Pahl G. Engineering design: a systematic approach. London: Springer; 1996. 2012.
10. Pugh S. Creating innovative products using total design: the living legacy of 43. Zuckerman DM, Brown P, Nissen SE. Medical device recalls and the FDA
Stuart Pugh. Reading, MA: Addison-Wesley; 1996. approval process. Arch Intern Med 2011;171:1006–11.
11. Cohen L. Quality function deployment: how to make QFD work for you. 44. Feld B, Mendelson J. Venture deals: be smarter than your lawyer and venture
Reading, MA: Addison-Wesley; 1995. capitalist. Hoboken, NJ: John Wiley & Sons; 2011.
12. Grady JO. System requirements analysis. New York: McGraw-Hill; 1993. 45. Chamber of Commerce, USA Directory, Visitor Bureaus United States
13. Blanchard BS. Systems engineering and analysis. Upper Saddle River, NJ: Directory. Available at www.2chambers.com. Accessed Sept 4, 2012.
Pearson Prentice Hall; 2006. 46. SCORE Counselors to America’s small business. Available at www.score.org/
14. Fries RC. Medical device quality assurance and regulatory compliance. New index.html. Accessed Sept 4, 2012.
York: Marcel Dekker; 1998. 47. America’s Small Business Development Center Network.
15. Cheng M, World HO. Medical device regulations: global overview and Available at www.asbdc-us.org. Accessed Sept 4, 2012.
guiding principles. Geneva: World Health Organization; 2003.
16. U.S. Food and Drug Administration—Center for Devices and Radiological
Health. at http://www.fda.gov/AboutFDA/CentersOfficesMedicalProductsand
Tobacco/CDRH/default.htm. Accessed Sept 4, 2012.
VII
Chapter
39  

Engineering Issues
Brett Trimble and Jens Bracht

Introduction death. For this reason ICP monitoring is important in neuro-


critical care. Nutrients such as oxygen and glucose are sup-
History and Background plied and waste products removed from the brain through the
By most accounts the modern era of neuromonitoring began blood circulation. Although the brain is a relatively small
in the 1950s with reports of continuous intracranial pressure organ at 2% of body weight, it receives 20% of the body’s
(ICP) measurements in humans by Janny et al.1,2 ICP was blood flow and accounts for 20% of oxygen and 25% of
measured by connecting a strain gauge pressure transducer to glucose consumption.3 Significantly, the brain does not store
a fluid-filled tube connected to a catheter with its distal end oxygen. Alterations in blood flow are common in neurocritical
implanted in the patient’s cerebral ventricle. The use of a care patients, and there is much interest in monitoring blood
strain gauge, as opposed to a manometer, to measure pressure flow in both the large vessels leading to and from the brain
made ICP monitoring safe, easy, and accurate. The frequency and the microvasculature within the brain parenchyma. Sys-
response of the sensor and associated electronics was suffi- temic blood pressure is commonly monitored because it is the
cient to allow visualization of the pulsatile ICP waveform. In driving force for cerebral blood flow (CBF). The concentra-
addition periodic measurements could be plotted over time, tion of carbon dioxide (CO2), which is a powerful vasodilator
leading to the discovery of the well-known and clinically and affects CBF is indirectly monitored in the expired breath.
useful Lundberg waves. Continuous ICP monitoring has CO2 also has been monitored directly in the brain. Oxygen
become the cornerstone of critical care monitoring for patients tension can be measured in both the circulation and in brain
admitted to neurocritical care units (NCCUs). tissue. The oxygen saturation of systemic blood is routinely
From an engineering point of view, it is interesting that the monitored, and estimates of regional saturation of cerebral
application of a simple electrical sensor and analog electronics blood also are possible.
resulted in a significant improvement in clinical practice. This It is common for alterations in cerebral metabolism to
is a pattern that has repeated itself in the years since Lund- occur in neurocritical care patients. The brain normally pro-
berg’s original work in the 1960s. The remarkable develop- duces energy in the form of adenosine triphosphate (ATP)
ments in electronics and sensing technologies driven by the through the Krebs cycle; oxygen and glucose are necessary for
computer, telecommunications, and aerospace and defense this ATP production. When cerebral metabolism is disturbed,
industries have been applied to many aspects of medicine for example, by a lack of oxygen availability, the brain relies
including monitoring devices used in neurocritical care. more on anaerobic glycolysis that produces less ATP. Under
these conditions, brain cells may not function normally and
may even lose structural integrity.4 This condition is some-
Clinical Background times called hyperglycolysis. This condition as well as other
An examination of engineering issues associated with neuro- metabolic disturbances can be inferred by various chemicals
monitoring devices must be made in the context of the goals released by the brain. A portable analyzer can test samples
and challenges of neurocritical care. Neurocritical care patients collected from the interstitial fluid by dialysis probes implanted
commonly suffer from traumatic brain injury (TBI), from directly in the brain to determine the concentration of the
neurovascular diseases such as subarachnoid hemorrhage, or chemical in question. The clinical utility of these measure-
have undergone a neurosurgical procedure such as resection ments is the subject of significant research.
of a brain tumor. In addition to the original injury or disease,
patients are at risk of “secondary” injury because of the unique
nature of the head and brain. One of the primary goals of Neuromonitoring Systems:
neurocritical care is the prevention of these secondary inju- An Engineering Prospective
ries. The purpose of monitoring equipment used in neuro-
critical care is to enable the clinician to identify signs and The Ideal Monitor
symptoms of the primary disease, to warn of impending sec- The ideal monitoring system would be noninvasive, provide
ondary insults, and to help judge the efficacy of treatment. continuous information, interrogate the entire brain, present
Because the brain is encased in the rigid skull, there is the information in a way that is easily understood, be compat-
limited room for the brain to swell when injured. Left ible with other monitoring and imaging systems including
unchecked, swelling (edema) can lead to serious morbidity or magnetic resonance imaging (MRI), take up little or no space,
384 © Copyright 2013 Elsevier Inc. All rights reserved.
Section VII—Computers, Engineering, and the Future 385

meet all regulatory requirements both medical and technical, secure and leakproof fit. The lead-in threads are usually
and cost very little. Few if any currently marketed devices meet tapered in a manner similar to pipe threads with the crest
all of these requirements. The following sections describe the height of the initial one to three pitches smaller than the
basic components of most common existing monitoring diameter of the twist drill used to cut the insertion hole in the
systems, give an overview of the types of sensing techniques skull. The compression fitting must resist axial movement of
that are or have been used, and assess the pros and cons of the probe and provide a leakproof seal around the outer
each technique. surface of the probe. The maximum compressive force that
Most monitoring systems used in neurocritical care consist can be exerted must be limited to an amount that will not
of a sensor in contact with the scalp or directly implanted into damage the probe. Providing strain relief at the junction
the brain, some kind of fixation device that keeps the sensor between the top of the bolt and the probe is a good practice.
in place during the monitoring period, connecting cables, and The ultimate strength of both the bolt and probe in bending
a stand-alone electronic monitor to operate the sensor and and tension must be carefully considered because the probe
display data or connect to a bedside monitor. and bolt often are subjected to significant loading when the
probe or a connecting cable is inadvertently pulled when it
catches on a fixed object during patient transport, the patient
Fixation Techniques falls out of bed with the cable wrapped on the bed frame or
Fixation is an extremely important aspect of monitoring other stationary object, or the patient pulls on the probe in a
systems. Fixation systems must ensure consistent alignment or semiconscious state. It is far better for the bolt or probe
contact between the sensor and the patient. Two primary fixa- (outside the patient) to break than the bolt to break in the
tion techniques are used for indwelling sensors: bolt fixation skull (Fig. 39.2).
and tunneling. Most indwelling sensors take the overall physi-
cal shape of a long slender cylinder usually referred to as a
catheter or probe with the sensor located at the tip of the probe Tunneling
(Fig. 39.1). Perhaps the oldest fixation method is tunneling. Tunneling
refers to the surgical technique of routing an elongated cath-
eter under the scalp toward the insertion site. This method has
Bolt Fixation the advantage of good fixation to the patient using stitches in
The bolt device is composed of a short metal cylinder with the scalp and better infection control than simply routing the
self-tapping threads at one end and some kind of compression catheter out of the scalp directly over the insertion site. This
fitting at the other end. The bolt is usually threaded to a depth technique is commonly used with fluid-filled ventriculostomy
of approximately the thickness of an adult skull of approxi- ICP monitoring. It also has become common for transducer-
mately 0.7 cm. The thread pitch is usually sized such that at tipped probes to be affixed to the patient via tunneling. It is
least three or four fully formed threads will be in contact with important to recognize that the probe must be capable of
the skull with the bolt in place. If the thread pitch is too large, making a right-angle turn into the skull into the insertion site
not enough contact is made by the thread crests to ensure a under the scalp without being damaged. Because the connec-
tors on the proximal end of most probes are too large to be
tunneled under the scalp, many of these devices are not

Fig. 39.1  Bolt fixation device. Licox oxygen sensing catheter and Fig. 39.2  Bolt fixation device. Camino intracranial pressure bolt in situ.
fixation bolt. (Used with permission of Integra LifeSciences Corporation.) (Used with permission of Integra LifeSciences Corporation.)
386 Section VII—Computers, Engineering, and the Future

Fig. 39.4  Hewlett-Packard bedside monitor connecting cables and


Fig. 39.3  Tunneling fixation. Ventrix intracranial pressure catheter Camino monitor. (Used with permission of Integra LifeSciences Corporation.)
tunneled under scalp. The inset shows the sensor in the tip of the
ventricular catheter. Note the shape of the probe tip (bullet-like) that
limits tissue injury during insertion. (Used with permission of Integra
LifeSciences Corporation.)
specific parameter type such as pressure or temperature.
Commonly displayed parameters include electrocardiogram
(ECG), heart rate, temperature, blood oxygen saturation,
tunneled per se but are routed through a plastic sleeve that cardiac output, and mean arterial blood pressure among other
has itself been tunneled under the scalp. This “tunneling options. Most physiologic pressures such as arterial pressure,
sleeve” usually takes the form of a plastic tube connected at pulmonary artery pressure, and pulmonary capillary wedge
one end to a solid trocar. After the sleeve has been tunneled pressure are monitored using strain gauges. BSMs are there-
under the scalp, the trocar is cut from the sleeve and the cath- fore designed to provide electrical excitation of the strain
eter is passed through the sleeve toward the insertion site. The gauge and to interpret pressure from the electrical signals
tip of the catheter is then implanted and the catheter and/or returned form the strain gauge. Neurologic-related parame-
sleeve are sutured to the scalp via loops (Fig. 39.3). ters, other than ICP signals measured by fluid filled systems,
are generally not included in the current generation of BSMs.
Devices that measure parameters that are not included as stan-
Other Fixation Methods dard features of the BSM commonly connect to it via analog
Noninvasive monitoring devices such as electroencephalo- strain gauge emulation or by serial means when available.
graph (EEG) electrodes, near infrared (NIR) oximeters, and Cable connectors must accommodate the various makes and
transcranial Doppler (TCD) probes are affixed by hand, by models of BSMs (Fig. 39.4).
mechanical means, or by the use of some kind of adhesive
tape–like substance. Heavy devices that are not usually used
to make continuous measurements such as TCD transducers
Primary Sensors
are simply held to the head by the technician or are sometimes The injured brain can be a difficult sensing environment. In
fitted to a device that looks like an eyeglass frame. Lighter particular intracranial hemorrhage of one form or another
transducers like EEG electrodes or oximeter leads usually are results in free and clotted blood both of which can damage or
held in place by adhesive patches. The adhesives used must confound many types of indwelling senor elements. Blood or
not irritate the skin during monitoring durations of up to large proteins in the cerebrospinal fluid (CSF) also can deposit
several days, must not cause hair to be removed when the on indwelling sensors and so damage or confound them.
patch is removed, and must securely hold the transducer in Monitoring durations may continue for several days and
place over the monitoring period. increase the chances that blood, blood clots or proteins may
deposit or form on implanted sensors.
Invasive catheters must be stiff enough to be pushed past
The Bedside Monitor an opening in the dura mater and into parenchyma anywhere
Most if not all neurocritical care patients are connected to from a few millimeters to several centimeters, such as with a
bedside monitors (BSMs). The BSM collects signals from ventricular catheter. However, once implanted, catheters
external monitors and displays them on a single screen. Gen- should be flexible enough to impart the least amount of force
erally each parameter is connected to the BSM via detachable on the tissue if overloaded. The hair, scalp, and skull of varying
“modules” that are part of the BSM and accept analog or serial thickness are all factors to account for when using noninvasive
input from an external monitor. Each module is tailored to a monitors.
Section VII—Computers, Engineering, and the Future 387

Many primary sensor technologies have been used in neu- Electroencephalogram


rocritical care monitors. Electrical, optical, chemical, and EEG sensors are simple electrodes typically connected by a
ultrasound are some of the broad categories. single wire to one side of a differential amplifier with the other
side connected to a common reference. Arrays of up to 256
Electrical Sensors leads are common and can measure the electrical activity of
the brain through the scalp, the dura, pia mater, or directly
Strain Gauge in the cortex. EEG monitoring is a very mature technology,
Doped silicon piezoresistive strain gauge sensors are used but there have been several recent improvements including
commonly to transduce ICP. Miniaturized versions can be noise suppression and signal processing techniques. In addi-
placed in the distal tip of catheters, whereas larger versions tion, improvements in analog electronics and the advent of
are placed outside the head in fluid communication with the digital signal processing (DSP) have made EEG monitoring
ventricle or less commonly the subarachnoid space. Both full- more reliable and easier to understand for the clinician. For
and half-bridge devices, with and without temperature com- example, useful time and frequency domain transformations
pensation, are common. These devices are accurate and are now easy and inexpensive to implement with commer-
reliable and in the larger sizes low cost, because these larger cially available DSP chips and software. Because of these tech-
devices are manufactured in large quantities for use in periph- nologic advances, continuous EEG is used more frequently in
eral arterial line blood pressure monitoring. The use of the NCCUs, and its use has provided insight into physiologic
strain gauge to sense pressure has the added advantage that effects of brain injury and how seizures themselves may cause
BSMs are designed to operate them directly; this eliminates secondary injury.5
the need for complicated electronics between the sensor and
the BSM. The wire bond junctions must be protected from Optical Sensors
corrosive environments, often with low modulus adhesive
coatings. The chip must be insulated from mechanical strain Intensity Modulation
other than that imparted by what is to be measured, such as Many optical methods have been used to sense neurologic
ICP. This is a difficult design requirement, and much intel- parameters such as pressure, oxygen tension, and blood oxygen
lectual property and many trade secrets revolve around this saturation. Most indwelling optical sensors rely on a fiber-
issue (Fig. 39.5). optic connection to optical components outside the head.
Optical ICP sensors use a primary mechanical sensor and an
optical proximity detector to transduce pressure. One com-
Polarographic Electrodes monly used device makes use of a miniaturized thin-walled
Electrochemical “polarographic” electrodes are used to mea­ bellows closed at one end as the primary transducer. The
sure oxygen tension in the brain parenchyma. A metal cathode intensity of light reflected by the closed end of the bellows
and anode and electrolyte solution are contained in an oxygen- from one optical fiber to another is used to sense the bellows’
permeable plastic tube. Oxygen molecules that have diffused position. The geometry of the reflections modulates the inten-
through the probe wall react with water at the surface of the sity of the returned light signal, which in turn is proportional
cathode to produce hydroxide ions. Free electrons then are
liberated, causing current flow between the cathode and
e−
anode. This current is sensed by the monitor and is propor-
tional to the partial pressure of oxygen in the tissues in contact
with the probe. This method has proven to be accurate and Current measurement
reliable and does not require complex optical or electronic
devices for operation (Fig. 39.6). + −
700 mV polarization

1
3

5
2 4

O2

Fig. 39.6  Licox oxygen sensor that uses polarograph electrodes.


Fig. 39.5  Strain gauge pressure transducer within NeuroSensor ICP/CBF 1, Oxygen-permeable plastic tube; 2, cathode; 3, anode; 4, electrolyte
probe. ICP/CBF, Intracranial pressure/cerebral blood flow. (Used with solution; 5, tissues in contact with probe. (Used with permission of Integra
permission of Integra LifeSciences Corporation.) LifeSciences Corporation.)
388 Section VII—Computers, Engineering, and the Future

to pressure. The only optical components used are the optical laser source in the near infrared. Light is scattered by station-
fibers, a light-emitting diode (LED), and two photodiodes, all ary structures in the tissues as well as the moving red blood
of which are contained in the probe. cells. Light reflected by the moving red blood cells undergoes
a Doppler phase shift dependent on the blood cells’ velocity.
This in turn causes the spectra of the returned light to be
Interferometer Proximity Detection broadened by some 20 Hz to 20 kHz. This spectral broadening
Another optical pressure sensor device used to measure ICP is used to estimate the velocity of the moving red blood cells.
uses an interferometer to measure the depth of an optical The intensity of the returned signal is used to estimate their
cavity formed by a silicon diaphragm bonded over a cavity concentration, and the product of the two terms, commonly
etched in a small cylinder of borosilicate glass. The cavity referred to as flux, is proportional to blood flow.7 This mea-
is vented to atmosphere on its proximal side; therefore, surement technique is attractive because it is continuous,
changes in cavity depth caused by displacement of the silicon allows for the visualization of the blood flow pulse waves, and
diaphragm are proportional to ICP. This sensor has the advan- can be used in febrile patients. The technique, however, is not
tage of being formed of all glass materials, which reduces the quantitative.
effects of differential thermal expansion or water take-up
causing apparent pressure changes not due to a change in the
patient’s ICP. Thermal Diffusion
Thermal diffusion techniques are used to measure blood flow
in the parenchyma. Two thermistors are located near the distal
Fluorescence Quenching tip of a probe. Current is passed through one thermistor to
Optical sensors have been used to measure oxygen and other raise its temperature a predetermined amount above the tissue
parameters in the brain using the technique of fluorescence temperature. Heat from this thermistor is transferred by both
quenching. Typically silicone room temperature vulcanizing conduction and convection. The convective heat transfer is
(RTV) adhesive is applied to the end of an optical fiber. The due to blood flowing past the probe. Knowledge of thermal
RTV is doped with dye that fluoresces at a specific wavelength. properties of the parenchyma, the temperature rise of the
The intensity or duration of the light signal generated by the thermistor, the power required to maintain the thermistor at
fluorescence also is altered by the presence of a target molecule temperature, and other factors can be used to estimate the rate
such as oxygen. The fluorescent dye also can be immobilized of convection and therefore the blood flow near the probe tip.8
by applying it to porous glass microbeads. The rate of fluores-
cent decay when the lamp or LED used for excitation is turned Other Sensors
off is used to measure temperature in some devices. These
systems can be reasonably accurate and cost effective. Microdialysis
Microdialysis probes are used to collect samples of chemical
substances from the interstitial fluid. The catheter is composed
Spectroscopy of a small-diameter tube with a membrane at the distal end
Optical techniques also may be used to measure concentration filled with a perfusion fluid. The proximal end of the catheter
of hemoglobin and the total oxygen saturation of hemoglobin ends in a collection vile. Chemicals diffuse across the dialysis
in blood. The absorption of light at convenient wavelengths membrane into a perfusion fluid inside the catheter. The
in near-infrared (NIR) near the isobestic point for hemoglo- probe membrane can be constructed such that chemicals of
bin are measured and used to determine the absolute con­ different molecular weight can be collected. The chemical of
centrations of oxyhemoglobin, deoxyhemoglobin, and total interest is collected in a sample vial connected to the catheter.
saturation in blood. The same principle is used in noninvasive The sample is then analyzed in a bedside chemical analyzer.
cerebral oximeters to estimate regional blood oxygen satura- The list of potential substances that can be measured is long,
tion in brain tissues. The forehead is illuminated by two emit- but in the clinical environment glucose, lactate, and pyruvate
ter-receiver pairs with NIR light at two wavelengths. The are the most frequently analyzed substances. Microdialysis is
attenuation of the returned light signals is used to estimate a excellent research tool, but its use in routine clinical practice
regional blood oxygen saturation.6 These systems appear to has been limited by the intermittent measurements and need
function reasonably well in uninjured brain but can be con- to transfer samples from the patient to the analyzer (Fig. 39.7).
founded by factors such as intracranial bleeding. They also
suffer from an inability to depth resolve the return signals; this
may lead to inaccuracies due to signals from scalp blood. More Doppler
sophisticated techniques can be used to overcome many of Ultrasound Doppler techniques are used to noninvasively
these obstacles; however, these techniques are not in commer- measure the blood flow velocity in the large conductive vessels
cially available systems. Chapter 33 contains a more detailed in the circle of Willis. The transducer head, including trans-
description of these methods. ducer and receiver, is positioned where the skull is thin (tem-
poral region) and the ultrasound energy, typically using
frequencies of around 2 MHz or a multiple thereof, is focused
Laser Doppler Flowmetry on the vessel of interest. The phase shifted return signal is used
Optical methods are used to estimate blood flow in brain to calculate velocity. Although accurate blood velocity mea-
parenchyma. The well-known laser Doppler flowmetery tech- surements can be made, it is harder to use as a continuous
nique has been used in many products over the past 15 to 20 monitor because it can be difficult to maintain contact and
years. The tissue is illuminated with collimated light from a alignment of the transducer head.
Section VII—Computers, Engineering, and the Future 389

Equipment—Part 1: General Requirements for Safety and


related documents. Medical devices sold in the United States,
the European Union, and many other parts of the world must
meet this standard as tested by an independent test laboratory
such as Underwriters Laboratories (UL), Technical Inspection
Association (TUV), British Standards Institution (BSI), and
the like.

Magnetic Resonance Imaging Safety


An area of increasing concern is the safety of medical devices
in the MRI environment. Devices made of or containing metal
can translate in the scanner due to the strong magnetic field
Fig. 39.7  Microdialysis system showing microdialysis catheter, pump and
and can rotate to align with the direction of the magnetic field,
analyzer. (Courtesy CMA Microdialysis.) imparting torsional forces. In addition, lead wires or other
elongated components capable of carrying current can couple
with the radiofrequency (RF) field in the scanner causing the
device to heat, particularly at the distal tip. There are reports
Safety of patients and others sustaining injuries due to each of these
Safety is a paramount consideration in the design, manufac- safety issues. The American Society for Testing and Materials
ture, and use of monitoring devices in neurocritical care. (ASTM) International in close cooperation with the U.S. Food
Safety hazards generally relate to the probe and sensor, ancil- and Drug Administration (FDA) has developed test standards
lary components such as fixation devices, connecting cables, to measure magnetically induced displacement force, mag-
and the electronic monitor used with the sensor. Manufactur- netically induced torque, and RF-induced heating of passive
ers of medical device products use formal risk analysis proce- implants along with a guide for MRI safety labeling.9-11 Of the
dures to identify hazards and mitigation methods and to three test methods RF heating is by far the most difficult and
determine whether the residual risks after mitigation are complex. The understanding of factors that influence RF
acceptable. The list of potential safety concerns is exhaustive; heating in the MRI environment is rapidly advancing and as
the more important issues posed by neurocritical care moni- such, testing requirements are in flux.12 As of this writing the
toring devices are discussed in the following text. revision of the standard for measurement of RF-induced
heating on or near passive implants is ASTM 2182 11a. In
addition, a joint working group of the IEC and ISO has devel-
Probe and Ancillary Components oped a test standard (TS) for MRI safety testing of active
The shape of the distal tip of implanted probes should be implantable medical devices (AIMDs) that is a precursor for
given careful consideration. An ogive or bullet-nose shape is an International Standards document. RF heating is influ-
probably the best and safest design. A completely blunt shape enced by the strength of the electromagnetic fields present in
with a sharp transition between the axial tip and the outer the scanner. These fields vary spatially depending on such
diameter of the catheter tip is probably the worst because it factors as location within the RF coil and interactions between
tends to tear tissues during implantation. The behavior under the electromagnetic fields and the device and patient. Current
unanticipated loading of components that are implanted in best practice appears to be to measure the heating of the
the patient, the connection to components implanted in the device in a phantom within an RF coil or MRI system while
patient, or which secure implanted parts should be considered controlling or measuring the electromagnetic fields to obtain
to limit transmission of these loads to the patient as much as a baseline understanding of the response of the device to a
possible. The methods of sealing implanted probes and fixa- well-understood set of conditions. This information is then
tion devices against leaks must be given careful thought to used in conjunction with computer modeling of the patient,
minimize the chances of infection. Materials that contact the device, and the scanner to predict heating in actual use.
patient must be chosen to minimize irritation and must be Regardless of the form the test method ultimately takes, it is
tested for biocompatibility, for example, per International important for devices to be tested for MRI safety and for clini-
Standards Organization (ISO)10993 Biological Evaluation of cians to closely follow the MRI safety instructions provided
Medical Devices. by manufacturers (Figs. 39.8 and 39.9).

Electronic Components The Regulatory Environment


Electronic monitors and cables pose their own set of safety No discussion of engineering aspects of medical devices would
concerns. Malfunctioning electrical devices can display erro- be complete without mention of the regulations that govern
neous data, cause burns and electrical shock, and interfere the design, manufacture, and distribution of these devices.
with the proper functioning of other electronic devices. Elec- Like the practice of medicine the business of medical devices
trical safety issues such as patient electrical isolation, leakage is highly regulated. In the United States the primary governing
currents, radiated emissions, susceptibility to radiated emis- laws are contained in the Code of Federal Regulations titled
sions, flammability, and a host of other potential safety issues Quality System Regulation.13 This set of regulations is enforced
are covered in the electrical safety standard International by the FDA. The Council of European Communities Medical
Electrotechnical Commission (IEC) 60601 Medical Electrical Device Directives of 1993 that was last amended in 200714
390 Section VII—Computers, Engineering, and the Future

Electric Field with ASTM Phantom in RF Coil regulatory standards: (1) 510(k) that requires the device be
200
similar to an already marketed device (i.e., it should be low
–0.4 risk), or (2) premarket approval (PMA) that requires clinical
testing and inspection. However, device classification errors
can be associated with patient safety and recalls15 and so the
–0.2
FDA asked the Institute of Medicine to review the classifica-
150 tion and review process in 2011; these recommendations are
pending.
0
Business Considerations
The majority of medical devices are sold by for-profit compa-
Z (m)

100
0.2 nies; therefore the decision to develop and market a medical
device is in large part an economic one. Most companies apply
a disciplined and analytical approach that uses one or more
forms of return on investment analysis. The cost of develop-
0.4
50 ing the product and the costs of making and selling it are
weighed against future revenue streams from sales of the
product. The time value of money is always a factor in these
0.6 calculations. One easy-to-understand approach is to deter-
mine the net present value of future revenues less all expenses.
0 This estimate can be compared with competing projects or
–0.2 0 0.2 simply the interest earned by placing the same amount of
X (m)
money in a savings account. It is important for engineers and
clinicians to understand the basic economic principals that
Fig. 39.8  Finite-difference time domain (FDTD) plot of electrical fields in govern how companies decide what products to develop and
and around American Society for Testing and Materials (ASTM) phantom when to develop them. Obviously products that address a
within magnetic resonance imaging radiofrequency (MRI RF) coil. (Used large market, are not costly to produce, command a high
with permission of Integra LifeSciences Corporation 2008.)
price, and are inexpensive and quick to develop are favored.

References
1. Gullaume J, Janny P. Continuous intracranial manometry; physiopathologic
and clinical significance of the method. Presse Med 1951;59(45):953–5.
2. Lundberg N. Continuous recording and control of ventricular fluid pressure
in neurosurgical practice. Acta Psych Neurol Scand 1960;36(Suppl. 149):
1–193.
3. Magistretti PJ, Pellerin L, Martin J. Brain energy metabolism. From the
American College of Neuropsychopharmacology (ACNP) website
publication Psychopharmacology—The Forth Generation of Progress, 2000.
http://www.acnp.org/g4/GN401000064/Default.htm
4. Duke T. Dysoxia and lactate. Arch Dis Child 1999;81:343–50.
5. Vespa PM, Miller C, McArthur D, et al. Nonconvulsive electrographic
seizures after traumatic brain injury result in a delayed, prolonged increase
in intracranial pressure and metabolic crisis. Crit Care Med 2007;35(12):
2830–6.
6. Chance B, Cope M, Gratton E, et al. Phase measurement of light absorption
and scatter in human tissue. Am Inst Physics 1998;69(10):3457–81.
7. Shepherd AP, Oberg PA. Laser-Doppler blood flowmetry. Boston: Kluwer
Academic; 1990.
8. Martin GT, Bowman HF. Validation of real-time continuous perfusion
Fig. 39.9  American Society for Testing and Materials (ASTM) phantom measurement. Med Bio Eng Comput 2000;38(3):319–26.
in 1.5T magnetic resonance imaging (MRI) scanner during device MRI 9. ASTM F 2052–00, Standard test method for measurement of magnetically
safety test. (Used with permission of Integra LifeSciences Corporation 2008.) induced displacement force on passive implants in the magnetic resonance
environment.
10. ASTM F 2213-04, Standard test method for measurement of magnetically
induced torque on medical devices in the magnetic resonance
environment.
governs the products made or sold in the European Union 11. ASTM F 2182-02, Standard test method for measurement of radio
frequency induced heating near passive implants during magnetic
(EU) and the manufacturers thereof. It is critical for engineers resonance imaging.
to understand these and many other regulations as they 12. Kainz W. MR heating tests of MR critical implants. J Magn Reson Imaging
impact most technical decisions. Furthermore, it is important 2007;26:1182–85.
to consider these regulatory issues early in design and devel- 13. 21 CFR Part 820—Quality system regulation, Revisions as of 31 March,
opment to ensure that any new idea is feasible in the clinical 2006.
14. Council Directive 93/42/EEC of 14 June 1993 concerning medical devices,
environment before expending time and labor. At present the Amended 11/10/2007.
FDA divides medical devices into three classes according 15. Zuckerman DM, Brown P, Nissen SE. Medical device recalls and the FDA
to their level of risk to a patient. There are two alternative approval process. Arch Intern Med 2011;171(11):1006–11. Epub 2011 Feb 14.
Chapter
40  
VII

Multimodality Monitoring
and Artificial Intelligence
Richard S. Moberg and J. Michael Schmidt

physiologic data can be lost if there is no means to store or


Introduction archive it. In most NCCUs today, physicians and nurses can
Multimodality monitoring can be defined as the simultaneous only view continuous physiologic data by looking at the
collection of data from multiple diverse sources pertaining to bedside monitor. This information, once it has scrolled across
a single patient coupled with the ability to view the data in an the screen, is lost, and if the staff is not at the bedside only
integrated and time-synchronized manner. Alternatively, mul- intermittently recorded data are available; that is, data collec-
timodality monitoring may be considered the use of more tion and storage is essential to NCCU informatics. Second, in
than one complementary method to monitor a single organ, the severely brain-injured patient, use of the clinical examina-
when no one single method can provide complete informa- tion to detect changes in patient state is severely limited. Thus
tion. It is the first step to create a bedside environment where physicians and nurses rely in large part on changes in physi-
advanced knowledge tools can be applied to the data to aid ologic metrics and on results from diagnostic tests to make
the clinician in patient management. However, few, if any, patient management decisions. During rounds of critically ill
neurocritical care units (NCCUs) have such an environment patients each morning, a physician may be confronted with
due to a variety of barriers (see Chapter 4). Assuming these more than 200 variables.6 The high data volume and the time-
problems will be solved, this chapter reviews the creation of a critical situations physicians are confronted with create a
“knowledge infrastructure” in neurocritical care. The ratio- problem.7 This is accentuated because human beings are not
nale and benefits are presented along with a historical perspec- able to judge the degree of relatedness between more than two
tive, current progress, and future prospects. variables. Furthermore, constant information overload is one
contributing cause for preventable medical errors.8 A knowl-
edge infrastructure can provide a closer coupling between the
Rationale clinician and the data with the promise to decrease errors and
An accepted goal of neuromonitoring in intensive care is to improve outcomes. Finally, as the aging population fills the
enable the detection of harmful physiologic events before they health care system, it is predicted there will be a significant
cause irreversible damage to the brain.1 Ideally monitoring shortage of caregivers, particularly in the ICU.9 Because of
should be used to predict “events” in the intensive care unit higher efficiencies, workflow software driven by multimodal
(ICU).2 However, the brain is a complex system with inter- patient data has the potential for minimizing the number of
related hemodynamic, metabolic, and electrical subsystems. staff needed to accomplish the same amount of work.
From simply a logical perspective, the most effective monitor- The current federal (and international) emphasis on
ing paradigm would be one that monitors multiple sites in this medical economics provides further justification for multi-
multifaceted system. But to date, no formal studies have been modal neuromonitoring. The rising costs of health care in the
conducted that pair multimodal monitoring with a treatment United States has led the government to conclude that better
protocol to demonstrate better outcome, although one such information in a variety of areas could eventually alter the way
trial, the Brain Oxygen and Outcome Study in Traumatic in which medicine is practiced and yield lower health care
Brain Injury (BOOST), a phase 2 trial to examine physiologic spending without having adverse effects on health.10 Other
efficacy of multimodal monitoring, is likely to complete countries have come to similar conclusions, and such thinking
enrollment in 2013.3 For the moment, however, a common- has prompted a rise in comparative effectiveness research that
sense rationale must suffice. evaluates the impact (primarily costs and benefits) of different
The need for a knowledge-rich infrastructure made possible treatment options.11 This emphasis on informatics under-
by multimodal data extends beyond the NCCU to all of high- scores the need for a more comprehensive and coordinated
acuity critical care. First, in intensive care, clinical information approach to data collection, something that multimodal mon-
systems acquire and store physiologic variables and device itoring can provide.
parameters online at least every minute.4 Physiologic signals There also are benefits to patient safety in creating a knowl-
may contain useful data in frequencies of ~0.2 kHz. These data edge infrastructure. There are a few reports of accidental pa­
can be lost when data less than 0.5 to 1 kHz is sampled over tient deaths that could have been prevented if medical devices
a short time (1 to 2 ms).5 In addition, high-resolution were able to intercommunicate. In one example, a ventilator
© Copyright 2013 Elsevier Inc. All rights reserved. 391
392 Section VII—Computers, Engineering, and the Future

was momentarily turned off to eliminate blurring of x-ray with 16 or more channels. Assuming a 256-Hz sampling
images that were being obtained. The film plate jammed rate, 16 channels, and 16-bit precision, storage of 1 day of
under the table and the anesthesiologist went to help. In so EEG requires more than 700 megabytes of space. Adding
doing, the anesthesiologist forgot to restart the ventilator and simultaneous video of the patient can bring the storage rate
the patient died.12 Had there been a method for the x-ray from 250 megabytes to more than 1 gigabyte per hour,
machine and the ventilator to communicate, the patient death depending on the frame rate and type of video compression
may have been prevented. used. Consequently, most institutions do not store all the
Neurocritical care has evolved as a distinct speciality dur­ video data but rather just the segments of interest, a process
ing the past decade. In large part patient management that requires manual intervention. Similarly manual inter-
depends on understanding patient physiology, so techniques vention may be necessary to exclude potential artifacts
to monitor the brain also have evolved. Increasingly, NCCUs or during periods when patients are disconnected from a
use more than one monitor of brain function, and an increas- monitor.
ing number of publications and reviews allude to the poten-
tial benefits of multimodal monitoring in various aspects of
neurocritical care, including traumatic brain injury (TBI),13- Comprehensive Monitoring
15,16
subarachnoid hemorrhage,17 management of cerebral Multimodal neuromonitoring must be comprehensive in that
hemodynamics,18-20 and determination of brain death,21 and all of the necessary data for particular monitoring goals
in understanding patient physiology in general.22-25,24,26-30 Fur- need to be collected simultaneously, time synchronized, and
thermore when multimodality monitoring is used, it has displayed in an integrated fashion. This is a challenge in
become apparent that single monitors may miss episodes of the NCCU because most monitors stand alone and are self-
cerebral compromise.31 contained, provided by a commercial vendor. However, this
challenge is likely to diminish since the American Society for
Testing and Materials adopted the concept of an “integrated
Requirements of Multimodal clinical environment” in 2009.35 Time synchronization of data
received from multiple devices in a variety of formats presents
Neuromonitoring Systems a significant problem to developers of multimodal data col-
Recording multiple simultaneous measurements from a lection systems. The data sampling rates may be widely differ-
variety of diverse sources brings about problems not ent and the data may be sent to the collection device
encountered in conventional monitoring. These problems continuously or in packets with varying time intervals. The
can be broadly classified as dealing with the following accuracy of the time synchronization task, however, depends
requirements of monitoring: continuous, comprehensive, on the use of the data. For slow varying trends where only a
and communicative. visual correlation is to be made, a high degree of synchroniza-
tion may not be required.
There are some applications where very accurate time syn-
Continuous Monitoring chronization of data is required. An example is combining
Continuous monitoring is required in critical care to not miss data from different sources to improve the quality of data. For
clinically significant events. More specifically, the frequency of example, EEG data contaminated with electrocardiogram
monitoring needs to be higher than the duration of the events (ECG) or pulse artifact can falsely trigger automated seizure
to be detected. Hemphill et al.32 compared medical records detection algorithms. Other physiologic signals can be used to
(with approximately hourly entries) to automatic continuous help remove or reduce such artifacts in the EEG using various
data recordings (approximately 1-second intervals) recorded analysis methods such as adaptive filtering36 or independent
simultaneously in 16 patients and found that the medical component analysis.37 For example, ECG can be used to help
record data tended to underestimate the number of secondary remove pulse artifact and a respiration signal can be used to
brain insults detected. They concluded that higher frequency aid in removing high-frequency ventilator artifact from the
data (i.e., higher than is normally acquired in the medical EEG. Figure 40.1 shows EEG data (left panel) contaminated
record) may be necessary for more precise evaluation of sec- with pulse artifact. Using simultaneously collected ECG, the
ondary brain injury in neurocritical care. A notable example EEG can be adaptively filtered to remove the artifact (right
of the value of continuous monitoring is with electroencepha- panel). The notation on the first channel of each panel shows
logram (EEG) recordings. In 1993, Jordan monitored EEG the difference in an amplitude envelope function caused by
continuously rather than intermittently and showed the the artifact (8.2 µV p-p with the artifact versus 5.4 µV p-p
number patients with seizures and the frequency of seizures without).
present in critical care patients was much greater than what
was conventionally thought.33 He also showed that continuous
monitoring of EEG made an impact on clinical decision Communicative Monitoring
making in 51% of patients (n = 124). Subsequently, Claassen Once collected, the information in multiple data streams must
and Mayer34 showed that continuous EEG influenced patient be extracted and communicated to the clinician. Perhaps the
management on 50% of monitored days. simplest and most effective way to extract information is to
The problem inherent in continuous monitoring is the observe relationships in the data by plotting time-synchronized
amount of data collected when monitoring from multiple trends on a single display. Such displays are standard on con-
sources. Storage of trend data and a few waveforms is insig- ventional vital-signs monitors because they facilitate the visual
nificant with current storage capacities. However, EEG is detection of correlations between measurements. Figure 40.2
generally recorded with a higher sampling rate and usually shows an example that plots trends of ICP, arterial blood
Section VII—Computers, Engineering, and the Future 393

EEG with ECG Artifact (shown at 50uV p-p) EEG after Adaptive Filtering with ECG Channel (shown at 50uV p-p)

T3-C3 T3-C3

90% PDF amp = 8.2 uV p-p 90% PDF amp = 5.4 uV p-p

C3-Cz C3-Cz

Cz-C4 Cz-C4

C4-T4 C4-T4

ECG ECG

180 181 182 183 184 185 186 187 188 189 190 180 181 182 183 184 185 186 187 188 189 190
Time (sec) Time (sec)

Fig. 40.1  Electroencephalographic data contaminated with electrocardiogram (ECG) artifacts (left panel) and after adaptive filtering with the ECG
(right panel) to remove the artifact.

50
ICP 40
[mm Hg] 30
20
10
200

ABP 150
[mm Hg] 100
50
200
150
FV 100
[cm/s] 50
0
90
85
TOI 80
[%] 75
70
22
20
PbtO2 18
[mm Hg] 16
22:10 22:15 22:20 22:25 22:30 22:35 22:40 22:45 22:50 22:55 23:00 23:05
Time [h:m]
Fig. 40.2  Simultaneous trends of 1 hour monitoring of intracranial pressure (ICP), arterial blood pressure (ABP), flow velocity (FV), tissue
oxygenation index (TOI), and brain tissue oxygenation (PbtO2). This provides a synchronized view of physiologic data. (Courtesy Dr. Marek Czosnyka.)

pressure (ABP), flow velocity, tissue oxygenation index (TOI), neurocritical care. These plots can be calculated with most
and brain tissue oxygenation. Episodes of ICP increases are spreadsheet software using data exported from a neuromoni-
associated with hyperemia due to unstable arterial pressure. toring data collection system or with more specialized statis-
Figure 40.3 shows a more comprehensive visual display, a tics software. These capabilities also exist in some of the
neurocritical care dashboard, which includes data represented neuromonitoring systems described in the next section.
by waveforms, images, and other formats in addition to trends. In summary, multimodality monitoring creates new prob-
Going further, simple statistical analyses on the recorded lems with data collection, but it is anticipated that most will
data can be revealing. For example, Figure 40.4 shows be solved with near-term technology. For data analysis, there
the change in brain oxygen versus cerebral perfusion pressure are many ways to process multiparametric data to extract and
at two points in time in the same patient using scatter display information useful to the clinician. This is an active
plots. It illustrates the dynamic nature of autoregulation in area of critical care research.
394 Section VII—Computers, Engineering, and the Future

Fig. 40.3  A neurocritical care dashboard that includes information from multiple domains. (Courtesy Dr. Val Nenov.)

Cerebral Perfusion Pressure by Brain Oxygen Cerebral Perfusion Pressure by Brain Oxygen
Patient 105, day 1 11:00 am − 1:00 pm Patient 105, day 1 11:00 am − 1:00 pm
r2 = 0.6199 r2 = 0.0.2586
34 34

32
32
30

28 30
Brain oxygen

Brain oxygen

26
28
24

22 26

20
24
18

16 22
45 50 55 60 65 70 75 80 85 90 95 100 54 56 58 60 62 64 66 68 70 72 74 76 78 80
Cerebral perfusion pressure Cerebral perfusion pressure
Fig. 40.4  Scatter plots of brain oxygen vs. cerebral perfusion pressure on subsequent days made using the Statistica Data Mining software.
(Courtesy Dr. Micheal Schmidt.)

Creating a Bedside Knowledge John McCarthy in 1956.38 It is a field that attempts to trans-
Environment form data into reasoning and uses a variety of tools such as
artificial neural networks (ANN) and statistical classifiers.
The Role of Artificial Intelligence Neural networks are composed of “neuron-like” software ele-
Once continuous multimodal data can be recorded, it will ments that, in the aggregate, can exhibit simple learning
be possible to create a bedside knowledge environment that behaviors. They have a particular use in the analysis of neuro-
couples decision support to patient management. These critical care data in that they can be trained to find patterns
methods are largely in the domain of artificial intelligence in data both in a generalized way and within a single patient.
(AI). AI is defined as the science and engineering of making Although the potential of AI is significant, publications
intelligent machines and was coined by computer scientist about using AI to analyze data specifically in neurocritical care
Section VII—Computers, Engineering, and the Future 395

are sparse, most likely due to the challenges to collecting mul- relationships to intracranial pressure and cerebral perfusion
tiparametric data. Some of the work is reviewed here and then pressure. Minhas et al.51 used decision tree analysis to iden-
the promise AI has to offer in creating a knowledge environ- tify which patients would be colonized with antibiotic-
ment at the bedside is discussed. A number of references can resistant bacteria on admission to the NCCU. Several groups
provide a more in-depth explanation of AI concepts.39,40 have studied the prognostic and predictive abilities of AI
An active area of AI research in critical care is to attempt to techniques. For example, Swiercz et al.52 used neural
improve the accuracy of alarms and the detection of events. network techniques with a wavelet algorithm incorporated
Most clinicians are familiar with the problem of alarms in a to predict ICP values, whereas Nikiforidis et al.53 constructed
multimonitor environment, such as critical care or surgery. a Bayesian belief network (BBN) to predict short-term prog-
Imhoff 41 recently reviewed the literature and showed that up nosis of TBI patients in the intensive care unit, and Väth
to 90% of all alarms in critical care monitoring are false et al. used ANN to analyze the accuracy of outcome predic-
alarms, which leads to a dangerous desensitization of the staff tion after TBI.54
toward true alarms.42 Several studies have shown encouraging Discriminate function analysis is used to determine which
results with the application of AI techniques to alarm manage- combination of variables discriminate between two or more
ment,43-45 but these techniques have not made their way to naturally occurring states. The resulting discriminate function
routine clinical use, most likely because of liability concerns can be used in monitoring to alert the clinician to a change in
of medical device vendors. the state of the patient. The bispectral index (BIS) is an
AI techniques can be used to assess the context of the example that integrates several disparate descriptors of the
patient, taking into consideration the variability among EEG55 and has gained wide use in surgery to assess anesthetic
patients, to provide more individualized monitoring. For effects on the brain and has been used as a measure of sedation
example, Apiletti and colleagues used real-time classification in critical care (see Chapters 9 and 25).
techniques to detect different severity levels of a monitored Another AI approach is used when “human expertise” is
patient’s clinical situation,46 whereas Zhang and Szolovits, needed to solve problems. Human expertise is formalized as a
using neural networks, showed patient-specific modeling in knowledge base, a series of rules, or other means (or a com-
real time to be feasible and effective to generate bedside bination of these) so the computer can use this information
alerts.47 An analysis of the dynamics of the critical care work to process the data. Expert systems, as these are called, often
environment as a complex cognitive system was performed by are used to augment statistical analyses when they are not
Patel et al.; this work has implications for the design of deci- robust enough to provide the desired results. Figure 40.5
sion support tools.48 shows the results of an automated seizure detection algorithm
Specific to neurocritical care, Nelson et al.49,50 analyzed developed using an expert systems approach. The first pass of
patterns of cerebral microdialysis in patients with TBI and, the analysis calculates metrics for a variety of features such as
with a neural network methodology, investigated pattern rhythmicity and amplitude. The second pass, using expert

Fig. 40.5  A display of automated seizure detection (red colored trace) that uses a combination of statistical techniques and an expert system approach
(in a neonatal electroencephalogram).
396 Section VII—Computers, Engineering, and the Future

systems tools, adds the knowledge of an expert epileptologist integrated display of signals in a patient in the NCCU whose
and follows a rule-based approach to evaluate the metrics for condition deteriorated to brain death.
conditions such as their stability or fluctuation over time and Starting in the early 1990s, researchers in the Department
symmetry of activity in adjacent channels. This combination of Neurosurgery at the University of California Los Angeles
of techniques significantly increases the accuracy of the seizure (UCLA) began collecting multichannel EEG signals continu-
detection and allows a more precise determination of seizure ously on every patient in neurocritical care. EEG is one of the
start and stop on a per-channel basis. more challenging signals to collect continuously and to review
because of the volumes of data collected. The UCLA group
therefore developed the percent alpha variability (PAV), which
Historical Notes and Review condensed certain features of the EEG into a variable that
could be trended along with other physiologic parameters in
of Existing Systems a multimodal fashion.59 The PAV is associated with outcome
There are very few commercial systems that integrate neuro- in TBI and among other diagnoses provides insight about
logic data. Consequently a few university-based NCCUs have ischemia.60,61 The data collection capabilities of the UCLA
developed their own systems. These various systems and range system also include other physiologic parameters, patient
of capabilities are reviewed briefly. demographics, and images. This integrated “dashboard” is
illustrated in Figure 40.3.
Investigators interested in ICP in the NCCU at Adden-
University-Developed Systems brooke’s Hospital in Cambridge, England, developed a system
Some of the earliest publications in multimodality neuro- that collects data from multiple sources so it could be corre-
monitoring came from the University of Graz, Austria, in the lated to ICP.22 This multimodality monitoring system is based
late 1980s.56,57 Beginning mostly with the simultaneous collec- on a standard personal computer equipped with a digital-to-
tion and integration of electrophysiologic signals (EEG and analog converter and serial data interfaces, and has evolved
evoked potentials), the researchers have added a full com­ from the1980s into a system, ICM+, for multimodal NCCU
plement of physiologic signals in their data collection system monitoring and ICP waveform analysis.22,62
and have used the system to investigate a variety of procedures At San Francisco General Hospital, the University of Cali-
in surgery and critical care.58 Figure 40.6 illustrates the fornia San Francisco (UCSF) Brain and Spinal Injury Center

EEG BAEP I-V IPL SSEP CCT HR HRV ICP BP Temp.


Zervikal Kortikal Sys Dis
16:51

<2%
15:40

N13 Hirntod
15:15

>2%

14:10

12:29

8:15

N20
6:50

ICP
0.5
0.1 µV 0.5
µV µV 6:20
0 24 0 24 0 10 3 6 5 20 15 30 3 12 20 120 0 20 0 50 60 200 60 140 36 39
Hz Hz msec msec msec msec msec 1/min % mm Hg °C

Fig. 40.6  Multimodal data recording from a 57-year-old woman with an intracerebral hemorrhage in the left hemisphere that shows a
progression to brain death following an intracranial pressure (ICP) increase. The recorded modalities from left to right are electroencephalo
graph (EEG) compressed spectral array (two channels), brainstem auditory evoked potentials (BAEP), the interpeak latency (IPL) of peak I to IV of the
BAEP, somatosensory evoked potentials (SSEP) with the N14 and N20 marked and the central conduction time (CCT) calculated, heart rate (HR), heart
rate variability (HRV), ICP, blood pressure ([BP], systolic and diastolic), and temperature. Zervical and Kortikal refer to the cervical and cortical responses
from the SSEP. Hirntod indicates brain death. (Courtesy Dr. Gerhard Litscher.)
Section VII—Computers, Engineering, and the Future 397

Fig. 40.7  ICM+ software developed at the Cambridge Neurosurgical Unit.

has an integrated data repository that attempts to link bedside


critical care physiology, critical care charting, and the hospital
patient electronic medical records. These represent three dis-
tinct systems: the locally developed Aristein system (bedside
continuous physiology data collected at least once each
minute from each ICU bed), Philips’ Carevue critical care
bedside nurse charting system, and Siemens’ Invision elec-
tronic medical record. Selected data from each system is
brought together using a distinct patient identifier into a
common server that is located behind the hospital firewall.
These data are then coded daily and Health Insurance Porta-
bility and Accountability Act identifiers removed before the
data are passed outside the firewall into a metadata ware-
house that can be used for advanced informatics analysis.
The ultimate goal is to make this metadata warehouse avail-
able for pooling of coded anonymous data from multiple
institutions.63

Commercial Systems
Several of the available commercial multimodal neuromoni-
toring systems originated from those developed in university
and research settings. Global Care Quest (Aliso Viejo, CA)
was formed to commercialize the data integration work at
UCLA (see Fig. 40.3). ICM+, a Windows program for multi-
modal neuromonitoring, grew out of the system used at
Cambridge University (Fig. 40.7). The author’s [R.M.] early
work in data integration (Fig. 40.8) led to a research system Fig. 40.8  An early multimodality neuromonitoring system deve­
(CNS Technology, Ambler, PA) used for data collection in loped by the author [RM] in 1996. It collected data from a vital signs
multicenter clinical trials funded by the National Institutes of monitor, transcranial Doppler, brain oxygen (RunMan from Dr. Britton
Health. It is now a commercial multimodal data collection Chance), laser Doppler flowmetry, ScVO2, and EEG. The system was used
and display system called the Component Neuromonitoring to investigate software architectures for multimodal data collection and
System (CNS) Monitor. in its current iteration is a single device operated through a touch screen.
398 Section VII—Computers, Engineering, and the Future

Fig. 40.9  ICUpilot screen shot.

Several companies that manufacture a monitor for a spe- At the NCCU at Columbia University College of Physicians
cific modality have developed research systems that record and Surgeons, efforts have been made to get multiple com-
additional data, and some of these systems are commercially mercial products to work together. Capsule Technologies
available. GMS (Kiel-Mielkendorf, Germany), which initially (Paris, France) is one of several companies that provide
developed the Licox brain tissue oxygen monitor, developed general purpose data integrating systems. Capsule Technolo-
a product called the MMM (Multi Modality Monitor) that gies provide a wide variety of device interfaces and hardware
collected data from a variety of related monitors. GMS to collect trends of numerical data on a central server.
became part of Integra (Plainsboro, NJ) that has the Although targeted at workflow and patient record applica-
Moebius system for collection of multimodality data. CMA tions, the technology has been used in the Columbia
Microdialysis (Solna, Sweden), the clinical arm of which is University NCCU as one component of their multimodal
now known as Dipylon Medical, developed the ICUpilot neuromonitoring system. Excel Medical Electronics (Jupiter,
software that is widely used for analysis of data. The soft- FL) produces the BedMaster software, which, like the Capsule
ware collects data from a variety of neuromonitoring devices Technologies product, integrates data from multiple sources
and other physiologic monitors, although it is integrated and provides an integrated display of that data. An advantage
into the CMA analyzer, and stores the data in an SQL data- of the BedMaster software is that it is able to emulate a
base. The user then can review the data in a variety of monitor display and it allows remote access to patient data
formats and process the data with built-in statistical tools information through a Citrix server (Fig. 40.11). To visualize
(Fig. 40.9). multimodality data, ICUpilot is used to query the BedMaster
Manufacturers of continuous EEG monitors have added the database. A separate system is used to collect EEG data from
capability to record and trend other parameters along with the each bedside.
EEG trends. Most notably, the Axon Systems (Hauppauge, In summary, current products or combinations of products
NY) Eclipse Workstation, although focused on intraoperative can provide multimodal data collection. Each system has
monitoring, can record EEG, evoked potentials, physiologic potential advantages and disadvantages but in large part each
trends, transcranial Doppler, and a variety of other signals is a means to display multiple data sources and at best inte-
(Fig. 40.10). grate them. However the ideal information environment in
Capabilities of vital sign monitors are constantly expand- neurocritical care has still to be developed and in particular
ing to incorporate new measurements and most can trend interpretative systems or decision support does not yet exist
ICP, cerebral perfusion pressure, brain oxygen, and a limited in the NCCU. This future is the focus of the next section.
number of EEG channels. The expectation is that multimodal
neuromonitoring will be integrated into conventional vital
signs monitoring in the future. But, at present, the comput-
The Future
ing power in most vital sign monitors is not adequate to While the future of neuromonitoring may rely on nanotech-
provide the advanced processing (e.g., multichannel spectral nology or molecular solutions,64 monitoring devices are here
analysis of EEG, seizure detection) that is required in neuro- to stay for the foreseeable future. There are several trends that
critical care. will influence the course of multimodal monitoring.
Section VII—Computers, Engineering, and the Future 399

Fig. 40.10  Display from Axon Systems


Eclipse Neurological Workstation.

Fig. 40.11  Display from the BedMaster


XA software.

Sensors drainage catheter is an exciting concept (Fig. 40.12).65,66 Pres-


Sensors, both invasive and noninvasive, are moving toward sure, temperature, oxygen, and glucose microsensors to
multimodal data collection to reduce the “real estate” on and measure these parameters in real time are rolled in a spiral and
in the head. For example, collecting tissue oxygen, ICP, and attached to the catheter.
temperature is now possible in the same catheter (Raumedic, Many sensors may truly be noninvasive in the future;
Münchberg, Germany). The recent announcement of a “lab- this likely will increase the number of patients who are moni-
on-a-tube” incorporated on the outside of a ventricular tored for ICP and even cerebrovascular reactivity.67-69 It is
400 Section VII—Computers, Engineering, and the Future

conceivable that a noninvasive sensor may combine EEG, introduced as a relative measure of trends of brain oxygen,
evoked potentials, near-infrared spectroscopy, and transcra- and now systems are available that provide absolute readings
nial Doppler (TCD) in an easy-to-apply headband. An inter- (see Chapter 33). Recent work shows that NIRS can be used
esting helmet for continuous monitoring of TCD, EEG, to track the kinetics of the tracer dye indocyanine green to
somatosensory evoked potentials, brainstem auditory evoked calculate an index of cerebral blood flow.71 Similarly, diffuse
potentials, and brain oxygen (via near-infrared spectroscopy correlation spectroscopy (DCS) uses near-infrared light to
[NIRS]) was constructed by Dr. Gerhard Litscher in Graz, assess changes in cerebral blood flow, but does not require a
Austria, in 1998.70 Although somewhat unwieldy for routine tracer. Although it is a relatively new modality, blood flow
use, the system demonstrated the feasibility of this concept calculations using DCS have been validated both in animal
and the ability to set up and start collecting these parameters and human studies,72-74 which show it to be a promising non-
in as little as 10 minutes. invasive measurement.

Monitoring Modalities Unified Information Architecture


New monitoring modalities or improved existing methods Neurocritical care monitoring, as with much of critical care
will enter routine use. For example, NIRS was originally medicine, will benefit greatly from a unified information
architecture. The architecture would consist of a bedside envi-
ronment where a seamless data pathway exists from the
patient, to processing methods, to displays, to the patient
record, to workflow software and decision support methods,
to a data warehouse for later use. The architecture would
support medical devices that would plug in and be automati-
cally recognized, as well as software-based knowledge modules
that would “plug in” to provide new calculations, new displays,
practice guidelines, decision support methods, and other
bedside-usable information. The system would understand
the context of the patient and customize the monitoring to
specific problems.
Although somewhat futuristic, work is currently underway
to design components of such an architecture, and to develop
standards and address regulatory issues. Figure 40.13 illus-
trates the concept and is based on the Integrated Clinical
Environment (ICE).75,76 ICE is a proposed standard for infor-
mation flow at the bedside that includes medical device “plug-
Fig. 40.12  The Lab-on-a-Tube consisting of a spiral-wound strip of and-play,” the creation of a patient-centric database, and the
sensors on a ventricular drainage catheter. (Printed with permission from the use of the data by knowledge tools such as workflows. The
Mayfield Clinic.) concept is being developed by a consortium of industry

Data Knowledge
manager manager

Patient record and


remote review Patient
context
Bedside
Guideline
engine
P
External database
Decision G G
Standard protocol support
Knowledge
Adapters plug-ins
Events
manager D D

Patient-specific
Monitors database Learning D L
manager

Fig. 40.13  A unified information architecture for neurocritical care.


