Sunteți pe pagina 1din 21

Intensive Care Med (2014) 40:1189–1209

DOI 10.1007/s00134-014-3369-6 CO NFERENCE REPORTS AND EXPERT PANEL

Peter Le Roux
David K. Menon
Consensus summary statement
Giuseppe Citerio of the International Multidisciplinary
Paul Vespa
Mary Kay Bader Consensus Conference on Multimodality
Gretchen M. Brophy
Michael N. Diringer Monitoring in Neurocritical Care
Nino Stocchetti
Walter Videtta A statement for healthcare professionals from the
Rocco Armonda Neurocritical Care Society and the European Society
Neeraj Badjatia of Intensive Care Medicine
Julian Böesel
Randall Chesnut
Sherry Chou
Jan Claassen
Marek Czosnyka
Michael De Georgia
Anthony Figaji
Jennifer Fugate
Raimund Helbok
David Horowitz
Peter Hutchinson
Monisha Kumar
Molly McNett
Chad Miller
Andrew Naidech
Mauro Oddo
DaiWai Olson
Kristine O’Phelan
J. Javier Provencio
Corinna Puppo
Richard Riker
Claudia Robertson
Michael Schmidt
Fabio Taccone

Care Medicine (ESICM), the Society for


Received: 26 May 2014 Critical Care Medicine (SCCM) and the P. Le Roux ())
Accepted: 7 June 2014 Latin America Brain Injury Consortium and Brain and Spine Center, Suite 370,
Published online: 20 August 2014 is being simultaneously published in Medical Science Building,
 Springer-Verlag Berlin Heidelberg and Neurocritical Care and Intensive Care Lankenau Medical Center,
ESICM 2014 Medicine. It will be published in Intensive 100 East Lancaster Avenue,
Care Medicine in an abridged version. Wynnewood, PA 19096, USA
The Neurocritical Care Society affirms the
value of this consensus statement as an e-mail: lerouxp@mlhs.org
Electronic supplementary material
educational tool for clinicians. Tel.: ?1 610 642 3005
The online version of this article
(doi:10.1007/s00134-014-3369-6) contains
This article is being simultaneously supplementary material, which is available D. K. Menon
published in Neurocritical Care and to authorized users. Neurosciences Critical Care Unit,
Intensive Care Medicine. Division of Anaesthesia,
University of Cambridge Consultant,
This article is being simultaneously Addenbrooke’s Hospital, Box 93,
published in Neurocritical Care and Cambridge CB2 2QQ, UK
Intensive Care Medicine.This article is
endorsed by the Neurocritical Care Society e-mail: dkm13@wbic.cam.ac.uk
(NCS), the European Society of Intensive
1190

G. Citerio S. Chou A. Naidech


NeuroIntensive Care Unit, Department of Department of Neurology, Brigham and Department of Neurology, Northwestern
Anesthesia and Critical Care, Ospedale San Women’s Hospital, 75 Francis Street, University Feinberg, SOM 710, N Lake
Gerardo, Via Pergolesi 33, 20900 Monza, Italy Boston, MA 02115, USA Shore Drive, 11th floor, Chicago, IL 60611,
e-mail: g.citerio@hsgerardo.org USA
J. Claassen
P. Vespa Neurological Intensive Care Unit, Columbia M. Oddo
David Geffen School of Medicine at UCLA, University College of Physicians and Surgeons, Department of Intensive Care Medicine,
Los Angeles, CA 90095, USA 177 Fort Washington Avenue, Milstein 8 Faculty of Biology and Medicine University
e-mail: PVespa@mednet.ucla.edu Center room 300, New York, NY 10032, USA of Lausanne, CHUV University Hospital,
BH 08-623, 1011 Lausanne, Switzerland
M. K. Bader M. Czosnyka
Neuro/Critical Care CNS, Mission Hospital, Department of Neurosurgery, University of D. Olson
Mission Viejo, CA 92691, USA Cambridge, Addenbrooke’s Hospital, Box Neurology, Neurotherapeutics and
e-mail: Marykay.Bader@stjoe.org 167, Cambridge CB2 0QQ, UK Neurosurgery, University of Texas
Southwestern, 5323 Harry Hines Blvd,
G. M. Brophy M. De Georgia Dallas, TX 75390-8897, USA
Virginia Commonwealth University, Medical Neurocritical Care Center, Cerebrovascular
College of Virginia Campus, 410N. 12th Center, University Hospital Case Medical K. O’Phelan
Street, Richmond, VA 23298-0533, USA Center, Case Western Reserve University Department of Neurology, University of
e-mail: gbrophy@vcu.edu School of Medicine, 11100 Euclid Avenue, Miami, Miller School of Medicine, JMH,
Cleveland, OH 44106, USA 1611 NW 12th Ave, Suite 405, Miami, FL
M. N. Diringer 33136, USA
Neurocritical Care Section, Department of A. Figaji
Neurology, Washington University, Campus University of Cape Town, 617 Institute for J. J. Provencio
Box 8111, 660 S Euclid Ave, St Louis, MO Child Health, Red Cross Children’s Hospital, Cerebrovascular Center and
63110, USA Rondebosch, Cape Town 7700, South Africa Neuroinflammation Research Center,
e-mail: diringerm@neuro.wustl.edu Lerner College of Medicine, Cleveland
J. Fugate Clinic, 9500 Euclid Ave, NC30, Cleveland,
N. Stocchetti Mayo Clinic, Rochester, MN 55905, USA OH 44195, USA
Department of Physiopathology and
Transplant, Milan University, Neuro ICU, R. Helbok C. Puppo
Fondazione IRCCS Cà Granda Ospedale Neurocritical Care Unit, Department of Intensive Care Unit, Hospital de Clinicas,
Maggiore Policlinico, Via F Sforza, 35, Neurology, Innsbruck Medical University, Universidad de la República, Montevideo,
20122 Milan, Italy Anichstr.35, 6020 Innsbruck, Austria Uruguay
e-mail: stocchet@policlinico.mi.it
D. Horowitz R. Riker
W. Videtta University of Pennsylvania Health System, Critical Care Medicine, Maine Medical
ICU Neurocritical Care, Hospital Nacional 3701 Market Street, Philadelphia, PA Center, 22 Bramhall Street, Portland, Maine
‘Prof. a. Posadas’, El Palomar, Pcia de 19104, USA 04102-3175, USA
Buenos Aires, Argentina
P. Hutchinson C. Robertson
R. Armonda Department of Clinical Neurosciences, Department of Neurosurgery, Center for
Department of Neurosurgery, MedStar University of Cambridge, Addenbrooke’s Neurosurgical Intensive Care, Ben Taub
Georgetown University Hospital, Medstar Hospital, Box 167, Cambridge CB2 2QQ, Hospital, Baylor College of Medicine, 1504
Health, 3800 Reservoir Road NW, UK Taub Loop, Houston, TX 77030, USA
Washington, DC 20007, USA
M. Kumar M. Schmidt
N. Badjatia Department of Neurology, Perelman School Columbia University College of Physicians
Department of Neurology, University of of Medicine, University of Pennsylvania, 3 and Surgeons, Milstein Hospital 8 Garden
Maryland Medical Center, 22 S Greene St, West Gates, 3400 Spruce Street, South, Suite 331, 177 Fort Washington
Baltimore, MD 21201, USA Philadelphia, PA 19104, USA Avenue, New York, NY 10032, USA

J. Böesel M. McNett F. Taccone


Department of Neurology, Ruprecht-Karls Nursing Research, The MetroHealth Laboratoire de Recherche Experimentale,
University, Hospital Heidelberg, Im System, 2500 MetroHealth Drive, Department of Intensive Care, Erasme
Neuenheimer Feld 400, 69120 Heidelberg, Cleveland, OH 44109, USA Hospital, Route de Lennik, 808, 1070
Germany Brussels, Belgium
C. Miller
R. Chesnut Division of Cerebrovascular Diseases and
Harborview Medical Center, University of Neurocritical Care, The Ohio State Abstract Neurocritical care
Washington, Mailstop 359766, 325 Ninth University, 395W. 12th Ave, 7th Floor, depends, in part, on careful patient
Ave, Seattle, WA 98104-2499, USA Columbus, OH 43210, USA monitoring but as yet there are little
1191

data on what processes are the most research, publication record, and Strong consideration was given to
important to monitor, how these expertise. They undertook a system- providing pragmatic guidance and
should be monitored, and whether atic literature review to develop recommendations for bedside neuro-
monitoring these processes is cost- recommendations about specific top- monitoring, even in the absence of
effective and impacts outcome. At the ics on physiologic processes high quality data.
same time, bioinformatics is a rapidly important to the care of patients with
emerging field in critical care but as disorders that require neurocritical Keywords
yet there is little agreement or stan- care. This review does not make Consensus development conference 
dardization on what information is recommendations about treatment, Grading of Recommendations
important and how it should be dis- imaging, and intraoperative monitor- Assessment Development and
played and analyzed. The ing. A multidisciplinary jury, selected Evaluation (GRADE) 
Neurocritical Care Society in collab- for their expertise in clinical investi- Brain metabolism  Brain oxygen 
oration with the European Society of gation and development of practice Clinical trials  Intracranial pressure 
Intensive Care Medicine, the Society guidelines, guided this process. The Microdialysis 
for Critical Care Medicine, and the GRADE system was used to develop Multimodal monitoring 
Latin America Brain Injury Consor- recommendations based on literature Neuromonitoring 
tium organized an international, review, discussion, integrating the Traumatic brain injury 
multidisciplinary consensus confer- literature with the participants’ col- Brain physiology  Bioinformatics 
ence to begin to address these needs. lective experience, and critical review Biomarkers  Neurocritical care 
International experts from neurosur- by an impartial jury. Emphasis was Clinical guidelines
gery, neurocritical care, neurology, placed on the principle that recom-
critical care, neuroanesthesiology, mendations should be based on both
nursing, pharmacy, and informatics data quality and on trade-offs and
were recruited on the basis of their translation into clinical practice.

Introduction pressure supports the management of critically ill


neurological patients. The use of these monitoring
The Neurocritical Care Society (NCS) in collaboration modalities has become routine despite limited level I
with the European Society of Intensive Care Medicine evidence to support their use. It is not our intention to
(ESICM), the Society for Critical Care Medicine make recommendations for such monitoring, except
(SCCM), and the Latin America Brain Injury Consortium where such recommendations are directly relevant to
(LABIC) commissioned a consensus conference on clinical care of the injured or diseased nervous system.
monitoring patients with acute neurological disorders that 2. We accept that imaging is indispensable in the
require intensive care management. diagnosis and management of the critically ill patient
Patient monitoring using some, many, or all of the with neurological disease, perhaps more so than any
techniques outlined in this consensus document is rou- other area of intensive care medicine. However, with a
tinely performed in most neurocritical care units (NCCU) few exceptions we have elected not to focus on
on patients with acute neurological disorders who require imaging but rather will concentrate on bedside tools
critical care. In many institutions the combined use of that can be used in the intensive care unit (ICU).
multiple monitors is common, a platform often termed 3. It is not our intent to discuss or recommend therapy in
‘‘multimodality monitoring’’ (MMM). The use of such any of the settings we address. This may seem to be a
tools to supplement the clinical examination is predicated somewhat arbitrary distinction, but the distinction
by the insensitivity of the neurologic examination to allows us to focus our questions on the act of
monitor for disease progression in patients in whom the monitoring rather than the act of treatment. It must
clinical features of disease are confounded by the effects be recognized that no monitor in the end will change
of sedation, analgesia, and neuromuscular blockade, or in outcome. Instead it is how that information is inter-
deeply comatose patients (e.g., malignant brain edema, preted and integrated into clinical decision-making and
seizures, and brain ischemia) where neurological then how the patient is treated that will influence
responses approach a minimum and become insensitive to outcome. For many of the processes monitored,
clinical deterioration. Several considerations frame our effective treatments have still to be fully elucidated
subsequent discussion: or remain empiric rather than mechanistic. In this
context, monitoring can be valuable in learning about
1. As with general intensive care, basic monitoring such pathophysiology after acute brain injury (ABI) and
as electrocardiography, pulse oximetry, and blood potentially help identify new therapies.
1192

