Sunteți pe pagina 1din 354

Multidisciplinary Management

of Rectal Cancer
Vincenzo Valentini
Hans-Joachim Schmoll
Cornelis J.H. van de Velde
Editors

Multidisciplinary
Management of Rectal
Cancer
Questions and Answers
Editors
Vincenzo Valentini Cornelis J.H. van de Velde
Department of Radiation Therapy Leiden University
Università Cattolica S.Cuore Medical Center
Rome Leiden
Italy Netherlands

Hans-Joachim Schmoll
Klinik und Poliklinik Innere Medizin IV
Abt. Hämatologie und Onkologie
Universitätsklinikum Halle-Wittenberg
Halle, Sachsen-Anhalt
Germany

ISBN 978-3-642-25004-0 ISBN 978-3-642-25005-7 (eBook)


DOI 10.1007/978-3-642-25005-7
Springer Heidelberg New York Dordrecht London

Library of Congress Control Number: 2012940207

© Springer-Verlag Berlin Heidelberg 2012


This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or
part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way,
and transmission or information storage and retrieval, electronic adaptation, computer software,
or by similar or dissimilar methodology now known or hereafter developed. Exempted from this
legal reservation are brief excerpts in connection with reviews or scholarly analysis or material
supplied specifically for the purpose of being entered and executed on a computer system, for
exclusive use by the purchaser of the work. Duplication of this publication or parts thereof is
permitted only under the provisions of the Copyright Law of the Publisher’s location, in its
current version, and permission for use must always be obtained from Springer. Permissions for
use may be obtained through RightsLink at the Copyright Clearance Center. Violations are liable
to prosecution under the respective Copyright Law.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
While the advice and information in this book are believed to be true and accurate at the date of
publication, neither the authors nor the editors nor the publisher can accept any legal responsibility
for any errors or omissions that may be made. The publisher makes no warranty, express or
implied, with respect to the material contained herein.

Printed on acid-free paper

Springer is part of Springer Science+Business Media (www.springer.com)


Foreword

During the past few decades there have been many advances in the
management of rectal cancer. Building on a more comprehensive
understanding of anatomy and patterns of local recurrence, new surgical
techniques such as total mesorectal resection and sphincter sparing coloanal
anastomosis have become standards. The move toward preoperative adjuvant
therapy has been facilitated by more effective chemoradiation programs.
Advances in radiation planning, delivery, and fractionation techniques
coupled with new cytotoxic and targeted chemotherapeutic agents hold the
promise of reduced toxicity and increased tumor response and control rates.
New diagnostic modalities such as high resolution MRI have helped identify
which therapeutic approaches and modalities are best suited to an individual
tumor, allowing a more selective approach. Lastly, a renewed focus on expert
pathologic analysis coupled with the evolving field of prognostic and
predictive molecular markers has facilitated the development of surrogate
endpoints of response.
Although each discipline has made their individual diagnostic and
therapeutic contributions, the cornerstone of success has been the unified
movement toward multidisciplinary management. It is the collaborative
efforts of surgeons, radiation oncologists, medical oncologists, radiologists,
and pathologists which have truly had the most significant impact on
outcome.
This exciting new book is a unique contribution to the field of rectal cancer.
In contrast to the traditional didactic approach, each chapter directly engages
the reader with timely questions and answers. Building on the value of
multidisciplinary management, Professors Valentini, Schmoll, and Van de
Velde have assembled an internationally known group of contributors from a
number of European centers of excellence. Broad areas of expertise include
risk factors, imaging, radiotherapy, chemotherapy, surgery, and pathology.
The advances of the past three decades as well as new emerging controversies
are discussed.
The editors have succeeded in providing us with the foundation, relevant
data, and guidance to multidisciplinary management of rectal cancer. This
team approach sets the standard for modern cancer management.

Chicago, IL, USA Bruce D. Minsky, M.D.

v
Preface

In an era where all patients are entitled to access healthcare systems that
enable the highest quality of treatment delivered within a safe healthcare
environment, and access to appropriate advice, support and long term follow-
up, the multidisciplinary team is of central importance and a critical require-
ment in the development of modern oncology.
Joint efforts of different specialists involved in the diagnosis, staging,
treatment and evaluation of outcomes in rectal cancer throughout Europe to
promote mutual understanding and collaboration by managing multidisci-
plinary consensus conferences (EURECA-CC1-2) and the publication of
their recommendation were undertaken.
This was extended to a multidisciplinary teaching course (5 editions across
Europe and 1 in China) and culminated to the endorsement of these activities
by European cancer societies like ESTRO, ESSO and ESMO and hence cre-
ating the background to the holistic approach in promoting the multidiscipli-
narity of this book.
The aim of this book is to report the most common questions that arose in
the practice of a multidisciplinary team, devoted to address the health request
of patients with rectal cancer. A recognised group of clinicians, mostly
involved in the management of the more significant trials published in Europe
in the last decade, were requested to provide simple and focused answers to
support the best choices in a multidisciplinary setting. We are very grateful to
their enthusiastic and fully supportive participation to this project.
With this book we hope to contribute to improve the overall care of the
patient, supporting the multidisciplinary teams in their unique responsibility
for patient’s on-going care and wellbeing.

Vincenzo Valentini
Hans-Joachim Schmoll
Cornelius J.H. van de Velde

vii
Contents

Part I Introduction

1 What Do We Consider Cancer of the Rectum?. . . . . . . . . . . . . 3


Marilyne M. Lange and Cornelis J.H. van de Velde
2 What Is the Ongoing Recommendation in the
Management of Rectal Cancer? . . . . . . . . . . . . . . . . . . . . . . . . . 9
Vincenzo Valentini, Hans-Joachim Schmoll,
and Cornelis J.H. van de Velde

Part II Q&As on Risk Factor Identification

3 What Prognostic Clinical Factors Must Be Considered


Before Treatment? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
Claus Rödel
4 What Are the Relevant Imaging Factors to Optimize
Treatment Decisions?. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
Manish Chand and Gina Brown
5 What Biochemical and Molecular Biological Factors
Have Greater Relevance to Treatment Decisions? . . . . . . . . . . 41
Guido Lammering and Jeroen Buijsen
6 Do Different Populations of Rectal Cancer Exist? . . . . . . . . . . 49
Vincenzo Valentini, Francesco Cellini, Maria Cristina Barba,
and Ruud van Stiphout

Part III Q&As on Imaging

7 How Can We Identify Tumour Penetration?. . . . . . . . . . . . . . . 59


Manish Chand and Gina Brown
8 How Can We Identify Mesorectal Fascia Involvement?. . . . . . 67
Regina G.H. Beets-Tan
9 How Can We Identify Nodal Involvement? . . . . . . . . . . . . . . . . 73
Regina G.H. Beets-Tan
10 How Can We Identify Pathologic Complete Responders
After Radiochemotherapy?. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
Brunella Barbaro and Lucia Leccisotti

ix
x Contents

11 How Can We Identify Local Relapse? . . . . . . . . . . . . . . . . . . . . 95


Doenja M.J. Lambregts and Regina G.H. Beets-Tan

Part IV Q&As on Radiotherapy

12 When Should Preoperative Short-Course Radiotherapy


or Long-Course Chemoradiotherapy Be Performed? . . . . . . . 105
David Sebag-Montefiore and Robert Glynne-Jones
13 Should We Tailor the Delineation of Pelvic Structures
According to Tumor Presentation? . . . . . . . . . . . . . . . . . . . . . . 117
Maria Antonietta Gambacorta and Vincenzo Valentini
14 What Is the Role of IMRT and IGRT in Rectal Cancer?. . . . . 129
Jasper Nijkamp, Karin Haustermans, and Corrie A.M. Marijnen
15 What Are the Dose-Volume Constraints to Reduce
Late Toxicity? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149
Krzysztof Bujko
16 What Is the Contribution of Intraoperative Radiotherapy
(IORT) in Tailoring Local Therapy in Primary or Recurrent
Rectal Cancer? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155
Felipe A. Calvo
17 What Is the Contribution of Brachytherapy in Tailoring
Local Therapy? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163
Jean-Pierre Gérard, Te Vuong, Jean-Michel Hannoun-Lévi,
and Arthur Sun Myint

Part V Q&As on Chemotherapy

18 Should Oxaliplatin Be Added to Preoperative


Chemoradiation? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173
Carlo Aschele
19 Should Biologic Targeted Agents Be Combined
with Preoperative Chemoradiation in Rectal Cancer? . . . . . . 181
Pieter-Jan Cuyle and Eric Van Cutsem
20 Should Upfront Chemotherapy Precede Preoperative
Chemoradiation and Surgery? . . . . . . . . . . . . . . . . . . . . . . . . . . 193
Carlos Fernandez-Martos and Xabier Garcia de Albeniz
21 How to Achieve Long-Term Survival in Patients
with Metastatic Rectal Cancer? . . . . . . . . . . . . . . . . . . . . . . . . . 205
Alexander Stein and Hans-Joachim Schmoll
22 Will Adjuvant Chemotherapy Improve Outcome
After Preoperative Chemoradiation?. . . . . . . . . . . . . . . . . . . . . 217
Bengt Glimelius and Peter Nygren
Contents xi

Part VI Q&As on Surgery

23 How to Evaluate the Quality of Surgery? Suggestions


for Critical Reading of Surgical and Pathological Reports . . . 229
Lars Påhlman
24 How Is Nerve-Sparing Surgery Well Performed?. . . . . . . . . . . 233
Zoran Krivokapic and Ivan Dimitrijevic
25 Is Laparoscopic Rectal Surgery the Gold Standard? . . . . . . . . 249
David Jayne and Laeeq Khan
26 Is a Diverting Stoma Always Necessary for a Low
Anterior Resection of a Rectal Cancer?. . . . . . . . . . . . . . . . . . . 257
Geerard L. Beets
27 Will Extralevator Abdominoperineal Excision Become
the New Gold Standard? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 261
Ingrid Martijnse, Nicholas West, Phil Quirke, Richard Heald,
Cornelius J.H. van de Velde, and Harm Rutten
28 Which Patients Do Benefit from Extended Resections
in Case of Locally Advanced Rectal Cancer? . . . . . . . . . . . . . . 275
Ralph L. Dudink, Miranda Kusters, and Harm Rutten
29 Can Standard Surgical Procedure Reliably Be Avoided
in Major Responders After Radio(chemo)therapy?. . . . . . . . . 291
Claudio Coco and Gianluca Rizzo

Part VII Q&As on Pathology

30 What Is the Correct Procedure for Handling


the Surgical Specimen? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
Nigel Scott
31 What Is the Prognostic Value of (y)pT and (y)pN?. . . . . . . . . . 319
Nadine Ectors
32 What Is the Prognostic Value of CRM Involvement?. . . . . . . . 327
Iris D. Nagtegaal
33 What Is the Prognostic Value of TRG? . . . . . . . . . . . . . . . . . . . 333
Fabio M. Vecchio

Part VIII Q&As on Multidisciplinary Team Management

34 What Are the Recommendations to Ensure a Successful


Multidisciplinary Team in Rectal Cancer? . . . . . . . . . . . . . . . . 341
Sujay Shah, Pawan Mathur, and Robert Glynne-Jones
xii Contents

35 What Is the Appropriate Timetable for Tailored


Follow-up? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 351
Lars Påhlman
36 How Should Data Be Shared and Rapid
Learning Health Care Promoted? . . . . . . . . . . . . . . . . . . . . . . . 355
Ruud van Stiphout, Erik Roelofs, Andre Dekker,
and Philippe Lambin
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 365
Part I
Introduction
What Do We Consider Cancer
of the Rectum? 1
Marilyne M. Lange and Cornelis J.H. van de Velde

Contents 1.1 Epidemiology


1.1 Epidemiology .............................................. 3
Rectal cancer constitutes one third of all col-
1.2 The Rectum ................................................ 4
orectal cancers, representing the cancer with the
1.2.1 Anatomy....................................................... 4
1.2.2 Function ....................................................... 5 second highest incidence and the second cause
of cancer death in the western society [1, 2]. An
1.3 Pathophysiology ......................................... 6
estimated 100,000 new cases of rectal cancer
1.4 Presentation ................................................ 7 are diagnosed each year in Europe. The inci-
1.5 Diagnosis ..................................................... 7 dence is increasing, mainly due to earlier detec-
References ................................................................. 7 tion and increasing age of the population, as the
highest incidence of rectal cancer is found in the
sixth and seventh decades. High incidence rates
are found especially in western world popula-
tions, i.e., Western Europe, North America and
Australia. This can probably be explained by a
combination of factors, including dietary pat-
terns with high amounts of red meat, obesity
and smoking [3]. The United States is the only
country with significantly decreasing incidence
rates in both males and females in the most
recent time period, which largely reflects detec-
tion and removal of precancerous lesions
through colorectal cancer screening [4]. Next to
M.M. Lange
Department of Surgery,
dietary and lifestyle factors, risk factors for rec-
Zaans Medical Center, tal cancer include inflammatory bowel disease
Koningin Julianaplein 58, and primary sclerosing cholangitis. Also, genetic
1500EE Zaandam, The Netherlands predisposition plays a role; however, rectal can-
e-mail: lange_marilyne@hotmail.com
cer most commonly occurs sporadically and is
C.J.H. van de Velde (*) inherited in only 5% of the cases. Five-year sur-
Leiden University Medical Center,
Albinusdreef 2,2300RC Leiden,
vival rate of rectal cancer is about 60% and
The Netherlands depends to a large extent on disease stage at
e-mail: c.j.h.van_de_velde@lumc.nl diagnosis [5].

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 3


DOI 10.1007/978-3-642-25005-7_1, © Springer-Verlag Berlin Heidelberg 2012
4 M.M. Lange and C.J.H. van de Velde

Fig. 1.1 Sagittal and transversal illustration of the male rectal artery (red), seminal vesicles (purple) (Illustrated
pelvis. Sympathetic and parasympathetic nerves (green), by J.F.M. Lange)
mesorectum (yellow), mesorectal fascia (blue), middle

1.2 The Rectum cancer as a tumour of which the major part is


located at or below the peritoneal reflection [10].
The anatomic relations and physiology of the rec- In women, the peritoneal reflection (4–7 cm from
tum makes rectal cancer treatment a potential cause the anal verge) can descend to 4 cm from the anal
of severe, long-term morbidity [6, 7]. In order to verge. The rectum forms an acute 90–115° ano-
comprehend rectal cancer, the principles of treat- rectal angle with the dorsally directed anal canal.
ment and its implications, it is necessary to under- This angle, widening during defaecation (more
stand the anatomy and the function of the rectum. than 130°), is caused by the puborectal sling of
the levator ani muscles, inserting just cranially to
the level of the mucocutaneous line, halfway the
1.2.1 Anatomy anal canal [11]. Circumferentially, the rectum is
surrounded by fatty and connective tissue, which
Anatomically, the rectum extends from the anal is known as the mesorectum (Fig. 1.1). Starting at
verge for about 12–15 cm, where it curves anteri- the sacral promontory, the mesorectum being most
orly and merges into the sigmoid. As a rule, one pronounced at the dorsal site of the rectum dimin-
third of the rectum is located intraperitoneally and ishes below the rectosacral fascia around the leva-
two thirds extraperitoneally. The definitions of tor ani muscles at the end of the distal third of the
rectum and low rectal cancer are highly variable. rectum. The mesorectum is enveloped by the vis-
Some publications define the rectum as 15 cm ceral rectal (pelvic, proper rectal) fascia, separat-
from the anal verge as measured by rigid endos- ing it from the parietal endopelvic fascia. In
copy, defining low rectal cancer within 5 cm from between is a dorsal layer of thin fat, containing
the anus [8, 9]. Other definitions are related to autonomic nerves to the pelvic organs, and the
anatomy rather than endoscopic measurement. more ventral retrorectal space, filled with loose
These define the rectum as located below the bor- areolar tissue (‘holy plane of Heald’). The midline
der of the second sacral vertebra and low rectal hindgut (rectum) and the mesorectum, containing
1 What Do We Consider Cancer of the Rectum? 5

Fig. 1.2 Three-dimensional reconstruction of the male pelvis. The levator ani nerve, running just cranially to the pelvic
floor (green), is closely related to the mesorectum (light blue)

its vessels, fat and most of its lymph glands, are mesenteric nodes and then the para-aortic nodes.
embryologically derived together as a single unit. The lower lymph drainage is variable both proxi-
The anatomy and embryological origin is respected mally and laterally along the middle rectal ves-
by the current golden standard for rectal cancer sels towards nodes at the internal iliac vessels.
resection (total mesorectal excision, TME) as it The nerve supply to the pelvic organs, i.e., the
involves en bloc resection of the rectum and the rectum, vagina, uterus, vestibular bulbs, clitoris,
mesorectal tissue to the level of the levator muscles bladder, urethra, penis, prostate and pelvic floor,
within the embryologically determined, avascular are closely related to the rectum. From the supe-
plane outside the mesorectum between the parietal rior hypogastric plexus (at level L4-S1, at the level
and visceral rectal fascia [12]. This allows for radi- of the promontory) within the aorta bifurcation,
cal resection of the tumour and preservation of the the two sympathetic hypogastric nerves descend
pelvic autonomic nerves which are essential for dorsally to the mesorectum, parallel to the ureters
urogenital and anorectal functions [6, 7]. towards the inferior hypogastric plexuses (plexi
The mesorectum is suspended to the pelvic pelvini), where they join the parasympathetic pel-
wall by: (1) the ‘lateral ligaments’ which are vic splanchnic nerves (nervi erigentes) coming
strands of condensed tissue, located ventrolater- from S2–4 (Fig. 1.1). The levator ani nerve, which
ally to the rectum, at the level of the seminal ves- is responsible for the innervation of the levator ani
icles in men, containing the middle rectal blood muscle, also arises from S3–4 and runs over the
vessels and lymphatics [13]—these adhere close surface of the pelvic floor muscles, only covered
to the sympathetic and parasympathetic inferior by the parietal fascia [14] (Fig. 1.2).
hypogastric plexuses—(2) the rectosacral fascia,
just cranially to the pelvic floor, at the anorectal
junction at level S4; (3) levator ani complex, cov- 1.2.2 Function
ered by fat and the parietal rectal fascia.
The arterial supply of the rectum is supported The anorectum is responsible for maintaining
by the superior rectal (haemorrhoidal) artery, rep- faecal continence and, when socially appropriate,
resenting the inferior mesenteric artery after defaecation. This is possible as the rectum has a
spring-off of the left colic and sigmoid arteries. capacity to store an amount of faeces, acting as a
The inferior haemorrhoidal arteries from the reservoir [15]. Furthermore, the anal canal con-
internal iliac and the middle rectal artery also tains a rich network of nerve endings sensitive to
contribute blood to the rectum. Venous return fol- pain, temperature and touch, which is used to dif-
lows the arteries. The lymphatic drainage mirrors ferentiate solid or liquid stool from flatus, and
its vasculature. The first nodal level is located in allows for selective passage of flatus. The anal
the mesorectum, draining mostly to the inferior sphincters keep the anal canal closed in a resting
6 M.M. Lange and C.J.H. van de Velde