Section VII—Computers, Engineering, and the Future 401

members, academic organizations, professional societies, and this field. The recent efforts to solve much broader problems
regulatory agencies and is moving through the standards in hospital informatics will help in the NCCU. Second, indus-
development process.75 try is starting to focus on automatic data collection, including
In ICE, data are collected from devices that use a standard a host of smaller companies that will challenge the single-
protocol to ensure consistency in how data are received, vendor monopolies and their proprietary data pathways.
labeled, and stored. Because there is a wide variety of medical Third, a renewed effort to achieve a device communications
device communications protocols and data naming conven- standard coupled to a bedside information environment has
tions, adapters are proposed that transport the medical device made significant progress. Fourth the ASTM has adopted the
data via a standard protocol that provides information about concept of an integrated clinical environment.35 Finally,
the device as well as the data from the device. In this manner, various professional societies have begun to document the
the receiving software does not have to know about every new increase in patient safety and worker efficiencies that device
device that is developed. interoperability and intelligent software can yield. Together
A key benefit of the information architecture is the ability these factors mean that working in a knowledge environment
to uncouple the knowledge components from basic data col- at the bedside is within sight.
lection devices and the ability to distribute them as separate
“knowledge plug-ins.” At present, at least in the United States,
a medical device manufacturer must “re-clear” a device with
Disclaimer
the Food and Drug Administration (FDA) if a new metric is The author (R.M.) is a founder of one of the companies (CNS
added to it that provides an additional use (e.g., a measure of Technology) whose product is mentioned in this chapter.
autoregulation or a seizure detection method). Device manu-
facturers often are not willing to spend the time or money to
go through this FDA process. This hurdle means that many References
innovative techniques for data analysis and decision support
1. Gupta AK. Monitoring the injured brain in the intensive care unit.
remain on the shelves of the inventor. Uncoupling these J Postgrad Med 2002;48:218–25.
knowledge components from the device manufacturer would 2. Cohen MJ, Grossman AD, Morabito D. Identification of complex metabolic
then shift the safety and quality requirements to the develop- states in critically injured patients using bioinformatic cluster analysis. Crit
ers of the components, and a process to certify them would Care 2010;14:R10.
need to be developed. 3. Sahuquillo J. Does multimodality monitoring make a difference in
neurocritical care? Eur J Anaesthesiol 2008;42(Suppl):83–6.
4. Imhoff M, Fried R, Gather U, et al. Dimension reduction for physiological
variables using graphical modeling. AMIA Annu Symp Proc 2003:313–17.
Annotated Database 5. Burykin A, Peck T, Krejci V, et al. Toward optimal display of physiologic
A wider adoption of multimodal neuromonitoring will facili- status in critical care. I. Recreating bedside displays from archived
physiologic data. J Crit Care 2010;26:105.e1–e9.
tate the creation of a comprehensive database of neurophysi- 6. Morris G, Gardner R. Computer applications. In: Hall J, Schmidt G, Wood L,
ologic signals (and related data) that can be used for editors. Principles of critical care. New York: McGraw-Hill; 1992. p. 500–14.
retrospective review, research, and teaching. It is proposed 7. Imhoff M. Detecting relationships between physiological variables using
that a standardized method to annotate these multimodal data graphical modeling. In: Proc AMIA Symp 2002; 340–44, AMIA (Bethesda),
sets be developed. Such a database could be used for validation 2002.
8. Jennings D, Amabile T, Ross L. Informal assessments: data-based versus
and conformance testing of “knowledge plug-ins,” such as theory-based judgments. In: Kahnemann D, Slovic P, Tversky A, editors.
event detectors, decision support modules, and protocol strat- Judgments under uncertainty: heuristics and biases. Cambridge: Cambridge
egies. It would expedite their development and commercializa- University Press; 1982. p. 211–30.
tion similar to the way the annotated ECG database (developed 9. Angus DC, Kelley MA, Schmitz RJ, et al. Caring for the critically ill patient.
Current and projected workforce requirements for care of the critically ill
at Massachusetts Institute of Technology more than 30 years and patients with pulmonary disease: can we meet the requirements of an
ago) ushered in modern day arrhythmia analysis.77 aging population? JAMA 2000;284:2762–70.
10. Congressional Budget Office. Research on the comparative effectiveness of
medical treatments: issues and options for an expanded federal role.
Conclusion Washington; 2007.
11. Maas AI, Menon DK, Lingsma HF, et al. Re-orientation of clinical research
The concept of multimodality monitoring in neurocritical in traumatic brain injury: report of an international workshop on
care is not new; it has been discussed for more than 30 years. comparative effectiveness research. J Neurotrauma 2011;29 [Epub ahead of
However, even today there are only a few institutions that print].
provide something that approaches multimodality monitor- 12. Turn your alarms on! Anesthesia Patient Safety Foundation Newsletter
2004:43. Available at www.apsf.org.
ing in a comprehensive manner. The primary reason for this 13. Hlatky R, Robertson CS. Multimodality monitoring in severe head injury.
is the lack of a medical device communications standard. It Curr Opin Anaesthesiol 2002;15:489–93.
is surprising that today it is possible to buy any number of 14. Kett-White R, Hutchinson PJ, Czosnyka M, et al. Multi-modal monitoring of
devices for a home computer such as cameras, printers, and acute brain injury. Adv Tech Stand Neurosurg 2002;27:87–134.
scanners, and they all connect to the computer “out of the 15. Tisdall MM, Smith M. Multimodal monitoring in traumatic brain injury:
current status and future directions. Br J Anaesth 2007;99:61–7.
box.” Yet with medical devices this is not the case. Without a 16. Nangunoori R, Maloney-Wilensky E, Stiefel M, et al. Brain tissue oxygen-
database of physiologic signals collected in a consistent based therapy and outcome after severe traumatic brain injury: a systematic
manner, it is difficult to make progress in creating a truly literature review. Neurocrit Care 2011;16 [Epub ahead of print].
integrated knowledge environment. 17. Levine JM. Critical care management of subarachnoid hemorrhage. Curr
Neurol Neurosci Rep 2008;8:518–25.
Future evolution of NCCU informatics is likely to accelerate 18. Czosnyka M, Kirkpatrick PJ, Pickard JD. Multimodal monitoring and
for several reasons. First, the problems in the information assessment of cerebral haemodynamic reserve after severe head injury.
infrastructure encountered in the NCCU are not unique to Cerebrovasc Brain Metab Rev 1996;8:273–95.
402 Section VII—Computers, Engineering, and the Future

19. Chan KH, Dearden NM, Miller JD, et al. Multimodality monitoring as a 36. He P, Wilson G, Russell C, et al. Removal of ocular artifacts from the EEG: a
guide to treatment of intracranial hypertension after severe brain injury. comparison between time-domain regression method and adaptive filtering
Neurosurgery 1993;32:547–53. method using simulated data. Med Biol Eng Comput 2007;45:495–503.
20. Stiefel MF, Verdiramo AM, Schuster JM, et al. Multimodality monitoring 37. Devuyst S, Dutoit T, Stenuit P, et al. Removal of ECG artifacts from EEG
in the management of refractory intracranial hypertension: a case report. using a modified independent component analysis approach. Conf Proc
J Trauma 2005;59:757–61. IEEE Eng Med Biol Soc 2008;2008:5204–7.
21. Shiogai T, Takeuchi K. Multimodal neuromonitoring in impending brain 38. McCarthy J, Minsky M, Rochester N, et al. A proposal for the Dartmouth
death. Minerva Anesthesiol 1994;60:583–8. Summer Research Project on Artificial Intelligence; 1965. Available at http://
22. Czosnyka M, Whitehouse H, Smielewski P, et al. Computer supported www-formal.standard.edu/jmc/history/dartmouth.html.
multimodal bed-side monitoring for neuro intensive care. Int J Clin Monit 39. Arbib MA. The handbook of brain theory and neural networks. 2nd ed.
Comput 1994;11:223–32. Cambridge, Mass: MIT Press; 2003.
23. De Georgia MA, Deogaonkar A. Multimodal monitoring in the neurological 40. Haykin SS. Neural networks and learning machines. 3rd ed. New York:
intensive care unit. Neurologist 2005;11:45–54. Prentice Hall; 2009.
24. Kirkpatrick PJ, Czosnyka M, Pickard JD. Multimodal monitoring in 41. Imhoff M, Kuhls S. Alarm algorithms in critical care monitoring. Anesth
neurointensive care. J Neurol Neurosurg Psychiatry 1996;60:131–9. Analg 2006;102:1525–37.
25. Sorani MD, Hemphill 3rd JC, Morabito D, et al. New approaches to 42. Meredith C, Edworthy J. Are there too many alarms in the intensive care
physiological informatics in neurocritical care. Neurocrit Care 2007;7: unit? An overview of the problems. J Adv Nurs 1995;21:15–20.
45–52. 43. Kalogeropoulos D, Carson ER, Collinson PO. Clinical-HINTS: integrated
26. Miller JD, Piper IR, Jones PA. Integrated multimodality monitoring in the intelligent ICU patient monitoring and information management system.
neurosurgical intensive care unit. Neurosurg Clin N Am 1994;5:661–70. Stud Health Technol Inform 1997;43(Pt B):906–10.
27. Mulvey JM, Dorsch NW, Mudaliar Y, et al. Multimodality monitoring in 44. Aboukhalil A, Nielsen L, Saeed M, et al. Reducing false alarm rates for
severe traumatic brain injury: the role of brain tissue oxygenation critical arrhythmias using the arterial blood pressure waveform. J Biomed
monitoring. Neurocrit Care 2004;1:391–402. Inform 2008;41:442–51.
28. Vespa PM. Multimodality monitoring and telemonitoring in neurocritical 45. Blum JM, Kruger GH, Sanders KL, et al. Specificity improvement for
care: from microdialysis to robotic telepresence. Curr Opin Crit Care network distributed physiologic alarms based on a simple deterministic
2005;11:133–8. reactive intelligent agent in the critical care environment. J Clin Monit
29. Wartenberg KE, Mayer SA. Multimodal brain monitoring in the neurological Comput 2009;23:21–30.
intensive care unit: where does continuous EEG fit in? J Clin Neurophysiol 46. Apiletti D, Baralis E, Bruno G, et al. Real-time analysis of physiological data
2005;22:124–7. to support medical applications. IEEE Trans Inf Technol Biomed
30. Wartenberg KE, Schmidt JM, Mayer SA. Multimodality monitoring in 2009;13:313–21.
neurocritical care. Crit Care Clin 2007;23:507–38. 47. Zhang Y, Szolovits P. Patient-specific learning in real time for adaptive
31. Chen HI, Stiefel MF, Oddo M, et al. Detection of cerebral compromise with monitoring in critical care. J Biomed Inform 2008;41:452–60.
multimodality monitoring in patients with subarachnoid hemorrhage. 48. Patel VL, Zhang J, Yoskowitz NA, Green R, Sayan OR. Translational
Neurosurgery 2011;69(1):53–63. cognition for decision support in critical care environments: a review.
32. Hemphill 3rd JC, Barton CW, Morabito D, et al. Influence of data resolution J Biomed Inform 2008;41:413–31.
and interpolation method on assessment of secondary brain insults in 49. Nelson DW, Bellander BM, Maccallum RM, et al. Cerebral microdialysis of
neurocritical care. Physiol Meas 2005;26:373–86. patients with severe traumatic brain injury exhibits highly individualistic
33. Jordan KG. Continuous EEG and evoked potential monitoring in the patterns as visualized by cluster analysis with self-organizing maps. Crit Care
neuroscience intensive care unit. J Clin Neurophysiol 1993;10:445–75. Med 2004;32:2428–36.
34. Claassen J, Mayer SA. Continuous electroencephalographic monitoring in 50. Nelson DW, Thornquist B, MacCallum RM, et al. Analyses of cerebral
neurocritical care. Curr Neurol Neurosci Rep 2002;2:534–40. microdialysis in patients with traumatic brain injury: relations to
35. ASTM subcommittee F29.21. ASTM Standard F2761–09 Medical devices and intracranial pressure, cerebral perfusion pressure and catheter placement.
medical systems: essential safety requirements for equipment comprising the BMC Med 2011;9:21.
patient-centric integrated clinical environment (ICE). Part 1. General
requirements and conceptual model. ASTM International http:// A complete list of references for this chapter can be found online at
www.astm.org/Standards/F2761.htm;2009. www.expertconsult.com.
Section VII—Computers, Engineering, and the Future 402.e1

References 29. Wartenberg KE, Mayer SA. Multimodal brain monitoring in the neurological
intensive care unit: where does continuous EEG fit in? J Clin Neurophysiol
1. Gupta AK. Monitoring the injured brain in the intensive care unit. 2005;22:124–7.
J Postgrad Med 2002;48:218–25. 30. Wartenberg KE, Schmidt JM, Mayer SA. Multimodality monitoring in
2. Cohen MJ, Grossman AD, Morabito D. Identification of complex metabolic neurocritical care. Crit Care Clin 2007;23:507–38.
states in critically injured patients using bioinformatic cluster analysis. Crit 31. Chen HI, Stiefel MF, Oddo M, et al. Detection of cerebral compromise with
Care 2010;14:R10. multimodality monitoring in patients with subarachnoid hemorrhage.
3. Sahuquillo J. Does multimodality monitoring make a difference in Neurosurgery 2011;69(1):53–63.
neurocritical care? Eur J Anaesthesiol 2008;42(Suppl):83–6. 32. Hemphill 3rd JC, Barton CW, Morabito D, et al. Influence of data resolution
4. Imhoff M, Fried R, Gather U, et al. Dimension reduction for physiological and interpolation method on assessment of secondary brain insults in
variables using graphical modeling. AMIA Annu Symp Proc 2003:313–17. neurocritical care. Physiol Meas 2005;26:373–86.
5. Burykin A, Peck T, Krejci V, et al. Toward optimal display of physiologic 33. Jordan KG. Continuous EEG and evoked potential monitoring in the
status in critical care. I. Recreating bedside displays from archived neuroscience intensive care unit. J Clin Neurophysiol 1993;10:445–75.
physiologic data. J Crit Care 2010;26:105.e1–e9. 34. Claassen J, Mayer SA. Continuous electroencephalographic monitoring in
6. Morris G, Gardner R. Computer applications. In: Hall J, Schmidt G, Wood L, neurocritical care. Curr Neurol Neurosci Rep 2002;2:534–40.
editors. Principles of critical care. New York: McGraw-Hill; 1992. p. 500–14. 35. ASTM subcommittee F29.21. ASTM Standard F2761–09 Medical devices and
7. Imhoff M. Detecting relationships between physiological variables using medical systems: essential safety requirements for equipment comprising the
graphical modeling. In: Proc AMIA Symp 2002; 340–44, AMIA (Bethesda), patient-centric integrated clinical environment (ICE). Part 1. General
2002. requirements and conceptual model. ASTM International http://
8. Jennings D, Amabile T, Ross L. Informal assessments: data-based versus www.astm.org/Standards/F2761.htm;2009.
theory-based judgments. In: Kahnemann D, Slovic P, Tversky A, editors. 36. He P, Wilson G, Russell C, et al. Removal of ocular artifacts from the EEG: a
Judgments under uncertainty: heuristics and biases. Cambridge: Cambridge comparison between time-domain regression method and adaptive filtering
University Press; 1982. p. 211–30. method using simulated data. Med Biol Eng Comput 2007;45:495–503.
9. Angus DC, Kelley MA, Schmitz RJ, et al. Caring for the critically ill patient. 37. Devuyst S, Dutoit T, Stenuit P, et al. Removal of ECG artifacts from EEG
Current and projected workforce requirements for care of the critically ill using a modified independent component analysis approach. Conf Proc
and patients with pulmonary disease: can we meet the requirements of an IEEE Eng Med Biol Soc 2008;2008:5204–7.
aging population? JAMA 2000;284:2762–70. 38. McCarthy J, Minsky M, Rochester N, et al. A proposal for the Dartmouth
10. Congressional Budget Office. Research on the comparative effectiveness of Summer Research Project on Artificial Intelligence; 1965. Available at http://
medical treatments: issues and options for an expanded federal role. www-formal.standard.edu/jmc/history/dartmouth.html.
Washington; 2007. 39. Arbib MA. The handbook of brain theory and neural networks. 2nd ed.
11. Maas AI, Menon DK, Lingsma HF, et al. Re-orientation of clinical research Cambridge, Mass: MIT Press; 2003.
in traumatic brain injury: report of an international workshop on 40. Haykin SS. Neural networks and learning machines. 3rd ed. New York:
comparative effectiveness research. J Neurotrauma 2011;29 [Epub ahead of Prentice Hall; 2009.
print]. 41. Imhoff M, Kuhls S. Alarm algorithms in critical care monitoring. Anesth
12. Turn your alarms on! Anesthesia Patient Safety Foundation Newsletter Analg 2006;102:1525–37.
2004:43. Available at www.apsf.org. 42. Meredith C, Edworthy J. Are there too many alarms in the intensive care
13. Hlatky R, Robertson CS. Multimodality monitoring in severe head injury. unit? An overview of the problems. J Adv Nurs 1995;21:15–20.
Curr Opin Anaesthesiol 2002;15:489–93. 43. Kalogeropoulos D, Carson ER, Collinson PO. Clinical-HINTS: integrated
14. Kett-White R, Hutchinson PJ, Czosnyka M, et al. Multi-modal monitoring of intelligent ICU patient monitoring and information management system.
acute brain injury. Adv Tech Stand Neurosurg 2002;27:87–134. Stud Health Technol Inform 1997;43(Pt B):906–10.
15. Tisdall MM, Smith M. Multimodal monitoring in traumatic brain injury: 44. Aboukhalil A, Nielsen L, Saeed M, et al. Reducing false alarm rates for
current status and future directions. Br J Anaesth 2007;99:61–7. critical arrhythmias using the arterial blood pressure waveform. J Biomed
16. Nangunoori R, Maloney-Wilensky E, Stiefel M, et al. Brain tissue oxygen- Inform 2008;41:442–51.
based therapy and outcome after severe traumatic brain injury: a systematic 45. Blum JM, Kruger GH, Sanders KL, et al. Specificity improvement for
literature review. Neurocrit Care 2011;16 [Epub ahead of print]. network distributed physiologic alarms based on a simple deterministic
17. Levine JM. Critical care management of subarachnoid hemorrhage. Curr reactive intelligent agent in the critical care environment. J Clin Monit
Neurol Neurosci Rep 2008;8:518–25. Comput 2009;23:21–30.
18. Czosnyka M, Kirkpatrick PJ, Pickard JD. Multimodal monitoring and 46. Apiletti D, Baralis E, Bruno G, et al. Real-time analysis of physiological data
assessment of cerebral haemodynamic reserve after severe head injury. to support medical applications. IEEE Trans Inf Technol Biomed
Cerebrovasc Brain Metab Rev 1996;8:273–95. 2009;13:313–21.
19. Chan KH, Dearden NM, Miller JD, et al. Multimodality monitoring as a 47. Zhang Y, Szolovits P. Patient-specific learning in real time for adaptive
guide to treatment of intracranial hypertension after severe brain injury. monitoring in critical care. J Biomed Inform 2008;41:452–60.
Neurosurgery 1993;32:547–53. 48. Patel VL, Zhang J, Yoskowitz NA, Green R, Sayan OR. Translational
20. Stiefel MF, Verdiramo AM, Schuster JM, et al. Multimodality monitoring in cognition for decision support in critical care environments: a review.
the management of refractory intracranial hypertension: a case report. J Biomed Inform 2008;41:413–31.
J Trauma 2005;59:757–61. 49. Nelson DW, Bellander BM, Maccallum RM, et al. Cerebral microdialysis of
21. Shiogai T, Takeuchi K. Multimodal neuromonitoring in impending brain patients with severe traumatic brain injury exhibits highly individualistic
death. Minerva Anesthesiol 1994;60:583–8. patterns as visualized by cluster analysis with self-organizing maps. Crit Care
22. Czosnyka M, Whitehouse H, Smielewski P, et al. Computer supported Med 2004;32:2428–36.
multimodal bed-side monitoring for neuro intensive care. Int J Clin Monit 50. Nelson DW, Thornquist B, MacCallum RM, et al. Analyses of cerebral
Comput 1994;11:223–32. microdialysis in patients with traumatic brain injury: relations to
23. De Georgia MA, Deogaonkar A. Multimodal monitoring in the neurological intracranial pressure, cerebral perfusion pressure and catheter placement.
intensive care unit. Neurologist 2005;11:45–54. BMC Med 2011;9:21.
24. Kirkpatrick PJ, Czosnyka M, Pickard JD. Multimodal monitoring in 51. Minhas P, Perl TM, Carroll KC, et al. Risk factors for positive admission
neurointensive care. J Neurol Neurosurg Psychiatry 1996;60:131–9. surveillance cultures for methicillin-resistant Staphylococcus aureus and
25. Sorani MD, Hemphill 3rd JC, Morabito D, et al. New approaches to vancomycin-resistant enterococci in a neurocritical care unit. Crit Care Med
physiological informatics in neurocritical care. Neurocrit Care 2007;7: 2011;39(10):2322–9.
45–52. 52. Swiercz M, Mariak Z, Krejza J, et al. Intracranial pressure processing with
26. Miller JD, Piper IR, Jones PA. Integrated multimodality monitoring in the artificial neural networks: prediction of ICP trends. Acta Neurochir (Wien)
neurosurgical intensive care unit. Neurosurg Clin N Am 1994;5:661–70. 2000;142:401–6.
27. Mulvey JM, Dorsch NW, Mudaliar Y, et al. Multimodality monitoring in 53. Nikiforidis GC, Sakellaropoulos GC. Expert system support using Bayesian
severe traumatic brain injury: the role of brain tissue oxygenation belief networks in the prognosis of head-injured patients of the ICU. Med
monitoring. Neurocrit Care 2004;1:391–402. Inform (Lond) 1998;23:1–18.
28. Vespa PM. Multimodality monitoring and telemonitoring in neurocritical 54. Vath A, Meixensberger J, Dings J, et al. Prognostic significance of advanced
care: from microdialysis to robotic telepresence. Curr Opin Crit Care neuromonitoring after traumatic brain injury using neural networks.
2005;11:133–8. Zentralbl Neurochir 2000;61:2–6.
402.e2 Section VII—Computers, Engineering, and the Future

55. Rampil IJ. A primer for EEG signal processing in anesthesia. Anesthesiology 67. Li C, Shutter LA, Wu PM, et al. Potential of a simple lab-on-a-tube for
1998;89:980–1002. point-of-care measurements of multiple analytes. Lab Chip
56. Pfurtscheller G, Druschky K, Kamp HD, et al. [Multimodal evoked potentials 2010;10(11):1476–9.
and heart rate variability in comatose patients. 1. Method of measuring and 68. Chacón M, Pardo C, Puppo C, et al. Non-invasive intracranial pressure
normal values]. EEG EMG Z Elektroenzephalogr Elektromyogr Verwandte estimation using support vector machine. Conf Proc IEEE Eng Med Biol Soc
Geb 1987;18:108–14. 2010;2010:996–9.
57. Pfurtscheller G, Schwarz G, Schroettner O, et al. Continuous and 69. Rajajee V, Vanaman M, Fletcher JJ, et al. Optic nerve ultrasound for the
simultaneous monitoring of EEG spectra and brainstem auditory and detection of raised intracranial pressure. Neurocrit Care 2011;Jul 19 [Epub
somatosensory evoked potentials in the intensive care unit and the operating ahead of print].
room. J Clin Neurophysiol 1987;4:389–96. 70. Kasprowicz M, Schmidt E, Kim DJ, et al. Evaluation of the cerebrovascular
58. Litscher G, Schwarz G. Noninvasive bioelectrical neuromonitoring in pressure reactivity index using non-invasive Finapres arterial blood pressure.
anaesthesia and critical care. Eur J Anaesthesiol 2001;18:785–8. Physiol Meas 2010;31(9):1217–28.
59. Vespa PM, Nuwer MR, Juhasz C, et al. Early detection of vasospasm after 71. Litscher G. A multifunctional helmet for noninvasive neuromonitoring.
acute subarachnoid hemorrhage using continuous EEG ICU monitoring. J Neurosurg Anesthesiol 1998;10:116–19.
Electroencephalogr Clin Neurophysiol 1997;103:607–15. 72. Bein B, Meybohm P, Cavus E, et al. A comparison of transcranial Doppler
60. Vespa PM, Boscardin WJ, Hovda DA, et al. Early and persistent impaired with near infrared spectroscopy and indocyanine green during hemorrhagic
percent alpha variability on continuous electroencephalography monitoring shock: a prospective experimental study. Crit Care 2006;10:R18.
as predictive of poor outcome after traumatic brain injury. J Neurosurg 73. Zhou C, Eucker SA, Durduran T, et al. Diffuse optical monitoring of
2002;97:84–92. hemodynamic changes in piglet brain with closed head injury. J Biomed Opt
61. Hebb MO, McArthur DL, Alger J, et al. Impaired percent alpha variability on 2009;14:034015.
continuous electroencephalography is associated with thalamic injury and 74. Buckley EM, Cook NM, Durduran T, et al. Cerebral hemodynamics in
predicts poor long-term outcome after human traumatic brain injury. preterm infants during positional intervention measured with diffuse
J Neurotrauma 2007;24:579–90. correlation spectroscopy and transcranial Doppler ultrasound. Opt Express
62. Smielewski P, Czosnyka M, Zabolotny W, et al. A computing system for the 2009;17:12571–81.
clinical and experimental investigation of cerebrovascular reactivity. Int J 75. Medical Device Plug and Play. <http://mdpnp.org/ICE.html>.
Clin Monit Comput 1997;14:185–98. 76. Essential safety requirements for equipment comprising the patient-centric
63. Hemphill C. Personal communication Oct. 5, 2009. integrated clinical environment (ICE). Part I. General requirements and
64. Kohli-Seth R, Oropello JM. The future of bedside monitoring. Crit Care Clin conceptual model. In: F2761: ASTM; 2009.
2000;16:557–78, vii-viii. 77. Moody GB, Mark RG. The impact of the MIT-BIH arrhythmia database.
65. Li C, Wu P, Narayan RK, et al. A novel lab-on-a-tube for multimodal monitoring IEEE Eng Med Biol Mag 2001;20:45–50.
of patients with traumatic brain injury. Lab Chip 2009;9(14):1988–90.
66. Li C, Wu P-M, Jung W, et al. A novel lab-on-a-tube for multimodal
monitoring of patients with traumatic brain injury. Proceedings from the
IEEE Transducers 2009 symposium. p. 1449-52, Denver; 2009.
Chapter
41  
VII

Simulation in the ICU


W. Andrew Kofke

Introduction can range from simple insertion of an intravenous cannula


into a simulated arm to a computerized virtual reality
Simulation: a situation or environment created to allow world that allows an operator to learn highly complex
persons to experience a representation of a real event for procedures such as transesophageal echocardiography,
the purpose of practice, learning, evaluation, testing, or to laparoscopy, or endoscopy procedures.4 One example of a
gain understanding of systems or human actions. new virtual reality simulator is that developed by Banerjee
—definition taught at the Center for Medical et al.5,6 to teach ventriculostomy placement (Fig. 41.1).
Simulation, Cambridge, Mass.  PC-based simulation. This entails use of PC-based pro-
In the 1960s, Red Cross first aid classes taught a new cardio- grams that are installed on laptop computers. They can be
pulmonary resuscitation (CPR) method, having discarded the set up to mimic real life scenarios such that the user has
chest pressure-arm lift method, the primary resuscitation to run through all of the cognitive decision algorithms that
method of the 1950s and 1960s and perhaps before. So how are required in clinical practice. Moreover, in accordance
was CPR practiced? On Resusci-Anne—a relatively new with the apparent preferences of the current younger
Laerdal product that was used everywhere to teach mouth-to- generation, the PC-based simulation also can be set up as
mouth and chest compressions following the contemporary a game.
 Standardized patients. What can be better than working on
recommendations of the American Heart Association. By
today’s standards this is a relatively primitive simulator, one a real human? This notion has resulted in the development
that is still in use. of standardized patient programs in many schools and
However, perhaps the most sophisticated simulator of all a standardized patient society (www.aspeducators.org).
had already been demonstrated in the late 1960s by Steven Standardized patients are real people, individuals who are
Abrahamson, the Dean of Education at University of South- trained to display the appropriate behaviors of patients
ern California.1 Abahamson et al. developed a high-fidelity with given diseases in given situations or individuals with
simulator—SIM-ONE—that was more sophisticated then the relevant physical findings. Both types of standardized
than some simulators that are currently available; it even fas- patients can be used to teach novices how to deal with a
ciculated after succinylcholine injection. However, SIM-ONE variety of problems.
 High-fidelity simulation. This uses simulators that bring
required a room full of computers, each with an attendant
technician in the manner of a NASA space launch. Dr. Abra- together all of the physiology and pharmacologic responses
hamson went on sabbatical, his simulator was dismantled, so of a human in a manikin, which breathes, may produce or
the story goes, and it took advances in computer technology consume a variety of different gases, and talks in an inter-
and the vision of subsequent simulation pioneers before active manner with the students. After appropriate suspen-
resurrection of this technology in the 1990s. Simulation is sion of disbelief, this manikin becomes for all intents and
now an exponentially growing field, driven by the patient- purposes a real human being with a real problem that
safety movement, changes in the educational opportunities students have to diagnose and treat appropriately.7
available to trainees, and evolving computer capabilities.
Medical-simulation centers that numbered in the 10s in the All of these elements of simulation can be further integrated
early 1990s now number in the 1000s worldwide. Further- to create scenarios that encompass any or all of these methods.
more there is accumulating evidence that simulation can be Students can perform procedures, make decisions, and inter-
used in training of physicians and health care providers to act with a patient (manikin or standardized patient who may
help reduce medical errors, improve technical skills for some be attached to a task trainer). At an even higher level, students
invasive procedures, and enhance communication skills.2,3 may interact with other health care providers on a team. This
Simulation in terms of a taxonomy can be characterized as had led increasingly to the use of such simulation tools to train
follows: health care providers in the principles of crew/crisis resource
management (CRM) (teamwork), another lesson from the
 Task training. This entails use of simulation devices meant aviation industry. And finally simulation can be used as an
to teach specific psychomotor tasks with complexity that essential tool for microsystem or macrosystem simulations.
© Copyright 2013 Elsevier Inc. All rights reserved. 403
404 Section VII—Computers, Engineering, and the Future

of using a team model to resuscitate victims of severe trauma.16


Providing further support to this concept, there is now com-
pelling data on the advantages of CRM team training on error
rates and practice process variables and, moreover, the value
of attending intensivist contribution to critical care outcome.
All of these observations and those of others clearly indicate
process change as an area for improvement that could improve
survival after in-hospital resuscitation. The notable disparity
in ROSC and discharge survival rates suggest that improve-
ments in processes of practice can reasonably be targeted as
aspects of patient care that could increase the rate of meaning-
ful survival after cardiac arrest or near arrest.
The aviation and nuclear energy industries, as well as the
military, all described as high reliability organizations, have a
Fig. 41.1  ImmersiveTouch. Ventriculostomy simulation is depicted, rich and successful history of using crisis simulation to train
showing the observer’s view and the operator’s view. (From Issenberg SB, personnel to deal with high-risk situations in complex
Chung HS, Devine LA. Patient safety training simulations based on competency environments.17-19 More recently, the pioneering work of anes-
criteria of the accreditation council for graduate medical education. Mt Sinai J Med thesiologists, critical care specialists, and others has extended
2011;78(6):842–53, with permission.) the knowledge developed in these industries to acute care
medicine and the operating room.20-23 These providers have
collaborated with industry to develop realistic computerized
manikin systems, which simulate medical crisis situations
Such simulations encompass handling multiple patients with typically encountered in the operating room and intensive
management of medical, interpersonal, and system issues. care unit (ICU) and that may be directed at physicians, nurses,
In the context of critical care, CRM is an important new advanced practitioners, residents, or students.24-27 The systems
vista that can be used to improve team performance. are being further developed and adapted to medical crisis situ-
ations outside of the operating room. For example, these units
can provide both inexperienced and experienced personnel in
Crisis Resource Management a number of fields with exposure to common and uncommon
In 2000, the American Heart Association established medical crises such as myocardial ischemia, hypothermia,
the National Registry of Cardiopulmonary Resuscitation acute respiratory distress syndrome, septic shock, anaphylaxis,
(NRCPR) to assist participating hospitals with systematic data complex arrhythmias, Advanced Cardiac/Trauma Life Support
collection on resuscitative efforts. In so doing, the AHA pro- protocols, pneumothorax, difficult airway situations, and
vided a prospective, multisite, observational mechanism to many others. Moreover, the realistic setting of the simulation
determine the epidemiology and outcomes from in-hospital “theater” can be used to create realistic situations that teach
resuscitation. Initial results from the registry were reported important lessons about optimal team dynamics and can help
by Peberdy et al.8 in 2003 from 14,720 cardiac arrests in adults improve leadership, teamwork, and self-confidence skills to
at 207 hospitals, of which 86% had an organized emergency manage medical emergencies. It should be remembered that
response team continuously available. The researchers effective instructor training is needed for successful imple-
reported that 44% of adult in-hospital cardiac arrest victims mentation of simulation based CRM learning.28
had a return of spontaneous circulation (ROSC), but only
17% survived to hospital discharge. Neurologic outcome was
reported as good in 86% of discharged survivors. This 17% What Is Crisis Resource
discharge survival rate is consistent with reports of others.9
Various models have been proposed to prevent cardiac
Management?
arrests and improve survival following cardiac arrest, such as Risser et al.29 in their analysis of medical teamwork failures
using emergency medicine residents as responders10 and creat- describe a core team as “a set of 3 to 10 clinically skilled care-
ing emergency response teams,11 but these recommendations givers who work together during a shift and have been trained
have yet to be translated into improved survival after inpatient to use specific teamwork behaviors to tightly coordinate and
cardiac arrest. Several reasons can be attributed to the rela- manage their clinical actions.” Risser et al. adapted knowledge
tively poor results after inpatient cardiac arrest. These cer- from aviation CRM to the emergency department acute care
tainly include patients’ premorbid severity of illness such that setting and derived five team dimensions:
any practice or process changes in resuscitation would not be
expected to have a meaningful impact. Nonetheless, several 1. Maintenance of team structure and climate. This refers ini-
studies emphasize the important role that various practices tially to:
have on patient outcomes after resuscitation. For example,  The establishment of the team leader and organization
time to CPR was identified as an important factor in pre­ of the team
dicting outcome for both inpatient9 and out-of-hospital  Cultivation of team climate
cardiac arrest,12 and organized early medical emergency  Constructive resolution of conflicts
response team involvement appears to positively affect Success in the subsequent four dimensions depends
outcomes.13-15 Trauma surgeons have been leaders in this on this one being successfully implemented. Examples
aspect of resuscitation, and have demonstrated the advantage of common team failures in this area include not
Section VII—Computers, Engineering, and the Future 405

assigning roles properly and not holding team members and mission. Baker et al., in general agreement with Risser
accountable. et al., indicates that each team member is able to do the fol-
Dimensions 2, 3, and 4, described next, are the opera- lowing to identify when errors occur and how to recover and
tional core of a high-quality team and are functionally correct for these errors:
intertwined, essentially occurring simultaneously. The  Anticipate the needs of others
caregivers may have to balance competing demands and  Adjust to each other’s actions and to the changing
must maintain a high level of situational awareness.30
environment
2. Application of problem-solving strategies. This describes: 

Have a shared understanding of how a procedure should
Conducting situational planning

happen
Application of decision-making methods
 Engaging in error correction actions Hunt et al.,33 in a review of teamwork training, indentifies
This dimension relies on adequate communication. This several factors listed in Table 41.1 as elements of high per-
means that all team members are a part of decision- forming teams.
making, that the leader asks for and is given relevant infor-
mation, and that relevant protocols are implemented.
This dimension also has caregivers being aware of the Simulation as a Strategy for CRM
three basic types of errors: slips, lapses, and mistakes,31 and
being assertive enough to identify and correct them.
Training in Critical Care
Examples of common team failures in this area include not Recent advances in simulation technology suggest that simu-
identifying relevant established protocols, not engaging lation can be used as an important tool to implement a strat-
other team members in decision making, not alerting the egy to improve critical care outcomes by using it as an essential
team to potential bias and error, and not asserting the need tool to teach CRM.
for corrective actions. Building on the seminal work of Abrahamson34 in the
3. Communication with the team. This refers to: 1960s to develop the first medical simulator; high-fidelity
 The use of local standards for effective communication patient simulation has become widely available with an expo-
 Provision of supporting information nential jump in progress and acceptance over the past 15
 Requesting supporting information years. The number of simulation centers worldwide num-
The emphasis of this dimension is the assurance bered about 10 in 1994, 200 in 2001, and 452 in 2005.35 Pres-
within the team of a common understanding of patient ently, 126 simulation centers are listed in the directory of the
and operational issues that affect the performance of Society for Simulation in Healthcare.36 Ongoing technical
the team and patient outcome. There is timely informa- improvements have been evident with a substantial amount
tion transfer and maintenance of team situational aware- of study to ascertain efficacy of human acute-care simulation,
ness. Examples of common team failures in this area as was first suggested by Abrahamson.34 Gaba et al. and others
include not using a check-back process to verify com- have described the use of a simulator suite to recreate the
munication, not offering information that supports a operating room for research and clinical training.37-40 Investi-
decision, not seeking needed information, and not com- gators have subsequently demonstrated that simulators can
municating plans to the team members. help reduce morbidity due to critical incidents,20,37,41-45 to
4. Execution of plans and management of workload. This provide CRM training,37,46 and to more effectively provide
refers to: resident trainees with a safe and effective milieu to learn
 Plan implementation appropriate procedures in acute care situations.47,48 Recent
 Primary and secondary triage research has demonstrated that simulation can be applied to
 Task prioritization effectively measure some quality measures in cardiac arrest
 Management of team workload and resources response teams49 and that it can be used in surgical proce-
 Cross-monitoring team member actions dures to increase efficiency and safety.50 Simulation is now an
 Maintaining situational awareness accepted tool in the improvement of patient safety in many
The emphasis of this dimension is to encourage willing- disparate situations.
ness of team members to ask for assistance and to assist From a psychological perspective, Rose et al.51 demon-
others to eliminate work overload and poor integration of strated the feasibility of the notion that prior training of a
everyone’s tasks. Examples of common team failures in nonmedical task in a simulated environment can be expected
this area include not executing the protocol or plan, not to result in transfer of skills to real life situations. For medical
integrating team member assessments of patient needs, procedural simulations, several investigators have demon-
poor task prioritization, and inadequate cross-monitoring strated that prior procedure simulation results in better sub-
of team members and the team’s plan execution. sequent performance in a simulated environment.52-54 Tuggy
5. Improvement of team skill. This refers to engaging in infor- et al.55 reported improved sigmoidoscopy performance on
mal and formal team improvement strategies. This entails human volunteers with prior simulation. Grantcharov et al.56
informal discussion at the time of the event and later and Seymour et al.50 using a prospective randomized design
formal review and discussion. demonstrated the capability of prior simulator training to
significantly decrease errors and improve efficiency in the
Baker et al.,32 in a recent review of this topic, outline knowl- operating room. This presumably translates into better patient
edge, skills, and attitudes that members of a high-quality team surgical outcome, but that element was not studied. Ahlberg
should possess that facilitate coordinated, adaptive perfor- et al.57 showed that previous training on a simulator resulted
mance and provide support of one’s teammates, objectives, in improved colonoscopy performance. Conversely, in another
406 Section VII—Computers, Engineering, and the Future

Table 41.1  Characteristics of High Performing Teams


Situation Awareness (SA) Team performance is improved when team members continually assess their environment and update
each other in a process call “shared cognition” so that they are making decisions based on current
information and can have a “shared mental model” of the current state of affairs and an updated
plan of action with contingencies. SA allows team to maintain “big picture” view of situation.
Effective military and aviation teams have higher SA than low-performing teams.
Leadership An effective team leader can both command the team and values input from team members.
Flattening the hierarchy improves safety as information can flow both directions, while leaders who
maintain an authoritarian type of leadership “reinforce large authority gradients, creating
unnecessary risk.” A leader should try not to perform procedures unless the procedure is essential
and no one else is capable of doing it. Stepping back and keeping a “bird’s eye view” allows the
leader to take in and process more information and contributes to situational awareness.
Followership The nonleader members of the team are called followers. Good “followership” is just as important
for good team functioning as good leadership. Followers need to know their individual role on the
team but also contribute to overall team functionality. They must contribute to situational
awareness by verbalizing observations about changes in the environment, ideas about diagnosis, to
decrease the leaders’ workload if necessary and finally to help the leader avoid mistakes; for
example, “the team leader might focus on an incorrect diagnosis and apply the wrong rule
(treatment) owing to a fixation error, or be incapacitated, hence everyone in the team should
always be alert.” Finally, followers must not assume that the leader knows everything and should
feel obligated to share observations that might impact outcome.
Closed loop communication Closed looped communication is used to ensure that the message that was sent is heeded and
understood, and involves: “(1) the sender initiating a message, (2) the receiver receiving the
message, interpreting it, and acknowledging its receipt, and (3) the sender following up to ensure
the intended message was received.”
Critical language and Critical language refers to the use of a catch phrase that means something to every member of an
standardized practices organization and requires specific action (standardized practices). United Airlines developed the
CUS program for “I’m concerned, I’m uncomfortable, this is unsafe, or I’m scared,” which is
adopted within the culture as meaning, “We have a serious problem; stop and listen to me.”
Another example of a standardized approach to improve the effectiveness of communication is
SBAR (situation, background, assessment, recommendation). This is a tool that gives an outline of
how “awareness and education regarding the fact that nurses, physicians, and other clinicians are
taught to communicate in very different styles.”
Assertive communication Safe patient care may depend on the ability for a team member to speak up and get the attention
of other team members when they believe something might be going wrong. This is more likely to
happen if a team member believes speaking up will not be held against him or her. A recurrent
phrase is that people can still show deference to expertise, but speak up in a “nonthreatening and
respectful manner.” The idea is that all team members may have valuable input, “regardless of
rank.” The “hint and hope” model has been described as a common and dangerous way of trying
to indirectly communicate with other team members.
Adaptive behaviors Teams whose members are flexible and perform as needed to optimize team functioning and
demonstrate “adaptive behaviors” are those that can truly benefit from the synergy of an effective
team. Examples of adaptive behavior that optimize team functionality include the following: “(1)
team members ask for help when overloaded, (2) team members monitor each other’s performance
to notice any performance decreases (mutual performance monitoring), or (3) team members take
an active role in assisting other team members who are in need of help (backup behavior). An
essential component to the above actions happening is trust among team members.”
Workload management Workload management depends on team members demonstrating adaptive behaviors. This principle
requires: (1) “proper allocation of tasks to individuals, (2) avoidance of work overloads in self and
in others, (3) prioritization of tasks during periods of high workload, and (4) preventing
nonessential factors from distracting attention from adherence to protocols, particularly those
relating to critical tasks.”
Debriefing Debriefing is the process of reviewing a simulation or real event after it is complete to optimize any
lessons that can be learned. When using simulation as a teaching tool, simulation with no
debriefing and feedback does not result in effective learning. In terms of real events, teams that
debrief themselves afterwards have been shown to be higher performing.

From Hunt EA, Shikofsk, NA, Stavroudis TA, et al. Simulation: translation to improved team performance. Anesthesiol Clin 2007;25(2):301-19.

study of technical skill on patients, Gerson58 compared resi- Notably lacking are studies that demonstrate that such prior
dents trained in endoscopy on patients with those trained on procedural training has a positive impact on measures of per-
a simulator and concluded that traditional methods were formance and quality care.
superior, although they did not account for possible morbidity Gaba and others have pioneered the application of simula-
in the patients who were subjects of the traditional training. tion training to the medical community, following many of
Section VII—Computers, Engineering, and the Future 407

the tenants of aviation training. His group initially used simu- scenarios can be conducted to practice difficult transports. For
lation technology to evaluate human performance during example, a sick intubated patient with multiple brain and car-
anesthesia crisis situations; this resulted in the development diovascular monitors could be taken to radiology in a simula-
of Anesthesia Crisis Resource Management.59,60 The principles tion scenario. Ensuring an adequate oxygen supply, having
from the early work are being adapted to critical care. airway equipment on hand, and accidental infusion discon-
CRM training applied to medicine is similar in aviation tinuation can be simulated. Application of teamwork princi-
training, including the three essential elements of CRM train- ples to such situations appears to decrease the incidence of
ing: knowledge, practice, and recurrence.61 Simulated scenar- transport mishaps.67,68 Similarly simulation-based training can
ios are created and applied to evaluate team performance, in be used to enhance “hand-off ” skills and communication.69
addition to preventing small management errors that can lead
to lethal results, such as in cardiopulmonary arrest or critical
care transport. Debriefing is then used as an essential element Does CRM Work in Medicine?
of CRM training. Simulation to teach CRM has been shown Many institutions presently are grappling with whether and
to be ineffective if there is no debriefing. For example, Savodelli how to invest in simulation and CRM training, but without
et al.62 demonstrated the importance of debriefing in a study scientific evidence it is difficult to determine who should
showing that those trainees who received audio or visual feed- participate, the level of training, and the impact on patient
back following a simulated scenario performed significantly care. While simulation-based learning has greatly enhanced
better on subsequent scenarios than their counterparts who the technical skill of physicians and other health care profes-
had not received debriefing. sionals in performing selected interventions, the benefits of
Neurocritical care units (NCCUs) are comprised of dispa- simulation-based learning coupled with CRM have not been
rately composed dynamic teams that must act correctly and adequately investigated in “real life” patient care situations.
fast when unexpected critical events arise. NCCU patients Furthermore, the perceived advantages of using human
typically require complex physiologic monitoring with new simulators in clinical laboratories as a teaching method have
technologies with which all members may not be familiar. not been translated into measurable benefits for patients
Simulation is helpful in such circumstances to replicate the and institutions. The benefits of simulation team training
NCCU environment in advance of the real crisis. Complex also may require repeated simulation exposure rather than a
scenarios that require successful team interactions can be single exposure.70 Cost-constrained hospitals are therefore
simulated, such as cardiac arrest, cerebral herniation, exsan- reluctant to commit the necessary manpower and equipment
guination, arrhythmias, transport, and team/family behav- resources to sustain these programs, although programs can
ioral problems. Specific human patient simulators, such as the be shaped to meet specific institutional needs and budget
Human Patient Simulator (HPS) produced by Medical Educa- limitations.
tion Technologies, have been used to enhance neurocritical Nonetheless, from this technology and background has
care education.63 sprung a growing interest among health care professionals
Procedures performed in the NCCU, such as intubation and across disciplines and over the past decade to develop and
central venous pressure (CVP) catheter placement, also can be train CRM principles. It is clear that simulation and medical
included in such simulations, including hand washing and education are now at a historic patient-safety interface. This
sterile procedures. For example, an improper sterile technique has been largely driven by the Institute of Medicine report71
could be used by an actor in a simulation with the teaching “To Err is Human.” This report recommended development
goal to reinforce assertive tactful communication by other of patient-safety programs that “establish interdisciplinary
team members. team training programs, such as simulation, that incorporate
Leadership failure has been quoted as a common error in proven methods for team management.” Their conclusions
simulated ICU crisis situations.64 The use of simulation and were derived from studies such as those of Risser et al.,21 which
associated debriefing sessions to create the need for leadership retrospectively demonstrated that lack of team behaviors
has been favorably received as a valuable teaching tool at likely contributed to adverse events in the emergency depart-
Stanford.64 ment. Moreover, Moorthy et al.72 demonstrated, in the absence
Use of standardized patients (SPs) is another means to of CRM training, many teamwork deficiencies in a simulated
augment team communication skills. For example, SPs can be surgery environment. More recently Morey et al.,22 in a multi-
used to create problems in team dynamic during a crisis or to institutional project, demonstrated significant reduction in
simulate problems in dealing with families, such as breaking errors when an intensive CRM team training, but without use
bad news, doing difficult consents, or simulating discussions of simulation, was implemented.
regarding end-of-life care. Several other studies have evaluated the impact of
Equipment problems can be an obstacle to successful team CRM. The outcome measures from these studies have reported
function. Simulation has a dual role here. The first is to test satisfaction by students with the training,20,73-76 improved
equipment before purchase for any issues in its use in the local team performance dynamics in clinical situations18 or in a
environment. The second is to ensure adequate familiarity of simulated environment,37,75,77-79 decreased incidence of bedside
team members with the equipment that is available to them. errors,18 and improved performance in a simulation environ-
Hunt et al.65 have demonstrated the utility of team training in ment.37,75,80,81 As a general rule, better teamwork translates into
a simulated environment to ensure that team members make better performance.82 Most recently DeVita et al.80 have pro-
correct crucial decisions regarding pacemaker use. vided some important data about the efficacy of training
Reports indicate that mishaps with transport related to un- teams in a simulated environment. They found that prior
familiarity with equipment and with potential obstacles that CRM training for medical emergency response teams results
may arise are frequent and important problems.66 Simulation in better later performance of the same teams in the same
408 Section VII—Computers, Engineering, and the Future

simulated environment. The potential benefits of simulation 10. Adams BD, Zeiler KK, Jackson WO, et al. Emergency medicine residents
effectively direct inhospital cardiac arrest teams. Am J Emerg Med 2005;
may extend beyond CRM. For example, simulation studies or
23(3):304–10.
simulation modeling may be used to inform infection control 11. Foraida MI, Devita MA, Braithwaite RS, et al. Improving the utilization of
methods,83 identify features of team efficiency, such as task medical crisis teams (Condition C) at an urban tertiary care hospital. J Crit
allocation and closed-loop communication, that can translate Care 2003;18:87–94.
into improved outcome,84 explain variance across patient 12. Holmberg M, Holmberg S, Herlitz J, et al. Factors modifying the effect of
bystander cardiopulmonary resuscitation on survival in out-of-hospital
cohorts,85 enhance drug dosing,86 evaluate performance of cardiac arrest patients in Sweden. Eur Heart J 2001;22(6):511–19.
common ICU monitors (e.g., glucometer),87 evaluate the 13. Bellomo R, Goldsmith D, Uchino S, et al. A prospective before-and-after trial
effects of fatigue on performance,88 or maximize design, of a medical emergency team. Med J Aust 2003;179:283–7.
enrollment, and execution of clinical trials,89 among others. 14. Goldhill DR, Worthington LM, Mulcahy AJ, et al. The patient-at-risk team:
identifying and managing seriously ill ward patients. Anaesthesia 1999;54:
853–60.
Conclusion 15. Buist MD, Moore GE, Bernard SA, et al. Effects of a medical emergency team
on reduction of incidence of and mortality from unexpected cardiac arrests
In summary, there has been significant progress in patient in hospital: preliminary study. Br Med J 2002;324(7334):387–90.
safety but there remains a need for practitioners to use CRM 16. Petrie D, Lane P, Stewart T. An evaluation of patient outcomes comparing
trauma team activated versus trauma team not activated using TRISS
methods that have been found to be helpful in high reliability analysis. Trauma and Injury Severity Score. J Trauma Inj Infect Crit Care
organizations such as the airline industry or nuclear energy 1996;41(5):870–3.
industries. Based on these organizations’ excellent safety 17. Wiener EL, Helmreich RL. Cockpit resource management. San Diego, CA:
records, it becomes logical to suggest that using simulation of Academic Press; 1993.
18. Orlady J. Airline pilot training today and tomorrow, cockpit resource
rare events for the experienced or simulation of common
management. In: Weiner EL, Kanki BG, Helmreich RL, editors. Cockpit
events for the inexperienced should be the next vista in patient resource management. San Diego: Academic Press; 1995. p. 447–78.
safety and in critical care taught largely in the context of CRM 19. Eisen LA, Savel RH. What went right: lessons for the intensivist from the
training with simulation as an important tool. crew of U.S. Airways Flight 1549. Chest 2009;136(3):910–17.
It is clear that many advances are emerging in this field and 20. Schroeder T, Schierbeck J, Howardy P, et al. Effect of labetalol on CBF and
middle cerebral arterial flow velocity in healthy volunteers. Neurol Res 1991;
that the medical culture of the twenty-first century is going to 13:10.
be extraordinarily different from that of the twentieth century. 21. Risser DT, Rice MM, Salisbury ML, et al. The potential for improved
How health care providers are educated also is changing; teamwork to reduce medical errors in the emergency department. The
alternative media resources such as Twitter,90 web-based edu- MedTeams Research Consortium. Ann Emerg Med 1999;34(3):373–83.
22. Morey JC, Simon R, Jay GD, et al. Error reduction and performance
cation, computer-assisted learning,91 and simulation-based
improvement in the emergency department through formal teamwork
medical education92 that apply to both students and estab- training: evaluation results of the MedTeams project. Health Serv Res 2002;
lished multidisciplinary teams93 now are commonplace. We 37(6):1553–81.
are going to transition from “see one, do one, teach one,” to a 23. Ziewacz JE, Arriaga AF, Bader AM, et al. Crisis checklists for the operating
culture of “practice it until proficient and then, and only then, room: development and pilot testing. J Am Coll Surg 2011;213(2):212–17.
24. Fernandez GL, Lee PC, Page DW, et al. Implementation of full patient
… do one.” Indeed, it should not be surprising to find the simulation training in surgical residency. J Surg Educ 2010;67(6):393–9.
twenty-first century routine to include the use of high-stakes 25. Lambden S, Martin B. The use of computers for perioperative simulation in
simulation for certification and hospital credentialing. As anesthesia, critical care, and pain medicine. Anesthesiol Clin 2011;29(3):
such these advances represent an important cultural shift in 521–31.
26. Mould J, White H, Gallagher R. Evaluation of a critical care simulation series
medicine, one that is still evolving and likely to affect the
for undergraduate nursing students. Contemp Nurse 2011;38(1-2):180–90.
practice of neurocritical care, including how various monitors 27. Pascual JL, Holena DN, Vella MA, et al. Short simulation training improves
are used. objective skills in established advanced practitioners managing emergencies
on the ward and surgical intensive care unit. J Trauma 2011;71(2):330–8.
References 28. Cheng A, Donoghue A, Gilfoyle E, et al. Simulation-based crisis resource
management training for pediatric critical care medicine: a review for
1. Abrahamson S. An interview of Professor Stephen Abrahamson. Educ Health instructors. Pediatr Crit Care Med 2011;14 [Epub ahead of print].
(Abingdon) 2003;16(3):378–84. 29. Risser DT, Rice MM, Salisbury ML, et al. The potential for improved
2. Smith CC, Huang GC, Newman LR, et al. Simulation training and its effect teamwork to reduce medical errors in the emergency department. Ann
on long-term resident performance in central venous catheterization. Simul Emerg Med 1999;34(3):373–83.
Healthc 2010;5(3):146–51. 30. Wright MC, Taekman JM, Endsley MR. Objective measures of situation
3. Issenberg SB, Chung HS, Devine LA. Patient safety training simulations awareness in a simulated medical environment. Qual Saf Health Care 2004;
based on competency criteria of the accreditation council for graduate 13(Suppl 1):165–71.
medical education. Mt Sinai J Med 2011;78(6):842–53. 31. Reason J. The contribution of latent human failures to the breakdown of
4. Bose RR, Matyal R, Warraich HJ, et al. Utility of a transesophageal complex systems. Philos Trans R Soc Lond B Biol Sci 1990;327(1241):
echocardiographic simulator as a teaching tool. J Cardiothorac Vasc Anesth 475–84.
2011;25(2):212–15. 32. Baker DP, Salas E, King H, et al. The role of teamwork in the professional
5. Luciano CJ, Banerjee PP, Lemole Jr GM, et al. Second generation haptic education of physicians: current status and assessment recommendations.
ventriculostomy simulator using the ImmersiveTouch system. Stud Health Joint Commission J Quality Patient Safety 2005;31(4):185–202.
Technol Inform 2006;119:343–8. 33. Hunt EA, Shilkofski NA, Stavroudis TA, et al. Simulation: translation to
6. Banerjee PP, Luciano CJ, Rizzi S. Virtual reality simulations. Anesthesiol Clin improved team performance. Anesthesiol Clin 2007;25(2):301–19.
2007;25(2):337–8. 34. Abrahamson S, Denson JS, Wolf RM. Effectiveness of a simulator in training
7. Kofke WA, Rosen KA, Barbaccia J, et al. The value of acute care simulation. anesthesiology residents. J Med Educ 1969;44:515–19.
W V Med J 2000;96(2):396–402. 35. Berman S. Individual lifetime achievement: Jeffrey B. Cooper, PhD. Joint
8. Peberdy MA, Kaye W, Ornato JP, et al. Cardiopulmonary resuscitation of Commission J Quality Patient Safety 2003;29(12):625–33.
adults in the hospital: a report of 14720 cardiac arrests from the National 36. Directory of Sim Centers. Available at http://ssih.org/directory-of-sim-
Registry of Cardiopulmonary Resuscitation. Resuscitation 2003;58:297–308. centers-2. The Society for Simulation in Healthcare. Accessed June 28, 2012.
9. Herlitz J, Bång A, Alsén B, et al. Characteristics and outcome among patients 37. Howard SK, Gaba DM, Fish KJ, et al. Anesthesia crisis resource management
suffering from in hospital cardiac arrest in relation to the interval between training: teaching anesthesiologists to handle critical incidents. Aviat Space
collapse and start of CPR. Resuscitation 2002;53(1):21–7. Environ Med 1992;63(9):763–70.
Section VII—Computers, Engineering, and the Future 409

38. Good ML, Gravenstein JS. Anesthesia simulators and training devices. Int 46. Holzman RS, Cooper JB, Gaba DM, et al. Anesthesia crisis resource
Anesthesiol Clin 1989;27:161–8. management: real-life simulation training in operating room crises. J Clin
39. Chopra V, Engbers FH, Geerts MJ, et al. The Leiden anesthesia simulator. Br Anesth 1995;7(8):675–87.
J Anaesth 1994;73(3):287–92. 47. Good ML, Gravenstein JS, Mahla ME, et al. Can simulation accelerate the
40. Devitt JH, Kurrek M, Cohen M, et al. Testing internal consistency and learning of basic anesthesia skills by beginning anesthesia residents.
construct validity during evaluation of performance in a patient simulator. Anesthesiology 1992;77:A1133.
Anesth Analg 1998;86(6):1160–4. 48. Chopra V, Gesink B, DeJong J, et al. Does training on an anesthesia
41. DeAnda A, Gaba DM. Unplanned incidents during comprehensive simulator lead to improvement in performance? Br J Anaesth 1994;73:293–7.
anesthesia simulation. Anesth Analg 1990;71(1):77–82. 49. Hunt EA, Nelson KL, Shilkofski NA, et al. Performance of crucial
42. DeAnda A, Gaba DM. Role of experience in the response to simulated resuscitation maneuvers during simulated pediatric cardiopulmonary
critical incidents. Anesth Analg 1991;72(3):308–15. arrests: the effect of an educational intervention. Simul Healthc 2006;
43. Schwid H, O’Donnell D. Anesthesiologists’ management of simulated critical 1(2):139 (abstract).
incidents. Anesthesiology 1991;76:495–501. 50. Seymour NE, Gallagher AG, Roman SA, et al. Virtual reality training
44. Botney R, Gaba DM, Howard SK, et al. The role of fixation error in improves operating room performance: results of a randomized, double-
preventing the detection and correction of a simulated volatile anesthetic blinded study. Ann Surg 2002;236(4):458–63.
overdose (abstract). Anesthesiology 1993;79:A1115.
45. Botney R, Gaba DM, Howard SK. Anesthesiologists performance during A complete list of references for this chapter can be found online at
simulated loss of pipeline oxygen (abstract). Anesthesiology 1993;79(A1118). www.expertconsult.com.
Section VII—Computers, Engineering, and the Future 409.e1

References 28. Cheng A, Donoghue A, Gilfoyle E, et al. Simulation-based crisis resource


management training for pediatric critical care medicine: a review for
1. Abrahamson S. An interview of Professor Stephen Abrahamson. Educ Health instructors. Pediatr Crit Care Med 2011;14 [Epub ahead of print].
(Abingdon) 2003;16(3):378–84. 29. Risser DT, Rice MM, Salisbury ML, et al. The potential for improved
2. Smith CC, Huang GC, Newman LR, et al. Simulation training and its effect teamwork to reduce medical errors in the emergency department. Ann
on long-term resident performance in central venous catheterization. Simul Emerg Med 1999;34(3):373–83.
Healthc 2010;5(3):146–51. 30. Wright MC, Taekman JM, Endsley MR. Objective measures of situation
3. Issenberg SB, Chung HS, Devine LA. Patient safety training simulations awareness in a simulated medical environment. Qual Saf Health Care
based on competency criteria of the accreditation council for graduate 2004;13(Suppl 1):165–71.
medical education. Mt Sinai J Med 2011;78(6):842–53. 31. Reason J. The contribution of latent human failures to the breakdown of
4. Bose RR, Matyal R, Warraich HJ, et al. Utility of a transesophageal complex systems. Philos Trans R Soc Lond B Biol Sci 1990;327(1241):
echocardiographic simulator as a teaching tool. J Cardiothorac Vasc Anesth 475–84.
2011;25(2):212–15. 32. Baker DP, Salas E, King H, et al. The role of teamwork in the professional
5. Luciano CJ, Banerjee PP, Lemole Jr GM, et al. Second generation haptic education of physicians: current status and assessment recommendations.
ventriculostomy simulator using the ImmersiveTouch system. Stud Health Joint Commission J Quality Patient Safety 2005;31(4):185–202.
Technol Inform 2006;119:343–8. 33. Hunt EA, Shilkofski NA, Stavroudis TA, et al. Simulation: translation to
6. Banerjee PP, Luciano CJ, Rizzi S. Virtual reality simulations. Anesthesiol Clin improved team performance. Anesthesiol Clin 2007;25(2):301–19.
2007;25(2):337–8. 34. Abrahamson S, Denson JS, Wolf RM. Effectiveness of a simulator in training
7. Kofke WA, Rosen KA, Barbaccia J, et al. The value of acute care simulation. anesthesiology residents. J Med Educ 1969;44:515–19.
W V Med J 2000;96(2):396–402. 35. Berman S. Individual lifetime achievement: Jeffrey B. Cooper, PhD. Joint
8. Peberdy MA, Kaye W, Ornato JP, et al. Cardiopulmonary resuscitation of Commission J Quality Patient Safety 2003;29(12):625–33.
adults in the hospital: a report of 14720 cardiac arrests from the National 36. Directory of Sim Centers. Available at http://ssih.org/directory-of-sim-
Registry of Cardiopulmonary Resuscitation. Resuscitation 2003;58:297–308. centers-2. The Society for Simulation in Healthcare. Accessed June 28, 2012.
9. Herlitz J, Bång A, Alsén B, et al. Characteristics and outcome among patients 37. Howard SK, Gaba DM, Fish KJ, et al. Anesthesia crisis resource management
suffering from in hospital cardiac arrest in relation to the interval between training: teaching anesthesiologists to handle critical incidents. Aviat Space
collapse and start of CPR. Resuscitation 2002;53(1):21–7. Environ Med 1992;63(9):763–70.
10. Adams BD, Zeiler KK, Jackson WO, et al. Emergency medicine residents 38. Good ML, Gravenstein JS. Anesthesia simulators and training devices. Int
effectively direct inhospital cardiac arrest teams. Am J Emerg Med Anesthesiol Clin 1989;27:161–8.
2005;23(3):304–10. 39. Chopra V, Engbers FH, Geerts MJ, et al. The Leiden anesthesia simulator.
11. Foraida MI, Devita MA, Braithwaite RS, et al. Improving the utilization of Br J Anaesth 1994;73(3):287–92.
medical crisis teams (Condition C) at an urban tertiary care hospital. J Crit 40. Devitt JH, Kurrek M, Cohen M, et al. Testing internal consistency and
Care 2003;18:87–94. construct validity during evaluation of performance in a patient simulator.
12. Holmberg M, Holmberg S, Herlitz J, et al. Factors modifying the effect of Anesth Analg 1998;86(6):1160–4.
bystander cardiopulmonary resuscitation on survival in out-of-hospital 41. DeAnda A, Gaba DM. Unplanned incidents during comprehensive
cardiac arrest patients in Sweden. Eur Heart J 2001;22(6):511–19. anesthesia simulation. Anesth Analg 1990;71(1):77–82.
13. Bellomo R, Goldsmith D, Uchino S, et al. A prospective before-and-after trial 42. DeAnda A, Gaba DM. Role of experience in the response to simulated
of a medical emergency team. Med J Aust 2003;179:283–7. critical incidents. Anesth Analg 1991;72(3):308–15.
14. Goldhill DR, Worthington LM, Mulcahy AJ, et al. The patient-at-risk team: 43. Schwid H, O’Donnell D. Anesthesiologists’ management of simulated critical
identifying and managing seriously ill ward patients. Anaesthesia 1999;54: incidents. Anesthesiology 1991;76:495–501.
853–60. 44. Botney R, Gaba DM, Howard SK, et al. The role of fixation error in
15. Buist MD, Moore GE, Bernard SA, et al. Effects of a medical emergency team preventing the detection and correction of a simulated volatile anesthetic
on reduction of incidence of and mortality from unexpected cardiac arrests overdose (abstract). Anesthesiology 1993;79:A1115.
in hospital: preliminary study. Br Med J 2002;324(7334):387–90. 45. Botney R, Gaba DM, Howard SK. Anesthesiologists performance during
16. Petrie D, Lane P, Stewart T. An evaluation of patient outcomes comparing simulated loss of pipeline oxygen (abstract). Anesthesiology 1993;79(A1118).
trauma team activated versus trauma team not activated using TRISS 46. Holzman RS, Cooper JB, Gaba DM, et al. Anesthesia crisis resource
analysis. Trauma and Injury Severity Score. J Trauma Inj Infect Crit Care management: real-life simulation training in operating room crises. J Clin
1996;41(5):870–3. Anesth 1995;7(8):675–87.
17. Wiener EL, Helmreich RL. Cockpit resource management. San Diego, CA: 47. Good ML, Gravenstein JS, Mahla ME, et al. Can simulation accelerate the
Academic Press; 1993. learning of basic anesthesia skills by beginning anesthesia residents.
18. Orlady J. Airline pilot training today and tomorrow, cockpit resource Anesthesiology 1992;77:A1133.
management. In: Weiner EL, Kanki BG, Helmreich RL, editors. Cockpit 48. Chopra V, Gesink B, DeJong J, et al. Does training on an anesthesia
resource management. San Diego: Academic Press; 1995. p. 447–78. simulator lead to improvement in performance? Br J Anaesth 1994;73:
19. Eisen LA, Savel RH. What went right: lessons for the intensivist from the 293–7.
crew of US Airways Flight 1549. Chest 2009;136(3):910–17. 49. Hunt EA, Nelson KL, Shilkofski NA, et al. Performance of crucial
20. Schroeder T, Schierbeck J, Howardy P, et al. Effect of labetalol on CBF and resuscitation maneuvers during simulated pediatric cardiopulmonary
middle cerebral arterial flow velocity in healthy volunteers. Neurol Res 1991; arrests: the effect of an educational intervention. Simul Healthc 2006;
13:10. 1(2):139 (abstract).
21. Risser DT, Rice MM, Salisbury ML, et al. The potential for improved 50. Seymour NE, Gallagher AG, Roman SA, et al. Virtual reality training
teamwork to reduce medical errors in the emergency department. The improves operating room performance: results of a randomized, double-
MedTeams Research Consortium. Ann Emerg Med 1999;34(3):373–83. blinded study. Ann Surg 2002;236(4):458–63.
22. Morey JC, Simon R, Jay GD, et al. Error reduction and performance 51. Rose FD, Attree EA, Brooks BM, et al. Training in virtual environments:
improvement in the emergency department through formal teamwork transfer to real world tasks and equivalence to real task training. Ergonomics
training: evaluation results of the MedTeams project. Health Serv Res 2002; 2000;43(4):494–511.
37(6):1553–81. 52. Eversbusch A, Grantcharov TP. Learning curves and impact of psychomotor
23. Ziewacz JE, Arriaga AF, Bader AM, et al. Crisis checklists for the operating training on performance in simulated colonoscopy: a randomized trial using
room: development and pilot testing. J Am Coll Surg 2011;213(2):212–17. a virtual reality endoscopy trainer. Surg Endosc 2004;18(10):1514–18.
24. Fernandez GL, Lee PC, Page DW, et al. Implementation of full patient 53. Wilhelm DM, Ogan K, Roehrborn CG, et al. Assessment of basic endoscopic
simulation training in surgical residency. J Surg Educ 2010;67(6):393–9. performance using a virtual reality simulator. J Am Coll Surg 2002;195(5):
25. Lambden S, Martin B. The use of computers for perioperative simulation in 675–81.
anesthesia, critical care, and pain medicine. Anesthesiol Clin 2011;29(3): 54. Uchal M, Raftopoulos Y, Tjugum J, et al. Validation of a six-task simulation
521–31. model in minimally invasive surgery. Surg Endosc 2005;19(1):109–16.
26. Mould J, White H, Gallagher R. Evaluation of a critical care simulation series 55. Tuggy ML. Virtual reality flexible sigmoidoscopy simulator training: impact
for undergraduate nursing students. Contemp Nurse 2011;38(1-2):180–90. on resident performance. J Am Board Fam Pract 1998;11(6):426–33.
27. Pascual JL, Holena DN, Vella MA, et al. Short simulation training improves 56. Grantcharov TP, Kristiansen VB, Bendix J, et al. Randomized clinical trial of
objective skills in established advanced practitioners managing emergencies virtual reality simulation for laparoscopic skills training. Br J Surg 2004;
on the ward and surgical intensive care unit. J Trauma 2011;71(2):330–8. 91(2):146–50.
409.e2 Section VII—Computers, Engineering, and the Future