4. The purpose of this consensus document is to provide clinical situation in individual patients on the basis of
evidence-based recommendations about monitoring in clinical judgment and resource availability. We therefore
neurocritical care patients, and to base these recommen- recognize that, while our recommendations provide
dations on rigorously evaluated evidence from the useful guidance, they cannot be seen as mandatory for all
literature. However, we also recognize that, in many individual clinician–patient interactions.
cases, the available evidence is limited for several reasons:
Given this background, and recognizing the clinical
equipoise for most of the brain monitors that will be
(a) Some monitors have strong anecdotal evidence of discussed, we assess basic questions about monitoring
providing benefit, and formal randomized evalua- patients with acute brain disorders who require critical
tion is limited by real or perceived ethical concerns care. Our recommendations for monitoring are based on a
about withholding potentially life-saving monitors systematic literature review, a robust discussion during
with an outstanding safety record. the consensus conference about the interpretation of the
(b) Important physiological information obtained literature, the collective experience of the members of the
from several monitors may translate into outcome group, and review by an impartial, international jury.
differences in select patients, but this benefit is not
universal and is diluted by the patients in whom
such effects are not seen. However, we still do not Process
have a clear basis for identifying the cohorts in
whom such benefit should be assessed. A fundamental goal in the critical care management of
(c) The process by which we identify treatment thresh- patients with neurological disorders is identification, pre-
olds based on monitoring and the process to integrate vention, and treatment of secondary cerebral insults that are
multiple monitors are still being elucidated. known to exacerbate outcome. This strategy is based on a
5. The monitoring tools we discuss fall into several variety of monitoring techniques that includes the neuro-
categories, and their nature and application predicate logical examination, imaging, laboratory analysis, and
how discussion of their utility is framed. Some of these physiological monitoring of the brain and other organ sys-
tools [e.g., intracranial pressure (ICP), brain oximetry, tems used to guide therapeutic interventions. The reasons
and microdialysis] meet the definition of bedside mon- why we monitor patients with neurological disorders are
itors, and are assessed in terms of their accuracy, safety, listed in Table 1. In addition rather than focus on individual
indications, and impact on prognostication, manage- devices we chose to review physiological processes that are
ment, and outcome. However, other tools (e.g., important to neurocritical care clinicians (Table 2). Each of
biomarkers and tests of hemostasis) are used intermit- these topics is further reviewed in individual sections con-
tently, and are best dealt with in a different framework. tained in the electronic supplementary information (ESM)
Our choice of review questions addresses this difference. and in a supplement to Neurocritical Care. The reader is
6. In addition to the discussion of individual monitors we referred to these sections for further details about the review
also include some correlative essays on the use of process, evidence to support the recommendations in this
monitoring in emerging economies, where we attempt summary document, and additional citations for each topic.
to identify how our recommendations might be applied Representatives of the NCS and ESICM respectively
under conditions where there are limited resources. chaired the review and recommendation process. Experts
This discussion also provides a useful framework for from around the world in the fields of neurosurgery,
minimum standards of monitoring and assessment of
the effects in a wider conversation. Table 1 Reasons why we monitor patients with neurologic dis-
7. This issue also includes two other correlative essays. orders who require critical care
One focuses on metrics for processes and quality of
care in neurocritical care that provides an organiza- Detect early neurological worsening before irreversible brain
tional context for the recommendations that we make. damage occurs
Individualize patient care decisions
Finally, we provide a separate discussion on the Guide patient management
integration of MMM, which draws on the rapid Monitor the physiologic response to treatment and to avoid any
advances in bioinformatics and data processing cur- adverse effects
rently available. In each of these cases we recognize Allow clinicians to better understand the pathophysiology of
that the field is currently in a state of flux, but have complex disorders
Design and implement management protocols
elected to provide some recommendations in line with Improve neurological outcome and quality of life in survivors of
the data currently available. severe brain injuries
8. The intent of this consensus statement is to assist Through understanding disease pathophysiology begin to develop
clinicians in decision-making. However, we recognize new mechanistically oriented therapies where treatments
currently are lacking or are empiric in nature
that this information must be targeted to the specific
1193

Table 2 Physiological processes that are important to neurocritical • Very low Any estimate of effect is very uncertain.
care clinicians that were selected for review in the International
Multidisciplinary Consensus Conference on Multimodality Moni- The GRADE system classifies recommendations as
toring in Neurocritical Care strong or weak, according to the balance among benefits,
Topic section
risks, burden, and cost, and according to the quality of
evidence. Keeping those components separate constitutes
Clinical evaluation a crucial and defining feature of this grading system. An
Systemic hemodynamics advantage of the GRADE system is that it allows for
Intracranial pressure and cerebral perfusion pressure strong recommendations in the setting of lower quality
Cerebrovascular autoregulation
Systemic and brain oxygenation evidence and therefore is well suited to the intended
Cerebral blood flow and ischemia monitoring questions. Recommendations are stated as
Electrophysiology either strong (‘‘we recommend’’) or weak (‘‘we suggest’’)
Cerebral metabolism and based on the following:
Glucose and nutrition
Hemostasis and hemoglobin
Temperature and inflammation • The trade-offs, taking into account the estimated size of
Biomarkers of cellular damage and degeneration the effect for the main outcomes, the confidence limits
ICU processes of care around those estimates, and the relative value placed on
Multimodality monitoring informatics integration,
display and analysis each outcome
Monitoring in emerging economies • Quality of the evidence
Future directions and emerging technologies • Translation of the evidence into practice in a specific
setting, taking into consideration important factors that
neurocritical care, neurology, critical care, neuroanesthesiol- could be expected to modify the size of the expected effects.
ogy, nursing, pharmacy, and informatics were recruited on the
basis of their expertise and publication record related to each Each topic was then presented and discussed at a 2-day
topic. Two authors were assigned to each topic and efforts conference in Philadelphia held on September 29 and 30,
were made to ensure representation from different societies, 2013. The chairs, co-chairs, jury, and each author attended
countries, and disciplines (Appendix 1 ESM). The review and the meeting. In addition representatives from each of the
recommendation process, writing group, and topics were endorsing organizations were invited and 50 additional
reviewed and approved by the NCS and ESICM. A jury of attendees with expertise in neurocritical care were allowed
experienced neurocritical care clinicians (physicians, a nurse, to register as observers. Industry representatives were not
and a pharmacist) was selected for their expertise in clinical allowed to participate. Each author presented a summary of
investigation and development of practice guidelines. the data and recommendations to the jury and other partic-
The authors assigned to each topic performed a critical ipants. Presentations were followed by discussion focused
literature review with the help of a medical librarian on refining the proposed recommendations for each topic.
according to the Preferred Reporting Items for Systematic Approximately one-third of the conference time was used
Reviews and Meta-Analyses (PRISMA) statement [1]. The for discussion. The jury subsequently held several confer-
review period included January 1980–September 2013 and ence calls, and then met again at a subsequent 2-day meeting
was limited to clinical articles that included more than five to review and abstract all manuscripts and finalize the
subjects and were published in English. The focus was on summary consensus statement presented here. They
adult patients and brain disorders. The literature findings reviewed selected key studies, the recommendations made
were summarized in tables and an initial summary that by the primary reviewers, and the discussion that took place
included specific recommendations was prepared. The at the conference. Strong consideration was given to pro-
chairs, co-chairs, and jury members, each assigned to spe- viding guidance and recommendations for bedside
cific topics as a primary or secondary reviewer, reviewed neuromonitoring, even in the absence of high quality data.
these drafts. The quality of the data was assessed and rec-
ommendations developed using the GRADE system [2–
10]. The quality of the evidence was graded as: Caveats and limitations to the process

• High Further research is very unlikely to change our The setting of these recommendations, monitoring, makes
confidence in the estimate of effect. it difficult to use all of the normal considerations used to
• Moderate Further research is likely to have an impor- make decisions about the strength of recommendations,
tant impact on our confidence in the estimate of effect typically of a treatment [4], which include the balance
and may change the estimate. between desirable and undesirable effects, estimates of
• Low Further research is very likely to have an effect based on direct evidence, and resource use since
important impact on our confidence in the estimate of monitoring has no proximate effects on outcome. Instead
effect and is likely to change the estimate. it typically modifies treatment and can only influence
1194

outcome through such modulation. Our confidence in the Summary


estimate of effects in most analyses was not derived from
methodologically rigorous studies, because few such All clinical scales of consciousness should account for the
studies exist, but often driven by epidemiological studies effects of sedation and neuromuscular blockade. Inter-
and investigations of clinical physiology, which usually rater reliability assessments of the GCS report a range of
provided indirect evidence, with several potential kappa scores, but the GCS is a strong prognostic marker
confounders. and indicator of need for surgery in traumatic brain injury
Given these limitations, decisions on recommenda- (TBI) [11], of clinical outcome in posterior circulation
tions are driven by an expectation of values and stroke [12], and following cardiac arrest [13]. In isolation,
preferences. Given the limited outcome data of both the GCS is disadvantaged by the confounders produced
benefit and harm associated with neuromonitoring, we by endotracheal intubation, and by the lack of measure-
relied on inferences from observational studies and ment of pupillary responses (which are strong predictors
extrapolation from pathophysiology to estimate the effect of outcome). However, the prognostic information pro-
and effect size of potential benefit and harm. We con- vided by pupillary responses can be integrated with the
cluded that the avoidance of permanent neurological GCS to provide greater specificity of outcome prediction
deficit would be the dominant driver of patient choice. [14]. Newer devices provide objective measurement of
Given that the diseases and disease mechanisms we pupillary diameter, and the amount and speed of pupillary
monitor are known to be damaging, and given that the response, but additional research is necessary to confirm
time available for intervention is limited, we made these the role of these devices in caring for brain-injured
extrapolations unless there was real concern about benefit patients.
or evidence of harm. This approach to deciding on rec- Sedation, potent analgesics (e.g., opioids), and neu-
ommendations was universally adopted by all members of romuscular blockade remain a problem for any clinical
the multispecialty, multidisciplinary, multinational panel. scale of consciousness. However, in the non-sedated (or
Though there was some variation in initial opinions, lightly sedated but responsive) patient, the recently
careful consideration of the available evidence and devised FOUR score, which measures ocular (as well as
options resulted in relatively tightly agreed consensus on limb) responses to command and pain, along with pupil-
recommendations. lary responses and respiratory pattern [15], may provide a
more complete assessment of brainstem function. Volume
assist ventilator modes may confound differentiation
Summary of recommendations from the individual between the two lowest scores of the respiratory compo-
consensus conference topics nent of the FOUR score. The FOUR score has been shown
to have good inter-rater reliability [16] and prognostic
Clinical evaluation content in a range of neurological conditions, and may
Questions addressed show particularly good discrimination in the most unre-
sponsive patients. However, experience with this
1. Should assessments with clinical coma scales be instrument is still limited when compared to the GCS.
routinely performed in comatose adult patients with Current evidence suggests that both the GCS and FOUR
ABI? score provide useful and reproducible measures of neu-
2. For adult comatose patient with ABI, is the Glasgow rological state, and can be routinely used to chart trends in
Coma Scale (GCS) or the Full Outline of Unrespon- clinical progress.
siveness (FOUR) score more reliable in the clinical Brain-injured patients in NCCU are known to expe-
assessment of coma? rience more significant pain than initially presumed [17].
3. Which pain scales have been validated and shown to While any level of neurological deficit can confound
be reliable among patients with brain injuries who assessment of pain and agitation, perhaps a greater barrier
require neurocritical care? arises from perceptions of clinicians who feel that such
4. Which pain scales have been validated and shown to assessments are simply not possible in such patient pop-
be reliable among patients with severe disorders of ulations. In actual fact, up to 70 % of neurocritical care
consciousness [minimally conscious state (MCS) and patients can assess their own pain using a self-reporting
unresponsive wakefulness syndrome (UWS)]? tool such as the Numeric Rating Scale (NRS), while cli-
5. Which ‘‘sedation’’ scales are valid and reliable in nician rated pain using the Behavioral Pain Scale (BPS) is
brain-injured patients who require neurocritical care? assessable in the remainder. Assessing pain in patients
6. What other sedation strategies may lead to improved with severe disorders of consciousness such as vegetative
outcomes for brain-injured patients? state (VS) and minimally conscious state (MCS) is a
7. Which delirium scales are valid and reliable in brain- greater challenge, but is possible with Nociception Coma
injured patients who require neurocritical care? Scale-revised (NCS-R) [18].
1195