state. In addition to the resting anal pressures, the entiated and with an unusual histologic type
mesenteric plexus of the internal anal sphincter (mucinous and marked intra- and peritumoural
enables the recto-inhibitory reflex, which implies lymphocytic infiltration). It is also the hallmark
relaxation of the internal anal sphincter in of hereditary nonpolyposis colorectal cancer,
response to increased pressure in the rectum. And HNPCC (Lynch syndrome). It has been observed
finally, the pelvic floor (levator ani muscles) is that MSI is most common in (right-sided) colon
responsible for the anorectal angle, flattening cancer and rare in rectal carcinoma [19].
during defaecation [16]. Nevertheless, compared with colon cancer, the
number of mutations detected is significantly
higher in rectal cancer [20]. Furthermore,
1.3 Pathophysiology cyclooxygenase-2 (COX2) is overexpressed in
90% of rectal tumours but in only 20% of colonic
The majority of rectal cancers develop from tumours [21]. These genetic characteristics
benign preneoplastic lesions: the adenomatous confirm that rectal cancer is a different entity in
polyps or adenomas. Polyps are histologically colorectal cancer.
classified as tubular (5% malignant), villous In the progression of rectal cancer microenvi-
(40% malignant) or mixed (20% malignant), ronmental interactions are important. Loss of
depending on glandular structure. Degree of cell adhesion leads to reorganisation of epithe-
dysplasia (atypical cells) is graded: chance of lial cells to make invasion and metastasis possi-
malignancy varies from about 5% (low grade) to ble [22]. Angiogenesis is vital for tumour growth
about 35% (high grade). Risk of malignancy is and is mediated by multiple molecules, such as
also collated with size: 90% of adenomas are less vascular endothelial growth factor (VEGF),
than 1 cm (1% risk of malignancy), 10% are big- which are released by tumour cells [23]. For a
ger than 1 cm (about 10% malignant). Progression full understanding of the process of normal cells
from a benign adenoma to a malignant carcinoma becoming malignant tumours, all the genetic
passes through a series of well-defined histologi- pathways and mechanisms need to be
cal stages, which is referred to as the adenoma- identified.
carcinoma sequence. Two major mechanisms of Direct spread of rectal cancer occurs intramu-
genomic instability lead to colorectal carcinoma rally and radially, resulting in invasion of adjacent
development and progression: chromosomal tissues or organs. Indirect spread through lymph
instability (CIN) and microsatellite instability and blood vessels was first described by Harrison
(MSI). The former mechanism is associated with Cripps in 1890 [24]. Consequently, his pupil,
a series of genetic changes that involve the acti- William Ernest Miles stretched the importance of
vation of oncogenes (uncontrolled cell growth; resecting the rectal tumour en bloc with its
k-ras gene) and inactivation of tumour suppres- mesorectum, lymph nodes and blood supply,
sor genes (uninhibited growth; APC gene, p53 introducing the first curative resection for rectal
gene, DCC/SMAD4 gene) and contributes pre- cancer [25]. Lymphatic spread occurs in stepwise
dominantly to carcinogenesis in the rectum [17, progression. Skip metastases appear in less than
18]. Familial adenomatous polyposis (FAP) and 5%. Haematogenous spread is the most important
its attenuated variant (AFAP) represents the pattern of spread, most commonly involving the
(hereditary) syndrome dealing with APC muta- liver. However, rectal cancer may also metastasise
tion. The MSI-pathway, in which mutations in initially to the lungs because the inferior rectal
DNA mismatch repair (MMR) genes result in a vein drains into the inferior vena cava rather than
failure to repair errors that occur during DNA into the portal venous system. Other infrequent
replications in repetitive sequences (microsatel- sites are the adrenal glands, kidneys, bones and
lites), results in an accumulation of frameshift brain. In addition, spread within the peritoneal
mutations. This failure leads to an MSI type of cavity happens, initially close to the tumour with
tumour, which is more frequently poorly differ- small nodules arising from cells shed from the pri-
1 What Do We Consider Cancer of the Rectum? 7

1.5 Diagnosis

Rectal cancer can be suspected from the symp-


toms and signs described above or may be asymp-
tomatic and discovered by routine screening
(faecal occult blood testing, colonoscopy).
Histological confirmation is sought through
colonoscopy and biopsy. In patients in whom
colonoscopy is impossible, computed tomogra-
phy (CT) colonoscopy can provide radiographic
diagnosis. The entire large bowel should be
examined for the presence of synchronous
Fig. 1.3 Ulcerative rectal carcinoma. Resection specimen
after abdominoperineal resection for ulcerative rectal car- lesions. Magnetic resonance imaging (MRI) and
cinoma located at the anal verge endorectal ultrasound (EUS; differentiate T1
from T2) is used for staging and evaluating
locoregional disease and predicting if negative
mary tumour. Later, plaques become more wide- surgical margins can be achieved, which is the
spread, omentum is involved and ascites is case in approximately 75% [28]. Colonic tomog-
produced. Peritoneal involvement is a poor prog- raphy and/or abdominal ultrasound are used to
nostic factor (median survival less than 6 months) identify extrapelvic metastases. Furthermore, a
[26, 27]. thorax x-ray is performed to identify lung metas-
tasis. Once the diagnosis is established and the
local and distant extent of disease spread is deter-
1.4 Presentation mined, therapy and prognosis are discussed in a
multidisciplinary setting. Modern multimodal
Next to polypoidal disease, a rectal carcinoma treatment of rectal cancer attempts to integrate
can appear as an atypical ulcer, with rolled edges surgery, radiotherapy and chemotherapy and uses
and a necrotic base (Fig. 1.3). This tends to the expertise and knowledge of pathology and
infiltrate more deeply and is more likely to perfo- radiology to optimise oncologic and functional
rate. Also stenosing or annular lesions have been results.
described. Lastly, rectal cancer can be a diffuse
infiltrative disease, appearing as an extensive
lesion infiltrating the bowel wall over many cen- References
timetres. The macroscopic appearance influences
the symptomatology of rectal cancer. Patients 1. Ferlay J et al (2007) Estimates of the cancer incidence
present either electively or as an emergency with and mortality in Europe in 2006. Ann Oncol
obstruction or perforation. Obstruction leads to 18:581–592
2. Boyle P, Ferlay J (2005) Cancer incidence and mortal-
abdominal distension, pain, nausea and vomit-
ity in Europe, 2004. Ann Oncol 16:481–488
ing. Elective symptoms include altered bowel 3. Jemal A et al (2011) Global cancer statistics. CA
habit, rectal bleeding, mucus discharge, abdomi- Cancer J Clin 61:69–90
nal pain, fatigue, weight loss and palpable 4. Edwards BK et al (2010) Annual report to the nation
on the status of cancer, 1975–2006, featuring colorec-
abdominal mass. Less common symptoms
tal cancer trends and impact of interventions (risk fac-
include tenesmus, in case of pelvic floor involve- tors, screening, and treatment) to reduce future rates.
ment, and neuropathic pain syndrome due to sci- Cancer 116:544–573
atic or obturator nerve involvement in locally 5. Zampino MG et al (2004) Rectal cancer. Crit Rev
Oncol Hematol 51:121–143
advanced disease. Approximately 20% of patients
6. Lange MM, van de Velde CJ (2008) Faecal and uri-
have distant metastatic disease at the time of pre- nary incontinence after multimodality treatment of
sentation [27]. rectal cancer. PLoS Med 5:e202
8 M.M. Lange and C.J.H. van de Velde

7. Lange MM, van de Velde CJ (2011) Urinary and sex- 18. Vogelstein B et al (1988) Genetic alterations during
ual dysfunction after rectal cancer treatment. Nat Rev colorectal-tumor development. N Engl J Med 319:
Urol 8:51–57 525–532
8. Martling A, Holm T, Johansson H, Rutqvist LE, 19. Nilbert M, Planck M, Fernebro E, Borg A, Johnson A
Cedermark B (2001) The Stockholm II trial on preoper- (1999) Microsatellite instability is rare in rectal carci-
ative radiotherapy in rectal carcinoma: long-term follow- nomas and signifies hereditary cancer. Eur J Cancer
up of a population-based study. Cancer 92:896–902 35:942–945
9. Kapiteijn E et al (1999) Total mesorectal excision 20. Frattini M et al (2004) Different genetic features asso-
(TME) with or without preoperative radiotherapy in ciated with colon and rectal carcinogenesis. Clin
the treatment of primary rectal cancer. Prospective Cancer Res 10:4015–4021
randomised trial with standard operative and histo- 21. Dimberg J, Samuelsson A, Hugander A, Soderkvist P
pathological techniques. Dutch ColoRectal Cancer (1999) Differential expression of cyclooxygenase 2 in
Group. Eur J Surg 165:410–420 human colorectal cancer. Gut 45:730–732
10. Czito BG, Willett CG (2010) Rectal cancer: interna- 22. Takeichi M (1991) Cadherin cell adhesion receptors as
tional perspectives on multimodality management. a morphogenetic regulator. Science 251:1451–1455
Humana Press, New York 23. Sinicrope FA et al (1996) Increased apoptosis accom-
11. Lange JF, Kleinrensink GJ (2002) Surgical anatomy panies neoplastic development in the human colorec-
of the abdomen. Elsevier, Maarssen tum. Clin Cancer Res 2:1999–2006
12. Heald RJ (1979) A new approach to rectal cancer. Br 24. Cripps H (1890) On diseases of the rectum and the
J Hosp Med 22:277–281 anus. J&A Churchill, London
13. Kusters M et al (2010) Origin of presacral local recur- 25. Miles WE (1971) A method of performing abdomino-
rence after rectal cancer treatment. Br J Surg perineal excision for carcinoma of the rectum and of
97:1582–1587 the terminal portion of the pelvic colon (1908). CA
14. Wallner C et al (2008) The contribution of the levator Cancer J Clin 21:361–364
ani nerve and the pudendal nerve to the innervation of 26. MacKay GJ, Dorrance HR, Richard GM, O’Dwyer PJ
the levator ani muscles; a study in human fetuses. Eur (2010) Colorectal surgery. Oxford University Press,
Urol 54:1136–1142 Oxford
15. Lange MM et al (2007) Risk factors for faecal incon- 27. Wibe A et al (2002) Prognostic significance of the cir-
tinence after rectal cancer treatment. Br J Surg cumferential resection margin following total
94:1278–1284 mesorectal excision for rectal cancer. Br J Surg
16. Wallner C et al (2008) Causes of fecal and urinary 89:327–334
incontinence after total mesorectal excision for rectal 28. Lahaye MJ et al (2005) Imaging for predicting the risk
cancer based on cadaveric surgery: a study from the factors – the circumferential resection margin and
cooperative clinical investigators of the Dutch total nodal disease – of local recurrence in rectal cancer: a
mesorectal excision trial. J Clin Oncol 26:4466–4472 meta-analysis. Semin Ultrasound CT MR 26:259–268
17. Conlin A, Smith G, Carey FA, Wolf CR, Steele RJ
(2005) The prognostic significance of K-ras, p53, and
APC mutations in colorectal carcinoma. Gut
54:1283–1286
What Is the Ongoing
Recommendation in the 2
Management of Rectal Cancer?
Vincenzo Valentini, Hans-Joachim Schmoll,
and Cornelis J.H. van de Velde

Contents During the past 20 years, we have seen major


References ................................................................. 17 changes in the way patients with rectal cancer are
investigated and treated. The key components are
the improvements in preoperative staging, in sur-
gical technique and histopathological assessment
of the resected specimen, and in combining mul-
timodality treatments to ameliorate the long-term
outcome.
The concept of “mesorectal excision,” where
meticulous dissection of the anatomical plane
surrounding the mesorectal fat, is of crucial
importance and has been reported to result in
significantly lower rates of local recurrence in
specialist centers, population-based audit, and
within the framework of randomized controlled
trials. By far, surgery remains the most important
treatment of rectal cancer; nevertheless, the man-
agement of this disease has evolved to become
multidisciplinary.
Conclusive studies established the ability of
MRI to demonstrate the relationship of the mac-
roscopic tumor to the surrounding well-defined
V. Valentini (*)
anatomical plane of surgical excision. This
Department of Radiotherapy,
Università Cattolica Sacro Cuore, Rome, Italy development is closely linked with the ability of
e-mail: vvalentini@rm.unicatt.it relatively simple histopathological techniques
H.J. Schmoll to assess and measure the distance of micro-
Department for Internal Medicine, scopic tumor to the circumferential resection
Oncology and Hematology, margin (CRM) of the resected rectal cancer
University Clinic Halle (Saale),
specimen, providing the choice of optimal sur-
Halle, Germany
gical planes. MRI showed a great reliability in
C.J.H. van de Velde
predicting mesorectal fascia (MRF) involvement,
Department of Surgery,
Leiden University Medical Center, and more properly it was recently proposed to
Leiden, The Netherlands report it as MRF +/−.

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 9


DOI 10.1007/978-3-642-25005-7_2, © Springer-Verlag Berlin Heidelberg 2012
10 V. Valentini et al.

In the last decade, several European phase III times a web-based document customized for the
trials evaluating the role of radiotherapy and che- consensus process. A meeting was openly held to
motherapy in rectal cancer have been published. debate by attendees the more controversial sen-
From these trials, the efficacy of both short-course tences. The total number of voted sentences was
preoperative radiotherapy and preoperative con- 207. Of the 207, 86% achieved large consensus,
current chemoradiotherapy was determined [1–7]. 13% achieved moderate consensus, and only 3 (1%)
Anyway, although the findings of large random- resulted in minimum consensus. The document
ized trials have addressed important questions, addresses a wide range of topics relating to the man-
there remain patient care issues that cannot be agement of rectal cancer and, of equal importance,
addressed by subgroup analyses of existing trials identifies areas where future research is a priority.
and large areas of controversies are still in place. A second document was addressed by ESMO
To support physicians to deliver more tailored inside their program of organ oriented guide-
choices—as the oncology profession moves into lines. A group of experts invited by ESMO
the era of individualized medicine—some debated in a 2-day meeting the key issues about
European Consensus guidelines were proposed the management of rectal and colon cancer and a
under the collaboration of the major Oncology document circulated between them till a final
Society ESTRO (European SocieTy of Radio- approval [9].
therapy and Oncology), ESSO (European Society To get more information about the different
of Surgical Oncology), and ESMO (European strategies regarding staging and treatment of rec-
Society of Medical Oncology) [8, 9]. tal cancer, we recommend the reading of these
Two papers summarized these efforts: The two documents as well as the different answers to
EURECA project elaborated the Consensus the main questions, which arose in a multidisci-
document using the Delphi method [8]. A group plinary group in the daily management of rectal
of experts delegated by the three Oncology cancer patients, reported in this book (Figs. 2.1–2.10
Societies voted sentence by sentence for three and Tables 2.1–2.3). In this chapter, we tried to

Clinical Rectal Cancer


presentation

T stage
MRF Sphincter
Aim Location N stage Metastases
involvement involvement

T1 T2–3 T4

1° Abdomen MDCT
Choice ERUS MRI MRI MRI MRI MRI +
MRI
Exam chest –X ray or
CT (to be preferred)

MDCT
IF 1° Rigid ERUS MDCT MDCT high
choice ERUS slow ERUS mid ERUS
proctocopy high
exam is rectum mid rectum rectum
not
available

Flexible

endoscopy MDCT Multidetector CT ERUS Endorectal US MRF Mesorectal Fascia
choice

Fig. 2.1 Imaging work-up


2 What Is the Ongoing Recommendation in the Management of Rectal Cancer? 11

Clinical cT1 N0 M0
stage

Local
TME
Primary excision
treatment

pT1NX
Pathological pT1NX high risk > pT2 or pN + pT1–2 N0
report Margin - features
or > pT1N0

Adjuvant Chemotherapy
Observation Observation
treatment + radiochemotherapy
(see table 20.1)

Fig. 2.2 Treatment strategy: cT1 N0 M0

Clinical cT2 N0 M0
stage

TME
Primary
treatment

pT3, CRM−, pN0


Pathological pT1−2 N0 pT2−3 and (CRM+ or pN+)
High Middle rectum
report

Adjuvant Observation Observation Chemotherapy


treatment + radiochemotherapy
(see Table 2.1)

Fig. 2.3 Treatment strategy: cT2 N0 M0


12 V. Valentini et al.

Clinical cT3 (MRF-) N0 -2 M0


stage

Preoperative
RT chemotherapy
Preoperative long course
Primary RT short course (see Table 2.1)
treatment

2−3 days 6−8 weeks

TME TME

Pathological
report CRM− CRM+ CRM− CRM+

Adjuvant Chemo+ Adjuvant


Adjuvant
treatment Observation RT chemo Observation chemo
chemo
A according to
(see Table 2.2)
(see Table 2.1) nomogram*
(see Table 2.2)

*V. Valentini JCO 2011 MRF Mesorectal Fascia

Fig. 2.4 Treatment modalities: cT3 (MRF−) N0–2 M0

Clinical
cT3 (MRF+) N0−2−M0 or cT4 any N M0
stage

Preoperative
RT chemotherapy
long course
Primary (see Table 2.1)
treatment

6–8 weeks

TME

Adjuvant
Adjuvant chemotherapy
treatment (see Table 2.2)

MRF Mesorectal Fascia

Fig. 2.5 Treatment modalities: cT3 (MRF+) N0–2–M0 or cT4 any N M0


2 What Is the Ongoing Recommendation in the Management of Rectal Cancer? 13

Clinical cT3 ab N0 M0 cT3 MRF+ M0


stage middle-upper cT3 (MRF–) N0–1 M0
cT4 any N M0
rectum

6−8 weeks
Preoperative
Preoperative
RT chemotherapy
RT short course
long course
TME
Primary
treatment 6−8 weeks

Preoperative
Restaging
CRM + chemotherapy
CRM− > clinical response?
or PN +
Pathological
report
Local excision TME

pT0 Nx pT1–2–3
CHEMO+
RT Chemo Adjuvant chemotherapy
Adjuvant Observation Observation TME (see Table 2.2)
(see Table 2.1)
treatment
pT0–2 N0 pT3 N+

Observation Adjuvant
Chemotherapy (seeTable 2.2)

MRF Mesorectal Fascia

Fig. 2.6 Treatment modalities under clinical evaluation

Clinical Local recurrence


stage

No previous RT Previous RT
for the primary for the primary

RT chem Resecability

Primary
treatment Resecability
Yes No

Yes No
Surgery RT chem Chemotherapy RT chem

Chemotherapy B
Resection + IORT Surgery
(see Table 2.2) Adjuvant Resecability
Chemotherapy
(see Table 2.2)
Yes No
adjuvant
chemotherapy
Adjuvant – Adjuvant (see Table 2.2) Surgery Chem
maintenance Chemotherapy B
treatment
Chem

Fig. 2.7 Treatment modalities local recurrence, cM0


14 V. Valentini et al.

Syncronous cancer with


intact primary and R0/R1
resectable metastases
Clinical
stage
< T3 N0
≥ T3 or N+

or
Preop RT (5 × 5) 3 months preop
3 months preop
Primary Or CRT FOLFOX
FOLFOX
treatment

3 months preop Preop RT (5 × 5)


FOLFOX

Resection of primary and met

Pathological Postop CRT if T3, CRM+,


report pN+, perforation

Adjuvant – 6 months postop


maintenance 3 months postop FOLFOX
FOLFOX
treatment

Fig. 2.8 Treatment modalities, cM1, resectable synchronous metastases

Syncronous unresectable met


Clinical with intact primary rectal cancer
stage
Upfront chemotherapy (see Table 2.3)

Primary
Resectability of mets achieved?
treatment

Yes No

< T3 N0 ≥ T3 or N+
Continue/change chemotherapy

Preop CRT or Resectability of mets achieved?


short course RT

Yes No
Resection of primary +
resection of metastases
Avoid radical and mutilating
Adjuvant – Resume initial treatment for a surgery
maintenance total of 6 months RCT or short course RT or brachiterapy
treatment for locally advanced tumors

Fig. 2.9 Treatment modalities, cM1, unresectable synchronous metastases


2 What Is the Ongoing Recommendation in the Management of Rectal Cancer? 15

Clinical Resectable liver/lung metastses


stage

Clearly R0 resectable Boderline/R0 resectable

Intensive chemotherapy
Single, <2 cm FOLFOX
3−4(−6) months
Primary liver met 3 months preop
(see Table 2.3)
treatment

Resection No resection

Adjuvant – R0/1 : complete


R0/1 : FOLFOX R0/1 : FOLFOX Escalate/change
maintenance periop. chemotherapy
6 months postop 3 months postop chemotherapy, than
treatment for total of 6 months resection, if possible

Fig. 2.10 Treatment modalities CM1, non-synchronous

Table 2.1 Chemotherapy-options and doses for concomitant chemotherapy during radiation
Regimen References
5FU 325–350 mg/m2 + LV 20 mg/m2 iv bolus, d1–5, week 1 and 5 [3, 10]
5FU 400 mg/m2 + LV 100 mg iv bolus, d1, 2, 11, 12, 21, 22 [7]
5FU 225 mg/m2 iv continuous infusion, 5 days per week, together [11, 12]
with radiotherapy
5FU 1,000 mg/m2 iv continuous infusion, d1–5, week 1 and 5 [1]
Capecitabine 800–825 mg/m2 bid po continuously, 5–7 days per week, [11, 13, 14]
together with radiotherapy
UFT (300–350 mg/m2/day) and LV (22.5–90 mg/day) po continu- [15–18]
ously, 5–7 days per week, together with radiotherapy
5FU 250 mg/m2 iv continuous infusion on days 1–14 and 22–35 and [19]
oxaliplatin 50 mg/m2 iv d1, 8, 22, 29, only preoperatively

Table 2.2 Standard adjuvant chemotherapy regimens in rectal cancer (number of cycles without chemoradiation are
given in brackets)
Regimen Cycles References
5FU 350–370 mg/m2, + LV 20–25 mg/m2 iv 4 (−6) [3, 20]
bolus, d1–5, q 4 weeks
5FU 500 mg/m2 iv, continuous infusion, d1–5, 4 [1]
q 4 weeks
5FU 500 mg/m2 + LV 100 mg, iv Bolus, d1, 2, 8 [7]
q 2 weeks
Capecitabine 2,000–2,500 mg/m2, po, d1–14, 5–6 (−8) [13, 21]
q 3 weeks
16 V. Valentini et al.