57. Ahlberg G, Hultcrantz R, Jaramillo E, et al. Virtual reality colonoscopy 76. Ali J, Dunn J, Eason M, et al. Comparing the standardized live trauma
simulation: a compulsory practice for the future colonoscopist? Endoscopy patient and the mechanical simulator models in the ATLS initial assessment
2005;37(12):1198–204. station. J Surg Res 2010;162(1):7–10.
58. Gerson LB, Van Dam J. A prospective randomized trial comparing a virtual 77. Shapiro MJ, Morey JC, Small SD, et al. Simulation based teamwork training
reality simulator to bedside teaching for training in sigmoidoscopy. for emergency department staff: does it improve clinical team performance
Endoscopy 2003;35(7):569-75; erratum appears in Endoscopy 2004;36(2):185. when added to an existing didactic teamwork curriculum? Qual Safet Health
59. Howard SK, Gaba DM, Fish KJ, et al. Anesthesia crisis resource management Care 2004;13(6):417–21.
training: teaching anesthesiologists to handle critical incidents. Aviat Space 78. Holcomb JB, Dumire RD, Crommett JW, et al. Evaluation of trauma team
Environ Med 1992;63(9):763–70. performance using an advanced human patient simulator for resuscitation
60. Cooper JB. Are simulation and didactic crisis resource management (CRM) training. J Trauma 2002;52(6):1078–85.
training synergistic? Qual Saf Health Care 2004;13(6):413–14. 79. Holcomb JB, Dumire RD, Crommett JW, et al. Evaluation of trauma team
61. Baker DP, Gustafson S, Beaubien JM, et al. Medical teamwork and patient performance using an advanced human patient simulator for resuscitation
safety: the evidence-based relation. Literature review. AHRQ Publication training. J Trauma 2002;52(6):1078–85.
No. 05-0053; 2005. Available from: http://www.ahrq.gov/qual/medteam. 80. DeVita MA, Schaefer J, Lutz J, et al. Improving medical emergency team
62. Savoldelli GL, Naik VN, Park J, et al. Value of debriefing during simulated (MET) performance using a novel curriculum and a computerized human
crisis management: oral versus video-assisted oral feedback. Anesthesiology patient simulator. Qual Safet Health Care 2005;14(5):326–31.
2006;105(2):279–85. 81. Aggarwal R, Undre S, Moorthy K, et al. The simulated operating theatre:
63. Musacchio Jr MJ, Smith AP, McNeal CA, et al. Neuro-critical care skills comprehensive training for surgical teams. Qual Saf Health Care 2004;
training using a human patient simulator. Neurocrit Care 2010;13(2): 13(Suppl 1):27–32.
169–75. 82. Siassakos D, Fox R, Crofts JF, et al. The management of a simulated
64. Lighthall GK, Barr J, Howard SK, et al. Use of a fully simulated intensive care emergency: better teamwork, better performance. Resuscitation 2011;82(2):
unit environment for critical event management training for internal 203–6.
medicine residents. Crit Care Med 2003;31(10):2437–43. 83. Karkada UH, Adamic LA, Kahn JM, et al. Limiting the spread of highly
65. Hunt EA, Nelson KL, Shilkofski NA. Simulation in medicine: addressing resistant hospital-acquired microorganisms via critical care transfers: a
patient safety and improving the interface between healthcare providers and simulation study. J Intensive Care Med 2011;37(10):1633–40.
medical technology. Biomed Instrum Technol 2006;40(5):399–404. 84. Siassakos D, Bristowe K, Draycott TJ, et al. Clinical efficiency in a simulated
66. Vos GD, Nissen AC, Nieman FH, et al. Comparison of interhospital pediatric emergency and relationship to team behaviours: a multisite cross-sectional
intensive care transport accompanied by a referring specialist or a specialist study. Br J Obstet Gyenaecol 2011;118(5):596–607.
retrieval team. Intensive Care Med 2004;30(2):302–8. 85. Chase JG, Le Compte AJ, Suhaimi F, et al. Tight glycemic control in critical
67. Flabouris A, Runciman WB, Levings B. Incidents during out-of-hospital care—the leading role of insulin sensitivity and patient variability: a review
patient transportation. Anaesth Intensive Care 2006;34(2):228–36. and model-based analysis. Comput Methods Programs Biomed 2011;
68. Lupton BA, Pendray MR. Regionalized neonatal emergency transport. Semin 102(2):156–71.
Neonatol 2004;9(2):125–33. 86. Roberts JA, Kirkpatrick CM, Lipman J. Monte Carlo simulations:
69. Chen JG, Mistry KP, Wright MC, et al. Postoperative handoff maximizing antibiotic pharmacokinetic data to optimize clinical practice for
communication: a simulation-based training method. Simul Healthc 2010; critically ill patients. J Antimicrob Chemother 2011;66(2):227–31.
5(4):242–7. 87. Karon BS, Boyd JC, Klee GG. Glucose meter performance criteria for tight
70. Stocker M, Allen M, Pool N, et al. Impact of an embedded simulation team glycemic control estimated by simulation modeling. Clin Chem 2010;56(7):
training programme in a paediatric intensive care unit: a prospective, 1091–7.
single-centre, longitudinal study. Intensive Care Med 2011 Oct 1 [Epub 88. Sharpe R, Koval V, Ronco JJ, et al. The impact of prolonged continuous
ahead of print]. wakefulness on resident clinical performance in the intensive care unit: a
71. Kohn L, Donaldson M. To err is human: building a safer health system. patient simulator study. Crit Care Med 2010;38(3):766–70.
Washington, DC: National Academy Press, 1999. 89. Brindley PG, Dunn WF. Simulation for clinical research trials: a theoretical
72. Moorthy K, Munz Y, Adams S, et al. Human factors analysis of technical and outline. J Crit Care 2009;24(2):164–7.
team skills among surgical trainees during procedural simulations in a 90. Mistry V. Critical care training: using Twitter as a teaching tool. Br J Nurs
simulated operating theatre. Ann Surg 2005;242(5):631–9. 2011;20(20):1292–6.
73. France DJ, Stiles R, Gaffney EA, et al. Crew resource management 91. Kleinpell R, Ely EW, Williams G, et al. Web-based resources for critical care
training: clinicians’ reactions and attitudes. AORN J 2005:82(2):212–24. education. Crit Care Med 2011;39(3):541–53.
74. Lighthall GK, Barr J, Howard SK, et al. Use of a fully simulated intensive care 92. McGaghie WC, Issenberg SB, Petrusa ER, et al. A critical review of
unit environment for critical event management training for internal simulation-based medical education research: 2003-2009. Med Educ 2010;
medicine residents. Crit Care Med 2003;31(10):2437–43. 44(1):50–63.
75. Blum RH, Raemer DB, Carroll JS, et al. A method for measuring the 93. Frengley RW, Weller JM, Torrie J, et al. The effect of a simulation-based
effectiveness of simulation-based team training for improving training intervention on the performance of established critical care unit
communication skills. Anesth Analg 2005;100(5):1375–80. teams. Crit Care Med 2011;39(12):2605–11.
VII
Chapter
42  

Robotic Telepresence in
Neurocritical Care: The Next
Paradigm in Critical Care
Paul M. Vespa

by an intensivist who can conduct remote rounds has the


Introduction potential to improve patient outcomes.
Timely assessment and treatment of intensive care unit (ICU)
patients is a major goal in intensive care.1,2 This requirement
can be difficult given the shortage of available intensive care
Physician Response Paradigms
physicians and surgeons1 and ICU beds. Delays in both patient Rapid response and implementation of goal-directed treat-
assessment and physician response can result in lost opportu- ment improve outcomes for critically ill patients with sepsis.18
nities to improve patient outcome and can result in increased Physician attention to patients is divided into prospective
morbidity and length of stay.3 These problems particularly and responsive attention.19 In most neurointensive care units
affect neurocritical care patients, because the dichotomy (NICU), prospective physician attention is conducted on
between the need for rapid intervention and the shortage of morning bedside rounds. Morning bedside rounds are made
highly trained experts is most acute.4 Most neurocritical care to evaluate the patient’s clinical condition (the physical exam-
patients are at high risk for further secondary brain injury ination), and to evaluate the laboratory studies, brain images,
such as brain ischemia or elevated intracranial pressure (ICP),5 and other data that accumulate over time. Morning rounds in
and the time window for intervention is quite short. Hence the NICU are a mixture of retrospective analysis of trends in
there is a great need to quickly identify and rapidly respond data and prospective planning of treatment for the day. These
to secondary brain insults once they occur. However, there is rounds are usually based on scheduled periods of time and
a nationwide shortage of neurointensivists. The use of infor- result in a well-formulated “plan of attack” for the day, which
mation technology may hold the answer to facilitate this goal likely will change several times during the course of the day
of quick diagnosis and rapid response despite the workforce based on intercurrent changes in patient condition. For a
shortage.6 typical brain injury patient with ICP monitoring, it has been
This chapter outlines the role of information technology to calculated that the intensivist evaluates more than 650 data
change the paradigm for patient care using robotic telepres- points during each episode of bedside rounds.2 These data
ence (RTP). Robots have been used increasingly in a variety points consist of hourly vital signs, waveform morphology
of surgical procedures, in large part to augment surgical visu- values, brain monitoring values such as brain tissue oxygen
alization, dexterity, and precision, although the effect on and microdialysis, radiologic studies, clinical examination
patient outcome and cost efficacy rather than on surrogate findings, laboratory data, verbal story-line information, and
endpoints is still debated.7-15 In recent years there also has been visual-cue information from body language and appearance.
a rapid expansion of ICU telemedicine that combines audio- These data points involve integration of graphic information,
visual technology with the electronic medical record to provide longitudinal trends of data, and cross-sectional review of data
critical care from a remote location.16 This includes continu- at fixed points in time. This analysis is done prospectively
ous offsite monitoring for the entire ICU or all ICUs in a during morning rounds to make a reasoned clinical treat-
hospital,17 or as is the focus of this chapter, targeted individual ment plan.
patient evaluation. This chapter outlines how RTP works, how Physicians also respond to changes in the clinical condition
it is used, how it impacts patient care, and how developers of the patient by being paged by the nurse or other health care
anticipate it will be used in the future in the neurointensive professionals. This can be called an urgent or emergent response,
care unit. There are essentially two components to the RTP and occurs throughout the day and night. During such an
system: the robot and the computerized information system. urgent response, the physician often needs to consider the
These two elements have synergy and facilitate high-quality, same set of data points and to determine the evolution
timely clinical care. RTP may represent the next clinical care of the patient over time. However, the physician may not be
paradigm in the NCCU. This approach, perhaps through at the bedside when paged and may not have all these data
implementation of evidence-based practice in the ICU applied points available to him or her; yet he or she still is required to
410 © Copyright 2013 Elsevier Inc. All rights reserved.
Section VII—Computers, Engineering, and the Future 411

make a clinical decision.1,4 The response is by telephone and, broadband connection. Security protocols enable Health
at best, a delayed face-to-face evaluation of the patient, often Insurance Portability and Accountability Act (HIPAA) com-
hours later. This means at the time of the initial call, the physi- pliant audiovisual communication across the public internet,
cian may not have access to visual, graphic, or waveform infor- via a hardwire or cellular connection.
mation, and perhaps the electronic medical records or From the control station, the health care professional can
electronic charting data. Thus the physician is at a disadvan- drive the robot throughout the facility and communicate in
tage in processing the clinical information and reaching a real time with patients, their families, and medical staff. The
clinical decision during the time of urgent patient need. This RP-7 is 5.4 feet tall, weighs 220 lb, and travels up to 2 miles
disadvantage may result in poor clinical judgment or medical per hour. The anthropomorphic design consists of a “head,”
errors. Medical errors can occur when incomplete informa- which contains a flat screen monitor, microphone, and digital
tion is presented and/or verbal information alone is used.3 camera with pan, tilt, and 10× zoom features. The central body
One way to avoid medical errors is to increase intensivist houses the speaker, robot-side volume control, and expansion
coverage in the ICU.1 Because there is a shortage of intensiv- bay that can include a handset for private conversation, a
ists, it is difficult to increase the number of intensivists, per se. printer for physician’s notes, a digital stethoscope, and other
However, using RTP technology, one may be able to increase video inputs. The control station has a share-and-display
the percentage of time in which the intensivist is telepresent feature that permits images on the control station to be dis-
in the ICU. As discussed later in this chapter, robotic telepres- played remotely on the head/face of the robot, visible to the
ence can help reduce or avoid medical errors. audience in the ICU (see Fig. 42.2). The robot rides on a

Robotic Telepresence Technology


InTouch Technologies Inc. (InTouch Health), a privately held
company based in Santa Barbara, CA, manufactures the
robotic telecommunication system currently in use in the
author’s ICU.2 The company has pioneered Remote Presence
technology for hospital providers. Through its RP-7 Remote
Presence Robotic System, a proprietary mobile robotic and
communications platform, health care professionals are able
to consult with hospital-based patients and staff more easily
and frequently. The InTouch Health solution leverages the
time and expertise of health care professionals across multiple
care facilities to improve the efficiency and effectiveness of
care delivery. The RP-7 Robotic System enables “remote pres-
ence” that allows individuals to project themselves from one
geographic location to another, via the wireless mobile robot,
such that they can interact from that remote location.
There are two main components to the Remote Presence
System: the RP-7 Robot (Fig. 42.1) and control station (Fig.
42.2). The technology is dependent on the use of broadband
internet connectivity and existing wireless network within the A B
hospital. Most hospitals already have the necessary network
Fig. 42.1  Telepresence robot (Intouch Technologies, Santa Barbara,
infrastructure in place. The system incorporates two-way
CA). A, the frontal view of the robot, with head and neck consisting of
audio and video that permits observation and face-to-face a flatscreen TV, and mobile neck device enabling 340-degree head
interaction between health care professionals and patients. movement, and attached camera system for wide view and focused
The robot operates in the hospital environment on an 802.11 views. Sensors along the base are designed to override manual controls
Wi-Fi network. It is controlled remotely by a health care pro- and enable collision avoidance. B, Rear projection of the robot showing
fessional using a laptop or desktop computer, called a control portals for digital and video diagnostic equipment, such as digital
station, which can be linked to the Internet through secure stethoscope.

Fig. 42.2  Desktop physician control station. The


control station contains software, camera, microphone,
and digital Internet connection system, as well as a control
stick device. The physician drives the robot like playing a
video game. The double screen facilitates simultaneous
display of radiology and laboratory images and the intensive
care unit environment.
412 Section VII—Computers, Engineering, and the Future

A B C
Fig. 42.3  Sample case display. These are picture files taken using the desktop control station of a new patient admission to the ICU. A, A
neurointensive care fellow is presenting the patient to the attending who is running the robot at 21:00. B, The appearance of a hard-copy computed
tomography scan, brought with the patient from an outside hospital, showing the Fisher grade 3 subarachnoid hemorrhage. C, The bloody output
from the ventriculostomy catheter 2 hours later, indicating release of blood via the ventriculostomy catheter. Note the intravenous pump display to
the left, indicating infusion of 3% sodium chloride.

patented holonomic platform about 20 inches wide, made up visual information was highlighted.20 The main types of criti-
of three balls that allow it to move in any direction, thus cal data acquired from the telemedicine interaction were
greatly increasing maneuverability in tight spaces. An array of judged by the attending physician to be important in the deci-
infrared proximity sensors placed around the lower section of sion pathway. The distribution of these critical data is as
the robot prevents it from colliding with people or objects. follows: 67% visual and 33% verbal information. Of the visual
The robot is powered by a rechargeable battery with a life of information, the breakdown was as follows: physical examina-
5 to 6 hours depending on usage. The robot moves from tion (40.6%), physiologic monitor graphics (11.5%), printed
bedside to bedside under the control of the physician, is information from the medical record (5.8%), body language
capable of traveling on elevators, and has been safely used in of the nurse (5.7%), information about a tube or catheter
operating and procedure rooms. Direct feeds from clinical (3.8%). In a randomized controlled trial of telemedicine for
information systems, such as digital radiology systems and lab acute ischemic stroke, telemedicine provided important visual
systems, are routinely done using the doctor’s workstation. At and verbal information, improved diagnostic accuracy, and
UCLA, caregivers use the Global Care Quest (GCQ, Mission enabled more complete data assessment as compared with the
Viejo, CA) clinical information system to view all types of telephonic paradigm.21 Similar reliance on visual and graphic
electronic medical record data. In this fashion, the physician information has been reported using the electronic-ICU
can discuss cases with colleagues, evaluate radiology, and eval- system (Visicu, Baltimore, MD).22 These data speak to the
uate the bedside environment (Fig. 42.3). need for visual information to be part of the physician response
HIPAA compliance and consent for telemedicine is required paradigm.
for any system.6,20 There are formal consent forms for all ICU
procedures, including RTP. Information technology and com-
pliance officer review of RTP is done routinely at centers that Uses of Robotic Telepresence
implement the technology; acceptance of RTP technology has
been uniform across various regions of the country. Tele- Rapid Response to Unstable Intensive Care
medicine has enjoyed a robust experience with medical-legal Unit Patient Condition
circles. Increased physician participation and enhanced data The RTP system enables the clinician to make a rapid response
review actually makes medical-legal positions better com- to a sudden deterioration in patient condition. In a recent
pared with telephonic communication. Multiple systems can study, Vespa et al20 demonstrated a marked reduction in physi-
be used to provide medical documentation and computer cian response time for a face-to-face evaluation of the patient.
order entry, depending on the particular hospital. A separate Overall there was a marked reduction in the attending physi-
internal documentation system, called Stroke-Respond, is cur- cian response latency using RTP. The mean response time was
rently available, and can integrate into a hospital-based elec- significantly shorter for patients with brain ischemia (7.8 ± 2.8
tronic medical record system for transparency. In July 2008, vs. 152 ± 85 minutes) and elevated intracranial pressure (11 ±
Medicare initiated special ICU billing codes for telemedicine, 14 vs. 108 ± 55 minutes) (P < .001). Univariate analysis dem-
called 0188T and 0189T. These codes are currently under onstrated a significant reduction in the response latency for
study by Medicare. each category and for the composite overall (P < .001). Figure
42.4 illustrates a comparison of face-to-face response times
using the RTP system by specific categories.
Changing the Physician The reduction in response time to elevated ICP and brain
ischemia is clinically meaningful in that the time window to
Response Paradigm intervention is quite short. For example, customary time for
The response to an urgent condition using RTP makes use of an attending physician to visually see a patient with elevated
visual information and real-time audiovisual communication ICP averaged 108 minutes. With the use of RTP, the mean
with the bedside nurse. In a recent study, the importance of time to a face-to-face contact was about 11 minutes. This
Section VII—Computers, Engineering, and the Future 413

reduction in latency is substantial and clinically meaningful would have similar overall benefits. Other telemedicine
because the duration of untreated, elevated ICP can be a systems have reported improved outcomes for critically ill
major determinant factor for morbidity and mortality after patients.5
traumatic brain injury. There was a similar reduction in
latency for other paroxysmal events that could contribute
irreversible brain damage, such as brain ischemia, seizures, Prospective Compliance Monitoring
hypotension, and hypoxemia. This reduction was mirrored by and Mentoring
a reduction in the length of stay in those patients who are A fundamental principal of safety in the ICU is compliance
most at risk for brain ischemia, namely those with subarach- and close mentoring of less experienced practitioners by their
noid hemorrhage, stroke, and brain trauma. The impact of senior colleagues.26 This requires presence in the ICU by the
rapid response on patient outcome remains untested. However, senior practitioner. The RTP system enables the clinician to
the concept of rapid response to patient instability has been make rounds during night hours and during times when tra-
found to reduce mortality overall in general critical care,22-25 ditionally the physician is not in the ICU, and facilitates face-
suggesting that special methods to facilitate rapid response to-face discussions with simultaneous evaluation of imaging
and laboratory values. This enables physicians to supervise
directly the ongoing care of the patient and to enable mentor-
ing of the bedside nurse. In a recent study, such compliance
monitoring and mentoring accounted for 12% of all encoun-
Comparison of Response Time ters, and together with avoidance of medical errors, accounted
350 for 20% of all encounters with the robot. Figure 42.5 illus-
300 trates a simultaneous evaluation of an ischemic limb with
Doppler ultrasound of the dorsalis pedis artery. The mutual
250
simultaneous evaluation and treatment of patients, with both
Minutes

200 nurse and remote doctor acting together, facilitates patient


150 care. In addition, there are a number of groups that are using
100
the robot to enhance mentoring of inexperienced nurses by a
senior nurse educator. This provides the bedside nurse with
50 an approachable person with whom he or she can discuss
0 ongoing nursing issues that would otherwise not be discussed
with the physician. The senior nurse educator has the poten-
ia

P
s

ia
on

es
ia

um
ia

IC
em

tial to staff numerous hospitals and ICUs from a central loca-


em
ur

ur
si

hm

iri
lig
en

ed

iz
ox

ch

el
yt

Se

tion. Similar telementoring, teleproctoring, and telestration


O
ot

D
at
yp

is
rh
yp

ev

n
H

Ar

systems also have been used to enable trainees and mentors to


ai
H

El

Br

remotely observe surgical cases in the operating room or


Actual response time provide intraoperative consultation by other surgeons at
Customary response time remote sites.27-29 These systems thus can enhance education
Fig. 42.4  Face-to-face response time using the robot (solid bars)
and mentoring in the ICU.
compared with the customary face-to-face response time using
the traditional in-person paradigm. The responses for a number of
neurologic emergencies are individually shown, with mean and standard
Improving Efficiency of Care
deviation data between 5 and 20 cases per category. ICP, Intracranial Given the universal shortage of hospital and intensive care
pressure. unit beds, a significant focus on improving efficiency of care

A B
Fig. 42.5  Nurse mentoring examples: A, An example of physician-operated robot discussion with a nurse at the patient’s bedside. Review of laboratory
and imaging data are being simultaneously done, and discussion of results and formulation of treatment plans are occurring. B, An example of
mentoring of a nurse while examining an ischemic limb using physical exam (note dusky appearance of dorsal leg) and Doppler ultrasound of the
dorsalis pedis. Clinical decision making and confirmation of the nurse’s diagnosis of limb ischemia were made.
414 Section VII—Computers, Engineering, and the Future

and bed allotment is underway. The robot may be a mecha-


nism to facilitate improved efficiency of care in the same way
that it is used to improve compliance with treatment proto-
cols. This concept has been studied in the postoperative
setting, in which clinical pathways have been established to
facilitate quality of care and expedite efficiency of the hospital
stay. Gandsas and colleagues30 recently presented results from
a retrospective trial of RTP versus traditional rounding on
routine postoperative patients in a gastric bypass surgery
practice. In this study, patients were very satisfied with the
telerounding concept, and postoperative morbidity was not
increased. The length of stay was reduced with the use of
robotic telerounding. The attending surgeon was able to
increase the time spent doing office and operating room work A
because of better efficiency in the rounding process. This
enabled an overall improvement in physician workflow and
resulted in overall hospital and physician profit.
In the UCLA study,20 a similar improvement in efficiency
was found for an academic neurointensive care unit. In this
study, a comparison of the length of stay and costs associated
with the primary diagnoses in a neurointensive care unit were
examined 1 year before implementing the robot and com-
pared with the initial year of routine robot use. The diagnoses
were subarachnoid hemorrhage, brain trauma, intracerebral
hemorrhage, brain tumors, and ischemic stroke. The use of
the robot was associated with a substantial reduction in mean
length of stay for most diagnoses except brain tumors, and a
net cost savings of more than $1.1 million during that year.
B
Supervision and Mentoring for Invasive
Fig. 42.6  A, Mentoring of a house staff member in the performance of
Procedures in the Intensive Care Unit an external ventriculostomy catheter insertion using robotic telepresence.
Because there is a shortage of neurosurgeons and intensivists, The robotic physician was able to determine and guide the trajectory
invasive procedures may be delayed or not done. Robotics angle for drilling the hole and for catheter insertion. B, Mentoring of a
provides an opportunity for telementoring and teleproctor- nurse practitioner inserting a radial arterial line. The angle of needle
ing, although there is very limited study of this concept in the approach, as well as needle depth, was discussed in real time and the
NCCU.31 In a pilot study, the ability to supervise invasive mentoring suggestions were provided in order to get the ideal arterial
procedures using RTP has been demonstrated. This experi- blood jet flow, before attempting to place the guidewire and catheter.
ence mirrors that of general surgeons who routinely supervise
laparoscopic surgery of junior colleagues. In the author’s
NICU, there have been 40 invasive procedures using RTP to
mentor junior house staff in a pilot protocol. Under this para- design remains to be defined.32,35-38 In the near future, robots
digm, the attending specialist is able to watch the procedure, may help establish a reproducible method to quantitate pro-
provide real-time feedback to the junior physician, and coach cedural performance and so help assess surgical or critical care
the entire procedure. There is no delay in image acquisition trainees. Robots also may be adapted to a hybrid operating
or audiovisual communication. Using this paradigm, 97% of room (radiology suite) to augment neuroendovascular proce-
procedures were successfully completed, while one procedure dures and integrate various imaging modalities.39 This may
was aborted because of inability to gain venous access using become of particular relevance to the NCCU and performance
the technique. Importantly, 60% of these procedures were of thrombolytic procedures for stroke.
judged to be emergent and provided stabilizing or life-saving
care to the patient. An example case of ventriculostomy place-
ment in a patient with traumatic brain injury and elevated Emergency Department Triage
ICP is shown in Figure 42.6. Arterial access with visualization and Consultation
of pulsatile blood flow is appreciated through this system. There is a growing experience with telemedicine in the emer-
More recently studies have addressed the prospect of semi- gency department setting for neurocritical care consulta-
autonomous surgery and specifically whether a robotic system tions.21,40-43 Telemedicine enables rapid and more accurate
is capable of performing a burr hole procedure in specific assessment of stroke patients, enables early decisions about
locations and along predetermined trajectories to a precise administration of tissue plasminogen activator, and facilitates
depth.32,33 The robot, however, does not replace a health care early goal-directed treatment that enhances cerebral blood
provider and cannot be performed without a human being flow and avoids hemorrhagic complications. Use of telemedi-
present.34 In addition, whether robot-assisted surgery in neu- cine for acute stroke can result in improved outcomes for large
rosurgery improves upon surgical outcome and optimal populations, both in rural and urban settings. A number of
Section VII—Computers, Engineering, and the Future 415

academic and private organizations have been formed to the potential to achieve a new paradigm in patient care and at
enable delivery of acute stroke care to underserved regions. the same time make the physician more mobile and less
This technology enables accurate triage of patients and enables hospital-bound. The concept of changing the paradigm of
appropriate transfers of complex patients to tertiary care delivering critical care is penetrating the collective conscious-
centers in a hub and spoke pattern.44,45 ness,47 although barriers such as the perceived ease of use
A similar experience has been demonstrated in emergency and complexity, perceived usefulness, perceived behavioral
neurosurgical evaluation and treatment.46 In this study, the control, and ethical and legal dilemmas may affect adaptation
use of a videoconferencing technology was associated with a of robotic telepresence in the ICU.48,49 With robotic telepres-
reduction in mortality using a combination of teleradiology ence, there is the potential to deliver the right care, right away,
and videoconferencing, compared with telephone-only con- to neurocritical care patients.
sultation (25% vs. 35%, P < 0.025). This study did not use the
robotic device, but used the Polycom view station (Polycom References
Inc, San Jose, CA). However, there was a high failure rate, 30%
of all cases, for this videoconferencing device, during this 1. Angus DC, Shorr AF, White A, et al, Committee on Manpower for
Pulmonary and Critical Care Societies (COMPACCS). Critical care delivery
study. Furthermore, these authors found that the teleradiology in the United States: distribution of services and compliance with Leapfrog
component had the largest impact on mortality. Thus atten- recommendations. Crit Care Med 2006;34:1016–24.
tion to the specific videoconferencing technology appears to 2. Vespa P. Multimodality monitoring and telemonitoring in neurocritical care:
be warranted. In contrast, in a 4-year experience with the from microdialysis to robotic telepresence. Curr Opin Crit Care 2005;11:
133–8.
Telepresence Robot, the failure rate has been less than 1%. 3. Heffner JE, Ellis R, Zeno B. Safety in training and learning in the intensive
In the Neurocritical Care Robotic Telepresence Network, care unit. Crit Care Clin 2005;21:129–48.
there is connectivity to multiple community-based hospitals 4. Pronovost PJ, Thompson DA, Holzmueller CG, et al. The organization of
and UCLA-affiliated hospitals. Triage in the emergency depart- intensive care unit physician services. Crit Care Med 2007;35:2256–61.
ment, with continuity of care in the ICU has become a very 5. Vespa PM. The implications of cerebral ischemia and metabolic dysfunction
for treatment strategies in neurointensive care. Curr Opin Crit Care 2006;12:
useful model. The most recent development of the robotic 119–23.
telepresence system is the feature called Multi-Presence. Multi- 6. Cummings J, Krsek C, Vermoch K, et al, University HealthSystem
Presence enables multiple physicians and nurses to be engaged Consortium ICU Telemedicine Task Force. Intensive care unit telemedicine:
simultaneously through the robot with the patient, and to review and consensus recommendations. Am J Med Qual 2007;22:239–50.
7. Barbash GI, Glied SA. New technology and health care costs: the case of
communicate with each other at the same time. This enables robot-assisted surgery. N Engl J Med 2010;363:701–4.
three- and four-way discussion groups and enables collective 8. Maeso S, Reza M, Mayol JA, et al. Efficacy of the Da Vinci Surgical System in
problem solving (Fig. 42.7). abdominal surgery compared with that of laparoscopy: a systematic review
and meta-analysis. Ann Surg 2010;252:254–62.
9. Barnett JC, Judd JP, Wu JM, et al. Cost comparison among robotic,
Conclusion laparoscopic, and open hysterectomy for endometrial cancer. Obstet Gynecol
2010;116:685–93.
RTP is a powerful tool to facilitate patient care. The health 10. Maan ZN, Gibbins N, Al-Jabri T, et al. The use of robotics in
care and ICU world is changing, with a 24-7-365 mentality otolaryngology-head and neck surgery: a systematic review. Am J
for access to information and discussion. Seemingly, the cur­ Otolaryngol 2011; Jun 7 [Epub ahead of print].
11. Kowalczyk KJ, Yu HY, Ulmer W, et al. Outcomes assessment in men
rent traditional paradigm for patient care, with intermittent undergoing open retropubic radical prostatectomy, laparoscopic radical
rounds and telephone-based care, is being challenged by the prostatectomy, and robotic-assisted radical prostatectomy. World J Urol
demands to be omnipresent and always available. RTP has 2011; Mar 2 [Epub ahead of print].
12. Atluri P, Woo YJ. Minimally invasive robotic mitral valve surgery. Expert Rev
Med Devices 2011;8(1):115–20.
13. Antoniou GA, Riga CV, Mayer EK, et al. Clinical applications of robotic
technology in vascular and endovascular surgery. J Vasc Surg 2011;53(2):
493–9.
14. Khatlani K, Sharma S, Mendoza PJ, et al. The current state of robot assisted
radical prostatectomy. Minerva Urol Nefrol 2010;62(2):193–201.
15. Hellenthal NJ, Hussain A, Andrews PE, et al. Surgical margin status after
robot assisted radical cystectomy: results from the International Robotic
Cystectomy Consortium. J Urol 2010;184(1):87–91.
16. New England Healthcare Institute. Tele-ICUs: remote management in
intensive care units. Cambridge, Mass: Massachusetts Technology
Collaborative and Health Technology Center; 2007.
17. Thomas EJ, Lucke JF, Wueste L, et al. Association of telemedicine for remote
monitoring of intensive care patients with mortality, complications, and
length of stay. JAMA 2009;302(24):2671–8.
18. Rivers E, Nguyen B, Havstad S, et al, Early Goal-Directed Therapy
Collaborative Group. Early goal-directed therapy in the treatment of severe
sepsis and septic shock. N Engl J Med 2001;345:1368–77.
19. Pronovost PJ, Angus DC, Dorman T, et al. Physician staffing patterns and
clinical outcomes in critically ill patients: a systematic review. JAMA 2002;
288:2151–62.
Fig. 42.7  Multipresence. Multiple users are able to see the patient via
20. Vespa, P, Miller, CM, Hu, X, et al. Intensive care unit robotic telepresence
the robot as well as see each other and have a group conversation facilitates rapid physician response to unstable patients and decreased cost in
simultaneously. Two consultant physicians are shown to the far left of neurointensive care. Surg Neurol 2007;67:331–7.
the image, and the attending is shown in the bottom screen evaluating 21. Meyer BC, Raman R, Hemmen T, et al. Efficacy of site-independent
the patient, who is shown in the large center screen. All participants can telemedicine in the STRokE DOC trial: a randomised, blinded, prospective
communicate with each other in real time. study. Lancet Neurol 2008;7:787–95.
416 Section VII—Computers, Engineering, and the Future

22. Breslow MJ, Rosenfeld BA, Doerfler M, et al. Effect of a multiple-site 36. Arata J, Tada Y, Kozuka H, et al. Neurosurgical robotic system for brain
intensive care unit telemedicine program on clinical and economic tumor removal. Int J Comput Assist Radiol Surg 2011;6(3):375–85.
outcomes: an alternative paradigm for intensivist staffing. Crit Care Med 37. Alric M, Chapelle F, Lemaire JJ, et al. Potential applications of medical and
2004;32:31–8. non-medical robots for neurosurgical applications. Minim Invasive Ther
23. Ranji SR, Auerbach AD, Hurd CJ, et al. Effects of rapid response systems on Allied Technol 2009;18(4):193–216.
clinical outcomes: systematic review and meta-analysis. J Hosp Med 2007;2: 38. Pandya S, Motkoski JW, Serrano-Almeida C, et al. Advancing neurosurgery
422–32. with image-guided robotics. J Neurosurg 2009;111(6):1141–9.
24. Rosenfeld BA, Dorman T, Breslow MJ, et al. Intensive care unit telemedicine: 39. Murayama Y, Irie K, Saguchi T, et al. Robotic digital subtraction angiography
alternate paradigm for providing continuous intensivist care. Crit Care Med systems within the hybrid operating room. Neurosurgery 2011 Jan 26 [Epub
2000;28:3925–31. ahead of print].
25. Winters BD, Pham JC, Hunt EA, et al. Rapid response systems: a systematic 40. Audebert HJ, Schenkel J, Heuschmann PU, et al, Telemedic Pilot Project for
review. Crit Care Med 2007;35:1238–4. Integrative Stroke Care Group. Effects of the implementation of a
26. Hussain, K. Medication safety and transfusion errors in the ICU and beyond. telemedical stroke network: the Telemedic Pilot Project for Integrative Stroke
Crit Care Clin 2005;21:91–110. Care (TEMPiS) in Bavaria, Germany. Lancet Neurol 2006;5:742–8.
27. Smith CD, Skandalakis JE. Remote presence proctoring by using a 41. Choi JY, Porche NA, Albright KC, et al. Using telemedicine to facilitate
wireless remote-control videoconferencing system. Surg Innov 2005;12(2): thrombolytic therapy for patients with acute stroke. Jt Comm J Qual Patient
139–43. Saf 2006;32:199–205.
28. Agarwal R, Levinson AW, Allaf M, et al. The RoboConsultant: telementoring 42. Schwab S, Vatankhah B, Kukla C, et al, TEMPiS Group. Long-term
and remote presence in the operating room during minimally invasive outcome after thrombolysis in telemedical stroke care. Neurology 2007;69:
urologic surgeries using a novel mobile robotic interface. Urology 2007; 898–903.
70(5):970–4. 43. Hess DC, Wang S, Gross H, et al. Telestroke: extending stroke expertise into
29. Lai F. Robotic telepresence for medical training and education. Stud Health underserved areas. Lancet Neurol 2006;5:275–8.
Technol Inform 2009;142:151–2. 44. Lai F. Stroke networks based on robotic telepresence. J Telemed Telecare
30. Gandsas A, Parekh M, Bleech MM, et al. Robotic telepresence: profit analysis 2009;15(3):135–6.
in reducing length of stay after laparoscopic gastric bypass. J Am Coll Surg 45. Jabbour P, Gonzalez LF, Tjoumakaris S, et al. Stroke in the robotic era. World
2007;205(1):72–7. Neurosurg 2010;73(6):603–4
31. Ereso AQ, Garcia P, Tseng E, et al. Usability of robotic platforms for remote 46. Wong HT, Poon WS, Jacobs P, et al. The comparative impact of video
surgical teleproctoring. Telemed J E Health 2009;15(5):445–53. consultation on emergency neurosurgical referrals. Neurosurgery 2006;59:
32. Louw DF, Fielding T, McBeth PB, et al. Surgical robotics: a review and 607–13.
neurosurgical prototype development. Neurosurgery 2004;54(3):525–37. 47. Groves Jr RH, Holcomb Jr BW, Smith ML. Intensive care telemedicine:
33. Brodie J, Eljamel S. Evaluation of a neurosurgical robotic system to make evaluating a model for proactive remote monitoring and intervention in the
accurate burr holes. Int J Med Robot 2011;7(1):101–6. critical care setting. Stud Health Technol Inform 2008;131:131–46.
34. Stüer C, Ringel F, Stoffel M, et al. Robotic technology in spine surgery: 48. Benmessaoud C, Kharrazi H, MacDorman KF. Facilitators and barriers to
current applications and future developments. Acta Neurochir Suppl 2011; adopting robotic-assisted surgery: contextualizing the unified theory of
109:241–5. acceptance and use of technology. PLoS One 2011;6(1):e16395.
35. Le Roux P, Das H, Esquenazi S, et al. Robot-assisted microsurgery: feasibility 49. Mavroforou A, Michalodimitrakis E, Hatzitheo-Filou C, et al. Legal and
in a rat microsurgical model. Neurosurgery 2001;48:584–9. ethical issues in robotic surgery. Int Angiol 2010;29(1):75–9.
Chapter
43  
VII

Information Processing, Data


Acquisition, and Storage
Per Enblad, Ian Piper, and Richard O. Sinnott

Introduction disorders that are treated there and so help design new treat-
Neurocritical care monitoring generates enormous amounts ment strategies.2
of various kinds of data from multiple sources. The informa- There is accumulating evidence that demonstrates that IT
tion obtained serves the purpose of detecting secondary can improve patient health care. Bates and Gawande describe
brain insults and guiding the immediate management of the trials in which technology was associated with a reduction in
individual patient. Stored data also may be used for quality medication errors and errors related to transitions of care, and
assurance and to capture the intracenter and intercenter in which technology aided in earlier detection of adverse
management variation that can be fundamental to being able events. For example, Kupermann et al.,4 in a randomized con-
to show positive effects in phase 3 trials. Furthermore, the trolled trial of technology for early detection of adverse events,
collection and storage of monitoring data into databases showed a reduction in time to treatment of 11% and a 29%
fosters research, for example, defining critical threshold levels reduction in the duration of dangerous conditions to patients.
for managing raised intracranial pressure (ICP). Such re- Rosenfield5 conducted a study of IT-based remote monitoring
search may increase the understanding of secondary brain of a 10-bed intensive care unit (ICU) and reported a reduction
injury mechanisms and define the proper use and value of in mortality of greater than 40% and a reduced length of stay
new monitoring techniques. Ultimately, this may lead both of 30% compared with historical controls.
to better detection of dangerous secondary brain insults and Critics of this type of research point out that better resolu-
to new treatment approaches. If the future expectations of tion of events is of no value unless their direct management
neurocritical care and multimodality monitoring are to be influences clinical outcome. Single-center studies with small
realized, it is essential to improve information processing and numbers of patients are not suited to answer such questions,
to standardize data acquisition storage and analysis of data. and multicenter randomized controlled trials to validate
In these aspirations, information technology (IT) plays a IT-driven management are not readily funded nor easily justi-
crucial role. fied as a research priority. Paradoxically, the patient popula-
tions that may benefit most from better IT-based event
detection, such as the brain injury population, are the most
Information Technology challenging in which to conduct a controlled management
IT is defined by the Information Technology Association of trial because of continuing intercenter management varia-
America (ITAA) as “the study, design, development, imple- tion6,7 fostered in large part by a lack of evidence for any type
mentation, support or management of computer-based infor- of effective therapy.8
mation systems, particularly software applications and Nevertheless, indirect evidence is available and continues to
computer hardware.” It deals with the use of electronic com- become available. There are increasing numbers of reports
puters and computer software to convert, store, protect, that indicate the importance of providing specialist neuro-
process, transmit, and securely retrieve information. Infor- critical care in the management of traumatic brain injury
matics in the neurosciences was first applied to basic science (TBI) patients.9-11 For example, the report by Patel9 of 2300
research but there now are several large databases, such as the patients treated in nonneurosurgical hospitals showed a 2.15-
Alzheimer’s Disease Neuroimaging Database (ADNI) or the fold increase in the odds of death compared with those treated
Glioma Molecular Diagnostic Initiative (GMDI), that provide in a neurosurgical center. These studies do not indicate which
researchers in any location open access to data that can drive aspects of critical care management are key, but management
research forward.1 It is clear that the computer science expert (whether surgically or medically focused) aimed at earlier
also must be included in the neurointensive care team today detection and treatment of adverse events must in part be
and in the future. The application and development of IT in responsible. In support of this and despite criticism from
the neurocritical care unit (NCCU) is the challenge faced to enthusiasts of evidence-based medicine (EMB), neuroin­
further develop neurocritical care. In particular, informatics tensive care centers with a track record in the aggressive man-
in the NCCU can be leveraged to help provide insight into the agement of secondary insults continue to report improvements
© Copyright 2013 Elsevier Inc. All rights reserved. 417
418 Section VII—Computers, Engineering, and the Future

in outcome statistics of patients compared with historical infrastructure aimed at improving the standards for multi-
controls.12 center studies of monitoring and managing TBI patients
The Brain Monitoring with Information Technology during their acute stay in intensive care. (These same princi-
(BrainIT) group has been a strong proponent for adopting IT ples can be adapted to patients with other disorders admitted
methods for the early detection and management of second- to the NCCU.) It was agreed that a different approach was
ary insults in patients with brain injury (www.brainit.org). needed, one that focused on using IT-based methods to
Analyses in progress from this group on the minute-by- increase the resolution of data capture and the quality and
minute physiologic data of 200 head-injured patients obtained validation of data captured. The pervasive nature of the
from 22 neurointensive care centers across Europe also may Internet and extensive use by clinicians of email systems fos-
indicate that more complex summary measures, such as the tered the creation of such a network as an Internet-based
Pressure-Time Index,13 better relate to clinical outcome than e-Infrastructure: the BrainIT group.15
do simple measures such as the mean, median, or total dura- The BrainIT group works collaboratively to develop stan-
tion of insult burden. dards to collect and analyze of data from brain-injured patients
Researchers continue to hope for definitive controlled trial toward providing a more efficient infrastructure to assess new
evidence that IT-led management yields improved patient health technology. In several international meetings, the group
outcome, but experience to date of funding and conducting has defined a core dataset designed to be collected using
such studies is limited. Perhaps research time and funds are PC-based tools and to provide a common minimal dataset for
better spent on conducting trials of new forms of manage- all studies, regardless of the underlying research question. This
ment without the need to trial new health care support systems data definition period was funded as part of a European Com-
as well? However, to demonstrate benefit of a therapy requires munity study (QLGT-2000-00454).16 The meetings brought
that all other aspects of management be homogenous. In together clinical and scientific experts from the domain of TBI
conditions as complex as TBI and subarachnoid hemorrhage, basic research and of multicenter clinical trials, such as the
there remain many opinions as to what constitutes “standard” European Brain Injury Consortium,17 as well as representa-
or “optimal” management, and without information about tives from the medical device and pharmaceutical industries.
this, even effective therapies may appear to be ineffective in A series of meetings and workshops conducted over 1 year
clinical practice because of variation in care. The authors are enabled the group to define a minimum set of data that could
inclined to agree with the sentiments of Socrates as portrayed be collected from all patients with TBI, which would be useful
in the “letter of dissent”14 when arguing against “Enthusiasti- in most research projects conducted in this population of
cus” that perhaps the loudest supporters of EBM are the hos- patients. To facilitate discussion, the core dataset was subdi-
pital accountants keen to keep health care costs down. There vided into four logical groups: (1) demographic and clinical
is no question that better monitoring and event detection information, (2) minute-by-minute monitoring information,
technology for health care is needed and that more research (3) intensive care management information, and (4) second-
to optimize that technology is also needed, but should their ary insult treatment information.
adoption depend upon large-scale clinical trials? Perhaps now From these meetings a consensus dataset was formed, which
the questions to focus upon are no longer if but when and no includes nine categories:
longer why but how.14a
1. Demographic and one-off clinical data (e.g., pre-
neurosurgical hospital data, first and worst computed
Data Acquisition tomography scan data)
Data acquisition is the sampling of the real world to generate 2. Daily management data (e.g., daily summary measures of
data that can be manipulated by a computer. Collection of the use of sedatives, analgesics, vasopressors, fluid input/
minute-by-minute physiologic monitoring data is routine in output balance)
many NCCUs. However, there are no agreed-upon standards 3. Laboratory data (e.g., blood gas, haematology, biochemis-
to define the collection of data. Cerebral perfusion pressure try data)
(CPP = Blood pressure[BP] − ICP) monitoring is a good 4. Event data (e.g., nursing maneuvers, physiotherapy, medical
example. When CPP values are reported, how does one correct procedures such as line insertion, calibrations)
for policies that differ between centers on the level to which 5. Surgical procedures
the BP transducer is zeroed? Tracking changes in bed tilt 6. Monitoring data summary (e.g., type and placement loca-
(which affects the size of hydrostatic pressure gradients tion of ICP sensors, BP lines)
between the head and heart) is also a significant technical issue 7. Neuroevent summary (e.g., Glasgow Coma Scale [GCS],
without defined standards. As a result, it is often difficult to pupil size and reactivity)
compare even the most common forms of monitoring when 8. Targeted therapies (e.g., mannitol given for raised ICP,
data are presented. Development of standards and guidelines pressor given for arterial hypotension)
in this area is an important step for the future design 9. Vital monitoring data (e.g., minute-by-minute BP, ICP,
and conduct of trials of new intensive care monitoring and systemic arterial oxygen saturation [SaO2] collected from
treatment methods. In an attempt to standardize how to the bedside monitoring)
approach standardizing correction of hydrostatic pressure
gradients, the BrainIT group has published a BrainIT web The full details of the core dataset definition and the collabo-
standard (http://www.brain-it.eu/). ration structure of the group can be found in the BrainIT
With this background in mind, certain individuals working publication: BrainIT Group: Core Concept and Data Defini-
within the field of TBI research met to discuss the founda- tion.16 This same approach has been undertaken in the United
tion and development of a network to create an IT-based States, sponsored by several federal agencies including the
Section VII—Computers, Engineering, and the Future 419

Ventilation change:
Normal
Mild hyperventilation (4-4.5 kPa)
Severe hyperventilation (<4.0 kPa)
Other (specify)
Sedation/Analgesia
Y/N
Volume expansion
Crystalloids
Colloids (including plasma)
Hypertonic saline
Erythrocytes
Other (specify)
Inotropes
Catecholamines
Other (specify)
Anti-hypertensive therapy ICP
Beta-blockers CCP
Alpha2 antagonists Neurologic deterioration
Other (specify) (eg: change in GCS, pupils)
Anti-pyretic therapy
Fever
Paracetamol
Cooling Hyponatremia
Other (specify) Hypoxia
Cerebral vasoconstriction Hypercapnia
Dihydroergotamine Hypotension
Indomethacin
SjvO2 desaturation
Other (specify)
Osmotic therapy Hyperglycemia
Mannitol Seizures
Glycerol Other (specify)
Other (specify)
Barbiturates
Dose/day
CSF drainage
Closed
Intermittent drainage
Open drainage
Steroids
Y/N
Hypothermia
Y/N
Head elevation
Y/N
Fig. 43.1  Tracking therapies and targets. CCP, Cerebral
Other treatments
Insulin perfusion pressure; CSF, cerebrospinal fluid; GCS,
Other (specify) Glasgow Coma Scale; LCP, intracranial pressure; SjvO2,
jugular venous oxygen saturation.

National Institutes of Health to develop the “Common Data Figure 43.1 summarizes the minimum choice of therapy cat-
Elements” (http://www.commondataelements.ninds.nih.gov/ egories and associated targets for the BrainIT core dataset.
and references 18 and 19). Unique to other dataset definitions, This therapy-tracking model is designed to be implemented
the BrainIT core dataset defined a special approach to quantify easily in software.
secondary insult management. This is medical management It is one thing to define on paper a dataset and another to
therapy given to patients specifically to treat secondary insults actually collect the data. Although a paper-based feasibility
that occur despite the patient’s baseline intensive care medical exercise established some baseline information, the acid test
management. To distinguish therapy given to patients to treat was still to develop a series of IT-based tools to collect the core
secondary insults from those of baseline intensive care, the dataset and to prospectively trial the collection of core data
authors have devised a coding system that allows specific cat- from a number of centers with NCCUs.
egories of therapy to be assigned a “therapy target.” For A 3-year follow-up EC-funded study (QLGC-2002-00160)
example, vasopressors may be given to treat systemic hypoten- enabled the group to develop IT methods to collect the core
sion or to treat reduced CPP secondary to raised ICP. Choos- dataset and to assess the feasibility and accuracy to collect this
ing an appropriate target for each secondary insult therapy core dataset from 22 neurointensive care centers. The main
will enhance the usefulness of the database on medical therapy. data collection instrument for the episodic nonmonitoring
Each therapy must be assigned a target chosen from a drop- data was a personal digital assistant (PDA)-based data collec-
down list. If drugs are given, then one can indicate continuous tion tool.
infusion if drugs are delivered by a continuous infusion pump Bedside nursing staff entered clinical data on hand held
or one can indicate boluses if it is delivered noncontinuously. PDAs that supported the BrainIT core dataset definition
420 Section VII—Computers, Engineering, and the Future

Center Glasgow

Data collection tool


Data converter tool Common data format

Regional
data validater
+ 20% sample Validation request tool
Data validation tool for validation

Fig. 43.2  BrainIT data validation flow of information (black lines, Incoming raw data flow; blue lines, validation requests/data flow).

through a Java Struts-based tool. This allowed the core dataset was uploaded via the BrainIT web-upload services where a
to be entered by roaming research nurses using a set of PDA server-side data-converter tool converted data from center-
documents accessed via a series of buttons and tabs. With this based formats into BrainIT data format to generate data cat-
system, indicators were present that showed data documents egory files that were imported into the BrainIT database (SQL
that were fully complete, partially complete, or totally incom- Server 2005). A validation request tool sampled 20% of the
plete. When convenient, the PDA was connected to a docking data sent for each data category and generated a validation
station and a client program allowed viewing and saving of request file listing the timestamps and data items to be
patient data collected. An anonymization routine removed checked by local data validators. Emails were generated to the
patient identification elements from the collected data and data validation staff that contained the validation data
labeled the patient data file with a unique BrainIT study code requested documents listing the data items to be checked.
generated from the BrainIT website. A local database held in Data validators entered into a data validation tool the
each center linked the anonymized data to local center patient requested data items for checking from source documenta-
ID information that was needed during the data checking tion held in each local center. Validation data then were
stage of the study. The multicenter ethics approval precluded uploaded to the BrainIT data coordinating center via the
connection of the PC client system holding the data to any website and using data validation checking software tools, the
computer connected to the Internet. Local research nurses validated data were checked against the data items originally
downloaded the anonymized data from the PDA system client sent, from which percentage accuracy data was calculated.
PC onto a memory stick or CD and transferred the data to an Figure 43.2 shows the flow of data between a remote center
Internet-connected PC for upload of the data to the BrainIT and the BrainIT database with the validation procedure
database via the website data upload page. selecting a random sample of 20% of data items uploaded per
Because this was a multicenter study collecting data from data type, which are sent to data validation nurses. The data
several countries with different languages, a multilanguage validation nurses enter requested data into a PC-based valida-
implementation was needed to foster ease of use by local tion data tool and upload the data to the data manager, who
nursing staff. A training course was held for the data valida- can then check the accuracy of data for each data category
tion nursing staff in Glasgow on the use of this data collection and estimate an overall error rate.
instrument, which also included using their medical term and As part of this validation process, in addition to the cate-
language expertise to translate all PDA labels and text output gorical and numeric clinical data being checked for accuracy,
into six European languages (English, French, German, the BrainIT system also assessed the minute-by-minute moni-
Spanish, Flemish, and Italian). Data could be entered in the toring data. Random samples of monitoring data channels
local language, exported in an XML file format where a table uploaded (e.g., ICP, SaO2) were selected and validation staff
lookup driven by an XSL transformation converted the data asked to manually enter the hourly recorded values from the
into a standard English language version. nurse’s chart (or local gold standard data source) for the first
Data validation research nurse staff were hired on a coun- and last 24-hour periods of bedside monitoring for a given
try-by-country basis to check samples of the collected data patient for a given channel. These validation values could then
against gold standard clinical record sources to quantify the be compared with a range of summary measures (e.g., mean,
accuracy for collection of the BrainIT core-dataset using the median) from the computer-based monitoring data acquired
group IT-based data collection methods. Anonymized data from the patient.
Section VII—Computers, Engineering, and the Future 421

well with nursing chart end-hour recordings. This allows the


Table 43.1  Percentage Error Rate by Data end-hour averaged computer records to be used in database
Type Class with Common Error Types analyses assessing nurses’ chart-recorded detection of events
Data Class Error Rate (%) Common Errors with computer-based sampling. These validation results cal-
culated on a subset of patients provides an estimate of the data
Laboratory 2 PaCO2, FiO2 value
quality for future analyses on the full patient cohort of 350
Demographic 4 Monitoring on arrival patients collected as part of the EC-funded study, which was
at neurosurgery, conducted over the same time period by the same staff using
intubation on arrival
at neurosurgery the same data methods. Clearly, though, future data collection
projects will generate datasets under differing data collection
Neuro 5 Pupil Size, GCS versus
observations (code 1 versus
conditions and will require a separate validation stage if con-
unknown) fidence is to be maintained in the level of data accuracy.
However, the costs of maintaining such a data validation
Monitoring 5 ICP type, ICP location
summary network are prohibitively high. To maintain a full-time data
validation nurse within each participating country costs in
Daily 5 Infusion type (bolus vs
management infusion or both),
excess of 1 million Euros per year. Such large running costs
summary drug number (1, >1) for an academic network are not sustainable in the long term
and a more cost-effective solution for data validation must
Targeted 6 Nonstandard target,
therapy no target specified be found.
Surgeries 34 ICP monitor; surgery
for skull fracture or Information Processing
mass lesion
Practical information processing can be described as a cycle,
FiO2, Fraction of inspired oxygen; GCS, Glasgow Coma Scale; ICP, intracranial wherein data that may have no inherent meaning to the
pressure; pCO2, partial pressure of carbon dioxide.
observer is converted into information that does have meaning
for the observer. In neurointensive care, improved informa-
tion processing is now a necessity. Furthermore, with advanced
By late 2008, 384 TBI patients’ core data had been collected bioinformatics that use analytic techniques, the complex
from 22 European neurointensive centres. The first 200 interrelationship between physiologic variables can be
patients, data were cleaned and validation analyses conducted. explored. In turn, this may predict future events in the NCCU
In total, 19,461 comparisons were made between collected rather than just outcome; that is, it can facilitate targeted
data elements and source documentation data.20 The number management.22 With increasing use of computer technology
of comparisons made per data category was in proportion to that generates large quantities of high-resolution datasets,
the size of the data received for that category with the largest interpretation of the data is now the limiting step. Three
number checked in laboratory data (5667) and the least in the examples of research information processing approaches that
surgery data (567). Table 43.1 summarizes error rates by data may address some of the problems with interpretation of large
class. Error rates were generally less than or equal to 6%, with amounts of neurocritical care data follow.
the exception being the surgery data class where a high error
rate of 34% was found.
The proportion of surgery errors was due primarily to the Mathematical Models
classification system used to simplify and thereby reduce the Cerebral autoregulation is the process by which cerebral blood
burden of data entry. For example, through discussions with flow (CBF) is maintained constant over a specific CPP range.
local nursing and data validation staff it was found that there Cerebral autoregulation can be an important factor in clinical
was particular confusion over when to record ICP sensor decisions about treatment of the patient’s injuries; if it is mis-
placement and a craniectomy or craniotomy when the proce- diagnosed, undesirable effects, such as ischemia or hypoxia,
dures occurred together as the primary surgical procedure. As could occur from inappropriate treatment. Autoregulation is
such, confusion over coding these two procedures by both the typically classed as either static or dynamic, depending on the
local research nurse and the data validation nurse accounted time frame over which it is assessed.23
for the majority of errors in this data category. Tests of static autoregulation tend to be based on the
This study conducted by the BrainIT group is one of only manipulation of BP by pharmacologic agents with measures
a few projects to attempt to prospectively assess the data of volume CBF at two different BP levels with measurements
capture error rate within an academic environment. It was occurring many minutes apart. The current gold standard
shown that it is feasible to collect the BrainIT dataset from methods to assess static autoregulation involve the use of
multiple centers in an international setting with human- ultrasound or invasive isotope infusion, or require the patient,
intensive (research nurse) IT-based methods and the accuracy who is critically ill, to be taken out of the NCCU to a posi-
of the data collected is greater than or equal to 94%, with the tron emission tomography/single photon emission computed
exception of the surgery data type implementation. One of the tomography (PET/SPECT) scanner for blood-flow measure-
challenges to implementing the system over multiple sites is ments. For patients in the ICU, this approach can be used at
data security. In the short term this is overcome in part by most only once or twice a day.
allowing voluntary entry of data by investigators at each site.21 Dynamic autoregulation assesses the short-term response
It was also shown that computer-collected minute-by-minute of the cerebrovascular bed over 10 to 20 seconds to perturba-
vital signs data, summarized as end-hour averages, correlate tions in BP caused by leg cuff deflation or brief unilateral
422 Section VII—Computers, Engineering, and the Future

carotid compression. There is no accepted standard method good correlation of the reworked Ursino Lodi model output
for dynamic autoregulation testing, although there is a con- G with clinical outcome over the later stages of patient ICU
sensus that dynamic testing of autoregulation is more clini- stay. Results from a pilot study in 12 patients showed that there
cally practical than static autoregulation testing.23 was a significantly greater mean variance in G in patients with
Ideally, what is needed is a method that can assess the status bad outcome than good outcome.21
of autoregulation on a continuous basis with minimal distur- Such modeling approaches based upon mathematical
bance to the patient’s normal clinical management or moni- models rather than data driven models can be inherently
toring. This is often difficult to achieve with techniques that more informative, because their underlying mechanism can
measure CBF or velocity, because they typically require spe- be studied and adapted as knowledge of the cerebrovascular
cialized monitoring and expert measurement. However, most system develops. Modeling approaches that rely solely on the
patients have minute-by-minute monitoring of BP and ICP as relationship between two or more physiologic parameters may
standard, and a number of groups have been studying the not be so easily extended.
time-dependent variation in BP and ICP (cerebrovascular
pressure transmission) with a view to revealing relationships
between them and highlighting patterns that may indicate Fractal Analysis
abnormal autoregulation. During the past two decades, analysis techniques have moved
Mathematical modeling of the autoregulatory process is from classic stochastic process analysis using basic statistics to
one approach that has been studied. Several models have more nonlinear systems or chaos theory–based approaches
already been produced using different analysis techniques that look at patterns in the variability of the time series. Physi-
including linear regression, spectral analysis, transfer function ologic time series exhibit complex multifractal properties,28 so
analysis, cross correlation function, or impulse response anal- changes and alterations in the underlying physiologic pro-
ysis.24 Some of these approaches have had their performance cesses can be detected when a classification and analysis based
assessed in a comparative study, but these methods are based on this nonlinear and chaotic nature of the time series is
upon single or pulse train analysis that requires specialized designed. The main idea used in the initial analysis and fractal
monitoring with high data-sampling rates.25 To be practical in characterization of the waveform leverages the relationship
the clinical environment, models should be developed and between the mathematical properties of a wavelet transforma-
assessed from data with minute-by-minute BP and ICP sam- tion and a signal’s localized fractal nature.
pling rates that are a more common data capture rate in many Wavelets29 can be thought of as a time-frequency analysis
ICUs. technique analogous to a Fourier transformation but without
There are several models of cerebral autoregulation; the the inherent frequency-related problems of Fourier analysis.
Ursino Lodi model is an example of a mathematical modeling As an example of this type of analysis, ICP waveform signals
approach.26 Further work by the BrainIT group20 has reworked were randomly sampled and cleaned from the BrainIT dataset,
the Ursino Lodi autoregulation model to derive an index of and a moving window approach was created to allow the
autoregulation (G) that allows comparison of autoregulatory repeated application of the mean Holder function (which is a
models and provides a standard measure of autoregulation measure of the fractal nature of a signal) along the time course
obtained from animal models. This modified Ursino index of the original signal (Fig. 43.4). Although only a sample
also correlates with the Bouma index27 of static autoregula- analysis, a clear downward trend in the Holder component
tion.21 The application of this type of autoregulatory modeling that indicates a decrease in the chaotic (fractal) nature of the
in TBI datasets is illustrated in Figure 43.3, which shows a signal is observed before increases in ICP instability occur.21
It is likely that further study of the fractal nature of physiologic
time series generated from patients in the NCCU may provide
new approaches to data interpretation.
Outcome Group Comparison

350 Neural Networks


300
* (p = 0.03) Artificial Neural Network
An artificial neural network (ANN) is a digital simulation of
250 a brain model, and can be considered a pattern classifier, that
relates input patterns to output patterns. This is a very robust
Variance of G

200 tool for classification of multiple factors and for multivariate


analysis that may facilitate recognition of patterns or clusters
150 of data, including in ICU studies that may otherwise be dif-
ficult to recognise.30 In an ANN, individual “neurons” are
100 interconnected from an input layer (the inputs to which are
the patient parameters) through one or more hidden layers to
50 an output layer (Fig. 43.5). Each neuron within the ANN
performs some numerical operation on its input connections
25 to generate the values for its output parameters. This is typi-
cally accomplished by assigning (initially random) weights to
Bad Good all of the inputs and outputs. All but the simplest ANNs
Fig. 43.3  Box plot representation of variance of G that compares require some amount of training for the output patterns to
outcome groups. become relevant to the input patterns. This training usually
Section VII—Computers, Engineering, and the Future 423

ICP Wave Form


200

150
ICP (mm Hg)

100

50

0
0 2000 4000 6000 8000 10000 12000 14000
A Time (min)

Trend of Mean Holder Exponent


1.5
Mean holder exponent

1.0

0.5
Fig. 43.4  Time series plots of (A) raw intracranial
0.0 pressure (ICP) data and (B) the mean Holder
exponent (as a measure of the fractal nature of
–0.5 the signal). As ICP becomes more unstable, the
Holder exponent drops and shows a clear trend
0 2000 4000 6000 8000 10000 12000 14000
many hours before the occurrence of the ICP
B Time (min) instability.

that fit the limited training set perfectly but that are highly
inaccurate when presented with new data. This problem is
Input layer Output layer referred to as “overfitting.” Also, because of the complexity of
the functions computed, the techniques used to compute con-
fidence intervals for simpler models (e.g., linear or polynomial
models) cannot be used for neural networks. In many domains,
and clearly in any domain that includes medical treatment
(such as is presented here), the confidence assigned to a pre-
Hidden layer(s) diction is of critical importance.
An appropriate approach to resolve these problems applies
Bayesian principles to the problem of neural network training.
Rather than generating a single network to model the training
set, many models (typically hundreds) are generated in a
Monte Carlo Markov Chain (MCMC). The series of models
generated by the MCMC has the property that it conforms to
Fig. 43.5  Schematic representation of an artificial neural network.
the Bayesian posterior distribution on the space of possible
models conditioned on the training data. Thus to generate a
prediction, the whole set of models derived during training is
requires that the weights on the network be adjusted after each used to generate a corresponding distribution of outputs.
training event to optimize the output and so depends on the Some summary measure of the Bayesian Artificial Neural
existence of a (typically large) dataset that matches the inputs Network (BANN) output such as the median or the mode of
to the required outputs. When training, great care must be this distribution can be used as the prediction, and the spread
taken to ensure that the network is not overtrained, which of the output values can be used to assign confidence intervals
typically results in the network being highly accurate for only for this prediction, derived on the basis of the Bayesian prin-
a small subset of the input pattern set, yet it must be ensured ciples of probability. The problem of overfitting is addressed,
that the network is sufficiently trained to offer realistic results because in the process of combining predictions from hun-
for previously unseen input patterns. dreds of models, the divergent predictions of the models that
overfit the training data average out.
The confidence intervals generated in such a model are
Bayesian Approach to Neural superior to those normally used for simpler models such as
Network Training linear and polynomial models. As an example, when a linear
The power of neural networks as general nonlinear models model is fit, confidence intervals are given based on the
can lead to problems. Overly complex models may be derived assumption that the data are in fact generated by a linear
424 Section VII—Computers, Engineering, and the Future

Edinburgh Uppsala

1.0 1.0
0.9 0.9

Probability of favorable outcome

Probability of favorable outcome


0.8 0.8
0.7 0.7
0.6 0.6
0.5 0.5
0.4 0.4
0.3 0.3
0.2 0.2
0.1 0.1
0.0 0.0

0 20 40 60 80 100 0 20 40 60 80 100
% Monitoring time with CPP <60 % Monitoring time with CPP <60
Fig. 43.6  Bayesian Artificial Neural Network–generated probability distribution plots for the mean (±95% confidence limits) for the
likelihood of a favorable clinical outcome with increasing time spent with cerebral perfusion pressure (CPP) less than 60 mm Hg. Data
are presented from two centers. Note the difference in predicted outcome with CPP management; this suggests that these two patient populations
have different underlying pathophysiologic mechanisms or clinical characteristics.

process. However, this assumption particularly in complex data sets often means that research data entry is duplicated
medical datasets is not justified, and as a result leads to unre- and often requires nursing staff to enter the same data on
alistically optimistic confidence intervals being assigned to more than one data-entry system. If one could directly access
model predictions. The confidence intervals derived by the hospital-based data remotely (and securely) for research pur-
Bayesian neural network approach realistically reflect uncer- poses, then considerable savings could be made to implement
tainty about both the form and the parameter settings of the and finance many clinical research projects.
model, as well as the variance of the training data. Grid technology provides software methods (middleware)
Bayesian networks have been applied to critical care that can provide distributed access and collection (federation)
research.31 This type of BANN approach also has found some of data to occur. However the medical domain is especially
success in neurocritical care applications. For example Howells demanding in its adherence to ethics and information gover-
et al.32 applied a BANN model to show how patient outcome nance, which in turn requires fine-grained security. Many
probabilities change as a function of physiologic variables, IT-based data storage solutions already exist that cross primary
such as the percentage of monitoring time that CPP is less care, secondary care, and a range of specialized resources such
than 60 mm Hg. Figure 43.6 shows graphically the output of as disease registries in the clinical domain. Many of these
such a model with the probability of a favorable outcome solutions have been developed in isolation and have different
(Glasgow Outcome Scale [GOS] 4 or 5) as a function of the data security policies, or in many cases, no security policies
percentage of monitoring time that a patient’s CPP was less other than protection at the hospital firewall level. This
than 60 mm Hg. In the patients in Edinburgh, longer dura- deficiency in security policies is magnified when crossing
tions of CPP insult were associated with death. In contrast, in national boundaries. Dealing with such heterogeneity from
those in Uppsala, longer durations of CPP insult suggested the data perspective has been one of the drivers behind grid
favorable outcome. Using this same approach, there are several technologies.
commercially available systems that offer prediction of forth- Given the sensitivity of data, the establishment of security
coming adverse events, such as the BioSign device (Fig. 43.7), policies and their enforcement is essential in the clinical
which derives a BioSign index value from five parameters domain. A cornerstone of these solutions is to ensure fine-
(heart rate, respiration rate, body temperature, oxygen satura- grained security. However, it is an unavoidable fact that the
tion, and BP), from which subsequent cardiovascular instabil- specific privileges required in a particular trial or study will
ity can be predicted.33 not be known when the system is first created. Similarly a
The BrainIT network approach to collaborative neurocriti- doctor in one hospital may have privileges to access various
cal care research used the accepted model of upload of local systems in that hospital, but these do not transfer directly
center data to a central data repository or database. To do so when this doctor attends a different hospital. Therefore
required hiring of research nurse staff employed specifically systems capable of adding and removing resources or privi-
to validate uploaded data. The costs of maintaining such a leges “on the fly” are necessary, wherein the corresponding
data validation network are expensive and not sustainable in allocation of privileges can be added or removed depending
the long term and so a more cost-effective solution for data on the needs of different trials or health care systems. In grid
validation must be found.21 Much of the data collected for parlance, the framework by which such rules and regulations
research purposes already is collected as part of the routine on the resources and the users that may access them and
clinical management. However, in many centers the lack of under what conditions is conceptually called a virtual organi-
transparent mechanism that allows access to hospital-based zation (VO).
Section VII—Computers, Engineering, and the Future 425

Fig. 43.7  BioSign technology to help predict cardiovascular instability.