The assessment of sedation in the context of brain 5. We recommend monitoring sedation with a validated
injury is challenging, since both agitation and apparent and reliable scale such as the SAS or RASS. (Strong
sedation may be the consequence of the underlying neu- recommendation, low quality of evidence.)
rological state, rather than simply a marker of suboptimal 6. We recommend against performing sedation interrup-
sedation. However, both the Richmond Agitation Seda- tion or wake-up tests among brain-injured patients
tion Scale (RASS) and the Sedation-Agitation Scale with intracranial hypertension, unless benefit out-
(SAS) [19] provide workable solutions in some patients. weighs the risk. (Strong recommendation, low
‘‘Wake-up tests’’ in patients with unstable intracranial quality of evidence.)
hypertension pose significant risks and often may lead to 7. We suggest assessment of delirium among neurocrit-
physiological decompensation [20], and show no proven ical care patients include a search for new neurologic
benefits in terms of in duration of mechanical ventilation, insults as well as using standard delirium assessment
length of ICU and hospital stay, or mortality. However we tools. (Weak recommendation, low quality of
recognize that in some patients (e.g., those with aneu- evidence.)
rysmal subarachnoid hemorrhage (SAH) requiring 8. We recommend attention to level of wakefulness, as
neurological assessment) a balance will need to be struck used in the ICDSC, during delirium screening to avoid
between the information gained from clinical evaluation confounding due to residual sedative effect. (Strong
and risk of physiological decompensation with a wake-up recommendation, low quality of evidence.)
test. In such circumstances, the benefit of a full neuro-
logical assessment may be worth a short period of modest
ICP elevation. The Confusion Assessment Method for the Systemic hemodynamics
ICU (CAM-ICU) or the Intensive Care Delirium
Questions addressed
Screening Checklist (ICDSC) was strongly recommended
for delirium assessment by the 2013 PAD Guidelines 1. What hemodynamic monitoring is indicated in patients
[19]. While delirium assessment has been reported in with ABI?
stroke [21], generalizability of this data is limited, and 2. What hemodynamic monitoring is indicated to diag-
even within this study, as the majority of patients were nose and support the management of unstable or at-
unassessable. The ICDSC may be preferred since it does risk patients?
not score changes in wakefulness and attention directly
attributable to recent sedative medication as positive IC-
DSC points. It is important to emphasize that a diagnosis Summary
of delirium in a neurocritical care patient may represent
evidence of progress of the underlying disease, and must Cardiopulmonary complications are common after ABI,
prompt an evaluation for a new neurologic deficit or and have a significant impact on clinical care and patient
specific neurologic process. outcome [22–26]. Among several hypotheses, the main
mechanism of cardiac injury following ABI (e.g., SAH) is
related to sympathetic stimulation and catecholamine
release [27–29]. All patients with ABI admitted to the
Recommendations ICU require basic hemodynamic monitoring of blood
1. We recommend that assessments with either the GCS pressure, heart rate, and pulse oximetry. Some stable
(combined with assessment of pupils) or the FOUR patients will require nothing more than this, but many will
score be routinely performed in comatose adult need more invasive and/or sophisticated hemodynamic
patients with ABI. (Strong recommendation, low monitoring. Monitoring of systemic hemodynamics con-
quality of evidence.) tributes to understanding the mechanisms of circulatory
2. We recommend using the NRS 0–10 to elicit patient’s failure, and detecting or quantifying inadequate perfusion
self-report of pain in all neurocritical care patients or organ dysfunction. Although there is limited evidence,
wakeful enough to attempt this. (Strong recommenda- cardiac output should be monitored (invasively or non-
tion, low quality of evidence.) invasively) in those patients with myocardial dysfunction
3. We recommend in the absence of a reliable NRS or hemodynamic instability [30]. Whether this also
patient self-report, clinicians use a behavior-based applies to patients on vasopressors to augment cerebral
scale to estimate patient pain such as the BPS or perfusion pressure (CPP) rather than for hemodynamic
CCPOT. (Strong recommendation, low quality of instability should be decided on a case-by-case basis. The
evidence.) various hemodynamic devices available have differing
4. We recommend use of the revised NCS-R to estimate technical reliability, clinical utility, and caveats, but
pain for patients with severely impaired consciousness limited studies are available in acute brain-injured
such as VS or MCS, using a threshold score of 4. patients. Baseline assessment of cardiac function with
(Strong recommendation, low quality of evidence.) echocardiography may be a useful approach when there
1196

are signs of cardiac dysfunction. Methods for evaluation associated with the variability in how mean arterial
of fluid responsiveness are similar to the ones used in the pressure (MAP) is measured to determine CPP [37] and
general ICU population. depend on disease state. In addition, management strate-
gies based on population targets for CPP rather than ICP
have not enhanced outcome [38], and rather than a single
Recommendations threshold optimal CPP, values may need to be identified
1. We recommend the use of electrocardiography and for each individual [39]. There are several devices
invasive monitoring of arterial blood pressure in all available to measure ICP; intraparenchymal monitors or
unstable or at-risk patients in the ICU. (Strong Rec- ventricular catheters are the most reliable and accurate,
ommendation, moderate quality of evidence.) but for patients with hydrocephalus a ventricular catheter
2. We recommend that hemodynamic monitoring be used is preferred. The duration of ICP monitoring varies
to establish goals that take into account cerebral blood according to the clinical context.
flow (CBF) and oxygenation. These goals vary Recently, our core beliefs in ICP have been challenged
depending on diagnosis and disease stage. (Strong by the BEST-TRIP trial [40]. While this study has high
recommendation, moderate quality of evidence.) internal validity, it lacks external validity and so the
3. We recommend the use of additional hemodynamic results cannot be generalized. Furthermore, the trial
monitoring (e.g., intravascular volume assessment, evaluated two treatment strategies for severe TBI, one
echocardiography, cardiac output monitors) in selected triggered by an ICP monitor and the other by the clinical
patients with hemodynamic instability. (Strong rec- examination and imaging rather than the treatment of
ommendation, moderate quality of evidence.) intracranial hypertension. In this context it must be
4. We suggest that the choice of technique for assessing emphasized that clinical evaluation and diagnosis of ele-
pre-load, after-load, cardiac output, and global sys- vated ICP was fundamental to all patients in BEST-TRIP,
temic perfusion should be guided by specific evidence and hence the study reinforces that evaluation and mon-
and local expertise. (Weak recommendation, moderate itoring, either by a specific monitor or by an
quality of evidence.) amalgamation of clinical and imaging signs, is standard of
care.
ICP treatment is important and is best guided by ICP
Intracranial pressure and cerebral perfusion pressure monitoring, clinical imaging, and clinical evaluation used
in combination and in the context of a structured protocol
Questions addressed [41–43]. We recognize that this may vary across different
1. What are the indications for monitoring ICP and CPP? diagnoses and different countries. Today, a variety of
2. What are the principal methods of reliable, safe, and other intracranial monitoring devices are available, and
accurate ICP and CPP monitoring? ICP monitoring is a mandatory prerequisite when other
3. What is the utility of ICP and CPP monitoring for intracranial monitors are used, to provide a framework for
prognosis in the comatose TBI patient? optimal interpretation.

Summary Recommendations
1. ICP and CPP monitoring are recommended as a part of
Monitoring of ICP and CPP is considered to be funda- protocol-driven care in patients who are at risk of
mental to the care of patients with ABI, particularly those elevated intracranial pressure based on clinical and/or
in coma, and is routinely used to direct medical and imaging features. (Strong recommendation, moderate
surgical therapy [31]. ICP and CPP monitoring are most quality of evidence.)
frequently studied in TBI, but can play a similar role in 2. We recommend that ICP and CPP monitoring be used
conditions such as SAH and ICH among other disorders. to guide medical and surgical interventions and to
Increased ICP, and particularly that refractory to treat- detect life-threatening imminent herniation; however,
ment, is a well-described negative prognostic factor, the threshold value of ICP is uncertain on the basis of
specifically for mortality [32–34]. There are well-estab- the literature. (Strong recommendation, high quality of
lished indications and procedural methods for ICP evidence.)
monitoring, and its safety profile is excellent [35]. The 3. We recommend that the indications and method for
threshold that defines intracranial hypertension is uncer- ICP monitoring should be tailored to the specific
tain but generally is considered to be greater than diagnosis (e.g., SAH, TBI, encephalitis). (Strong
20–25 mmHg, although both lower and higher thresholds recommendation, low quality of evidence.)
are described [36]. The recommendations for an optimal 4. While other intracranial monitors can provide useful
CPP have changed over time and may in part be information, we recommend that ICP monitoring be
1197

used as a prerequisite to allow interpretation of data permit us to conclusively recommend one approach in
provided by these other devices. (Strong recommen- preference to another.
dation, moderate quality of evidence.) In broad terms, the preservation or absence of pressure
5. We recommend the use of standard insertion and autoregulation can influence blood pressure management
maintenance protocols to ensure safety and reliability following brain injury. Patients who show preserved
of the ICP monitoring procedure. (Strong recommen- autoregulation may benefit from higher mean arterial and
dation, high quality of evidence.) CPP as part of an integrated management scheme for ICP
6. Both parenchymal ICP monitors and external ventric- control, while those who show pressure passive responses
ular catheters (EVD) provide reliable and accurate data may be better served by judicious blood pressure control.
and are the recommended devices to measure ICP. In the Critical autoregulatory thresholds for survival and favor-
presence of hydrocephalus, use of an EVD when safe able neurological outcome may be different, and may
and practical is preferred to parenchymal monitoring. vary with age and sex. The brain may be particularly
(Strong recommendation, high quality of evidence.) vulnerable to autoregulatory dysfunction during rewarm-
7. We recommend the continuous assessment and mon- ing after hypothermia and within the first days following
itoring of ICP and CPP including waveform quality injury [46].
using a structured protocol to ensure accuracy and More refined monitoring of autoregulatory efficiency
reliability. Instantaneous ICP values should be inter- is now possible through online calculation of derived
preted in the context of monitoring trends, CPP, and indices such as the pressure reactivity index (PRx) [45].
clinical evaluation. (Strong recommendation, high About two-thirds of TBI patients have an optimum CPP
quality of evidence.) range (CPPopt) where their autoregulatory efficiency is
8. While refractory ICP elevation is a strong predictor of maximized, and that management at or close to CPPopt is
mortality, ICP per se does not provide a useful associated with better outcomes [47]. The safety of
prognostic marker of functional outcome; therefore, titrating therapy to target CPPopt requires further study,
we recommend that ICP not be used in isolation as a and validation in a formal clinical trial before it can be
prognostic marker. (Strong recommendation, high recommended.
quality of evidence.)

Recommendations
Cerebral autoregulation
1. We suggest that monitoring and assessment of auto-
Questions addressed
regulation may be useful in broad targeting of cerebral
1. Does monitoring of cerebral autoregulation help guide perfusion management goals and prognostication in
management and contribute to prognostication? ABI. (Weak recommendation, moderate quality of
2. Which technique and methodology most reliably evidence.)
evaluates the state of autoregulation in ABI? 2. Continuous bedside monitoring of autoregulation is
now feasible, and we suggest that should be considered
as a part of MMM. Measurement of pressure reactivity
Summary has been commonly used for this purpose, but many
different approaches may be equally valid. (Weak
Pressure autoregulation is an important hemodynamic recommendation, moderate quality of evidence.)
mechanism that protects the brain against inappropriate
fluctuations in CBF in the face of changing CPP. Both
static and dynamic autoregulation have been monitored in Systemic and brain oxygenation
neurocritical care to aid prognostication and contribute to
Questions addressed
individualizing optimal CPP targets in patients [44].
Failure of autoregulation is associated with a worse out- 1. What are the indications for brain and systemic oxy-
come in various acute neurological diseases [45]. For genation in neurocritical care patients?
monitoring, several studies have used ICP (as a surrogate 2. What are the principal methods of reliable and
of vascular caliber and reactivity), transcranial Doppler accurate brain oxygen monitoring?
ultrasound, and near-infrared spectroscopy (NIRS) to 3. What is the safety profile of brain oxygen monitoring?
continuously monitor the impact of spontaneous fluctua- 4. What is the utility of brain oxygen monitoring to
tions in CPP on cerebrovascular physiology, and determine prognosis in the comatose patient?
calculated derived variables of autoregulatory efficiency. 5. What is the utility of brain oxygen monitoring to direct
However, the inconsistent approaches to using such medical and surgical therapy?
devices to monitor autoregulation make comparison dif- 6. What is the utility of brain oxygen monitoring to
ficult, and there are no good comparative studies that improve neurological outcome?
1198