Table 2.3 Choice of first line treatment


2.3 a
Group Clinical presentation Treatment aim Treatment intensity
1 liver and/or lung metastases only
which • maximum upfront most active
• might become resectable after induction chemotherapy tumour combination regimen
• ±limited/localized metastases to other sites, e.g. locore- shrinkage
gional lymphnodes
• physically able to undergo major surgery (biological age,
heart/lung condition)
Group KRAS wildtype Recommenda KRAS mutant Recommenda
1 • FOLFIRI+Cet +++ • FOLFOX/XELOX+Bev +++
• FOLFOX+Pan/Cet +++ • FOLFOXIRI ++(+) b
• FOLFOX/XELOX+Bev ++(+) • FOLFIRI/XELIRI+Bev ++(+) c
• FOLFOXIRI ++(+)b • FOLFOX/XELOX +
• FOLFIRI/XELIRI+Bev ++(+) c • FOLFIRI/XELIRI +
• FOLFOX/XELOX + • IRIS +
• FOLFIRI/XELIRI +
• IRIS +
2.3 b
Group Clinical presentation Treatment aim Treatment intensity
2 multiple metastases/sites, with
• rapid progression and/or • clinically relevant tumour upfront active combination: at
• tumour-related symptoms/risk shrinkage as soon as possible least doublet
of rapid deterioration • at least achieve control of
• co-morbidity allows intensive progressive disease
treatment
Group KRAS wildtype Recommenda KRAS mutant Recommenda
2 • FOLFIRI+Cet +++ • FOLFOX/XELOX+Bev +++
• FOLFOX+Pan +++ • FOLFIRI/XELIRI+Bev ++(+)c
• FOLFOX/XELOX+Bev +++ • FOLFOX/XELOX ++
• FOLFIRI/XELIRI+Bev ++(+)c • FOLFIRI/XELIRI ++
• FOLFOXIRI +(+) b • FOLFOXIRI ++b
• FOLFOX+Cet +(+) • IRIS +
• FOLFOX/XELOX +
• FOLFIRI/XELIRI +
• IRIS +
2.3 c
Group Clinical presentation Treatment aim Treatment intensity
3 multiple metastases/sites, with
• never option for resection • abrogation of further treatment selection according to
• and/or no major symptoms or risk progression disease characteristics and
of rapid deterioration • tumour shrinkage less patients preference re toxicity
• and/or severe comorbidity relevant and efficacy:
(excluding from later surgery and/ • low toxicity most relevant • “watchful waiting”
or intensive systemic treatment, as • sequential approach: start with
for groups 1+2) – single agent, or
– doublet with low toxicity
• exceptional triplets
2 What Is the Ongoing Recommendation in the Management of Rectal Cancer? 17

Group KRAS wildtype Recommenda KRAS mutant Recommenda


3 • FUFOL/Cape (mono) +++ • FUFOL/Cape (mono) +++
• FUFOL/Cape+Bev +++ • FUFOL/Cape+Bev +++
• XELOX/FOLFOX ++ • XELOX/FOLFOX ++
• FOLFIRI/XELIRI ++ • FOLFIRI/XELIRI ++
• IRIS + • IRIS +
• Cet/Pan (mono) (+) • watchful waiting + selected pts.d
• watchful waiting + selected ptsd • triplets (+/-Bev) + option for spec.
triplets (+/-Bev or Cet/Pan) + option for spec. situations e
situationse
a
Rec: consented recommendation, however decision might be modified based on individual objective and subjective
parameters
b
FOLFOXIRI: only 2 (small) phase III trials with contradictory results
c
no randomized data for FOL(XEL)IRI + Bev
d
option in case of low tumor burden, asymptomatic, indolent disease: close control until definitive progression (not until
symptoms!)
e
Patients who are group 3 but deserve (and tolerate) more intensive treatment due to specific reasons

6. Sebag-Montefiore D, Stephens RJ, Steele R, Monson J,


summarize by charts the contents of those two Grieve R, Khanna S et al (2009) Preoperative radio-
documents, which largely represent the view of therapy versus selective postoperative chemoradio-
the experts of the three major European Oncology therapy in patients with rectal cancer (MRC CR07 and
Societies (ESTRO, ESMO, and ESSO), to facili- NCIC-CTG C016): a multicentre, randomised trial.
Lancet 373:811–820
tate the understanding of the approach to man- 7. Braendengen M, Tveit KM, Berglund A et al (2008)
agement of rectal cancer. Randomized phase III study comparing preoperative
radiotherapy with chemoradiotherapy in nonresect-
able rectal cancer. J Clin Oncol 26:3687–3694
8. Valentini V, Aristei C, Glimelius B et al (2009)
References Multidisciplinary Rectal Cancer Management: 2nd
European rectal cancer consensus conference
1. Sauer R, Becker H, Hoyhenberger W, For the German (EURECA-CC2). Radiother Oncol 92(2):148–163
Rectal Cancer Study Group et al (2004) Pre-operative 9. Schmoll HJ, Van Cutsem E, Stein A, Valentini V,
versus post-operative chemoradiotherapy for rectal Glimelius B, Haustermans K et al Standards for diagno-
cancer. N Engl J Med 351:1731–1740 sis and treatment of patients with colon and rectal can-
2. Gérard JP, Conroy T, Bonnetain F et al (2006) cer. ESMO consensus guidelines. Ann Oncol, in press
Preoperative radiotherapy with or without concurrent 10. Roh MS, Colangelo LH, O’Connell MJ et al (2009)
fluorouracil and leucovorin in T3–4 rectal cancers: Preoperative multimodality therapy improves disease-
results of FFCD 9203. J Clin Oncol 24:4620–4625 free survival in patients with carcinoma of the rectum:
3. Bosset JF, Collette L, Calais G, Mineur L, Maingon P, NSABP R-03. J Clin Oncol 27:5124–5130
Radosevic-Jelic L, EORTC Radiotherapy Group Trial 11. Roh JK, Yothers G (2011) The impact of capecitabine
22921 et al (2006) Chemotherapy with preoperative and oxaliplatin in the preoperative multimodality
radiotherapy in rectal cancer. N Engl J Med treatment in patients with carcinoma of the rectum:
355:1114–1123 NSABP R-04. J Clin Oncol 29:abstract 3503
4. Bujko K, Nowacki MP, Nasierowska-Guttmejer A, 12. O’Connell MJ, Martenson JA, Wieand HS et al (1994)
Michalski W, Bebenek M, Kryj M (2006) Long-term Improving adjuvant therapy for rectal cancer by combin-
results of a randomized trial comparing preoperative ing protracted-infusion fluorouracil with radiation ther-
short-course radiotherapy with preoperative conven- apy after curative surgery. N Engl J Med 331:502–507
tionally fractionated chemoradiation for rectal cancer. 13. Hofheinz RD, Wenz F (2011) Capecitabine (Cape)
Br J Surg 93:1215–1223 versus 5-fluorouracil (5-FU)–based (neo)adjuvant
5. Peeters KC, Marijnen CA, Nagtegaal ID, Kranenbarg chemoradiotherapy (CRT) for locally advanced rectal
EK, Putter H, Wiggers T et al (2007) Dutch Colorectal cancer (LARC): long-term results of a randomized,
Cancer Group the TME trial after a median follow-up phase III trial. J Clin Oncol 29:abstract 3504
of 6 years: increased local control but no survival 14. Gerard JP, Azria D, Gourgou-Bourgade S et al (2010)
benefit in irradiated patients with resectable rectal Comparison of two neoadjuvant chemoradiotherapy
carcinoma. Ann Surg 246:693–701 regimens for locally advanced rectal cancer: results of
18 V. Valentini et al.

the phase III trial ACCORD 12/0405-Prodige 2. radiotherapy (60 Gy) and UFT/l-leucovorin in patients
J Clin Oncol 28:1638–1644 with non-resectable locally advanced rectal cancer
15. Bystrom P, Frodin JE, Berglund A, Wilking N, (LARC). Acta Oncol 47:428–433
Glimelius B (2004) Phase I study of UFT plus leuco- 19. Roedel C, Becker H (2011) Preoperative chemoradio-
vorin with radiotherapy in patients with inextirpable therapy and postoperative chemotherapy with
non-rectal gastrointestinal cancer. Radiother Oncol 5-fluorouracil and oxaliplatin versus 5-fluorouracil
70:171–175 alone in locally advanced rectal cancer: first results of
16. Schiebe ME, Reese T, Wenz F et al (2002) Phase I the German CAO/ARO/AIO-04 randomized phase III
study of oral uracil and Tegafur plus leucovorin and trial. J Clin Oncol 29:abstract LBA3505
pelvic radiation in patients with recurrent rectal can- 20. Gray R, Barnwell J, McConkey C, Hills RK, Williams
cer. Anticancer Drugs 13:1005–1009 NS, Kerr DJ (2007) Adjuvant chemotherapy versus
17. Jakobsen A, Appelt AL (2011) The dose-effect rela- observation in patients with colorectal cancer: a ran-
tionship in preoperative chemoradiation of locally domised study. Lancet 370:2020–2029
advanced rectal cancer: preliminary results of a phase 21. Twelves C, Wong A, Nowacki MP et al (2005)
III trial. J Clin Oncol 29:abstract 3512 Capecitabine as adjuvant treatment for stage III colon
18. Vestermark LW, Jacobsen A, Qvortrup C et al (2008) cancer. N Engl J Med 352:2696–2704
Long-term results of a phase II trial of high-dose
Part II
Q&As on Risk Factor Identification
What Prognostic Clinical Factors
Must Be Considered Before 3
Treatment?

Claus Rödel

Contents 3.1 Patient-Related


3.1 Patient-Related Characteristics: Characteristics: Sex, Age,
Sex, Age, and Body Mass Index ................ 21 and Body Mass Index
3.2 Tumor-Related Characteristics................. 22
3.2.1 Tumor Size and Location ............................. 22 It has been observed in some reports that sur-
3.2.2 Clinical T- and N-Stage ............................... 24 vival after colorectal cancer resection is longer
3.2.3 Distance of the Tumor in women than men. With the use of the
to Mesorectal Fascia .................................... 24
Surveillance, Epidemiology, and End Results-
3.3 Histopathology on Pretreatment Medicare-linked database of a cohort of 30,975
Biopsies ....................................................... 25 colorectal cancer patients (8,350 with rectal can-
3.4 Pretreatment Laboratory Findings .......... 26 cer, treated from 1996 to 2003), one group from
References ................................................................. 26 Philadelphia, Pennsylvania, could show that for
both colon and rectal cancer, women presented
at an older age and underwent less aggressive
therapy but had longer adjusted survival com-
pared with men. In a recent pooled analysis from
five European clinical trials of preoperative con-
ventionally fractionated radiotherapy (with or
without 5-fluorouracil-based concurrent chemo-
therapy), female sex was also associated with a
more favorable overall survival, albeit there was
no difference in local or distant tumor control
between men and women (Table 3.1) [1].
Understanding of this difference is worthy of
further investigation; however, based on these
data, it is unlikely that there are gender-specific
differences in tumor response and rectal cancer-
specific survival rates. Conversely, several
reports have confirmed that acute organ toxicity
C. Rödel during chemoradiotherapy (CRT) is increased in
Department of Radiotherapy and Oncology, women. Gender differences in the dihydropy-
Johann Wolfgang Goethe-University Frankfurt,
rimidine dehydrogenase expression (the initial,
Theodor-Stern-Kai 7, Frankfurt am Main
60590, Germany rate-limiting enzyme in the catabolism of
e-mail: claus.roedel@kgu.de 5-fluorouracil) may contribute to higher toxicity

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 21


DOI 10.1007/978-3-642-25005-7_3, © Springer-Verlag Berlin Heidelberg 2012
22 C. Rödel

Table 3.1 Impact of clinical factors on local and distant control


5-year local 5-year distant 5-year overall
No. control (%) P control (%) P survival (%) P
Total no. of 2,795 87 69 70
patients
Sex
Male 1,961 87 n.s. 68 n.s. 68 <0.001
Female 843 89 72 74
Age
£49 378 85 n.s. 68 n.s. 72 <0.001
50–59 746 87 70 73
60–69 1,131 89 67 67
³70 540 90 73 69
Tumor location
Low 953 86 n.s. 64 0.001 65 0.001
Mid 1,461 88 71 72
High 369 91 75 74
cT stage
cT2 18 95 <0.001 66 n.s. 83 <0.001
cT3 2,228 89 70 71
cT4 275 78 64 57
cN stage
cN0 309 93 n.s. 78 0.009 78 0.006
cN1–2 544 94 72 74
Data from five European randomized trials on preoperative radiotherapy with or without 5-fu-based concurrent
chemotherapy

in women treated with 5-fluorouracil-based CRT. patients, who may require aggressive supportive
Furthermore, an increased risk of dyspareunia management to complete therapy.
and vaginal dryness was observed in women fol- An increased body mass index, particularly in
lowing surgery combined with (chemo-)radio- males, was associated with a decreased likelihood
therapy compared with women treated with of sphincter preservation and an increased risk of
surgery alone. local recurrences in a pooled analysis of 1,688
There is limited information on how elderly rectal cancer patients treated within postoperative
patients tolerate radiotherapy or combined CRT 5-fluorouracil-based CRT trials. Interestingly,
for rectal cancer. Recent analyses of the Dutch obese patients – both women and men – had a
TME trial and two large Dutch population-based significantly lower rate of grade 3–4 acute toxic-
registries showed that, unlike younger patients, ity (mainly leucopenia, stomatitis) when com-
patients >75 years of age did not benefit in terms of pared to normal-weight individuals, indicating
overall survival from the introduction of preopera- that actual body weight dosing of 5-fluorouracil
tive radiotherapy and TME surgery [2]. Randomized may be justified in obese patients [3].
trials in rectal cancer commonly include few
patients more than 75 years of age. Data from the
pooled analysis of the five European trials 3.2 Tumor-Related Characteristics
(Table 3.1) do not indicate worse outcome in terms
of local and distant control in the age cohorts from 3.2.1 Tumor Size and Location
below 50 years to above 70 years, respectively.
However, several nonrandomized and population- Through digital rectal examination (DRE, the
based datasets indicate higher rates of toxicity and average finger can reach approximately 8 cm
treatment deviation during CRT among elderly above the anal verge), tumors can be assessed for
3 What Prognostic Clinical Factors Must Be Considered Before Treatment? 23

Table 3.2 Impact of distance from anal verge by univariate analysis on local failure rates
Swedish rectal cancer trial
Local failure rate at 5 years (%)
Preop. RT Surgery P value
High >11 cm 8 12 n.s.
Middle 6–10 cm 9 26 <0.001
Low £5 cm 10 27 0.03
Dutch trial
High >10 cm 3.7 6.2 0.12
Middle 5.1–10 cm 3.7 13.7 <0.001
Low £5 cm 10.7 12 0.58
Medical Research Council MRC CR07
High >10–15 cm 1.2a 6.2a HR 0.19 (0.07–0.47)
Middle >5–10 cm 5.0a 9.8a HR 0.50 (0.28–0.9)
Low 0–5 cm 4.8a 10.4a HR 0.45 (0.23–0.88)
a
Three-year data

size, ulceration, and fixation to surrounding struc- randomized trials which were not stratified by
tures. DRE also permits a cursory evaluation of distance. These trials used short-course preopera-
the patient’s sphincter function, which is critical tive radiation and included patients with cT–
when determining whether a patient is a candi- –2N0 disease (Swedish Rectal Cancer Trial from
date for a sphincter-sparing procedure. Several the pre-TME era, Dutch Trial, Medical Research
studies identified the pretreatment tumor size Council MRC CR07) [4–6]. As seen in Table 3.2,
(<3 cm) as a significant factor for pathologic by univariate analysis, “high” tumors generally
complete response after preoperative chemora- had a lower incidence of local recurrence com-
diotherapy (CRT) in rectal cancer. Other known pared with mid and lower tumors. On multivari-
risk factors include circumferential lesions, teth- ate analysis, tumor location was an independent
ered or fixed tumors on palpation, near-obstruct- prognostic variable in the Dutch trial. The MRC
ing lesions, and low-lying tumors. Rigid CR07 trial also identified tumors involving the
proctosigmoidoscopy allows direct visualization anterior quadrant as an independent risk factor
of the lesion and provides an estimation of the for time to local recurrence, whereas distal extent
size of the lesion and degree of obstruction. This was not significantly associated with local recur-
procedure is used to obtain biopsies and gives an rence on multivariate analysis.
accurate measurement of the distance of the It is interesting to note that radiation did
lesion from the anal verge. significantly decrease local recurrence for all
The rectum is subdivided into three parts tumor positions in the MRC CR07 trial, whereas
according to the distance of the lower margin of this effect was significant only for middle and
the tumor from the anal verge: upper third low tumors in the Swedish and for middle
12–16 cm, middle third 6–£12 cm, and lower tumors only in the Dutch trial, respectively. If
third <6 cm. Alternative definitions (0–5 cm, analyzed as a continuous factor (rather than with
>5–10 cm, >10–15 cm) have also been used. The arbitrary cutoff points), the benefit of radiother-
anterior peritoneal reflexion represents the point apy in the Dutch trial became significant as the
at which the rectum exits the peritoneal cavity distance from anal verge increased. However, if
and becomes retroperitoneal (approximately patients with positive circumferential resections
8–12 cm from the anal verge, extremely variable margins (which occurs more often in distal rec-
between females and males). There are no pro- tal cancer) were excluded, the effect of radio-
spective randomized data examining the impact therapy on reduction of local recurrences was
of the distance from the anal verge on local recur- independent of the distance of the tumor from
rence. The available data are subset analysis from the anal verge.
24 C. Rödel