The basic models suggested for the majority of grid-based framework. A more sophisticated grid infrastructure (AVERT-
security systems can be broadly broken down into the “AAA” IT project; http://www.avert-it.org) is under design.34 In this
categories: virtual environment near real-time information is captured
from six NCCUs across Europe (located in Barcelona, Glasgow,
 Authentication. Establish the identity of the person who
Heidelberg, Monza, Uppsala, and Vilnius) to explore the
requests access to a resource.

potential prediction of hypotensive events. To support this
Authorization. Having established identity, establish and
work, researchers are exploring how hypotensive events might
enforce what that person is allowed to do on a given
be predicted using a novel BANN, to be trained against an
resource.

existing BrainIT dataset before a prospective clinical trial is
Accountability. Establish the activities, and time of activi-
undertaken to demonstrate the effectiveness of the AVERT-IT
ties, of a particular person on that resource (or resources)
project concept (AVERT-IT portal). Figure 43.9 is a block
so that they cannot subsequently deny potential misuse
diagram that illustrates the virtual organization structure.
later on (nonrepudiation).
With the AVERT-IT systems, models are proposed based
The following example illustrates the interplay of role-based upon pushed data transfer only; that is, the clinical data pro-
access to federated resources. A particular BrainIT trial is vider firewalls will reject all incoming data requests, but will
undertaken with the Southern General Hospital in Glasgow. push data out of the firewalls into demilitarized zones set up
When users attempt to access the BrainIT portal, they are as a buffer between the grid re++search domain and the
redirected to their home institutions when they are asked to domain of live health care provision. One significant challenge
authenticate. After authenticating, the attributes needed to that remains to be solved in the AVERT-IT system and within
access the BrainIT portal for authorized access to the federated the wider e-research community is with real-time or near real-
clinical data are accessible. The portal then uses these attri- time data. Monitoring data that needs to be streamed to
butes to configure the contents of the portal; that is, the users support the Bayesian adverse event prediction algorithms
are restricted to see and do what their roles dictate. Figure 43.8 offers new challenges that have hitherto not been addressed.
shows two different users have logged in to the system, one
with the brainIT-investigator role (right side) and one with the
brainIT-nurse role (left side). The investigator role has more
Conclusion
privileges in this particular trial and hence is allowed access to The future of information processing, data acquisition, and
a wider range of information within the context of that trial storage within the neurocritical care environment must be a
(e.g., GCS, ICP, glucose levels, and patient identification). The collaborative approach. Gone are the days when neurosurgeons
nurse role on the other hand is restricted to a subset of non– or intensivists were able to follow and implement technical
patient-identifying information. developments outside their own specific area of expertise. Col-
The systems described show the proof of concept upon laborations between clinicians and information technologists
which clinical data can be accessed and used within a secure are key, but often too large a knowledge gap must be bridged
426 Section VII—Computers, Engineering, and the Future

Fig. 43.8  Role-based access to portal and restrictions to data access.

AVERT IT Existing
data
HypoPredict Bedside Bedside Bedside
collection
web app Local monitor monitor monitor
HypoPredict
engine
Patient
Web Pages
WebServer for
(for both HypoPredict HypoPredict
and GRID system)
Web Pages Physiologic
for
GRID GRID data
secure
access
services
Event/treatment
Local data
HypoPredict
database

GRID Demographic
role DB data
Existing hospital IT
GRID
Secure internet
access to/from This software runs on a infrastrucure
secure
clinical trail
access computer supplied by
management
Web app services the Neurosurgery center
to an AVERT-IT spec
Neurosurgery ICU
Key: = C3Amulet, = GRID

Fig. 43.9  Block diagram that shows the virtual organization design where hospital datasets are transferred to a local grid repository in
each neurocritical care center. Secure grid access services support role-based access to the repository with monitoring of all six centers’ data remotely
by a data manager via grid-based web services.
Section VII—Computers, Engineering, and the Future 427

to make such collaborations functional. Many hospitals now 14. Smith D. Evidence based medicine: Socratic dissent. Br Med J
1995;310:1126–7.
employ clinical physicists or scientists who are seconded into
14a.  Piper I. Not if but when; no longer why but how. Critical Care
areas of specialization, including neurosurgery and intensive 2007:11(1):117.
care. It may well be that such staff could help bridge this gap. 15. Brain Monitoring with Information Technology (BrainIT) Research Group:
Clinical scientists are the natural interface between clinicians http://www.brainit.org.
and their data. Often they are responsible for assisting clinical 16. Piper I, Citerio C, Chambers I, et al. The BrainIT Group: concept and core
dataset definition. Acta Neurochir 2003;145:615–29.
staff to develop and maintain data collection and analysis 17. European Brain Injury Consortium – EBIC: http:www.ebic.nl.
tools and, as such, they understand better than many the details 18. Maas AI, Harrison-Felix CL, Menon D, et al. Common data elements for
and limitations of the data and the methods to collect and traumatic brain injury: recommendations from the interagency working
condition that data. Clinical scientists are not IT experts, but group on demographics and clinical assessment. Arch Phys Med Rehabil
2010;91(11):1641–9.
given their numerate and technical background, are better
19. Manley GT, Diaz-Arrastia R, Brophy M, et al. Common data elements for
able to act as a communication channel between the informat- traumatic brain injury: recommendations from the biospecimens and
ics experts and clinicians. Whatever the formula is, the modern biomarkers working group. Arch Phys Med Rehabil 2010;91(11):1667–72.
NCCU is best based upon functional collaborations between 20. Shaw M, Piper I, Chambers I, et al. on behalf of the BrainIT Group. The
domain experts who all pull in the same direction. And that Brain Monitoring with Information Technology (BrainIT) collaborative
network: data validation results. Acta Neurochir Suppl 2008;102:217–21.
direction is to provide a sound evidence base to help improve 21. Piper I, Chambers I, Citerio G, et al. The brain monitoring with Information
patient monitoring and management. Technology (BrainIT) collaborative network: EC feasibility study results and
future direction. Acta Neurochir (Wien) 2010;152:1859–71.
References 22. Buchman TG. The digital patient: predicting physiologic dynamics with
mathematical models. Crit Care Med 2009;37:1167–8.
1. Riddick G, Fine HA. Integration and analysis of genome-scale data from 23. Tieks F, Lam AM, Aaslid R, et al. Comparison of static and dynamic cerebral
gliomas. Nat Rev Neurol 2011;7(8):439–50. autoregulation measurements. Stroke 1995;26:1014–9.
2. Hemphill JC, Andrews P, De Georgia M. Medscape. Multimodal monitoring 24. Panerai RB. Assessment of cerebral pressure autoregulation in humans—
and neurocritical care bioinformatics. Nat Rev Neurol 2011;7(8):451–60. a review of measurement methods. Physiol Meas 1998;19:305–38.
3. Bates D, Gawande A. Improving safety with information technology. N Engl 25. Panerai RB. Neural network modelling of dynamic cerebral autoregulation:
J Med 2003;348:2526–34. assessment and comparison with established methods. Med Eng Phys
4. Kuperman GJ, Teich JM, Tanasijevi MJ, et al. Improving response to critical 2004;26(1):43–52.
laboratory values with automation: results of a randomised controlled trial. 26. Ursino M, Lodi CA. A simple mathematical model of the interaction
J Am Med Inform Assoc 1999;6:512–22. between intracranial pressure and cerebral hemodynamics. J Appl Physiol
5. Rosenfield BA, Dorman T, Breslow MJ, et al. Intensive care unit 1997;82(4):1256–69.
telemedicine; alternative paradigm for providing continuous intensive care. 27. Bouma G, Muizelaar P, Bandow K, et al. Blood pressure and intracranial
Crit Care Med 2000;28:3925–31. pressure-volume dynamics in severe head injury: relationship with cerebral
6. Stochetti N, Penny K, Dearden M, et al. Intensive care management of blood flow. J Neurosurg 1992;77:15–9.
head-injured patients in Europe: a survey from the European Brain Injury 28. Bassigthwaighte JB, Liebowitch LS, West BJ. Fractal physiology. Oxford:
Consortium. Intensiv Care Med 2001;27:400–6. OxfordUniversity Press; 1994.
7. Bulger E, Nathens A, Rivara F, et al. Management of severe head injury: 29. Muzy JF, Bacry E, Arneodo A. Wavelets and multifractal formalism for
institutional variations in care and effect on outcome. Crit Care Med singular signals: application to turbulence data. Phys Rev Lett
2002;30:1870–6. 1991;67(25):3515–8.
8. Narayan R, Michel M, Clinical Trials in Head Injury Study Group. Clinical 30. Cohen MJ, Grossman AD, Morabido D, et al. Identification of complex
trials in head injury. J Neurotrauma 2002;19:503–57. metabolic states in critically injured patients using bioinformatic cluster
9. Patel H, Bouamra O, Woodford M, et al. Trends in head injury outcome analysis. Crit Care 2010;14:R10.
from 1989 to 2003 and the effect of neurosurgical care: an observational 31. Peelen L, de Keizer NF, Jonge E, et al. Using hierarchical dynamic Bayesian
study. Lancet 2005;366:1538–44. networks to investigate dynamics of organ failure in patients in the intensive
10. Varelas PM, Eastwood D, Yun H, et al. Impact of a neurointensivist on care unit.
outcomes in patients with head trauma treated in a neurosciences intensive J Biomed Inform 2010;43:273–86.
care unit. J Neurosurg 2006;104:713–9. 32. Howells T, Elf K, Jones P, et al. Pressure reactivity as a guide in the treatment
11. Clayton T, Nielson R, Manara A. Reduction in mortality from severe head of cerebral perfusion pressure in patients with brain trauma. J Neurosurg
injury following introduction of a protocol for intensive care management. 2005;102:311–7.
Br J Anaesth 2004;93:761–7. 33. BioSign Technology. http://www.isis-innovation.com/documents/Biosign.pdf.
12. Elf K, Nilsson P, Enblad P. Outcome after traumatic brain injury improved 34. Stell A, Sinnott R, Jiang J, et al. Federating distributed clinical data for the
by an organised secondary insult program and standardised care. Crit Care prediction of adverse hypotensive events. Philos Transact A Math Phys Eng
Med 2002;30:2129–34. Sci 2009;367(1898):2679–90.
13. Chambers I, Jones P, Milly Lo T, et al. Critical thresholds of intracranial
pressure and cerebral perfusion pressure related to age in paediatric head
injury. J Neurol Neurosurg Psychiatry 2006;77:234–40.
VII
Chapter
44  

VISICU and the eICU Program


Pamela J. Amelung and Martin E. Doerfler

has been done in the past, or what is being done currently, but
Introduction rather by using new tools to help solve core problems. The
Neurocritical care originated from the principles of respira- report also called for use of information technology to help
tory intensive care established during the poliomyelitis epi- drive the quality improvements. In response to the IOM
demic of the mid-twentieth century, evolving to a discipline reports, The Leapfrog Group, an organization of Fortune 500
that provides comprehensive medical and specialized neuro- companies and large private and public health care purchas-
logic support for patients with life-threatening neurologic ers, identified four hospital quality and safety practices that
diseases.1 The early neurologic critical care unit (NCCU) ini- could help prevent errors and save lives, including intensive
tially focused on the management of postoperative neurosur- care units (ICUs) staffed by intensivists 24 hours a day and all
gical patients. Today neurocritical care has expanded to week long. Estimates suggest that 50,000 lives and $4.3 billion
include the management of patients with intracranial hemor- may be saved each year in the United States through 24-hour
rhage, traumatic brain injury (TBI), stroke, complex seizures, intensivist staffing.6,7
elevated intracranial pressure (ICP), and the systemic compli- Too few ICU patients are cared for by critical care special-
cations of brain injury. Rapid advances in neurologic and ists; not every hospital has the patient volume or the financial
neurosurgical interventions and interventional neuroradiol- resources to hire dedicated intensivists. In addition there is a
ogy in the last two decades, and the appreciation that a brain shortage of trained intensivists.8,9 About 6000 intensivists are
affected by a primary injury is influenced by systemic altera- estimated to be in practice in the United States today, and less
tions that affect its function (secondary injury), have led to than 15% of all ICU beds have dedicated intensivists. There
the development of specialized NCCUs and specialized train- are approximately 6000 ICUs in the United States, which rep-
ing for physicians and nurses, all focused on treating life- resents 5% to 10% of hospital beds. On a daily basis, these
threatening neurologic diseases.2 ICUs care for approximately 55,000 patients. Only one third
Today, neurocritical care is one of the newest and fastest of these patients are treated by an intensivist, either as the
growing specialties in medicine, and an increasing number of primary physician or as a consultant.10 In addition, there is a
institutions throughout the world now have dedicated neuro- current nursing shortage and aging of the most experienced
critical care units. Additionally, there are a number of hospi- critical care nurses. The first of the “baby boomers” are reach-
tals without dedicated neurocritical care units but with ing retirement age at a time when technology provides for
neurointensivists acting as consultants.3 unprecedented life expectancy and recovery from illness and
injury. However the relative lack of intensivists and critical
care nurses is not likely to improve.
Development of VISICU and
the eICU Program
ICU Outcomes with an Intensivist Model
The Leapfrog Initiative Many variables determine outcomes in critically ill patients,
As neurocritical care has come to the forefront in the care of including the primary diagnosis that leads to admission,
seriously ill or injured neurologic patients, so has the concept patient characteristics, the evolution of secondary disorders,
of specialty critical care medicine in general become more and the care that these patients receive. There is a wealth of
widely accepted over the past two decades. In its 1999 report, data to suggest that intensivists, having been trained specifi-
“To Err Is Human: Building a Safer Health System,” the cally in the care of the critically ill patient, are better at man-
National Academy of Sciences’ Institute of Medicine (IOM) agement of comorbidities and avoidance of complications,
estimated at least 44,000 and as many as 98,000 people die and so can improve outcomes in ICU patients. This has stimu-
each year in hospitals from preventable medical errors.4 In a lated considerable interest in ICU performance. Patients
subsequent report, “Crossing the Quality Chasm: A New managed by a dedicated critical care service in a single hospital
Health System for the 21st Century,” published in 2001, the surgical ICU have a shorter length of stay (LOS), fewer com-
IOM called for a redesign of the U.S. health care system to plications, and lower hospital charges than patients cared for
help achieve the goals stated in the first report and stated: “You by general surgeons and house staff.11 In 1999, a study by
can’t get there from here.”5 This means that the problem of Pronovost et al12 found that daily rounds by an intensivist
quality improvement will not be solved by doing more of what were associated with improved outcomes in patients who
428 © Copyright 2013 Elsevier Inc. All rights reserved.
Section VII—Computers, Engineering, and the Future 429

underwent abdominal aortic surgery. Others have demon- individual patients21 to continuous offsite monitoring for the
strated reduction in mortality,13 including a systematic litera- whole ICU,22 but all combine audiovisual technology (video-
ture review in which 16 of 17 studies showed a decrease in conferencing technology and telemetry) and electronic
hospital mortality with high-intensity ICU staffing (the inten- medical records to provide critical care from a remote site. It
sivist was the primary attending or there was a mandatory is estimated now that about 10% of the total ICU beds in the
critical care consultation for all ICU patients).10 However, United States use this technology.23 The telemedicine coverage
Levy et al14 have debated the benefit of the intensivist model may come in several forms, including (1) type of coverage
and suggest that intensive care provided by critical care spe- (proactive, reactive, or mixed), (2) structure (a single consult-
cialists does not improve outcomes and may be detrimental ing “hub” that covers multiple ICU “spokes,” a “parent” hos-
to patients. In this study, hospital mortality was higher for pital that covers another hospital in its system, or a network
patients managed by critical care physicians, even after adjust- of clinicians who monitor from multiple sites), and (3) hours
ment for severity of illness and other factors that may have of operation (24 hours or night only). In part, the telemedi-
influenced the probability of being cared for by a critical care cine setup depends on physical ICU staffing (open or closed),
physician. However, this study is observational in nature, intensivist availability at physical ICUs, number and type of
making cause-and-effect conclusions difficult. Furthermore, ICUs, and hospital resources or affiliation with a tele-ICU
heterogeneity beyond that accounted for by applying adjust- vendor.
ments may limit result interpretation.
Most studies identify ICU physician staffing as an opportu-
nity to reduce in-hospital mortality. Assuming 3.5 million Telemedicine and General ICU Outcome
patients are admitted each year to ICUs in the United States Several factors may influence how a telemedicine program
and a 40% reduction in mortality with dedicated intensivists, affects clinical outcome: (1) integration of the information
it is estimated that 54,000 lives could be saved with a full systems of the tele-ICU and the monitored units, (2) physician
intensivist model as described by the Leapfrog Group. acceptance, and (3) how the telemedicine system is used. In
However, only 10% to 15% of U.S. hospitals have such a addition, other delivery systems and work-process innova-
program in place, in large part because of a shortage of inten- tions rather than technology alone can impact outcome.24
sivists. This shortage of intensivists is expected to worsen in Several studies have examined how telemedicine in general
coming years. The technology tools of the eICU program, ICU care may influence outcome.19,22,25-30 Some suggest a
including virtual presence through audiovisual technology, benefit in the sickest patients,22 because these may be the
provides an alternative means to bring intensivists expertise patients most likely to have unexpected events. In 2011, Young
to hospitals that lack sufficient numbers of intensivists. et al30 published a systematic review and metaanalysis that
described the clinical impact of ICU telemedicine. They iden-
tified 13 studies, only seven of which were published in peer-
Telemedicine to Leverage Scarce Resources reviewed literature that examined the association between
Grundy and colleagues15,16 first introduced telemedicine as a ICU telemedicine and clinical outcomes. Each study used a
tool for critical care in the late 1970s. Interactive television was before-and-after design, but severity of illness and case-mix
used to provide consultation with university-based critical was not always adjusted for. On metaanalysis there was an
care physicians for patients in the ICU of a 100-bed hospital association between reduced ICU mortality and LOS and a
that lacked physicians with critical care expertise. During an telemedicine ICU. However, there was no association between
18-month period, 1548 telemedicine “visits” were made to 395 admission to an ICU under a telemedicine model and
patients. The process was thought to be a reliable method to in-hospital mortality or hospital LOS. The confidence inter-
extend the availability of specialist expertise and to be benefi- vals were wide, and so a clinically significant effect could not
cial to patient care. However it was another 15 years before the be excluded. In addition, most of the studies included in the
concept of telemedicine in critical care reemerged, albeit in a analysis used telemedicine at night to enhance patient safety
very different form. In 2000, Rosenfeld17 reported that intro- in ICUs that already had high-quality daytime staffing. In
duction of around-the-clock intensivist oversight through some of the studies, authority was delegated to the telemedi-
telemedicine produced reductions in mortality and LOS cine physicians only in life-threatening events. The value of
similar to those reported by implementation of an intensivist telemedicine may not be so much in improved safety but
program.17 Subsequently, Breslow and Rosenfeld founded rather in the ability to provide evidence-based care on a
VISICU, a start-up company that provided the first con­ 24-hour basis in a proactive fashion.31-33 Furthermore the
tinuous, remote, centralized, computer-assisted intensivist benefit may be greatest in smaller ICUs with limited staffing
program for critical care. Sentara Healthcare in southeast Vir- where the telemedicine intensivists can interact with bedside
ginia was the first to implement this program in 2000. After staff to optimize proactive evidence-based management rather
the first year, hospital mortality decreased by 27%, ICU LOS than reactive care.26
by 17%, and hospital LOS by 13% compared with the previous
year.18 Variable costs decreased and hospital revenue increased
because of increased patient throughput. These improvements Evolution of VISICU
have been sustained over subsequent years. Since the original Sentara project, VISICU has implemented
What is telemedicine? Because onsite intensivist staffing is more than 35 programs in 26 states, bringing the eICU
associated with reduced mortality,13,19,20 telemedicine in the program to more than 200 hospitals and more than 8% of the
ICU has evolved in part to address limited clinician supply adult ICU beds in the United States. More than 300,000
through a technologic solution. ICU telemedicine can come patients now have their ICU care supplemented by a remote
in several forms that range from consultations regarding care team annually (Fig. 44.1). These eICU programs can
430 Section VII—Computers, Engineering, and the Future

residents, significant reductions in ICU (63%) and hospital


(45%) mortality were observed; that is, 336 lives were saved.34
Hospital and ICU LOS also were reduced with an estimated
$5.8 million savings in direct costs. Studies have also demon-
strated an eICU program to be useful in rural hospitals.35 For
example, in the Avera-McKennan hospital of the Avera Health-
care System in South Dakota, significant reductions in stan-
dardized (APACHE III) ICU and hospital mortality ratios
(0.63 and 0.67 to 0.25 and 0.53, respectively) and ICU and
hospital LOS were observed with use of an eICU program.35

Setup of an eICU
A variety of clinical process changes accompany the introduc-
tion of an eICU. The eICU program is not a single technology
or a single product. It is a set of tools and an innovation stream
that results in a technology-supported team approach to the
care of ICU patients. It can be transformational because
it reorganizes work processes and care models. The eICU
program does not replace bedside doctors and nurses or sig-
nificantly alter their roles and responsibilities, but instead adds
an additional layer of support for the care of the most critically
ill patients whose conditions can, and often do, change rapidly.
Critically ill patients require frequent assessment throughout
the day and night, which is probably an important mechanism
by which intensivists improve care by providing regular avail-
ability and focus on the constant titration of critical care. This
allows for more rapid interventions such as fine-tuning thera-
pies and prompt detection of emerging problems, which if not
Fig. 44.1  eICU programs are used to monitor critically ill patients in
intensive care units, emergency departments, and postanesthesia care
recognized early can lead to life-threatening complications.
units, as well as other areas in university hospitals, community hospitals, These tasks (assessment/titration/intervention) represent a
and critical access hospitals. core activity of the remote care team and allow health systems
to leverage existing intensivists to provide 24/7 coverage to all
critically ill patients.
The eICU program is designed to ensure intensivist involve-
Table 44.1  Realized Benefits of the eICU ment in care for every ICU patient, and to provide the inten-
Program sivists with tools that improve efficiency. Cardiologists
frequently say “time is heart.” Stroke specialists now say “time
CLINICAL BENEFITS is brain.” In trauma there is the concept of “the golden hour”
Decreased severity-adjusted mortality to prevent major systemic damage. In the ICU, there fre-
Decreased adverse outcomes in low-risk patients quently are many important things going on such that only
Decreased frequency of complications emergent situations truly generate rapid responses and many
Increased compliance with best practices
pathophysiologic processes progress further than is ideal
FINANCIAL BENEFITS before they are readdressed. Often the traditional systems that
Decreased severity-adjusted ICU LOS are in place to take care of patients become overwhelmed. In
Decreased severity-adjusted floor LOS this setting, improvements in efficiency lead to improved care
Decreased cost-per-day for remaining days in the ICU as patient-related events are addressed at an earlier stage in
Decreased nursing staff turnover
Avoided capital for new ICU beds their evolution.
Improved throughput
Increased hospital revenue
Increased physician pro-fee billing eICU Technology
ICU, Intensive care unit; LOS, length of stay. An essential part of the eICU system that leads to improved
efficiency is the technology infrastructure that manages the
flow of clinical information from the hospitals to the eICU
center. This includes the real-time data from bedside moni-
contribute to potential clinical and financial benefits including tors, and all the supporting data (i.e., lab results, medications,
at large academic medical centers and level I trauma centers patient registration and administration data, and medical
(Table 44.1). For example, in the 30-bed surgical trauma unit images such as x-rays and magnetic resonance images), and
at the Hospital of the University of Pennsylvania, Kohl et al audio and video streams for remote observation of patients
compared care in the year before implementation of the eICU (Fig. 44-2). Using evidence-based algorithms and clinician-set
program with care in the 2 1 2 years following. In an ICU that parameters, the software parses patient data in real time to
already was staffed with dedicated intensivists, fellows, and identify incipient clinical events and alerts clinicians before
Section VII—Computers, Engineering, and the Future 431

many significant adverse events develop. Smart Alert prompts technology. The hospital admitting or managing physician
evaluate patients’ electronic monitoring data in real time and remains the attending physician of record and is responsible
flag potentially worrisome trends and readings that exceed for the patient care plan and determining which on-site con-
specified thresholds (Fig. 44-3). This focuses the intensivist’s sultants assist with care delivery. This concept is particularly
attention and can promote timely intervention to avoid crises. important in understanding how an eICU program staffed
The eCareManager System provides a “dashboard-like” inter- with intensivists can be of great assistance in the care of sub-
face that helps the remote intensivists coordinate patient care specialty patients such as the neurocritical care patient. It is
with doctors and nurses at the bedside. An online decision- not the role or responsibility of the eICU team to determine
support tool called The Source draws on the latest evidence- the specific needs of an ICU patient or to define the primary
based care guidelines. In addition, the eCareManager system course of treatments; those evaluations and decisions are best
is built upon a relational database, which allows data organiza- done at the bedside by the physicians and surgeons who know
tion to drive best practices such as simplified multidisciplinary the patient. The role of the eICU team is to closely monitor
rounds and the tracking of performance according to evidence- the patient’s data for evidence that care needs are changing or
based best practices. that expected data are available, and then to alter or initiate
care in accordance with the previously expressed plan and
direction of the bedside health care providers. If there is a
Clinical Transformation Process question whether the original plan is still best considering the
The real significance of the eICU program lies in the way it changes or new data, the eICU intensivist seeks clarification
changes care delivery. The technology is only enabling; the key from the managing bedside team and then resumes giving
is a clinical process transformation that accompanies the support accordingly.

eICU Team Responsibilities


The eICU team can regularly fine-tune the acute care of ICU
patients through observation of trends. Consider a patient
who presents with an intracerebral hemorrhage. The plan of
care following initial evaluation by a neurologist and neuro-
surgeon may include admission to an ICU with an ICP
monitor, frequent follow-up neurologic evaluation, blood
pressure control, and intubation for airway protection. The
remote eICU physician monitoring the patient follows the
plan of the bedside team and monitors ICP and cerebral per-
fusion pressure (CPP) in the patient, and regularly confers
with the bedside nurse via camera to determine stability in the
Fig. 44.2  eICU clinicians monitor patient data from a remote site, using patient’s neurologic findings. Several hours into the patient’s
Smart Alerts, real-time monitor data, laboratory results, and other ICU course, the eICU staff is alerted via Smart Alerts (see
ancillary test results to intervene on the patients’ behalf. Fig. 44-3) that, although the patient’s vital signs are not out

Reload Patient Status Font Size: 16-point Select Units Log out

Selected Patient -Additional Alert Details- Selected Units:


<No Selection> Please select an active alert to see the additional information There are 10 unit(s) being
monitored in 6 hospital(s).

Time Site Bed Patient HR/MAP MAP Limit MAP Trend HR Limit HR Trend 02 Sat/RR

11:59 M-MICU 3 GEHRIG, L HR 175, MAP 44

11:55 C-SICU 8 COBB, T H [101] 124

11:47 M-CICU 12 WAGNER, H H [130] 132

11:45 G-CICU 4 CLEMENTE, R H [100] 104

11:30 G-MICU 5 MAYS, W H [120] 126 02 [92] 90, RR [26] 30

11:28 M-SICU 10 STARGELL, W L [70] 65

11:20 M-MICU 3 MUSIAL, S L [92] 77

Fig. 44.3  Smart Alerts software continuously track vital signs and trends, and notify eICU clinicians of aberrancies, enabling quick action and thereby
potentially preventing problems from escalating. H, High; HR, heart rate; L, low; MAP, mean arterial pressure; O2 Sat, oxygen saturation; RR, respiratory
rate.
432 Section VII—Computers, Engineering, and the Future

of the set range of acceptable values, the trend has changed of therapies, as necessary, to achieve care plan objectives;
such that the heart rate has dropped 25 beats per minute and (3) identification of emerging problems and initiation of
the mean arterial pressure has risen 30 mm Hg. Inspection of appropriate countermeasures; (4) facilitation of communica-
the ICP trend reveals that it has increased. Concerned about tion among members of the care team; and (5) responsibility
a potential Cushing response, the eICU physician contacts the for best practice compliance.38
bedside nurse, orders an emergent repeat head computed Several lines of evidence suggest that allowing the remote
tomography (CT) scan, and alerts the attending physician of team to fulfill these responsibilities is associated with improved
record, consulting neurologist, and neurosurgeon. While ICU practices in both urban and rural hospitals, including
awaiting formal input from the neurologic experts, the eICU many evidence-based practices, such as DVT prophylaxis
physician begins simple measures to treat elevated ICP, such compliance, glycemic control,37 and 6- and 24-hour care
as sedation and elevation of the head of bed. When the CT bundles in patients with sepsis, often resulting in reduced
scan is completed and demonstrates extension of the hemor- mortality.39 In addition to best practice improvements,
rhage with worsening edema, the eICU intensivist confers improvements in other aspects of patient care have been
with the neurologist or neurosurgeon immediately to discuss observed with an eICU program. For example, Shafer et al
further patient treatment. observed a reduction in the number of cardiopulmonary
The eICU team also can intervene in emergencies and arrests in their ICUs and the number of patients who died of
manage best practices (e.g., glucose control, deep vein throm- cardiopulmonary arrest once an eICU program was instituted
bosis (DVT) and stress ulcer prophylaxis, low tidal volume at their hospital.40
ventilation). In many eICU programs, the attending physician
defines in advance the requirements for communication by
the eICU center before nonemergent actions are taken for The Impact Beyond the ICU
their patients. This may range from specific review of all non- There are several potential advantages to a tele-ICU that
emergent decisions to allowing the eICU intensivist to act as extend beyond the ICU. For example, tele-ICU care may con-
a surrogate decision maker, calling the bedside team as indi- tribute to more judicious decisions about which patients are
cated by specific conditions and degree of expertise in those to benefit from ICU admission and which patients should
decisions. Examination of communication methods suggests receive palliative care, that is, better resource use.41 Further-
better outcomes when the remote care team is allowed more more, any telemedicine program means that there is a large
autonomy to address new problems and institute best prac- volume of electronic data that is collected in real time. Used
tices.36 Independent of communication preferences, the eICU correctly, this can drive clinical practice improvement through
physician reviews all patient data at regular intervals (from “instantaneous” quality metrics. Alternatively, the aggregated
hourly for the sickest patients to every 4 to 6 hours for the data can be used to answer important research questions. For
most stable). Each eICU caregiver monitors and delivers care this to be a reality, however, informatics infrastructure and
from a workstation comprised of several desktop monitors protection of patient privacy must be addressed.42,43
that provide access to the eCareManager application suite, A remote eICU monitoring program can help increase
including audiovisual tools, the hospital information system, attending physician oversight and availability. This may be
picture archiving and communication system, and remote- particularly helpful in hospitals with residency programs and
view applications for the bedside monitors (see Fig. 44.2). enhance education.44 House officers have an immediate point
Caregivers consist of physicians, nurses, clerical staff, and, of contact to discuss complex cases or even ask simple ques-
in some programs, a pharmacologist or pharmacist. Staff tions. Often a new house officer may be hesitant to call an
numbers vary based on the number of beds in the network. attending physician in the middle of the night, especially if it
With smaller networks (less than 70 beds being monitored), is not clear to the house officer that the question is urgent.
the remote care team is comprised of one intensivist and one The eICU physician can serve as a buffer for the house officer,
critical care nurse. As the number of monitored beds increases, even if it is simply to advise the house officer whether an
the number of care providers increases, with additional nurses issue for the bedside attending physician should be addressed
typically added after 60 to 70 beds are reached and a second immediately or can wait until morning. Intensivists in the
physician (not necessarily a second intensivist) added after eICU center have time to educate house officers and can
100 to 120 beds. There can be variability in how tasks are quickly email or fax relevant information including publica-
allocated among the various care providers, however, several tions, using The Source for topical information or as a
core features must be in place to achieve quality goals, such as conduit to the medical literature and papers of interest. Staff
a comprehensive daily care plan that addresses key clinical in the eICU center should be regarded as a resource for house
issues (e.g., ventilator weaning, nutrition, risk assessment, staff, but should not usurp house staff autonomy. In that
social issues).37 The care plan is developed by the on-site team regard, eICU physicians are trained to include house officers
and optimally encompasses the input of all relevant care pro- in any decision making that is of educational value. In addi-
viders. The eICU team monitors the care plan and assists with tion, the presence of the eICU clinicians can help reduce the
implementing the various components, and can take primary stress that critical care nurses often experience by providing
responsibility for processes such as compliance with best prac- them a constant safety net. Together these resources for nurse
tices. High compliance rates can be achieved by centralizing and house staff can provide better outcomes for ICU patients
these essential activities and assigning them to dedicated per- by eliminating the wait time often needed to reach a physi-
sonnel. Because the remote team has no other clinical respon- cian. An eICU program allows an “ICU without walls”; it is
sibilities, it is efficient and continuously attentive. The already available in some emergency departments (EDs) and
responsibilities of the remote team include (1) continuous postanesthesia care units (PACUs) and is being deployed
monitoring of the progress of each patient; (2) titration through wireless mobile units to assist with rapid response in
Section VII—Computers, Engineering, and the Future 433

general acute care settings. Several eICU programs also are smaller community hospitals.56 Those issues tackled by the
adding stroke assessment and intervention programs using eICU program are fundamental to medical care today:
their eICU technology and operational backbones. Office-  The shortage of appropriate specialists
based or at-home stroke specialists then can work collabora-  The shortage and high burn-out rate of specialized nurses
tively with the existing eICU teams and so more rapidly assess  The need to handle large amounts of data and filter
and intervene in acute stroke cases from the ED through the
through the noise and find the important data that needs
ICU care stages.
attention
 The need for decision support systems to synthesize evolv-
ing medical knowledge and supply it at the point of care
Neurocritical Care Today  The demand for detailed documentation of care, and the
In Support of Dedicated Neurointensivists cost and complexity of increasingly sick patients in the
hospital
In the same way patients with acute cardiac dysfunction are
best cared for in coronary care units and by cardiologists,
patients with acute neurologic disorders should be cared for Most NCCUs are equipped with advanced treatments that
in an NCCU with dedicated staff. Neurointensivists combine may improve prognosis in patients with severe neurologic
knowledge of neurologic diseases with the techniques of conditions that can threaten both survival and brain function.
intensive care and defragment care by focusing on the inter- The advancement of the specialty of neurocritical care, the
play between the brain and other systems. Specific areas of creation of specialized NCCUs, and the continued expansion
expertise of neurocritical care specialists include ICP manage- in number and size of eICU programs are all occurring in
ment, hemodynamic augmentation to improve cerebral blood response to the growing needs for evidence-based, timely care
flow, therapeutic hypothermia, and advanced neuromonitor- in an efficient, effective manner to produce the best possible
ing (e.g., continuous electroencephalography, brain-tissue outcomes for patients, health care providers, and health
oxygen, and microdialysis).45 Patients with neurologic disor- systems. This creates an opportunity for synergy between
ders such as TBI, intracerebral hemorrhage, and stroke who these evolving concepts.
are admitted to NCCUs appear to have reduced mortality and The future likely has the most complex neurocritical care
better functional outcomes than those admitted to general patients cared for in specialized units by specialized clinicians
ICUs.46-48 In addition, there is reduced LOS and lower overall using specialized monitoring equipment. It also likely contin-
costs when patients with acute neurologic disorders are cared ues to have some highly complex patients in less complex set-
for by neurointensivists in a dedicated NCCU.49-52 These out- tings cared for by less specialized caregivers. Less complex but
comes likely are associated with developments in several areas, quite ill or injured patients may also remain in nonspecialized
including improved understanding of disease pathophysiol- settings. In short, critically ill patients will continue to exist in
ogy, neuromonitoring, new and effective treatments for many setting from complex specialized ICUs to general med-
complex neurologic conditions, and staff with specialized surg units to PACUs to waiting for beds in EDs or neuroradiol-
neurocritical care skills. Multimodality neuromonitoring may ogy suites. These patients may be monitored by and their care
play an important role because the importance of early iden- supported by dedicated eICU teams led by intensivists. For
tification and treatment of secondary insults after acute brain example, again consider a patient who presents with an intra-
injury is well established.53 More important than the monitor cerebral hemorrhage to a small critical access hospital. Such a
is the interpretation of the data and what is done with the facility may have a general intensivist on staff but is not a neu-
information. This is where the neurointensivist becomes rointensivist. However, if this critical-access facility is part of
essential; monitoring enables the neurointensivist to antici- an eICU program, one centered at a large community or aca-
pate, prevent, and treat potentially dangerous secondary demic hospital, the ED staff can contact the eICU intensivist
insults. However, there still is a national shortage of neuroin- for advice when the patient presents to the ED. The intensivist
tensivists and many hospitals do not have dedicated NCCUs; is able to quickly assess the patient, determine the need for and
that is, few centers have the expertise and technology to arrange immediate transfer to a tertiary care setting, and at the
provide this level of care. Thus for the majority of neurocriti- same time can arrange neuroimaging upon arrival and confer
cal care patients there is a gap in actual care compared to what with the on-call neurologist or neurosurgeon at the referral
is now known to be the best care; constant surveillance by center; that is, the eICU intensivist can coordinate patient care
specialized nurses, immediate availability of neuroimaging, and communication between health care providers.
specially trained physicians, and specialized interventions that When the patient is admitted to the ICU, the eICU clini-
together can provide constant observation and immediacy of cians can assist the bedside neurologic team in providing
action to detect and treat neurologic deterioration or patient- round-the-clock clinical surveillance and interpretation of
related factors that may aggravate the initial brain injury. In monitoring technology, and guide specialized treatments to
addition, the technology can be leveraged to enhance safety, reduce any immediate or delayed brain damage. Monitoring
reduce errors, or avoid adverse drug reactions.54,55 the injured brain has become an integral part of the manage-
ment of severely brain-injured patients in intensive care but
has evolved in recent years such that a variety of techniques
Possibilities for the Practice of now are available (Table 44.2). These monitoring techniques
Neurocritical Care with an eICU Program can provide the neurointensivist with crucial information
Innovations can help solve important problems. For example, about brain physiology and metabolism. Combined, these
several stroke networks based on telepresence have evolved so techniques (“multimodal monitoring”) can produce an even
that stroke expertise is immediately available in remote or more accurate overall picture. This approach, however, is labor
434 Section VII—Computers, Engineering, and the Future

Table 44.2  Potential Neurologic Procedures/ Outcome Analysis


Monitoring/Interventions by an eICU Data suggest that neurology-based ICUs may benefit from an
Program eICU program. Saint Lukes Health System in Kansas City
Neuro-ICU Monitoring Techniques implemented an eICU program in 2006 which monitored,
INTRACRANIAL PRESSURE MONITORING
among others, a neurosurgical ICU. APACHE III scores did
not change; however, compared with pre-eICU, in the year and
Intraventricular catheters
Fiberoptic monitors
a half following the start of eICU, risk-adjusted hospital mor-
Subarachnoid bolts tality for neurosurgical ICU patients (standardized mortality
Epidural monitors ratio, or observed to predicted mortality) decreased from 0.86
Continuous brain tissue monitoring and cerebral blood flow to 0.67 (leading to 15% fewer hospital deaths in that popula-
probes tion) and ICU LOS decreased from 1.52 to 1.11 (22% fewer
Jugular venous oxygen saturation monitoring
Invasive hemodynamic monitoring ICU days).57
Continuous electroencephalogram (EEG) monitoring
PROCEDURES MONITORED Conclusion
Ventricular drainage Treatment of TBI, stroke, and cerebral hemorrhage are just
Thrombolysis
Plasmaphersis some of the several pursuits of NCCUs and neurointensivists
Hypothermia today. Future directions include the clinical implementation
INTERVENTIONS
of brain resuscitation and brain-sparing therapies, sophisti-
cated monitoring of electrophysiologic and intracranial physi-
Administration of blood products for urgent indications ologic indices, and further understanding of the dysfunction
Code supervision
Airway management of other organs that follows brain and nerve failure. The intro-
Fluid bolus for hypotension duction of neurointensivists and NCCUs is associated with
Ventilator management reduced hospital mortality and resource use. However, there
Management of seizure activity is no change overall in readmission rates or long-term mortal-
Intensive insulin therapy
Acute management of delirium, psychosis, or severe agitation
ity. This suggests that, although neurocritical care has evolved
Management of severe hypertension recently, there is further room for improvement. Implementa-
Culture-directed or empiric antibiotics tion of an eICU program potentially can help expand the
Pain management reach of neurointensivists and bring about needed improve-
ROUTINE FLUID AND ELECTROLYTE MANAGEMENT ments so that all neurocritical care patients can benefit from
Fluid bolus for low urine output the expertise of specially trained physicians.
Routine electrolyte replacement
Interpretation of electrocardiogram
Use of diuretics References
1. Menon D. Neurocritical care: turf label, organizational construct, or clinical
asset? Curr Opin Crit Care 2004;10:91–3.
2. Suarez J. Outcome in neurocritical care: advances in monitoring and
treatment and effect of a specialized neurocritical care team. Crit Care Med
intensive and often requires specialized knowledge to best 2006;34:S232–8.
interpret the information. This is something eICU staff can 3. Wright W. Putting neurocritical care units on the map. 2007. Neurocritical
help with, and through communication with the bedside Care Society: http://www.neurocriticalcare.org/files/public/
advocacyarticle4.pdf.
team, alert them of detrimental trends and variations, and 4. Kohn L, Corrigan J, Donaldson M. To err is human: building a safer health
intervene, if needed, to elicit corrections as directed by the system. In: IOM Committee on Healthcare in America, editor. Washington,
patient’s care plan developed by the bedside team. DC: The National Academies Press; 1999.
eICU clinicians can evaluate and intervene in response to 5. Committee on Quality of Health Care in America, IOM. Crossing the quality
chasm: a new health system for the 21st century. Washington, DC: The
compromise of any organ system (see Table 44.2). For example, National Academies Press; 2001.
suppose the patient with intracerebral bleed develops sympa- 6. Lwin AK, Shepard DS. Estimating lives and dollars saved from universal
thetic nervous system overdrive that may occur with intracra- adoption of the Leapfrog safety and quality standards: 2008 update.
nial hemorrhage. When this occurs it may be necessary to Washington, DC: Leapfrog Group; 2008.
stress the heart and avoid lower CPP to resuscitate the brain 7. Milstein A, Galvin RS, Delbanco SF, et al. Improving the safety of health
care: the Leapfrog initiative. Eff Clin Pract 2000;3(6):313–6.
and allow the heart function to recover later. eICU program 8. Gutsche JT, Kohl BA. Who should care for intensive care unit patients? Crit
technology allows careful monitoring to ensure the blood Care Med 2007;35(2):S18–S23.
pressure remains in a range that would not jeopardize CPP. 9. Kahn JM, Matthews FA, Angus DC, et al. Barriers to implementing the
An early warning system known as Smart Alert Prompts con- Leapfrog Group recommendations for intensivist physician staffing: a survey
of intensive care unit directors. J Crit Care 2007;22(2):97–103.
tinuously analyzes each patient’s vital signs and tracks changes 10. Pronovost PJ, Angus DC, Dorman T, et al. Physician staffing patterns and
and trends to identify problems that require rapid attention. clinical outcomes in critically ill patients: a systematic review. JAMA 2002;
These alerts allow the eICU team to respond immediately to 288:2151–62.
the slightest changes in the patient’s condition and quickly 11. Hanson C, Deutschman C, Anderson HL, et al. Effects of an organized
identify the most appropriate treatment. The criteria for critical care service on outcomes and resource utilization: a cohort study.
Crit Care Med 1999;27:270–4.
prompts can be individualized for each patient so that eICU 12. Pronovost PJ, Jenckes MW, Dorman T, et al. Organizational characteristics of
clinicians can maintain blood pressure or other parameters intensive care units related to outcomes of abdominal aortic surgery. JAMA
within a range specified by the bedside team. 1999;281:1310–17.
Section VII—Computers, Engineering, and the Future 435

13. Young MB, Birkmeyer JD. Potential reduction in mortality rates using an 32. Girard TD, Kress JP, Fuchs BD, et al. Efficacy and safety of a paired sedation
intensivist model to manage intensive care units. Eff Clin Pract 2000;3:284–9. and ventilator weaning protocol for mechanically ventilated patients in
14. Levy MN, Rapoport J, Lemeshow S, et al. Association between critical care intensive care (Awakening and Breathing controlled trial): a randomised
physician management and patient mortality in the intensive care unit. Ann controlled trial. Lancet 2008;371(9607):126–34.
Intern Med 2008;148:801–9. 33. Sairanen T, Soinila S, Nikkanen M, et al. Finnish Telestroke Task Force. Two
15. Grundy BL, Crawford P, Jones PK, et al. Telemedicine in critical care: an years of Finnish Telestroke: thrombolysis at spokes equal to that at the hub.
experiment in health care delivery. JACEP 1977;6:439–44. Neurology 2011;76(13):1145–52.
16. Grundy BL, Jones PK, Lovitt A. Telemedicine in critical care: problems in 34. Kohl BA, Gutsche JT, Kim P, et al. Effect of telemedicine on mortality and
design, implementation, and assessment. Crit Care Med 1982;10:471–5. length of stay in a university ICU. Crit Care Med 2007;35:A22.
17. Rosenfeld BA, Dorman T, Breslow MJ, et al. Intensive care unit telemedicine: 35. Zawada ET, Herr P, Erickson D, et al. Financial benefit of a teleintensivist
alternate paradigm for providing continuous intensivist care. Crit Care Med program to a rural health system. Chest 2007;132:444.
2000;28:3925–31. 36. Cowboy EN, Rajamani S, Jamil MG, et al. Impact of remote ICU
18. Breslow MJ, Rosenfeld BA, Doerfler M, et al. Effect of a multiple-site management on ventilator days. Crit Care Med 2005;33:A1.
intensive care unit telemedicine program on clinical and economic 37. Breslow M. Remote ICU care programs: current status. J Crit Care 2007;22:
outcomes: an alternative paradigm for intensivist staffing. Crit Care Med 66–76.
2004;32:31–8. 38. Aaronson ML, Zawada ET, Herr P. Role of a telemedicine intensive care unit
19. Angus DC, Kelley M, Schmitz R, et al. Committee on Manpower for program (TISP) on glycemic control (GC) in seriously ill patients in a rural
Pulmonary and Critical Care Societies (COMPACCS). Caring for the health system. Chest 2006;130:226S.
critically ill patient: current and projected workforce requirements for care 39. Rincon T, Bourke G, Ikeda D. Centralized, remote care improves sepsis
of the critically ill and patients with pulmonary disease: can we meet the identification, bundle compliance and outcomes. Chest 2007;132:557b.
requirements of an aging population? JAMA 2000;284(21):2762–70. 40. Shaffer JP, Breslow MJ, Johnson JW, et al. Remote ICU management
20. Gajic O, Afessa B, Hanson AC, et al. Effect of 24-hour mandatory versus improves outcomes in patients with cardiopulmonary arrest. Crit Care Med
on-demand critical care specialist presence on quality of care and family and 2005;33:A5.
provider satisfaction in the intensive care unit of a teaching hospital. Crit 41. Ramsay A, Rabinowitz T. Developing a rural telemedicine program to
Care Med 2008;36 (1):36–44. provide palliative care consultation. Paper presented at 14th Annual Meeting
21. Vespa PM, Miller C, Hu X, et al. Intensive care unit robotic telepresence and Exposition of the American Telemedicine Association, Las Vegas, NV;
facilitates rapid physician response to unstable patients and decreased cost in April 27, 2009.
neurointensive care. Surg Neurol 2007;67(4):331–7. 42. Gamble KH. Critical care network. ICU telemedicine can help ease the
22. Thomas EJ, Lucke JF, Wueste L, et al. Association of telemedicine for remote burden of caring for critically ill patients—provided all the right pieces are
monitoring of intensive care patients with mortality, complications, and in place. Healthcare Inform 2009;26(12):26–30.
length of stay. JAMA 2009;302(24):2671–78. 43. McShea M, Holl R, Badawi O, et al. The eICU research institute—a
23. New England Healthcare Institute. Tele-ICUs: remote management in collaboration between industry, health-care providers, and academia. IEEE
intensive care units. Cambridge, MA: Massachusetts Technology Eng Med Biol Mag 2010;29(2):18–25.
Collaborative and Health Technology Center; 2007. 44. Conde JG, De S, Hall RW, et al. Telehealth innovations in health education
24. Berenson RA, Grossman JM, November EA. Does telemonitoring of and training. Telemed J E Health 2010;16(1):103–6.
patients—the eICU—improve intensive care? Health Aff (Millwood) 2009; 45. Rincon F, Mayar SA. Neurocritical care: a distinct discipline? Curr Opin Crit
28(5):w937–47. Care 2007;13:115–21.
25. Gracias V. Outcomes of SICU patients after implementation of an electronic 46. Diringer MN, Edwards DF. Admission to a neurologic/neurosurgical
ICU (“eICU”) system and off-site intensivist. Paper presented at 42nd World intensive care unit is associated with reduced mortality rate after
Congress of Surgery, Montreal, Quebec; August 2007. intracerebral hemorrhage. Crit Care Med 2001;29:635–40.
26. Zawada ET Jr, Herr P, Larson D, et al. Impact of an intensive care unit 47. Mirski MA, Chang CW, Cowan R. Impact of a neuroscience intensive care
telemedicine program on a rural health care system. Postgrad Med 2009; unit on neurosurgical patient outcomes and cost of care: evidence-based
121(3):160–70. support for an intensivist-directed specialty ICU model of care. J Neurosurg
27. Van der Kloot T, Riker R, Goran S, et al. Improved hospital survival after Anesthesiol 2001;13:83–92.
implementing a remote ICU telemedicine program. Paper presented at 14th 48. Elf K NP, Enblad P. Outcome after traumatic brain injury improved by an
Annual Meeting and Exposition of the American Telemedicine Association, organized secondary insult program and standardized neurointensive care.
Las Vegas, NV; April 27, 2009. Crit Care Med 2002;30:2129–34.
28. McCambridge M, Jones K, Paxton H, et al. Association of health information 49. Mirski MA, Chang CW, Cowan R. Impact of a neuroscience intensive care
technology and teleintensivist coverage with decreased mortality and unit on neurosurgical patient outcomes and cost of care: evidence-based
ventilator use in critically ill patients. Arch Intern Med 2010;170(7):648–53. support for an intensivist-directed specialty ICU model of care. J Neurosurg
29. Morrison JL, Cai Q, Davis N, et al. Clinical and economic outcomes of the Anesthesiol 2001;13:83–92.
electronic intensive care unit: results from two community hospitals. Crit 50. Suarez JI. Outcome in neurocritical care: advances in monitoring and
Care Med 2010;38(1):2–8. treatment and effect of a specialized neurocritical care team. Crit Care Med
30. Young LB, Chan PS, Lu X, et al. Impact of telemedicine intensive care unit 2006;34:S232–8.
coverage on patient outcomes: a systematic review and meta-analysis. Arch
Intern Med 2011;171(6):498–506. A complete list of references for this chapter can be found online at
31. Luyt CE, Combes A, Aegerter P, et al. Mortality among patients admitted to www.expertconsult.com.
intensive care units during weekday day shifts compared with “off ” hours.
Crit Care Med 2007;35(1):3–11.
Section VII—Computers, Engineering, and the Future 435.e1

References 26. Zawada ET Jr, Herr P, Larson D, et al. Impact of an intensive care unit
telemedicine program on a rural health care system. Postgrad Med
1. Menon D. Neurocritical care: turf label, organizational construct, or clinical 2009;121(3):160–70.
asset? Curr Opin Crit Care 2004;10:91–3. 27. Van der Kloot T, Riker R, Goran S, et al. Improved hospital survival after
2. Suarez J. Outcome in neurocritical care: advances in monitoring and implementing a remote ICU telemedicine program. Paper presented at 14th
treatment and effect of a specialized neurocritical care team. Crit Care Med Annual Meeting and Exposition of the American Telemedicine Association,
2006;34:S232–8. Las Vegas, NV; April 27, 2009.
3. Wright W. Putting neurocritical care units on the map. 2007. Neurocritical 28. McCambridge M, Jones K, Paxton H, et al. Association of health information
Care Society: http://www.neurocriticalcare.org/files/public/ technology and teleintensivist coverage with decreased mortality and
advocacyarticle4.pdf. ventilator use in critically ill patients. Arch Intern Med 2010;170(7):648–53.
4. Kohn L, Corrigan J, Donaldson M. To err is human: building a safer health 29. Morrison JL, Cai Q, Davis N, et al. Clinical and economic outcomes of the
system. In: IOM Committee on Healthcare in America, editor. Washington, electronic intensive care unit: results from two community hospitals. Crit
DC: The National Academies Press; 1999. Care Med 2010;38(1):2–8.
5. Committee on Quality of Health Care in America, IOM. Crossing the quality 30. Young LB, Chan PS, Lu X, et al. Impact of telemedicine intensive care unit
chasm: a new health system for the 21st century. Washington, DC: The coverage on patient outcomes: a systematic review and meta-analysis. Arch
National Academies Press; 2001. Intern Med 2011;171(6):498–506.
6. Lwin AK, Shepard DS. Estimating lives and dollars saved from universal 31. Luyt CE, Combes A, Aegerter P, et al. Mortality among patients admitted to
adoption of the Leapfrog safety and quality standards: 2008 update. intensive care units during weekday day shifts compared with “off ” hours.
Washington, DC: Leapfrog Group; 2008. Crit Care Med 2007;35(1):3–11.
7. Milstein A, Galvin RS, Delbanco SF, et al. Improving the safety of health 32. Girard TD, Kress JP, Fuchs BD, et al. Efficacy and safety of a paired sedation
care: the Leapfrog initiative. Eff Clin Pract 2000;3(6):313–6. and ventilator weaning protocol for mechanically ventilated patients in
8. Gutsche JT, Kohl BA. Who should care for intensive care unit patients? Crit intensive care (Awakening and Breathing controlled trial): a randomised
Care Med 2007;35(2):S18–S23. controlled trial. Lancet 2008;371(9607):126–34.
9. Kahn JM, Matthews FA, Angus DC, et al. Barriers to implementing the 33. Sairanen T, Soinila S, Nikkanen M, et al. Finnish Telestroke Task Force. Two
Leapfrog Group recommendations for intensivist physician staffing: a survey years of Finnish Telestroke: thrombolysis at spokes equal to that at the hub.
of intensive care unit directors. J Crit Care 2007;22(2):97–103. Neurology 2011;76(13):1145–52.
10. Pronovost PJ, Angus DC, Dorman T, et al. Physician staffing patterns and 34. Kohl BA, Gutsche JT, Kim P, et al. Effect of telemedicine on mortality and
clinical outcomes in critically ill patients: a systematic review. JAMA length of stay in a university ICU. Crit Care Med 2007;35:A22.
2002;288:2151–62. 35. Zawada ET, Herr P, Erickson D, et al. Financial benefit of a teleintensivist
11. Hanson C, Deutschman C, Anderson HL, et al. Effects of an organized program to a rural health system. Chest 2007;132:444.
critical care service on outcomes and resource utilization: a cohort study. 36. Cowboy EN, Rajamani S, Jamil MG, et al. Impact of remote ICU
Crit Care Med 1999;27:270–4. management on ventilator days. Crit Care Med 2005;33:A1.
12. Pronovost PJ, Jenckes MW, Dorman T, et al. Organizational characteristics of 37. Breslow M. Remote ICU care programs: current status. J Crit Care
intensive care units related to outcomes of abdominal aortic surgery. JAMA 2007;22:66–76.
1999;281:1310–17. 38. Aaronson ML, Zawada ET, Herr P. Role of a telemedicine intensive care unit
13. Young MB, Birkmeyer JD. Potential reduction in mortality rates using an program (TISP) on glycemic control (GC) in seriously ill patients in a rural
intensivist model to manage intensive care units. Eff Clin Pract 2000;3: health system. Chest 2006;130:226S.
284–9. 39. Rincon T, Bourke G, Ikeda D. Centralized, remote care improves sepsis
14. Levy MN, Rapoport J, Lemeshow S, et al. Association between critical care identification, bundle compliance and outcomes. Chest 2007;132:557b.
physician management and patient mortality in the intensive care unit. Ann 40. Shaffer JP, Breslow MJ, Johnson JW, et al. Remote ICU management
Intern Med 2008;148:801–9. improves outcomes in patients with cardiopulmonary arrest. Crit Care Med
15. Grundy BL, Crawford P, Jones PK, et al. Telemedicine in critical care: an 2005;33:A5.
experiment in health care delivery. JACEP 1977;6:439–44. 41. Ramsay A, Rabinowitz T. Developing a rural telemedicine program to
16. Grundy BL, Jones PK, Lovitt A. Telemedicine in critical care: problems in provide palliative care consultation. Paper presented at 14th Annual Meeting
design, implementation, and assessment. Crit Care Med 1982;10:471–5. and Exposition of the American Telemedicine Association, Las Vegas, NV;
17. Rosenfeld BA, Dorman T, Breslow MJ, et al. Intensive care unit telemedicine: April 27, 2009.
alternate paradigm for providing continuous intensivist care. Crit Care Med 42. Gamble KH. Critical care network. ICU telemedicine can help ease the
2000;28:3925–31. burden of caring for critically ill patients—provided all the right pieces are
18. Breslow MJ, Rosenfeld BA, Doerfler M, et al. Effect of a multiple-site in place. Healthcare INFORM 2009;26(12):26–30.
intensive care unit telemedicine program on clinical and economic 43. McShea M, Holl R, Badawi O, et al. The eICU research institute—a
outcomes: an alternative paradigm for intensivist staffing. Crit Care Med collaboration between industry, health-care providers, and academia. IEEE
2004;32:31–8. Eng Med Biol Mag 2010;29(2):18–25.
19. Angus DC, Kelley M, Schmitz R, et al. Committee on Manpower for 44. Conde JG, De S, Hall RW, et al. Telehealth innovations in health education
Pulmonary and Critical Care Societies (COMPACCS). Caring for the and training. Telemed J E Health 2010;16(1):103–6.
critically ill patient: current and projected workforce requirements for care 45. Rincon F, Mayar SA. Neurocritical care: a distinct discipline? Curr Opin Crit
of the critically ill and patients with pulmonary disease: can we meet the Care 2007;13:115–21.
requirements of an aging population? JAMA 2000;284(21):2762–70. 46. Diringer MN, Edwards DF. Admission to a neurologic/neurosurgical
20. Gajic O, Afessa B, Hanson AC, et al. Effect of 24-hour mandatory versus intensive care unit is associated with reduced mortality rate after
on-demand critical care specialist presence on quality of care and family and intracerebral hemorrhage. Crit Care Med 2001;29:635–40.
provider satisfaction in the intensive care unit of a teaching hospital. Crit 47. Mirski MA, Chang CW, Cowan R. Impact of a neuroscience intensive care
Care Med 2008;36(1):36–44. unit on neurosurgical patient outcomes and cost of care: evidence-based
21. Vespa PM, Miller C, Hu X, et al. Intensive care unit robotic telepresence support for an intensivist-directed specialty ICU model of care. J Neurosurg
facilitates rapid physician response to unstable patients and decreased cost in Anesthesiol 2001;13:83–92.
neurointensive care. Surg Neurol 2007;67(4):331–7. 48. Elf K NP, Enblad P. Outcome after traumatic brain injury improved by an
22. Thomas EJ, Lucke JF, Wueste L, et al. Association of telemedicine for remote organized secondary insult program and standardized neurointensive care.
monitoring of intensive care patients with mortality, complications, and Crit Care Med 2002;30:2129–34.
length of stay. JAMA 2009;302(24):2671–78. 49. Mirski MA, Chang CW, Cowan R. Impact of a neuroscience intensive care
23. New England Healthcare Institute. Tele-ICUs: remote management in unit on neurosurgical patient outcomes and cost of care: evidence-based
intensive care units. Cambridge, MA: Massachusetts Technology support for an intensivist-directed specialty ICU model of care. J Neurosurg
Collaborative and Health Technology Center; 2007. Anesthesiol 2001;13:83–92.
24. Berenson RA, Grossman JM, November EA. Does telemonitoring of 50. Suarez JI. Outcome in neurocritical care: advances in monitoring and
patients—the eICU—improve intensive care? Health Aff (Millwood) treatment and effect of a specialized neurocritical care team. Crit Care Med
2009;28(5):w937–47. 2006;34:S232–8.
25. Gracias V. Outcomes of SICU patients after implementation of an electronic 51. Suarez JI, Zaidat OO, Suri MF, et al. Length of stay and mortality in
ICU (“eICU”) system and off-site intensivist. Paper presented at 42nd World neurocritically ill patients: impact of a specialized neurocritical care team.
Congress of Surgery, Montreal, Quebec; August 2007. Crit Care Med 2004;32:2311–7.
435.e2 Section VII—Computers, Engineering, and the Future