Summary ischemia is present when SjvO2 is less than 55 % [56], but


cannot reliably be assumed to be absent at higher values
Maintenance of adequate oxygenation is a critical objec- since regional abnormalities may not be detected [57].
tive of managing critically ill patients with neurological The majority of SjvO2 studies are in severe TBI patients
disorders. Assessing tissue oxygenation provides vital with limited studies in SAH, ICH, or ischemic stroke
information about oxygen supply and consumption in patients. SjvO2 values can guide therapy [58] but have not
tissue beds. Inadequate systemic and brain oxygen been shown to improve outcomes. NIRS has several
aggravates secondary brain injury. Multimodality brain limitations in adult use [59]. There are limited small
monitoring includes measuring oxygenation systemically observational studies with conflicting results about de-
and locally in the brain. Systemic oxygenation and carbon saturations related to cerebral perfusion, vasospasm, head
dioxide (CO2) can be measured invasively with blood gas positioning during impending herniation, pharmacologic
sampling and non-invasively with pulse oximetry and interventions, and changes in MAP/CPP. There are no
end-tidal CO2 devices. There is extensive research in the studies that demonstrate that data from NIRS use alone
general critical care population on safety and applicability can influence outcomes in adult neurocritical care.
of systemic oxygen and carbon dioxide monitoring. PaO2,
SaO2, and SpO2 are indicators of systemic oxygenation
and useful to detect oxygenation decreases. Periodic
Recommendations
measurements of PaO2 and SaO2 and continuous SpO2
measurements should be used to guide airway and ven- 1. We recommend systemic pulse oximetry in all patients
tilator management in patients who require neurocritical and end-tidal capnography in mechanically ventilated
care [48, 49]. PaCO2 is a reliable measurement of hyper- patients, supported by arterial blood gases measure-
or hypocapnia and is superior to ETCO2 monitoring. The ment. (Strong recommendation, high quality of
continuous monitoring of ETCO2 and periodic monitoring evidence.)
of PaCO2 assists in ventilator management [50]. The 2. We recommend monitoring brain oxygen in patients
optimal target values for PaO2, SaO2, and SpO2 specific to with or at risk of cerebral ischemia and/or hypoxia,
the NCCU patient population are still being elucidated. using brain tissue (PbtO2) or/and jugular venous bulb
Normoxemia and avoidance of hypoxemia and hyperox- oximetry (SjvO2)—the choice of which depends on
emia should be targeted. patient pathology. (Strong recommendation, low qual-
Brain oxygen measurements include two invasive ity of evidence.)
bedside techniques, brain parenchymal oxygen tension 3. We recommend that the location of the PbtO2 probe
(PbtO2) and jugular bulb oxygen saturation (SjvO2), or a and side of jugular venous oximetry depend on the
non-invasive bedside method, NIRS. Normal PbtO2 is diagnosis, the type and location of brain lesions, and
23–35 mmHg [51]. A PbtO2 threshold of less than technical feasibility. (Strong recommendation, low
20 mmHg represents compromised brain oxygen and is a quality of evidence.)
threshold at which to consider intervention. Decreases 4. While persistently low PbtO2 and/or repeated episodes
below this are associated with other markers of cerebral of jugular venous desaturation are strong predictors of
ischemia or cellular dysfunction although exact values mortality and unfavorable outcome, we recommend
vary slightly with the type of parenchymal monitor used that brain oxygen monitors be used with clinical
and should be interpreted on the basis of probe location indicators and other monitoring modalities for accurate
identified on a post-insertion CT [52, 53]. However, prognostication. (Strong recommendation, low quality
PbtO2 is not simply a marker of ischemia or CBF. PbtO2 of evidence.)
monitoring is safe and provides accurate data for up to 5. We suggest the use of brain oxygen monitoring to
10 days with measured responses to interventions (e.g., assist titration of medical and surgical therapies to
changes in CPP, ventilator targets, pharmacologic seda- guide ICP/CPP therapy, identify refractory intracranial
tion, and transfusion) and can be used to guide therapy hypertension and treatment thresholds, help manage
[54]. Observational studies suggest a potential benefit delayed cerebral ischemia, and select patients for
when PbtO2-guided therapy is added to a severe TBI second-tier therapy. (Weak recommendation, low
management protocol, but there remains clinical quality of evidence.)
equipoise.
SjvO2 values differ from PbtO2 in what is measured
and can be used to detect both ischemia and hyperemia. Cerebral blood flow
Positioning, clot formation on the catheter, and poor
Questions addressed
sampling technique can influence SjvO2 accuracy and
errors are common so making SjvO2 monitoring more 1. What are the indications for CBF monitoring?
difficult to use and less reliable than PbtO2 monitoring 2. Do the various CBF monitors reliably identify those
[55]. Normal SjvO2 is between 55 and 75 %. Cerebral patients at risk for secondary ischemic injury?
1199

3. What CBF neuromonitoring thresholds best identify vasospasm after aneurysmal SAH. (Weak recommen-
risk for ischemic injury? dation, moderate quality of evidence.)
4. Does use of CBF neuromonitoring improve outcomes 3. We suggest that TCCS is superior to TCD in the
for those patients at risk for ischemic injury? detection of angiographically proven vasospasm after
aneurysmal SAH. (Weak recommendation, low quality
of evidence.)
Summary 4. We suggest that TCD or TCCS monitoring can help
predict vasospasm after traumatic SAH. (Weak rec-
Measurement of CBF has long been used in experimental ommendation, very low quality of evidence.)
models to define thresholds for ischemia leading to 5. We suggest that a TDF probe may be used to identify
interest in monitoring CBF in patients, in large part patients with focal ischemic risk within the vascular
because ischemia can underlie secondary cerebral injury. territory of the probe. (Weak recommendation, very
In addition to radiographic methods (not covered here) low quality of evidence.)
several devices can be used at the patient’s bedside to 6. We suggest use of a TCD screening paradigm using
monitor for CBF changes. These radiographic studies, Lindegaard ratios or comparisons of bi-hemispheric
particularly PET, have demonstrated that cellular injury middle cerebral artery mean velocities to improve
often can occur in the absence of ischemia [60, 61]. sensitivity for identification of vasospasm-associated
Advances in our understanding of the pathophysiology of ischemic damage. (Weak recommendation, low qual-
TBI and ICH suggest, however, that traditional ischemic ity of evidence.)
thresholds may not always apply and CBF data should be 7. We suggest that TDF probes used to assess ischemic
coupled with measurements of metabolic demand. risk after aneurysmal SAH should be placed in the
Flow can be continually monitored in a single small vascular territory of the ruptured aneurysm. (Weak
region of brain using invasive thermal diffusion flowmetry recommendation, very low quality of evidence.)
(TDF) or, less commonly, laser Doppler flowmetry (LDF)
[62, 63]. The utility of these probes is limited by their invasive
nature, small field of view, and uncertainly as to where they Electrophysiology
should be placed. TDF use is limited by reduced reliability in
Questions addressed
patients with elevated systemic temperatures. There are few
data regarding ischemic thresholds for these devices. 1. What are the indications for electroencephalography
Blood flow in larger regions of brain can be estimated (EEG)?
by transcranial Doppler ultrasonography (TCD), although 2. What is the utility of EEG following convulsive status
accuracy may be limited by operator variability. TCD is epilepticus (cSE) and refractory status epilepticus?
primarily used to monitor for vasospasm following aneu- 3. What is the utility of EEG or evoked potentials (EPs)
rysmal SAH. TCD also can be used to identify TBI patients in patients with and without ABI, including cardiac
with hypoperfusion or hyperperfusion and so guide their arrest, and unexplained alteration of consciousness?
care. However there is a far greater body of literature 4. What is the utility of EEG to detect ischemia in
describing TCD use in SAH. TCD can predict angiographic patients with SAH or acute ischemic stroke (AIS)?
vasospasm with good sensitivity and specificity [64, 65] 5. Should scalp and/or intracranial EEG be added to
but is less accurate in predicting delayed ischemic neuro- patients undergoing invasive brain monitoring?
logical deficits [66]. Predictive power is improved with the
use of transcranial color-coded duplex sonography (TCCS)
[67]. Inclusion of the Lindegaard ratio [68] and the rate of Summary
the increase in velocities [69] in interpreting the data
improves performance. There are no published studies that Electroencephalography and EPs are the most frequently
demonstrate enhanced outcomes that result from imple- used electrophysiological techniques used in the ICU
mentation of a treatment strategy directed only by [70]. EEG provides information about brain electrical
neuromonitoring devices that assess CBF or ischemic risks. activity and it is essential to detect seizures, including
duration and response to therapy and can help outcome
prediction after coma [71–74]. Seizures are frequent with
and without ABI in the ICU, and are mostly nonconvul-
Recommendations sive. Further, some patients will have cyclic seizure
1. We recommend TCD or TCCS monitoring to predict patterns, which will only be detectable by continuously
angiographic vasospasm after aneurysmal SAH. (cEEG) recorded data [75]. However, data to support the
(Strong recommendation, high quality of evidence.) benefit of continuous over routine EEG recordings, typi-
2. We suggest that trends of TCD or TCCS can help cally no longer than 30-min duration (sometimes called
predict delayed ischemic neurological deficits due to spot EEG), to detect seizures is very limited. Routine
1200

EEG will miss nonconvulsive seizures (NCSz) in 2. What is the preferred location for a microdialysis
approximately half of those with seizures when compared probe?
to prolonged monitoring [76]. Advances in neuroimaging 3. What is the utility of cerebral microdialysis in
have limited the application of EPs in many ICUs, but in determining patient prognosis?
select patients EPs can help in outcome prediction. 4. What is the utility of cerebral microdialysis in guiding
The optimal montage and number of electrodes to record medical and surgical therapy?
EEG in the ICU is uncertain and the practicality of placing
many electrodes in an electrophysiologically unfriendly
environment needs to be considered. Quantitative EEG Summary
algorithms have been developed to support the time-con-
suming expert review of cEEG recordings in the ICU setting. Brain metabolism in humans can be monitored at
Several studies have highlighted concern regarding the use bedside using cerebral microdialysis. Brain extracellular
of bispectral index score (BIS) measurements as an EEG concentrations of energy metabolism markers, including
quantification tool, stressing large intra- and inter-individual lactate, pyruvate, and glucose, are accurately measured
variability, as well as interferences. Data do not support the by microdialysis. Their variations over time, and in
use of BIS for brain-injured patients in the ICU. response to therapy, can help clinical management [77,
78] and are not markers of ischemia alone but also
reflect energy metabolism in the brain [79, 80]. In TBI,
Recommendations cerebral microdialysis may contribute to prognostication
1. We recommend EEG in all patients with ABI and and abnormalities appear to be associated with long-
unexplained and persistent altered consciousness. term tissue damage [81, 82]. In SAH microdialysis may
(Strong recommendation, low quality of evidence.) provide insight into inadequate energy substrate deliv-
2. We recommend urgent EEG in patients with cSE that ery [83] and on markers of delayed cerebral ischemia
do not return to functional baseline within 60 min after [84].
seizure medication and we recommend urgent (within Cerebral microdialysis has an excellent safety record.
60 min) EEG in patients with refractory SE. (Strong However, there are limitations in that it is a focal mea-
recommendation, low quality of evidence.) surement, disclosing different metabolite concentrations
3. We recommend EEG during therapeutic hypothermia when inserted in pathological or preserved brain areas and
and within 24 h of rewarming to exclude NCSz in all so microdialysis should be interpreted on the basis of
comatose patients after cardiac arrest (CA). (Strong location defined by post-insertion CT [85]. The technique
recommendation, low quality of evidence.) can be labor intensive for bedside point of care moni-
4. We suggest EEG in comatose ICU patients without an toring and interpretation. Metabolite collection also
acute primary brain condition and with unexplained occurs over time (e.g., 60 min) and so data is delayed
impairment of mental status or unexplained neurolog- rather than real-time. Microdialysis when used with other
ical deficits to exclude NCSz, particularly in those with monitors can enhance understanding of brain physiology
severe sepsis or renal/hepatic failure. (Weak recom- and also when used for research may provide novel
mendation, low quality of evidence.) insights into pathophysiological mechanisms and on
5. We suggest EEG to detect delayed cerebral ische- various treatment modalities that directly affect brain
mia (DCI) in comatose SAH patients, in whom metabolism and function.
neurological examination is unreliable. (Weak recom-
mendation, low quality of evidence.)
6. We suggest continuous EEG monitoring as the Recommendations
preferred method over routine EEG monitoring when- 1. We recommend monitoring cerebral microdialysis in
ever feasible in comatose ICU patients without an patients with or at risk of cerebral ischemia, hypoxia,
acute primary brain condition and with unexplained energy failure, and glucose deprivation. (Strong rec-
impairment of mental status or unexplained neurolog- ommendation, low quality of evidence.)
ical deficits to exclude NCSz. (Weak recommendation, 2. We recommend that the location of the microdialysis
low quality of evidence.) probe depend on the diagnosis, the type and location of
brain lesions, and technical feasibility. (Strong recom-
mendation, low quality of evidence.)
Cerebral metabolism 3. While persistently low brain glucose and/or an
Questions addressed elevated lactate/pyruvate ratio is a strong predictor of
mortality and unfavorable outcome, we recommend
1. What are the indications for cerebral microdialysis that cerebral microdialysis only be used in combina-
monitoring? tion with clinical indicators and other monitoring
1201

modalities for prognostication. (Strong recommenda- All relevant patient data, acquired at various resolution
tion, low quality of evidence.) rates, have to be integrated, since dynamic systems are
4. We suggest the use of cerebral microdialysis to assist based on relationships that can only be understood by data
titration of medical therapies such as systemic glucose integration. However, there are several obstacles to this,
control and the treatment of delayed cerebral ischemia. such as proprietary drivers from commercial vendors and
(Weak recommendation, moderate quality of time-synchronization among others. Hence, standardiza-
evidence.) tion of an informatics infrastructure including data
5. We suggest the use of cerebral microdialysis monitor- collection, data visualization, data analysis, and decision
ing to assist titration of medical therapies such as support is essential [141]. The goal of data visualization
transfusion, therapeutic hypothermia, hypocapnia, and and a clinical informatics program is to provide clinical
hyperoxia. (Weak recommendation, low quality of decision support that enhances clinician situational
evidence.) awareness about the patient state. Ergonomic data dis-
plays that present results from analyses with clinical
Please consult the relevant ESM for the following information in a sensible uncomplicated manner improves
topics: Glucose and nutrition (ESM 2), hemostasis and clinical decision-making [142]. This field of bioinfor-
hemoglobin (ESM 3), temperature and inflammation matics is rapidly evolving and dynamic and so its role in
(ESM 4), cellular damage and degeneration (ESM 5), and critical care is still to be fully elucidated.
ICU processes of care and quality assurance (ESM 6).