3.2.2 Clinical T- and N-Stage presence of mixed signal intensity and irregular-
ity of the borders of the lymph nodes may be more
The primary imaging modalities to assess the reliable. However, adequate clinical detection of
extent of the primary tumor (cT-, cN-status) are lymph node involvement remains a challenge that
endorectal ultrasound (ERUS), multidetector 4–16 is critically important with respect to the increas-
slice CT, and phased-array MRI. ERUS is the most ing use of preoperative treatment strategies.
accurate tool in predicting T stage of rectal can- It is clear that the most important prognostic
cers, especially T1 versus T2. Thus, ERUS has factors for survival are the extent of disease as
been recommended as the investigation of choice determined histopathologically after surgery
in selection of potentially curative local excision (with or without neoadjuvant treatment) by the
which should be restricted to patients with T1 degree of bowel wall penetration; the presence or
tumors without further risk factors, as well as in absence of lymph node metastases or distant
selection of patients with early T3 tumors (vs. T2) metastases, i.e., the (y)pTNM stage, as well as
for neoadjuvant treatment. ERUS cannot be reli- the tumor resections margins (R0, R1, R2; cir-
ably used in patients with high or stenosing tumors. cumferential resection margin £1 mm). The prog-
Of note, overestimation of staging with this tech- nostic value of the clinically determined cT- and
nique occurs more often than understaging. In the cN-stage is less validated. Figure 3.1a depicts the
German CAO/ARO/AIO-94 study, 18% of patients association of the pretreatment, clinical T- and
in the immediate surgery arm – clinically staged N-stage with disease-free survival within the
by ERUS to have cT3–4 and/or cN + disease – had German Rectal Cancer Study [8]. In this trial,
lymph node negative tumors confined to the rectal staging procedures included DRE, proctoscopy,
wall (pT1/2 pN0) on pathologic assessment of the endorectal ultrasound, and a pelvic CT scan (MRI
resected specimen. This probably is due to the was not mandatory). Only patients with cT3–4
inflammatory and desmoplastic processes caused and/or cN + disease were eligible. Interestingly,
by the tumor (or by taken tumor biopsy within clinical T and N staging was not significantly
1 week before ERUS was performed) but is also associated with long-term outcomes in this trial,
operator dependent. Endorectal coil MRT is an reflecting the inaccuracy of pretreatment staging,
alternative to ERUS but appears to be not superior especially with respect to lymph node involve-
to ERUS for T staging. Because of limited acous- ment. Data from the pooled analysis of the five
tic penetration by ERUS, invasion of bulky tumors European trials (Table 3.1), however, indicate
into the perirectal fat and adjacent organs and pel- that cT-stage and cN-stage are significantly asso-
vic side walls is better evaluated by multislice-CT ciated with the risk of local and distant failure,
scan and phased-array MRI. The involvement of respectively, albeit the absolute difference in out-
the anal sphincter and levator ani muscles cannot comes was only small. Conversely, positive
be truly seen on CT scans, whereas high-resolu- lymph nodes after preoperative CRT indicate
tion MRI techniques using phased-array coils have both an aggressive potential of the malignant
led to better spatial resolution and particularly cells to spread to the regional lymph nodes and
have been shown to identify the anal sphincter, the resistance of these cells toward CRT. As a
puborectalis, and particularly the mesorectal fas- result, these patients have a very unfavorable
cia. This latter is an important feature to predict prognosis and ypN0 versus ypN1–2 largely sepa-
negative circumferential margins between the rates different prognostic groups (Fig. 3.1b).
tumor and the mesorectal fascia and makes phase
array MRI superior to CT especially for lower
third rectal tumors [7]. 3.2.3 Distance of the Tumor
The identification of positive lymph nodes is to Mesorectal Fascia
more difficult. Involvement is mainly assessed by
size criteria (>8 mm), although enlarged lymph Histopathologic tumor involvement of the mesorec-
nodes are not pathognomonic of tumor involve- tal resection margin is a well-established indepen-
ment, and morphological features such as the dent predictor of local and distant metastases
3 What Prognostic Clinical Factors Must Be Considered Before Treatment? 25

Fig. 3.1 (a) Impact of clinical a


cT- and cN-stage on disease- 1.0 1.0
free survival after preoperative
chemoradiotherapy. (b) Impact
of pathologic ypT- and .8 cT2 (n=16) .8 cN+ (n=213)
ypN-stage on disease-free

Disease-free survival
survival after preoperative
.6 cT4 cT3 .6
chemoradiotherapy (Data from cN0 (n=154)
the German Rectal Cancer (n=24) (n=268)
Study)
.4 .4

.2 .2
P=0.92 P=0.34

0 0
0 20 40 60 80 100 0 20 40 60 80 100
Months Months
b
1.0 ypT1 1.0
ypT0
ypNO
.8 ypT2 .8
Disease-free survival

ypT3 ypN1
.6
ypT4 .6

.4
.4
ypN2
.2
P=0.001 .2
P<0.0001
0
0
0 20 40 60 80 100 0 20 40 60 80 100
Months Months

both after primary surgery and after preoperative prognostic tumors (e.g.,T4; predicted CRM
CRT/RT plus surgery. High-resolution phased- £1 mm, cN2; extramural venous invasion).
array MRI techniques are highly accurate for the
prediction of the circumferential resection margin
(CRM), especially CRM £1 mm versus >1 mm. 3.3 Histopathology on
Moreover, substaging of T3 tumors (T3a/b with Pretreatment Biopsies
tumor spread £5 mm into the mesorectal compart-
ment) has been validated prospectively by the The majority (over 90%) of rectal cancers are ade-
MERCURY study group and has shown direct nocarcinomas. Some adenocarcinomas have mucin
agreement between pretreatment imaging and which can be extracellular (colloid) or intracellular
corresponding pathology [9]. Both information (signet ring cell). Colloid cancer, which occurs in
may help to stratify patients for preoperative treat- 15–20% of adenocarcinomas, is not an indepen-
ment alternatives, including omission of RT in dent prognostic factor, whereas signet ring cell,
good prognostic tumors (e.g., cT3a/b, cN0, mid- which occurs in 1–2% of adenocarcinomas, is an
dle/upper part of rectum, predicted CRM >1 mm) independent poor prognostic factor for survival.
or short-course RT versus CRT in intermediate Other histological types are rare and include carci-
(e.g., >cT3a/b, predicted CRM >1 mm) and poor noid tumors, leiomyosarcomas, lymphoma, and
26 C. Rödel

squamous cell cancers. The grading system used References


for adenocarcinomas refers to the degree of dif-
ferentiation. Some institutions use a three-grade 1. Valentini V, van Stiphout RG, Lammering G et al
system (well, moderate, poor), and others use a (2011) Nomograms for predicting local recurrence,
distant metastases, and overall survival for patients
four-grade system. Despite substantial inter- and with locally advanced rectal cancer on the basis of
intraobserver variability in tumor grading, poorly European randomized clinical trials. J Clin Oncol
differentiated tumors have consistently found to be 29:3163–3172
associated with a worse prognosis in multivariate 2. Rutten HJ, den Dulk M, Lemmens VE et al (2008)
Controversies of total mesorectal excision for rectal
analysis. cancer in elderly patients. Lancet Oncol 9:494–501
3. Tepper JE, O’Connell M, Niedzwiecki D et al (2002)
Adjuvant therapy in rectal cancer: analysis of stage,
3.4 Pretreatment Laboratory sex, and local control – final report of intergroup 0114.
J Clin Oncol 20:1744–1750
Findings 4. van Gijn W, Marijnen CA, Nagtegaal ID et al (2011)
Preoperative radiotherapy combined with total
Carcinoembriogenic antigen (CEA) is the most mesorectal excision for resectable rectal cancer:
frequently used tumor marker in colorectal can- 12-year follow-up of the multicentre, randomised
controlled TME trial. Lancet Oncol 12:575–582
cer. An elevated CEA level at the time of presen- 5. Sebag-Montefiore D, Stephens RJ, Steele R et al
tation has an adverse impact on survival (2009) Preoperative radiotherapy versus selective
independent of tumor stage, and a reduction after postoperative chemoradiotherapy in patients with rec-
radical surgery has been associated with improved tal cancer (MRC CR07 and NCIC-CTG C016): a mul-
ticentre, randomised trial. Lancet 373:811–820
disease-free survival. Prechemoradiotherapy 6. Birgisson H, Pahlman L, Gunnarsson U et al (2005)
(CRT) CEA concentrations >5 ng/ml has been Adverse effects of preoperative radiation therapy for
associated with poor tumor response. Another rectal cancer: long-term follow-up of the Swedish rec-
study found post-CRT CEA concentrations tal cancer trial. J Clin Oncol 23:8697–8705
7. Valentini V, Beets-Tan R, Borras JM et al (2008)
<5 ng/ml to be associated with increased rates of Evidence and research in rectal cancer. Radiother
complete clinical and pathologic response. Little Oncol 87:449–474
is known about the relationship between the 8. Sauer R, Becker H, Hohenberger W et al (2004)
degree of CEA reduction from before to after Preoperative versus postoperative chemoradiotherapy
for rectal cancer. N Engl J Med 351:1731–1740
neoadjuvant CRT and prognosis. A recent retro- 9. MERCURY Study Group (2007) Extramural depth of
spective study from Korea suggests that in tumor invasion at thin-section MR in patients with
patients with elevated CEA levels pre-CRT rectal cancer: results of the MERCURY study.
(>6 ng/ml), a ³70% reduction in post-CRT levels Radiology 243:132–139
10. Kim CW, Yu CS, Yang SS et al (2011) Clinical
predicted for improved disease-free survival [10]. significance of Pre- to post-chemoradiotherapy s-CEA
The pretreatment serum hemoglobin level has reduction ratio in rectal cancer patients treated with
also been correlated to pathologic response to preoperative chemoradiotherapy and curative resec-
neoadjuvant CRT. According to one study, pre- tion. Ann Surg Oncol 18:3271–3277
11. Lee SD, Park JW, Park KS et al (2009) Influence of
treatment anemia (<9.0 g/dl of Hb) was associ- anemia on tumor response to preoperative chemora-
ated with poor tumor response to CRT, and diotherapy for locally advanced rectal cancer. Int J
correction of anemia for rectal cancer patients Colorectal Dis 24:1451–1458
with such low Hb levels was recommended [11].
What Are the Relevant Imaging
Factors to Optimize Treatment 4
Decisions?

Manish Chand and Gina Brown

Contents 4.1 Introduction


4.1 Introduction ................................................ 27
The optimal management of rectal cancer is
4.2 The Common Imaging Modalities ............ 28
influenced by several different factors relating to
4.2.1 Magnetic Resonance Imaging (MRI) .......... 28
4.2.2 Endoanal Ultrasound.................................... 28 the primary tumour. These may be specific char-
4.2.3 Computed Tomography (CT)....................... 29 acteristics of the tumour itself as well as pattern of
4.3 Local Factors Which Influence spread both locally and to distant sites. Identifying
Treatment Decisions................................... 29 these factors at an early stage is beneficial to long-
4.3.1 Circumferential Resection Margin (CRM) .. 29 term outcome and leads to a reduction in the risk
4.3.2 Height of Tumour......................................... 30 of local recurrence and distant spread. The aim of
4.3.3 Extramural Venous Invasion (EMVI) .......... 30
4.3.4 T-Staging ...................................................... 32 treatment must be to alleviate any symptoms, to
4.3.5 Nodal Staging .............................................. 36 deal with the immediate threat of cancer and to
4.4 Distant Factors ........................................... 36
reduce the risk of recurrence. This may involve
4.4.1 Hepatic Metastases ...................................... 36 surgery, radiotherapy and chemotherapy or a com-
4.4.2 Extra-hepatic Disease .................................. 37 bination of all three depending on the prognostic
4.5 Summary..................................................... 38 features of the tumour. Imaging provides an
important role in identifying factors which may
References ................................................................. 38
influence treatment decisions both at the time of
diagnosis and during treatment.
Traditionally, clinicians have relied on a com-
bination of digital rectal examination (DRE),
sigmoidoscopy or colonoscopy and tissue biop-
sies to confirm the diagnosis of rectal cancer.
However, these techniques provided minimal
information on tumour spread and characteris-
tics. Assessment of the surgical specimen would
M. Chand (*) be the basis for decisions on adjuvant therapy
Colorectal Research Fellow
depending on identification of specific prognostic
and Registrar in Colorectal Surgery,
Royal Marsden Hospital, Downs Road, Sutton features on histology. The introduction of increas-
e-mail: mans001@aol.com ingly accurate imaging modalities such as mag-
netic resonance imaging (MRI) and computerized
G. Brown
tomography (CT) have helped identify additional
Consultant Radiologist, Royal Marsden Hospital,
Downs Road, Sutton prognostic information about the tumour pre-
e-mail: gina.brown@rmh.nhs.uk operatively.

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 27


DOI 10.1007/978-3-642-25005-7_4, © Springer-Verlag Berlin Heidelberg 2012
28 M. Chand and G. Brown

Optimal treatment strategy must account


for the risk of local recurrence and distant metas-
tases. The decision to offer patients oncologi-
cal therapies such as chemo- and radiotherapy is
now strongly influenced by the results of early
imaging studies – “staging scans”. However, it
is not only important to be able to select patients
who are at a higher risk of poor outcomes for
neoadjuvant therapy but also not to offer
patients such treatment unnecessarily. Therefore,
the role of imaging in adding to management
decisions must be to avoid both under- and
overtreatment.
The current repertoire of imaging modalities
aim to provide detailed information on tumour
size, location and type; pattern and depth of inva-
sion; relationship to mesorectum; nodal involve-
ment and distant metastases; and, most recently,
angiotropism. Ultimately, this information allows
clinicians to risk stratify patients and optimize
treatment decisions.

4.2 The Common Imaging


Modalities Fig. 4.1 Axial image showing correct field alignment.
The red lines indicate correct alignment so to accurately
measure and stage the tumour
4.2.1 Magnetic Resonance Imaging
(MRI)
trast enhancement is considered to be far less
The most common MRI techniques for assessing reliable.
rectal cancer involve either an endorectal coil or Correct alignment of the axial images, perpen-
surface coil. The endorectal coil suffers some of dicular to the long axis of the rectum is essential
the drawbacks associated with EAUS, as they for accurate assessment of the tumour. Figure 4.1
must be positioned in the patient’s anorectum. shows the normal axis of the rectum. The rectum
This makes it difficult to assess stenosing tumours angles posteriorly and inferiorly rather than
and limits visualization of structures immediately straight down. Mal-alignment of the imaging
outside the rectum. The imaging field is limited field, for example, perpendicular to the vertical
to a small area, and although the images can be plane, will lead to inaccurate staging. The field
highly detailed, the external phased-array surface must be perpendicular to the rectal tube as seen in
coil has a greater advantage of providing compa- Fig. 4.1. Incorrect alignment of the imaging field
rable views of the rectal wall whilst being non- can have a major influence on pre-operative deci-
invasive and able to evaluate the remainder of the sion making.
pelvis. Regardless of which coil technique is
used, the protocol may be the same. There have
in the past been wide variations in sequences 4.2.2 Endoanal Ultrasound
used, but all the recently published data have
confirmed the improved accuracy achievable Endoanal ultrasound is useful in the local assess-
using high-resolution T2-weighted images. The ment of rectal cancer. A recent review comparing
use of T1-weighted scans with or without con- EAUS to MRI did not find one technique superior
4 What Are the Relevant Imaging Factors to Optimize Treatment Decisions? 29

over the other when assessing rectal cancer pre- Patients with synchronous metastatic disease now
operatively; rather, they are complimentary tech- receive systemic chemotherapy and may be con-
niques [1]. Local staging and the usefulness of sidered for metastasectomy with consequent
EAUS is discussed below. High spatial resolution of improvements in overall survival.
EAUS images is particularly suited to identification Limitations of CT include poor spatial resolu-
of the structure of the bowel wall. However, whilst tion which makes identification of the individual
MRIs drawbacks relate to tolerance and compatibil- layers of the bowel wall difficult. Tumour depth is
ity with the magnet, EAUS is not reliable for certain an important consideration in decision making as
types of tumour morphology. For example, the regards neoadjuvant therapy, and accurate local
endoanal technique produces inaccuracy with staging is a must. Recent studies have investigated
stenosing tumours, polypoidal tumours and high the use of multi-detector CT as an alternative to
rectal cancer. In addition, it does not demonstrate MRI for local staging of rectal cancer [3, 4].
fully the mesorectal fascia and thus the proximity of However, results of these studies have not favoured
tumour to the potential circumferential resection CT. Although for “higher” rectal tumours CT is
margin. Peritumoral inflammation, fibrosis and fae- accurate in identifying a potentially involved
cal material can give the appearance of artefact or CRM, it has poor accuracy for low rectal cancer.
overcall tumour. Accuracy may be further reduced
due to a combination of technical and user errors
including incorrect field alignment, improper bal- 4.3 Local Factors Which Influence
loon inflation, physical or refraction artefacts and if Treatment Decisions
the tumour is close to the anal verge [2].
Therefore, EUS is severely limited in support- The main risk factors for local recurrence in
ing preoperative treatment decisions on the fol- rectal cancer are histopathological involvement
lowing basis: of the circumferential resection margin (CRM)
1. Inability to identify patients at risk of local and height of tumour from anal verge. More
recurrence based on predicted CRM status recently, the presence of extramural venous inva-
2. Limited assessment of bulky and stricturing sion (EMVI) has been shown to be a prognostic
tumours and of identifying disease remote factor for poor outcome in terms of both local
from the lumen within mesorectum and distant failure [5]. In patients undergoing
3. Poor accuracy in reassessment of tumour stage TME surgery, nodal status no longer predicts
and safety of surgical margins following local failure but remains a risk factor for distant
chemoradiotherapy or radiotherapy metastases as well as increasing depth of tumour
spread. All these factors can be most accurately
assessed with MRI, although there may be some
4.2.3 Computed Tomography (CT) role for CT and endoanal ultrasound (EAUS).