52. Varelas PN, Conti MM, Spanaki MV, et al. The impact of a neurointensivist- 55. Hassan E, Badawi O, Weber RJ, et al. Using technology to prevent adverse drug
led team on a semiclosed neurosciences intensive care unit. Crit Care Med events in the intensive care unit. Crit Care Med 2010;38(6 Suppl):S97–S105.
2004;32:2191–8. 56. Lai F. Stroke networks based on robotic telepresence. J Telemed Telecare
53. DeGeorgia MA, Krieger DW, Abou-Chebl DA, et al. Cooling for acute 2009;15(3):135–6.
ischemic brain damage, a feasibility trial of endovascular cooling. Neurology 57. Lem V, eICU medical director, St. Luke’s Health System, Kansas City.
2004;63:312–7. Personal communication.
54. Meidl TM, Woller TW, Iglar AM, et al. Implementation of pharmacy services in
a telemedicine intensive care unit. Am J Health Syst Pharm 2008;65(15):1464–9.
VII
Chapter
45  

Medical Informatics
J. Claude Hemphill III, Marco D. Sorani, Stuart Russell, and Geoffrey T. Manley

Every NCCU already uses some form of informatics technol­


Introduction ogy for these purposes. However, in most circumstances, this
The term medical informatics frequently is used to describe “technology” consists of monitors that display but do not
systems applied to hospital and physician office record capture and analyze the data. Often raw data are simply visu­
keeping, operations, and regulatory documentation, such as ally extracted from the monitor and recorded on a paper chart
electronic medical records (EMR), computerized order entry at defined intervals. Bedside nurses and physicians then review
(CPOE), and databases for billing or review by regulatory this paper record of the data and rely on their expertise and
bodies such as The Joint Commission. However, these systems intuition, without the aid of additional analytic tools, to apply
generally provide an electronic recapitulation of written it to clinical care. At a simple level, this leads to data cleaning
records rather than new insight into disease treatments. In because obvious artifacts, for example, a monitor that was
the context of monitoring in neurocritical care, medical infor- turned off during patient transport, are accounted for. The
matics is more appropriately defined as the acquisition, presence of large amounts of data that require complex inte­
storage, and analysis of neuromonitoring data to (1) facilitate gration and robust analytic tools is now a routine problem
knowledge and discovery about pathogenesis and treatment for the corporate world, such as the financial, airline, and
of disease, and (2) improve methods to integrate the com­ computer industries. These industries have made significant
plex array of clinical, physiologic, and genetic data into advances in information technology over the past 20 years.
practical bedside care.1 Unfortunately, although monitoring— Remarkably, only recently have attempts been initiated to
specifically neuromonitoring—has advanced tremendously in push critical care informatics beyond the paper chart and
the four decades since the origins of critical care, for the clinician intuition to address the questions posed earlier.
purposes of clinical care, data are considered in a way similar Figure 45.1 provides an example of the complexity and
to what was used a generation ago. However, it is clear that volume of even a short duration of critical care data.
the complexity of data in the neurocritical care unit (NCCU)
requires a fresh and novel approach to medical informatics
to complement the development of new neuromonitoring
What Is Not Known
paradigms.2,3 This chapter provides insight into this clinical The fundamental purpose of neurocritical care (for central
problem and describes some initial efforts toward this end. nervous system problems) is the identification, prevention,
and treatment of secondary brain injury. Physiology matters
and much of the focus in the NCCU is on management of
The Data Environment secondary brain insults (SBIs), which may include hypoten­
The NCCU is an extremely data-intense environment. The art sion, hypoxia, fever, elevated ICP, hyperglycemia, and seizures.
of neurocritical care is to use these data in real-time to make In fact, much of the moment-to-moment treatment provided
decisions about patient care.2 Patients routinely are monitored in neurocritical care involves ensuring that these physiologic
frequently or continuously using 30 or more parameters parameters, which can be seen as surrogate or intermediate
including blood pressure (BP), heart rate (HR), systemic targets with presumed impact on survival and long-term
arterial oxygen saturation (SaO2), respiratory rate (RR), tidal functional outcome, are within specified ranges. This often
volume, peak inspiratory pressures, intracranial pressure leads to a lengthy series of bedside orders that delineate accept­
(ICP), and body temperature (T), to name a few. Add to these able ranges of one parameter at a time (e.g., “Call medical
the variety of new advanced neuromonitoring tools such as doctor if systolic BP <90 mm Hg”; “Call medical doctor if
brain tissue oxygen tension (PbtO2), cerebral blood flow (CBF), SaO2 <94%”; “Call medical doctor if ICP >20 mm Hg”). The
and microdialysis parameters and several obvious questions bedside nurse then calls the physician and abnormalities in a
emerge: single parameter are treated in a reactive fashion (e.g., acet­
aminophen after a temperature elevation, mannitol after con­
1. What do we do with all these data? tinued ICP elevation) with little attention to how multiple
2. Are all of these data useful and important? parameters may interact. Yet, while patients are treated in a
3. How are artifacts “cleaned?” univariate manner, they live in a multivariate world in which
4. When and how often are the data collected? many factors interact to determine injury and outcome. Fur­
5. Which data are discarded and when? thermore, the clinical response is to a certain threshold rather
436 © Copyright 2013 Elsevier Inc. All rights reserved.
Section VII—Computers, Engineering, and the Future 437

Fig. 45.1  Critical care data are voluminous and complex. These graphs depict two days of continuous neurocritical care physiologic data from
a single patient represented on a time-series plot. The top panel shows blood pressure, central venous pressure, heart rate, and intracranial pressure.
The middle panel shows brain tissue oxygen, brain temperature, and jugular venous oxygen saturation. The bottom panel shows measures obtained
from the mechanical ventilator. These data are too noisy and compressed for visual inspection to be informative to identify single events, trends, or
relationships between parameters, thus necessitating summary measures, indices, or informatics-based analyses.

issues in a more comprehensive and sophisticated manner,


Table 45.1  Several Existing Neurocritical Care data acquisition tools well beyond the paper chart are required.
Informatics Questions This is simply not a recapitulation of a paper record in an
Do secondary brain insults (e.g., fever, elevated intracranial electronic form as occurs in many commercial EMRs4 but
pressure [ICP], hypotension) have a dose-response rather one in which all data, including patient records, labora­
relationship with outcome? tory analysis, imaging, and continuous physiologic data, are
Are there multivariate interactions and relationships among not only acquired but also archived, integrated, and synchro­
various physiologic parameters that influence outcome
beyond individual parameters?
nized.5,6 Many monitors used in the NCCU are stand-alone
Are there event signatures that would predict the future and self-contained; that is, they are not designed to operate
occurrence of a deleterious problem (e.g., elevated ICP or together. This has been an obstacle to NCCU informatics but
organ failure) that would allow proactive measures to should become less of a barrier since the American Society for
prevent its occurrence? Testing and Materials adopted the concept of an integrated
How do we integrate new measures (e.g., PbtO2)?
How often do we need to collect physiologic data to optimize clinical environment in 2009.7 Other chapters in this text focus
patient care? on the various monitors and the challenges to integration
How can we classify patients to understand their methods, and they should be seen as both complementary and
heterogeneity? required aspects of neurocritical care informatics.
How can we integrate physiologic data with data from
genomic, proteomic, imaging, and other types of data?
How can physiologic data be presented in real-time and in
a user-friendly manner to provide decision-enabling Are More Data Better? The Example
information to clinicians?
To what extent is prior medical history (from before
of Defining “Dose” of Secondary
hospitalization or earlier in hospital course) important in Brain Insults
interpreting current physiologic values and events?
Many studies have identified SBIs, such as hypotension,
hypoxia, elevated ICP, and fever, as being associated with poor
outcome after acute neurologic injury or illness.8-13 However,
than to trends over time and with little insight into the the manner in which these events have been defined often
complex interactions between various physiologic factors. varies widely across studies. Some studies have used any
Table 45.1 enumerates several questions about secondary occurrence of an SBI (e.g., systolic BP <90 mm Hg), while
brain injury in neurocritical care that remain elusive because others have used the number of times an event has occurred,
of limitations in existing approaches to informatics. It should and others have used the duration below a specific threshold.
be emphasized that a fundamental limitation that has ham­ Many times, the choice of a specific definition depends on the
pered attempts to address these issues has been inadequacies limitation imposed by the resolution, or frequency, with which
in data acquisition and storage. To begin to address these physiologic vital signs were collected in the medical record or
438 Section VII—Computers, Engineering, and the Future

research database. For example, in a study that identified fever New Measures Derived from Old—
as a predictor of hospital length of stay, only the maximum
daily temperature was recorded in the database used for study
The Example of PRx
analysis.10 In another study of the impact of hypotension in A compelling reason to pursue critical care informatics is the
the emergency department (ED) on outcome after TBI, the possibility that measurements derived from analysis of single
number of times the systolic BP was less than 90 mm Hg was or multiple parameters might be more informative than the
assessed, regardless of depth or duration of the episodes.14 All raw data of a single parameter alone. Although a very simpli­
of these various methods are attempts to define a “dose” of the fied example, the most common measurement routinely used
deleterious event, a concept familiar in pharmacology but in this fashion is cerebral perfusion pressure (CPP). CPP is
often not used in the NCCU. not measured directly but rather is a derived measure calcu­
In pharmacokinetics, dose or “exposure” is usually defined lated as the difference between the mean arterial pressure
as area under the curve of concentration versus time. Intui­ (MAP) and the ICP. CPP is routinely calculated by monitors,
tively this makes sense for SBIs; a longer duration of a lower charting systems, or by hand and is used in real time at the
BP should be more injurious than a brief event that just barely bedside in neurocritical care. However, current systems and
passed the threshold definition. However, appreciating this bedside charting methods are poorly equipped to derive mea­
dose is not readily apparent from current paper or electronic sures that involve anything more complex than simple math­
bedside charting. Even this seemingly simple assessment is an ematics. Even so, some early neurocritical care informatics
example of informatics technology; the resolution at which systems have provided the ability to assess some new param­
data are collected must be defined and analysis must be per­ eters derived from relationships between existing parameters.
formed on raw collected data to derive a summary dose PRx, the pressure reactivity index, is one example.
term—SBI burden. Figure 45.2 demonstrates the example of Cerebral autoregulation is a critical component to ensure
fever burden15 as area under the curve above a defined thresh­ adequate blood flow to normal and injured brain tissue.
old. Importantly, initial studies suggest that it makes a differ­ Impaired autoregulation is associated with worsened outcome
ence both how frequently data are collected and how “dose” is after TBI and management of CPP based on an individual
defined. It is not surprising that more frequent data collection patient’s autoregulation status may improve outcome. How­
identifies more SBIs. The extension of this is the conclusion ever, autoregulation status is difficult to assess from usual
that current intensive care unit (ICU) charting practices monitored parameters. PRx is a moving correlation coefficient
(usually writing down a value hourly) are missing many events between ICP and MAP and is one measure of whether auto­
of potential clinical significance. Furthermore, integrating regulation is intact or impaired.18,19 PRx ranges between −1
these data as “area under the curve” seems to be more infor­ and 1, with a positive number indicating some correlation
mative than just identifying presence or number of events. between the two parameters and a negative number indicating
Hypotension burden is more associated with outcome than an absence of correlation. If ICP and MAP are correlated, this
just number of events of hypotension.16 Fever burden is a means that the ICP increases as the MAP increases; this occurs
broader method of assessing normothermia control than just when the brain’s autoregulation status is impaired or the MAP
time over a threshold.15,17 Thus just the assessment of SBIs as is shifted off the normal autoregulation curve. If there is no
“dose” is a straightforward and seemingly simple example of correlation, then ICP and MAP are independent and auto­
ICU informatics that is still not clinically routine because of regulation is presumably intact. A PRx of less than 0.3 is often
shortcomings in data acquisition, charting, and bedside data considered as a threshold to assess intact autoregulation. PRx
analysis tools. has been used in studies of both TBI and nontraumatic intra­
cerebral hemorrhage to define optimal CPP, the CPP at which
PRx is brought within the noncorrelating range.20 Impor­
tantly, PRx is determined, not as a simple arithmetic function
39 between single current values of existing parameters, but
rather as a statistically analyzed relationship between time-
series data from existing parameters (ICP and MAP); that is,
Body temperature (°C)

38 it is a form of real-time multivariable modeling. Although a


seemingly straightforward example of a single derived para­
37
meter, PRx represents a leap forward in applying real-time
analytics to raw data, which then can be used to drive clinical
care. It does, however, require time-synchronization and inte­
36 gration of digital data acquisition to allow real-time analysis.
Such systems now exist for that purpose (e.g., ICM+).21
35
0 24 48 72 96 120 144 168 Advanced Critical Care
Hours from hospital admission Informatics I: Data-Driven
Fig. 45.2  Fever curve with cutpoint at 38.0° C (dashed line). Depending Methods for Prediction
on the measurement method, this patient could be considered: 
(1) positive for any fever during hospitalization, (2) had 15 febrile The previous examples represent fairly straightforward appli­
episodes, (3) had a total duration of fever of 78.2 hours, or (4) had a cations of medical informatics that become possible only
total fever dose (area under the curve [AUC]) of 28.1 degree-hours. AUC when data are collected electronically in a high-resolution
is calculated as the sum of all the solid regions over the 38.0° C cutpoint. format. The methods of analysis (e.g., area under the curve,
Section VII—Computers, Engineering, and the Future 439

correlation coefficients) are conventional in statistics and For example, Van Santbrink et al.24 investigated the prognostic
provide targeted summaries of measurements collected over value of a derived index known as brain tissue oxygen response
an extended period for a single patient. Far more is possible, (TOR). Patients with an unfavorable outcome had a higher
however. This section goes beyond summarization of mea­ TOR during the first 24 hours. Logistic regression analysis
surements into the realm of data-driven methods that inter­ supported the independent predictive value of TOR for
pret measurements and predict outcomes, or better yet events outcome. Oddo et al.25 examined the burden of brain hypoxia
that may occur in the ICU22 based on experience represented and intracranial hypertension after TBI and found that brain
by repositories of data collected from many patients. The hypoxia was associated with poor short-term outcome after
next section considers model-based methods that combine severe TBI independently of elevated ICP, low CPP, and injury
repository data with expert knowledge of physiology to reason severity. Amin and Kulkarni26 used the opinions of clinical
about underlying patient states and their anticipated evolu­ experts to develop a “fuzzy” GCS to predict cognitive recovery
tion over time. and found that it improved the information content for pre­
It is has been said that critical care is “the art of managing diction. Chambers et al.27 developed pressure-time indices and
extreme complexity.”23 Informatics should thus be seen as a identified age-related thresholds for ICP and CPP. These
potential tool to help manage this complexity. Both data- indices also were predictive of score on the Glasgow Outcome
driven and model-based methods do this by analyzing raw Scale (GOS) and could be compared independently of age.
data and generating outputs that are much closer to actionable Hornero et al.28 studied changes in ICP complexity, estimated
decisions. Of course, expert bedside clinicians do this already. by the approximate entropy of the ICP signal, as subjects
Through accumulated experience, they have learned that progressed from normal to elevated ICP. They concluded that
certain patterns in the data—often, patterns across time and decreased complexity coincides with episodes of hypertension,
across multiple parameters and monitoring tools (heretofore suggesting that regulatory mechanisms that govern ICP are
described as sensors)—may indicate future problems that can disrupted during acute rises. These different examples all dem­
be avoided with preemptive intervention. Advanced informat­ onstrate early efforts to develop informatics methods to aid in
ics techniques can support this process through data visualiza­ outcome prediction. Regression analysis, however, assumes
tion and by extracting features not readily available from raw that all data are of value and usually assumes a linear relation­
data and clinical charting and, to some extent, they can auto­ ship between the parameters and outcome. In addition it is
mate this process, at least in part. difficult to include time-series data in this form of analysis,
The fields of statistics and machine learning have produced and so other tools may be better suited to bioinformatics in
a huge range of methods to analyze data and make predic­ the NCCU.
tions. The methods are typically trained on previously col­
lected data and can be tested on unseen data to evaluate the
accuracy of their predictions. Most of the methods used in Neural Networks
medical informatics are supervised learning methods, which Real-world systems rarely behave in a simple, linear fashion
means that the training data contain ground-truth or expert- throughout their entire dynamic range, and physiologic
supplied answers for each of the instances. (Unsupervised systems are no exception. For example, if MAP is used as an
methods such as clustering algorithms are useful for explor­ input variable in an outcome predictor, poor outcomes would
atory analysis—“making sense” of a large data collection or be expected for very low and very high MAP with better out­
discovering unsuspected regularities through “data mining.”) comes in between. A linear model cannot generate this behav­
Within the class of supervised learning methods, many differ­ ior; instead, a nonlinear predictor such as a neural network is
ent prediction methods are used. Three are considered here: needed. A neural network is essentially a nonlinear input-
linear models (which include most indices), neural networks, output function with many tunable weights. (The “network”
and decision trees. simply displays the internal structure of the nonlinear func­
tion, with weights on the links and intermediate values com­
puted at the internal nodes.) The training process iteratively
Linear Models and Indices modifies the weights to improve the degree of fit between the
The simplest way to build a predictor from multivariate ICU network’s predictions and the actual training data.
data is to combine them linearly—that is, by taking a weighted Several recent studies have described the use of neural net­
sum. Linear regression sets the weights to give the best linear works in critical care patients29 and in particular those with
fit to a continuous output variable such as length of ICU stay, acute neurologic insults. For example, Vath et al.30 used neural
whereas logistic regression passes the sum through a soft networks to predict outcome after TBI for different combina­
threshold function to give an estimated probability for a tions of clinical and neuromonitoring parameters. Different
binary output variable such as mortality. The Glasgow Coma network models were composed using clinical data plus addi­
Score (GCS) can be thought of as a linear model that sums tional inputs for the number of events related to ICP and PbtO2
three terms: eye, verbal, and motor responses, with weights 4, levels.30 Lang et al.31 attempted to determine whether neural
5, and 6, respectively. Although there is no explicit outcome network modeling would improve outcome prediction com­
predictor, thresholds often are applied to the GCS to predict pared with logistic regression analysis. They confirmed the
outcome. As the GCS example illustrates, selecting or design­ importance of age, GCS, and hypotension in outcome predic­
ing highly predictive indices that aggregate or summarize the tion. Model performance was similar. Becalick and Coats32
raw data is a crucial contribution. developed a neural network using anatomic and physiologic
Recent techniques have included the development of indices, predictors. Head injury, age, and chest injury were the most
such as total fever burden, elevated ICP dose, or burden of important predictors. Li et al.33 compared three different
brain hypoxia, based on area-under-the-curve calculations. models to predict a particular TBI decision about open-skull
440 Section VII—Computers, Engineering, and the Future

GCS

,5 6,
7,
3,4 8

Pupillary Glucose level


response on the 2nd day

Unilaterally or
Bilaterally
bilaterally
normal
absent ≤169 mg/dL
>169 mg/dL
n = 119
Age
Unfav 96.6% SAH
1
>48 yrs Intracranial
≤48 yrs
Absent Present diagnosis
n = 12
Unfav 91.7% n = 67 n = 49
Glucose level Epidural hematoma Acute subdural hematoma
Unfav 1.5% Unfav 28.6%
on admission Diffuse brain injury Intracerebral hematoma
2
9 6

WBC count n = 28
≤179 mg/dL >179 mg/dL Unfav 89.3%
on admission
3
n = 10 n = 17 ≤15700 mm3 >15700 mm3
Unfav 10% Unfav 70.6%
8 4 n = 29 n = 14
Unfav 15.6% Unfav 42.8%
7 5
Fig. 45.3  Decision tree to predict outcome based on sequential partitioning of patients based on specific characteristics. Structure of the
tree is based on classification and regression analysis that creates a hierarchical set of binary decisions (in this case) based on optimizing outcome
prediction rather than separating patients based on clinically driven characteristics or cutpoints. SAH, Subarachnoid hemorrhage; Unfav, unfavorable
outcome; WBC, white blood cell. (Reprinted with permission from Rovlias A, Kotsou S. Classification and regression tree for prediction of outcome after severe head
injury using simple clinical and laboratory variables. J Neurotrauma 2004;21:886–93. Copyright Mary Ann Liebert, Inc.)

surgery: logistic regression, a multilayer perceptron neural in Figure 45.3. For example, a set of patients in a study might
network, and a radial basis function neural network. Sensitiv­ all be grouped together at the “top” of a tree. At each branch,
ity and specificity for the regression model were lower than the set of patients can be divided into two (e.g., “Is a patient
for the network models to predict a neurosurgeon’s decision, female or male?”) or more (e.g., “Did a patient receive 5 mg,
so confirming the need for a nonlinear predictor. Recently 10 mg, or 25 mg of drug?”) subsets. These subsets can further
Nelson et al.34 used mixed effects models and nonlinear (arti­ be subdivided. The nature and sequence of the rules can be
ficial neural networks) analysis to examine microdialysis data optimized according to various algorithms. Ultimately, the
in TBI. This analysis showed multiple perturbations in metab­ final subsets are evaluated based on an endpoint (e.g., females
olism and that the long-term metabolic patterns were weakly who received 5 mg of drug showed 70% improvement, females
correlated to ICP and CPP; that is, factors other than pressure who received 10 mg showed 80% improvement). This type of
and/or flow likely influence the microdialysis findings. evaluation might suggest a decision such as increasing drug
dosage for poor responders.
Decision trees are useful methods to analyze nonlinear,
Decision Trees multivariate data, particularly with discrete inputs, and have
Whereas linear models and neural networks generate smoothly been implemented with growing frequency for prognosis.
varying functions of the input variables, decision trees use They often incorporate demographic as well as physiologic
hard thresholds (or discrete inputs, such as gender or injury parameters. Although not necessarily more accurate than
type) to split the input space into distinct regions, each of neural networks, trees are much easier to understand—the
which can be treated separately from the others. The training patterns that the learning algorithms (e.g., demographic risk
algorithm starts with a “stump” and adds branches to create factors or critical physiologic thresholds) find are very clear
subsets of the training data that are homogeneous with respect when one looks at the main branches of the tree. Furthermore,
to the output variable. Decision trees are essentially flowcharts the prediction for any given patient is easily explained based
that can be used to categorize items such as patients, as shown on the corresponding path through the tree.
Section VII—Computers, Engineering, and the Future 441

Decision tree analysis has been used to predict TBI outcome produced a series of quantitatively rigorous studies on cere­
or verify other methods of outcome prediction. For example, bral vascular reserve, cerebral hemodynamics during arterial
Rovlias and Kotsou 35 developed a tree model to predict neu­ and CO2 pressure changes, cerebral vasospasm, ICP dynamics,
rologic outcome after TBI; their results indicated that GCS and relationships between CPP and autoregulation. Zhu and
was the best predictor. The overall cross-validated predictive Diao44 developed a model to examine brain temperature gra­
accuracy was 87%. Andrews et al.36 compared decision tree dients during selective cooling of the brain surface after head
analysis and logistic regression and used decision tree analysis injury. Thoman et al.45 developed the ability to visualize intra­
to identify patient subgroups and critical values in assessed cranial dynamics during simulated clinical scenarios. Previous
variables after TBI. The best predictors of mortality were studies had isolated physiologic variables and shown their
duration of hypotensive, pyrexic, and hypoxemic insults. effects on brain dynamics, but no studies had shown the com­
Decision tree analysis confirmed some of the results of logistic bined effects of these variables on intracranial dynamics.
regression and challenged others. Temkin et al.37 developed Finally, Zhang et al.46 investigated differences in brain response
prediction trees of verbal IQ, Halstead’s Impairment Index, due to impacts based on a finite element model with anatomic
and work status at 1 year after injury. They found that the features of a human head. The model predicted higher posi­
trees were simple enough to be used in a clinical setting, and tive pressures accompanied by a large localized skull deforma­
predictions were accurate enough for clinical utility. Other tion at the impact site from a lateral impact compared to a
studies have used decision tree analysis to predict memory frontal impact. Overall, dynamic systems modeling has yielded
recovery, disability, or productive activity after TBI based on a great deal of qualitative understanding of human physiology
measures of initial injury severity, tests of attention, demo­ in general and intracranial dynamics in particular.
graphic characteristics,38 or the TBI Model Systems database.39 The drawback of classic dynamic systems models is the
Choi et al.40 employed a tree to predict outcomes among the need for exact and complete knowledge of the underlying
five Glasgow Outcome Scale categories. Analysis revealed that system. Whereas it is possible to estimate the numerical coef­
the important prognostic factors differed among patient sub­ ficients of differential equation models by laboratory experi­
groups. The overall accuracy of the technique was 78%. Deci­ ment or by fitting the model to data from patient populations,
sion tree analysis has also been used to predict which patients a generic model is seldom correct for any particular patient.
admitted to the NCCU are likely to have colonization with Sometimes this can be overcome by further training to fit
methicillin-resistant Staphylococcus aureus or vancomycin- coefficients to patient-specific data. For example, Ursino’s
resistant Enterococcus.41 group incorporated estimation of intracranial compliance
into its predictive modeling protocol.42,43 A more serious
problem is the assumption of determinism in the model; for
Advanced Critical Care example, no reasonable model can predict the exact time and
Informatics II: Model-Based characteristics of a myocardial infarction, rupture of an aortic
aneurysm, or occurrence of a cerebral hemorrhage, because
Methods the amount of detail and exactness of initial conditions
Although data-driven prediction systems have obvious uses required are simply too great. Put another way, deterministic
and reflect some aspects of expert understanding, they typi­ models have no need for observation of the patient because
cally lack any notion of underlying mechanisms. To give a they already “know” what is going to happen.
trivial example, the GCS, taken literally, gives incorrect pre­
dictions for a deaf-blind patient who may make no verbal
response to spoken commands. A physician obviously under­ Dynamic Bayesian Networks
stands how the patient’s condition affects the ability to respond A dynamic Bayesian network, or DBN, can be thought of as a
to verbal stimuli, and if the patient’s history is unknown, the generalization of dynamic system models that allows for
physician might well suspect the presence of such a condition uncertainty about how the underlying system state—in this
if other responses appear normal. This section describes two case, the patient state—evolves over time (the “transition
methods to represent the causal processes that underlie mea­ model”) and how that system state is manifested in sensor
sured data. To the extent that specific causal processes are measurements (the “sensor model”). This may predict “transi­
understood, this knowledge can be applied directly to inter­ tions” between states in the ICU.47 The network structure of
pret data and make predictions, possibly augmented by the DBN reflects dependencies among the variables. In addi­
machine learning methods and patient-specific adaptation. tion, each node is annotated with the conditional probability
distribution for that variable given its parents in the network.
Both network structure and conditional distributions may be
Dynamic System Models learned from data or specified based on expert knowledge.
Dynamic system models are sets of algebraic and differential Similar to dynamic system models, DBNs provide a natural
equations whose variables represent physiologic quantities. and intuitive format to express knowledge of causal processes.
The models specify exactly how a system evolves over time, Figure 45.4 shows a simple example to predict heart rate (HR)
given the initial conditions. They are among the most intuitive from multiple sensors.
mathematical tools to develop because they are based on ordi­ Given values for the measurement variables in a DBN
nary physical understanding, for example, of pressure-volume- (ECG-HR, Pleth-HR [heart rate from the oxygen saturation
flow relationships in cerebrospinal fluid dynamics, or on monitor], and SpO2 in the example), posterior probability
experimentally derived relationships such as autoregulation. distributions can be inferred (by standard algorithms) for all
Several examples of dynamic system models relevant to neu­ the unobserved variables in the model. This includes the
rocritical care exist. In the late 1990s, Ursino et al.42,43 current state of the patient (True-HR, patient-movement), the
442 Section VII—Computers, Engineering, and the Future

Time t Time t+1 Time t+2...

Resting
HR

True-HR True-HR True-HR

Patient Patient Patient


movement movement movement

ECG Pleth ECG Pleth ECG Pleth


detached detached detached detached detached detached

ECG-HR SpO2 Pleth-HR ECG-HR SpO2 Pleth-HR ECG-HR SpO2 Pleth-HR

Fig. 45.4  Qualitative structure of a dynamic Bayesian network (DBN) that models a patient’s true heart rate changing over time, its
measurement by electrocardiogram and Pleth (heart rate from the oxygen saturation monitor), and three possible causes of artifacts
in those measurements. The network also shows measured oxygen saturation (SpO2) and two causes of artifact in that measurement. It also shows
the influence of an underlying, constant patient parameter (resting heart rate) on the actual, moment-to-moment heart rate. ECG, Electrocardiograph;
HR, heart rate.

current state of the sensors (ECG-detached, Pleth-detached) Neurocritical data can be highly variable. Such data are not
that might affect the readings, and underlying patient param­ necessarily “bad,” but they can be difficult to interpret. Vari­
eters (Resting-HR). Notice the important distinction between ance can be introduced by factors such as faulty instru­
measured and true values of physiologic variables; a DBN mentation, clinical interventions, or physiologic variability.
typically models sensor noise, drift, miscalibration, and failure, Approaches to process missing data include interpolation/
whereas deterministic dynamic system models typically make extrapolation, random filling, removal of the record, or
no distinction between true state and measured state. inference from DBNs. Filters, simple yet robust statistical
Detailed models of physiology need to be coupled to parameters such as medians, change-point algorithms, and
detailed sensor models if real, artifact-ridden data are to be information theory may be applied to simplify highly variable
understood properly. For example, careful modeling of the data.50,51 Thus fundamental goals of physiologic informatics
process by which arterial BP is measured, including artifactual must be to ensure reliability of the data and establish relation­
sensor states causesd by rezeroing and blood draws, can yield ships between physiologic data and clinical events.
almost perfect detection and discounting of artifactual sensor Vast amounts of neurocritical data are collected. For
values and allows correct inference of the true BP even during example, if HR is monitored every minute for a day, 1440 data
extended interruptions.48 In another study, Celi et al.49 used points are generated. If 20 parameters are continuously cap­
this type of modeling to predict fluid requirements for criti­ tured from 20 patients for a week, then 4,032,000 data points
cally ill patients. are generated. Data rapidly reaches “genomic” orders of mag­
nitude. Ironically, even with large data volumes, neurocritical
care data can be sparse. For example, even if physiologic
Current Challenges in Physiologic parameters are recorded every minute, laboratory values may
only be measured every day, and images may only be captured
Data Analysis once. Furthermore, events of interest such as rapid ICP
There are at least three significant challenges in translating increases are sporadic. Thus development of medical infor­
analytic methods for clinical utility. These are the multivariate, matics goes hand-in-hand with the development of data
highly variable, and vast nature of the data generated by neu­ acquisition methods and repositories of large amounts of
rocritical care monitoring. Within these challenges are issues critical care data. Just as efforts to develop specimen and data
related to artifact detection, integration of event data (e.g., repositories for genetic data have had to address evolving
medical administration), and noise or variability in sensors. issues of privacy, heterogeneity, scale, and standardization of
Neurocritical data is multivariate (e.g., ICP, MAP), multi­ terminology, emerging approaches to physiologic critical care
modal (e.g., numerical, text, image), and multidomain (e.g., data registries for the purposes of knowledge discovery likely
physiologic, biochemical, clinical). Decisions usually cannot will be confronted with the same regulatory issues as well.
be made on the basis of just one parameter. For example, Overcoming these challenges (Table 45.2), along with issues
treatment of arterial pressure may change intracranial blood of user-friendly data visualization that could be used at the
volume and result in a change of ICP, and the CPP also may bedside for real-time clinical decision making, lies at the heart
be affected. Furthermore, parameters may be directly, inversely of current and future approaches to data acquisition and
or even sporadically correlated. Information that is multi­ analysis in critical care.
modal also must be quantified and integrated when making
patient decisions. Computed tomography (CT) scan images,
while complex, can be scored, if not evaluated pixel by pixel. Medical Informatics and Decision
However, this type of analysis may not capture the qualitative
overall impression of injury state assessed by an experienced
Support—Enhancing Expertise
clinician or radiologist. Thus analysis of these data likely Medical informatics is a broad, multidisciplinary field con­
requires multiple domains of expertise. cerned with the processing of data and knowledge in medicine
Section VII—Computers, Engineering, and the Future 443

and analysis to understand how to use these data for maximum


Table 45.2  Challenges to the Development patient benefit. It is hoped that as medical informatics em­
of the Field of Neurocritical Care Informatics braces translational bioinformatics, public access “genomic-
Data acquisition electronically as a standard in intensive care
style” repositories of physiologic data and software could do
units much to support global clinical and research efforts. As moni­
Interoperability of different monitoring devices to acquire data toring and neurocritical care informatics evolve, it will be
Standardization of global controlled vocabularies of important to constantly reevaluate how they can aid decision
terminology making, standardize care, serve as a training resource, or
Volume and resolution of data
Regulatory aspects of data collection (e.g., Health Insurance provide better information access. The potential benefits are
Portability and Accountability Act and privacy boards) alluring, but the complexity and regulatory, ethical, and ana­
Automated artifact detection and cleaning of acquired data lytic challenges should not be underestimated (Table 45.2).
before analysis
Missing data (e.g., due to artifact, disconnection, different
time domains)
Varying types of data (e.g., numeric, imaging, text, References
continuous, ordinal)
Integration with electronic medical records and other 1. Sorani MD, Hemphill JC 3rd, Morabito D, et al. New approaches to
patient data physiological informatics in neurocritical care. Neurocrit Care 2007;7:45–52.
Whether to use all data or discard selectively or empirically 2. Hemphill JC, Andrews P, De Georgia M. Multimodal monitoring and
Real-time analysis and feedback to the bedside neurocritical care bioinformatics. Nat Rev Neurol 2011;7(8):451–60.
Clinician acceptance and use of analyzed data 3. Diedler J, Czosnyka M. Merits and pitfalls of multimodality brain
monitoring. Neurocrit Care 2010;12(3):313–6.
4. Kumar S, Aldrich K. Overcoming barriers to electronic medical record
(EMR) implementation in the US healthcare system: a comparative study.
Health Informatics J 2010;16:306–18.
and health care. A primary goal of medical informatics is to 5. Gomez H, Camacho J, Yelicich B, et al. Development of a multimodal
provide decision support for clinicians.52 The field of medical monitoring platform for medical research. Conf Proc IEEE Eng Med Biol
Soc 2010;1:2358–61.
informatics can be characterized in many ways, but three large 6. Saeed M, Villarroel M, Reisner AT, et al. Multiparameter Intelligent
subdomains are information systems, knowledge representa­ Monitoring in Intensive Care II (MIMIC-II): a public-access intensive care
tion, and data analysis.53 Recently, the American Medical unit database. Crit Care Med 2011;39:952–60.
Informatics Association augmented its activities to encompass 7. ASTM subcommittee F29.21. ASTM Standard F2761-09 Medical devices and
medical systems—essential safety requirements for equipment comprising
“translational bioinformatics,” including “analytic and inter­ the patient-centric integrated clinical environment (ICE)—Part 1: general
pretive methods to optimize the transformation of increas­ requirements and conceptual model. ASTM International [online]; 2009.
ingly voluminous biomedical data into proactive, predictive, http://www.astm.org/Standards/F2761.htm.
preventive, and participatory health.”54 This effort lies at the 8. Chesnut RM. Avoidance of hypotension: conditio sine qua non of successful
intersection of information systems, knowledge representa­ severe head-injury management. J Trauma 1997;42:S4–9.
9. Chesnut RM, Marshall LF, Klauber MR, et al. The role of secondary brain
tion, and data analysis. injury in determining outcome from severe head injury. J Trauma 1993;34:
Here, the focus is on the analysis of physiologic data cap­ 216–22.
tured from neuromonitoring, but soon, increased efforts will 10. Diringer MN, Reaven NL, Funk SE, et al. Elevated body temperature
be made to integrate such data with computer-based deci­ independently contributes to increased length of stay in neurologic intensive
care unit patients. Crit Care Med 2004;32:1489–95.
sion support systems, EMRs, and other technologies. In 11. Kilpatrick MM, Lowry DW, Firlik AD, et al. Hyperthermia in the
addition to standard methods for systems to communicate neurosurgical intensive care unit. Neurosurgery 2000;47:850–56.
(HL7) and submit claims (ICD-10), other standards could 12. Robertson CS, Valadka AB, Hannay HJ, et al. Prevention of secondary
improve the access and exchange of patient information. ischemic insults after severe head injury. Crit Care Med 1999;27:2086–95.
Advances in the Unified Medical Language System (UMLS) 13. Struchen MA, Hannay HJ, Contant CF, et al. The relation between acute
physiological variables and outcome on the glasgow outcome scale and
of the National Library of Medicine have made medical disability rating scale following severe traumatic brain injury. J Neurotrauma
record information standards (Systemized Nomenclature of 2001;18:115–25.
Medicine, [SNOMED]) more accessible.55 14. Manley G, Knudson MM, Morabito D, et al. Hypotension, hypoxia, and head
injury: frequency, duration, and consequences. Arch Surg 2001;136:1118–23.
15. Diringer MN. Treatment of fever in the neurologic intensive care unit with a
Conclusion catheter-based heat exchange system. Crit Care Med 2004;32:559–64.
16. Barton CW, Hemphill JC, Morabito D, et al. A novel method of evaluating
Neurocritical care is complex, and informatics methods hold the impact of secondary brain insults on functional outcomes in traumatic
some promise to manage this complexity. Initial approaches brain-injured patients. Acad Emerg Med 2005;12:1–6.
using comparatively simple analytical advances have already 17. Schmutzhard E, Engelhardt K, Beer R, et al. Safety and efficacy of a novel
intravascular cooling device to control body temperature in neurologic
demonstrated the early potential of new informatics tech­ intensive care patients: a prospective pilot study. Crit Care Med
niques. However, a recent systematic review concluded that 2002;30:2481–88.
while prognostic models are frequently published, they are 18. Steiner LA, Czosnyka M, Piechnik SK, et al. Continuous monitoring of
“developed from small samples of patients, their methodolog­ cerebrovascular pressure reactivity allows determination of optimal cerebral
perfusion pressure in patients with traumatic brain injury. Crit Care Med
ical quality is poor and they are rarely validated on external 2002;30:733–38.
populations. Furthermore, they are not clinically practical as 19. Czosnyka M, Smielewski P, Kirkpatrick P, et al. Continuous monitoring of
they are not presented to physicians in a user-friendly way.”56 cerebrovascular pressure-reactivity in head injury. Acta Neurochir Suppl
Similarly, commercial products suffer from lack of flexibility, 1998;71:74–7.
problems with interoperability, complexity, and high cost. 20. Diedler J, Sykora M, Rupp A, et al. Impaired cerebral vasomotor activity in
spontaneous intracerebral hemorrhage. Stroke 2009;40:815–9.
Along with advances in neuromonitoring devices (e.g., brain- 21. Smielewski, P, Czosnyka M, Steiner LA, et al. ICM+: software for on-line
tissue oxygen monitors, microdialysis), it is likely necessary to analysis of bedside monitoring data after severe head trauma. Acta
develop user-friendly methods of advanced data visualization Neurochir Suppl 2005;95:43–9.
444 Section VII—Computers, Engineering, and the Future

22. Buchman TG. The digital patient: predicting physiologic dynamics with 37. Temkin NR, Holubkov R, Machamer JE, et al. Classification and regression
mathematical models. Crit Care Med 2009;37:1167–8. trees (cart) for prediction of function at 1 year following head trauma.
23. Gawande A. The checklist. The New Yorker. December 10, 2007. J Neurosurg 1995;82:764–71.
24. van Santbrink H, vd Brink WA, Steyerberg EW, et al. Brain tissue oxygen 38. Stuss DT, Binns MA, Carruth FG, et al. Prediction of recovery of continuous
response in severe traumatic brain injury. Acta Neurochir (Wien) 2003;145: memory after traumatic brain injury. Neurology 2000;54:1337–44.
429–38. 39. Brown AW, Malec JF, McClelland RL, et al. Clinical elements that predict
25. Oddo M, Levine JM, Mackenzie L, et al. Brain hypoxia is associated with outcome after traumatic brain injury: a prospective multicenter recursive
short-term outcome after severe traumatic brain injury independently of partitioning (decision-tree) analysis. J Neurotrauma 2005;22:1040–51.
intracranial hypertension and low cerebral perfusion pressure. Neurosurgery 40. Choi SC, Muizelaar JP, Barnes TY, et al. Prediction tree for severely
2011;69(5):1037–45. head-injured patients. J Neurosurg 1991;75:251–5.
26. Amin AP, Kulkarni HR. Improvement in the information content of the 41. Minhas P, Perl TM, Carroll KC, et al. Risk factors for positive admission
Glasgow Coma Scale for the prediction of full cognitive recovery after head surveillance cultures for methicillin-resistant Staphylococcus aureus and
injury using fuzzy logic. Surgery 2000;127:245–53. vancomycin-resistant enterococci in a neurocritical care unit. Crit Care Med
27. Chambers IR, Jones PA, Lo TY, et al. Critical thresholds of intracranial 2011;39(10):2322–9.
pressure and cerebral perfusion pressure related to age in paediatric head 42. Ursino M, Giulioni M, Lodi CA. Relationships among cerebral perfusion
injury. J Neurol Neurosurg Psychiatry 2006;77:234–40. pressure, autoregulation, and transcranial Doppler waveform: a modeling
28. Hornero R, Aboy M, Abasolo D, et al. Interpretation of approximate entropy: study. J Neurosurg 1998;89:255–66.
analysis of intracranial pressure approximate entropy during acute 43. Ursino M, Lodi CA, Rossi S, et al. Estimation of the main factors affecting
intracranial hypertension. IEEE Trans Biomed Eng 2005;52:1671–80. icp dynamics by mathematical analysis of PVI tests. Acta Neurochir Suppl
29. Cohen MJ, Grossman AD, Morabito D, et al. Identification of complex 1998;71:306–9.
metabolic states in critically injured patients using bioinformatic cluster 44. Zhu L, Diao C. Theoretical simulation of temperature distribution in the
analysis. Crit Care 2010;14:R10. brain during mild hypothermia treatment for brain injury. Med Biol Eng
30. Vath A, Meixensberger J, Dings J, et al. Prognostic significance of advanced Comput 2001;39:681–7.
neuromonitoring after traumatic brain injury using neural networks. 45. Thoman WJ, Lampotang S, Gravenstein D, et al. A computer model of
Zentralbl Neurochir 2000;61:2–6. intracranial dynamics integrated to a full-scale patient simulator. Comput
31. Lang EW, Pitts LH, Damron SL, et al. Outcome after severe head injury: an Biomed Res 1998;31:32–46.
analysis of prediction based upon comparison of neural network versus 46. Zhang L, Yang KH, King AI. Comparison of brain responses between frontal
logistic regression analysis. Neurol Res 1997;19:274–80. and lateral impacts by finite element modeling. J Neurotrauma 2001;
32. Becalick DC, Coats TJ. Comparison of artificial intelligence techniques with 18:21–30.
UKTISS for estimating probability of survival after trauma. UK Trauma and 47. Peelen L, de Keizer NF, de Jonge E, et al. Using hierarchical dynamic
Injury Severity Score. J Trauma 2001;51:123–33. Bayesian networks to investigate dynamics of organ failure in patients in the
33. Li YC, Liu L, Chiu WT, et al. Neural network modeling for surgical decisions intensive care unit. J Biomed Inform 2010;43:273–86.
on traumatic brain injury patients. Int J Med Inform 2000;57:1–9. 48. Aleks N, Russell S, Madden MG, et al. Probabilistic detection of short events,
34. Nelson DW, Thornquist B, MacCallum RM, et al. Analyses of cerebral with application to critical care monitoring. In: Advances in neural
microdialysis in patients with traumatic brain injury: relations to information processing systems 21. Cambridge, MA: MIT Press; 2009.
intracranial pressure, cerebral perfusion pressure and catheter placement. 49. Celi LA, Hinske LC, Alterovitz G, et al. An artificial intelligence tool to
BMC Med 2011;2(9):21. predict fluid requirement in the intensive care unit: a proof-of-concept
35. Rovlias A, Kotsou S. Classification and regression tree for prediction of study. Crit Care 2008;12:R151.
outcome after severe head injury using simple clinical and laboratory 50. Aboy M, McNames J, Thong T, et al. An automatic beat detection algorithm
variables. J Neurotrauma 2004;21:886–93. for pressure signals. IEEE Trans Biomed Eng 2005;52:1662–70.
36. Andrews PJ, Sleeman DH, Statham PF, et al. Predicting recovery in patients
suffering from traumatic brain injury by using admission variables and A complete list of references for this chapter can be found online at
physiological data: a comparison between decision tree analysis and logistic www.expertconsult.com.
regression. J Neurosurg 2002;97:326–36.
Section VII—Computers, Engineering, and the Future 444.e1

References 27. Chambers IR, Jones PA, Lo TY, et al. Critical thresholds of intracranial
pressure and cerebral perfusion pressure related to age in paediatric head
1. Sorani MD, Hemphill JC 3rd, Morabito D, et al. New approaches to injury. J Neurol Neurosurg Psychiatry 2006;77:234–40.
physiological informatics in neurocritical care. Neurocrit Care 2007;7:45–52. 28. Hornero R, Aboy M, Abasolo D, et al. Interpretation of approximate entropy:
2. Hemphill JC, Andrews P, De Georgia M. Multimodal monitoring and analysis of intracranial pressure approximate entropy during acute
neurocritical care bioinformatics. Nat Rev Neurol 2011;7(8):451–60. intracranial hypertension. IEEE Trans Biomed Eng 2005;52:1671–80.
3. Diedler J, Czosnyka M. Merits and pitfalls of multimodality brain 29. Cohen MJ, Grossman AD, Morabito D, et al. Identification of complex
monitoring. Neurocrit Care 2010;12(3):313–6. metabolic states in critically injured patients using bioinformatic cluster
4. Kumar S, Aldrich K. Overcoming barriers to electronic medical record analysis. Crit Care 2010;14:R10.
(EMR) implementation in the US healthcare system: a comparative study. 30. Vath A, Meixensberger J, Dings J, et al. Prognostic significance of advanced
Health Informatics J 2010;16:306–18. neuromonitoring after traumatic brain injury using neural networks.
5. Gomez H, Camacho J, Yelicich B, et al. Development of a multimodal Zentralbl Neurochir 2000;61:2–6.
monitoring platform for medical research. Conf Proc IEEE Eng Med Biol 31. Lang EW, Pitts LH, Damron SL, et al. Outcome after severe head injury: an
Soc 2010;1:2358–61. analysis of prediction based upon comparison of neural network versus
6. Saeed M, Villarroel M, Reisner AT, et al. Multiparameter Intelligent logistic regression analysis. Neurol Res 1997;19:274–80.
Monitoring in Intensive Care II (MIMIC-II): a public-access intensive care 32. Becalick DC, Coats TJ. Comparison of artificial intelligence techniques with
unit database. Crit Care Med 2011;39:952–60. UKTISS for estimating probability of survival after trauma. UK Trauma and
7. ASTM subcommittee F29.21. ASTM Standard F2761-09 Medical devices and Injury Severity Score. J Trauma 2001;51:123–33.
medical systems—essential safety requirements for equipment comprising 33. Li YC, Liu L, Chiu WT, et al. Neural network modeling for surgical decisions
the patient-centric integrated clinical environment (ICE)—Part 1: general on traumatic brain injury patients. Int J Med Inform 2000;57:1–9.
requirements and conceptual model. ASTM International [online]; 2009. 34. Nelson DW, Thornquist B, MacCallum RM, et al. Analyses of cerebral
http://www.astm.org/Standards/F2761.htm. microdialysis in patients with traumatic brain injury: relations to
8. Chesnut RM. Avoidance of hypotension: conditio sine qua non of successful intracranial pressure, cerebral perfusion pressure and catheter placement.
severe head-injury management. J Trauma 1997;42:S4–9. BMC Med 2011;2(9):21.
9. Chesnut RM, Marshall LF, Klauber MR, et al. The role of secondary brain 35. Rovlias A, Kotsou S. Classification and regression tree for prediction of
injury in determining outcome from severe head injury. J Trauma 1993;34: outcome after severe head injury using simple clinical and laboratory
216–22. variables. J Neurotrauma 2004;21:886–93.
10. Diringer MN, Reaven NL, Funk SE, et al. Elevated body temperature 36. Andrews PJ, Sleeman DH, Statham PF, et al. Predicting recovery in patients
independently contributes to increased length of stay in neurologic intensive suffering from traumatic brain injury by using admission variables and
care unit patients. Crit Care Med 2004;32:1489–95. physiological data: a comparison between decision tree analysis and logistic
11. Kilpatrick MM, Lowry DW, Firlik AD, et al. Hyperthermia in the regression. J Neurosurg 2002;97:326–36.
neurosurgical intensive care unit. Neurosurgery 2000;47:850–56. 37. Temkin NR, Holubkov R, Machamer JE, et al. Classification and regression
12. Robertson CS, Valadka AB, Hannay HJ, et al. Prevention of secondary trees (cart) for prediction of function at 1 year following head trauma. J
ischemic insults after severe head injury. Crit Care Med 1999;27:2086–95. Neurosurg 1995;82:764–71.
13. Struchen MA, Hannay HJ, Contant CF, et al. The relation between acute 38. Stuss DT, Binns MA, Carruth FG, et al. Prediction of recovery of continuous
physiological variables and outcome on the glasgow outcome scale and memory after traumatic brain injury. Neurology 2000;54:1337–44.
disability rating scale following severe traumatic brain injury. J Neurotrauma 39. Brown AW, Malec JF, McClelland RL, et al. Clinical elements that predict
2001;18:115–25. outcome after traumatic brain injury: a prospective multicenter recursive
14. Manley G, Knudson MM, Morabito D, et al. Hypotension, hypoxia, and partitioning (decision-tree) analysis. J Neurotrauma 2005;22:1040–51.
head injury: frequency, duration, and consequences. Arch Surg 2001;136: 40. Choi SC, Muizelaar JP, Barnes TY, et al. Prediction tree for severely
1118–23. head-injured patients. J Neurosurg 1991;75:251–5.
15. Diringer MN. Treatment of fever in the neurologic intensive care unit with a 41. Minhas P, Perl TM, Carroll KC, et al. Risk factors for positive admission
catheter-based heat exchange system. Crit Care Med 2004;32:559–64. surveillance cultures for methicillin-resistant Staphylococcus aureus and
16. Barton CW, Hemphill JC, Morabito D, et al. A novel method of evaluating vancomycin-resistant enterococci in a neurocritical care unit. Crit Care Med
the impact of secondary brain insults on functional outcomes in traumatic 2011;39(10):2322–9.
brain-injured patients. Acad Emerg Med 2005;12:1–6. 42. Ursino M, Giulioni M, Lodi CA. Relationships among cerebral perfusion
17. Schmutzhard E, Engelhardt K, Beer R, et al. Safety and efficacy of a novel pressure, autoregulation, and transcranial Doppler waveform: a modeling
intravascular cooling device to control body temperature in neurologic study. J Neurosurg 1998;89:255–66.
intensive care patients: a prospective pilot study. Crit Care Med 2002;30: 43. Ursino M, Lodi CA, Rossi S, et al. Estimation of the main factors affecting
2481–88. icp dynamics by mathematical analysis of PVI tests. Acta Neurochir Suppl
18. Steiner LA, Czosnyka M, Piechnik SK, et al. Continuous monitoring of 1998;71:306–9.
cerebrovascular pressure reactivity allows determination of optimal cerebral 44. Zhu L, Diao C. Theoretical simulation of temperature distribution in the
perfusion pressure in patients with traumatic brain injury. Crit Care Med brain during mild hypothermia treatment for brain injury. Med Biol Eng
2002;30:733–38. Comput 2001;39:681–7.
19. Czosnyka M, Smielewski P, Kirkpatrick P, et al. Continuous monitoring of 45. Thoman WJ, Lampotang S, Gravenstein D, et al. A computer model of
cerebrovascular pressure-reactivity in head injury. Acta Neurochir Suppl intracranial dynamics integrated to a full-scale patient simulator. Comput
1998;71:74–7. Biomed Res 1998;31:32–46.
20. Diedler J, Sykora M, Rupp A, et al. Impaired cerebral vasomotor activity in 46. Zhang L, Yang KH, King AI. Comparison of brain responses between frontal
spontaneous intracerebral hemorrhage. Stroke 2009;40:815–9. and lateral impacts by finite element modeling. J Neurotrauma
21. Smielewski, P, Czosnyka M, Steiner LA, et al. ICM+: software for on-line 2001;18:21–30.
analysis of bedside monitoring data after severe head trauma. Acta 47. Peelen L, de Keizer NF, de Jonge E, et al. Using hierarchical dynamic
Neurochir Suppl 2005;95:43–9. Bayesian networks to investigate dynamics of organ failure in patients in the
22. Buchman TG. The digital patient: predicting physiologic dynamics with intensive care unit. J Biomed Inform 2010;43:273–86.
mathematical models. Crit Care Med 2009;37:1167–8. 48. Aleks N, Russell S, Madden MG, et al. Probabilistic detection of short events,
23. Gawande A. The checklist. The New Yorker. December 10, 2007. with application to critical care monitoring. In: Advances in neural
24. van Santbrink H, vd Brink WA, Steyerberg EW, et al. Brain tissue oxygen information processing systems 21. Cambridge, MA: MIT Press; 2009.
response in severe traumatic brain injury. Acta Neurochir (Wien) 49. Celi LA, Hinske LC, Alterovitz G, et al. An artificial intelligence tool to
2003;145:429–38. predict fluid requirement in the intensive care unit: a proof-of-concept
25. Oddo M, Levine JM, Mackenzie L, et al. Brain hypoxia is associated with study. Crit Care 2008;12:R151.
short-term outcome after severe traumatic brain injury independently of 50. Aboy M, McNames J, Thong T, et al. An automatic beat detection algorithm
intracranial hypertension and low cerebral perfusion pressure. Neurosurgery for pressure signals. IEEE Trans Biomed Eng 2005;52:1662–70.
2011;69(5):1037–45. 51. Eide PK. Assessment of quality of continuous intracranial pressure
26. Amin AP, Kulkarni HR. Improvement in the information content of the recordings in children. Pediatr Neurosurg 2006;42:28–34.
Glasgow Coma Scale for the prediction of full cognitive recovery after head 52. Ubeyli ED. Medical informatics: a model developed for diabetes education
injury using fuzzy logic. Surgery 2000;127:245–53. via telemedicine. J Med Syst 2009;33:113–9.
444.e2 Section VII—Computers, Engineering, and the Future

53. Schuemie MJ, Talmon JL, Moorman PW, et al. Mapping the domain of 55. Coonan KM. Medical informatics standards applicable to emergency
medical informatics. Methods Inf Med 2009;48:76–83. department information systems: making sense of the jumble. Acad Emerg
54. Butte AJ. Translational bioinformatics: coming of age. J Am Med Inform Med 2004;11:1198–205.
Assoc 2008;15:709–14. 56. Perel P, Edwards P, Wentz R, et al. Systematic review of prognostic models in
traumatic brain injury. BMC Med Inform Decis Mak 2006;6:38.
Chapter
46  
VII

Noninvasive Brain Monitoring


Marek Czosnyka, Bernhard Schmidt, Eric Albert Schmidt, Rohan Ramakrishna,
Pierre D. Mourad, and Michel Kliot

encephalopathy in which an underlying coagulopathy increases


Introduction the risk of hemorrhage from insertion of an invasive monitor-
During the first decade of the 21st century, many major ing device) or when the potential benefits of invasive monitor-
advances have revolutionized neurocritical care. These changes ing are unclear (e.g., moderate stroke, moderate and mild
have been achieved mainly because neurocritical care now is head injury, or eclampsia, among others).
regarded as a distinct specialty in its own right, necessitating This chapter reviews some of the noninvasive technologies
specialists familiar with patients with neurologic injury. Appli- available now as well as on the horizon that may enhance
cation of new technologies toward the central nervous system neurocritical care.
(CNS) in an effort to understand and manage the physiology
of the brain has been central to this mission. Such technolo-
gies include intracranial pressure monitoring, brain oxygen Noninvasive Intracranial
and cerebral blood flow (CBF) monitoring, microdialysis Pressure and Cerebral
techniques, and jugular venous blood saturation monitoring,
to name a few. Each of these technologies used in neurocritical
Perfusion Pressure Monitoring
care, however, have important limitations. First, their use is ICP can be estimated noninvasively using various methods:
generally limited to major medical centers in part because of (1) tympanic membrane displacement,1 (2) fontanometry,2
the personnel required to install and manage these devices, the (3) analysis of transcranial Doppler (TCD) pulse waveform,3,4
training required, and the cost of implementation. Second, or (4) ocular nerve sonography.5,6 However, none of these
some of devices are frequently criticized as being too global noninvasive methods can be considered reliable or precise
and therefore nonspecific in their measurements, or con- enough to be used routinely in the clinical management of
versely too focal and then not representative of the entire intracranial hypertension. Nor is there a validated noninvasive
neurologic system. Third and perhaps most important, most system that can replace invasive ICP monitors for quantifica-
of the devices and in particular those to measure the gold tion of ICP.7 Instead current noninvasive monitors provide
standard parameter, intracranial pressure (ICP), are invasive supporting qualitative data that may be helpful in making
and so there is a reluctance to insert these devices. It seems decisions about use of more invasive direct measurement
that technologies able to take neurocritical care to the next devices.
level will need to be noninvasive to garner widespread use, as Various methods of noninvasive ICP measurement are
well as multimodal, because clinical experience has indicated described in the contemporary literature8:
that the neurologic system cannot be optimally managed
without an integrated approach that factors the multiple  Prediction of ICP from noninvasive transocular venous
important determinants of brain pathophysiology. and arterial hemodynamic measurements (reported 95%
Because of its anatomy, the brain is not an easy organ on confidence interval for prediction ±12 mm Hg)9
which to perform noninvasive monitoring. The surrounding  Change in a skull diameter10 (no consistent 95% percent
skull represents an important barrier to ultrasonic insonation, limit)
for example. Thanks to technologic refinements, various  Change in blood flow velocity in the straight sinus11 (esti-
direct sensors used in neurocritical care have become safer and mated 95% confidence limit is low, ±8.8 mm Hg, but this
less invasive. They have been miniaturized, sometimes made is a difficult measurement to make in adults)
wireless, and the infection risk has decreased as these monitors  Methods based on electroencephalography (reported
are excluded from intracranial contents such as the cerebro- lowest 95% limits for prediction ±8 mm Hg)12
spinal fluid (CSF). In the majority of acute neurologic injuries  Methods based on magnetic resonance imaging (MRI)
the small risk of sensor placement is outweighed by the clinical analysis of CSF volume, as well as arterial blood inflow and
benefits of having better insight into assessment of the timing venous blood outflow from the cranium13 (there is no
and extent of secondary insults such as ischemia. Nevertheless, consistent 95% confidence limit reported)
the benefits of noninvasive brain monitoring are obvious, par-  MRI or ultrasound-based measurement of the optic nerve
ticularly in cases with known contraindications (e.g., hepatic sheath diameter14 (95% confidence limit ±20 mm Hg)
© Copyright 2013 Elsevier Inc. All rights reserved. 445
446 Section VII—Computers, Engineering, and the Future

to be accurate enough because many other factors can


Intracranial Pressure Monitoring Based influence PI such as arterial pulse, heart rate, PaCO2, vascu-
on Transcranial Doppler lar tone, proximal stenosis, and spasm, among others.
Methods using TCD ultrasonography are discussed in more However, data from Bellner et al.21 suggest a very narrow
detail because this technology is commonly used to noninva- 95% confidence limit for prediction in head-injured pa-
sively assess ICP. Aaslid’s design of TCD sonography in 198215 tients: ±4.2 mm Hg. The same prediction limit derived from
permitted bedside monitoring of CBF velocity (the instanta- data obtained at Cambridge and the University of Washing-
neous speed of flowing blood, typically in units of cm/second), ton is plus or minus 20 mm Hg. Such a huge discrepancy
which may be used assuming vasospasm is absent as an index ought to be explained.
of the CBF (the instantaneous volume of flowing blood, typi- A moving-average model of transmission between ABP and
cally in units of mL/second; see also Chapter 30). The measure- ICP, modified by the relationship between ABP and FV gives
ment may be conducted noninvasively, repeatedly, and even a mean absolute error of 6 mm Hg and 95% confidence limit
continuously. This is a “big tube technique,” which measures for predictor of plus or minus 16 mm Hg.22 The method is
arterial blood flow velocity in the major branches of the circle based on analysis of a large database of patients with homo-
of Willis, most commonly the middle cerebral artery (MCA). geneous pathology who received ICP, ABP, and FV direct
A compliant branch of the MCA may be interpreted as an monitoring and is most probably pathology dependent. The
implanted pressure transducer, in that the pattern of blood method is useful for monitoring ICP changes over time (Fig.
flow within the MCA is modulated by transmural pressure 46.2). However, a change in PaCO2, vessel spasm and proximal
(i.e., cerebral perfusion pressure [CPP]) and the distal vascular stenosis can confound the results.
resistance (also modulated by CPP), and therefore analysis of TCD methods (with easy access to bilateral Doppler
MCA blood flow should produce a measure of the net pressure machines) theoretically allow the assessment of inter­
acting on the “transducer” (again, the MCA). But the calibra- hemispheric gradients of ICP or CPP. As long as CSF com-
tion factor for the MCA, the nonlinear distortion, and other municates freely between different fluid cavities within the
factors (in particular, their stability in time) are unknown. brain, there should not be any substantial differences in
There is a reasonable correlation between the pulsatility regionally measured ICP. Direct measurements of pressures in
index (PI) of the MCA blood flow velocity and CPP after head two CSF compartments are performed rarely. In head injury,
injury, but absolute measurements of CPP cannot be extrapo- intrahemispheric pressure gradients are reported23 with inter-
lated.16 Others suggest that critical closing pressure (CCP), hemispheric ICP differences greater than 20 mm Hg mea-
derived from flow velocity and arterial pressure waveform, sured in one severely injured patient.24 Other studies of
approximates ICP.17 The accuracy of this method has, however, patients with focal injuries or intracranial masses show a
never been satisfactorily demonstrated.18 Aaslid et al. pro- median interhemispheric difference of 5.5 mm Hg after cra-
posed19 that an index of CPP could be derived from the ratio niotomy but before parenchymal resection, ranging from 3 to
of the amplitudes of the first harmonics of the arterial blood 16 mm Hg.25 There are intrahemispheric pressure gradients in
pressure and the MCA velocity (detected by TCD sonography) critical closing pressure and noninvasive CPP associated with
multiplied by the mean flow velocity in the MCA: midline shift, side of contusion (assessed using computed
tomography [CT]), or side of craniectomy.26 Both nCPP and
nCPP = A1/F1 × FVm
CCP measurements indicate that cerebral perfusion or level
where F1 and A1 are first harmonic components of flow veloc- of cerebrovascular dilation is greater on the side of a contusion
ity and arterial pressure pulse waveforms, and FVm is time- or more swollen brain in the case of midline shift. This may
averaged flow velocity. However, the 95% confidence limit for support the hypothesis that vascular expansion and not a
predicted values is as wide as plus or minus 20 mm Hg. brain tissue volume increase is associated with side-to-side
CPP affects the shape of the blood flow velocity waveform. differences seen on CT scan. This hypothesis may be further
However, arterial pulse waveform, heart rate, tension of arte- supported by the observation that cerebral autoregulation is
rial carbon dioxide (CO2), distal vascular resistance, and even worse on the side of contusion or brain expansion.27
age, all affect the flow velocity (FV) waveform as well. Some
simple formulas to assess CPP noninvasively from arterial
blood pressure (ABP) and FV waveforms have been proposed. Intracranial Pressure Monitors Based
One particular calculation has achieved satisfactory accuracy on the Optic Nerve
(error less than ±10 mm Hg in more than 80% measure- The optic nerve sheath (ONS) is an extension of the dura mater,
ments20; including in traumatic brain injury [TBI] patients, and the space within the sheath is continuous with the intra-
shown in work by Mourad, Kliot et al.): cranial contents. Therefore pressure in the ONS increases when
ICP increases.28 This can be detected by physical examination
nCPP = MAP × FVd/FVm + 14 (i.e., funduscopy) to detect papilledema. The Frisen scale can
where FVd is diastolic flow velocity; FVm is mean flow veloc- be used to grade the severity of papilledema (0, normal, through
ity, and 14 mm Hg is “calibrating constant” for TBI patients. 5, severe). Although papilledema indicates increased ICP, fun-
nCPP is useful both to estimate absolute CPP and to duscopy is not suited to monitoring patients in the NCCU.
monitor changes in CPP in time (Fig. 46.1). The 95% confi- First, it is a qualitative “point-in-time” assessment. Second, the
dence limit for estimation of CPP is 12 mm Hg (prospective sensitivity and specificity to detect papilledema vary among
trial)20 and 18 mm Hg in retrospective analysis of these medical specialists and may depend on pupillary dilation.
authors’ own database. Although this seems to be satisfactory Third, in acute brain injury, papilledema may take several
for CPP, such precision is not good enough to estimate ICP. hours to develop after ICP increases. Fourth, the absence of
The pulsatility index (measured by TCD) increases as ICP papilledema does not preclude increased ICP.29 In addition,
increases.3 The prediction of absolute ICP using PI may not spontaneous venous pulsations, the absence of which are
120
100
MAP 80
60
[mm Hg] 40
20
0
120
FVI 100
FVr
[cm/s] 50
0

100
80
nCPPr 60
[mm Hg] 40
20
0

100
80
nCPPI 60
[mm Hg] 40
20
0
10:00 11:00 00 13:00 14:00 15:00 16:00
Dissection Ahepatic Reperfusion
Fig. 46.1  An example of noninvasive cerebral perfusion pressure (nCPP) monitoring based on transcranial Doppler (TCD) obtained during
a liver transplant. nCPP fluctuated quite considerably during dissection and ahepatic and reperfusion phases, with an overall tendency to fall during
the operation. This was most probably caused by a gradual fall in mean arterial pressure (MAP). The difference between arterial blood pressure (ABP)
and nCPP was stable over the whole data collection period and did not exceed 20 mm Hg. Blood flow velocity on left and right was symmetrical,
with a specific increase during the reperfusion phase (vasodilation). nCPP was symmetrical with a 20-minute period of discrepancy (about 10:40 on
x-axis), when short-term irregularity of ABP was noticed and at the beginning of the reperfusion period, with absolute values of difference between
left and right never greater than 10 mm Hg. From a technical standpoint it was very difficult to keep the TCD probes in place during the 6-hour-long
operation. FVl, Flow velocity, left; FVr, flow velocity right; nCCPr, noninvasive cerebral perfusion pressure right; nCPPl, noninvasive cerebral perfusion
pressure left.