Multimodality monitoring: informatics, data Recommendations


integration, display, and analysis 1. We recommend utilizing ergonomic data displays that
Questions addressed present clinical information in a sensible uncompli-
cated manner to reduce cognitive load and improve
1. Should ergonomic data displays be adopted to reduce judgments of clinicians. (Strong recommendation,
clinician cognitive burden? moderate quality of evidence.)
2. Should clinical decision support tools be adopted to 2. We suggest using clinical decision support tools such
improve clinical decision-making? as algorithms that automatically process multiple data
3. Should high-resolution physiologic data be integrated streams with the results presented on a simple,
with lower resolution data? uncomplicated display. (Weak recommendation, mod-
4. Should human-centered design principles and methods erate quality of evidence.)
be used to develop technology interventions for the 3. We recommend adopting a database infrastructure that
ICU? enables the integration of high-resolution physiologic
5. Should devices use data communication standards to data (including EEG recordings) with lower resolution
improve data connectivity? data from laboratory and electronic health care systems.
6. Should multiparameter alarms and other methods of (Strong recommendation, low quality of evidence.)
‘smart’ alarms be adopted to comply with the Joint 4. We recommend following an iterative, human-centered
Commission mandate requiring hospitals to address design methodology for complex visualization displays
alarm fatigue? to avoid adversely affecting clinical decision-making.
(Strong recommendation, moderate quality of evidence.)
5. We recommend that device manufacturers utilize data
Summary communication standards including time synchroniza-
tion on all devices to improve usability of its data.
Multimodal monitoring generates an enormous amount of (Strong recommendation, low quality of evidence.)
data, including written, ordinal, continuous, and imaging 6. We recommend adopting ‘‘smart’’ alarms in the ICU to
data, in the typical patient with a neurologic disorder in help address alarm fatigue. (Strong recommendation,
the ICU. The frequency and resolution at which physio- low quality of evidence.)
logical data are acquired and displayed may vary
depending on the signal, technology, and purposes [137, Please consult ESM 7 for the discussion on monitor-
138]. Clinicians may be confronted with more than 200 ing in emerging economies.
variables when evaluating a patient [139], with the risk of
‘‘information overload’’ that can lead to preventable
medical errors [140]. In addition, data are essentially Future directions and emerging technologies
meaningless unless annotated so that providers can search
for ‘‘epochs of interest’’, effects of therapies, or identify Multimodality monitoring including clinical and labora-
potential artifacts. tory evaluation, imaging, and continuous physiologic data
1202

is an important feature of neurocritical care. The future careful observation will help understand how care based
appears bright and likely will be driven by studies that on monitoring impacts outcome including long-term
address the principal limitations to our knowledge, doc- outcome and quality of life after ICU care. In the end,
umented in this consensus, and by the desire to develop MMM is an extension of the clinical exam and cognitive
more specific and less invasive brain monitors. It is dif- skill set of the clinician, and is only as good or as useful
ficult to demonstrate that any single monitor or as the clinical team who is using the monitor and avail-
combination of monitors has a positive effect on outcome, able therapeutic options.
since outcome is influenced by the therapeutic plan driven
by monitoring, not by monitoring itself. Furthermore, Acknowledgments We would like to thank Janel Fick and Joanne
information derived from monitors of when and how to Taie for their administrative support.
treat or how to integrate information from various mon- Conflicts of interest Each author and each member of the jury
itors is still being elucidated. Hence, we need to develop reported any potential conflicts of interest (COI). The author and
more evidence on how various monitors used in neuro- NCS Guideline Committee Chairs determined any required reso-
critical care can influence care and outcome. To that end, lutions according to NCS COI process and resolution guidelines
small, randomized studies that focus on intermediate before appointment to the writing committee. The following
methods were used to resolve any potential COI: (1) Perform peer
outcomes or biomarker outcomes seem to be a reasonable review for evidence-based content, (2) provide faculty with alter-
approach [149] although careful observational studies can nate topic, (3) provide alternate faculty for specific topics, (4) limit
also help advance understanding of physiology. content to evidence with no recommendations, (5) perform review
Important enhancements in data display, integration, of all materials associated with the activity by planning committee,
(6) abstain from discussions related to the conflict, (7) abstain from
and analysis will be forthcoming as the field of bioin- voting related to the conflict, (8) request reassignment to a com-
formatics continues to evolve. However, this will depend mittee that will not result in a conflict. NCS Guidelines state: ‘‘The
on close collaboration between industry, engineers, cli- chair or co-chairs cannot have any financial or other important
nicians, and regulatory bodies to ensure standardization of conflicts of interest related to the guideline topic.’’ PLR proposed
the subject and initiated the project and therefore was appointed
device, data element terminology, and technologies. chair by the NCS. To be compliant with NCS Guidelines he did not
During the next 5 years, we likely will see the develop- vote on any of the recommendations that followed jury delibera-
ment and implementation of several visualization and tions because of potential COI associated with industry
presentation interfaces that will serve to integrate the data relationships.
into a time-aligned stream of information. Advanced data Peter Le Roux receives research funding from Integra Lifesciences,
Neurologica, the Dana Foundation, and the National Institutes of
visualization and interpretation systems, which include Health (NIH); is a consultant for Integra Lifesciences, Codman,
algorithms to detect (1) trends in physiological changes Synthes, and Neurologica; and is a member of the scientific advi-
[150]; (2) autoregulation [45]; (3) optimum CPP [151]; sory board of Cerebrotech, Brainsgate, Orsan, and Edge
(4) patient-specific rather than population-specific Therapeutics.
Mary Kay Bader receives honoraria from Bard, The Medicines
thresholds [137]; (5) reasons for physiologic alterations Company, and Neuroptics and has Stock options in Neuroptics.
[152] and other predictive methods [153, 154] to find the Neeraj Badjatia receives consulting fees from Bard and Medivance
ideal physiological state for each individual throughout and is a Scientific Advisor to Cumberland Pharmaceuticals.
their clinical course, will become commonplace. There Julian Boesel receives honoraria from Covidien, Sedana Medical,
and Orion Pharma.
will be development and validation of several monitors Gretchen Brophy receives research funding from the NIH and the
that are currently just being introduced at the bedside or Department of Defense (DoD); is on the scientific advisory board of
are planned, such as next generation NIRS-DCS [155], Edge Therapeutics; has acted as a consultant for CSL Behring; and
optic nerve sheath ultrasound [156], pupillometry [157], has received honoraria from UCB Pharma.
direct current EEG for cortical spreading depolarization Sherry Chou receives research funding from the NIH and Novartis.
Giuseppe Citerio receives speaker honoraria from Codman and has
(CSD) [158], and TCD-based non-invasive measures of received research funding from Italian government agencies (AIFA,
ICP [159]. Ministero Salute, Regione Lombardia).
Devices used to monitor patients with neurologic Marek Czosnyka is a consultant for Cambridge Enterprise Ltd and
disorders are experiencing technological advancements serves on the Speakers Bureau for Bard Medical.
Michael Diringer receives research funding from the NIH and the
leading to high functionality, non-invasive devices, ease AHA and is a consultant for Cephalogics LLC.
of operation, and miniaturization. These technologies and Monisha Kumar receives research funding from Haemonetics.
others likely will become increasingly used to better Molly McNett is a consultant for Bard Medivance and a scientific
monitor patients who are at risk of neurological deterio- advisor for Cumberland Pharmaceuticals.
David Menon has acted as a consultant or a member of Steering or
ration. The challenge will be to integrate some or all of Data Management Committees for Solvay Ltd, GlaxoSmithKline
the multimodality monitors in an organized way to Ltd, Brainscope Ltd, Ornim Medical, Shire Medical, and Neurovive
enhance patient care, and to avoid data misinterpretation Ltd.
[160, 161]. This challenge will likely be met through J. Javier Provencio receives research funding from the NIH, Bard
rigorous training of clinicians with expertise in neuro- Medivance, and Advanced Circulatory Systems, and is on the sci-
entific advisory board of Edge Therapeutics and Minnetronix.
critical care rather than by one or more definitive studies. Nino Stocchetti is a consultant for Orsan.
However multicenter collaborative research through
1203

Paul Vespa receives grant funding from the NIH, DOD; is a con- Horowitz, Peter Hutchinson, Chad Miller, Andrew Naidech, Mauro
sultant for Edge Therapeutics; and has Stock Options with Intouch Oddo, DaiWai Olson, Kristine O’Phelan, Corinna Puppo, Richard
Health. Riker, Claudia Robertson, Michael Schmidt, Fabio Taccone have
Walter Videtta receives NIH funding. declared no conflicts of interest.
Rocco Armondo, Randall Chesnut, Jan Classen, Michael De
Georgia, Anthony Figaji, Jennifer Fugate, Raimund Helbok, David

References
1. Moher D, Liberati A, Tetzlaff J, 7. Wilt TJ, Guyatt G, Kunz R, Macnee 14. Marmarou A, Lu J, Butcher I,
Altman DG, PRISMA Group (2009) W, Puhan MA, Viegi G, Woodhead McHugh GS, Murray GD, Steyerberg
Preferred reporting items for M, Akl EA, Schünemann HJ, ATS/ EW, Mushkudiani NA, Choi S, Maas
systematic reviews and meta-analyses: ERS Ad Hoc Committee on AI (2007) Prognostic value of the
the PRISMA statement. BMJ Integrating and Coordinating Efforts Glasgow Coma Scale and pupil
339:b2535 in COPD Guideline Development reactivity in traumatic brain injury
2. Jaeschke R, Guyatt GH, Dellinger P (2012) Deciding what type of evidence assessed pre-hospital and on
et al (2008) Use of GRADE grid to and outcomes to include in guidelines: enrollment: an IMPACT analysis.
reach decisions on clinical practice article 5 in Integrating and J Neurotrauma 24:270–280
guidelines when consensus is elusive. coordinating efforts in COPD 15. Wijdicks EF, Bamlet WR,
BMJ 337:a744 guideline development. An official Maramattom BV, Manno EM,
3. Rochwerg B, Alhazzani W, Jaeschke ATS/ERS workshop report. Proc Am McClelland RL (2005) Validation of a
R (2014) Clinical meaning of the Thorac Soc 9(5):243–250 new coma scale: the FOUR score. Ann
GRADE rules. Intensive Care Med 8. Woolf S, Schünemann HJ, Eccles MP, Neurol 58:585–593
40:877–879 Grimshaw JM, Shekelle P (2012) 16. Kramer AA, Wijdicks EF, Snavely
4. Andrews JC, Schünemann HJ, Oxman Developing clinical practice VL, Dunivan JR, Naranjo LL, Bible S,
AD, Pottie K, Meerpohl JJ, Coello PA, guidelines: types of evidence and Rohs T, Dickess SM (2012) A
Rind D, Montori VM, Brito JP, Norris outcomes; values and economics, multicenter prospective study of
S, Elbarbary M, Post P, Nasser M, synthesis, grading, and presentation interobserver agreement using the Full
Shukla V, Jaeschke R, Brozek J, and deriving recommendations. Outline of Unresponsiveness score
Djulbegovic B, Guyatt G (2013) Implement Sci 4(7):61 coma scale in the intensive care unit.
GRADE guidelines: 15. Going from 9. Hsu J, Bro_zek JL, Terracciano L, Crit Care Med 40:2671–2676
evidence to recommendation- Kreis J, Compalati E, Stein AT, 17. Gelinas C, Klein K, Naidech AM,
determinants of a recommendation’s Fiocchi A, Schünemann HJ (2011) Skrobik Y (2013) Pain, sedation, and
direction and strength. J Clin Application of GRADE: making delirium management in the
Epidemiol 66(7):726–735 evidence-based recommendations neurocritically ill: lessons learned
5. Guyatt G, Oxman AD, Akl EA, Kunz about diagnostic tests in clinical from recent research. Semin Respir
R, Vist G, Brozek J, Norris S, Falck- practice guidelines. Implement Sci Crit Care Med 34:236–243
Ytter Y, Glasziou P, DeBeer H, 10(6):62 18. Chatelle C, Majerus S, Whyte J,
Jaeschke R, Rind D, Meerpohl J, 10. Guyatt GH, Oxman AD, Kunz R, Laureys S, Schnakers C (2012) A
Dahm P, Schünemann HJ (2011) Atkins D, Brozek J, Vist G, Alderson sensitive scale to assess nociceptive
GRADE guidelines: 1 Introduction- P, Glasziou P, Falck-Ytter Y, pain in patients with disorders of
GRADE evidence profiles and Schünemann HJ (2011) GRADE consciousness. J Neurol Neurosurg
summary of findings tables. J Clin guidelines: 2. Framing the question Psychiatry 8(3):1233–1237
Epidemiol 64(4):383–394 and deciding on important outcomes. 19. Barr J, Fraser GL, Puntillo K, Ely EW,
6. Schünemann HJ, Wiercioch W, J Clin Epidemiol 64(4):395–400 Gélinas C, Dasta JF, Davidson JE,
Etxeandia I, Falavigna M, Santesso N, 11. Gill M, Windemuth R, Steele R, Green Devlin JW, Kress JP, Joffe AM,
Mustafa R, Ventresca M, SM (2005) A comparison of the Coursin DB, Herr DL, Tung A,
Brignardello-Petersen R, Laisaar KT, Glasgow Coma Scale score to Robinson BR, Fontaine DK, Ramsay
Kowalski S, Baldeh T, Zhang Y, Raid simplified alternative scores for the MA, Riker RR, Sessler CN, Pun B,
U, Neumann I, Norris SL, Thornton J, prediction of traumatic brain injury Skrobik Y, Jaeschke R, American
Harbour R, Treweek S, Guyatt G, outcomes. Ann Emerg Med 45:37–42 College of Critical Care Medicine
Alonso-Coello P, Reinap M, Brozek J, 12. Tsau JW, Hemphill JC, Johnston SC, (2013) Clinical practice guidelines for
Oxman A, Akl EA (2014) Guidelines Smith WS, Bonovich DC (2005) the management of pain, agitation, and
2.0: systematic development of a Initial Glasgow Coma Scale score delirium in adult patients in the
comprehensive checklist for a predicts outcome following intensive care unit. Crit Care Med
successful guideline enterprise. CMAJ thrombolysis for posterior circulation 41:263–306
186(3):E123–E142 stroke. Arch Neurol 62:1126–1129 20. Helbok R, Kurtz P, Schmidt MJ, Stuart
13. Schefold JC, Storm C, Kruger A, MR, Fernandez L, Connolly SE, Lee
Ploner CJ, Hasper D (2009) The K, Schmutzhard E, Mayer SA,
Glasgow Coma Score is a predictor of Claassen J, Badjatia N (2012) Effects
good outcome in cardiac arrest of the neurological wake-up test on
patients treated with therapeutic clinical examination, intracranial
hypothermia. Resuscitation pressure, brain metabolism and brain
80:658–661 tissue oxygenation in severely brain-
injured patients. Crit Care 16:R226
1204