The use of CT is mainly that of identifying the


possibility of extra-colonic tumour spread and 4.3.1 Circumferential Resection
metastatic deposits. Although it is not used pri- Margin (CRM)
marily for local assessment of the tumour, it does
provide information on the tumour relationship to One of the main advantages of MRI is in its abil-
adjacent organs such as the vagina or bladder and ity to accurately identify the mesorectal fascia
thus pelvic disease. The main advantage of CT is which forms the radial margins of excision and
in assessment of the common sites of metastases, thus the circumferential resection margin in total
i.e. lungs, liver and lymph nodes. It has high sen- mesorectal excision surgery. Local recurrence of
sitivity and specificity for identifying tumour rectal cancer following surgical resection has
deposits in the liver and lungs. In the past, involve- been dramatically reduced by the introduction of
ment of these sites has had a negative effect on total mesorectal excision (TME). Obtaining a
clinical outcome in terms of overall survival. clear circumferential resection margin following
30 M. Chand and G. Brown

TME is the “gold standard” as CRM involvement basis for neoadjuvant therapy for CRM positive
is a positive predictive factor for local recurrence. tumours reduced the need for CRT by 35% with-
Whereas histopathological analysis of the resec- out compromising outcome [11].
tion specimen provides confirmation of oncolog-
ically successful surgery, knowledge of the
relationship between the tumour and the mesorec- 4.3.2 Height of Tumour
tum is a mandatory in treatment planning. MRI
has been shown to accurately predict the rela- Rectal cancer is defined in terms of distance from
tionship between tumour and mesorectal fascia the anal verge. The rectum is measured as
with a high degree of concordance (92%) with 12–15 cm from the anal verge, and tumours found
histology [6]. within the distal 6 cm are classified as “low” rec-
Tumour spread which threatens the potential tal cancers. The distance from the anal verge or
CRM has been shown to be a predictive factor of height of the tumour is important due to its ana-
local recurrence [7]. It is now generally accepted tomical relationships as the rectum leaves the
that tumour spread within 1 mm of the potential peritoneal cavity. It is prognostically significant
CRM is a strong influence in local recurrence. for local recurrence [12, 13] and anastomotic leak
Recent work by Taylor et al. has shown that rates following surgery [14, 15].
of local recurrence decreased from 53% with Traditional assessment of the height of tumour
tumour less than 1 mm from the potential CRM was done using a combination of digital rectal
to less than 8% when the tumour distance from examination and rigid sigmoidoscopy. Both these
the mesorectal fascia was between 1 and 5 mm methods can be greatly inaccurate. The level of the
[8]. Knowledge of the proximity of the tumour to tumour has implications on surgical planning and
the mesorectal fascia at an early stage is impor- may be the difference between performing an
tant when planning treatment. anterior resection or abdominoperineal resection
Involvement of the potential circumferential or deciding to create a defunctioning stoma. In this
resection margin may be subdivided and appears regard, demonstrating the relationship between the
as distinct entities on imaging (Fig. 4.2). It is tumour and pelvic floor as well as the sphincter
therefore important to define the type of threat to complex is vital. In the era of laparoscopic surgery,
the potential CRM as not all subtypes may be many surgeons find laparoscopic rectal cancer
prognostic. For example, a local well-encapsu- resection for low tumours inappropriate, so accu-
lated lymph node lying close to the mesorectal rate objective identification of tumour height is
fascia is not associated with an increased risk in important, particularly the distance between the
local recurrence [9] (Table 4.1). distal edge of the tumour and the puborectal sling.
Being able to accurately identify tumour Axial images on MRI can clearly demonstrate
proximity to the mesorectal fascia within a mil- the level and longitudinal spread of the tumour
limetre on MRI has been a challenge. A mea- (Fig. 4.1). However, correct field alignment is
sured distance of 5 mm on MRI has been shown imperative and taking images through a plane
to strongly correlate with negative CRM on his- which is not exactly 90° to the long axis of the
tology, which led to patients being offered rectum and over- or under-call tumour height.
chemoradiotherapy when tumours are within
5 mm of the mesorectal fascia. However, this
results in substantial overtreatment of patients 4.3.3 Extramural Venous
with safe margins. More recently, the MERCURY Invasion (EMVI)
Study Group showed that margins could be
identified more accurately and reproducibly Invasion of extramural veins has been identified
using a 1-mm cut-off [10]. One recent study has as a poor prognostic factor for overall survival
shown that using MRI-based measurement as a and, more recently, local recurrence [1, 2]. This
4 What Are the Relevant Imaging Factors to Optimize Treatment Decisions? 31

a b

Fig. 4.2 (a) Grade 2 EMVI on MRI. (b) Grade 3 EMVI. (c) Grade 4 EMVI. The red arrows indicate the extent of
venous invasion

is independent of tumour stage. The definition be identified pre-operatively on MRI with great
of EMVI is the presence of malignant cells accuracy [3]. Knowledge of the appearances
within the endothelial-lined blood vessels and position of normal rectal vasculature is
beyond the muscularis propria. It can occur in important to be able to accurately identify
up to 50% of rectal cancer patients. EMVI can EMVI.
32 M. Chand and G. Brown

Table 4.1 Subtypes of histopathology CRM involvement of the UICC TNM classification. Depth of extra-
Type of spread Frequency (%) mural invasion is recognized as an independent
Direct tumour spread 18–29 prognostic factor [1, 2]. Within the broad T-stage
Discontinuous spread 14–67 classification, there are certain groups of patients
Lymph node metastases 12–14 who are considered high risk and thus may benefit
Venous invasion 14–57 from adjuvant therapy. This has led to a sub-
Lymphatic invasion 9 classification of T-stage. For example, T3 tumours
Perineural spread 7–14 can be further classified according to maximal
tumour invasion beyond the outer border of the
Table 4.2 MRI classification of EMVI muscularis propria – into the mesorectum. T4
MRI- tumours can be further classified depending on
EMVI the presence of invasion into adjacent organs with
score EMVI status Description or without perforation of visceral peritoneum [3].
0 Negative No vessels adjacent to areas A more detailed discussion of T sub-staging will
of tumour penetration be addressed in further chapters.
1 Negative Minimal stranding but well
Accurate assessment of this relies on the
away from vessels
2 Negative Stranding within vicinity of
identification of the layers of the bowel wall. A
vessel but no definite tumour meta-analysis of almost 5,000 patients compar-
signal ing accuracy of T-staging between MRI, CT and
3 Positive Intermediate signal within EAUS showed 84%, 73% and 87%, respectively
slightly expanded vessel [4]. Although this would suggest greater accu-
4 Positive Irregular contour of vessel by racy with EAUS, MRI techniques have improved
definite tumour signal
since, and it has the advantage in being able to
evaluate the peri-rectal structures and not be lim-
Using high spatial resolution MRI, veins are ited by access or user problems. EAUS may be
identified as serpiginous or tortuous linear struc- more useful for T1 and T2 tumours where accu-
tures on T2-weighted images. The depth of inva- rate identification of the mesorectal fascia has
sion of the tumour bears relevance as a tumour less importance, particularly if the lesion is ame-
limited to the muscularis propria will not be able to nable to local resection.
spread into extramural veins. Essentially, this An understanding of the MRI appearances of
means that EMVI positive tumours will be T-stage normal rectal anatomy is essential to appreciate
3 or 4. Assessment of EMVI using MRI must con- tumour depth and spread. MRI can readily iden-
sider the following components: pattern of tumour tify the layers of mucosa and muscle through dis-
margin which gives the appearance of nodularity, tinct signal characteristics. T2-weighted images
location of tumour to relevant vessels which makes are particularly useful for this. The mucosal layer
tumour invasion more likely, calibre of vessel as is seen as a very fine line of low signal intensity
tumour infiltration can cause an increase in lumi- overlying the much thicker and higher signal of
nal size, and vessel border if the tumour disrupts the submucosa (Fig. 4.3). Outside this, the mus-
the vessel itself [4]. Table 4.2 shows classification cularis propria can be seen as a dual layer repre-
of EMVI in rectal cancer. Extension of the primary senting the inner circular and the outer longitudinal
tumour into a vascular structure indicates EMVI. muscle layers. The latter has a typically irregular
appearance due to vessels traversing the rectal
wall. The peri-rectal fat is identified as a high sig-
4.3.4 T-Staging nal with signal void areas surrounding the rela-
tively low signal intensity of the muscularis. This
Staging of rectal cancer involves determining the is all enveloped by the fine layer of low signal
depth of invasion of the tumour in relation to the intensity representing the mesorectal fascia – the
composition of the bowel wall and makes up part defining plane of the TME.
4 What Are the Relevant Imaging Factors to Optimize Treatment Decisions? 33

indicate T3 spread, as this more commonly repre-


sents small vessels penetrating this layer
(Fig. 4.4).
More recently, importance has been given to the
extent of tumour growth beyond the muscularis into
the mesorectal fat. Patients who have tumours

Fig. 4.3 MRI axial image showing tumour relationship to


the mesorectal fascia

Table 4.3 MRI T-staging of rectal cancer


T-stage Description
1 Low signal in submucosal layer but no
signal abnormality in the circular muscle
layer
2 Intermediate signal intensity within muscle.
No extension of signal abnormality into the
peri-rectal fat b
3 Broad based bulge or nodular projection of
intermediate signal intensity projecting
beyond outer muscle coat
4 Extension of abnormal signal into adjacent
organ or peritoneal reflection

The MRI T-staging of rectal cancer is shown


in Table 4.3. Determining the T-stage of the
tumour depends on careful assessment of the
images with respect to the signal characteristics
and correct field alignment. Abnormal or unex-
pected signal intensity in the bowel wall layers
defines the T-stage. Of particular importance are
the characteristics of T3 tumours. A T3 tumour
must demonstrate invasion beyond the muscu-
laris propria into the peri-rectal fat. This is seen
as a bulging projection into the high signal layer
of the peri-rectal fat. Small irregularities along Fig. 4.4 (a) T1 lesion on MRI. (b) T2 lesion on MRI. (c)
the border of the muscularis does not necessarily Subclassification of T3 lesions. (d) T4 tumour
34 M. Chand and G. Brown

70.5

Fig. 4.4 (continued)


4 What Are the Relevant Imaging Factors to Optimize Treatment Decisions? 35

Table 4.4 Bowel wall layers in EAUS


d
Layer of Signal
bowel wall Description characteristic
Superficial The layer between the Hyperechoic
mucosa probe and the superficial
mucosa
Mucosa/ Second layer of bowel Hypoechoic
muscularis wall
mucosae
Submucosa Interface between Hyperechoic
mucosa and submucosa
Muscularis Fourth layer outside Hypoechoic
propria submucosa
Serosa The layer between the Hyperechoic
serosa and peri-rectal fat

Fig. 4.4 (continued)

which show greater invasion into the mesorectum


(>5 mm) have been shown to have lower 5-year sur-
vival and is independent of lymph node involve-
ment [2]. Those patients whose tumours demonstrate
less than 1 mm invasion into the mesorectum have
a favourable prognosis [1]. This has led to a sub-
classification of T3 tumours. Local recurrence rates
are similar in patients staged T2 and T3a, whereas Fig. 4.5 Endoanal ultrasound demonstrating layers of
rates significantly differ between T3a and T3b – bowel wall
this is independent of lymph node status [5]. MRI is
not particularly accurate in separating T1 and T2
tumours and in distinguishing sessile or polypoid larly useful when planning mucosal resection or
adenomas from T1 lesions. EAUS in comparison is transanal excision. Sensitivity and specificity for
highly accurate in assessment of these tumours [6]. T1 cancers is 87.8% and 98.3%, respectively [7].
Similar to the images produced by MRI, As transanal endoscopic microsurgery (TEMS)
EAUS identifies different signal characteristics to and endoscopic submucosal resections become
correspond to the layers of the bowel wall. Five more popular, greater detail of the bowel wall is
alternate layers of hyper- and hypoechoic rings essential to select appropriate patients. Assessing
are produced from the differences in acoustic whether a tumour has breached the submucosa
impedance. A carcinoma is seen as an irregular becomes an important decision, and other imaging
hypoechoic mass which disrupts the normal ana- modalities may not be as accurate as EAUS. EAUS
tomical layers. The extent of this disruption may be particularly useful in distinguishing T2
determines the depth of invasion and this T-stage. from T3a tumours, more so than MRI. However,
The normal anatomy is shown in Table 4.4. whether this is relevant in terms of oncological
EAUS has increased accuracy for defining the treatment is debatable. This is discussed in more
detail of the bowel wall structure which is particu- detail in future chapters (Fig. 4.5).
36 M. Chand and G. Brown

4.3.5 Nodal Staging Location of lymph nodes may be an important


consideration. Lymph nodes distal to the tumour
Identifying nodal involvement has traditionally are rarely involved. Similarly, the sparing of
been a difficult issue. Suspicious appearance of extra-mesorectal lymph nodes is helpful in under-
local nodes can be due to size or differences in standing the signal characteristics of malignant
signal characteristics. The problem with using nodes. A contentious issue has been the relevance
size is defining a cut-off number. A more sophis- of lymph nodes to the mesorectal fascia. Shihab
ticated method may be identifying particular sig- et al. showed that a solitary lymph node in close
nal characteristics. MRI criteria involve proximity to the mesorectal fascia and therefore
identification of spiculated or indistinct lymph involvement of the CRM is uncommon [13]
node borders as well as mottled heterogeneous (Fig. 4.6).
signal intensity which may predict nodal involve-
ment [8]. More recently, the use of specific par-
ticles unique to normal lymph nodes as compared 4.4 Distant Factors
with cancerous nodes which are identifiable on
MRI has been used, although further study is The presence of distant disease not only has an
required – Ultra-small Superparamagnetic implication on oncological therapy but also tim-
Particles of Iron Oxide (USPIO) [9]. As a whole, ing of treatment. Knowledge of hepatic metasta-
MRI, EAUS and CT show sensitivity and ses may lead to deferral of rectal surgery, for
specificity for detection of malignant lymph instance. Common sites of metastases include
nodes to be between 55% and 78%, respectively liver, lung, bones and adrenal glands. CT exami-
[10, 11]. nation of the chest, abdomen and pelvis is man-
EAUS does not predict lymph node involve- datory. For most patients, CT will be sufficient to
ment any better. Sensitivity and specificity for examine the extent of extra-abdominal disease.
detection of cancerous lymph nodes in rectal However, indeterminate lesions of the liver or
cancer is 73.2% and 75.8%, respectively [12], lungs may require furthermore detailed imaging
although more likely to be accurate in the more to exclude metastases. Additional anatomical
proximal parts of the rectum. Swollen reactive information may also be needed for surgical plan-
nodes, small blood vessels and even local struc- ning of metastasectomy.
ture such as the seminal vesicles may mimic
malignant nodes.
4.4.1 Hepatic Metastases

Accurate imaging of the liver is of vital impor-


tance in the optimal management for rectal can-
cer. Metastatic involvement of the liver can have
a profound effect on treatment decisions both in
terms of oncological therapy and surgery. Ideally,
liver imaging should provide the following:
• Accurate delineation of the anatomical distri-
bution of metastases to facilitate planning of
potential surgical resection
• Presence or absence of widespread micro-
metastatic disease within the liver
• Presence or absence of extra-hepatic disease
• Discrimination between benign and malignant
liver lesions
• A method of road mapping lesions to ensure a
Fig. 4.6 Nodal assessment on MRI margin of greater than 10 mm
4 What Are the Relevant Imaging Factors to Optimize Treatment Decisions? 37

Fig. 4.8 MR image showing same liver metastases

margin definition and rim enhancement apply [15].


Haemangiomas enhance peripherally in a nodular
fashion and persistence of enhancement [16].
Artefact can produce occasional wedge-shaped
defects; however, assessment of these areas on the
pre-contrast images can usually avoid confusion.

4.4.1.2 MRI
Fig. 4.7 CT and PET-CT of liver metastases
MRI assessment includes T1- and T2-weighted
images with a contrast medium such as gadolin-
It is important to appreciate that it is unlikely ium providing sufficient anatomical detail, delin-
that a single imaging modality will be able to eating perfusion defects seen on CT images from
satisfy all the above questions; however, it is impor- fatty infiltration. Differentiating benign liver
tant to select an appropriate combination of sequen- lesions from malignant ones is an important abil-
tial imaging techniques to optimize outcome. ity of MRI. Specific contrast agents are taken up
by functioning Kupffer cells and are seen as dark-
4.4.1.1 CT ened areas on MR imaging. Mangafodipir triso-
CT exploits the relative hypovascularity of dium (Mn-DPDP) is taken up by functioning
hepatic metastases compared with normal liver hepatocytes and leads to an elevation in signal
parenchyma and has accuracy rates of up to 85% intensity of T1-weighted images of normal liver
[14]. The use of multi-detector CT can be highly parenchyma within 10 min of injection. There is
accurate in demonstrating liver lesions (Fig. 4.7). a 400% increase in conspicuity between the
The entire liver can be imaged within 10 s during hypointense hepatic metastases and the surround-
several phases of hepatic enhancement. Liver ing normal liver tissue [17]. It is important to be
metastases are seen as hypodense lesions with able to distinguish metastases from haeman-
complimentary rim enhancement. Larger lesions giomas, fatty infiltration and cysts (Fig. 4.8).
may exhibit signs of central necrosis such as a
central low density ‘cystic nidus’.
Differentiating metastases from benign lesions 4.4.2 Extra-hepatic Disease
is more straightforward for larger lesions as they
are well defined and lack rim enhancement. Smaller Patients who demonstrate single metastases in
lesions, particularly less than 10 mm, are more one lobe only are much less likely to have
difficult to characterize, but the same principles of unrecognized irresectable disease when
38 M. Chand and G. Brown

compared with those patients with multi-lobar tumour features being identified, thus giving cli-
metastases [18]. Although laparoscopy may be nicians more information to base treatment deci-
a useful tool in assessing the presence of extra- sions. This also highlights the advantages of a
hepatic disease, a combination of CT and PET multi-disciplinary approach to cancer.
scanning are more common.
PET imaging with 18-fluoro-2-deoxyglucose
(FDG) has been shown to have the greatest poten-
tial in detecting extra-hepatic disease not found
References
on conventional imaging [19]. Tumour tissue can 1. Willett CG et al (1999) Prognostic factors in stage
be detected on the basis of its increased glucose T3N0 rectal cancer: do all patients require postopera-
utilization which is characteristic of malignant tive pelvic irradiation and chemotherapy? Dis Colon
cells. Sensitivity to tissue glucose metabolism Rectum 42(2):167–173
2. Cawthorn SJ et al (1990) Extent of mesorectal spread
allows differentiation from equivocal lesions. and involvement of lateral resection margin as prog-
Upstaging of disease resulting in a change in sur- nostic factors after surgery for rectal cancer. Lancet
gical management can be seen in up to 40%, and 335(8697):1055–1059
this is more sensitive than contrast-enhanced CT 3. Wittekind C, International Union against Cancer
(2003) TNM supplement: a commentary on uniform
[20]. Its role in the evaluation of liver disease is use, 3rd edn. Wiley-Liss, Hoboken, p xix, 168 p
well defined, but it remains inferior to MRI, 4. Kwok H, Bissett IP, Hill GL (2000) Preoperative stag-
which is the most effective imaging modality for ing of rectal cancer. Int J Colorectal Dis 15(1):9–20
liver anatomy [21]. One of the drawbacks of PET 5. Merkel S et al (2001) The prognostic inhomogeneity
in pT3 rectal carcinomas. Int J Colorectal Dis
imaging has been its lack of spatial resolution 16(5):298–304
and anatomical detail. The solution to this has 6. Akasu T et al (2000) Endorectal ultrasonography and
been a single examination combining PET and treatment of early stage rectal cancer. World J Surg
CT. PETCT is most useful in detecting small 24(9):1061–1068
7. Puli SR et al (2009) How good is endoscopic ultra-
mesenteric or peritoneal disease not apparent on sound in differentiating various T stages of rectal can-
CT. It has been shown to reduce the rate of recur- cer? Meta-analysis and systematic review. Ann Surg
rence in patients undergoing hepatic resection for Oncol 16(2):254–265
colorectal metastases [12]. 8. Brown G et al (2003) Morphologic predictors of
lymph node status in rectal cancer with use of high-
One of the limitations of PET is the false-pos- spatial-resolution MR imaging with histopathologic
itive rate. This is particularly the case with granu- comparison. Radiology 227(2):371–377
lomatous disease and inflammatory processes of 9. Will O et al (2006) Diagnostic precision of nanoparti-
the lung. False-negatives can be diagnosed in cle-enhanced MRI for lymph-node metastases: a
meta-analysis. Lancet Oncol 7(1):52–60
lesions less than 1 cm in size, as well as micro- 10. Lahaye MJ et al (2005) Imaging for predicting the risk
metastatic disease in lymph nodes. factors – the circumferential resection margin and nodal
disease – of local recurrence in rectal cancer: a meta-
analysis. Semin Ultrasound CT MR 26(4):259–268
11. Bipat S et al (2004) Rectal cancer: local staging and
4.5 Summary assessment of lymph node involvement with endolu-
minal US, CT, and MR imaging – a meta-analysis.
A shift of strategy has become apparent in recent Radiology 232(3):773–783
years with more efforts being concentrated on 12. Puli SR et al (2009) Accuracy of endoscopic ultra-
sound to diagnose nodal invasion by rectal cancers: a
pre-operative rather post-operative oncological meta-analysis and systematic review. Ann Surg Oncol
therapy. Implementing successful neo-adjuvant 16(5):1255–1265
treatment relies on prognostic and predictive 13. Shihab OC et al (2010) Magnetic resonance imaging-
factors of outcome being known early in diagno- detected lymph nodes close to the mesorectal fascia
are rarely a cause of margin involvement after total
sis. This has led to increased importance in mesorectal excision. Br J Surg 97(9):1431–1436
accurately identifying these factors at an early 14. Valls C et al (2001) Hepatic metastases from colorectal
stage. An improvement in the quality of imaging cancer: preoperative detection and assessment of resec-
modalities has led to increasingly more subtle tability with helical CT. Radiology 218(1):55–60
4 What Are the Relevant Imaging Factors to Optimize Treatment Decisions? 39