160
120
FV
80
[cm/s]
40
0
200 ABP
160
ABP 120
[mm Hg] 80
40
0
100 ICP
80
ICP 60
[mm Hg] 40
20
0
750 LDF
60
LDF 450
[a.u.] 300
150
0
nICP
100
80
nICP 60
[mm Hg] 40
20
0
0 2814 5629 8443 11258 TIME [sec]
Fig. 46.2  Traces showing noninvasive intracranial pressure (nICP) assessment in a traumatic brain injury (TBI) patient. Middle cerebral
artery (MCA) blood flow velocity (FV), arterial blood pressure (ABP), intraparenchymal intracranial pressure (ICP), and cortical blood flow using laser-
Doppler flowmetry (LDF) were recorded. Retrospectively, simulated nICP using TCD and ABP recordings indicated good agreement between real and
simulated elevations of ICP during plateau waves.
448 Section VII—Computers, Engineering, and the Future

consistent with increased ICP, are absent in about 10% of FVr 150 B
normal subjects. Fifth, the optic nerve does not swell in atro- [cm/s] A
120
phic areas30 (i.e., papilledema is not reliable in patients who
have had previous optic nerve injury). To overcome the limita- 90
tions of funduscopy, several techniques based on evaluation of 60
the ONS have been used to evaluate ICP, including (1) ophthal- 30
modynamometry, (2) scanning laser tomography (SLT), (3)
0
optical coherence tomography, and (4) ONS ultrasound. In 0 5 10 15 20
addition, color Doppler ultrasonography to insonate the oph- Time [sec.]
thalmic artery and the central retinal artery and vein can be
Fig. 46.3  The transient hyperemic response test is an example of
used.31,32 Although these techniques can be used to quantify a noninvasive test of autoregulation. Only a transcranial Doppler
ICP, none is suitable for ICP monitoring in the ICU at present. (TCD) machine is needed. The hyperemic phase (B) after short-term
compression of the ipsilateral common carotid artery (CCA) is compared
to a baseline (A) that in this case represents evidence of preserved
Ophthalmodynamometry autoregulation. The method is not suitable for continuous monitoring
This technique is an attempt to quantify ICP. First, the baseline and is contraindicated when there is significant atherosclerotic carotid
intraocular pressure (IOP) is measured. Then a suction cup is disease. FVr, Flow velocity right obtained from the middle cerebral artery.
placed on the eye, and IOP increased until the central retinal
vein collapses. This pressure added to the baseline IOP indi-
cates the central retinal vein venous outflow pressure that has
a linear relationship with ICP.33,34 the other hand some therapies for severe TBI, in particular the
Lund concept, are based on the premise that autoregulation is
impaired.45 Methods for noninvasive assessment of autoregu-
Scanning Laser Tomography lation through study of CBF can be divided into (1) tests in
SLT uses a laser to scan the retinal surface. It is primarily used which an external stimulus to challenge regulation mecha-
in patients with glaucoma but has been used to quantify pap- nisms is applied or (2) methods suitable for continuous moni-
illedema in patients with idiopathic intracranial hypertension toring, that is, without an external stimulus. Methods to
in which optic nerve height and volume measurements are examine autoregulation include (1) static rate of autoregula-
associated with opening pressure on lumbar puncture.37,38 tion, (2) lower body negative pressure, (3) tilt test, (4) CO2
reactivity, (5) leg-cuff test, and (6) transient hyperemic
response (Fig. 46.3). These tests provide information at a point
Optical Coherence Tomography in time, but in many conditions, cerebral autoregulation may
Infrared light to measure total retinal thickness, retinal nerve fluctuate quite rapidly, particularly in NCCU patients. In such
fiber layer thickness, and optic nerve head morphology is used cases, continuous monitoring of autoregulation is clinically
to quantify papilledema. This technique can be used to distin- useful. These methods rely on analysis of spontaneous con-
guish normal nerves from mild papilledema but at present is tinuous fluctuations of MAP or CPP. Thanks to refinement of
not suited to NCCU monitoring.35,36 computer-assisted methodology, fluctuations of MAP greater
than 5 mm Hg are suitable to retrieve information about auto-
regulation. In some cases information is less precise than in
Optic Nerve Sheath Ultrasound tests using an external stimulus, but the ability to time-average
Measurement of optic nerve diameter using ultrasound, typi- continuously obtained autoregulation indices can improve the
cally using a frequency between 5 and 10.5 MHz, is the most signal-to-noise ratio of measures of autoregulation.46
studied noninvasive method of ICP assessment based on the
ONS.6,39-41 The technique is relatively easy to learn and interob- System Analysis: Phase Shift and Transfer
server variation is low.42,43 The ONS begins to expand when
the ICP is greater than 15 mm Hg, and beyond this level is a Function Between Arterial Pressure and
linear correlation between ONS enlargement and ICP Flow Velocity Waves
increases. An ONS diameter on ultrasound between 4.8 and In this methodology it is presumed that autoregulation
5.9 mm measured 3 mm posterior to the globe is considered mechanisms can be described by a linear dynamic system
the threshold value to identify ICP greater than 20 mm Hg.14,40,41 with arterial blood pressure as input and blood flow velocity
Although this technique has promise, it has not been used to the output. The fast pulsatile component associated with the
monitor ICP in the ICU because eye injury or swelling may cardiac cycle is probably too fast to help assess autoregula-
limit its use, off-axis measurement of the ONS diameter pro- tion, in part because cerebral autoregulation acts primarily
vides an incorrect value, and artifacts may limit quality.44 through altering the diameter of the secondary cerebral arte-
rioles on time scales of a few seconds in healthy individuals,
and longer for injured patients. However, respiratory waves
Continuous Monitoring (from 0.4-0.16 Hz) and slow waves (0.05-0.005 Hz) are
slower and therefore they can carry information about cere-
of Cerebral Autoregulation bral autoregulation. The first step in processing FV and ABP
Cerebral autoregulation is an essential intrinsic, self-protecting time-series is therefore filtering out pulsatile components,
brain mechanism in many disorders that require NCCU care typically with a low pass filter of cutoff frequency of 0.5 Hz.
(e.g., TBI, subarachnoid hemorrhage [SAH], and stroke). On A noninvasive method to derive autoregulatory status from
Section VII—Computers, Engineering, and the Future 449

110
ABP 100
[mm Hg] 90
80

60
FV
[cm/s] 55
50
10:36 10:38 10:40 10:42 10:44 10:46 10:48
Time [h:m]
0.8
Coherence 0.7
0.6
0.5
0.4
0.3
0.2
0.1
0
0 1 2 3 4 5 6 7 8 9 10
Frequency [b/min]
180
160
140
120
Phase 100
[deg] 80
60
40
20
0
0 1 2 3 4 5 6 7 8 9 10
Frequency [b/min]
Fig. 46.4  With working autoregulation, slow waves of flow velocity (FV) are leading changes in arterial pressure (ABP) (upper graph), with pulse and
respiratory waveforms filtered out. Coherence shows strong association (around 0.8) between waves in FV and ABP at a frequency around 2 cycles
per minute. Therefore in system analysis, calculated ABP to FV phase shift (bottom panel) is positive (in this case little bit above 60 degrees). b/min,
Beats per minute.

natural fluctuations in MCA flow velocity and ABP involves spontaneous ABP fluctuations, values of ARI are obtained by
the assessment of phase shift between the superimposed fitting the second-order linear model proposed by Tiecks
respiratory or slow ABP and blood FV waves during sponta- et al.50 that describes the FV response to a step-change in ABP.
neous recordings or slow breathing. Coherence function, Transfer function analysis is used to quantify the dynamic
which is a Fourier transform of normalized cross-correlation relationship between mean ABP (input) and mean FV
between ABP and FV series is calculated. The modulus of (output). The inverse Fourier transformation then is per-
coherence indicates how strong components of given fre- formed to obtain the FV impulse response in the time domain.
quency are associated with each other. Another transforma- Impulse response is in turn integrated to yield an estimate of
tion method is a transfer function between ABP and FV. It is the FV response to a hypothetical step-change in ABP. Each of
a complex function, having its gain and phase shift. Phase the 10 models,50 corresponding to ARI values from 0 (absence
indicates which variable changes before another. A zero- of autoregulation) to 9 (best autoregulation) is fitted to the
degree phase shift (e.g., both ABP and FV change at the same first 10 seconds of the FV step response and the best fit, as
time, without delay) indicates absent autoregulation, whereas selected by the minimum squared error, is considered the
a positive phase shift (>30 degrees, i.e., FV changes before representative value of ARI for that segment of data. A similar
ABP) indicates intact autoregulation47,48 (Fig. 46.4). Higher technique using repetitive thigh cuff tests is able to generate
FV (output) and ABP (input) transfer function also indicate rapid increases or decreases of MAP of plus or minus
worse autoregulation.49 In both methods, the absolute value 15 mm Hg and so examine the symmetry of cerebral auto-
of coherence between FV and MAP should be more than 0.5 regulation (i.e., is there an equal compensatory response to
in the analyzed frequency bandwidth. Both methods presume abrupt increases or decreases in ABP).51 Alternatively, inter-
a linear relationship between changes in FV and MAP within mittent application of thigh cuffs using square wave sequences
a studied range of fluctuations. can be used to modulate ABP and so assess CA.52

Calculation of Autoregulation Index Time Correlation Method (Mx Index)


Autoregulation index (ARI) is an “extension” of system analy- This method is based on assumptions that the predominant
sis using modeling of the step-response of the system generat- source of FV fluctuations over a short period of time is MAP
ing changes of flow velocity from changes in arterial pressure. fluctuations. By continuous monitoring over consecutive
To assess dynamic cerebral autoregulation (CA) based on time-averaged samples of FV and MAP (30 to 60, 5- to
450 Section VII—Computers, Engineering, and the Future

100
ABP 90
[mm Hg] 80
70
60
ICP 40
[mm Hg]
20

60
CPP 50
[mm Hg] 40
30
80
FV
[cm/s] 60
40
1
Mx 0.5
0
11:10 11:20 11:30 11:40 11:50 12:00
A Time [h:m]

120
ABP 100
[mm Hg] 80
60
25
ICP 20
[mm Hg] 15

25
CPP 20
[mm Hg] 15
10
120
FV 100
[cm/s] 80
60
40
1
Mx 0.5
0
B 13:56 14:00 14:04 14:08 Time [h:m]
Fig. 46.5  Autoregulation is a dynamic process and can be disturbed over relatively short time periods, such as during plateau waves of intracranial
pressure (ICP) (A). The Mx index increases during the plateau wave and then recovers to baseline values around 0. A similar observation occurs during
hypotension (B). These examples illustrate why, in neurocritical care, autoregulation should be monitored continuously rather than occasionally tested.
ABP, Arterial blood pressure; CCP, cerebral perfusion pressure; FV, flow velocity; Mx, autoregulation.

10-second averages are usually taken), a correlation coefficient in autoregulation in response to secondary insults such as
between mean MAP and mean FV is calculated. This coeffi- arterial hypotension or intracranial hypertension (Fig. 46.5).
cient termed the mean index (Mxa; Mx used in head injury53 Both Mxa and ARI methods are associated with TBI
is a correlation between CPP and FV. Mxa can be calculated outcome.53,54 Some studies, however, suggest that systolic flow
noninvasively, providing a noninvasive measure of blood pres- indices (Sx and Sxa) demonstrate a stronger association with
sure is available). A positive coefficient signifies a positive outcome after TBI than the mean flow indices (Mx and Mxa).55
association between FV and MAP (i.e., disturbed autoregula-
tion). A zero or negative correlation coefficient (i.e., absent or
negative association) implies intact autoregulation. The calcu- Wavelet Analysis
lation may be repeated with a moving time window in which When the time series are either nonstationary or nonlinearly
analysis occurs; therefore Mxa may form new variable and related to one another, the justification for transfer function
indicate changes of cerebral autoregulation with time. This theory is lost. To address this, a procedure has been introduced
index seems to be ideal to monitor transient changes that overlaps with transfer function theory in the linear
Section VII—Computers, Engineering, and the Future 451

90

FV
[cm/s]
80

70
62

TOI 60
[%]
58

12:45 12:50 12:55 13:00 13:05 13:10 13:15


Time [h:m]
Fig. 46.6  Contemporary near infrared machines now are faster, are better able to reduce the influence of extracranial circulation, and
have an improved signal-to-noise ratio. A slow fluctuation of blood flow velocity (FV) recorded by transcranial Doppler (0.05-0.005 Hz), from
which information for continuous monitoring of cerebral autoregulation is generated, is replicated, without visible phase shift, by the tissue oxygen
index (TOI) monitored by near infrared spectroscopy (NIRS). It suggests that TOI can be used as a noninvasive index of autoregulation.

domain, but in this method complex continuous wavelet CBF, cerebral oxygen diffusivity, and CMRO2. Over a short
transforms are used to study the phase difference between period of time (a few minutes), CBF probably is the major
fluctuations in ABP and MCA FV. Its application does not determinant of fluctuations of detected TOI (Fig. 46.6). NIRS
require the assumption that the relation between the two time optodes do not require precise focus on the MCA or continu-
series is or is not linear. Three parameters are used to charac- ous correction for shifts. Preliminary studies based on slow
terize recorded signals: (1) variability of the signal, (2) syn- respiration in patients with carotid artery stenosis,58 transfer
chronization that is analogous of coherence between two functions between slow waves in neonates,59 and a moving
variables, and (3) wavelet “gain,” which characterizes amplifi- linear correlation coefficient between CPP and INVOS cere-
cation of the output signal, flow velocity compared to input bral oximeter waveforms in a pig model suggest that artifact-
(i.e., arterial pressure).56 Increased gain (with good coherence) free continuous monitoring of CA using NIRS can become
may be interpreted as worse cerebral autoregulation. clinically feasible.60 For example, Brady et al.60 defined a cere-
bral oximetry index (COx) in pigs that had 92% sensitivity
(73%-99%) and 63% specificity (48%-76%) to detect loss of
Multimodal Decomposition autoregulation associated with hypotension when COx was
To further overcome problems related to nonstationary and above a threshold of 0.36. Further studies in a swine model of
nonlinearity, a novel computational method called multi- hypoxic-asphyxic cardiac arrest suggest that use of NIRS to
modal pressure-flow (MMPF) analysis was developed to study calculate the lower limit of autoregulation (LLA) is compa-
the ABP-FV relationship during the Valsalva maneuver. The rable to LLA calculated using laser Doppler flow measure-
MMPF method enables evaluation of autoregulatory dynam- ments.61 In the clinical environment NIRS-based studies in
ics based on instantaneous phase analysis of blood pressure adults who undergo cardiac surgery suggest there is a wide
(BP) and blood FV oscillations induced by the intervention. range of MAP at the LLA62 thus emphasizing a role for moni-
While applying this technique, a characteristic phase lag toring rather than an empiric choice of MAP.
between blood flow volume and BP oscillations induced by NIRS also has been used to assess cerebrovascular reactivity
the Valsalva maneuver was found in healthy subjects, and this noninvasively using the total hemoglobin reactivity index
phase lag was reduced in patients with hypertension and (THx) that is analogous to the pressure reactivity index (PRx).
stroke.57 These findings suggested that ABP-FV phase lag This NIRS-based cerebrovascular reactivity index THx can be
could serve as an index of CA. The same method can be used used as a noninvasive substitute for PRx, but only during
for continuous monitoring of CA, analyzing spontaneous phases with sufficient slow wave power in the input signal.63
fluctuation of ABP.
Brain Oxygenation
Possible Use of Near Infrared Spectroscopy NIRS is a noninvasive method to study brain oxygen satura-
Transcranial Doppler is operator dependent, and in some tion (see Chapter 33). In this method an NIRS sensor is
patients it is difficult to find and insonate the MCA. Further- applied to the forehead and its probe emits and detects wave-
more, once found the ultrasound probe may shift in position lengths of light millimeters from its tip. As the near infrared
over time scales of a few minutes to hours. Therefore other absorption characteristics of hemoglobin are known, the
noninvasive modalities proportional to changes in CBF have system calculates brain tissue oxygen saturation by measuring
been used instead of blood flow velocity. For example, near differences in intensity of light as it passes through the brain.
infrared spectroscopy (NIRS)–derived oxygenated hemoglo- The sensor then converts these wavelengths to a measurement
bin (HbO2), tissue oxygen index (TOI), and cerebral oxygen- that represents regional oxygen saturation. The saturations
ation index (CO) are associated with arterial oxygen saturation, range from 15% to 95%. In the brain the major source of
452 Section VII—Computers, Engineering, and the Future

80

ABP 60
[mm Hg] 40
20
0
65
60
FV 55
[cm/s] 50
45
40
35
30
71
70
TOI 69
[%] 68
67
66

5.5
5
EtCO2 4.5
[kPa] 4
3.5
3
17:36 17:39 17:42 17:45 17:48 17:51
Time [h:m]
Fig. 46.7  Line graphs illustrating carbon dioxide (CO2) reactivity testing with a near infrared spectroscopy (NIRS) machine. The increase
in blood flow velocity (FV) in the hypercapnic phase and tissue oxygen index (TOI) are almost correlated. If calibration of TOI is stable across a population
of patients, NIRS may become a viable test cerebrovascular reactivity. ABP, Arterial blood pressure; etCO2, end-tidal carbon dioxide.

tissue oxygen content is the blood, which is influenced by sensitivity of the NIRS technique was very limited unless the
brain oxygen delivery, consumption, and the arteriovenous extracranial tissues were removed from the measurement, in
blood volume ratio in the microcirculation. this study made possible by clamping the external carotid
There is a larger experience with this technology in cardiac artery (ECA).68 When the ECA was clamped, there was better
surgery patients and in the pediatric population than in concordance between ischemia, defined by TCD and EEG
NCCU use. Such studies indicate that optimal saturations are and NIRS readings. Newer NIRS designs apply an algorithm
about 75% and that levels less than 50% or a 20% reduction called spatially resolved spectroscopy, to calculate the tissue-
from baseline are associated with worse neurologic outcome.64,65 oxygen index. This index appears independent of the extra-
These values are consistent with animal data showing that cranial circulation and is associated with a CBF decrease
baseline values are about 70%, cerebral lactate increases when during carotid artery clamping.69 Although this makes this
NIRS brain oxygen saturation is 45%, the electroencephalo- technique useful during carotid surgery, the application of
gram (EEG) changes when saturation is 42%, and adenosine NIRS as a noninvasive modality in NCCU patients is still
triphosphate (ATP) levels are reduced when saturation is being elucidated but includes detection of secondary cerebral
33%.66 In patients with severe TBI, noninvasive transcranial ischemia, assessment of cerebrovascular reactivity to PaCO2
oxygen saturation (StcO2) values greater than 70% are associ- changes (Fig. 46.7), or assessment of cerebral pressure
ated with better outcome and fewer interventions to treat ICP. autoregulation.70,71
However, the difference in StcO2 between those who died or
survived was only 10% (70% vs. 60%).67 NIRS also can be used
to detect peri-infarct depolarizations (PIDs) in ischemic
Imaging
stroke. This has significant implications; if penumbral isch- Imaging modalities offer the advantage of giving both quan-
emia can be predicted by PIDs and is associated with NIRS titative and qualitative measurements of cerebral physiology.
recordings, then peri-infarct tissue may be salvaged by therapy The disadvantage of any imaging modality is that only a snap-
to improve cerebral perfusion.54 shot in time is examined—they cannot provide continuous
In adults there are several limitations to NIR-derived cere- data. Nonetheless, different imaging modalities can aid in
bral oxygen levels: (1) high scattering and attenuation of light noninvasive monitoring of patients in the NCCU.
in tissue, especially the cranium, (2) the difficulty of estimat-
ing the path length of light through tissue, (3) the inability
to distinguish between blood contained in arteries and veins, Magnetic Resonance Imaging
and (4) contamination of NIRS readings by extracranial The role of MRI in neurocritical care is discussed in Chapter
tissues. Indeed, Kirkpatrick et al. in a study of patients under- 28. In particular, advanced MRI techniques can help in iden-
going carotid endarterectomy, showed that the specificity or tifying patients with severe diffuse axonal injury (DAI) and in
Section VII—Computers, Engineering, and the Future 453

outcome prediction. For example, advanced techniques such quantitative, SPECT is qualitative but can be used to examine
as diffusion tensor imaging (DTI) are more sensitive than for hypoperfusion and so is suited to examination for condi-
conventional MRI sequences to detect white matter injury. tions such as vasospasm after SAH, particularly when per-
These techniques can also be used to examine the extent of formed with TCD evaluation. PET and SPECT have been used
structural integrity after hypoxic-ischemic injury and thus in a variety of neurologic diseases including Alzheimer’s
inform care.72 There are a variety of imaging sequences but disease, Huntington’s chorea, cerebral microangiopathy, and
one, MRI spectroscopy (MRS), lends itself to noninvasive TBI.78-82 In TBI, brain hypometabolism is frequent and the
monitoring of cellular function. In MRS intracellular metabo- severity of trauma correlates well with degree of hypometabo-
lites such as N-acetyl aspartate (NAA), choline, creatine (Cr), lism.83,84 This can be detected using PET or SPECT, although
and lactate are measured. These metabolites provide a reliable the results may always be concordant such as when edema
assessment of cellular function: (1) NAA indicates the propor- compromises blood flow without causing ischemia. In this
tion of dead or living neuronal cells; (2) choline levels are case PET would be normal but SPECT abnormal.
associated with cell membrane activity; (3) Cr provides insight In CT perfusion imaging, iodinated contrast is adminis-
into global metabolic level; and (4) increased lactate suggests tered and then regions of interest are referenced to known
cellular ischemia. In TBI, the NAA/Cr ratio is decreased by as blood vessels such as the pericallosal arteries or superior sagit-
much as 20% in areas of DAI.73 The combination of MRI and tal sinus. From these data, values such as CBV, CBF, and mean
MRS also can aid in TBI outcome prediction, particularly transit time (MTT) can be generated.85,86 If the perfusion scan
when the MRI is near normal.74 is obtained at two different blood pressures, the perfusion
With the advent of functional MRI (fMRI), it has become characteristics can be examined for loss of autoregulation. In
possible to assess the integrity of vascular coupling and neural normal autoregulation, reduced CBF is thought to trigger
function at risk in areas of brain injury and stroke. Thus fMRI vasodilation with a subsequent increase of CBV elevation to
can help assess function of tissues at risk noninvasively. fMRI maintain adequate tissue perfusion. Reduced CBV in the pres-
can be difficult to perform in sedated patients but spontane- ence of ischemia in turn indicates that autoregulation is
ous blood oxygen level–dependent (BOLD) fluctuations can impaired.
be observed without patient cooperation (i.e., resting state
fMRI) The BOLD contrast originates from intravoxel mag-
netic field inhomogeneity induced by paramagnetic deoxyhe- New Directions: Optoacoustic Techniques
moglobin in erythrocytes. This imaging is based on the The optoacoustic (or photoacoustic) technique uses short
concept that neural activity is intricately coupled to regional near-infrared laser pulses to generate ultrasonic waves in
CBF. When a task is performed, regional CBF increases dis- tissue due to fast thermoelastic expansion of heated volumes.
proportionately, which can be measured by techniques such Using advanced signal processing, Petrov et al.87 demon-
as arterial spin labeling (ASL). The CBF increase overcompen- strated a good correlation with known oxygenation values in
sates for the stimulus-evoked increase in oxygen consumption the internal jugular vein and those obtained by their opto-
needed to fuel the elevated neural activity relative to basal acoustic probe in normal volunteers. Further development of
conditions. In animal studies of cerebral ischemia, neural this technique could provide an alternative to invasive
activity can be lost in primary somatosensory cortex but vas- methods of brain oxygenation monitoring and provide a
cular coupling remains intact before changes in apparent dif- noninvasive insight into edema or hemodynamic changes in
fusion coefficient on MRI occur,75 that is, fMRI could be a the brain.88-90
predictor of at-risk brain. In TBI, intact task-correlated
sensory and cognitive BOLD hemodynamic responses to
stimuli have been observed in a comatose patient in whom Electrophysiology
electrophysiologic studies indicated absent thalamocortical
processing, suggesting that fMRI may help predict outcome in
in Neurocritical Care
nonresponsive TBI patients.76 Evaluation of brain function using electrophysiologic tech-
niques such as the EEG, bispectral index (BIS), evoked poten-
tial monitoring, and the rheoencephalogram is noninvasive
Other Noninvasive Methods of Metabolic and these techniques are useful adjuncts to patient care in the
and Flow Imaging NCCU.
Positron emission tomography (PET) scanning is an imaging
modality that uses cellular consumption of substances such as
glucose (18 F [fluorodeoxyglucose]) as its imaging substrate EEG and BIS
(see Chapter 29). This allows identification of glucose use in Electrical activity continuously emanates from the superficial
the brain. Alternatively, oxygen metabolism can be studied cerebral cortex; this can be examined using the EEG. Several
using PET by injection of 15-O–labeled water. Regional CBV lines of evidence provide a compelling argument for bedside
can be assessed by inhalation of 15-O–labeled carbon mon- continuous EEG (cEEG) in the NCCU to detect seizures,
oxide and CMRO2 measured using 15-O–labeled oxygen.77 nonconvulsive seizures, ICP changes, and cerebral ischemia,
PET can then be combined with CT or MRI for dual imaging outcome prediction, and to titrate therapies such as barbitu-
that gives more accurate anatomic localization. Single photon rates in a variety of conditions such as TBI, intracerebral
emission computed tomography (SPECT) is another “meta- hemorrhage, stroke, SAH, CNS infections, and after cardiac
bolic imaging technique” in which technetium-99-m- arrest91-98 (see also Chapter 25). cEEG provides good tempo-
hexamethyl propyleneamine is intravenously injected into ral and spatial resolution but can be technically difficult to
the patient’s bloodstream (see Chapter 29). Whereas PET is implement and is susceptible to drug effects and artifacts
454 Section VII—Computers, Engineering, and the Future

such as eye movements, patient movement, or electrical standard monitor, and in TBI, guidelines recommend that ICP
interference.99 New software algorithms make it easier for should be monitored invasively in patients with severe injury
non-electrophysiologists and non-epileptologists to identify (i.e., Glasgow Coma Scale score ≤8). Similar indications are
relevant information (e.g., compressed spectral array for sei- used for patients in coma associated with pathologies such as
zures and relative alpha variability for ischemia). However, SAH or stroke. For other patients—those with moderate
vast amounts of data are still generated and successful use of TBI—the role for invasive ICP monitoring is unclear. In these
cEEG requires efficient computer networks and information patients noninvasive estimation of ICP may help select patients
technology (IT) support and is resource dependent. This may for invasive monitoring. In addition, noninvasive monitoring
limit cEEG use to larger centers.100 However, a decreased may be useful when the risk of invasive monitoring is high
length of stay may offset this increased cost.101 such as in hepatic encephalopathy.
BIS represents a summary of EEG activity in which 0 rep- There is a relatively long history and several approaches—
resents a flat EEG and 100 represents normal wakefulness. To imaging (CT and MRI), TCD, NIRS, evoked potentials, EEG—
perform BIS, a sensor is placed over the forehead from which and techniques to evaluate the optic nerve sheath among
data are gathered. Although not as sensitive as cEEG, BIS is others in noninvasive monitoring of intracranial hemody-
simple and values greater than 60 are associated with better namics. These techniques remain largely exploratory in large
outcome and better control of ICP.102 part because none of methods are accurate enough and easy
to use at same time. For example, ophthalmodynamometry,
although precise, is unsuitable for continuous monitoring and
Evoked Potentials cumbersome for repeated measurement. In addition, the mea-
Evoked potential monitoring includes modalities such as sured characteristic often is not only a function of ICP but
somatosensory evoked potentials (SSEPs), visual evoked also other factors such as ABP (TCD, ultrasound) or func-
potentials (VEPs), auditory evoked potentials (AEPs), and tional integrity of a specific brain region (EEG). At present
brainstem auditory evoked potentials (BAEPs; see Chapter ultrasound measurements of the ONS diameter and Doppler
24). Evoked potentials (EPs) differ from EEG in that they flow are the most promising noninvasive techniques. These
represent the brain’s response to repetitive stimuli along a methods may be useful in select patients, but future improve-
specific nerve pathway. In addition EPs are an order of mag- ments will depend on improvement in accuracy. Ideally, in the
nitude smaller in voltage than the voltages seen on EEG. SSEPs future noninvasive monitoring could replace invasive intra-
are the most frequently used of these methods in the intensive cranial monitors.
care unit (ICU). In this technique, responses to median nerve
(or tibial nerve) stimulation are recorded at the cortical level. References
A variety of parameters are measured and useful in the NCCU:
cervical (C7, C2) and cortical evoked potentials; the interpeak 1. Samuel M, Burge DM, Marchbanks RJ. Tympanic membrane displacement
testing in regular assessment of intracranial pressure in eight children with
latencies C2/N20, also referred to as the central conduction shunted hydrocephalus. J Neurosurg 1998;88(6):983–95.
time (CCT); and the interrelation of the amplitudes N20/N13. 2. Plandsoen WCG, de Jong DA, Maas AIR, et al. Fontanelle pressure
SSEPs may be abnormal even when there are demonstrable monitoring in infants with the Rotterdam teletransducer: a reliable
lesions on imaging studies. EP monitoring is primarily used technique. Med Prog Technol 1987;13:21–271.
3. Klingelhofer J, Conrad B, Benecke R, et al. Evaluation of intracranial
to predict outcome rather than as a guide to therapy. In pressure from transcranial Doppler studies in cerebral disease. J Neurol
general, a significant prolongation of CCT, reduced amplitude 1988;235:159–62.
of cortical responses, a low amplitude quotient N20/N13, and 4. Prunet B, Asencio Y, Lacroix G, et al. Noninvasive detection of elevated
bilateral loss of the cortical response are associated with poor intracranial pressure using a portable ultrasound system. Am J Emerg Med
outcome.102 2011; Jun 10. Epub ahead of print.
5. Soldatos T, Chatzimichail K, Papathanasiou M, et al. Optic nerve
sonography: a new window for the non-invasive evaluation of intracranial
pressure in brain injury. Emerg Med J 2009;26(9):630–4. Review.
Rheoencephalography 6. Moretti R, Pizzi B. Ultrasonography of the optic nerve in neurocritically ill
Disrupted cerebral vasomotor autoregulation is associated patients. Acta Anaesthesiol Scand 2011;55(6):644–52.
7. Behrens A, Lenfeld N, Ambarki K, et al. Trans-cranial Doppler pulsatility
with poor outcome. CT perfusion (discussed earlier) or elec- index: not an accurate method to assess intracranial pressure. Neurosurgery
trical impedance studies, namely rheoencephalography (REG) 2010;66(6):1050–7.
can be used to examine this phenomenon. REG works by 8. Rosenberg JB, Shiloh AL, Savel RH, et al. Non-invasive methods of
applying electrodes on the scalp in a bipolar arrangement and estimating intracranial pressure. Neurocrit Care 2011;15(3):599–608.
is based on monitoring pulse-synchronous variations in 9. Ragauskas A, Daubaris G, Dziugys A, et al. Innovative non-invasive method
for absolute intracranial pressure measurement without calibration. Acta
cranial electrical impedance over time to derive CBF measure- Neurochir Suppl 2005;95:351–61.
ments. Animal studies suggest REG can be used to noninva- 10. Ueno T, Ballard RE, Shuer LM, et al. Intracranial pressure dynamics during
sively assess CBF in intracerebral hemorrhage and to assess simulated microgravity using a new noninvasive ultrasonic technique.
compliance of the cerebral arterial bed.103 Scalp thickness, J Gravit Physiol 1998;5(1):P39–40.
11. Schoser BG, Riemenschneider N, Hansen HC. The impact of raised
however, can affect the measurement quite significantly, intracranial pressure on cerebral venous hemodynamics: a prospective venous
despite rigorous efforts to mathematically exclude it104 and so transcranial Doppler ultrasonography study. J Neurosurg 1999;91(5):744–9.
REG is infrequently used in the ICU. 12. Zhao YL, Zhou JY, Zhu GH. Clinical experience with the non-invasive ICP
monitoring system. Acta Neurochir Suppl 2005;95:351–3.
13. Raksin PB, Alperin N, Sivaramakrishnan A, et al. Noninvasive intracranial
Conclusion compliance and pressure based on dynamic magnetic resonance imaging of
blood flow and cerebrospinal fluid flow: review of principles,
A variety of invasive intracranial monitors are used in the implementation, and other noninvasive approaches. Neurosurg Focus
NCCU. At present invasive ICP measurement is the gold 2003;14(4):e4.
Section VII—Computers, Engineering, and the Future 455

14. Geeraerts T, Merceron S, Benhamou D, et al. Non-invasive assessment of 33. Querfurth HW, Arms SW, Lichy CM, et al. Prediction of intracranial
intracranial pressure using ocular sonography in neurocritical care patients. pressure from noninvasive transocular venous and arterial hemodynamic
Intensive Care Med 2008;34(11):2062–7. measurements: a pilot study. Neurocrit Care 2004;1:183–94.
15. Aaslid R, Markwalder TM, Nornes H. Noninvasive transcranial Doppler 34. Querfurth HW, Lieberman P, Arms S, et al. Ophthalmodynamometry for
ultrasound recording of flow velocity in basal cerebral arteries. J Neurosurg ICP prediction and pilot test on Mt. Everest. BMC Neurol 2010 1;10:106.
1982;57(6):769–74. 35. Scott CJ, Kardon RH, Lee AG, et al. Diagnosis and grading of papilledema in
16. Chan KH, Miller DJ, Dearden M, et al. The effect of changes in cerebral patients with raised intracranial pressure using optical coherence
perfusion pressure upon middle cerebral artery blood flow velocity and tomography vs clinical expert assessment using a clinical staging scale. Arch
jugular bulb venous oxygen saturation after severe brain trauma. Ophthalmol 2010;128:705–11.
J Neurosurg 1992;77:55–61. 36. Rebolleda G, Munoz-Negrete FJ. Follow-up of mild papilledema in
17. Thees C, Scholz M, Schaller MDC, et al. Relationship between intracranial idiopathic intracranial hypertension with optical coherence tomography.
pressure and critical closing pressure in patients with neurotrauma. Invest Ophthalmol Vis Sci 2009;50:5197–200.
Anesthesiology 2002;96(3):595–9. 37. Trick GL, Vesti E, Tawansy K, et al. Quantitative evaluation of papilledema in
18. Czosnyka M, Smielewski P, Piechnik S, et al. Critical closing pressure in pseudotumor cerebri. Invest Ophthalmol Vis Sci 1998;39:1964–71.
cerebrovascular circulation. J Neurol Neurosurg Psychiatry 38. Heckmann JG, Weber M, Junemann AG, et al. Laser scanning tomography of
1999;66(5):606–11. the optic nerve vs CSF opening pressure in idiopathic intracranial
19. Aaslid R, Lundar T, Lindegaard K-F, et al. Estimation of cerebral perfusion hypertension. Neurology 2004;62:1221–3.
pressure from arterial blood pressure and transcranial Doppler recordings. 39. Tayal VS, Neulander M, Norton HJ, et al. Emergency department
In: Miller JD, Teasdale GM, Rowan JO, et al, editors. Intracranial pressure VI. sonographic measurement of optic nerve sheath diameter to detect findings
Berlin/Heidelberg: Springer Verlag; 1986. p. 229–31. of increased intracranial pressure in adult head injury patients. Ann Emerg
20. Schmidt EA, Czosnyka M, Gooskens I, et al. Preliminary experience Med 2007;49:508–14.
of the estimation of cerebral perfusion pressure using transcranial 40. Moretti R, Pizzi B, Cassini F, et al. Reliability of optic nerve ultrasound for
Doppler ultrasonography. J Neurol Neurosurg Psychiatry 2001;70(2): the evaluation of patients with spontaneous intracranial hemorrhage.
198–204. Neurocrit Care 2009;11:406–10.
21. Bellner J, Romner B, Reinstrup P, et al. Transcranial Doppler sonography 41. Rajajee V, Vanaman M, Fletcher JJ, et al. Optic nerve ultrasound for the
pulsatility index (PI) reflects intracranial pressure (ICP). Surg Neurol detection of raised intracranial pressure. Neurocrit Care 2011;Jul 19. Epub
2004;62(1):45–51; discussion 51. ahead of print.
22. Schmidt B, Klingelhofer J, Schwarze JJ, et al. Noninvasive prediction of 42. Potgieter DW, Kippin A, Ngu F, et al. Can accurate ultrasonographic
intracranial pressure curves using transcranial Doppler ultrasonography and measurement of the optic nerve sheath diameter (a non-invasive measure of
blood pressure curves. Stroke 1997;28(12):2465–72. intracranial pressure) be taught to novice operators in a single training
23. Reulen HJ, Graham R, Spatz M, et al. Role of pressure gradient and session? Anaesth Intensive Care 2011;39:95–100.
bulk flow in dynamics of vasogenic brain edema. J Neurosurg 1977;46: 43. Ballantyne SA, O’Neill G, Hamilton R, et al. Observer variation in the
24–35. sonographic measurement of optic nerve sheath diameter in normal adults.
24. Carhuapoma JR, Qureshi AI, Bhardwaj A, et al. Interhemispheric intracranial Eur J Ultrasound 2002;15:145–9.
pressure gradients in massive cerebral infarction. J Neurosurg Anesthesiol 44. Copetti R, Cattarossi L. Optic nerve ultrasound: artifacts and real images.
2002;14(4):299–303. Intensive Care Med 2009;35:1488–9.
25. Bundgaard H, Cold GE. Studies of regional subdural pressure gradients 45. Sharma D, Vavilala MS. Lund concept for the management of traumatic
during craniotomy. Br J Neurosurg 2000;14(3):229–34. brain injury: a physiological principle awaiting stronger evidence.
26. Kumar A, Schmidt EA, Hiler M, et al. Asymmetry of critical closing J Neurosurg Anesthesiol 2011;23(4):363–7.
pressure following head injury. J Neurol Neurosurg Psychiatry 2005;76(11): 46. Aoi MC, Matzuka BJ, Olufsen MS. Toward online, noninvasive, nonlinear
1570–3. assessment of cerebral autoregulation. Conf Proc IEEE Eng Med Biol Soc
27. Schmidt EA, Czosnyka M, Steiner LA, et al. Asymmetry of pressure 2011;2011:2410–3.
autoregulation after traumatic brain injury. J Neurosurg 2003;99(6): 47. Birch AA, Dirnhuber MJ, Hartley-Davies R, et al. Assessment of
991–8. autoregulation by means of periodic changes in blood pressure. Stroke
28. Hansen HC, Helmke K, Kunze K. Optic nerve sheath enlargement in acute 1995;26(5):834–7.
intracranial hypertension. Neuroophthalmology 1994;14:345–54. 48. Reinhard M, Müller T, Guschlbauer B, et al. Transfer function analysis for
29. Steffen H, Eifert B, Aschoff A, et al. The diagnostic value of optic disc clinical evaluation of dynamic cerebral autoregulation: a comparison
evaluation in acute elevated intracranial pressure. Ophthalmology 1996;103: between spontaneous and respiratory-induced oscillations. Physiol Meas
1229–32. 2003;24(1):27–43.
30. Hayreh SS. Optic disc edema in raised intracranial pressure. V. Pathogenesis. 49. Zhang R, Zuckerman JH, Giller CA, et al. Transfer function analysis of
Arch Ophthalmol 1977;95:1553–65. dynamic cerebral autoregulation in humans. Am J Physiol 1998;274
31. Querfurth HW, Lagrèze WD, Hedges TR, et al. Flow velocity and pulsatility (1 Pt 2):H233–41.
of the ocular circulation in chronic intracranial hypertension. Acta Neurol 50. Tiecks FP, Lam AM, Aaslid R, et al. Comparison of static and dynamic
Scand 2002;105:431–40. cerebral autoregulation measurements. Stroke 1995;26(6):1014–9.
32. Miller MM, Chang T, Keating R, et al. Blood flow velocities are reduced in
the optic nerve of children with elevated intracranial pressure. J Child A complete list of references for this chapter can be found online at
Neurol 2009;24:30–5. www.expertconsult.com.
Section VII—Computers, Engineering, and the Future 455.e1

References 26. Kumar A, Schmidt EA, Hiler M, et al. Asymmetry of critical closing
pressure following head injury. J Neurol Neurosurg Psychiatry
1. Samuel M, Burge DM, Marchbanks RJ. Tympanic membrane displacement 2005;76(11):1570–3.
testing in regular assessment of intracranial pressure in eight children with 27. Schmidt EA, Czosnyka M, Steiner LA, et al. Asymmetry of pressure
shunted hydrocephalus. J Neurosurg 1998;88(6):983–95. autoregulation after traumatic brain injury. J Neurosurg 2003;99(6):991–8.
2. Plandsoen WCG, de Jong DA, Maas AIR, et al. Fontanelle pressure 28. Hansen HC, Helmke K, Kunze K. Optic nerve sheath enlargement in acute
monitoring in infants with the Rotterdam teletransducer: a reliable intracranial hypertension. Neuroophthalmology 1994;14:345–54.
technique. Med Prog Technol 1987;13:21–271. 29. Steffen H, Eifert B, Aschoff A, et al. The diagnostic value of optic disc
3. Klingelhofer J, Conrad B, Benecke R, et al. Evaluation of intracranial evaluation in acute elevated intracranial pressure. Ophthalmology
pressure from transcranial Doppler studies in cerebral disease. J Neurol 1996;103:1229–32.
1988;235:159–62. 30. Hayreh SS. Optic disc edema in raised intracranial pressure. V.
4. Prunet B, Asencio Y, Lacroix G, et al. Noninvasive detection of elevated Pathogenesis. Arch Ophthalmol 1977;95:1553–65.
intracranial pressure using a portable ultrasound system. Am J Emerg Med 31. Querfurth HW, Lagre’ze WD, Hedges TR, et al. Flow velocity and pulsatility
2011; Jun 10. Epub ahead of print. of the ocular circulation in chronic intracranial hypertension. Acta Neurol
5. Soldatos T, Chatzimichail K, Papathanasiou M, et al. Optic nerve Scand 2002;105:431–40.
sonography: a new window for the non-invasive evaluation of intracranial 32. Miller MM, Chang T, Keating R, et al. Blood flow velocities are reduced in
pressure in brain injury. Emerg Med J 2009;26(9):630–4. Review. the optic nerve of children with elevated intracranial pressure. J Child
6. Moretti R, Pizzi B. Ultrasonography of the optic nerve in neurocritically ill Neurol 2009;24:30–5.
patients. Acta Anaesthesiol Scand 2011;55(6):644–52. 33. Querfurth HW, Arms SW, Lichy CM, et al. Prediction of intracranial
7. Behrens A, Lenfeld N, Ambarki K, et al. Trans-cranial Doppler pulsatility pressure from noninvasive transocular venous and arterial hemodynamic
index: not an accurate method to assess intracranial pressure. Neurosurgery measurements: a pilot study. Neurocrit Care 2004;1:183–94.
2010;66(6):1050–7. 34. Querfurth HW, Lieberman P, Arms S, et al. Ophthalmodynamometry for
8. Rosenberg JB, Shiloh AL, Savel RH, et al. Non-invasive methods of ICP prediction and pilot test on Mt. Everest. BMC Neurol 2010 1;10:106.
estimating intracranial pressure. Neurocrit Care 2011;15(3):599–608. 35. Scott CJ, Kardon RH, Lee AG, et al. Diagnosis and grading of papilledema
9. Ragauskas A, Daubaris G, Dziugys A, et al. Innovative non-invasive method in patients with raised intracranial pressure using optical coherence
for absolute intracranial pressure measurement without calibration. Acta tomography vs clinical expert assessment using a clinical staging scale. Arch
Neurochir Suppl 2005;95:351–61. Ophthalmol 2010;128:705–11.
10. Ueno T, Ballard RE, Shuer LM, et al. Intracranial pressure dynamics during 36. Rebolleda G, Munoz-Negrete FJ. Follow-up of mild papilledema in
simulated microgravity using a new noninvasive ultrasonic technique. idiopathic intracranial hypertension with optical coherence tomography.
J Gravit Physiol 1998;5(1):P39–40. Invest Ophthalmol Vis Sci 2009;50:5197–200.
11. Schoser BG, Riemenschneider N, Hansen HC. The impact of raised 37. Trick GL, Vesti E, Tawansy K, et al. Quantitative evaluation of papilledema
intracranial pressure on cerebral venous hemodynamics: a prospective in pseudotumor cerebri. Invest Ophthalmol Vis Sci 1998;39:1964–71.
venous transcranial Doppler ultrasonography study. J Neurosurg 38. Heckmann JG, Weber M, Junemann AG, et al. Laser scanning tomography
1999;91(5):744–9. of the optic nerve vs CSF opening pressure in idiopathic intracranial
12. Zhao YL, Zhou JY, Zhu GH. Clinical experience with the non-invasive ICP hypertension. Neurology 2004;62:1221–3.
monitoring system. Acta Neurochir Suppl 2005;95:351–3. 39. Tayal VS, Neulander M, Norton HJ, et al. Emergency department
13. Raksin PB, Alperin N, Sivaramakrishnan A, et al. Noninvasive intracranial sonographic measurement of optic nerve sheath diameter to detect findings
compliance and pressure based on dynamic magnetic resonance imaging of of increased intracranial pressure in adult head injury patients. Ann Emerg
blood flow and cerebrospinal fluid flow: review of principles, Med 2007;49:508–14.
implementation, and other noninvasive approaches. Neurosurg Focus 40. Moretti R, Pizzi B, Cassini F, et al. Reliability of optic nerve ultrasound for
2003;14(4):e4. the evaluation of patients with spontaneous intracranial hemorrhage.
14. Geeraerts T, Merceron S, Benhamou D, et al. Non-invasive assessment of Neurocrit Care 2009;11:406–10.
intracranial pressure using ocular sonography in neurocritical care patients. 41. Rajajee V, Vanaman M, Fletcher JJ, et al. Optic nerve ultrasound for the
Intensive Care Med 2008;34(11):2062–7. detection of raised intracranial pressure. Neurocrit Care 2011;Jul 19. Epub
15. Aaslid R, Markwalder TM, Nornes H. Noninvasive transcranial Doppler ahead of print.
ultrasound recording of flow velocity in basal cerebral arteries. J Neurosurg 42. Potgieter DW, Kippin A, Ngu F, et al. Can accurate ultrasonographic
1982;57(6):769–74. measurement of the optic nerve sheath diameter (a non-invasive measure
16. Chan KH, Miller DJ, Dearden M, et al. The effect of changes in cerebral of intracranial pressure) be taught to novice operators in a single training
perfusion pressure upon middle cerebral artery blood flow velocity and session? Anaesth Intensive Care 2011;39:95–100.
jugular bulb venous oxygen saturation after severe brain trauma. 43. Ballantyne SA, O’Neill G, Hamilton R, et al. Observer variation in the
J Neurosurg 1992;77:55–61. sonographic measurement of optic nerve sheath diameter in normal adults.
17. Thees C, Scholz M, Schaller MDC, et al. Relationship between intracranial Eur J Ultrasound 2002;15:145–9.
pressure and critical closing pressure in patients with neurotrauma. 44. Copetti R, Cattarossi L. Optic nerve ultrasound: artifacts and real images.
Anesthesiology 2002;96(3):595–9. Intensive Care Med 2009;35:1488–9.
18. Czosnyka M, Smielewski P, Piechnik S, et al. Critical closing pressure in 45. Sharma D, Vavilala MS. Lund concept for the management of traumatic
cerebrovascular circulation. J Neurol Neurosurg Psychiatry brain injury: a physiological principle awaiting stronger evidence.
1999;66(5):606–11. J Neurosurg Anesthesiol 2011;23(4):363–7.
19. Aaslid R, Lundar T, Lindegaard K-F, et al. Estimation of cerebral perfusion 46. Aoi MC, Matzuka BJ, Olufsen MS. Toward online, noninvasive, nonlinear
pressure from arterial blood pressure and transcranial Doppler recordings. assessment of cerebral autoregulation. Conf Proc IEEE Eng Med Biol Soc
In: Miller JD, Teasdale GM, Rowan JO, et al, editors. Intracranial pressure 2011;2011:2410–3.
VI. Berlin/Heidelberg: Springer Verlag; 1986. p. 229–31. 47. Birch AA, Dirnhuber MJ, Hartley-Davies R, et al. Assessment of
20. Schmidt EA, Czosnyka M, Gooskens I, et al. Preliminary experience of the autoregulation by means of periodic changes in blood pressure. Stroke
estimation of cerebral perfusion pressure using transcranial Doppler 1995;26(5):834–7.
ultrasonography. J Neurol Neurosurg Psychiatry 2001;70(2):198–204. 48. Reinhard M, Müller T, Guschlbauer B, et al. Transfer function analysis for
21. Bellner J, Romner B, Reinstrup P, et al. Transcranial Doppler sonography clinical evaluation of dynamic cerebral autoregulation: a comparison
pulsatility index (PI) reflects intracranial pressure (ICP). Surg Neurol between spontaneous and respiratory-induced oscillations. Physiol Meas
2004;62(1):45–51; discussion 51. 2003;24(1):27–43.
22. Schmidt B, Klingelhofer J, Schwarze JJ, et al. Noninvasive prediction of 49. Zhang R, Zuckerman JH, Giller CA, et al. Transfer function analysis of
intracranial pressure curves using transcranial Doppler ultrasonography dynamic cerebral autoregulation in humans. Am J Physiol 1998;274(1 Pt
and blood pressure curves. Stroke 1997;28(12):2465–72. 2):H233–41.
23. Reulen HJ, Graham R, Spatz M, et al. Role of pressure gradient and bulk 50. Tiecks FP, Lam AM, Aaslid R, et al. Comparison of static and dynamic
flow in dynamics of vasogenic brain edema. J.Neurosurg 1977;46:24–35. cerebral autoregulation measurements. Stroke 1995;26(6):1014–9.
24. Carhuapoma JR, Qureshi AI, Bhardwaj A, et al. Interhemispheric 51. Aaslid R, Blaha M, Sviri G, et al. Asymmetric dynamic cerebral
intracranial pressure gradients in massive cerebral infarction. J Neurosurg autoregulatory response to cyclic stimuli. Stroke 2007;38(5):1465–9.
Anesthesiol 2002;14(4):299–303. 52. Katsogridakis E, Bush G, Fan L, et al. Random perturbations of arterial
25. Bundgaard H, Cold GE. Studies of regional subdural pressure gradients blood pressure for the assessment of dynamic cerebral autoregulation.
during craniotomy. Br J Neurosurg 2000;14(3):229–34. Physiol Meas 2012;33(2):103–16.
455.e2 Section VII—Computers, Engineering, and the Future

53. Czosnyka M, Smielewski P, Piechnik S, et al. Cerebral autoregulation 78. Boecker H, Kuwert T, Langen K. SPECT with HMPAO compared to PET
following head injury. J Neurosurg 2001;95(5):756–63. with FDG in Huntingdon disease. J Comput Assist Tomogr 1994;19:
54. Panerai RB, Kerins V, Fan L, et al. Association between dynamic cerebral 542–8.
autoregulation and mortality in severe head injury. Br J Neurosurg 79. Davis P, Micra S, Alazraki N. The brain in older persons with and without
2004;18(5):471–9. dementia: findings on MRI, PET, and SPECT. Am J Roentgenol
55. Budohoski KP, Reinhard M, Aries MJ, et al. Monitoring cerebral 1994;162:1267–78.
autoregulation after head injury: which component of transcranial 80. Gross H, Kling A, Henry G, et al. Local cerebral glucose metabolism in
Doppler flow velocity is optimal? Neurocrit Care 2012;17(2):211–8. patients with long term behavioral and cognitive deficits following mild
56. Latka M, Turalska M, Glaubic-Latka M, et al. Phase dynamics in cerebral TBI. J Neuropsych Clin Neurosci 1996;8:324–34.
autoregulation. Am J Physiol Heart Circ Physiol 2005;289(5):H2272–9. 81. Mielke R, Petrzyk U, Jacobs A. HMPAO SPECT and FDG PET in
57. Novak V, Yang AC, Lepicovsky L, et al. Multimodal pressure-flow method to Alzheimer’s and vascular dementia: comparison of perfusion and metabolic
assess dynamics of cerebral autoregulation in stroke and hypertension. pattern. Eur J Nuc Med 1994;10:1052–60.
Biomed Eng Online 2004 5;3(1):39. 82. Yamaki T Imahori Y, Ohomori Y. Cerebral hemodynamics and metabolism
58. Reinhard M, Wehrle-Wieland E, Grabiak D, et al. Oscillatory cerebral of severe diffuse brain injury measured by PET. J Nuc Med 1996;37:
hemodynamics–the macro- vs. microvascular level. J Neurol Sci 1166–70.
2006;250(1-2):103–9. 83. Alavi A. Functional and anatomical studies of head injury.
59. Wong FY, Leung TS, Austin T, et al. Impaired autoregulation in preterm J Neuropsychiatry Clin Neurosci 1989;1(Suppl):S45–50.
infants identified by using spatially resolved spectroscopy. Pediatrics 84. Alavi A, Fazekas T, Alves W. PET in the evaluation of head injury.
2008;121(3):e604–11. J Cerebral Blood Flow Metab 1987;(Suppl):646.
60. Brady KM, Lee JK, Kibler KK, et al. Continuous time-domain analysis of 85. Furukawa M, Kashiwagi S, Matsunaga N, et al. Evaluation of cerebral
cerebrovascular autoregulation using near-infrared spectroscopy. Stroke perfusion parameters measured by perfusion CT in chronic cerebral
2007;38(10):2818–25. ischemia: comparison with xenon CT. J Comput Assist Tomogr 2002;26:
61. Lee JK, Yang ZJ, Wang B, et al. Noninvasive autoregulation monitoring in a 272–8.
Swine model of pediatric cardiac arrest. Anesth Analg 2012;114(4):825–36. 86. Soustiel J, Mahamid E, Goldsher D, et al. Perfusion CT for early assessment
Epub 2012, Feb 7. of traumatic cerebral contusions. Neuroradiology 2008;50:189–96.
62. Joshi B, Ono M, Brown C, et al. Predicting the limits of cerebral 87. Petrov YY, Petrova IY, Patrikeev IA, et al. Multiwavelength optoacoustic
autoregulation during cardiopulmonary bypass. Anesth Analg system for noninvasive monitoring of cerebral venous oxygenation: a pilot
2012;114(3):503–10. Epub 2011m Nov 21. clinical test in the internal jugular vein. Opt Lett 2006;31(12):1827–9.
63. Diedler J, Zweifel C, Budohoski KP, et al. The limitations of near-infrared 88. Jo J, Yang X. Functional photoacoustic imaging to observe regional brain
spectroscopy to assess cerebrovascular reactivity: the role of slow activation induced by cocaine hydrochloride. J Biomed Opt
frequency oscillations. Anesth Analg 2011;113(4):849–57. Epub 2011, 2011;16(9):090506.
Aug 4. 89. Zhang Y, Hong H, Cai W. Photoacoustic imaging. Cold Spring Harb Protoc
64. Murkin J Adams S, Schaefer B, et al. Monitoring cerebral oxygen saturation 2011;2011(9):pii.
significantly decreases stroke rate in CABG patients: a randomized blinded 90. Xu Z, Zhu Q, Wang LV. In vivo photoacoustic tomography of mouse
study. Presented at the Outcomes 2004: The Key West Meeting, Fla., May cerebral edema induced by cold injury. J Biomed Opt 2011;16(6):066020.
19–24, 2004. The Heart Surgery Forum 7(6), 2004[abstract]. 91. Ronne-Engstrom E, Winkler T. Continuous EEG monitoring in patients
65. Papadimos T, Marcos P. Cerebral oximetry and an unanticipated circulatory with traumatic brain injury reveals a high incidence of epileptiform
arrest. Anaesthesia 2004;59:309–10. activity. Acta Neurol Scand 2006;114:47–53.
66. Sakamoto T, Zurakowski D, Duebner LF, et al. Cerebral Ischemia caused by 92. Vespa PM, Nuwer MR, Nenov V. Increased incidence and impact of
obstructed SVC cannula is detected by NIRS. J Thorac Cardiovasc Surg nonconvulsive and convulsive seizures after traumatic brain injury as
2004;128:220–32. detected by continuous EEG monitoring. J Neurosurg 1999;91:750–60.
67. Dunham C, Ransom K, McAuley C, et al. Severe brain injury ICU 93. Young GB, Doig GS. Continuous EEG monitoring in comatose intensive
outcomes are associated with cranial-arterial pressure index and care patients: epileptiform activity in etiologically distinct groups.
noninvasive bispectral index and transcranial oxygen saturation: a Neurocrit Care 2005;2:5–10.
prospective, preliminary study. Crit Care 2006;10(6):1–9. 94. Hirsch LJ. Continuous EEG monitoring in the intensive care unit: an
68. Kirkpatrick J, Lam K, Al-Rawi P, et al. Defining thresholds for critical overview. J Clin Neurophysiology 2004;21:323–40.
ischemia by using near-infrared spectroscopy in adult brain. J Neurosurg 95. Carrera E, Claassen J, Oddo M, et al. Continuous electroencephalographic
1998;89(3):389–94. monitoring in critically ill patients with central nervous system infections.
69. Al-Rawi PG, Kirkpatrick PJ. Tissue oxygen index: thresholds for cerebral Arch Neurol 2008;65(12):1612–8.
ischemia using near-infrared spectroscopy. Stroke 2006;37(11):2720–5. 96. Oddo M, Carrera E, Claassen J, et al. Continuous electroencephalography
70. Zweifel C, Castellani G, Czosnyka M, et al. Continuous assessment of in the medical intensive care unit. Crit Care Med 2009;37(6):2051–6.
cerebral autoregulation with near-infrared spectroscopy in adults after 97. Rossetti AO, Urbano LA, Delodder F, et al. Prognostic value of continuous
subarachnoid hemorrhage. Stroke 2010;41(9):1963–8. Epub 2010, Jul 22. EEG monitoring during therapeutic hypothermia after cardiac arrest. Crit
71. Yokose N, Sakatani K, Murata Y, et al. Bedside monitoring of cerebral blood Care 2010;14(5):R173. Epub 2010, Sep 29.
oxygenation and hemodynamics after aneurysmal subarachnoid 98. Mani R, Schmitt SE, Mazer M, et al. The frequency and timing of
hemorrhage by quantitative time-resolved near-infrared spectroscopy. epileptiform activity on continuous electroencephalogram in comatose
World Neurosurg 2010;73(5):508–13. post-cardiac arrest syndrome patients treated with therapeutic
72. Wang S, Wu E, Tam C, et al. Characterization of white matter injury in a hypothermia. Resuscitation 2012;Feb 23. Epub ahead of print.
hypoxic ischemic neonatal rat model by diffusion tensor MRI. Stroke 99. Young GB. Continuous EEG monitoring in the ICU: challenges and
2008;38:2348–53. opportunities. Can J Neurol Sci 2009;36(Suppl 2):S89–91.
73. Cecil KM, Hills EC, Sandel ME, et al. Proton magnetic resonance 100. Young GB. Continuous EEG monitoring in the ICU. Acta Neurol Scand
spectroscopy for detection of axonal injury in the splenium of the corpus 2006;114:67–8.
callosum of brain-injured patients. J Neurosurg 1998;88(5):795–801 101. Vespa PM, Nenov V, Nuwer MR. Continuous EEG monitoring in the
74. Carpentier A, Galanaud D, Puybasset L, et al. Early morphologic and intensive care unit: early findings and clinical efficacy. J Clin
spectroscopic magnetic resonance in severe traumatic brain injuries can Neurophysiology 1999;16:1–13
detect “invisible brain stem damage” and predict “vegetative states.” 102. Freye E. Cerebral monitoring in the operating room and the intensive care
J Neurotrauma 2006;23(5):674–85. unit: an introductory for the clinician and a guide for the novice wanting
75. Shen Q, Ren H, Cheng H, et al. Functional perfusion and diffusion MRI of to open a window to the brain. J Clin Monit Comput 2005;19:77–168,
acute focal ischemic brain injury. J Cerebr Blood Flow Metab 2005.
2005;25:1265–79. 103. Traczewski W, Moskala M, Kruk D, et al. The role of computerized
76. Moritz C, Rowley H, Haughton V, et al. Functional MR Imaging assessment rheoencephalography in the assessment of normal pressure hydrocephalus.
of a non-responsive brain injured patient. Magn Reson Imaging J Neurotrauma 2005;22(7):836–43.
2001;19:1129–32. 104. Pérez JJ Guijarro E, Barcia JA. Influence of the scalp thickness on the
77. Driniger M, Aiyagari V, Zazulia A, et al. Effect of hyperoxia on cerebral intracranial contribution to rheoencephalography. Phys Med Bio
metabolic rate for oxygen measured using positron emission tomography 2004;49(18):4383–94.
in patients with acute severe head injury. J Neurosurg 2007;106:526–9.
VII
Chapter
47  