21. Mitasova A, Kostalova M, Bednarik J, 30. Mutoh T, Kazumata K, Ajiki M, 40. Chesnut RM, Temkin N, Carney N,
Michalcakova R, Kasparek T, Ushikoshi S, Terasaka S (2007) Goal- Dikmen S, Rondina C, Videtta W,
Balabanova P, Dusek L, Vohanka S, directed fluid management by bedside Petroni G, Lujan S, Pridgeon J, Barber
Ely EW (2012) Poststroke delirium transpulmonary hemodynamic J, Machamer J, Chaddock K, Celix
incidence and outcomes: validation of monitoring after subarachnoid JM, Cherner M, Hendrix T, Global
the Confusion Assessment Method for hemorrhage. Stroke 38(12):3218–3224 Neurotrauma Research Group (2012)
the Intensive Care Unit (CAM-ICU). 31. Mendelson AA, Gillis C, Henderson A trial of intracranial-pressure
Crit Care Med 40(2):484–490 WR, Ronco JJ, Dhingra V, Griesdale monitoring in traumatic brain injury.
22. Davies KR, Gelb AW, Manninen PH, DE (2012) Intracranial pressure N Engl J Med 367(26):2471–2481
Boughner DR, Bisnaire D (1991) monitors in traumatic brain injury: a 41. Arabi YM, Haddad S, Tamim HM, Al-
Cardiac function in aneurysmal systematic review. Can J Neurol Sci Dawood A, Al-Qahtani S, Ferayan A,
subarachnoid haemorrhage: a study of 39(5):571–576 Al-Abdulmughni I, Al-Oweis J,
electrocardiographic and 32. Andrews PJ, Sleeman DH, Statham Rugaan A (2010) Mortality reduction
echocardiographic abnormalities. Br J PF, McQuatt A, Corruble V, Jones PA, after implementing a clinical practice
Anaesth 67(1):58–63 Howells TP, Macmillan CS (2002) guidelines-based management
23. Kopelnik A, Fisher L, Miss JC, Banki Predicting recovery in patients protocol for severe traumatic brain
N, Tung P, Lawton MT, Ko N, Smith suffering from traumatic brain injury injury. J Crit Care 25(2):190–195
WS, Drew B, Foster E, Zaroff J (2005) by using admission variables and 42. Gerber LM, Chiu YL, Carney N, Härtl
Prevalence and implications of physiological data: a comparison R, Ghajar J (2013) Marked reduction
diastolic dysfunction after between decision tree analysis and in mortality in patients with severe
subarachnoid hemorrhage. Neurocrit logistic regression. J Neurosurg traumatic brain injury. J Neurosurg
Care 3(2):132–138 97(2):326–336 119(6):1583–1590
24. Naidech AM, Kreiter KT, Janjua N, 33. Badri S, Chen J, Barber J, Temkin NR, 43. Alali AS, Fowler RA, Mainprize TG,
Ostapkovich ND, Parra A, Dikmen SS, Chesnut RM, Deem S, Scales DC, Kiss A, de Mestral C, Ray
Commichau C, Fitzsimmons BF, Yanez ND, Treggiari MM (2012) JG, Nathens AB (2013) Intracranial
Connolly ES, Mayer SA (2005) Mortality and long-term functional pressure monitoring in severe
Cardiac troponin elevation, outcome associated with intracranial traumatic brain injury: results from the
cardiovascular morbidity, and pressure after traumatic brain injury. American College of Surgeons
outcome after subarachnoid Intensive Care Med 38(11):1800–1809 Trauma Quality Improvement
hemorrhage. Circulation 34. Treggiari MM, Schutz N, Yanez ND, Program. J Neurotrauma
112(18):2851–2856 Romand JA (2007) Role of 30(20):1737–1746
25. Naidech AM, Bassin SL, Garg RK, intracranial pressure values and 44. Steiner LA, Coles JP, Johnston AJ,
Ault ML, Bendok BR, Batjer HH, patterns in predicting outcome in Chatfield DA, Smielewski P, Fryer
Watts CM, Bleck TP (2009) Cardiac traumatic brain injury: a systematic TD, Aigbirhio FI, Clark JC, Pickard
troponin I and acute lung injury after review. Neurocrit Care. 6(2):104–112 JD, Menon DK, Czosnyka M (2003)
subarachnoid hemorrhage. Neurocrit 35. Bekar A, Doğan S, Abaş F, Caner B, Assessment of cerebrovascular
Care 11(2):177–182 Korfali G, Kocaeli H, Yilmazlar S, autoregulation in head-injured
26. Mayer SA, Lin J, Homma S, Solomon Korfali E (2009) Risk factors and patients: a validation study. Stroke
RA, Lennihan L, Sherman D, Fink complications of intracranial pressure 34(10):2404
ME, Beckford A, Klebanoff LM monitoring with a fiberoptic device. 45. Czosnyka M, Smielewski P,
(1999) Myocardial injury and left J Clin Neurosci 16(2):236–240 Kirkpatrick P, Laing RJ, Menon D,
ventricular performance after 36. Resnick DK, Marion DW, Carlier P Pickard JD (1997) Continuous
subarachnoid hemorrhage. Stroke (1997) Outcome analysis of patients assessment of the cerebral vasomotor
30(4):780–786 with severe head injuries and reactivity in head injury. Neurosurgery
27. Tung P, Kopelnik A, Banki N, Ong K, prolonged intracranial hypertension. 41(1):11–17
Ko N, Lawton MT, Gress D, Drew B, J Trauma 42(6):1108–1111 46. Lavinio A, Timofeev I, Nortje J,
Foster E, Parmley W, Zaroff J (2004) 37. Kosty JA, Le Roux PD, Levine J, Park Outtrim J, Smielewski P, Gupta A,
Predictors of neurocardiogenic injury S, Kumar MA, Frangos S, Maloney- Hutchinson PJ, Matta BF, Pickard JD,
after subarachnoid hemorrhage. Stroke Wilensky E, Kofke WA (2013) Brief Menon D, Czosnyka M (2007)
35(2):548–551 report: a comparison of clinical and Cerebrovascular reactivity during
28. Sugimoto K, Inamasu J, Kato Y, research practices in measuring hypothermia and rewarming. Br J
Yamada Y, Ganaha T, Oheda M, cerebral perfusion pressure: a Anaesth 99(2):237–244
Hattori N, Watanabe E, Ozaki Y, literature review and practitioner 47. Steiner LA, Czosnyka M, Piechnik
Hirose Y (2013) Association between survey. Anesth Analg 117(3):694–698 SK, Smielewski P, Chatfield D,
elevated plasma norepinephrine levels 38. Robertson CS, Valadka AB, Hannay Menon DK, Pickard JD (2002)
and cardiac wall motion abnormality HJ, Contant CF, Gopinath SP, Cormio Continuous monitoring of
in poor-grade subarachnoid M, Uzura M, Grossman RG (1999) cerebrovascular pressure reactivity
hemorrhage patients. Neurosurg Rev Prevention of secondary ischemic allows determination of optimal
36(2):259–266 insults after severe head injury. Crit cerebral perfusion pressure in patients
29. Banki NM, Kopelnik A, Dae MW, Care Med 27(10):2086–2095 with traumatic brain injury. Crit Care
Miss J, Tung P, Lawton MT, Drew BJ, 39. Aries MJ, Czosnyka M, Budohoski Med 30(4):733–738
Foster E, Smith W, Parmley WW, KP, Steiner LA, Lavinio A, Kolias 48. Jubran A, Tobin MJ (1990) Reliability
Zaroff JG (2005) Acute AG, Hutchinson PJ, Brady KM, of pulse oximetry in titrating
neurocardiogenic injury after Menon DK, Pickard JD, Smielewski P supplemental oxygen therapy in
subarachnoid hemorrhage. Circulation (2012) Continuous determination of ventilator-dependent patients. Chest
112(21):3314–3319 optimal cerebral perfusion pressure in 97:1420–1425
traumatic brain injury. Crit Care Med
40(8):2456–2463
1205

49. Sulter G, Elting JW, Stewart R, den 59. Zweifel C, Castellani G, Czosnyka M, 67. Turek G, Kochanowicz J, Rutkowski
Arend A, De Keyser J (2000) Helmy A, Manktelow A, Carrera E, R, Krejza J, Lyson T, Gorbacz K,
Continuous pulse oximetry in acute Brady KM, Hutchinson PJ, Menon Zielinska-Turek J, Mariak Z (2012)
hemiparetic stroke. J Neurol Sci DK, Pickard JD, Smielewski P (2010) Accuracy of transcranial colour-coded
179:65–69 Noninvasive monitoring of sonography in the diagnosis of anterior
50. Anderson CT, Breen PH (2000) cerebrovascular reactivity with near cerebral artery vasospasm. Neurol
Carbon dioxide kinetics and infrared spectroscopy in head-injured Neurochir Pol 46(3):233–238
capnography during critical care. Crit patients. J Neurotrauma 27:1951–1958 68. Vora YY, Suarez-Almazor M, Steinke
Care 4:207–215 60. Menon DK, Coles JP, Gupta AK, DE, Martin ML, Findlay JM (1999)
51. Pennings FA, Schuurman PR, van den Fryer TD, Smielewski P, Chatfield Role of transcranial Doppler
Munckhof P, Bouma GJ (2008) Brain DA, Aigbirhio F, Skepper JN, Minhas monitoring in the diagnosis of cerebral
tissue oxygen pressure monitoring in PS, Hutchinson PJ, Carpenter TA, vasospasm after subarachnoid
awake patients during functional Clark JC, Pickard JD (2004) Diffusion hemorrhage. Neurosurgery
neurosurgery: the assessment of limited oxygen delivery following 44:1237–1247
normal values. J Neurotrauma head injury. Crit Care Med 69. Naval NS, Thomas CE, Urrutia VC
25:1173–1177 32:1384–1390 (2005) Relative changes in flow
52. Doppenberg EM, Zauner A, Watson 61. Vespa P, Bergsneider M, Hattori N, velocities in vasospasm after
JC, Bullock R (1998) Determination of Wu HM, Huang SC, Martin NA, subarachnoid hemorrhage: a
the ischemic threshold for brain Glenn TC, McArthur DL, Hovda DA transcranial Doppler study. Neurocrit
oxygen tension. Acta Neurochir Suppl (2005) Metabolic crisis without brain Care 2(2):133–140
71:166–169 ischemia is common after traumatic 70. Claassen J, Taccone FS, Horn P,
53. Ponce LL, Pillai S, Cruz J, Li X, Julia brain injury: a combined microdialysis Holtkamp M, Stocchetti N, Oddo M
H, Gopinath S, Robertson CS (2012) and positron emission tomography (2013) Recommendations on the use
Position of probe determines study. J Cereb Blood Flow Metab of EEG monitoring in critically ill
prognostic information of brain tissue 25:763–774 patients: consensus statement from the
PO2 in severe traumatic brain injury. 62. Vajkoczy P, Roth H, Horn P, Lucke T, neurointensive care section of the
Neurosurgery 70(6):1492–1502 Thomé C, Hubner U, Martin GT, ESICM. Intensive Care Med
54. Pascual JL, Georgoff P, Maloney- Zappletal C, Klar E, Schilling L, 39(8):1337–1351
Wilensky E, Sims C, Sarani B, Stiefel Schmiedek P (2000) Continuous 71. Varelas PN, Hacein-Bey L, Hether T,
MF, LeRoux PD, Schwab CW (2011) monitoring of regional cerebral blood Terranova B, Spanaki MV (2004)
Reduced brain tissue oxygen in flow: experimental and clinical Emergent electroencephalogram in the
traumatic brain injury: are most validation of a novel thermal diffusion intensive care unit: indications and
commonly used interventions microprobe. J Neurosurg diagnostic yield. Clin EEG Neurosci
successful? J Trauma 70:535–546 93(2):265–274 35:173–180
55. Coplin WM, O’Keefe GE, Grady MS, 63. Gesang DZ, Zhang D, Zhao JZ, Wang 72. Rossetti AO, Oddo M, Logroscino G,
Grant GA, March KS, Winn HR, Lam S, Zhao YL, Wang R, Sun JJ, Meng Z Kaplan PW (2010) Prognostication
AM (1997) Thrombotic, infectious, (2009) Laser Doppler flowmeter study after cardiac arrest and hypothermia: a
and procedural complications of the on regional cerebral blood flow in prospective study. Ann Neurol
jugular bulb catheter in the intensive early stage after standard superficial 67:301–307
care unit. Neurosurgery 41:101–107 temporal artery-middle cerebral artery 73. Zandbergen EG, Hijdra A, Koelman
56. Schoon P, Benito Mori L, Orlandi G, bypass surgery for Moyamoya disease. JH, van Dijk JG, Ongerboer de Visser
Larralde C, Radrizzani M (2002) Chin Med J (Engl) 122(20):2412–2418 BW, Spaans F, Tavy DL, Koelman JH
Incidence of intracranial hypertension 64. Kincaid MS, Souter MJ, Treggiari (2006) Prediction of poor outcome
related to jugular bulb oxygen MM, Yanez ND, Moore A, Lam AM within the first 3 days of postanoxic
saturation disturbances in severe (2009) Accuracy of transcranial coma. Neurology 66:62–68
traumatic brain injury patients. Acta Doppler ultrasonography and single- 74. Rossetti AO, Carrera E, Oddo M
Neurochir Suppl 81:285–287 photon emission computed (2012) Early EEG correlates of
57. Coles JP, Fryer TD, Smielewski P, tomography in the diagnosis of neuronal injury after brain anoxia.
Chatfield DA, Steiner LA, Johnston angiographically demonstrated Neurology 78:796–802
AJ, Downey SP, Williams GB, cerebral vasospasm. J Neurosurg 75. Young GB, Jordan KG, Doig GS
Aigbirhio F, Hutchinson PJ, Rice K, 110(1):67–72 (1996) An assessment of
Carpenter TA, Clark JC, Pickard JD, 65. Lysakowski C, Walder B, Costanza nonconvulsive seizures in the
Menon DK (2004) Incidence and MC, Tramèr MR (2001) Transcranial intensive care unit using continuous
mechanisms of cerebral ischemia in Doppler versus angiography in EEG monitoring: an investigation of
early clinical head injury. J Cereb patients with vasospasm due to a variables associated with mortality.
Blood Flow Metab 24(2):202–211 ruptured cerebral aneurysm: a Neurology 47:83–89
58. Schneider GH, von Helden A, systematic review. Stroke 76. Claassen J, Mayer SA, Kowalski RG,
Lanksch WR, Unterberg A (1995) 32(10):2292–2298 Emerson RG, Hirsch LJ (2004)
Continuous monitoring of jugular bulb 66. Suarez JI, Qureshi AI, Yahia AB, Detection of electrographic seizures
oxygen saturation in comatose Parekh PD, Tamargo RJ, Williams with continuous EEG monitoring in
patients—therapeutic implications. MA, Ulatowski JA, Hanley DF, critically ill patients. Neurology
Acta Neurochir (Wien) 134:71–75 Razumovsky AY (2002) Symptomatic 62:1743–1748
vasospasm diagnosis after
subarachnoid hemorrhage: evaluation
of transcranial Doppler ultrasound and
cerebral angiography as related to
compromised vascular distribution.
Crit Care Med 30(6):1348–1355
1206