15. Jang HJ et al (2002) Small hypoattenuating lesions in 19. Strasberg SM et al (2001) Survival of patients evalu-
the liver on single-phase helical CT in preoperative ated by FDG-PET before hepatic resection for meta-
patients with gastric and colorectal cancer: preva- static colorectal carcinoma: a prospective database
lence, significance, and differentiating features. study. Ann Surg 233(3):293–299
J Comput Assist Tomogr 26(5):718–724 20. Kong G et al (2008) The use of (18)F-FDG PET/CT
16. Nino-Murcia M et al (2000) Focal liver lesions: pat- in colorectal liver metastases-comparison with CT
tern-based classification scheme for enhancement at and liver MRI. Eur J Nucl Med Mol Imaging 35:
arterial phase CT. Radiology 215(3):746–751 1323–1329
17. Young SW et al (1990) Detection of hepatic malig- 21. Ong KO, Leen E (2007) Radiological staging of col-
nancies using Mn-DPDP (manganese dipyridoxal orectal liver metastases. Surg Oncol 16(1):7–14
diphosphate) hepatobiliary MRI contrast agent. Magn
Reson Imaging 8(3):267–276
18. Jarnagin WR et al (1999) Liver resection for meta-
static colorectal cancer: assessing the risk of occult
irresectable disease. J Am Coll Surg 188(1):33–42
What Biochemical and Molecular
Biological Factors Have Greater 5
Relevance to Treatment Decisions?

Guido Lammering and Jeroen Buijsen

Contents 5.1 Introduction


5.1 Introduction ................................................... 41
The current standard therapy for rectal cancer in
5.2 Biochemical and Molecular Biological
Factors for Radiochemotherapy .................. 42
most countries around the world consists of a pre-
operative treatment either with long-term radio-
5.3 Biochemical and Molecular Biological
therapy (RT) and a 5-FU-based chemotherapy
Factors for Surgical Decisions...................... 44
(CT) or a short course RT followed by
5.4 Biochemical and Molecular surgery or surgery alone, depending on the pre-
Biological Factors for Adjuvant
and Systemic Therapy .................................. 45 therapeutic staging resulting in a 5-year cumula-
tive rate of local relapse of less than 10% and an
References ................................................................. 47
incidence of distant metastases of about 35%
[2, 27]. The preoperative treatment has a major
impact on local control, however, with only mini-
mal impact on survival and disease-free survival.
So far, no clear evidence has been produced for
the benefit of adjuvant chemotherapy, leading to
different protocols in different countries and
regions around the world. Generally, all parts of
the current standard treatment of rectal cancer
have its toxicity profiles. Preoperative chemora-
diotherapy (CRT) can lead to diarrhoea, radiation
dermatitis and increased risk for anastomotic
leakage and perineal wound complications [29],
while short course radiotherapy generally
increases the risk for faecal incontinence, anal
blood loss, mucus loss and erectile dysfunction
[18, 28]. The surgical treatment itself is generally
accompanied with an increased risk for urinary,
bowel and erectile dysfunction, anastomic leak-
age, perineal wound complications and a mortality
G. Lammering (*) • J. Buijsen rate of 4–11%, depending on age and co-morbid-
Maastro Clinic, 1345, 6201 BH,
ity [12, 21]. Thus, all treatments come with a
Maastricht, The Netherlands
e-mail: guido.lammering@maastro.nl; price to pay, namely, the risk for over- or under-
jeroen.buijsen@maastro.nl treatment based on the current imaging-related

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 41


DOI 10.1007/978-3-642-25005-7_5, © Springer-Verlag Berlin Heidelberg 2012
42 G. Lammering and J. Buijsen

treatment decisions leading to probably unneces- The percentage of patients developing a patho-
sary toxicities and subsequently impact on quality logical complete response (pCR) varies, but lies
of life [8, 26]. However, rectal cancer is a biologi- typically around 20%. In treatment decision mak-
cal tumour, characterized by several biological ing, it would be very helpful to identify good and
features, which are nowadays better and better poor responders in order to give a tailored treat-
understood. Dramatic technical improvements in ment. Until now, the predictive value of several
detection assays have made it possible to charac- biomarkers has been studied. In these studies,
terize more and more biochemical and biological various endpoints have been used, but the most
features and phenotypes related to individual rec- common endpoints are overall survival, tumour
tal cancer patients. Thus, biological markers regression grade (TRG), pCR and downstaging.
might be able to better tailor the treatment of rec- Kuremsky et al. published a review evaluating
tal cancer, thereby minimizing the risk for over- the potential of genetic biomarkers in predicting
and undertreatment with also an improvement in the outcome of locally advanced rectal cancer
outcome and quality of life. However, this needs patients treated with chemoradiation [9]. In their
prospective evaluation and stringent validation. It review, they focused on gene products with more
is clear that there is a biological basis for the dif- than five studies in the literature. The six bio-
ferent responses of individual tumours to certain markers that met this criterion were p53, epider-
treatments, like CRT, RT and CT. The tumour mal growth factor receptor (EGFR), thymidylate
microenvironment, e.g. angiogenesis and hypo- synthase (TYMS), Ki-67, p21 and bax/bcl-2.
xia, repopulation and the intrinsic radiosensitivity The most studied marker is p53, a gene that
are currently the best known biological tumour plays a key role in apoptosis, tumorigenesis and
phenotypes, which undoubtedly have an impor- sensitivity to chemotherapeutic agents. Twenty-
tant impact on the degree of response to anti- one studies studying the predictive value of p53
tumour treatments [11]. status were identified. Seventeen of these stud-
This chapter will focus on the question where ies could not identify a correlation between p53
we are now in the detection and analysis of bio- expression and tumour response. Of the four
chemical and biological factors influencing treat- studies that did show a relationship between
ment decisions and how far we have already p53 and outcome, three identified a p53 muta-
come in our attempts to better individualize treat- tion as a predictor of poor response, whereas the
ment based on biomarkers. other study found a positive predictive value for
Section 5.2 will give an update on the current mutated p53. Therefore, p53 does not seem to
knowledge on biomarkers in treatment deci- be a good predictor for tumour response to
sions for neoadjuvant RT and CRT. Section 5.3 chemoradiation.
will focus on biomarkers for surgical treatment EGFR is important in the regulation of various
decisions, while Sect. 5.4 will discuss the bio- cellular responses, like proliferation, apoptosis
markers known to influence decisions for the and differentiation. Overexpression of EGFR has
adjuvant and systemic treatment of rectal can- been shown in 60–80% of all colorectal cancers
cer. Finally, Sect. 5.5 will provide a conclusion and has been associated with a poor prognosis
and a perspective. independent of lymph node status. Five studies
have been published studying the value of EGFR
expression as response predictor. These were
5.2 Biochemical and Molecular studies looking at chemoradiation only, without
Biological Factors for addition of EGFR inhibitors. Although these
Radiochemotherapy studies looked at different endpoints, the studies
that found a correlation all point towards a better
The combination of chemotherapy and radiother- response to chemoradiation in tumours with low
apy leads to some degree of pathological down- EGFR expression. It seems to be more important
staging in approximately 40–60% of patients. to quantify the level of EGFR expression rather
5 What Biochemical and Molecular Biological Factors Have Greater Relevance to Treatment Decisions? 43

Good-risk group
TYMS

Surgical Resection
*2/*2,*2/*3, RT 45 Gy
*2/*4 FU 225 mg/m2/day
T3/T4
Rectal TYMS genotyping
Cancer RT 45 Gy
FU 225 mg/m2/day
TYMS Cpt-11 50 mg/m2
*3/*3,*3/*4 q wk × 5
Staging
Poor-risk group
(TRUS, CT, MRI) Staging
Biopsy (TRUS, CT, MRI)

Fig. 5.1 Design of a phase II trial stratifying rectal cancer patients in good- and poor-risks according to TYMS status

than divide tumours into positive or negative. One group (TYMS overexpression) (Fig. 5.1) [31].
study identified a single nucleotide polymorphism The good-risk group was treated with standard
that may be useful as a biomarker for response. chemoradiation consisting of radiotherapy and
Thymidylate synthase is an important enzyme continuous 5-FU infusion, while the poor-risk
in DNA synthesis and is the main target of patients received weekly irinotecan in addition to
5-fluorouracil (5-FU). Overexpression of this standard chemoradiation. A pCR and tumour
enzyme leads to 5-FU resistance. Nine studies downstaging was found in 18.9% and 64.4% of
were identified in the Kuremsky review analysing good-risk patients, respectively, and in 35.5%
the influence of TYMS expression and SNPs in and 64.5% of poor-risk patients. However, the
the TYMS enhancer region on tumour response, combination with irinotecan turned out to cause
and very recently, two additional studies have more toxicity. RFS and OS were comparable
been published. Four studies used immunohis- between both risk groups. This study is the first
tochemical staining, and five studies used PCR attempt to stratify the treatment of rectal cancer
for genotyping. Three studies did not show a cor- patients according to TYMS status. Although
relation between TYMS and tumour response. further phase III trials are needed, it was remark-
The remaining six studies found in general a bet- able that the proportion of patients showing
ter response to chemoradiation in tumours with downstaging was comparable between the good-
low TYMS expression. Two studies also included and poor-risk group and the percentage that
a group of patients treated with radiotherapy only, developed a pCR was higher in the poor-risk
and as could be expected in this group, no corre- group.
lation between TYMS expression and response Ki-67 is a proliferation marker which has not
was observed. Only one study found an inverse shown a clear relationship with chemo- or radio-
correlation; however, this was a small study with sensitivity. Thirteen articles have been published
only 19 patients, who received both 5-FU and about the predictive value of Ki-67, of which
oxaliplatin, which could have confounded the only 2 showed a correlation. One study found a
results. Recently, however, two studies found a better response in tumours with high Ki-67 stain-
correlation between high expression of TYMS ing; the other showed an association between low
and better response after CRT for rectal cancer Ki-67 expression and response. Therefore, Ki-67
[10, 17]. Although there is not enough evidence is not a good biological marker in rectal cancer.
to use TYMS in daily practice, incorporation of The tumour suppression gene p21 is activated
TYMS expression measurements in chemoradio- by DNA damage and causes cell cycle arrest. Four
therapy trials would be worthwhile. Recently, the of eight studies found a correlation between p21
results of a phase II study have been published in expression and response to chemoradiation.
which patients were stratified in a good-risk However, the results of these four studies are
group (no TYMS overexpression) and a poor-risk conflicting. Two studies found better responses or
44 G. Lammering and J. Buijsen

survival in patients with low or negative p21 shown to have a predictive role in radiosensitivity
expression, while the other two studies found the of malignant tumours. The first study included 81
opposite. Based on in vitro studies, one would patients with locally advanced rectal cancer
expect better results in tumours with low p21 treated with chemoradiation. Of the patients with
expression because p21 suppresses apoptosis in an AG phenotype, 47% showed a major response,
case of DNA damage. The two studies which as compared to 22% in the AA and GG pheno-
showed better results in positive p21 tumours used type group. The second study analysed 93 patients
other treatments in addition to radiotherapy and treated with chemoradiation for rectal cancer.
5-FU which may have confounded the results. Genotyping was done on peripheral blood mono-
The bax and Bcl-2 proteins are involved in cytes. They found a better response in G/G carri-
apoptosis. Loss of bax function is correlated with ers as compared with G/A carriers (OR 4.180;
chemoresistance in colorectal cancer, and Bcl-2 p = 0.003). In the third study, DNA from mono-
overexpression has been linked with resistance to cytes of 128 rectal cancer patients was analysed.
different chemotherapeutic agents and inhibition In this study, no statistical significant association
of radiation-induced apoptosis. Three studies with tumour response was found. In conclusion,
evaluated the role of bax expression in response results of XRCC1 as a predictor of response in
to chemoradiation. Only one study found a rectal cancer are conflicting.
significant correlation. In that study, the percent- Another protein that attracted interest in the
age of bax-positive tumours was significantly past years is survivin, an inhibitor of apoptosis
higher in the complete-response group than in the [7, 16, 25]. Again, results are conflicting. Two
partial-response group (54% vs. 29%). Bcl-2 was recent studies reported a correlation between sur-
analysed in 12 studies, but a correlation with vivin expression and tumour response. Higher
response was found in only one study including survivin expression correlated with worse
only 17 patients. response to chemoradiation. One of these studies
Two rather small studies have tested the value compared survivin expression in pretreatment
of microarray in prediction of response. The first biopsies and surgical resection specimens and
study tested 54 genes in 23 patients. A different found a worse survival in patients with tumours
expression pattern was found between respond- expressing high levels of survivin after chemora-
ers and non-responders based on downstaging, diation. However, other studies could not find a
but this difference was no longer significant if correlation between survivin expression and clin-
response evaluation was based on TRG. The neg- ical outcome, which underlines the need for fur-
ative predictive value (NPV) was 86%, and the ther studies.
positive predictive value (PPV), 78%. The second
study included 43 patients and found 42 genes that
were differentially expressed between respond- 5.3 Biochemical and Molecular
ers and non-responders based on TRG. The PPV Biological Factors
using this gene set was 71% and the NPV 86%. for Surgical Decisions
Although these studies show for the first time
that gene expression profiles may be helpful in The surgical treatment strategy for rectal cancer
response prediction, they certainly need further has dramatically changed over the last years.
validation with larger patient groups. Coming from a standard radical surgical proce-
Quite recently, X-ray repair cross-comple- dure performed in strictly defined TME protocols
menting 1 (XRCC1) has been identified in three with only radicality being the primary goal, it
studies as a potential useful marker for response nowadays changes to a more tailored approach
prediction in rectal cancer [4, 10, 17]. XRCC1 with also the aim to reduce toxicity with better
plays an important role in DNA repair as it is quality of life. The surgical treatment usually
involved in the base excision repair (BER) path- takes place initially after diagnosis in early stage
way. One polymorphism, A399G, has been cancer or after a neoadjuvant treatment consisting
5 What Biochemical and Molecular Biological Factors Have Greater Relevance to Treatment Decisions? 45

of either radiotherapy alone (5 × 5 Gy) or RCT. In higher in earlier stages [23]. This however has
case of a neoadjuvant treatment, a short-course not been validated yet and warrants further evalu-
radiotherapy will usually be followed within ation. Generally, the only molecular markers
1–3 days by an immediate surgical resection of being currently prognostically relevant in rectal
the TME, as has been published by the DUTCH cancer are the deficient mismatch repair and pos-
TME trial [6]. Thus, the short-course radiother- sibly the KRAS mutation and the BRAF muta-
apy does not induce any downsizing or down- tions. All else, like expression assays, copy
staging, since the timing of the followed surgical number variation tests and even proteomics is
replacement of the rectum tumour does not allow experimental and have not shown any validated
any tumour shrinkage [15]. However, newer trials correlation with prognosis yet. The recently
have suggested that the short-course preoperative published analysis of the QUASAR study, which
radiotherapy indeed could be used to also down- mainly analysed stage II cases, showed MMR,
size and downstage the tumour, if the surgical KRAS or BRAF abnormalities in rectal cancer in
treatment is postponed by at least 6–8 weeks after 1–4%, 30–35% and 2–3%, respectively [5]. All
radiotherapy [19, 22]. This strategy might three abnormalities had prognostic impact at dif-
increase the potential for better sphincter- and ferent levels, with MMR being the strongest
even organ-sparing surgical treatment techniques, marker for prognostic impact. The defective
etc., as has been already proposed after neoadju- MMR genes were hMLH-1, hMSH-2 PMS-2 and
vant chemoradiotherapy in selected patients [14]. hMSH-6, detected by either immunohistochem-
However, accurate tumour response assessment istry or microsatellite testing. However, up until
is crucial in these modern adaptive surgical strat- now, no biomarkers have reached the level of
egies after neoadjuvant radio- or radiochemo- clinical relevance which would allow inclusion in
therapy. Biomarkers could help in the treatment surgical decision making. Recently, the first
decisions for less aggressive conservative surgi- cohort analysis of 20 patients selected for a non-
cal treatments, beside modern imaging and clini- surgical wait-and-see strategy after RCT has been
cal examinations. This applies not only for published with very promising local and distant
preoperatively pretreated rectal cancer patients controls [14]. The decision making for such a
but also for initially diagnosed patients, in which non-surgical approach in patients with good to
early stage disease is suggested. These patients complete clinical response after RCT, which is
could benefit from conservative surgery (e.g. currently only based on clinical and imaging
TEM) instead of radical surgery, if accurate bio- parameters due to the lack of sufficient biomark-
markers associated with disease progression, par- ers, should be stepwise improved with also bio-
ticularly mesorectal nodal metastasis, would chemical and molecular markers, as soon as they
become available. have been approved and validated.
One recently published study compared pat-
terns of gene-specific hypermethylations in radi-
cally excised rectal cancers with histopathological 5.4 Biochemical and Molecular
stage and came to the conclusion that locus- Biological Factors for Adjuvant
specific hypermethylation was more prevalent in and Systemic Therapy
early- than late-stage disease and that the hyper-
methylation of two or more of a panel of five The value of adjuvant chemotherapy in node-pos-
tumour suppressor genes was associated with itive colon cancer is clear. Since rectal tumours
localized disease [13]. Another study by Rasheed originate in an organ that is in anatomic continuity
et al. assessed the microvessel density (MVD) with the colon and tumours show a similar histol-
and the CA9 expression in more than 100 rectal ogy, it is often argued that results of adjuvant trials
cancer specimen and came to the conclusion that in colon cancer can be translated to rectal can-
the MVD was higher in more advanced T and N cers. However, until now, randomized trials in
stages, whereas the CA9 expression was generally rectal cancer have failed to show a clear benefit of
46 G. Lammering and J. Buijsen