Auditory Signals for


Noninvasive Monitors
Richard P. Dutton and John M. Sewell

Introduction difficult to study given the logistic constraints involved in use


Noninvasive monitoring of cerebral blood flow (CBF) among of the imaging tools, such as positron emission tomography
other aspects of neurologic function will add important tools (PET) scanning, in trauma or stroke patients in the emergency
to the armamentarium of the clinician. A noninvasive monitor setting. Acoustics, however, offers an alternative approach to
could be applied to a much wider range of patients than exist- cerebrovascular monitoring that is less expensive and easier to
ing intracranial pressure (ICP) monitors, which require that apply early after injury.
the patient be in an intensive care unit (ICU). It is now feasible Acoustics, the science of sound wave transmission, is com-
to measure ICP with optic nerve ultrasound or with venous monly used in medicine to characterize the flow of blood and
ophthalmodynamometry.1-4 Investigators have also combined air through the body. The stethoscope, the very symbol of the
ICP monitors with other noninvasive devices or used near- physician, is one of the oldest and most useful of all medical
infrared spectroscopy to measure cerebrovascular pressure devices. A stethoscope is a simple machine that amplifies natu-
reactivity.5,6 The role of transcranial Doppler or color-coded rally produced sound waves to an amplitude at which they can
duplex sonography that also can be used to noninvasively be detected and discriminated by the human ear. In this way
assess ICP and cerebral perfusion pressure7 is described in physicians can examine blood flow through the heart and
Chapter 30. It also is possible that a noninvasive monitor great vessels and airflow through the lungs. Although normal,
could provide more physiologically relevant information laminar, flow is relatively inaudible, turbulence produced by
about intracranial physiology (e.g., blood flow rather than obstructions or narrowing is clearly heard, and characterized
blood pressure). This chapter describes one approach to this variously as bruits, rales, wheezing, or the like. This is the
challenge, based on detection and processing of sound waves principle that underlies the function of the BAM. Blood flow
generated by the heart and transmitted through arterial vessels in the cranium is not audible to an external stethoscope, even
to the brain. Discussion of this brain acoustic monitor with electronic amplification. The cranium does flex or vibrate
(BAM)8,9 begins with a description of the pathophysiology of in time to the arterial pulse, however, on a microscopic level.
brain injury and the physics of the cranium that make acoustic A highly sensitive contact sensor can be used to detect these
monitoring possible. Following is a discussion of how the vibrations and display them digitally. The BAM is thus an
BAM evolved to its present state, with evidence from a number electronic stethoscope that “listens” to the blood moving
of studies of its use in traumatic brain injury (TBI) and stroke. through the brain and detects alterations in flow. Studies at
Illustrations of the output of the BAM as well as interpretation the R Adams Cowley Shock Trauma Center of the University
steps used to determine the existence of CBF anomalies are of Maryland have found that the brain acoustic signal the
provided. A discussion of challenges and future directions of BAM measures is identical in morphology and component
this technology concludes the chapter. frequencies to the arterial pulse signal captured from any
other artery in the body in normal patients, but substantially
different following any trauma to the brain.
Pathophysiology of Detectable The observation of what the BAM measured provided the
principal impetus for the study of this technology and evolved
Brain Injury into assessing the signal itself and its characteristics as they
The brain has the most highly regulated vasculature in the differed in “normal” volunteers and patients with TBI. The
body. Neuronal function depends on aerobic metabolism, process involved signal analysis in both time and frequency
which in turn requires continuous delivery of oxygenated domains as is shown in the following examples, in which the
blood. Local autoregulatory processes act to maintain even brain-emitted signals are compared with those from a refer-
blood flow and pressure within the brain across a wide range ence artery, such as the radial or digital artery. In the latest
of systemic pressures. This autoregulation is exquisitely sensi- embodiment of the BAM system the metrics that indicate
tive to disruption by TBI and other pathologies,10 but very critical differences between reference and brain are coded into
456 © Copyright 2013 Elsevier Inc. All rights reserved.
Section VII—Computers, Engineering, and the Future 457

software that will identify anomalies in signals immediately. at the basic level of those with and without TBI. Following this
To broaden the basis of comparison, the BAM findings have proof of principle study, further studies of in-hospital, pre-
been compared with computed tomography (CT) scan results, hospital and stroke applications of passive acoustic monitor-
and with a group of indicators such as evidence of head insult, ing applied to cerebral and systemic blood flow have been
alteration of consciousness, Glasgow Coma Scale (GCS) score conducted.
of less than 14, postconcussive symptoms, and general presen-
tation. When these indicators are included, there is a high
incidence of abnormal BAM scores in patients with evidence The Acoustic Science
of TBI, compared with a very low incidence of abnormalities of Brain Monitoring
in normal volunteers. There is a limited literature about passive brain monitoring;
Although in some ways this approach is novel, it is actually the approach described here relies on the first principles
an extension of general auscultation with a visual display used of acoustics.11 Fundamentally, stethoscopes are air-coupled
instead of detection of audible sound differences. It is possible devices designed primarily to detect sounds generated in air-
to amplify a BAM signal into the audible range, but the human filled structures (the lungs). To better detect variations in
ear is unable to detect the subtle differences in amplitude and brain sound emissions, sensors were built to “match” the
frequency associated with TBI that become evident when a brain, which is close in density to water. The sensor systems
visual display is used to characterize the wave. Quantification relied on “hydrophone” technology,12 instead of diaphragm-
of digitized data also makes it possible to track the progress based air-coupled stethoscope technology. This approach was
of an individual patient over the course of treatment and used because the cranial sensors were not immersed in the
recovery. fluid of the brain, but rather “coupled” to the outside of the
The diagnosis and subsequent clinical management of any skull at a point anterior to the upper portions of the forehead
patient with an acute brain injury depends on clinicians above the sinus cavity. A similar approach was used to capture
who integrate information from the patient’s history, from systemic arterial signals directly over the radial or digital
physical examination, from imaging studies, from laboratory artery.
tests, and from changes that occur over time. No one device In the application of these principles to monitoring, the
can possibly provide an absolute black or white diagnosis in first finding was that the resulting system did not detect
every patient. Instead the BAM is seen as an adjunctive tech- audible signals from the brain, although in some cases the
nology that is simple, rapid, logistically compatible with the resolution of the system could be increased to detect anoma-
early care of trauma patients, and able to provide objective lous responses (refer to Moretti and Pizzi’s discussion of the
data on the quality of blood flow in the potentially injured basic technology as applied to active and passive stethoscopy
brain. in high-noise environments13). Rather, the critical and subtle
features of the signal that contained most of the information
had to rely on a high-resolution digital display.
The second finding was that the brain signal was out of
Evolution of the Acoustic phase by nearly 180 degrees with signals that came from a
Monitoring Approach and Current location over the temporal artery, and from the reference
sensor. This is a useful feature of this technology because it
Embodiment enables the monitoring staff to readily distinguish the brain
Some notes on the somewhat serendipitous evolution of this signal from one that might come from the superficial vascu-
approach will shed light on the mechanisms involved in brain lature over the forehead. This distinction is shown in Figure
acoustics. The study began with a visit from a group of U.S. 47.1. When there is uniform pressure from the arterial pulse
Army Rangers to acoustic engineers now working at Active wave entering the cranium, the side of the head follows the
Signal Technologies. Their objectives were to determine if the phase of the pulse, and the front of the head where the sensors
science of exploring the ocean with sound could be used to are placed is receding. This is the general rule, although there
explore the brain and to develop an ICP monitor the size of a are some exceptions based on anatomic variations among
cigarette pack—the space the rangers had for such equipment. patients.
The initial efforts following these discussions indicated that
when small exciters at the temple interrogated the brain and
the results were measured at the forehead (a system based on Initial Studies
active sonar), variations in ICP would cause a difference in The initial BAM evaluation took place in a study of 30 patients
acoustic response. The approach encountered the challenge of who were admitted to the ICU with severe TBI and invasive
calibrating this difference against absolute levels of ICP across ICP monitors. The results of this study were based on the
different patients due to individual differences. During the time-variant waveforms or “time-domain” signals collected.
course of this effort, a physician noted that the passive sensing The original results proved promising14 and the analysis tech-
itself generated waveforms on the data-gathering analyzer that niques moved to conventional time and frequency domain
resembled those of the arterial line displays on the patient signal analysis. Further studies were performed in stroke
monitors. Furthermore, there was a difference in the mor- patients15 and other environments including the admitting
phology of the signals between healthy patients (those admit- area of a trauma center to determine early response to brain
ted with decreased level of consciousness due to alcohol, but insult and the prehospital environment. In addition studies
with no brain injury) and those with documented TBI and have been performed to examine how BAM relates to conven-
elevated ICP. Unlike the ICP values themselves, these differ- tional TBI metrics, and how the BAM tracked TBI effects over
ences appeared to be constant over a range of patients, at least a period of time following the original incident.
458 Section VII—Computers, Engineering, and the Future

0.60 Plot 0

Plot 1

Plot 2

0.40

0.20

0.00

–0.20

–0.40
B 1.00 1.25 1.50 1.75 2.00 2.25 2.50 2.75 3.00 3.25 3.50 3.75 4.00

Fig. 47.1  A, Outward flexion of the cranium in response to the arterial pulse. The top of the figure represents the anterior skull, which “recedes”
from a forehead sensor with each pulse. P, Pulse. B, A representative brain acoustic monitor signal from a healthy patient. The red trace is from the
radial artery, the green is from the temporal artery, and the black from the brain. The true brain signal is out of phase with the more superficial
signals.

Current Status 3. Activates the acquisition system, which records the sounds
The BAM system continues to evolve (an example of a BAM from all three locations, two head and one reference
used between 2005 and 2008 is shown in Fig. 47.2). The radial 4. Checks the integrity of the signals
and finger sensor locations are shown, as well as the contact 5. Repeats if necessary until signals are representative
points of the head sensors. All sensors are held in place with 6. Saves the data file and removes sensors and bands
bands except for the finger sensor, which is attached in much
the same way as a pulse oximeter probe. To operate the system This process normally takes approximately 5 minutes and can
the researcher: be performed without disrupting indicated clinical care (e.g.,
the study may be performed after initial physical assessment,
1. Places the sensors on the patient while the patient is waiting for a CT scan). The BAM monitor-
2. Enters patient data in the acquiring laptop (or personal ing procedure also includes observing the autograding results
digital assistant [PDA]) of the system.
Section VII—Computers, Engineering, and the Future 459

C
Fig. 47.2  The brain acoustic monitor in place on a volunteer. A, Laptop to acquire data. The radial and finger sensors also are visible.
B, Radial sensor. C, Head sensor on forehead.

The system then performs a fast-Fourier transformation


BAM Measurements in Normal Volunteers (FFT) on these signals that determines the component fre-
and Traumatic Brain Injury Patients quencies in the raw signal in a band from less than 1 Hz to
The BAM measures both the time variant amplitude of pulse 80 Hz—the frequency responses displayed in the middle of
waveforms and the frequency response of those waveforms. each panel (see Fig. 47.3, B and E). In the absence of severe
Examples are shown in the following text to illustrate the pathology, the highest peak in the frequency response should
acoustic properties of normal subjects (controls) compared be the first one (the fundamental), which is determined by
with those with TBI, beginning with data from a normal and the arterial pulse rate. In a normal individual this should be
a mild TBI patient shown in Figure 47.3. around 72 beats per minute, or just greater than 1 Hz. The
In Figure 47.3 A and D show the “time domain” signal in frequency responses of the 3 signals from the normal volun-
volts, representing the amplitude and direction of skull deflec- teer (see Fig. 47.3, B) essentially overlap starting at the
tion in real time from two points and a reference. The green approximately 1 Hz fundamental and descending to the
trace is from a sensor on the right forehead, the black trace is approximately 90 dB noise floor of the instrument, whereas
from a sensor on the left forehead, and the red trace is from the TBI patient’s brain responses (see Fig. 47.3, E) diverge
the reference sensor over either the radial artery or the finger. from the arterial reference greater than 20 Hz.
In a non-TBI patient (see Fig. 47.3, A), these signals are analo- The current BAM system has a signal-processing algorithm
gous, and morphologically similar, to the conventional trace that subtracts the frequency response of the artery from the
produced by an arterial pressure catheter. brain after averaging, and highlights the divergence by
460 Section VII—Computers, Engineering, and the Future

0.20 LB (0)

ref (1)

RB (2)
0.15

0.10

0.05

0.00

–0.05

–0.10
A 1.00 1.25 1.50 1.75 2.00 2.25 2.50 2.75 3.00 3.25 3.50 3.75 4.00

–20.00

–40.00

–60.00

–80.00

–100.00

–110.00
B 0.00 10.00 20.00 30.00 40.00 50.00 60.00 70.00 80.00

Fig. 47.3  Representative normal and abnormal signals from the brain acoustic monitor (BAM). A, Normal raw or “time domain” signal from
the BAM. The red trace is the radial artery signal; the black and green traces are from the left and right sides of the brain. B, Normal “frequency
domain” signal from the BAM, showing the superposition of frequencies in the right and left brain and the radial artery.
Section VII—Computers, Engineering, and the Future 461

25.00

20.00

15.00

10.00

5.00

0.00

–5.00

–10.00

–15.00
5.00 30.00 55.00 80.00
C
0.60 LB (0)

ref (1)
0.50
RB (2)

0.40

0.30

0.20

0.10

0.00

–0.10

–0.20

–0.30
D 1.00 1.25 1.50 1.75 2.00 2.25 2.50 2.75 3.00 3.25 3.50 3.75 4.00

Fig. 47.3 cont’d  C, Normal “subtraction display” signal from the BAM, showing that brain and systemic (radial artery) signals have equivalent
frequency distribution. D, Abnormal raw or time domain signal from the BAM, in a patient with traumatic brain injury (TBI). The red trace is the radial
artery signal; the black and green traces are from the left and right sides of the brain.
Continued
462 Section VII—Computers, Engineering, and the Future

–20.00

–40.00

–60.00

–80.00

–100.00

–110.00
E 0.00 10.00 20.00 30.00 40.00 50.00 60.00 70.00 80.00

25.00

20.00

15.00

10.00

5.00

0.00

–5.00

–10.00

–15.00
5.00 25.00 50.00 80.00
F
Fig. 47.3 cont’d  E, Abnormal frequency domain signal from the BAM, in a patient with TBI, showing the deviation of frequency response between
the right and left brain signals (black and green traces) and the systemic signal (red). F, Abnormal subtraction display signal from the BAM, in a patient
with TBI, quantifying the deviation in frequency response between the brain and systemic signals shown in (E). LB, Left brain; RB, right brain.

calculating mean values of these relative frequency responses. the arterial pressure trace should be transmitted cleanly
The relative frequency responses of the left and right forehead throughout the entire arterial tree, and so the relative fre-
signals are shown in panels of Figure 47.3 from 10 to 80 Hz— quency response of the brain (see Fig. 47.3, C) should be a
the band determined on the basis of correlation of critical nearly flat line close to 0 dB. Indeed the normal relative fre-
frequencies within the arterial spectrum from both trauma quency response curves in Figure 47.3, C are scattered around
and stroke patients. In normal individuals the morphology of 0 dB and their mean values are close to 0 dB: left brain
Section VII—Computers, Engineering, and the Future 463

approximately minus 3 dB, right brain approximately minus criteria—both have amplitudes of at least 0.1 V in their brain
2 dB. Turbulence in the brain signal resulting from disrupted signals bilaterally. However, the ratios of the normal are
autoregulation will cause a discrepancy between the brain and greater than 2 (see Fig. 47.3, A) with both left and right brain
systemic arterial frequency response, yielding a relative fre- signals exhibiting good pulsatile shape, whereas the brain-
quency response (see Fig. 47.3, F) that rises to greater than injured patient has ratios less than 2 and the brain signals are
20 Hz. uneven—an unevenness not caused by artifact. The diver-
gences of the normal brain signals in Figure 47.3, C are 7 and
8 dB for the right and left brain, respectively, indicating BAM
Analysis Approach of negative for TBI, whereas the TBI signals have a maximum
Acoustic Response divergence of 20 dB (see Fig. 47.3, F, left brain), indicating
Of the three principal metrics used for numerical assessment BAM positive for TBI.
of the BAM signal, the first two (amplitude and ratio) are This analysis now has been automated, so that the results
measured directly from the time domain signal (the raw data) of the amplitude, ratio and divergence calculations appear on
and the third (divergence) is measured from the relative fre- a pop-up panel with a color coding of yellow or green that
quency response, which uses a systemic arterial control. These indicate whether the person either is out of the bounds of the
metrics are defined in the following text. metrics established for controls or, in the case of green, that
the person passes all criteria. In the event of a yellow, the
1. Amplitude: An acceptable and representative amplitude is
screen shows the sensor(s) and metric(s) that are out of
sometimes difficult to obtain for the brain because it is
bounds and the procedure is to adjust the sensor position(s),
based on skull excursion for each arterial pulse and
band tension, and so on to accommodate for individual dif-
is only about 0.001 inch per heartbeat. The solid state
ferences and retest. When the results appear to repeat (at least
sensors of the BAM can consistently measure a signal
three times) they are considered representative, whether
equal to about 0.1 V from positive to negative peak in a
yellow or green, and the data are saved. In general, it is difficult
healthy normal subject and so amplitude less than 0.1 V
to obtain a green reading when there is any anomaly in the
is considered a positive indication for TBI. The systemic
patient’s blood flow, but yellow readings can often be cor-
arterial reference signal is much stronger and conse-
rected through adjustments to sensor placement. An example
quently the system attenuates it in hardware or normal-
of these displays is shown in Figure 47.4, in which the yellow
izes it in software so that it is consistently comparable to
reading is checked and the left brain sensor adjusted to yield
the brain signal.
a green reading with all metrics being within acceptable limits.
2. Ratio: The second metric is an initial crude assessment of
The system now has evolved from initial time domain read-
signal morphology that calculates the peak excursion
ings early in the study, to the later time and frequency domain
above the mean line divided by the peak excursion below
data collection, comparison with a systemic arterial reference,
the mean. Positive excursion should be at least twice as
and analysis with the autograding now in a clearly readable
great as negative excursion and a ratio less than 2 is con-
format.
sidered a positive indication for TBI. This is similar to the
normal calculation of mean arterial pressure (MAP) in
which the MAP is equal to (diastolic plus 1/3 [systolic Example of the Acoustic Response
minus diastolic]). Interestingly, this is a telling indicator. of a Stroke Patient
There can be some slight deviations, but the ratio cannot
Passive acoustics may have place in the field of stroke. Acoustic
go below 2 positive to negative deflection to be safely con-
technology could be especially useful if it allowed for a rapid
sidered normal.
and early discrimination of ischemic versus hemorrhagic
3. Divergence: The final and most important metric is
stroke, and could thus indicate or facilitate rapid treatment
derived from the frequency analysis. In a normal subject
with clot-busting drugs, where efficacy is time dependent.17
noise levels as indicated by the frequency-response slope
Currently, the diagnosis of ischemic stroke is made by a com-
should drop off rapidly at higher frequencies, whereas in
bination of physical examination and CT or magnetic reso-
the presence of brain trauma there will be higher ampli-
nance imaging (MRI) scan, the imaging often being needed to
tudes persisting into higher frequencies in the band. The
exclude cerebral hemorrhage that would contraindicate
divergence is defined as the maximum deviation of the
thrombolytics. BAM technology has been used in an attempt
brain signal’s relative frequency response above 0 dB.
to discriminate between these two forms of stroke.16 The
When the divergence is 10 dB or more, the patient is con-
results are best appreciated using two data examples followed
sidered to have an anomalous brain noise compared to
by a summary of the study results about the applicability of
reference. In a mundane comparison, it is as if there is a
passive acoustics to stroke.
quiet or laminar flow in the arteries when there is no dis-
Figure 47.5 illustrates a patient with an ischemic stroke who
turbance, and an increase caused by some alteration of
was monitored with the BAM within 3 hours of symptom
arterial tone causing disturbed flow (severely disturbed
onset and who received tissue-plasminogen activator (rt-PA).
flow would be turbulent flow similar to that caused by a
The monitoring took place both before and after administra-
hose constriction).16
tion of the drug and the data examples are interesting for
All three metrics on both forehead sensors must register a several reasons. Only the divergence measurements (see Anal-
“pass” to indicate a normal BAM; control and TBI patient ysis Approach of Acoustic Response) are shown here (see Fig.
BAM outputs are shown in Figure 47.3. Here both the 47.5) as they were the most telling in the stroke studies. As can
normal and mild TBI patient pass the amplitude be seen in Figure 47.5, A, the divergence rises in the middle of
464 Section VII—Computers, Engineering, and the Future

A B
Fig. 47.4  A, Automated interpretive display of a brain acoustic monitor session. Yellow indicates a deviation from normal in the signal, which may
be due to pathology or to misapplication of a sensor. B, Automated interpretive display of a brain acoustic monitor session. Green indicates a normal
signal.

the band to a level greater than 10 dB and this in several fre-


quency points. After rt-PA (see Fig. 47.5, B), the divergence is Future Potential Uses of Brain
uniformly reduced and resembles a normal response. Similar Acoustic Monitor Technology
results were observed in other patients, although in some cases
in which the divergence was reduced, the patient did not The Challenge of Prehospital Triage
recover clinically. These results suggest that brain acoustics More effective triage tools can help prehospital emergency
can be used to identify ischemic insults. medicine. For example, any patient who may have suffered a
TBI must be transported to a trauma center, where a head CT
scan is obtained. However, greater than 90% of admission CT
Hemorrhagic Stroke scans to rule out brain trauma are normal. In addition, many
Patients with hemorrhagic stroke were examined using BAM. of the patients with normal CT scans may still have brain
The primary focus of the study was to examine whether isch- injury such as a concussion. The diagnosis of concussion is a
emic or hemorrhagic stroke could be differentiated before 3 clinical one, and there currently is not an objective physiologic
hours, that is, determine eligibility for rt-PA. There were two test; this has important implications in sports medicine and
categories of BAM measurements of hemorrhagic stroke: less in the military. A simple noninvasive test that could facilitate
than 3 hours of symptom onset or greater than 3 hours. triage of TBI patients or inform the diagnosis of mild TBI may
Studies within 3 hours had a normal divergence or a less noisy be feasible using acoustic technology.
signal than the ischemic patients. When examined more than
3 hours after symptom onset, patients often had higher than
normal divergence. This may represent “mass effect” contrib- Initial Issues and Challenges
uting to ischemia rather than active bleeding. The difference There are several challenges and questions when BAM is used
between the ischemic (noisy or high in divergence) and the as a “triage tool.” First, can passive acoustics detect an anomaly
hemorrhagic (quiet, low, or normal divergence) acoustic very early after injury, or does it take time for the disruption
response within 3 hours suggests that BAM may be used to of blood flow to become apparent? Second, can a BAM system
differentiate stroke subtype. Further studies are needed to be configured for prehospital, sports field, or far-forward mili-
validate this. tary application? This would require a modified sensor suite
Section VII—Computers, Engineering, and the Future 465

20.00

10.00

0.00

–10.00

–20.00
0.00 10.00 20.00 30.00 40.00 50.00 60.00 70.00 80.00
A

20.00

10.00

0.00

–10.00

–20.00
0.00 10.00 20.00 30.00 40.00 50.00 60.00 70.00 80.00 90.00 100.00
B
Fig. 47.5  A, Abnormal brain acoustic monitor (BAM) subtraction display from a patient with an evolving ischemic stroke. Note the deviation between
brain and systemic signals in the midfrequency range (arrow). B, Improved BAM signal in the same patient, following thrombolytic therapy.

and a small and rugged electronics package. Third, can the


BAM system operate well in a noisy outdoor environment, or Delay of Initial Acoustic Response?
in a medevac unit (ground or helicopter) where vibration and Tissue injury appears to cause almost instantaneous acousti-
noise may limit BAM signal? To address these issues 150 cally detectable changes in the brain. Brain acoustic monitor-
patients were evaluated during transport to a trauma center ing often occurred less than 15 minutes after brain injury, and
(prehospital) and compared with data from patients exam- in all the injury was sensitive to acoustic detection. However,
ined in-hospital. some patients without injury had an abnormal BAM response,
466 Section VII—Computers, Engineering, and the Future

suggesting that early BAM information could not be used decision is the triage decision that is made before loading
reliably for a triage decision, but only as a what-if guide. the patient, thus there is no concern for vibration, although
noise is present. The system has been tested to less than
110 dBA noise environment with no adverse effects.
Could a Device Be Made Small Enough  Longer-term medevac or ambulance transport: Whether
for Triage Purposes? military or civilian, the platform excitation levels tend to
The electronics and associated software for the BAM are overwhelm the BAM signal. When this occurs it is not
simple and in theory adaptable to small platforms, especially possible to obtain representative measurements. Noise
with recent advances in technology. In early studies the cancellation approaches have been explored, but when the
Compaq iPAQ was used in the configuration shown in Figure signal to noise is greater than 20 in amplitude as is often
47.6. This particular device did not prove rugged enough over the case in these instances, even noise cancellation tech-
a long period of time, but showed that there was no inherent niques are limited.
reason that the BAM could not be reduced to a size compatible  Longer-term fixed-wing transport: Here there are more
with prehospital triage or handheld use. constant “lines” of excitation and these can be canceled,
even in the band of the BAM. Projects funded by the U.S.
Air Force long-distance critical care aeromedical transpor-
Could the Brain Acoustic Monitor Work tation teams continue to explore this question. It appears,
in Noisy or “Hostile” Environments? however, that reliable BAM signals can be obtained after
BAM has been studied in a variety of ground ambulances, noise cancellation in fixed-wing transports.
helicopters, and fixed-wing transport aircraft. Better record-
ings are found when the environment was quiet and the Thus the BAM is versatile, but in very high amplitude vibra-
vehicle was not moving; however, the technologic challenges tion excitation environments that have few periodic compo-
presented do not seem insurmountable. They fall into three nents, it will not operate well. The information must be
categories: gathered before or after the transport.

Short-term medevac or ambulance transport: In these


Acoustic Technology for

instances, medics indicate that the most important


In-Hospital Monitoring
BAM technology is unlikely to replace CT or MRI once a
patient reaches the hospital, but it does have several potential
uses including evaluation of evolving pathophysiology over
time to predict problems, use as a continuous monitoring
system, or use during surgical procedures, in which neurologic
monitoring is indicated but invasive intracranial monitoring
is not justified.

The Brain Acoustic Monitor


as a Leading Indicator
BAM changes appear to have a predictive nature. Table 47.1
shows the sensitivity and specificity of BAM abnormalities
(measured at the time of admission in patients with mild TBI)
associated with the presence of three or more postconcussive
symptoms 7 to 10 days after the injury, and compares it with
other potential indicators: CT scan; GCS score less than 15;
evidence of head trauma; and documented loss of conscious-
Fig. 47.6  The brain acoustic monitor configured to run on a ness (LOC). BAM changes have high sensitivity (most patients
personal digital assistant, as deployed with the U.S. Army. This with symptoms had an abnormal BAM) although low speci-
can be used for prehospital monitoring. ficity (some patients with abnormal BAM did not develop

Table 47.1  Sensitivity and Specificity of BAM Abnormalities in Patients with Mild TBI and
Postconcussive Symptoms 7 to 10 Days after the Injury Compared with Other Potential
Indicators of TBI
BAM CT Scan GCS <15 Evidence of Head Trauma Documented LOC
Sensitivity 87% 6% 7% 38% 43%
Specificity 19% 99% 92% 60% 58%
Total subjects 284 286 288 288 282

BAM, Brain acoustic monitor; CT, computed tomography; GCS, Glasgow Coma Scale; LOC, level of consciousness.
Section VII—Computers, Engineering, and the Future 467

symptoms). The patients studied all had mild TBI and so CT patients with mild TBI most likely to have postconcussive
and GCS had low sensitivity. None of the indications used symptoms.19
alone appeared to predict outcome. Future studies need to
explore whether the use of BAM together with other param-
eters may create a more accurate predictive tool. Concussion
A concussion sub-study was performed in a prehospital trial
of the BAM. Data were analyzed post hoc. Concussion was
The Brain Acoustic Monitor considered if a patient with a closed head injury and a GCS
as a Continuous Monitor of 14 or 15 had documented LOC (brief). In those with no
Acoustic technology may allow for continuous brain monitor- documented LOC, a GCS of 14 or 15, and negative CT scan
ing of patients with the potential for deterioration of neuro- 48% had anomalies in their acoustic response similar to the
logic status, but who are in good enough condition that an overall negative CT rate population. Those with a documented
invasive intracranial monitor is not justified. Because BAM LOC were more likely to show acoustic anomalies particularly
abnormalities occur in real time relative to changes in cerebral if the GCS was 14 (60%).
perfusion, the BAM may be useful. Anecdotally observation
has shown rapid deterioration of BAM signals in patients with
ICP spikes associated with suctioning, pain, or hypoxia, and Blast
rapid improvement of signals in severe TBI patients treated by Whether blast injury causes a change in brain acoustics is
head elevation, sedation, or osmotherapy. However, in some more difficult to answer. Blast should generate physical distur-
cases in which the ICP changed, the BAM did not. The signifi- bances in the brain20; however, BAM assessment of these
cance of this finding is not clear because changes in intra­ injuries in the battlefield has not been possible, and there are
cranial pressure may not be the appropriate comparator few blast patients seen in civilian trauma centers. There is a
for changes in intracranial blood flow. Preliminary studies in great need in the military for TBI diagnostics. BAM signals
severe TBI patients suggest that the BAM may predict outcome improve relatively quickly after injury (blood flow returning
in some patients better than ICP,18 but these were patients who to normal even as neuronal and glial damage persists). Delayed
received treatment for ICP. A limitation to use of the BAM for BAM evaluation after soldiers have returned from war zones
continuous monitoring is that close coupling of the BAM requires study to determine whether there is any association
sensor to the forehead is required, because a certain amount between changes in BAM signals over time and neuropsycho-
of pressure is required to achieve good signals. This may be logical evaluation.
feasible for a 15-second “snapshot” BAM recording even when
repeated, but if maintained for longer periods likely will
produce skin breakdown or patient discomfort. Challenges and Next Steps
in System Development
The Brain Acoustic Monitor Multicenter Testing
in the Operating Room Acoustic brain monitoring is in its infancy, but its develop-
Surgical procedures (e.g., orthopedic) may be delayed in poly- ment can be driven in a number of different directions by both
trauma patients with moderate or severe TBI because the clinical need and technologic possibility. At the time of pub-
anesthetic and surgical stress may produce secondary brain lication, BAM has only been tested in a single center, the
injury. In addition, the ability to monitor neurologic function R Adams Cowley Shock Trauma Center, albeit on more than
and detect deterioration through clinical examination is lost 1000 patients and in one of the largest trauma centers in the
in an anesthetized patient. Short-term BAM monitoring world. The BAM requires multicenter testing before U.S. Food
during anesthesia may permit earlier surgery or procedures and Drug Administration (FDA) clearance.
such as angiography. The hypothesis requires further testing
but it appears that anesthesia per se—as seen in patients with
intoxication but not brain injury—may not cause abnormal Sensitivity and Specificity Verification
BAM readings. To date, the BAM has been tested on patients who have had a
head CT due to suspected TBI; BAM sensitivity is more than
90% with a specificity between 15% and 40%. It is conceivable
Special Cases: Blast and Concussion that BAM detects “physiologic” anomalies that are not so
Two clinical questions outside the ICU may lend themselves severe that an anatomic defect can be seen on CT, but this
to BAM use: (1) improvised explosive device (IED)–induced hypothesis still needs study. In 2011, Dutton et al published a
blast injury in which a triage decision to move the patient to prospective study on 369 patients with a mechanism of injury
the next echelon of care or return to duty is critical; and (2) consistent with TBI who were studied with CT, BAM, and
sports-related concussion that involves a decision whether the clinical evaluation.19 In addition, BAM data was obtained
athlete can return to competition. Both injuries can generate from 50 normal volunteers and 49 trauma control patients
changes in cerebral autoregulation, and thus blood flow thought not to have TBI. BAM discriminated between patients
anomalies that change the acoustic signature of the brain, with mild TBI versus without TBI. An abnormal BAM signal
similar to those noted in general TBI. Detailed studies are was suggestive of a new abnormality on CT. The sensitivity of
needed to confirm this. An exploration of BAM as a diagnostic BAM abnormality for head CT abnormality was 100%, with
tool in mild TBI was conducted in 2008 at the Shock Trauma a specificity of 30.14%. The results suggested that BAM screen-
Center, and suggested that BAM could help discriminate those ing may be a useful diagnostic adjunct in patients with mild
468 Section VII—Computers, Engineering, and the Future

TBI. In particular when there is loss of consciousness, an This may be why the “stroke” signatures are easier to dis-
abnormal BAM reading adds significance to LOC as a predic- tinguish than in trauma, e.g. subarachnoid hemorrhage
tor of a new abnormality on head CT. (SAH) in a trauma patient will look different and often
“noisier” than SAH in a stroke patient, possibly because in
TBI there is more than one cause of brain tissue
What the Brain Acoustic Monitor Measures disturbance.
The BAM measures only injuries that create a global acoustic 2. Recent pooled analysis of the ATLANTIS,21, 22 ECASS,23,24
effect on the brain and likely measures the effects of an inter- and NINDS25 rt-PA trials shows that earlier treatment is
ference in the normal perfusion patterns of the brain. Studies better with the greatest benefit in patients whose treatment
that can verify exactly what is measured should help define a started within 90 minutes of symptom onset.26 During this
role for passive brain acoustics in brain injury and the neuro- time period and in patients who also have a very high
critical care unit. This may require PET imaging or be feasible stroke scale (i.e., >20), there is a higher divergence by a
with CT perfusion scanning. factor of 10 dB more than the normal threshold for
“typical” ischemic stroke. Thus passive acoustics may help
determine a role for rt-PA.
Emergency Medical Services Applications 3. There also is a higher incidence of arterial “noise” in
Because existing BAM technology is handheld, it most likely patients prone to stroke, such as plaque in cerebral arteries.
will find a home in the emergency medical services (EMS) This may be an adjunct to an evaluation of stroke risk.
arena.
Conclusion
Will Future BAM Configurations Be Passive brain acoustics in its current embodiment in the BAM
Compatible with the EMS Environment? has revealed a new and potentially fruitful embodiment of a
The latest embodiment of the BAM for EMS is in a rugged very traditional tool: the stethoscope. Many of the develop-
PDA. An early prototype is shown in Figure 47.6. The Air ments that have contributed to this technology come from the
Force’s Critical Care Air Transport Team is funding the devel- arena of underwater acoustics and face the same issues that
opment of a plug-in type module, whereby the BAM can be many such transition technologies face. Studies to date,
used with other types of vital sign assessment modalities. This however, demonstrate enough potential to suggest that the
would allow a medic to quickly assess for intracranial pathol- BAM system is a low-risk method to examine how trauma and
ogy and then track a patient’s course over a period of extended stroke affect the brain.
transport.

References
Is the Brain Acoustic Monitor
1. Geeraerts T, Merceron S, Benhamou D, et al. Non-invasive assessment of
Rugged Enough? intracranial pressure using ocular sonography in neurocritical care patients.
This is a constant question of the EMS groups. In its original Intensive Care Med 2008;34(11):2062–7. Epub 2008 May 29.
2. Rajajee V, Vanaman M, Fletcher JJ, et al. Optic nerve ultrasound for the
EMS embodiment, the iPAQ, the system was prone to failure detection of raised intracranial pressure. Neurocrit Care 2011;Jul 19. Epub
due to connector, battery, and other issues. Newer plug-in ahead of print.
versions appear to survive the rigors of field use better than 3. Moretti R, Pizzi B. Ultrasonography of the optic nerve in neurocritically ill
early prototypes. patients. Acta Anaesthesiol Scand 2011;55(6):644–52. Epub 2011 Apr 4.
4. Querfurth HW, Lieberman P, Arms S, et al. Ophthalmodynamometry for
ICP prediction and pilot test on Mt. Everest. BMC Neurol 2010;10:106.
What About Cost? 5. Kasprowicz M, Schmidt E, Kim DJ, et al. Evaluation of the cerebrovascular
pressure reactivity index using non-invasive finapres arterial blood pressure.
The BAM system and the technology that underlies it are Physiol Meas 2010;31(9):1217–28. Epub 2010 Jul 28.
simple in nature (i.e., an electronic stethoscope with display 6. Zweifel C, Castellani G, Czosnyka M, et al. Noninvasive monitoring of
cerebrovascular reactivity with near infrared spectroscopy in head-injured
and simple interpretation algorithms). Cost is an important patients. J Neurotrauma 2010; 27:1951–8.
question to EMS groups, many of which are volunteer services 7. Brandi G, Bechir M, Sailer S, et al. Transcranial color-coded duplex
with limited budgets. Indications are that the BAM will be sonography allows to assess cerebral perfusion pressure noninvasively
relatively inexpensive, particularly when compared with other following severe traumatic brain injury. Acta Neurochir (Wien) 2010;152:
diagnostic tools for brain injury now in use. 965–72.
8. Apparatus and method for measurement of intracranial pressure with lower
frequencies of acoustic signal, US Patent Number 5,919,144, issued July 6,
1999.
Stroke Applications 9. Brain assessment monitor, US Patent Number 6,887,199, issued May3, 2005.
BAM technology may have applications in stroke diagnosis. 10. Goodman JC. Pathophysiology. In: Marion D, editor. Traumatic brain injury.
New York: Thieme; 1999. p. 147.
Several observations support this concept although more 11. Kinsler LE, Frey AR. Fundamentals of acoustics. 2nd ed. New York: John
study is required to verify this. Wiley & Sons; 1962. p. 108. ff.
12. Burdic WS. Underwater acoustic system analysis. Englewood Cliffs, NJ:
1. Stroke appears to offer a “purer” case for passive acoustics, Prentice-Hall; 1984. p. 64.ff.
unlike trauma, which can involve diffuse, focal, or a com- 13. Houtsma AJ, Sewell JM. A dual mode noise-immune stethoscopes for use in
noisy vehicles. Army Sciences Conference. Orlando: Sept. 2006.
bination of injuries. Stroke generally is of two types— 14. Dutton R, Sewell J, Aarabi B, et al. preliminary trial of a noninvasive brain
ischemic or hemorrhagic, and at least within 3 hours after acoustic monitor in trauma patients with severe closed head injury.
the onset of symptoms there is a different BAM signature. J Trauma 2002;53(5):857–63.
Section VII—Computers, Engineering, and the Future 469

15. LaMonte MP, Sewell JM. A non-invasive portable acoustic diagnostic system 22. Clark WM, Wissman S, Albers GW, et al. The rt-PA (alteplase) 0- to 6-hour
to differentiate ischemic from hemorrhagic stroke. J Neuroimaging 2005;15: acute stroke trial. Part A (A0267g): results of a double-blind, placebo-
57–63. controlled multicenter study. Stroke 2000;31(4):811-6.
16. Hademenenos GJ, Massoud TF. The physics of cardiovascular disease. New 23. Hacke W, Kaste M, Fieschi C, et al. For the ECASS Study Group. Intravenous
York: Springer; 1998. p. 104. thrombolysis with recombinant tissue plasminogen activator for acute
17. LaMonte MP, Medlin T, Gaasch W. Intravenous thrombolytic therapy for hemispheric stroke: the European Cooperative Acute Stroke Study (ECASS).
stroke: a review of recent studies and controversies. Ann Emerg Med 1999; JAMA 1995;274:1017–25.
34(2):244–55. 24. Hacke W, Kaste M, Fieschi C, et al. Randomized double-blind, placebo-
18. Dutton RP, Sewell J, Aarabi B, et al. preliminary trial of a noninvasive brain controlled trial of thrombolytic therapy with intravenous alteplase in acute
acoustic monitor in trauma patients with severe closed head injury. ischaemic stroke (ECASS II). Lancet 1998;352;1245–51.
J Trauma 2002;53:857–63. 25. The National Institute of Neurological Disorders and Stroke (NINDS) rt-PA
19. Dutton RP, Prior K, Cohen R, et al. Diagnosing mild traumatic brain injury: Stroke Study Group. Tissue plasminogen activator for acute ischemic stroke.
where are we now? J Trauma 2011;70(3):554–9. N Engl J Med 1995;333(24): 1581–7.
20. Bayly P. MRI measurement of acceleration-induced brain deformation, 26. Association of outcome with early stroke treatment: pooled analysis of
neurological effects of blast injury conference. Bethesda, MD, April 2008. ATLANTIS, ECASS, and NINDS rt-PA stroke trials. Lancet 2004;768–74.
21. Clark WM, Albers GW, Madden KP, et al. Recombinant tissue-type
plasminogen activator (alteplase) for ischemic stroke 3 to 5 hours after
symptom onset: the ATLANTIS study—a randomized controlled trial. JAMA
1999;282:2019–26.
VII
Chapter
48  

Past, Present, and Future


Developments of Intracranial
Monitoring
David M. Benglis, Jr., Brett Trimble, and M. Ross Bullock

“Monitors themselves do not save lives, but the insightful are identified, and measures to correct these abnormalities
interpretation of the information derived from them can.” can be instituted. Ideally these methods should also include
—Saul and Ducker understanding the mechanisms associated with brain damage
at the cellular level. This chapter briefly discusses the current
neuromonitoring devices and techniques available, as well as
Introduction new technologies projected to be available in the upcoming
Traumatic brain injury (TBI) in the United States affects more years. In addition it covers the role of industry in the
than 1.5 million individuals per year costing more than $60 advancement of new ideas and nanotechnology, and envis-
billion in both direct medical and indirect costs such as loss ages the intensive care unit (ICU) of the future. Key issues
of productivity.1-4 Following the initial insult, a series of relevant to modern ICUs are whether improved brain moni-
complex pathophysiologic cascades are set in place, which toring techniques can ultimately affect individuals’ quality of
synergistically interact to cause delayed, secondary brain life and outcome.
damage, over hours, weeks, and even up to a year after the TBI.
Physicians have the capability to monitor and reverse these
secondary damage mechanisms. A host of modalities, includ- The Basics: Glasgow Coma Scale
ing cerebral blood flow (CBF), brain oxygen, intracranial pres-
sure (ICP), biomarkers, and electrical cortical activity, among
and Physical Examination
others, are available in state-of-the-art neurocritical care The brain is 2% of total body mass yet receives 15% to 20%
units (NCCU). of the cardiac output.8 Because this tissue is extremely sensitive
Despite this, no single brain monitoring method has yet to hypoxia and ischemia, maneuvers to prevent critical reduc-
emerged to supersede serial neurologic observations by a well- tions in arterial blood pressure (invasive arterial monitor or
trained neuroscience critical care nurse. Many promising noninvasive cuff), peripheral arterial oxygen content (oxygen
brain-monitoring technologies have been evaluated, but after saturation monitor), intrinsic cardiac function (electrocardio-
initial enthusiastic use, have not continued to be used in gram), and pulmonary function (end-tidal CO2 or arterial
patient care. Examples include jugular bulb oximetry and the blood gas analysis) output assessment are important in critical
“Neurotrend” system. Each of these represents ingenious tech- care. A standardized neurologic exam (Glasgow Coma Scale
nology able to generate meaningful data, yet they have failed [GCS]) introduced by Teasdale and Jennett in 1974 can be
to affect patient care in the long term. Nevertheless, technolo- used for a basic assessment of brain function. It has prevailed
gies to monitor the brain continue to evolve and be developed, as a simple yet critical bedside neuromonitoring technique
and much of what is done in the NCCU is to recognize changes because of its sensitivity to detect meaningful changes and its
in brain physiology and function and how the brain and the ability to be reproduced with a little variation.9
rest of the body interact. Ideally this is done through multi- When possible, new technologies should be compared to
modality monitoring with a bioinformatics support platform, established gold standard techniques for specific measure-
two concepts that will drive the future of monitoring in ment of changes in the injured brain. For example, a decrease
neurocritical care.5-7 in brain oxygen partial pressure reported by an intraparenchy-
mal probe, should be confirmed (e.g., by xenon-enhanced
computed tomography [Xe-CT] CBF) that those changes are
Aims of This Chapter actually accurate and relevant. With the development of por-
In an ideal system, information derived from monitoring table computed tomography (CT) scanners that also allow
should be specific, online or real-time, and filtered so that the evaluation of perfusion and flow, this may become a com-
pertinent information is presented, changes from the norm monly accessible modality in many NCCUs.10
470 © Copyright 2013 Elsevier Inc. All rights reserved.
Section VII—Computers, Engineering, and the Future 471

or the ventricle, and may have greater accuracy over time


(e.g., microstrain catheter), but cannot be recalibrated once
placed.11-13 Wet/dry systems are now available, such as the
Spiegelberg (Hamburg, Germany) transducer, which keep the
compartments to measure ICP and drain CSF separate. These
devices also have automatic zeroing functions. In conclusion,
ICP monitoring currently plays a major role in monitoring
brain status in the ICU. It should be considered in all patients
where ICP is a question until alternative methods prove to be
better. Developments will see an evolution of noninvasive
devices, although at present none are able to replace invasive
ICP monitors.14-17 In addition, it is well recognized that ICP is
A more than a number and that use of a single threshold at
which to treat may be an oversimplification. Hence further
D mm Hg evolution of ICP monitoring will include use of trend analysis
–100 and indices of autoregulation, pressure reactivity, and compli-
ance to name a few.18-22
–80

–60 SjvO2, PbtO2, CBF, NIRS


–40 Because brain ischemia and hypoxia are common following
TBI, the maintenance of tissue oxygenation above critical
–20
values is essential in modern NCCUs.23 This reduction in
–0 blood flow and oxygen delivery can occur within minutes after
TBI; this stresses the importance of early intervention and
120 105 90 75 60 45 30 15 0 initiation of monitoring.24 A mismatch or uncoupling between
Minutes substrate demand and substrate supply also may be present
because of the loss of cerebral autoregulation, and these
B D = Tapping of fluid (8 mL)
changes can be present on a regional or more global scale.25
Fig. 48.1  A, Ventricular monitoring device used by Nils Lundberg These tissues are thus at risk for permanent damage from
(Swedish neurosurgeon).163 B, Ventricular pressure tracing before and critical reductions in the continuous delivery of glucose,
after drainage of cerebrospinal fluid. (Courtesy Wilkins RH, Wilkins G, et al. oxygen, and other essential substrates.19 Aerobic metabolism
Neurosurgical classics II. American Association of Neurological Surgeons, Park becomes superseded by anaerobic mechanisms, ionic homeo-
Ridge, IL, 2000.) stasis is compromised, and calcium influx into cells along
with potassium efflux causes widespread neuronal dysfunc-
tion and edema.
Monitors of Brain Substrate
Delivery
SjvO2
ICP Monitoring Jugular venous oxygen saturation (SjvO2) monitoring was
Although ICP monitoring has been in use for greater than 50 implemented in the early 1990s as a promising method to
years (Fig. 48.1, A and B), its utility is still questioned by detect post-traumatic changes in global cerebral oxygenation
some, who allege its use has not been demonstrated to save (see Chapter 32).26 The average SjvO2 in normal subjects is
lives or improve outcome, in controlled trials. Such a trial 62%; a decrease less than 50% is defined as critical ischemia
sponsored by the National Institutes of Health (NIH) in Latin and is associated with a worse outcome.27 Mainstream use of
America to address this criticism was completed in late 2012. this monitoring device has been limited, however, because of
Nevertheless, these issues have been extensively reviewed in problems encountered with catheter migration or poor posi-
the latest revision of the severe TBI management guidelines, tioning, calibration errors (e.g., light absorbance), limited
and level II evidence exists to supports ICP treatment in indi- “time of good data quality,” and the inability to detect all
viduals with sustained ICPs greater than 20 mm Hg.11 With causes of decreased brain oxygenation.26,28-30 Thus in some
ICP and mean arterial blood pressure (MAP) measurements, studies, more than 50% of measurements were erroneous,
cerebral perfusion pressure (CPP) can then be derived. The leading to far less use of the method in most centers. SjvO2
CPP value necessary to prevent ischemia varies according to also requires intensive supervision by specialized personnel.31
the individual, but in general is between 50 and 70 mm Future use of retrograde jugular catheters may also include
Hg.11,12 Requirements for ICP monitors should be that the the sampling of biomarkers, cytokines, or other factors such
device is low cost, accurate, and reliable. Fluid-coupled exter- as endothelin that may influence CBF.32,33
nal strain gauge intraventricular catheters traditionally cost
less, but may experience significant measurement drifts over
time and have a greater incidence of malfunction. Transducer- PbtO2
tipped devices (micro strain gauge [Codman, Raynham, MA] A more direct approach to measure focal fluctuations in brain
versus fiber-optic (Integra NeuroSciences, Plainsboro, NJ) oxygen tension (PbtO2) can be accomplished with Licox (GMS-
cost slightly more, may be placed in either brain parenchyma Integra-Kiel, Mielkendorf, Germany) or Neurotrend (Codman,
472 Section VII—Computers, Engineering, and the Future

Raynham, MA) sensors (see Chapter 35).34 Optical sensors clinical practice, small companies may be unable to sustain
contained within the Neurotrend probe detect dye color production costs for sophisticated microsensor systems
changes resulting from differences in gas concentrations and without sufficient volumes of sales. The intrinsic conservatism
pH of the surrounding tissues. One advantage of this particu- of clinicians and hospital purchasing systems compound the
lar device is the capability to measure oxygen concentrations problem. The award of Small Business Innovation Research
and other modalities including carbon dioxide (PbtCO2), pH, (SBIR) grants may serve as a mechanism to help defray
and temperature (T).34 The Licox sensor consists of a minia- production costs, until uptake of new technologies achieves
turized Clark electrode with a polyethylene casing enclosing a critical levels for commercial viability. In late 2012, an NIH-
solution of buffered potassium phosphates. Oxygen concen- sponsored phase II randomized clinical trial to compare
tration is correlated with the electrical current created across guided management of severe TBI using ICP alone to ICP and
a silver anode and gold-coated silver cathode.8 Sampling area PbtO2 (based on a Licox monitor) was halfway through patient
is approximately 14 mm2, which is seven times the sampling recruitment. This trial hopefully will help clarify the utility
area of the Neurotrend probe.34 and cost effectiveness of PbtO2 monitoring.
Critical thresholds of oxygen concentration have been
determined for this technique. The oxygen value picked up by
the sensor is a function of diffusion of oxygen from the eryth- Cerebral Blood Flow (LDF, TDF Probes)
rocyte or plasma to the extracellular space and it integrates all Laser Doppler flowmetry (LDF) consists of an implanted sub-
of the venous and arterial vessel oxygen tensions in the vicin- cortical fiber-optic laser probe (0.5-1 mm in diameter) that
ity.23 Following placement, an increase of inspiratory oxygen measures the shift of reflected laser light induced by move-
fraction (FiO2) to 100% can confirm the sensitivity of the ment of red blood cells (RBCs). Data are presented in real
device.35 With Neurotrend, sustained values of PbtO2 that are time, and variations in blood flow are monitored (see Chapter
less than 20 to 25 mm Hg are associated with poor outcome.10 31). The information is not quantitative, however, and repre-
Using Licox technology, PbtO2 concentrations between 25 and sents only relative changes. Absolute values may be obtained
30 mm Hg are found in TBI patients with CPP and ICP in the when LDF is calibrated against other quantitative measures of
normal range, but values less than 10 to 15 mm Hg are con- CBF such as xenon-enhanced CT (see Chapter 27).12,46 Thermal
sidered a sign of tissue at risk, and values less than 5 to 10 mm dilution flow (TDF) methods (e.g., Bowman or Carter probes)
Hg indicate ongoing critical values, or, rarely, sensor malfunc- are based on conductance of small amounts of heat between
tion. Furthermore, brain hypoxia (<10 mm Hg) appears to be two small gold electrodes, one equipped with a sensitive ther-
an independent factor associated with poor outcome.36,37 Mea- mocouple sensor (see Chapter 31). CBF is proportional to heat
sures to increase PbtO2 then need to be instituted.11,30,38,39 When conductance, and the values are thus absolute, but the probes
PO2 is maintained at normal values, it is associated with better are usually configured for placement in deep white matter
outcomes in TBI patients.40 Persistent decreases in PbtO2 also rather than gray matter. CBF changes therefore are less (e.g.,
correlate with increases in brain lactate, and can result from normal CBF in white matter is ∼22 mL/100 g/min versus
with sustained ICP elevations, respiratory distress, and hypo- 80 mL/100 g/min in gray cortex). These CBF sensors usually
tension. In these studies, however, it was not possible to cor- are placed through a “bolt” system and can be placed at cra-
relate brain PaCO2 and pH with cerebral blood flow (CBF) niotomy (e.g., for aneurysm clipping). Clinical acceptance of
and PbtO2.10,25 For example, van den Brink et al. in a study of both these monitors of CBF is limited in part because relative
101 patients with severe nonpenetrating TBI (GCS ≤8), the CBF values are reported, because of questionable reliability
magnitude and length of time of low PbtO2, measured by Licox and validity, and because of questions on how to use the
were an independent factor associated with an unfavorable information.47 In particular it is difficult to make conclusions
outcome and death. In addition, they observed that the devices about CBF unless the state of metabolism is known. Neverthe-
demonstrated little zero drift and values were reliable over the less, use of these devices can supplement other monitors and
time course of monitoring.41 provide information about autoregulation.48,49 Noninvasive
It remains unclear what is the optimal brain territory in methods to assess regional and global blood flow and oxygen-
which to place the probe.42 When placed in contused brain, ation are under deve­lopment and may one day replace current
increasing the FiO2 does not result in increases in PbtO2 to the LDF and TDF monitors.
same degree it does in noncontused brain, thus suggesting
that CBF is too low to provide adequate substrate delivery
in these contused areas.43 Therefore to recognize critical Noninvasive Monitoring of Brain
decreases of PbtO2 some groups recommend placement in Oximetry: NIRS
nonlesioned white matter in the frontal lobe for diffuse cere- Near-infrared spectroscopy (NIRS) technology was initially
bral injury or on the affected side for a unilateral injury.34,43 pioneered by Jobsis and Chance, and is an emerging method
Because of this heterogeneity of measured values between dif- to monitor cerebral oxygenation, blood flow, blood volume,
ferent areas of brain and the potential influence of local cerebrovascular reactivity, and perhaps even edema in both
factors (e.g., microhemorrhages) that may contribute to the adults and children (see Chapter 33).31,50-56 The basis of NIRS
measurement, CT analysis of probe position is necessary.41,44,45 theory rests on the unique properties of light in the near
Most authors agree that PbtO2 monitoring should not be used infrared range to be scattered and absorbed when passing
as a “stand-alone” monitoring technique and is best com- through biologic tissues by factors found within those tissues
bined with ICP monitoring.23 (e.g., chromophores such as water, lipids, cytochrome c
The Neurotrend sensors are no longer available because of oxidase, and various hemoglobin compounds). In basic terms
commercial reasons. This emphasizes that although neuro- brain NIRS involves transmission of light from an emitting
monitoring technologies may be accurate and useful in source to a sensor placed nearby. Although transmission NIRS
Section VII—Computers, Engineering, and the Future 473

works well in neonates because of their semitransparent skull cannot be identified and subtracted. Second, signals from
and scalp, NIRS monitoring in adults is degraded by the blood in tissues overlying the brain such as the scalp cannot
increase in skull and scalp tissue density.57,58 To overcome this be reliably identified and subtracted.62 Third, hair on the scalp
limitation, reflectance mode NIRS is used, where an emitter also complicates measurements, and current systems only can
and detector are separated by a specific distance on the scalp be used in areas of the head not covered by hair such as the
with the premise that a fixed amount of transmitted, reflected, forehead. A few small clinical series, however, describe the
and scattered light follows an elliptical path whose depth of potential application in both the operating room and in
penetration is proportional to the distance of separation the ICU. For example, Brawanski et al. described a correlation
between the emitter and the detector.59 between intraparenchymal oxygen probes and NIRS data in
nine TBI patients.63 Kirkpatrick et al. found that NIRS regis-
tered changes in cerebral oxygenation that recovered once
Devices Based on Absorption blood flow was restored during vessel cross-clamping in
carotid endarterectomy.64 In 12 TBI patients the same investi-
Measurements gators reported an increased frequency of correlative changes
The absorption spectra of deoxyhemoglobin (HHb) and oxy- (e.g., decrease in peripheral saturation, intracranial hyperten-
hemoglobin (HbO2) in the infrared spectra between 700 and sion, hyperemia) in NIRS versus SjvO2.31
900 nm has long been used to measure blood oxygen satura- Several of the drawbacks of the first generation of cerebral
tion.54,59 The attenuation of optical signals at convenient wave- oximeters have been addressed using techniques and equip-
lengths in the near infrared range around the isosbestic point ment borrowed from radar and communications applica-
for hemoglobin (800 nm), are measured and used to deter- tions.59,65-68 This includes time of flight or time delay methods
mine the molar concentrations of HHb and HbO2 and oxygen and multifrequency, multiwavelength phase modulation
saturation in blood.53,60 This is possible because water and methods similar to the well-known time division multiplex
most other chromophores are functionally transparent or do (TDM) and wavelength division multiplex (WDM) methods
not dramatically vary in absorption characteristics within the used extensively in telecommunications applications. In short,
bandwidth interrogated61 (Fig. 48.2). a radiofrequency carrier wave is applied to the transmitted
The INVOS Cerebral Oximeter (Somanetics Inc., Troy, MI) optical signal. Phase shifts in the returned optical signals
uses a similar technique to estimate blood oxygen saturation coupled with the attenuations can be used to deconvolute scat-
in the brain. Optical transceiver pairs are placed on the left tering and absorption independently. In addition, the phase
and right forehead over the prefrontal cortex. Typically, the shift information can be used to spatially resolve the measure-
attenuation of light signals at two wavelengths is used to esti- ments and construct an image.60 In the future it is conceivable
mate regional blood oxygen saturation. The separation that this technique could be applied to the whole brain and
between emitter-detector pairs is optimized to give as much provide a continuous image of brain oxygen saturation or a
signal as possible from tissues that underlie the skull. related parameter. However, no commercial product based on
These systems are used often during cardiac surgery to these techniques is available despite the significant body of
detect cerebral blood desaturation episodes and show promise research (Fig. 48.3).
in cerebral hematoma detection. However, there is limited
enthusiasm for their use in NCCU monitoring, in part because
of several limitations. First, attenuation due to scattering Devices Based on Light
0.30
Scattering Measurements
The scattering of light in tissues is caused by refractive index
mismatch between adjoining materials and structures within
0.25 the tissue. In the near infrared bandwidth of 700 to 900 nm,
Water
Absorption factor (cm–1)

0.20

0.15
MRI
B
0.10 HHb

0.05
OxyHb

0
4

A
cm

600 700 800 900 1000 1100


cm

C
9

Wavelength (nm)
Fig. 48.3  Functional magnetic resonance imaging (fMRI) (A) versus near
Fig. 48.2  Absorption spectra of hemoglobin, deoxyhemoglobin (HHb), infrared phased array device (B, C) images of hemoglobin increase. (From
and water. OxyHb, Oxyhemoglobin. (Reused with permission from Chance B, Chance B, Nioka S, Zhao Z. A wearable brain imager. IEEE Eng Med Biol Mag
Cope M, Gratton E, et al. Rev Sci Instrum 1998;69:3457–81.) 2007;26(4):30–7.)
474 Section VII—Computers, Engineering, and the Future

scattering is the dominant effect thought to outstrip absorp- TBI, subarachnoid hemorrhage (SAH), and stroke among
tion by two orders of magnitude.59 In vivo scattering mea­ other disorders treated in the NCCU are associated with a
surements show promise in the ability of NIRS to detect cascade of events: excitatory amino acid (EAA) release, the
experimental brain edema in rodents.69,70 In these experiments opening of various ion channels that results in calcium influx
the optical density of brain tissues was directly interrogated and potassium efflux, failure of the sodium-potassium ade-
using NIRS signals and compared with the specific gravity of nosine triphosphatase (Na+/K+-ATPase) transport, mitochon-
tissue samples harvested during the experiment and to ICP drial derangement, cytoskeletal breakdown, and a shift from
measurements. Water intoxication by direct parenchymal aerobic to anaerobic metabolism that evolves over time82 (see
injection was used to induce brain edema and intracranial Fig. 48.1). These changes have been evaluated in humans using
hypertension. The NIRS-measured optical density correlated microdialysis and show that adverse clinical events (e.g., ele-
with the specific gravity of the tissue and decreased with tissue vated ICP, hypotension, or hypoxia) are associated with dialy-
swelling.69 In follow-up experiments, transgenic aquaporin-4 sate concentration increases (e.g., lactate, potassium, EAAs,
knockout mice and wild-type control animals were compared or decreases such as glucose.75,83-86 There is a suggestion that
under the same conditions. The knockout mice had less the changes—elevated lactate-to-pyruvate (LP) ratios and
decrease in optical density, suggesting that water uptake by glycerol—may precede an increase in ICP.87 In addition, a well-
cells may be responsible for the increased scattering and the described and independent association exists between extra-
subsequent decline in optical density of the tissue.71 cellular metabolic markers and outcome after TBI.88 However,
Further development of this device holds the promise of a it remains unclear whether treatment guided by microdialysis
continuous brain edema detector. If optical density is not findings can alter outcome, although its use can be used to
affected by blood volume or indicates an increase in optical identify episodes of cerebral compromise not detected by con-
density, the technique may be able to differentiate between ventional techniques (ICP and CPP)89 and to better stratify or
edema from cellular swelling or from vascular engorgement. target therapies based on metabolic disturbances.90
The potential shortcomings associated with this technique Microdialysis in the NCCU is largely a research tool, and
include: (1) the effect of solutes such as mannitol and (2) that more research and technology development are required
the measurement by its nature must be made in situ. Produc- before it is adapted into routine clinical use.91 Development
tion of a “piggyback” device added to current intraparenchy- likely will include the ability to obtain both regional and non-
mal monitors may soon be feasible. In addition, prototype regional information; the construction of analyzers (e.g.,
devices based on NIRS technology have been used for intra- direct electrochemical detectors) that can give real-time read-
cranial hematoma detection, and although at present these outs of an analyte such as lactate or glutamate; and evolution
devices cannot replace CT they can in the future play an of dialysis membranes. However, the “best” analyte to be mea-
important role in the NCCU if the need for patient transport sured remains unclear.
is avoided.72