77. Bellander BM, Cantais E, Enblad P, 85. Nordstrom CH, Reinstrup P, Xu W, 95. Smoller BR, Kruskall MS, Horowitz
Hutchinson P, Nordström CH, Gardenfors A, Ungerstedt U (2003) GL (1998) Reducing adult phlebotomy
Robertson C, Sahuquillo J, Smith M, Assessment of the lower limit for blood loss with the use of pediatric-
Stocchetti N, Ungerstedt U, Unterberg cerebral perfusion pressure in severe sized blood collection tubes. Am J
A, Olsen NV (2004) Consensus head injuries by bedside monitoring of Clin Pathol 91:701–703
meeting on microdialysis in regional energy metabolism. 96. Levine J, Kofke A, Cen L, Faerber J,
neurointensive care. Intensive Care Anesthesiology 98(4):805–807 Elliott JP, Winn HR, Le Roux P
Med 30(12):2166–2169 86. Hartl R, Gerber LM, Ni Q, Ghajar J (2010) Red blood cell transfusion is
78. Nortje J, Coles JP, Timofeev I, Fryer (2008) Effect of early nutrition on associated with infection and
TD, Aigbirhio FI, Smielewski P, deaths due to severe traumatic brain extracerebral complications after
Outtrim JG, Chatfield DA, Pickard JD, injury. J Neurosurg 109:50–56 subarachnoid hemorrhage.
Hutchinson PJ, Gupta AK, Menon DK 87. Desachy A, Vuagnat AC, Ghazali AD, Neurosurgery 66:312–318
(2008) Effect of hyperoxia on regional Baudin OT, Longuet OH, Calvat SN, 97. Naidech AM, Bernstein RA,
oxygenation and metabolism after Gissot V (2008) Accuracy of bedside Levasseur K, Bassin SL, Bendok BR,
severe traumatic brain injury: glucometry in critically ill patients: Batjer HH, Bleck TP, Alberts MJ
preliminary findings. Crit Care Med influence of clinical characteristics and (2009) Platelet activity and outcome
36(1):273–281 perfusion index. Mayo Clin Proc after intracerebral hemorrhage. Ann
79. Larach DB, Kofke WA, Le Roux P 83:400–405 Neurol 65:352–356
(2011) Potential non-hypoxic/ 88. Lonjaret L, Claverie V, Berard E, Riu- 98. Goldenberg NA, Jacobson L, Manco-
ischemic causes of increased cerebral Poulenc B, Geeraerts T, Genestal M, Johnson MJ (2005) Duration of
interstitial fluid lactate/pyruvate ratio Fourcade O (2012) Relative accuracy platelet dysfunction after a 7-day
(LPR): a review of available literature. of arterial and capillary glucose meter course of ibuprofen. Ann Intern Med
Neurocrit Care 15(3):609–622 measurements in critically ill patients. 142:506–509
80. Vespa P, Bergsneider M, Hattori N, Diabetes Metab 38:230–235 99. Naidech AM, Jovanovic B, Liebling S,
Wu HM, Huang SC, Martin NA, 89. Finkielman JD, Oyen LJ, Afessa B Garg RK, Bassin SL, Bendok BR,
Glenn TC, McArthur DL, Hovda DA (2005) Agreement between bedside Bernstein RA, Alberts MJ, Batjer HH
(2005) Metabolic crisis without brain blood and plasma glucose (2009) Reduced platelet activity is
ischemia is common after traumatic measurement in the ICU setting. Chest associated with early clot growth and
brain injury: a combined microdialysis 127:1749–1751 worse 3-month outcome after
and positron emission tomography 90. Oddo M, Schmidt JM, Mayer SA, intracerebral hemorrhage. Stroke
study. J Cereb Blood Flow Metab Chiolero RL (2008) Glucose control 40:2398–2401
25(6):763–774 after severe brain injury. Curr Opin 100. Dansirikul C, Lehr T, Liesenfeld KH,
81. Timofeev I, Carpenter KL, Nortje J, Clin Nutr Metab Care 11:134–139 Haertter S, Staab A (2012) A
Al-Rawi PG, O’Connell MT, 91. Vespa P, Boonyaputthikul R, combined pharmacometric analysis of
Czosnyka M, Smielewski P, Pickard McArthur DL, Miller C, Etchepare M, dabigatran etexilate in healthy
JD, Menon DK, Kirkpatrick PJ, Gupta Bergsneider M, Glenn T, Martin N, volunteers and patients with atrial
AK, Hutchinson PJ (2011) Cerebral Hovda D (2006) Intensive insulin fibrillation or undergoing orthopaedic
extracellular chemistry and outcome therapy reduces microdialysis glucose surgery. Thromb Haemost
following traumatic brain injury: a values without altering glucose 107:775–785
microdialysis study of 223 patients. utilization or improving the lactate/ 101. Stangier J, Feuring M (2012) Using
Brain 134(Pt 2):484–494 pyruvate ratio after traumatic brain the HEMOCLOT direct thrombin
82. Marcoux J, McArthur DA, Miller C, injury. Crit Care Med 34(3):850–856 inhibitor assay to determine plasma
Glenn TC, Villablanca P, Martin NA, 92. Ferrie S, Allman-Farinelli M (2013) concentrations of dabigatran. Blood
Hovda DA, Alger JR, Vespa PM Commonly used ‘‘nutrition’’ indicators Coagul Fibrinolysis 23:138–143
(2008) Persistent metabolic crisis as do not predict outcome in the critically 102. Chee YL, Crawford JC, Watson HG,
measured by elevated cerebral ill: a systematic review. Nutr Clin Greaves M (2008) Guidelines on the
microdialysis lactate-pyruvate ratio Pract 28:463–484 assessment of bleeding risk prior to
predicts chronic frontal lobe brain 93. Reignier J, Mercier E, Le Gouge A, surgery or invasive procedures. British
atrophy after traumatic brain injury. Boulain T, Desachy A, Bellec F, Committee for Standards in
Crit Care Med 36(10):2871–2877 Clavel M, Frat JP, Plantefeve G, Haematology. Br J Haematol
83. Skjøth-Rasmussen J, Schulz M, Quenot JP, Lascarrou JB, Clinical 140:496–504
Kristensen SR, Bjerre P (2004) Research in Intensive Care and Sepsis 103. West KL, Adamson C, Hoffman M
Delayed neurological deficits detected (CRICS) Group (2013) Effect of not (2011) Prophylactic correction of the
by an ischemic pattern in the monitoring residual gastric volume on international normalized ratio in
extracellular cerebral metabolites in risk of ventilator-associated neurosurgery: a brief review of a brief
patients with aneurysmal subarachnoid pneumonia in adults receiving literature. J Neurosurg 114:9–18
hemorrhage. J Neurosurg 100(1):8–15 mechanical ventilation and early 104. Mun JH, Cho KY, Lim BC, Lim JS,
84. Sarrafzadeh AS, Copin JC, Jimenez enteral feeding: a randomized Lee RS (2013) Factors related to
Bengualid J, Turck N, Vajkoczy P, controlled trial. JAMA 309:249–256 catheter-induced hemorrhage after
Bijlega P, Schaller K, Gasche Y 94. Kramer AH, Zygun DA, Bleck TP, brain parenchymal catheterization.
(2012) Matrix metaloproteinase-9 Dumont AS, Kassell NF, Nathan B Chonnam Med J 49:113–117
concentration in the cerebral (2009) Relationship between 105. Krisl JC, Meadows HE, Greenberg
extracellular fluid of patients during hemoglobin concentrations and CS, Mazur JE (2011) Clinical
the actue phase of aneurismal outcomes across subgroups of patients usefulness of recombinant activated
subarachnoid hemorrhage. Neurol Res with aneurysmal subarachnoid factor VII in patients with liver failure
34(5):455–461 hemorrhage. Neurocrit Care undergoing invasive procedures. Ann
10:157–165 Pharmacother 45:1433–1438
1207