adjuvant chemotherapy [3]. Especially in patients confirmed for metastatic colorectal cancer in a
who have a good response after preoperative recent meta-analysis [1].
chemoradiation, the expected gain of adjuvant
treatment is small. Recently, data of five large rec- Conclusion and Perspective of Biomarkers for
tal cancer trials including chemoradiation have Treatment Decisions
been pooled to build nomograms for the prediction Biochemical and molecular biological factors
of local and distal recurrence as well as survival. could help in treatment decision making along
This analysis confirms that in patients showing a the different treatment steps in the treatment of
good response after chemoradiation, the added rectal cancer. This would allow more tailored
benefit of adjuvant chemotherapy is small [32]. treatment approaches, thereby improving qual-
Biomarkers could be helpful to identify ity of life with even the benefit of more effec-
patients who are at higher risk for recurrent dis- tive treatments. When analysing the current
ease. When looking at the value of biomarkers, it value of these biomarkers in treatment decision
is important to distinguish prognostic from pre- making, it is important to identify markers as
dictive factors. A prognostic factor gives infor- being prognostic or predictive or even both. In
mation about the risk of recurrence of disease rectal cancer, the only markers reaching clini-
irrespective of a certain treatment, while a predic- cal relevance in prognosis so far are deficient
tive factor predicts the chance that a patient will MMR, KRAS and BRAF; however, its fre-
benefit from a treatment. Of course, a marker can quency in rectal cancer is rather low with only
be both prognostic and predictive. 1–4% deficient MMR, 30–35% KRAS muta-
In colorectal cancer, most evidence is available tion and 2–3% BRAF mutation (QUASAR).
for the prognostic value of mismatch repair (MMR) The defective MMR genes hMLH-1, hMSH-2,
gene status. Tumours that are MMR deficient have PMS-2 and hMSH-6, detected by immunohis-
a better prognosis. There is some debate whether tochemistry or by microsatellite testing,
MMR status also has a predictive value. However, showed a prognostic value in a meta-analysis
the proportion of rectal tumours that is MMR [20], in the PETACC trial [24], the QUASAR
deficient is small (around 1%). Therefore, it is not and numerous single-centre studies, which
a useful marker for rectal cancer. might lead to MMR-related treatment deci-
The role of BRAF status seems to have a pre- sions in the near future. KRAS status has also
dictive value, but the debate is going on about a been shown to be of prognostic value. KRAS
possible predictive role. Because the proportion of mutation tumour has a worse prognosis and a
rectal tumours carrying a BRAF mutation is very higher chance of recurrence. For EGFR-
low, BRAF does not seem to be a very important inhibitor therapy, the KRAS status even reaches
biomarker in rectal cancer. a predictive value.
KRAS status also has been shown to be of prog- With regard to prediction, six genetic bio-
nostic value. In about 40% of all rectal tumours, markers were identified with the potential of
KRAS is mutated. KRAS mutant tumours have a being predictive in the outcome of locally
poor prognosis and a higher chance of recurrence. advanced rectal cancer after CRT. These were
In the QUASAR trial, this difference was even p53, EGFR, TYMS, Ki-67, p21 and bax-bcl-2.
more pronounced for tumours located in the rec- While EGFR expression levels seem to pro-
tum as compared to tumours in other parts of the vide some prediction to CRT response, p53
colon. In this trial, the reduced risk of recurrence does not seem to serve as a good predictor, nei-
with chemotherapy was comparable between ther does Ki-67 or p21 nor bax and Bcl-2.
KRAS wild-type and KRAS-mutated tumours. TYMS expression measurements are worth-
Other studies also found that KRAS has a prognos- while to further study in future trials, while
tic but not predictive value [30]. However, for the survivin showed mostly conflicting results and
prediction of response to EGFR inhibitors, KRAS gene expression profiles with microarrays need
status has a predictive value. This has been further validation with larger patient groups.
5 What Biochemical and Molecular Biological Factors Have Greater Relevance to Treatment Decisions? 47

Taken together, the current value of bio- 10. Lamas MJ, Duran G, Gomez A et al (2012) X-ray
chemical and molecular biological factors in cross-complementing group 1 and thymidylate syn-
thase polymorphisms might predict response to
treatment decision making is rather low, how- chemoradiotherapy in rectal cancer patients. Int J
ever, with many promising developments in Radiat Oncol Biol Phys. Jan 1;82(1):138–144
the pipeline. Especially whole genome DNA 11. Lammering G (2005) Molecular predictor and prom-
or RNA analysis with copy number variation, ising target: will EGFR now become a star in radio-
therapy? Radiother Oncol 74:89–91
multiple mutation testing, full sequencing and 12. Lange MM, Maas CP, Marijnen CA et al (2008)
the association of genotype with phenotype, Urinary dysfunction after rectal cancer treatment is
will ultimately lead to more biomarker-based mainly caused by surgery. Br J Surg 95:1020–1028
treatment decisions in rectal cancer. However, 13. Leong KJ, Wei W, Tannahill LA et al (2011)
Methylation profiling of rectal cancer identifies novel
successful biomarker development needs col- markers of early-stage disease. Br J Surg 98:724–734
laboration, external validation and meta-analy- 14. Maas M, Beets-Tan RG, Lambregts DM et al (2011)
ses to reach the level of accuracy necessary to Wait-and-see policy for clinical complete responders
base treatment decisions on these markers. after chemoradiation for rectal cancer. J Clin Oncol.
Dec 10;29(35):4633–4640
15. Marijnen CA, Nagtegaal ID, Klein Kranenbarg E et al
(2001) No downstaging after short-term preoperative
radiotherapy in rectal cancer patients. J Clin Oncol
19:1976–1984
References 16. McDowell MT (2009) Increased spontaneous apopto-
sis, but not survivin expression, is associated with
1. Adelstein BA, Dobbins TA, Harris CA et al (2011) A histomorphologic response to neoadjuvant chemora-
systematic review and meta-analysis of KRAS status diation in rectal cancer. Int J Colorectal Dis 24:
as the determinant of response to anti-EGFR antibod- 1261–1269
ies and the impact of partner chemotherapy in meta- 17. Páez D et al (2011) Pharmacogenetic study in rectal
static colorectal cancer. Eur J Cancer. Jun;47(9): cancer patients treated with preoperative chemoradio-
1343–1354 therapy: polymorphisms in thymidylate synthase,
2. Bosset JF, Collette L, Calais G et al (2006) Chemo- epidermal growth factor receptor, GSTP1, and
therapy with preoperative radiotherapy in rectal DNA repair genes. Int J Radiat Oncol Biol Phys
cancer. N Engl J Med 355:1114–1123 81:1319–1327, Epub 2011 May 11
3. Bujko K, Glynne-Jones R, Bujko M (2010) Adjuvant 18. Peeters KC, van de Velde CJ, Leer JW et al (2005)
chemotherapy for rectal cancer. Ann Oncol. Late side effects of short-course preoperative radio-
Dec;21(12):2443 therapy combined with total mesorectal excision for
4. Grimminger PP (2010) XRCC1 gene polymorphism rectal cancer: increased bowel dysfunction in irradi-
for prediction of response and prognosis in the multi- ated patients– a Dutch colorectal cancer group study.
modality therapy of patients with locally advanced J Clin Oncol 23:6199–6206
rectal cancer. J Surg Res 164:e61–e66 19. Pettersson D, Cedermark B, Holm T et al (2010)
5. Hutchins G, Southward K, Handley K et al (2011) Value Interim analysis of the Stockholm III trial of preoper-
of mismatch repair, KRAS, and BRAF mutations in ative radiotherapy regimens for rectal cancer. Br J
predicting recurrence and benefits from chemotherapy Surg 97:580–587
in colorectal cancer. J Clin Oncol 2011(29):1261–1270 20. Popat S, Hubner R, Houlston RS (2005) Systematic
6. Kapiteijn E, Marijnen CA, Nagtegaal ID et al (2001) review of microsatellite instability and colorectal can-
Preoperative radiotherapy combined with total cer prognosis. J Clin Oncol 23:609–618
mesorectal excision for resectable rectal cancer. N Engl 21. Pucciarelli S, Del Bianco P, Efficace F et al (2011)
J Med 345:638–646 Patient-reported outcomes after neoadjuvant chemo-
7. Kim K, Chie EK, Wu HG et al (2011) High survivin radiotherapy for rectal cancer: a multicenter prospec-
expression as a predictor of poor response to preop- tive observational study. Ann Surg 253:71–77
erative chemoradiotherapy in locally advanced rectal 22. Radu C, Berglund A, Påhlman L et al (2008) Short-
cancer. Int J Colorectal Dis. Aug;26(8):1019–1023 course preoperative radiotherapy with delayed surgery
8. Koeberle D, Burkhard R, von Moos R et al (2008) in rectal cancer - a retrospective study. Radiother
Phase II study of capecitabine and oxaliplatin given Oncol 87:343–349
prior to and concurrently with preoperative pelvic 23. Rasheed S, Harris AL, Tekkis PP et al (2009)
radiotherapy in patients with locally advanced rectal Assessment of microvessel density and carbonic anhy-
cancer. Br J Cancer 98:1204–1209 drase-9 (CA-9) expression in rectal cancer. Pathol Res
9. Kuremsky JG (2009) Biomarkers for response to neo- Pract 205:1–9
adjuvant chemoradiation for rectal cancer. Int J Radiat 24. Roth AD, Tejpar S, Delorenzi M et al (2010)
Oncol Biol Phys 74:673–688 Prognostic role of KRAS and BRAF in stage II and
48 G. Lammering and J. Buijsen

III resected colon cancer: results of the translational 29. Swellengrebel HA, Marijnen CA, Verwaal VJ et al
study on the PETACC-3, EORTC 40993, SAKK (2011) Toxicity and complications of preoperative
60-00 trial. J Clin Oncol. Jan 20;28(3):466–474. chemoradiotherapy for locally advanced rectal cancer.
Epub 2009 Dec 14 Br J Surg 98:418–426
25. Rödel F (2002) High survivin expression is associated 30. Tol J, Dijkstra JR, Vink-Börger ME, et al (2010) High
with reduced apoptosis in rectal cancer and may predict sensitivity of both sequencing and real-time PCR
disease-free survival after preoperative radiochemo- analysis of KRAS mutations in colorectal cancer tis-
therapy and surgical resection. Strahlenther Onkol sue. J Cell Mol Med. Aug;14(8):2122–2131
178:426–435 31. Tan BR (2011) Thymidylate synthase genotype-
26. Rutten HJ, den Dulk M, Lemmens VE et al (2008) directed neoadjuvant chemoradiation for patients
Controversies of total mesorectal excision for rectal with rectal adenocarcinoma. J Clin Oncol 29:
cancer in elderly patients. Lancet Oncol 9:494–501 875–883
27. Sauer R, Becker H, Hohenberger W et al (2004) 32. Valentini V, Van Stiphout RGPM, Lammering G et al
Preoperative versus postoperative chemoradiotherapy (2011) Nomograms for predicting local recurrence,
for rectal cancer. N Engl J Med 351:1731–1740 distant metastases, and overall survival for patients
28. Stephens RJ, Thompson LC, Quirke P et al (2010) with locally advanced rectal cancer on the basis of
Impact of short-course preoperative radiotherapy for European randomized clinical trials. J Clin Oncol
rectal cancer patients’ quality of life: data from the 29:3163–3172
medical research council CR07/national cancer insti-
tute of Canada clinical trials group C016 randomized
clinical trial. J Clin Oncol 28:4233–4239
Do Different Populations of Rectal
Cancer Exist? 6
Vincenzo Valentini, Francesco Cellini,
Maria Cristina Barba, and Ruud van Stiphout

Contents The impact of new treatments for rectal cancer


6.1 Is It Possible to Obtain an Adequate has substantially improved patients clinical out-
Characterization of Tumour comes in the last years, but still worldwide col-
Heterogeneity, Based on Biological orectal cancer remains the third most common. In
Evidences?..................................................... 50 2000, colorectal cancer was responsible for 7.9%
6.2 Is It Possible to Obtain an Adequate of the world’s cancer deaths, with 492,000 deaths;
Characterization of Tumour it accounted for 9.4% of the world’s new cancers
Heterogeneity, Based on Clinical
Observation?................................................. 50
registered, with 945,000 cases diagnosed [1, 2].
Still remaining fundamental the role of surgery
6.3 Is It Possible to Identify These
for the curative approach to rectal cancer, the pub-
Different Groups Using Surrogate
Endpoints? .................................................... 52 lished clinical evidences of the last two decades
moved the management of such disease towards a
6.4 What Could Be a Methodology
That Provides Good Prediction multidisciplinary integrated treatment. In the
of Tumour Behaviour Basing frame of long course preoperative radiochemo-
on Surrogate Endpoints? ............................. 54 therapy, there are strong evidences that pathologi-
References ............................................................... 55 cal stage better discriminates different survivals in
a uniform patient population of locally advanced
presentations accrued in randomized clinical tri-
V. Valentini (*) • M.C. Barba als published after 2000. It supports the assump-
Department of Radiotherapy, tion that different populations of patients have
Università Cattolica Sacro Cuore, different sensibility to the same treatment [3].
Rome, Italy
All of these studies were based on the combi-
Policlinico Universitario ‘A. Gemelli’, nation of preoperative treatment and surgery, but
L. go Gemelli 8, Rome 00168, Italy
also, in series with patients treated with radiother-
e-mail: vvalentini@rm.unicatt.it; crissile@libero.it
apy alone, it is possible to observe the same het-
F. Cellini
erogeneity. Between 1978 and 1997, a group of
Radioterapia Oncologica,
Università Campus Biomedico, 271 patients affected by locally advanced rectal
Via E. Longoni 47, Rome 00155, Italy cancer underwent to primary radical external beam
e-mail: f.cellini@unicampus.it radiotherapy and was observed with long-term
R. van Stiphout follow-up. Patient received primary radical radio-
Department of Radiation Oncology (MAASTRO), therapy alone (without induction or concomitant
GROW – School for Oncology and Developmental
chemotherapy): treatment-administered doses
Biology, Maastricht University Medical Centre+,
Dr. Tanslaan 12, Maastricht 6201 BN, The Netherlands ranged from 40 to 60 Gy [4]. The 5-year cancer-
e-mail: ruud.vanstiphout@maastro.nl specific survival for the subgroup presenting a

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 49


DOI 10.1007/978-3-642-25005-7_6, © Springer-Verlag Berlin Heidelberg 2012
50 V. Valentini et al.

mobile lesion was 59% (95% CI 49–70%), for that of each evidence, understanding not only its
with partially fixed it was 33% (95% CI 21–54%), specific strength but also how the main evidences
and for the fixed tumour presentations was 9% mutually correlate. A detailed analysis is reported
(95% CI 4–20%). Even if tumour spread through in the chapter dealing with this issue [7].
rectal wall is prognostically significant for sur-
vival, each of the tumour presentation reported
subset (i.e., mobile, partially fixed and fixed) 6.2 Is It Possible to Obtain an
showed a rate of long-surviving patients (varying Adequate Characterization
according to the extension of the tumour): for the of Tumour Heterogeneity,
locally advanced lesions, up to 33% were cured by Based on Clinical Observation?
only intermediate dose of radiotherapy. This
behaviour highlights a different sensibility to the On the basis of the data reported by a recent study
same treatments that is irrespective of the macro- on pooled data from five large European random-
scopic initial presentation and based on intrinsic ized trials about the clinical behaviour of the differ-
tumour heterogeneity, supporting the concept that ent populations in locally advanced rectal cancer,
biological tumour heterogeneity leads to different the four subgroups [8] identified are as follows:
clinical patterns of responses and tumour progres- • Very good (15–20%): curable patients, highly
sion. As Mohiuddin et al. highlighted, indeed rec- sensible to radiation, in whom organ preserva-
tal cancer represents a broad spectrum of diseases tion is to be pursued
requiring tailored treatment regimens to maximize • Good (40–50%): curable patients, who require
the outcome [5]. It aims to use the most aggressive good local treatment and could have some
therapies only for the high-risk-featured presenta- benefit by adjuvant chemotherapy
tions, avoiding the less biologically aggressive • Bad (10%): patients with late recurrent dis-
tumours from the more intense treatment sched- ease, who require radical treatment when
ules. To tailor treatments, there are some questions tumour recurs
which have to be considered in the multidisci- • Ugly (15–20%): patients with early metasta-
plinary management of rectal cancer. ses onset, whom ongoing chemotherapy has
still not been able to cure
The analysis of overall risk ratio between DM
6.1 Is It Possible to Obtain an rate and LR rate (Fig. 6.1) reveals that at the start
Adequate Characterization
of Tumour Heterogeneity, 6
Based on Biological Evidences?
5.5
There is nowadays a great expectation about the
5
likelihood to determine tumour heterogeneity bas-
DM rate/LR rate [−]

ing on biological evidences. From the main clinical 4.5


evidences on biomarkers associated to rectal can-
cer heterogeneity, we could summarize that EGFR 4
expression have shown a role in the prediction of
3.5
the response, as well as mutation status of BRAF,
dMMR and KRAS (even if only in a reduced pro- 3
portion of cases for the last). However, since many
studies available in literature seem promising, the 2.5

identification of a validated and well correlated to 2


the clinical outcomes biological marker cluster 0 2 4 6 8 10
needs yet a lot of investigation and is still lacking Time of follow− up [years]
[6]. Some studies obtained controversial results, Fig. 6.1 Ratio of distant metastasis rate and local recur-
and it is still also difficult to handle the importance rence rate over the time of follow-up in years
6 Do Different Populations of Rectal Cancer Exist? 51

Fig. 6.2 Percentages of the 40


outcomes LR, DM and death
over the follow-up time as a
total of the outcome events 30

LR [% of total LR]
20

10

0
1 2 3 4 5 6 7 8 9 10

40
DM [% of total DM]

30

20

10

0
1 2 3 4 5 6 7 8 9 10

40

30
Death [% of total]

20

10

0
1 2 3 4 5 6 7 8 9 10
Time of follow-up [years]

of follow-up, there are six times more metastases Early recurrent patients (ugly), which are
than local recurrences. This number decreases to affected by highly aggressive tumour. This group
around 2.5 in the first 2 years and remains con- affects 15–20% of locally advanced tumour pre-
stant afterwards. Figure 6.2 shows that around sentations. They do not seem to get benefit from
80% of the recurrences occur within the first the current treatment intensification and chal-
4 years, while the occurrence of death is more lenge clinicians for better strategies.
frequent from the second to the fourth year Late recurrent patients (bad) probably with
consequently. slightly better pattern of features respect the pre-
Overall, in recurrent patients, two typologies vious one and late onset as for a slow growth, but
of behaviour are identified: that can occur early although aggressive on itself. This presentation
for the most aggressive or late for the less occurs in at least 10–15% of patients, and it
aggressive but still bad prognosis associated benefits from aggressive approach by integrated
tumours. treatment to optimize the curative chance.
52 V. Valentini et al.