Microdialysis Functional Monitoring: Glasgow


Coma Scale, Electroencephalogram,
Microdialysis (MD) was introduced to the clinical environ-
ment in the 1980s.73 It consists of a fine probe (0.62 mm in Electrocorticogram, Evoked
diameter) placed directly into the brain and may be combined Potentials, Pupillometer
with ICP and PbtO2 probes.74 The probe is perfused with a
sterile “mock extracellular fluid [ECF]” solution at a slow rate Glasgow Coma Scale and
that allows continuous sampling of the parenchymal extracel- Neurologic Assessment
lular environment with the transfer of substances less than Outcome following acute brain injury including TBI, SAH,
20 kDa across a dialysis membrane.8 Assays are based on and stroke is associated with the initial neurologic condition.
high-performance liquid chromatography (HPLC) techniques Hence assessment of consciousness, pupils, and limb move-
in small aliquots of dialysate fluid (60 µL). This allows iden- ments is a foundation of neuromonitoring (see Chapter 10).
tification and measurement of various molecules including This neurologic assessment is a continual process and can
glucose, lactate, potassium, pyruvate, nitric oxide, glutamate, provide reliable and dynamic information about the injured
and glycerol.75-77 The use of MD probes with much higher brain. The examination, however, may be limited following
molecular weight cutoff (e.g., 70 or 100 kDa) allows protein intubation, sedation, and administration of chemical paraly-
fragments, and small peptides, such as cytokines (e.g., inter- sis, and so standardized and objective scales are valuable. Since
leukin [IL]-1, IL-6, trophic factors, and amyloid precursor Teasdale and Jennett’s initial description of the GCS in 1974,
protein fragments) to be measured.78 Microdialysis has several it has evolved into the gold standard neuromonitoring tech-
limitations. First, measurements are representative of relative nique because it is simple, reproducible, objective, and has
concentrations of the extracellular molecules and do not rep- limited intra- and interobserver variability. Other monitors
resent actual concentrations. This can limit data comparison therefore should be used with the GCS.8
between different machines and centers.79 Second, the infor-
mation is not truly online, although newer devices, such as
the CMA600 (CMA Microdialysis, Stockholm, Sweden) have Brainstem Auditory Evoked Potentials
shortened times of bedside substrate analysis to approxi- Brainstem auditory evoked potentials (BAEPs) assess the
mately 1 to 2 hours.79,80 Third, result specificity may be affected integrity of the auditory efferent pathways. Normally there are
by cell edema, and changes in the ECF space around the five waveforms: waves I through V represent signals generated
catheter.12,29,77,81 at the vestibulocochlear nerve, cochlear nuclei, superior
Section VII—Computers, Engineering, and the Future 475

olivary complex, lateral lemniscus, and inferior colliculi, long-term effects in patients and what they are is still to be
respectively. BAEPs are useful in posterior fossa surgery but fully elucidated.104,109-111
have a limited role in assessment of patients in the NCCU, Animal studies dating back 60 years show that several
although they may help confirm the diagnosis of brain death mechanisms including potassium and glutamate in the ECF
(e.g., absence of all waveforms or presence of wave I).8 On the can influence CSD. However, the presence of CSD in humans
other hand, somatosensory evoked potentials (SSEPs) may after TBI has only recently been confirmed and depends in
play a role in prediction of outcome.92 This is further reviewed part on the ability to place ECoG monitors in perilesional
in Chapter 24, and guidelines on the use of clinical neuro- zones rather than standard frontal sites that are often distant
physiology in the ICU are available.93 from a lesion.103,104,112,113 CSD may be observed in a third of
these patients and is more likely to occur in younger patients.
Electroencephalogram, Electrocorticogram, In addition, CSD may occur with or precede a seizure, although
this observation is inconsistent and further study of this rela-
Cerebral Function Monitoring: Seizures tionship is required. Both CSD and PID events can be attenu-
and Spreading Depression ated with N-methyl-D-aspartic acid receptor blockade in
Subclincal, nonmotor, or partial seizures may occur in about animals, and so monitoring for these changes in humans may
20% of patients in the NCCU if looked for, and these noncon- provide a selection tool for those patients who could benefit
vulsive seizures can adversely influence outcome.94 Overt from these therapies in the future.112 Implantable fiber-based
motor seizures, however, may be masked by chemical paraly- optical probes also have been used to detect preictal changes
sis, so monitoring for epileptiform activity can be an impor- in the rodent hippocampus and determine whether scattering
tant adjunct to ICP, brain oximetry, or CBF monitors.12,95,96 coefficients can predict electrographic seizure onset in vivo.70
Electroencephalogram (EEG) data also are associated with
outcome and can be used to guide therapeutic interventions
before irreversible damage ensues.97-99 The presence of epilep- Pupillometer
tiform activity also is linked to episodic increases in ICP and Pupillary assessment is important when defining prognosis
the LP ratios.100 Finally, EEG data can be used as a surrogate after brain injury and to identify herniation; this component
endpoint to test the efficacy of new drugs without having to of the neurologic assessment has the potential to become
wait for functional outcome measures (typically 6 months to “automated” and more quantitative and sensitive through use
1 year). of a pupillometer.114-119 The neurologic pupillary index (NPi)
EEG monitoring is noninvasive, and machines used in the is an objective algorithm based on the pupillary indices
NCCU today are small and portable, contain data reduction obtained from healthy subjects that uses a handheld digital
software (e.g., microprocessor technology that splits informa- camera pupillometer that automatically tracks and analyzes
tion into spectra dependent on frequency, amplitude, and pupil dynamics (NeurOptics Inc., Irvine, CA)120 (Fig. 48.4).
location), provide continuous data, and eliminate the need for This technology has the potential to limit subjective descrip-
a full-time technical assistant at the bedside.8,101 The simplest tions of pupillary change and reduce interobserver variabil-
form of EEG monitoring includes a noninvasive strip of 2 to ity.121 The results of pupil assessment are stored in the device’s
4 electrodes (e.g., bispectral index [BIS]) that analyzes the memory and projected onto a liquid crystal display (LCD)
depth of sedation and whether seizure activity is present. screen. Use of the pupillometer may provide an early warning
These findings correlate with the GCS. However, BIS cannot of ICP changes that cannot be appreciated by visual inspec-
regionalize information. Larger EEG units including 8- to tion by the health care provider alone (i.e., clinical measures
14-channel 10- to 12-electrode montages provide better spatial
resolution and can collect specific information such as alpha,
delta, beta, and theta proportions of the raw EEG.97,98,100
Sophisticated units display complex information in more
simple formats that are easy to read and interpret in the NCCU
and can present the information in color spectra similar to the
appearance of a positron emission tomography (PET) scan.102
Following craniotomy, invasive EEG monitoring is feasible
with placement of an electrocorticogram (ECoG) recording
strip or grid on the cortex near the injury site.103,104 The use of
a single linear strip facilitates removal at the bedside once the
monitoring period is complete. Collected data can be stored
and analyzed offsite by specialized personnel. Use of ECoG
provides insight into cortical spreading depression (CSD) and
peri-infarct depolarization (PID). CSD consists of a wave of
depressed cortical activity or amplitude that spreads slowly
across cerebral gray matter.105 The depression is accompanied
by a transient loss of ion homeostasis, changes in glucose and
lactate, and an increase in CBF and oxygen followed by olige-
mia. ECoG changes are similar to those recorded during PIDs,
which occur in the periphery of infarct regions.103,106-109 Fig. 48.4  NeurOptics ForSite quantitative dynamic pupillometer.
However, PID and CSD are different, in that PID may contrib- Liquid crystal display screen during data targeting. Data acquisition
ute to irreversible ischemic damage. Whether CSD harbors phase. (Permission granted from NeurOptics, Irvine, Calif.)
476 Section VII—Computers, Engineering, and the Future

could be instituted early, to prevent ICP peaks and CPP underway to examine multiple biomarkers in TBI (Banyan
reduction). Biomarkers, Inc. Alachua, FL). Once an ideal biomarker is
defined, an enzyme-linked immunosorbent assay (ELISA) or
electrochemical detection system could be developed, and a
New Technologies That May Change “chip” or kit devised to allow cheap, rapid, serial measure-
Neuromonitoring: Biomarkers, ments at the bedside. The U.S. military has invested heavily in
research designed to produce such a biomarker detection
Nanotechnology, Microelectronics, device that is the size of a cell phone and can be deployed with
Wireless Systems, “Lab on a Chip” medics in combat zones.
Biomarkers: The Search for a Troponin
Equivalent in the Brain
The term biomarker has different meanings to different users,
Brain Temperature Monitoring
but in the NCCU it implies a substance or parameter that Hyperthermia is harmful after TBI, stroke, SAH, and other
represents either organ outcome or function. Ideally bio- disorders treated in the NCCU, and so monitoring of brain
markers also need to respond to treatment. Two examples are and systemic temperature is important (see Chapter 37).140
the reduction in viral load levels and increase in CD4 cells in Miniaturized thermocouples incorporated with brain oxygen
human immunodeficiency virus (HIV) with proper antiret- sensors are available for parenchymal monitoring because
roviral treatment and the reduction in cardiac specific tropo- cerebral temperatures may differ from core temperatures.82 A
nin levels following reestablishment of blood flow to the large body of evidence in animals and humans suggests that
heart. The ideal “brain biomarker” is still being sought (see induction of mild hypothermia (32° C to 35° C) early after
Chapter 18) but has the potential to improve understanding injury may arrest or slow secondary mechanisms and in
of the mechanisms associated with cellular, organ, and sys- humans can control elevated ICP.141-143 However, the results of
temic damage to facilitate future drug development and to randomized clinical trials using hypothermia other than in
alert clinicians when changes occur to guide manage- cardiac arrest have not demonstrated a consistent benefit of
ment.122,123 Similar to how cardiac troponins are specific to therapeutic hypothermia after brain injury.144 In part, this
cardiac muscle injury, the ideal neurologic biomarker should failure of clinical trials may be associated with patient
be accessible, specific to the nervous system, identify progres- selection—only specific patients will benefit—when and how
sion of secondary damage, and predict outcome.124,125 Ideally hypothermia is induced, time to target temperature, duration
it should appear in blood and CSF immediately after an of hypothermia, and speed of rewarming.142 Use of a brain
injury.124 Serum biomarkers are advantageous because of temperature monitor, perhaps as part of a micromachined
sampling ease.126 smart catheter that measures many parameters may facilitate
Potential biomarkers include cytokines (e.g., IL-6, IL-1, answers to these questions or simply better target fever control
tumor necrosis factor [TNF], and transforming growth in the ICU.145-147
factor-β [TGF-β]) that increase in blood 48 hours post injury,77
brain-specific creatine kinase (CK), glial fibrillary acidic
protein (GFAP), lactate dehydrogenase (LDH), myelin basic
protein (MBP), neuron-specific enolase (NSE), S-100β
ELISA for the Brain?
(protein expressed constitutively by brain astrocytes), and Electronic and microsensor technology has always been the
spectrin breakdown products (SBP 120), among others, or a limiting factor for neuromonitoring methods development.
combination of these markers.124 NSE, a glycolytic enzyme, Huge advances in computer memory integration, micropro-
appears rapidly in serum after brain injury and is specific to cessor speeds, microelectronics, and etched microcircuitry
brain tissue124,127 but results are conflicting and in some studies and immunologic linking have been made. These advances
has failed to separate persons with mild TBI from normal have led to a “quantum leap” in microanalytic technologies,
controls.124,128,129 S-100β also has undergone extensive study such as ELISA techniques (Fig. 48.5) and protein microarray
and its levels are associated with the GCS, imaging, and technologies. This has revolutionized the concept of “point
outcome. However, it also is found in tissue outside the brain, of care” testing (POCT), which may, in years to come,
adipose tissue, and chondrocytes.127,130-132 GFAP is released fol- render the practice of daily blood drawing in the ICU obso-
lowing TBI but not after multiple trauma without TBI; the lete. These technologies have yet to be incorporated into
GFAP levels correspond to TBI severity and outcome.133,134 neuromonitoring, but may offer the prospect of CSF or
Alpha II-spectrin is a cytoskeletal protein found in axons microdialysis based continuous “flow-through” ELISA, for a
and presynaptic membranes that is cleaved by caspase-3 cluster of biomarkers, which could detect incipient brain
and calpain, two enzymes that are activated following TBI and damage and thus offer new ways to treat or prevent such
can be measured in both blood and CSF. This protein is ele- damage.
vated in patients following TBI compared with controls,124,135
and secondary increases in CSF are observed with clinical
deterioration. Nanotechnology, Microprocessing,
Despite much research there is no ideal biomarker for prac-
tical clinical use in TBI.124,136 It also is conceivable that rather
and Long-Term Projections
than a single biomarker, a panel of several biomarkers may be “Nanotechnology is concerned with materials and systems
important.137,138 Proteomic analysis may provide a vehicle for whose structure and components exhibit novel and signifi-
new molecular discovery.139 A large multicenter trial is cantly improved physical, chemical, and biologic properties,
Section VII—Computers, Engineering, and the Future 477

Absorbent pad
Plastic backing

Control reagent

Test reagent
Membrane
with/without
blocking reagent

Conjugate pad
with reagent
Sample pad
with/without reagent
Wicking pad with reagent Sample flow Fig. 48.6  Silicone nanowire sensors for real-time, electronically
transduced, label-free biomolecular detection of electrolytes.
Fig. 48.5  Enzyme-linked immunosorbent assay (ELISA)–based Nanowires are 14 nm each. (From Cheng M, Cuda G, Bunimovich Y, et al.
flow-through system. The membrane is placed at the bottom of a Nanotechnologies for biomolecular detection and medical diagnostics. Curr Opin
sample reservoir and on top of an absorbent pad. Reagents added to Chem Biol 2006;10[1]:11–9.)
the sample reservoir flow through the membrane into the pad, allowing
discrete steps without having to use bags or other containers.

phenomena, and processes because of their small size.”148 Nanowires have been used to construct a dopamine sensor
Dimensions of these devices are in the range of 10 to 100 nm.149 for in vivo use in an implantable deep brain stimulator
This infers control of the orientation of matter at the molecu- (DBS) electrode.151 It is not hard to imagine miniaturized
lar or atomic level. At this scale quantum mechanical effects implantable sensor systems based on nanotechnology capable
sometimes cause these structures to have properties different of an array of sensing functions that communicate wirelessly
from bulk materials. Nanostructures such as nano­wires and to monitors outside the body. Production relevant to the
quantum dots hold promise in electronics, medicine, and NCCU may also come in the form of biomolecular detection
other applications. In medicine, nanostructures might be used devices, whereby 40-nm fiber-optic probes that use fluores-
to interact with individual cells, molecules, proteins, or deoxy- cence are able to monitor intra- and extracellular biochemi-
ribonucleic acid (DNA). In electronics, nanostructures may cal events. When the probes are removed, cellular events may
allow molecular level electronics and computing that would continue to be monitored due to deposition of these fluores-
enable integrated circuits to operate near the theoretical limit cent molecules.152 Delays in sampling and analysis of micro-
for power consumption per bit of processing, resulting in the dialysate may become things of the past with the introduction
smallest, fastest electronic devices possible. of “lab on a chip” technology that will give online answers
Nanowires are structures with a diameter on the order of about the parenchymal microenvironment or specific bio-
tens of nanometers and an unconstrained length. For this markers released following brain injury.153 Other forms of
reason nanowires are sometimes said to be one dimensional. biochemical detection include micro and nano electrome-
Many materials are used in their construction including chanical systems (MEMS/NEMS), which consist of carbon
metals (e.g., nickel and gold), insulators (e.g., titanium dioxide nanotubes that transduce biochemical events into electrical
and silicon dioxide), and inorganic or organic molecules (e.g., signals. These signals could then be processed, analyzed, and
DNA). Electrons in nanowires are constrained laterally; there- presented in a format to allow for protocolized decision
fore certain structures such as carbon nanotubes exhibit dis- making in the NCCU.153,154 The tools needed to create even
crete values of conductance. The future use of nanowires may the basic structures in the sizes and aspect ratios needed are
be to connect nanodevices in the construction of complex in their infancy. For example, even such exotic methods as
circuits for computing, sensing, or wireless data transfer. Sig- electron beam lithography cannot yet produce the small
nificant research is being conducted into the use of nanowires width, high-aspect ratio semiconductor materials needed for
as electronic sensors of genes and proteins. One operating nano­wire biochemical sensor arrays. New methods such as
principle is that a change in chemical potential occurring superlattice nanowire pattern (SNAP) transfer are being
during DNA hybridization can act as a field effect gate on a developed that use novel processing techniques, in this case a
doped, single-crystal nanowire. The effect is similar to a silicon fluidics approach (Fig. 48.6). Given the tremendous aca-
field effect transistor (FET). An advantage of such sensors is demic interest in this area, it is hoped that materials and
that the limitation on the number of sensor elements rests techniques soon will be developed and applied to neurocriti-
only in the ability to connect to the individual nanowires. cal care and other aspects of neurology and neurosurgery.
Theoretically, dense sensor arrays could be fabricated on very Applications such as continuous EEG mapping, in situ
small devices, enabling measurement of large numbers of protein, and DNA or other chemical analysis are tantalizing
genes or proteins from small tissue samples or even cells.150 but not yet clinically feasible.
478 Section VII—Computers, Engineering, and the Future

Design of the Future Neuroscience that consultation fees are often rewards for “loyal” users of a
particular product, drug, or device. By contrast, royalty
Intensive Care Unit revenue is distributed to those who develop a particular
“Superdocs,” Central Consoles, Data product, that is to say, innovation is rewarded. On the other
hand, the Bayh-Dole Act for universities of 1980 has catalyzed
Routing, and the Challenges in Integrating an interest by academic institutions to promote innovation
These Methods because it allows universities to patent and exclusively license
Advances in microchip technology are occurring rapidly. federally funded inventions. In turn this facilitates the transla-
These improvements will enable more refined means of data tion of ideas generated on the bench to patients at the bedside
processing, storage, and acquisition in the NCCU. The ICU of that requires close collaboration between scientists, entrepre-
the future may be a console operated system in which a central neurs, venture capitalists, and industry.162
physician is the “air traffic controller” who has access to all
data continuously and can thus make decisions and institute
therapies at remote locations with wireless systems, possibly
Conclusion
in a different hospital or even city (see also Chapters 4, 42, 43, The field of neuromonitoring is continually expanding with
and 44).155 In central California, such systems have allowed new developments. This chapter has covered past, present, and
smaller level II hospitals to continue to function with remote future techniques used to monitor patients in the NCCU.
intensive care specialists linked in by telemedicine. Early Much of the research and applications have been in TBI.
research suggests this telemedicine approach may help improve Industry plays a major role in which concepts receive financial
patient outcome, in part because it can answer staffing backing and ultimately end up in mainstream use. Monitors
needs.156,157 Linked drug-device combinations or “smart should present pertinent, online, and specific information.
systems” may automate certain functions such as administer- Furthermore, the information gathered may also facilitate an
ing benzodiazepines during the presence of seizure activity, understanding of the mechanisms involved in brain injury,
venting an ICP device, or administering mannitol during ICP including TBI, stroke, SAH, and cerebral edema. This may lead
elevations.158 However, there are many hurdles, including to the engineering of better medicines, the production of
technologic and legal to overcome before such systems become more refined clinical trials, and the improvement of patient
commonplace.159 outcomes. Promising methods that may be used increasingly
in the future include biomarkers, seizure and CSD sensing
equipment, and global blood oxygen saturation analysis
The Role of Industry through NIRS, and further in the future, nanotechnology and
Industry is a major driving force in whether certain concepts LOCs.
receive the financial support to be marketed and developed. Given the enormously responsive research and develop-
Furthermore, crossover technologic advances, such as those ment environment in the modern world, the challenge to all
made with microprocessors in the computer industry, trans- who work in NCCU is clear; there is a need to devise new ideas
late into new developments downstream for medical devices. and partner with industry, to advance the field of brain moni-
Products are in a constant flux, undergoing development and toring and improve outcome for patients. In this manner
replacement. The big neuroscience companies involved in patient management can be moved from empiric to targeted
neurocritical care (e.g., Integra Neuroscience-Plainsboro, NJ; stratified care and ultimately to individualized care based on
Medtronic, Goleta, CA; Codman, Raynham, MA) serve as a genomic and mechanistic analysis.
“choke point” for the integration and commercialization of
new ideas in NCCU. Development of devices sometimes
depends on the potential market and return on investment References
rather than the engineering and clinical expertise, and there 1. Selassie AW, Zaloshnja E, Langlois JA, et al. Incidence of long-term disability
have been visionary, futuristic ideas that have not yet received following traumatic brain injury hospitalization, United States, 2003. J Head
the financial support required for widespread production. For Trauma Rehabil 2008;23(2):123–31.
2. Rutland-Brown W, Langlois JA, Thomas KE, et al. Incidence of traumatic
example, one idea that is not yet used in NCCUs is the concept brain injury in the United States, 2003. J Head Trauma Rehabil 2006;21(6):
of monitoring oxidized and reduced states of nicotinamide 544–8.
adenine dinucleotide (NADH).160 Alternate methods for the 3. Langlois JA, Rutland-Brown W, Wald MM. The epidemiology and impact of
financial jumpstarting of ideas include SBIR grants funded by traumatic brain injury: a brief overview. J Head Trauma Rehabil 2006;21(5):
the National Institutes of Health (NIH). The SBIR program 375–8.
4. Finkelstein E, Corso PS, Miller TR. The incidence and economic burden of
supports small businesses to stimulate technologic innovation injuries in the United States. New York: Oxford University Press; 2006.
in biomedical research (see Chapter 38).161 5. Wijman CA, Smirnakis SM, Vespa P, et al. Research and technology in
Innovation is the key to progress in neuromonitoring. This neurocritical care. Neurocrit Care 2012;16(1):42–54.
depends on the interaction between physicians, surgeons and 6. Hemphill JC, Andrews P, De Georgia M. Multimodal monitoring and
neurocritical care bioinformatics. Nat Rev Neurol 2011;7(8):451–60.
scientists, information technologists, or engineers from many doi:10.1038/nrneurol.2011.101.
disciplines. However, legislation, at least in the United States 7. Diedler J, Czosnyka M. Merits and pitfalls of multimodality brain
has created intense scrutiny of the relationship between health monitoring. Neurocrit Care 2010;12(3):313–6.
care providers and industry. This scrutiny comes from both 8. Reilly P, Bullock R. Head injury: pathophysiology and management. 2nd ed.
the public and from Congress (e.g., the Grassley-Kohl Physi- London/New York: Hodder Arnold; 2005.
9. Teasdale G, Jennett B. Assessment of coma and impaired consciousness: a
cian Payments Sunshine Act of 2007). However, it is important practical scale. Lancet 1974;2(7872):81–4.
to recognize there is a difference between industry-provided 10. Zauner A, Doppenberg E, Soukup J, et al. Extended neuromonitoring:
consultation fees and royalty revenue. Critics argue new therapeutic opportunities? Neurol Res 1998;20(Suppl 1):S85–90.
Section VII—Computers, Engineering, and the Future 479

11. Guidelines for the management of severe traumatic brain injury. 33. Chatfield DA, Brahmbhatt DH, Sharp T, et al. Juguloarterial endothelin-1
J Neurotrauma 2007;24 Suppl 1:S1–106. gradients after severe traumatic brain injury. Neurocrit Care 2011;14(1):
12. Gupta AK, Bullock MR. Monitoring the injured brain in intensive care: 55–60.
present and future. Hosp Med 1998;59(9):704–13. 34. Lang EW, Mulvey JM, Mudaliar Y, et al. Direct cerebral oxygenation
13. Czosnyka M, Czosnyka Z, Pickard JD. Laboratory testing of three monitoring: a systematic review of recent publications. Neurosurg Rev
intracranial pressure microtransducers: technical report. Neurosurgery 2007;30(2):99–106; discussion 107.
1996;38(1):219–24. 35. Scheufler K. Tissue oxygenation and capacity to deliver O2 do the two go
14. Behrens A, Lenfeld N, Ambarki K, et al. Transcranial Doppler pulsatility together? Transfus Apheresis Sci 2004;31:45–54.
index: not an accurate method to assess intracranial pressure. Neurosurgery 36. Maloney-Wilensky E, Gracias V, Itkin A, et al. Brain tissue oxygen and
2010;66(6):1050–7. outcome after severe traumatic brain injury: a systematic review. Crit Care
15. Querfurth HW, Lieberman P, Arms S, et al. Ophthalmodynamometry for Med 2009;37(6):2057–63.
ICP prediction and pilot test on Mt. Everest. BMC Neurol 2010;10:106. 37. Oddo M, Levine JM, MacKenzie L, et al. Brain hypoxia is a predictor of
16. Moretti R, Pizzi B. Ultrasonography of the optic nerve in neurocritically ill short-term outcome after severe traumatic brain injury independent of
patients. Acta Anaesthesiol Scand 2011;55(6):644–52. intracranial hypertension and low cerebral perfusion pressure. Neurosurgery
17. Rosenberg JB, Shiloh AL, Savel RH, et al. Non-invasive methods of 2001;69:1037–45.
estimating intracranial pressure. Neurocrit Care 2011;15(3):599–608. 38. Pascual JL, Georgoff P, Horan A, et al. Brain tissue hypoxia in traumatic
18. Aoi MC, Matzuka BJ, Olufsen MS. Toward online, noninvasive, nonlinear brain injury: are most commonly used interventions successful at correcting
assessment of cerebral autoregulation. Conf Proc IEEE Eng Med Biol Soc brain tissue oxygen deficits? J Trauma 2011;70(3):535–46.
2011;2011:2410–3. 39. Bohman LE, Heuer GG, Macyszyn L, et al. Medical management of
19. Aries MJ, Czosnyka M, Budohoski KP, et al. Continuous monitoring of compromised brain oxygen in patients with severe traumatic brain injury.
cerebrovascular reactivity using pulse waveform of intracranial pressure. Neurocrit Care 2011;14:361–9.
Neurocrit Care 2012;Apr 3. Epub ahead of print. 40. Nangunoori R, Maloney-Wilensky E, Stiefel MD, et al. Brain tissue oxygen
20. Sorrentino E, Diedler J, Kasprowicz M, et al. Critical thresholds for based therapy and outcome after severe traumatic brain injury: a systematic
cerebrovascular reactivity after traumatic brain injury. Neurocrit Care literature review. Neurocrit Care 2011;Aug 16. Epub ahead of print.
2012;16(2):258–66. 41. van den Brink WA, van Santbrink H, Steyerberg EW, et al. Brain oxygen
21. Radolovich DK, Aries MJ, Castellani G, et al. Pulsatile intracranial pressure tension in severe head injury. Neurosurgery 2000;46(4):868–76; discussion
and cerebral autoregulation after traumatic brain injury. Neurocrit Care 876–8.
2011;15(3):379–86. 42. Ponce LL, Pillai S, Cruz J, et al. Position of probe determines prognostic
22. Lavinio A, Rasulo FA, De Peri E, et al. The relationship between the information of brain tissue PO2 in severe traumatic brain injury.
intracranial pressure-volume index and cerebral autoregulation. Intensive Neurosurgery 2012;Jan 27. Epub ahead of print.
Care Med 2009;35(3):546–9. Epub 2008, Oct 11. 43. Sarrafzadeh AS, Kiening KL, Bardt TF, et al. Cerebral oxygenation in
23. Mazzeo AT, Bullock R. Monitoring brain tissue oxymetry: will it change contusioned vs. nonlesioned brain tissue: monitoring of PtiO2 with Licox
management of critically ill neurologic patients? J Neurol Sci 2007;261(1-2): and Paratrend. Acta Neurochir 1998;71:186–9.
1–9. 44. van den Brink WA, Haitsma IK, Avezaat CJ, et al. Brain parenchyma/pO2
24. Bouma GJ, Muizelaar JP, Bandoh K, et al. Blood pressure and intracranial catheter interface: a histopathological study in the rat. J Neurotrauma 1998;
pressure-volume dynamics in severe head injury: relationship with cerebral 15(10):813–24.
blood flow. J Neurosurg 1992;77(1):15–9. 45. Lubbers DW, Baumgartl H, Zimelka W. Heterogeneity and stability of local
25. Doppenberg EM, Zauner A, Bullock R, et al. Correlations between brain PO2 distribution within the brain tissue. Adv Exper Med Biol 1994;345:567–74.
tissue oxygen tension, carbon dioxide tension, pH, and cerebral blood flow– 46. Carter LP, Weinand ME, Oommen KJ. Cerebral blood flow (CBF)
a better way of monitoring the severely injured brain? Surg Neurol 1998; monitoring in intensive care by thermal diffusion. Acta Neurochir Suppl
49(6):650–4. (Wien) 1993;59:43–6.
26. Sheinberg M, Kanter MJ, Robertson CS, et al. Continuous monitoring of 47. Vajkoczy P, Roth H, Horn P, et al. Continuous monitoring of regional
jugular venous oxygen saturation in head-injured patients. J Neurosurg cerebral blood flow: experimental and clinical validation of a novel thermal
1992;76(2):212–7. diffusion microprobe. J Neurosurg 2000;93(2):265–74.
27. Robertson C. Desaturation episodes after severe head injury: influence on 48. Rosenthal G, Sanchez-Mejia RO, Phan N, et al. Incorporating a parenchymal
outcome. Acta Neurochir Suppl (Wien) 1993;59:98–101. thermal diffusion cerebral blood flow probe in bedside assessment of
28. Siggaard-Andersen O, Fogh-Andersen N, Gothgen IH, et al. Oxygen status of cerebral autoregulation and vasoreactivity in patients with severe traumatic
arterial and mixed venous blood. Crit Care Med 1995;23(7):1284–93. brain injury. J Neurosurg 2011;114(1):62–70. Epub 2010, Aug 13.
29. Sarrafzadeh AS, Kiening KL, Unterberg AW. Neuromonitoring: brain 49. Hänggi D, Participants in the International Multi-Disciplinary Consensus
oxygenation and microdialysis. Cur Neurol Neurosci Rep 2003;3(6):517–23. Conference on the Critical Care Management of Subarachnoid Hemorrhage.
30. Kiening KL, Unterberg AW, Bardt TF, et al. Monitoring of cerebral Monitoring and detection of vasospasm. II: EEG and invasive monitoring.
oxygenation in patients with severe head injuries: brain tissue PO2 versus Neurocrit Care 2011;15(2):318–23
jugular vein oxygen saturation. J Neurosurg 1996;85(5):751–7. 50. McCormick PW, Stewart M, Goetting MG, et al. Noninvasive cerebral optical
31. Kirkpatrick PJ, Smielewski P, Czosnyka M, et al. Near-infrared spectroscopy spectroscopy for monitoring cerebral oxygen delivery and hemodynamics.
use in patients with head injury. J Neurosurg 1995;83(6):963–70. Crit Care Med 1991;19(1):89–97.
32. Helmy A, Carpenter KL, Menon DK, et al. The cytokine response to human
traumatic brain injury: temporal profiles and evidence for cerebral A complete list of references for this chapter can be found online at
parenchymal production. J Cereb Blood Flow Metab 2011;31(2):658–70. www.expertconsult.com.
Section VII—Computers, Engineering, and the Future 479.e1

References 29. Sarrafzadeh AS, Kiening KL, Unterberg AW. Neuromonitoring: brain
oxygenation and microdialysis. Cur Neurol Neurosci Rep 2003;3(6):
1. Selassie AW, Zaloshnja E, Langlois JA, et al. Incidence of long-term 517–23.
disability following traumatic brain injury hospitalization, United States, 30. Kiening KL, Unterberg AW, Bardt TF, et al. Monitoring of cerebral
2003. J Head Trauma Rehabil 2008;23(2):123–31. oxygenation in patients with severe head injuries: brain tissue PO2 versus
2. Rutland-Brown W, Langlois JA, Thomas KE, et al. Incidence of traumatic jugular vein oxygen saturation. J Neurosurg 1996;85(5):751–7.
brain injury in the United States, 2003. J Head Trauma Rehabil 2006;21(6): 31. Kirkpatrick PJ, Smielewski P, Czosnyka M, et al. Near-infrared spectroscopy
544–8. use in patients with head injury. J Neurosurg 1995;83(6):963–70.
3. Langlois JA, Rutland-Brown W, Wald MM. The epidemiology and impact 32. Helmy A, Carpenter KL, Menon DK, et al. The cytokine response to
of traumatic brain injury: a brief overview. J Head Trauma Rehabil 2006; human traumatic brain injury: temporal profiles and evidence for
21(5):375–8. cerebral parenchymal production. J Cereb Blood Flow Metab 2011;31(2):
4. Finkelstein E, Corso PS, Miller TR. The incidence and economic burden of 658–70.
injuries in the United States. New York: Oxford University Press; 2006. 33. Chatfield DA, Brahmbhatt DH, Sharp T, et al. Juguloarterial endothelin-1
5. Wijman CA, Smirnakis SM, Vespa P, et al. Research and technology in gradients after severe traumatic brain injury. Neurocrit Care
neurocritical care. Neurocrit Care 2012;16(1):42–54. 2011;14(1):55–60.
6. Hemphill JC, Andrews P, De Georgia M. Multimodal monitoring and 34. Lang EW, Mulvey JM, Mudaliar Y, et al. Direct cerebral oxygenation
neurocritical care bioinformatics. Nat Rev Neurol 2011;7(8):451–60. monitoring: a systematic review of recent publications. Neurosurg Rev
doi:10.1038/nrneurol.2011.101. 2007;30(2):99–106; discussion 107.
7. Diedler J, Czosnyka M. Merits and pitfalls of multimodality brain 35. Scheufler K. Tissue oxygenation and capacity to deliver O2 do the two go
monitoring. Neurocrit Care 2010;12(3):313–6. together? Transfus Apheresis Sci 2004;31:45–54.
8. Reilly P, Bullock R. Head injury: pathophysiology and management. 2nd ed. 36. Maloney-Wilensky E, Gracias V, Itkin A, et al. Brain tissue oxygen and
London/New York: Hodder Arnold; 2005. outcome after severe traumatic brain injury: a systematic review. Crit Care
9. Teasdale G, Jennett B. Assessment of coma and impaired consciousness: Med 2009;37(6):2057–63.
a practical scale. Lancet 1974;2(7872):81–4. 37. Oddo M, Levine JM, MacKenzie L, et al. Brain hypoxia is a predictor of
10. Zauner A, Doppenberg E, Soukup J, et al. Extended neuromonitoring: new short-term outcome after severe traumatic brain injury independent of
therapeutic opportunities? Neurol Res 1998;20(Suppl 1):S85–90. intracranial hypertension and low cerebral perfusion pressure.
11. Guidelines for the management of severe traumatic brain injury. Neurosurgery 2001;69:1037–45.
J Neurotrauma 2007;24(Suppl 1):S1–106. 38. Pascual JL, Georgoff P, Horan A, et al. Brain tissue hypoxia in traumatic
12. Gupta AK, Bullock MR. Monitoring the injured brain in intensive care: brain injury: are most commonly used interventions successful at
present and future. Hosp Med 1998;59(9):704–13. correcting brain tissue oxygen deficits? J Trauma 2011;70(3):535–46.
13. Czosnyka M, Czosnyka Z, Pickard JD. Laboratory testing of three 39. Bohman LE, Heuer GG, Macyszyn L, et al. Medical management of
intracranial pressure microtransducers: technical report. Neurosurgery compromised brain oxygen in patients with severe traumatic brain injury.
1996;38(1):219–24. Neurocrit Care 2011;14:361–9.
14. Behrens A, Lenfeld N, Ambarki K, et al. Transcranial Doppler pulsatility 40. Nangunoori R, Maloney-Wilensky E, Stiefel MD, et al. Brain tissue
index: not an accurate method to assess intracranial pressure. Neurosurgery oxygen based therapy and outcome after severe traumatic brain injury:
2010;66(6):1050–7. a systematic literature review. Neurocrit Care 2011;Aug 16. Epub ahead of
15. Querfurth HW, Lieberman P, Arms S, et al. Ophthalmodynamometry for print.
ICP prediction and pilot test on Mt. Everest. BMC Neurol 2010;10:106. 41. van den Brink WA, van Santbrink H, Steyerberg EW, et al. Brain oxygen
16. Moretti R, Pizzi B. Ultrasonography of the optic nerve in neurocritically ill tension in severe head injury. Neurosurgery 2000;46(4):868–76; discussion
patients. Acta Anaesthesiol Scand 2011;55(6):644–52. 876–8.
17. Rosenberg JB, Shiloh AL, Savel RH, et al. Non-invasive methods of 42. Ponce LL, Pillai S, Cruz J, et al. Position of probe determines prognostic
estimating intracranial pressure. Neurocrit Care 2011;15(3):599–608. information of brain tissue PO2 in severe traumatic brain injury.
18. Aoi MC, Matzuka BJ, Olufsen MS. Toward online, noninvasive, nonlinear Neurosurgery 2012;Jan 27. Epub ahead of print.
assessment of cerebral autoregulation. Conf Proc IEEE Eng Med Biol Soc 43. Sarrafzadeh AS, Kiening KL, Bardt TF, et al. Cerebral oxygenation in
2011;2011:2410–3. contusioned vs. nonlesioned brain tissue: monitoring of PtiO2 with Licox
19. Aries MJ, Czosnyka M, Budohoski KP, et al. Continuous monitoring of and Paratrend. Acta Neurochir 1998;71:186–9.
cerebrovascular reactivity using pulse waveform of intracranial pressure. 44. van den Brink WA, Haitsma IK, Avezaat CJ, et al. Brain parenchyma/pO2
Neurocrit Care 2012;Apr 3. Epub ahead of print. catheter interface: a histopathological study in the rat. J Neurotrauma 1998;
20. Sorrentino E, Diedler J, Kasprowicz M, et al. Critical thresholds for 15(10):813–24.
cerebrovascular reactivity after traumatic brain injury. Neurocrit Care 45. Lubbers DW, Baumgartl H, Zimelka W. Heterogeneity and stability of local
2012;16(2):258–66. PO2 distribution within the brain tissue. Adv Exper Med Biol 1994;345:
21. Radolovich DK, Aries MJ, Castellani G, et al. Pulsatile intracranial pressure 567–74.
and cerebral autoregulation after traumatic brain injury. Neurocrit Care 46. Carter LP, Weinand ME, Oommen KJ. Cerebral blood flow (CBF)
2011;15(3):379–86. monitoring in intensive care by thermal diffusion. Acta Neurochir Suppl
22. Lavinio A, Rasulo FA, De Peri E, et al. The relationship between the (Wien) 1993;59:43–6.
intracranial pressure-volume index and cerebral autoregulation. Intensive 47. Vajkoczy P, Roth H, Horn P, et al. Continuous monitoring of regional
Care Med 2009;35(3):546–9. Epub 2008, Oct 11. cerebral blood flow: experimental and clinical validation of a novel thermal
23. Mazzeo AT, Bullock R. Monitoring brain tissue oxymetry: will it change diffusion microprobe. J Neurosurg 2000;93(2):265–74.
management of critically ill neurologic patients? J Neurol Sci 48. Rosenthal G, Sanchez-Mejia RO, Phan N, et al. Incorporating a
2007;261(1-2):1–9. parenchymal thermal diffusion cerebral blood flow probe in bedside
24. Bouma GJ, Muizelaar JP, Bandoh K, et al. Blood pressure and intracranial assessment of cerebral autoregulation and vasoreactivity in patients with
pressure-volume dynamics in severe head injury: relationship with cerebral severe traumatic brain injury. J Neurosurg 2011;114(1):62–70. Epub 2010,
blood flow. J Neurosurg 1992;77(1):15–9. Aug 13.
25. Doppenberg EM, Zauner A, Bullock R, et al. Correlations between brain 49. Hänggi D, Participants in the International Multi-Disciplinary Consensus
tissue oxygen tension, carbon dioxide tension, pH, and cerebral blood Conference on the Critical Care Management of Subarachnoid
flow–a better way of monitoring the severely injured brain? Surg Neurol Hemorrhage. Monitoring and detection of vasospasm. II: EEG and invasive
1998;49(6):650–4. monitoring. Neurocrit Care 2011;15(2):318–23
26. Sheinberg M, Kanter MJ, Robertson CS, et al. Continuous monitoring of 50. McCormick PW, Stewart M, Goetting MG, et al. Noninvasive cerebral
jugular venous oxygen saturation in head-injured patients. J Neurosurg optical spectroscopy for monitoring cerebral oxygen delivery and
1992;76(2):212–7. hemodynamics. Crit Care Med 1991;19(1):89–97.
27. Robertson C. Desaturation episodes after severe head injury: influence on 51. Brazy JE. Cerebral oxygen monitoring with near infrared spectroscopy:
outcome. Acta Neurochir Suppl (Wien) 1993;59:98–101. clinical application to neonates. J Clin Monit 1991;7(4):325–34.
28. Siggaard-Andersen O, Fogh-Andersen N, Gothgen IH, et al. Oxygen 52. Jobsis FF. Noninvasive, infrared monitoring of cerebral and myocardial
status of arterial and mixed venous blood. Crit Care Med 1995;23(7): oxygen sufficiency and circulatory parameters. Science 1977;198(4323):
1284–93. 1264–7.
479.e2 Section VII—Computers, Engineering, and the Future

53. McCormick PW, Stewart M, Goetting MG, et al. Regional cerebrovascular 78. Hillman J, Aneman O, Anderson C, et al. A microdialysis technique for
oxygen saturation measured by optical spectroscopy in humans. Stroke routine measurement of macromolecules in the injured human brain.
1991;22(5):596–602. Neurosurgery 2005;56(6):1264–8; discussion 1268–70.
54. Lee JK, Kibler KK, Benni PB, et al. Cerebrovascular reactivity measured by 79. Johnston AJ, Gupta AK. Advanced monitoring in the neurology intensive
near-infrared spectroscopy. Stroke 2009;40(5):1820–6. Epub 2009, Mar 12. care unit: microdialysis. Curr Opin Crit Care 2002;8(2):121–7.
55. Zweifel C, Castellani G, Czosnyka M, et al. Noninvasive monitoring of 80. Zauner A, Doppenberg E, Woodward JJ, et al. Multiparametric continuous
cerebrovascular reactivity with near infrared spectroscopy in head-injured monitoring of brain metabolism and substrate delivery in neurosurgical
patients. J Neurotrauma 2010(11):1951–8. Epub 2010, Oct 28. patients. Neurol Res 1997;19(3):265–73.
56. Brady K, Joshi B, Zweifel C, et al. Real-time continuous monitoring of 81. Meixensberger J, Kunze E, Barcsay E, et al. Clinical cerebral microdialysis:
cerebral blood flow autoregulation using near-infrared spectroscopy in brain metabolism and brain tissue oxygenation after acute brain injury.
patients undergoing cardiopulmonary bypass. Stroke 2010;41(9):1951–6. Neurol Res 2001;23(8):801–6.
Epub 2010, Jul 22. 82. Mathew P, Gentleman D, Bullock MR. Brain monitoring in severe head
57. Brown JI, Moulton RJ, Konasiewicz SJ, et al. Cerebral oxidative metabolism injury: a practical guide. Trauma 1999;1:105–14.
and evoked potential deterioration after severe brain injury: new evidence 83. Bullock R, Zauner A, Woodward JJ, et al. Factors affecting excitatory amino
of early posttraumatic ischemia. Neurosurgery 1998;42(5):1057–63; acid release following severe human head injury. J Neurosurg
discussion 1063–4. 1998;89(4):507–18.
58. Germon TJ, Evans PD, Manara AR, et al. Sensitivity of near infrared 84. Bullock R, Zauner A, Myseros JS, et al. Evidence for prolonged release of
spectroscopy to cerebral and extra-cerebral oxygenation changes is excitatory amino acids in severe human head trauma: relationship to
determined by emitter-detector separation. J Clin Monitor Comput clinical events. Ann N Y Acad Sci 1995;765:290–7; discussion 298.
1998;14(5):353–60. 85. Persson L, Hillered L. Chemical monitoring of neurosurgical intensive care
59. Chance B, Cope M, Gratton E, et al. Phase measurement of light absorption patients using intracerebral microdialysis. J Neurosurg 1992;76(1):72–80.
and scatter in human tissues. Rev Sci Instrum 1998;69(10):3457–81. 86. Hutchinson PJ, al-Rawi PG, O’Connell MT, et al. On-line monitoring of
60. Chance B, Nioka S, Zhao Z. A wearable brain imager. IEEE Eng Med Biol substrate delivery and brain metabolism in head injury. Acta Neurochir
Mag 2007;26(4):30–7. 2000;76:431–5.
61. Izzetoglu M, Bunce SC, Izzetoglu K, et al. Functional brain imaging using 87. Belli A, Sen J, Petzold A, et al. Metabolic failure precedes intracranial
near-infrared technology. IEEE Eng Med Biol Mag 2007;26(4):38–46. pressure rises in traumatic brain injury: a microdialysis study. Acta
62. Kytta J, Ohman J, Tanskanen P, et al. Extracranial contribution to cerebral Neurochir (Wien) 2008;150(5):461–9; discussion 470.
oximetry in brain dead patients: a report of six cases. J Neurosurg 88. Timofeev I, Carpenter KL, Nortje J, et al. Cerebral extracellular chemistry
Anesthesiol 1999;11(4):252–4. and outcome following traumatic brain injury: a microdialysis study of 223
63. Brawanski A, Faltermeier R, Rothoerl RD, et al. Comparison of near- patients. Brain 2011;134(Pt 2):484–94. Epub 2011, Jan 18.
infrared spectroscopy and tissue p(O2) time series in patients after severe 89. Chen HI, Stiefel MF, Oddo M, et al. Detection of cerebral compromise with
head injury and aneurysmal subarachnoid hemorrhage. J Cereb Blood Flow multimodality monitoring in patients with subarachnoid hemorrhage.
Metab 2002;22(5):605–11. Neurosurgery 2011;69:53–63.
64. Kirkpatrick PJ, Smielewski P, Whitfield PC, et al. An observational study of 90. Oddo M, Levine J, Frangos S, et al. Brain lactate metabolism in humans
near-infrared spectroscopy during carotid endarterectomy. J Neurosurg with subarachnoid haemorrhage. Stroke 2012;Feb 16. Epub ahead of print.
1995;82(5):756–63. 91. Goodman JC, Robertson CS. Microdialysis: is it ready for prime time? Curr
65. Hueber DM, Franceschini MA, Ma HY, et al. Non-invasive and quantitative Opin Crit Care 2009;15(2):110–7.
near-infrared haemoglobin spectrometry in the piglet brain during hypoxic 92. Houlden DA, Taylor AB, Feinstein A, et al. Early somatosensory evoked
stress, using a frequency-domain multidistance instrument. Phys Med Biol potential grades in comatose traumatic brain injury patients predict
2001;46(1):41–62. cognitive and functional outcome. Crit Care Med 2010;38:167–74.
66. Tu T, Chen Y, Zhang J, et al. Analysis on performance and optimization of 93. Guerit JM, Amantini A, Amodio P, et al. Consensus on the use of
frequency-domain near-infrared instruments. J Biomed Opt neurophysiological tests in the intensive care unit (ICU):
2002;7(4):643–9. electroencephalogram (EEG), evoked potentials (EPs), and
67. Chen Y, Tailor DR, Intes X, et al. Correlation between near-infrared electroneuromyography (ENMG). Neurophysiol Clin 2009;39:71–83.
spectroscopy and magnetic resonance imaging of rat brain oxygenation 94. Vespa PM, McArthur DL, Xu Y, et al. Nonconvulsive seizures after
modulation. Phys Med Biol 2003;48(4):417–27. traumatic brain injury are associated with hippocampal atrophy. Neurology
68. Chance B, Ma HY, Nioka S. Quantitative brain tissue oximetry, phase 2010;75:792–8.
spectroscopy and imaging the range of homeostasis in piglet brain. Adv 95. Vespa P. Continuous EEG monitoring for the detection of seizures in
Exp Med Biol 2003;530:13–8. traumatic brain injury, infarction, and intracerebral hemorrhage: “to detect
69. Owen C, Gill A, Binder D. Detection of cerebral edema through an and protect.” J Clin Neurophysiol 2005;22(2):99–106.
implanted optical probe. Seattle, WA: American Epilepsy Society; 2008. 96. Vespa PM, Nuwer MR, Nenov V, et al. Increased incidence and impact of
70. Owen C, Mathews M, Abokasis D, et al. Pre-ictal optical changes in rodent nonconvulsive and convulsive seizures after traumatic brain injury as
hippocampus detected with an implanted optical probe. American Epilepsy detected by continuous electroencephalographic monitoring. J Neurosurg
Society December 5-9 Seattle, 2008. 1999;91(5):750–60.
71. Binder D. Early detection of cerebral edema through an implanted optical 97. Hebb MO, McArthur DL, Alger J, et al. Impaired percent alpha variability
probe. In: Proceedings of the AANS National Meeting 2008; Chicago, 2008 on continuous electroencephalography is associated with thalamic injury
(abstract). and predicts poor long-term outcome after human traumatic brain injury.
72. Robertson CS, Zager EL, Narayan RK, et al. Clinical evaluation of a J Neurotrauma 2007;24(4):579–90.
portable near-infrared device for detection of traumatic intracranial 98. Vespa PM, Boscardin WJ, Hovda DA, et al. Early and persistent impaired
hematomas. J Neurotrauma 2010;27(9):1597–604. percent alpha variability on continuous electroencephalography monitoring
73. Meyerson BA, Linderoth B, Karlsson H, et al. Microdialysis in the human as predictive of poor outcome after traumatic brain injury. J Neurosurg
brain: extracellular measurements in the thalamus of parkinsonian patients. 2002;97(1):84–92.
Life Sci 1990;46(4):301–8. 99. Rossetti AO, Oddo M, Logroscino G, et al. Prognostication after cardiac
74. Sarrafzadeh AS, Sakowitz OW, Callsen TA, et al. Bedside microdialysis for arrest and hypothermia: a prospective study. Ann Neurol 2010;67:301–7.
early detection of cerebral hypoxia in traumatic brain injury. Neurosurg 100. Vespa PM, Miller C, McArthur D, et al. Nonconvulsive electrographic seizures
Focus 2000;9(5):e2. after traumatic brain injury result in a delayed, prolonged increase in
75. Reinert M, Khaldi A, Zauner A, et al. High level of extracellular potassium intracranial pressure and metabolic crisis. Crit Care Med 2007;35(12):2830–6.
and its correlates after severe head injury: relationship to high intracranial 101. Friedman D, Claassen J, Hirsch LJ. Continuous electroencephalogram
pressure. J Neurosurg 2000;93(5):800–7. monitoring in the intensive care unit. Anesth Analg 2009;109:506–23.
76. Khaldi A, Zauner A, Reinert M, et al. Measurement of nitric oxide and 102. Wallace BE, Wagner AK, Wagner EP, et al. A history and review of
brain tissue oxygen tension in patients after severe subarachnoid quantitative electroencephalography in traumatic brain injury. J Head
hemorrhage. Neurosurgery 2001;49(1):33–8; discussion 38–40. Trauma Rehabil 2001;16(2):165–90.
77. Bullock MR, Doppenberg E, Zauner A, et al. New techniques for 103. Strong AJ, Fabricius M, Boutelle MG, et al. Spreading and synchronous
multi-modality monitoring of the injured brain: opportunities for therapy? depressions of cortical activity in acutely injured human brain. Stroke
Jap J Int Crit Care Med 1997;9(11):1295–300. 2002;33(12):2738–43.
Section VII—Computers, Engineering, and the Future 479.e3

104. Fabricius M, Fuhr S, Willumsen L, et al. Co-occurrence of spreading 132. Raabe A, Menon DK, Gupta S, et al. Jugular venous and arterial
depression and seizures after acute brain injury. Clin Neurophysiol 2008; concentrations of serum S-100B protein in patients with severe head injury:
119(9):1973–84. a pilot study. J Neurol Neurosurg Psychiatry 1998;65(6):930–2.
105. Leao A. Spreading depression of activity in cerebral cortex. J Neurophysiol 133. Pelinka LE, Kroepfl A, Schmidhammer R, et al. Glial fibrillary acidic protein
1944;7:359–90. in serum after traumatic brain injury and multiple trauma. J Trauma 2004;
106. Hopwood SE, Parkin MC, Bezzina EL, et al. Transient changes in cortical 57(5):1006–12.
glucose and lactate levels associated with peri-infarct depolarisations, 134. Papa L, Lewis LM, Falk JL, et al. Elevated levels of serum glial fibrillary
studied with rapid-sampling microdialysis. J Cereb Blood Flow Metab acidic protein breakdown products in mild and moderate traumatic brain
2005;25(3):391–401. injury are associated with intracranial lesions and neurosurgical
107. Lauritzen M, Jorgensen MB, Diemer NH, et al. Persistent oligemia of rat intervention. Ann Emerg Med 2011;Nov 8. Epub ahead of print.
cerebral cortex in the wake of spreading depression. Ann Neurol 1982; 135. Cardali S, Maugeri R. Detection of alphaII-spectrin and breakdown
12(5):469–74. products in humans after severe traumatic brain injury. J Neurosurg Sci
108. Hossman K. Periinfarct depolarizations. Cerebrovasc Brain Metab Rev 2006;50(2):25–31.
1996;8:195–208. 136. Laskowitz DT, Grocott H, Hsia A, et al. Serum markers of cerebral
109. Kraig RP, Nicholson C. Extracellular ionic variations during spreading ischemia. J Stroke Cerebrovasc Dis 1998;7(4):234–41.
depression. Neuroscience 1978;3(11):1045–59. 137. Siman R, Toraskar N, Dang A, et al. A panel of neuron-enriched proteins as
110. Busch E, Gyngell M, Elis M, et al. Potassium-induced cortical spreading markers for traumatic brain injury in humans. J Neurotrauma
depressions during focal cerebral ischemia in rats: contribution to lesion 2009;26(11):1867–77
growth assessed by diffusion-weighted NMR and biochemical imaging. 138. Siman R, Giovannone N, Toraskar N, et al. Evidence that a panel of
J Cereb Blood Flow Metab 1996;16:1090–9. neurodegeneration biomarkers predicts vasospasm, infarction, and outcome
111. Dreier JP. The role of spreading depression, spreading depolarization and in aneurysmal subarachnoid hemorrhage. PLoS One 2011;6(12):e28938.
spreading ischemia in neurological disease. Nat Med 2011;17:1–9. Epub 2011, Dec 9.
112. Fabricius M, Fuhr S, Bhatia R, et al. Cortical spreading depression and 139. Prieto DA, Ye X, Veenstra TD. Proteomic analysis of traumatic brain injury:
peri-infarct depolarization in acutely injured human cerebral cortex. Brain the search for biomarkers. Expert Rev Proteomics 2008;5(2):283–91.
2006;129(Pt 3):778–90. 140. Metz C, Holzschuh M, Bein T, et al. Moderate hypothermia in patients with
113. Mayevsky A, Doron A, Manor T, et al. Cortical spreading depression severe head injury: cerebral and extracerebral effects. J Neurosurg
recorded from the human brain using a multiparametric monitoring 1996;85(4):533–41.
system. Brain Res 1996;740(1-2):268–74. 141. Busto R, Globus MY, Dietrich WD, et al. Effect of mild hypothermia on
114. Chesnut RM, Gautille T, Blunt BA, et al. The localizing value of asymmetry ischemia-induced release of neurotransmitters and free fatty acids in rat
in pupillary size in severe head injury: relation to lesion type and location. brain. Stroke 1989;20(7):904–10.
Neurosurgery 1994;34(5):840–5; discussion 845–6. 142. Polderman KH. Induced hypothermia and fever control for prevention and
115. Andrews BT, Pitts LH. Functional recovery after traumatic transtentorial treatment of neurological injuries. Lancet 2008;371(9628):1955–69.
herniation. Neurosurgery 1991;29(2):227–31. 143. Sadaka F, Veremakis C. Therapeutic hypothermia for the management of
116. Braakman R, Gelpke GJ, Habbema JD, et al. Systematic selection of intracranial hypertension in severe traumatic brain injury: a systematic
prognostic features in patients with severe head injury. Neurosurgery 1980; review. Brain Inj 2012;Mar 26. Epub ahead of print.
6(4):362–70. 144. Hutchison JS, Ward RE, Lacroix J, et al. Hypothermia therapy after
117. Sakas DE, Bullock MR, Teasdale GM. One-year outcome following traumatic brain injury in children. N Engl J Med 2008;358(23):2447–56.
craniotomy for traumatic hematoma in patients with fixed dilated pupils. 145. Badjatia N. Hyperthermia and fever control in brain injury. Crit Care Med
J Neurosurg 1995;82(6):961–5. 2009;37(7 Suppl):S250–7.
118. Chen JW, Gombart ZJ, Rogers S, et al. Pupillary reactivity as an early 146. Childs C, Wieloch T, Lecky F, et al. Report of a consensus meeting on
indicator of increased intracranial pressure: the introduction of the human brain temperature after severe traumatic brain injury: its
Neurological Pupil index. Surg Neurol Int 2011;2:82. Epub 2011, Jun 21. measurement and management during pyrexia. Front Neurol 2010;1:146.
119. Fountas KN, Kapsalaki EZ, Machinis TG, et al. Clinical implications of 147. Li C, Wu PM, Wu Z, et al. Brain temperature measurement: a study of in
quantitative infrared pupillometry in neurosurgical patients. Neurocrit Care vitro accuracy and stability of smart catheter temperature sensors. Biomed
2006;5(1):55–60. Microdevices 2011;Sep 28. Epub ahead of print.
120. Hults KN, Knowlton SL, Oliver JW, et al. A study of pupillary assessment in 148. Leary SP, Liu CY, Yu C, et al. Toward the emergence of nanoneurosurgery.
outpatient neurosurgical clinics. J Neurosci Nurs 2006;38(6):447–52. Part I: progress in nanoscience, nanotechnology, and the comprehension of
121. Litvan I, Saposnik G, Maurino J, et al. Pupillary diameter assessment: need events in the mesoscale realm. Neurosurgery 2005;57(4):606–34; discussion
for a graded scale. Neurology 2000;54(2):530–1. 634.
122. Ingebrigtsen T, Romner B. Biochemical serum markers of traumatic brain 149. Lakhtakia A. Nanometer structures: theory, modeling, and simulation.
injury. J Trauma 2002;52(4):798–808. Bellingham, WA: SPIE Press; 2004.
123. Narayan RK, Michel ME, Ansell B, et al. Clinical trials in head injury. 150. Cheng M, Cuda G, Bunimovich Y, et al. Nanotechnologies for biomolecular
J Neurotrauma 2002;19(5):503–57. detection and medical diagnostics. Curr Opin Chem Biol 2006;10(1):11–9.
124. Pineda JA, Wang KK, Hayes RL. Biomarkers of proteolytic damage 151. Varadan VK, Society of Photo-optical Instrumentation Engineers, et al.
following traumatic brain injury. Brain Pathol 2004;14(2):202–9. Nanosensors, microsensors, and biosensors and systems 2007. 21-22 March,
125. Antman EM, Tanasijevic MJ, Thompson B, et al. Cardiac-specific troponin 2007, San Diego.
I levels to predict the risk of mortality in patients with acute coronary 152. Vo-Dinh T, Kasili P. Fiber-optic nanosensors for single-cell monitoring.
syndromes. N Engl J Med 1996;335(18):1342–9. Anal Bioanal Chem 2005;382(4):918–25.
126. Hergenroeder G, Redell JB, Moore AN, et al. Identification of serum 153. Elder JB, Liu CY, Apuzzo ML. Neurosurgery in the realm of 10(-9). Part 1:
biomarkers in brain-injured adults: potential for predicting elevated stardust and nanotechnology in neuroscience. Neurosurgery
intracranial pressure. J Neurotrauma 2008;25(2):79–93. 2008;62(1):1–20.
127. Johnsson P. Markers of cerebral ischemia after cardiac surgery. 154. Patolsky F, Zheng G, Lieber CM. Nanowire-based biosensors. Anal Chem
J Cardiothorac Vasc Anesth 1996;10(1):120–6. 2006;78(13):4260–9.
128. Ross SA, Cunningham RT, Johnston CF, et al. Neuron-specific enolase as 155. Elder JB, Liu CY, Apuzzo ML. Neurosurgery in the realm of 10(-9). Part 2:
an aid to outcome prediction in head injury. Br J Neurosurg 1996;10(5): applications of nanotechnology to neurosurgery: present and future.
471–6. Neurosurgery 2008;62(2):269–84; discussion 284–5.
129. Yamazaki Y, Yada K, Morii S, et al. Diagnostic significance of serum 156. Zawada ET Jr, Herr P, Larson D, et al. Impact of an intensive care unit
neuron-specific enolase and myelin basic protein assay in patients with telemedicine program on a rural health care system. Postgrad Med
acute head injury. Surg Neurol 1995;43(3):267–70; discussion 270–1. 2009;121(3):160–70.
130. Woertgen C, Rothoerl RD, Metz C, et al.. Comparison of clinical, 157. Young LB, Chan PS, Lu X, et al. Impact of telemedicine intensive care unit
radiologic, and serum marker as prognostic factors after severe head injury. coverage on patient outcomes: a systematic review and meta-analysis. Arch
J Trauma 1999;47(6):1126–30. Intern Med 2011;171(6):498–506
131. Raabe A, Grolms C, Seifert V. Serum markers of brain damage and 158. Gomez EJ, Hernando Perez ME, Vering T, et al. The INCA system: a further
outcome prediction in patients after severe head injury. Br J Neurosurg step towards a telemedical artificial pancreas. IEEE Trans Inf Technol
1999;13(1):56–9. Biomed 2008;12(4):470–9.
479.e4 Section VII—Computers, Engineering, and the Future

159. Gamble KH. Critical care network. ICU telemedicine can help ease the 162. Thursby JG, Thursby MC. Research and development: where is the new
burden of caring for critically ill patients–provided all the right pieces are science in corporate R&D? Science 2006;8(5805):1547–8.
in place. Healthcare Inform 2009;26(12):26–30. 163. Lundberg N. Continuous recording and control of ventricular fluid
160. Mayevsky A, Chance B. Oxidation-reduction states of NADH in vivo: from pressure in neurosurgical practice. Acta Psychiatr Scand Suppl 1960;36:
animals to clinical use. Mitochondrion 2007;7(5):330–9. 1–193.
161. Small business innovation research (SBIR) and small business technology
transfer (STTR) programs. Available at http://grants.nih.gov/grants/
funding/sbirs. Accessed 2008.

S-ar putea să vă placă și