106. Lisman T, Bakhtiari K, Pereboom IT, 115. Choi HA, Ko SB, Presciutti M, 122. Chou SH-Y, Feske SK, Simmons SL,
Hendriks HG, Meijers JC, Porte RJ Fernandez L, Carpenter AM, Lesch C, Konigsberg RG, Orzell SC,
(2010) Normal to increased thrombin Gilmore E, Malhotra R, Mayer SA, Marckmann A, Bourget G, Bauer DJ,
generation in patients undergoing liver Lee K, Claassen J, Schmidt JM, De Jager PL, Du R, Arai K, Lo EH,
transplantation despite prolonged Badjatia N (2011) Prevention of Ning MM (2011) Elevated peripheral
conventional coagulation tests. shivering during therapeutic neutrophils and matrix
J Hepatol 52:355–361 temperature modulation: the Columbia metalloproteinase 9 as biomarkers of
107. Oddo M, Frangos S, Milby A, Chen I, anti-shivering protocol. Neurocrit Care functional outcome following
Maloney-Wilensky E, Murtrie EM, 14(3):389–394 subarachnoid hemorrhage. Transl
Stiefel M, Kofke WA, Le Roux PD, 116. Provencio JJ, Fu X, Siu A, Rasmussen Stroke Res 2(4):600–607
Levine JM (2009) Induced PA, Hazen SL, Ransohoff RM (2010) 123. Papa L, Lewis LM, Silvestri S, Falk
normothermia attenuates cerebral CSF neutrophils are implicated in the JL, Giordano P, Brophy GM, Demery
metabolic distress in patients with development of vasospasm in JA, Liu MC, Mo J, Akinyi L,
aneurysmal subarachnoid hemorrhage subarachnoid hemorrhage. Neurocrit Mondello S, Schmid K, Robertson CS,
and refractory fever. Stroke Care 12(2):244–251 Tortella FC, Hayes RL, Wang KK
40(5):1913–1916 117. Oconnor E, Venkatesh B, (2012) Serum levels of ubiquitin
108. Puccio AM, Fischer MR, Jankowitz Mashongonyika C, Lipman J, Hall J, C-terminal hydrolase distinguish mild
BT, Yonas H, Darby JM, Okonkwo Thomas P (2004) Serum procalcitonin traumatic brain injury from trauma
DO (2009) Induced normothermia and C-reactive protein as markers of controls and are elevated in mild and
attenuates intracranial hypertension sepsis and outcome in patients with moderate traumatic brain injury
and reduces fever burden after severe neurotrauma and subarachnoid patients with intracranial lesions and
traumatic brain injury. Neurocrit Care haemorrhage. Anaesth Intensive Care neurosurgical intervention. J Trauma
11(1):82–87 32(4):465–470 Acute Care Surg 72:1335–1344
109. Thompson HJ, Pinto-Martin J, Bullock 118. Tiainen M, Roine RO, Pettila V, 124. Siman R, Giovannone N, Toraskar N,
MR (2003) Neurogenic fever after Takkunen O (2003) Serum neuron- Frangos S, Stein SC, Levine JM,
traumatic brain injury: an specific enolase and S-100B protein in Kumar MA (2011) Evidence that a
epidemiological study. J Neurol cardiac arrest patients treated with panel of neurodegeneration
Neurosurg Psychiatry 74(5):614–619 hypothermia. Stroke 34:2881–2886 biomarkers predicts vasospasm,
110. Fernandez A, Schmidt JM, Claassen J, 119. Diaz-Arrastia R, Wang KK, Papa L, infarction, and outcome in aneurysmal
Pavlicova M, Huddleston D, Kreiter Sorani MD, Yue JK, Puccio AM, subarachnoid hemorrhage. PLoS One
KT, Ostapkovich ND, Kowalski RG, McMahon PJ, Inoue T, Yuh EL, 6(12):e28938
Parra A, Connolly ES, Mayer SA Lingsma HF, Maas AI, Valadka AB, 125. Kazmierski R, Michalak S, Wencel-
(2007) Fever after subarachnoid Okonkwo DO, Manley GT, Track-Tbi Warot A, Nowinski WL (2012) Serum
hemorrhage: risk factors and impact Investigators, Casey IS, Cheong M, tight-junction proteins predict
on outcome. Neurology Cooper SR, Dams-O’Connor K, hemorrhagic transformation in
68(13):1013–1019 Gordon WA, Hricik AJ, Menon DK, ischemic stroke patients. Neurology
111. Singh V, Sharma A, Khandelwal R, Mukherjee P, Schnyer DM, Sinha TK, 79:1677–1685
Kothari K (2000) Variation of axillary Vassar MJ (2014) Acute biomarkers of 126. Knopf L, Staff I, Gomes J,
temperature and its correlation with traumatic brain injury: relationship McCullough L (2012) Impact of a
oral temperature. J Assoc Physicians between plasma levels of ubiquitin neurointensivist on outcomes in
India 48(9):898–900 C-terminal hydrolase-L1 and glial critically ill stroke patients. Neurocrit
112. Smith LS (2004) Temperature fibrillary acidic protein. Care 16:63–71
measurement in critical care adults: a J Neurotrauma 31(1):19–25 127. Kramer AH, Zygun DA (2013)
comparison of thermometry and 120. Czeiter E, Mondello S, Kovacs N, Declining mortality in neurocritical
measurement routes. Biol Res Nurs Sandor J, Gabrielli A, Schmid K, care patients: a cohort study in
6(2):117–125 Tortella F, Wang KK, Hayes RL, Southern Alberta over 11 years. Can J
113. Hata JS, Shelsky CR, Hindman BJ, Barzo P, Ezer E, Doczi T, Buki A Anesth 60:966–975
Smith TC, Simmons JS, Todd MM (2012) Brain injury biomarkers may 128. Damian MS, Ben-Shlomo Y, Howard
(2008) A prospective, observational improve the predictive power of the R, Bellotti T, Harrison D, Griggs K,
clinical trial of fever reduction to IMPACT outcome calculator. Rowan K (2013) The effect of secular
reduce systemic oxygen consumption J Neurotrauma 29:1770–1778 trends and specialist neurocritical care
in the setting of acute brain injury. 121. Mercier E, Boutin A, Lauzier F, on mortality for patients with
Neurocrit Care 9(1):37–44 Fergusson DA, Simard J-F, intracerebral hemorrhage, myasthenia
114. Oddo M, Frangos S, Maloney- Zarychanski R et al (2013) Predictive gravis and Guillain–Barré syndrome
Wilensky E, Andrew Kofke W, Le value of S-100B protein for prognosis admitted to critical care. Intensive
Roux P, Levine JM (2010) Effect of in patients with moderate and severe Care Med 39:1405–1412
shivering on brain tissue oxygenation traumatic brain injury: systematic 129. Kramer AH, Zygun DA (2011) Do
during induced normothermia in review and meta-analysis. BMJ neurocritical care units save lives?
patients with severe brain injury. 346:f1757 Measuring the impact of specialized
Neurocrit Care 12(1):10–16 ICUs. Neurocrit Care 14:329–333
130. Patel HC, Menon DK, Tebbs S,
Hawker R, Hutchinson PJ, Kirkpatrick
PJ (2002) Specialist neurocritical care
and outcome from head injury.
Intensive Care Med 28(5):547–553
1208

131. Elf K, Nilsson P, Enblad P (2002) 140. De Turck F, Decruyenaere J, 151. Lazaridis C, Smielewski P, Steiner
Outcome after traumatic brain injury Thysebaert P, Van Hoecke S, LA, Brady KM, Hutchinson P, Pickard
improved by an organized secondary Volckaert B, Danneels C, Colpaert K, JD, Czosnyka M (2013) Optimal
insult program and standardized De Moor G (2007) Design of a flexible cerebral perfusion pressure: are we
neurointensive care. Crit Care Med platform for execution of medical ready for it? Neurol Res
30(9):2129–2134 decision support agents in the 35(2):138–148
132. Naval NS, Chang T, Caserta F, intensive care unit. Comput Biol Med 152. Oddo M, Levine J, Frangos S,
Kowalski RG, Carhuapoma JR, 37:97–112 Maloney-Wilensky E, Carrera E,
Tamargo RJ (2012) Impact of pattern 141. Jacono FF, DeGeorgia MA, Wilson Daniel R, Magistretti PJ, Le Roux P
of admission on outcomes after CG, Dick TE, Loparo KA (2010) Data (2012) Brain lactate metabolism in
aneurysmal subarachnoid hemorrhage. acquisition and complex systems humans with subarachnoid
J Crit Care 27:532 analysis in critical care: developing haemorrhage. Stroke 43(5):1418–1421
133. Rincon F, Mayer SA, Rivolta J, the intensive care unit of the future. 153. Güiza F, Depreitere B, Piper I, Van
Stillman J, Boden-Albala B, Elkind J Healthc Eng 1:337–338 den Berghe G, Meyfroidt G (2013)
MS, Marshall R, Chong JY (2010) 142. Zhang J (2005) Human-centered Novel methods to predict increased
Impact of delayed transfer of critically computing in health information intracranial pressure during intensive
ill stroke patients from the emergency systems Part 1: analysis and design. care and long-term neurologic
department to the neuro-ICU. J Biomed Inform 38:1–3 outcome after traumatic brain injury:
Neurocrit Care 13:75–81 143. Mock C, Kobusingye O, Joshipura M, development and validation in a
134. English SW, Turgeon AF, Owen E, Nguyen S, Arreola-Risa C (2005) multicenter dataset. Crit Care Med
Doucette S, Pagliarello G, McIntyre L Strengthening trauma and critical care 41(2):554–564
(2013) Protocol management of severe globally. Curr Opin Crit Care 154. Narotam PK, Morrison JF, Schmidt
traumatic brain injury in intensive care 11:568–575 MD, Nathoo N (2014) Physiological
units: a systematic review. Neurocrit 144. Sim SK, Lim SL, Lee HK, Liew D, complexity of acute traumatic brain
Care 18:131–142 Wong A (2011) Care of severe head injury in patients treated with a brain
135. Rhodes A, Moreno RP, Azoulay E, injury patients in the Sarawak General oxygen protocol: utility of symbolic
Capuzzo M, Chiche JD, Eddleston J, Hospital: intensive care unit versus regression (SR) in predictive modeling
Endacott R, Ferdinande P, Flaatten H, general ward. Med J Malays of a dynamical system. J Neurotrauma
Guidet B, Kuhlen R, León-Gil C, 66:138–141 31(7):630–641
Martin Delgado MC, Metnitz PG, 145. De Silva MJ, Roberts I, Perel P, 155. Kim MN, Durduran T, Frangos S,
Soares M, Sprung CL, Timsit JF, Edwards P, Kenward MG, Fernandes Edlow BL, Buckley EM, Moss HE,
Valentin A, Task Force on Safety and J, Shakur H, Patel V, CRASH Trial Zhou C, Yu G, Choe R, Maloney-
Quality of European Society of Collaborators (2009) Patient outcome Wilensky E, Wolf RL, Grady MS,
Intensive Care Medicine (ESICM) after traumatic brain injury in high-, Greenberg JH, Levine JM, Yodh AG,
(2012) Prospectively defined middle- and low-income countries: Detre JA, Kofke WA (2010)
indicators to improve the safety and analysis of data on 8927 patients in 46 Noninvasive measurement of cerebral
quality of care for critically ill countries. Int J Epidemiol 38:452–458 blood flow and blood oxygenation
patients: a report from the Task Force 146. Biestro AA, Alberti RA, Soca AE, using near-infrared and diffuse
on Safety and Quality of the European Cancela M, Puppo CB, Borovich B correlation spectroscopies in critically
Society of Intensive Care Medicine (1995) Use of indomethacin in brain- brain-injured adults. Neurocrit Care
(ESICM). Intensive Care Med injured patients with cerebral 12(2):173–180
38(4):598–605 perfusion pressure impairment: 156. Cammarata G, Ristagno G,
136. Lilly CM, Zuckerman IH, Badawi O, preliminary report. J Neurosurg Cammarata A, Mannanici G, Denaro
Riker RR (2011) Benchmark data 83:627–630 C, Gullo A (2011) Ocular ultrasound
from more than 240,000 adults that 147. Rohlwink UK, Zwane E, Fieggen AG, to detect intracranial hypertension in
reflect the current practice of critical Argent AC, Le Roux P, Figaji AA trauma patients. J Trauma 71:779–781
care in the United States. Chest (2012) The relationship between 157. Chen JW, Gombart ZJ, Rogers S,
140(5):1232–1242 intracranial pressure and brain Gardiner SK, Cecil S, Bullock RM
137. Effken JA, Loeb RG, Kang Y, Lin ZC oxygenation in children with severe (2011) Pupillary reactivity as an early
(2008) Clinical information displays to traumatic brain injury. Neurosurgery indicator of increased intracranial
improve ICU outcomes. Int J Med 70(5):1220–1230 pressure: the introduction of the
Inform 77:765–777 148. Whitmore RG, Thawani JP, Grady neurological pupil index. Surg Neurol
138. Koch S, Staggers N, Weir C, Agutter MS, Levine JM, Sanborn MR, Stein Int 2:82
J, Liu D, Westenskow D (2010) SC (2012) Is aggressive treatment of 158. Hartings JA, Bullock MR, Okonkwo
Integrated information displays for traumatic brain injury cost-effective? DO, Murray LS, Murray GD,
ICU nurses: field observations, display J Neurosurg 116:1106–1113 Fabricius M, Maas AI, Woitzik J,
design, and display evaluation. Proc 149. Diaz-Arrastia R (2012) Brain tissue Sakowitz O, Mathern B, Roozenbeek
Hum Fact Ergon Soc Annu Meet oxygen monitoring in traumatic brain B, Lingsma H, Dreier JP, Puccio AM,
54:932–936 injury (TBI) (BOOST 2). Shutter LA, Pahl C, Strong AJ (2011)
139. Morris G, Gardner R (1992) Computer http://clinicaltrials.gov/ct2/show/ Co-Operative Study on Brain Injury
Applications. In: Hall J, Schmidt G, NCT00974259. Accessed 20 May Depolarisations. Spreading
Wood L (eds) Principles of critical 2014 depolarisations and outcome after
care. McGraw-Hill, New York, 150. Hu X, Xu P, Asgari S, Vespa P, traumatic brain injury: a prospective
pp 500–514 Bergsneider M (2010) Forecasting ICP observational study. Lancet Neurol
elevation based on prescient changes 10(12):1058–1064
of intracranial pressure waveform
morphology. IEEE Trans Biomed Eng
57(5):1070–1078
1209

159. Ragauskas A, Bartusis L, Piper I, 160. Vespa PM (2005) Multimodality 161. Sivaganesan A, Manly GT, Huang MC
Zakelis R, Matijosaitis V, Petrikonis monitoring and telemonitoring in (2014) Informatics for neurocritical
K, Rastenyte D (2014) Improved neurocritical care: from microdialysis care: challenges and opportunities.
diagnostic value of a TCD-based non- to robotic telepresence. Curr Opin Crit Neurocrit Care 20(1):132–141
invasive ICP measurement method Care 11(2):133–138
compared with the sonographic ONSD
method for detecting elevated
intracranial pressure. Neurol Res
36:607–614

S-ar putea să vă placă și