On the other hand, two typologies of behav- antigen (PSA) as a surrogate endpoint for sur-
iour are both characterized by chance of cure: the vival in prostate cancer. In colon cancer, 2–3 years
good are patients for whom the cure is achievable of disease-free survival (DFS) correlated better
by the use of the fully integrated treatment, as with 6-year overall survival after adjuvant che-
neoadjuvant chemoradiation or radiation alone motherapy in an analysis of a large pooled data-
followed by surgery ± adjuvant chemotherapy. base from randomized trials [9].
They represent 40–50% of locally advanced rec- For rectal cancer, pathologic complete response
tal cancer patients. (pCR) was argued to identify this more favour-
Finally, there is a group of patients in locally ably in population with less local recurrences
advanced presentations undergoing to the inte- (LR) and distant metastases (DM) and better
grated schedule that shows a high response to overall survival (OS) [10, 11]. Anyway, no ran-
neoadjuvant therapy (the best), suggesting that domized studies showed a significant benefit for
are potentially curable also avoiding surgery. survival for increasing pCR rates [12], which
The random distribution of these four groups questions the role of promoting pCR in preopera-
of patients in the different series could justify the tive approaches.
missing of evidences of clinical benefit for the In a recent analysis of a pooled database of
treatment strategies when they are addressed to five randomized trials, the comparison between
one of them, namely, but applied to the whole the pCR and non-pCR population with respect to
cohort of patients; all single trials of neoadjuvant long-term outcome, all KM curves differed
chemoradiotherapy followed by surgery failed to significantly (Fig. 6.3). After 10 years, the pCR
show a 5-year survival benefit because the ugly population had 11% more local control than the
and bad component hid the benefit of this treat- non-pCR population (96% vs. 85%). For DM and
ment modality in the good and the best ones. The OS, these numbers are 18% (82% vs. 64%) and
main problem is how to distinguish them to tailor 16% (73% vs. 57%).
treatment properly. There are evidences that pCR patients have a
better outcome, but this marker does not have an
impact on the outcome of any single randomized
6.3 Is It Possible to Identify These trial, and the relative small benefit in survival
Different Groups Using (16%) support the value of this surrogate end-
Surrogate Endpoints? point in identifying only a group of patients with
favourable outcome, for whom surgery could be
In a time of limited sources, there is a need to avoided, but not in providing a reliable prediction
have fast validation of new hypotheses to improve for the whole cohort of locally advanced rectal
cancer treatment. One way is to have reliable sur- cancer patients and, namely, about the impact of
rogate endpoints, for example, prostate-specific each treatment in the ugly ones.

1.0 1.0 1.0

0.9 0.9 0.9

0.8 0.8 0.8


DC (-)

OS (-)
LC (-)

0.7 0.7 0.7

0.6 0.6 0.6

0.5 0.5 0.5


0.00 60.00 120.00 180.00 0.00 60.00 120.00 180.00 0.00 60.00 120.00 180.00
Time of follow−up [months] Time of follow−up [months] Time of follow−up [months]

Fig. 6.3 Outcome rates local control (LC), distant control (DC) and overall survival (OS) for pCR subpopulation
(green) versus the non-pCR population (blue)
6 Do Different Populations of Rectal Cancer Exist? 53

1.0 1.0

0.8 pCR 0.8


Disease free at 2 years

0.6 0.6
OS (–)

Non−pCR
0.4 0.4

0.2 0.2

Recurrence within 2 years


0.0 0.0
0.00 50.00 100.00 150.00 0.00 50.00 100.00 150.00

Fig. 6.4 Comparison between the status of pCR and disease free at 2 years for overall survival (OS)

Fig. 6.5 Overall survival rate


(OS) over time for subpopula- 1.0
tions having different status pCR+disease free
for pCR and disease free at
2 years
0.8
Non−pCR+disease free

0.6
OS (–)

0.4

pCR+recurrence
0.2

Non−pCR+recurrence
0.0

0.00 50.00 100.00 150.00


Time of follow−up [months]

In the same study, a further analysis compared recurrence + non-pCR is 0.083, which means that
overall survival of the populations of pCR and being disease free after 2 years can be used as a
disease free after 2 years (Fig. 6.4). Disease free surrogate endpoint for pCR, since knowing the
after 2 years is more beneficial than having just a pCR status does not change overall survival
pCR. A recurrence within 2 years results in 13% significantly when knowing the recurrence state
chance of survival within 10 years, while having after 2 years (Fig. 6.5).
no PCR results in 58% chance of survival after Two-year disease-free survival as an endpoint
10 years. The p value between disease-free + may however be useful to predict survival for the
pCR versus disease-free + non-PCR is 0.396, and subpopulations in the group of more aggressive
the p value between recurrence + pCR versus tumours. This endpoint may promote different
54 V. Valentini et al.

chemotherapy regimens to reduce distant metas- in many forms [13]. Interpretation of the model
tases rates for this subpopulation, but could hide results is very much dependent on model
the overtreatment for the best and the good representation. One highly interpretable way
patients when all are treated by the same approach. to represent a prediction model is the nomogram.
Diversifying the options to personalize treatment A nomogram is nothing more than a visual repre-
is essential with the current increasing complex- sentation of mathematical equations involving
ity of clinical decision making. These hypotheses variables, weights and constants. An example for
require finalized analyses based on large database rectal cancer is given in Fig. 6.6. In the example,
with pooled individual patients, namely, from nomogram six variables were found to be predic-
randomized trials, to identify the different patient tive for the outcome local control. Each of these
population by reliable and validate predictive predictors is represented by a scale with possible
models as early as possible along the multiphase values. When using the nomogram to make a pre-
treatment programmes. diction for a single patient, one may fill in each of
the values for the predictors and read out the score
scale on top by drawing a vertical line between the
6.4 What Could Be a Methodology scales. These scores need to be summed and filled
That Provides Good Prediction in on the scale ‘total score.’ Subsequently, this
of Tumour Behaviour Basing final score corresponds to a probability for the
on Surrogate Endpoints? outcome by reading out the bottom scale. Often,
risk groups are defined or calculated based on this
The presented patient and tumour heterogeneity probability, even if the prediction gives a continu-
allows prediction of primary and surrogate end- ous probability score, because in this way patients
points with the aim of individualizing cancer are assigned to a certain subgroup which could be
treatment. Statistical prediction models able to treated less or more aggressive. Trials could be
find multivariate correlations between patient and designed based on these developed prediction
tumour characteristics and outcome are available models to encourage treatment individualization.

Single score
−2 −1 0 1 2
pT-stage
0 1 2 3 4
pN-stage
0 1 2
Concomitant chemo
Yes No
Adjuvant chemo
Yes No
Radiotherapy dose [Gy]
>45 45 <45
Tumor location
High Mid Low

Total score
−3 −2 −1 0 1 2 3 4 5
Low Medium High
Probability of LR [−]
0.05 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 0.95

Fig. 6.6 Example of a nomogram for locally advanced rectal cancer; prediction of local recurrence (LR) probability
based on clinicopathological data. Three risk groups were defined based on the outcome probabilities
6 Do Different Populations of Rectal Cancer Exist? 55

High prediction accuracy is however a necessity predicts disease-free survival in patients with locally
which can only be achieved with high-quality, advanced rectal cancer receiving neoadjuvant chemo-
radiotherapy. BMC Cancer 11:363
unbiased data from different origins. 7. Lammering G, Buijsen J (2012) What biochemical
and molecular biological factors have greater rele-
Conclusions vance to treatment decisions? In: Multidisciplinary
It is a priority to take in the right account that management of rectal cancer – questions and
answers
a strong component of heterogeneity exists in 8. Valentini V, van Stiphout R, Lammering G et al
rectal cancers: that implies the needing to per- 2-Year Disease Free vs Pathological Complete
sonalize treatments. The correct use of surro- Response as a Surrogate Endpoint by using Pooled
gated endpoints and nomograms can aid to Data of Randomized Trials for Locally Advanced
Rectal Cancer. In Press
tailor the treatments. 9. Sargent D, Shi Q, Yothers G (2011) Two or three year
disease-free survival (DFS) as a primary end-point in
stage III adjuvant colon cancer trials with fluoropy-
rimidines with or without oxaliplatin or irinotecan:
data from 12,676 patients from MOSAIC, X-ACT,
References PETACC-3, C-06, C-07 and C89803. Eur J Cancer
47(7):990–996, Epub 2011 Jan 21
1. Parkin DM (2001) Global cancer statistics in the year 10. Valentini V, Coco C, Picciocchi A (2002) Does down-
2000. Lancet Oncol 2(9):533–543 staging predict improved outcome after preoperative
2. National Cancer Institute: PDQ® Rectal Cancer chemoradiation for extraperitoneal locally advanced
Treatment. Bethesda, MD: National Cancer Institute. rectal cancer? A long-term analysis of 165 patients.
(2011) http://cancer.gov/cancertopics/pdq/treatment/ Int J Radiat Oncol Biol Phys 53(3):664–674
rectal/HealthProfessional 11. Capirci C, Valentini V, Cionini L (2008) Prognostic
3. Valentini V, van Stiphout RG, Lammering G, value of pathologic complete response after neoadju-
Gambacorta MA, Barba MC et al (2011) Nomograms vant therapy in locally advanced rectal cancer: long-
for predicting local recurrence, distant metastases, term analysis of 566 ypCR patients. Int J Radiat Oncol
and overall survival for patients with locally advanced Biol Phys 72(1):99–107, Epub 2008 Apr 11
rectal cancer on the basis of European randomized 12. Sebag-Montefiore D, Bujko K, Valentini V (2009)
clinical trials. J Clin Oncol 29:3163–3172 Rectal cancer multidisciplinary management: evi-
4. Wang Y, Cummings B, Catton P et al (2005) Primary dences and future landscape. Radiother Oncol
radical external beam radiotherapy of rectal adenocar- 92(2):145–147, Epub 2009 Jul 10
cinoma: long term outcome of 271 patients. Radiother 13. van Stiphout R, Roelofs E, Dekker A (2012) How to
Oncol 77:126–132 share data and promote a rapid learning health
5. Mohiuddin M, Ahmad N, Marks G (1993) A selective medicine? In: Multidisciplinary management of rectal
approach to adjunctive therapy for cancer of the rec- cancer – questions and answers
tum. Int J Radiat Oncol Biol Phys 27(4):765–772
6. Schwaab J, Horisberger K, Ströbel P (2011)
Expression of transketolase like gene 1 (TKTL1)
Part III
Q&As on Imaging
How Can We Identify Tumour
Penetration? 7
Manish Chand and Gina Brown

Contents 7.1 Introduction


7.1 Introduction ..................................................... 59
Tumour penetration is seen as the depth of inva-
7.2 Endoanal Ultrasound (EAUS) ........................ 61
sion through the layers of the bowel wall and into
7.3 Magnetic Resonance Imaging (MRI) ............ 61 the surrounding mesorectum. It is an important
7.4 Extent of Mesorectal Spread .......................... 62 component of the staging system during assess-
ment of rectal cancer. Historically, the Duke’s
7.5 CRM Involvement ........................................... 63
staging system has been used to risk-stratify
7.6 Summary .......................................................... 65 patients and influence treatment decisions. In
References ................................................................. 65 addition to tumour depth, it includes assessment
of local lymph node infiltration. In recent years
with increasing use of oncological therapy to
accompany surgical treatment of rectal cancer,
traditional staging systems have been found lack-
ing in detail. The UICC TNM classification has
largely replaced the Duke’s staging system,
although many pathologists still report both.
Stage at presentation strongly influences progno-
sis and in particular tumour depth. The risk of
recurrence for stage T1, T2 and T3 independent
of lymph node involvement is 5%, 10% and 25%,
respectively [1]. As the trend has shifted away
from adjuvant therapy to neoadjuvant therapy
due to improved outcomes and less morbidity,
staging information on the tumour is ideally
needed at an early stage in the diagnosis. Accurate
M. Chand (*) tumour stage is important in determining the
Colorectal Research Fellow
appropriateness of preoperative chemoradiother-
and Registrar in Colorectal Surgery,
Royal Marsden Hospital, Downs Road, Sutton apy (CRT), which has been shown to improve
e-mail: mans001@aol.com overall outcomes in more advanced tumours.
Sufficient detail cannot be obtained from biop-
G. Brown
sies alone.
Consultant Radiologist, Royal Marsden Hospital,
Downs Road, Sutton Dukes originally noted the link between
e-mail: gina.brown@rmh.nhs.uk increasing depth of spread and risk of nodal

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 59


DOI 10.1007/978-3-642-25005-7_7, © Springer-Verlag Berlin Heidelberg 2012
60 M. Chand and G. Brown

Table 7.1 Standard TNM classification of rectal cancer


Primary tumour (T)
TX Primary tumour cannot be assessed
T0 No evidence of primary tumour
Tis Carcinoma in situ
T1 Tumour invades submucosa
T2 Tumour invades muscularis propria
T3 Tumour invades through muscularis propria into the subserosa
T4 Tumour directly invades other structures and/or perforates
visceral peritoneum
Regional lymph nodes (N)
NX Regional nodes cannot be assessed
N0 No node metastases
N1 Metastases in 1–3 regional nodes
N2 Metastases in four or more nodes
Distant metastases (M)
MX Distant metastases cannot be assessed
M0 No distant metastases
M1 Distant metastases

involvement and the importance of tumour spread wall and surrounding mesorectum, and this has
in millimetres beyond the muscularis propria – significant bearing on treatment decisions. The
extramural spread. It has been further noted in only preoperative modality that has been shown
several histopathology studies. As a result, to measure depth of tumour spread with precision
Hermanek proposed a modification of the TNM is high-resolution MRI (Table 7.1).
classification to take into account the survival Accurate preoperative identification of tumour
variation amongst patients with T3 tumours depth is required for:
according to depth of extramural spread. 1. Successful TME surgery
The single-most important factor which has 2. Patient selection for neoadjuvant CRT
led to reduction in local recurrence rates is the 3. Limiting overtreatment of patients with CRT
acceptance of total mesorectal excision (TME) The importance of accurate identification of
as the ‘gold-standard’ surgical technique for T-stage relates to its treatment implications and
rectal cancer surgery. Sharp, precision dissec- prognosis. In essence, tumours which are confined
tion along the mesorectal fascia ensures that to the bowel wall, that is, T1 and T2 tumours, are
the tumour and all local draining lymph nodes referred for immediate surgery and do not need
are excised in a single package. Knowledge of preoperative chemoradiotherapy. In comparison,
the relationship between the tumour edge and tumours which penetrate beyond the bowel wall
the mesorectal fascia is essential to facilitate a into the surrounding mesorectum and beyond,
successful dissection and give a tumour-free e.g. T3 and T4 tumours, are considered for pre-
resection margin. operative chemoradiotherapy to improve overall
As imaging modalities have improved, prog- outcomes. Furthermore, distinguishing early
nostic risk factors in rectal cancer can be demon- T-stage tumours has implications in the type of
strated with much greater accuracy. Tumour surgery. For example, T1 lesions may be suitable
depth or T-stage is one such factor; however, for a less traumatic local excision such as an
there is no single optimal staging classification endoscopic mucosal resection rather than ante-
which can be applied to the entire spectrum of rior resection and total mesorectal excision.
T-stage. Tumour spread can be measured to Clearly, accurate T-staging is essential so not to
within millimetres beyond the layers of the bowel over- or undertreat patients.
7 How Can We Identify Tumour Penetration? 61

7.2 Endoanal Ultrasound (EAUS) 7.3 Magnetic Resonance


Imaging (MRI)
Endoanal ultrasound is considered as the modality
of choice in the local assessment of early rectal MRI is the optimal method for local assessment of
cancer by many clinicians. Its suitability for early primary rectal cancer as a whole. Technique is
lesions relates to identification of those patients important to obtain accurate images and not under-
where local resection may be more appropriate or over-stage tumours. This requires correct field
rather than TME surgery. This is because it has alignment in relation to the long axis of the rectum.
greater accuracy than MRI in visualizing the sub- The structure of the rectal wall can be readily
mucosa which is essential in planning local resec- identified on MRI. T2-weighted images are most
tion. However, its main limitation is its inability to suitable for distinguishing the layers of the bowel
define the mesorectal facia accurately and thus wall. The different layers of the bowel wall are
identify whether the circumferential resection mar- identified from their unique signal characteristic.
gin is threatened. In a large multi-centre trial, EAUS An appreciation of the normal anatomy is essential
was shown to substantially over-stage patients and for accurate identification of tumour invasion.
thus over-treat patients with preoperative CRT [2]. Disruption through the normal signal characteristic
Involvement of the circumferential resection mar- pattern determines the depth of invasion (Fig. 7.1).
gin is known to be a significant factor in local recur- As mentioned, the main advantage over EAUS
rence and is essential in surgical planning. is that MRI can accurately delineate the mesorectal

Fig. 7.1 MRI axial image


showing a rectal cancer and
the different signal character-
istics of the rectal wall and
surrounding tissues
62 M. Chand and G. Brown

fascia. The circumferential resection margin is early T3 tumours may not actually be as clini-
defined by the relationship between the tumour cally relevant as first thought.
edge and the mesorectal fascia. It is known to be an
independent risk factor for local recurrence. In
those patients where the CRM is threatened, neo- 7.4 Extent of Mesorectal Spread
adjuvant aims to downsize the tumour to obtain
tumour-free resection margins following TME sur- The mesorectum is the lymphovascular enve-
gery. Accurate identification and definition of the lope which surrounds the rectum to varying
mesorectal fascia is therefore mandatory in plan- degrees. It completely surrounds the middle
ning optimal treatment. third of the rectum but is virtually absent in the
Brown et al. first demonstrated the accuracy lower third and only covers the posterior aspect
of high-resolution MRI in preoperative predic- of the upper third of the rectum. The extent to
tion of rectal cancer stage [3]. In their study of which tumour penetrates the mesorectal fat is an
28 patients, they not only correlated tumour independent risk factor for recurrence. This is
stage on MRI with histological analysis but not adequately represented in the traditional
showed that MRI can accurately identify extent staging classifications as the level of penetra-
of extramural spread. One of the difficulties in tion into the mesorectum has prognostic
predicting tumour stage on MRI is distinguish- implications.
ing between T2 and T3 tumours. The patho- As described above, total mesorectal exci-
logical difference between T2 and T3 tumours sion (TME) involves excision of the rectum
is invasion of the mesorectal fat. Defining the along with the mesorectum. T3 tumours are
MR characteristics for T3 tumours is therefore those which have traversed the bowel wall and
important so to avoid over-staging. The pres- penetrated into the mesorectum. The majority
ence of spiculation within the extramural fat of patients present with T3 tumours; however,
can lead to misinterpretation and over-staging there is wide variety in survival rates of these
of tumours. Both the tumour itself and the patients. Jass and colleagues were the first to
effects of radiotherapy can produce a host des- describe the importance of the extent of
moplastic reaction resulting in fibrosis. This mesorectal invasion on prognosis [5]. Further
fibrotic reaction can cause spiculation into the work by Cawthorn and Merkel demonstrated an
mesorectal fat and should not be confused with improvement in 5-year survival in patients who
tumour. Through careful correlation with his- had ‘slight’ mesorectal invasion. Cawthorn
topathology whole-mount sections, Brown described ‘slight’ mesorectal invasion as 4 mm
et al. defined T3 tumours by the presence of or less and found 5-year survival to be 55%
tumour signal intensity permeating into the compared to 25% when more than 4 mm [6].
mesorectal fat with a ‘broad-based’ or nodular Another study by Merkel et al. studied patient’s
configuration and in continuity with the intra- survival characteristics with T3 tumours. Those
mural portion of the tumour. Such features patients with extramural spread of more than
could only be appreciated on small field of 5 mm had 5-year survival rate of 54% compared
view high-resolution imaging (3-mm slices, with 85% for those patients whose tumours had
16 cm field of view). Further study of prognos- extramural spread of less than 5 mm [7]. These
tic factors using high-resolution MRI by Brown results were independent of lymph node involve-
and colleagues demonstrated 94% concordance ment. These early studies highlight the impor-
between MRI and histology in terms of T-stage tance of accurate measurement of tumour
in addition to 92% agreement in prediction of penetration into the mesorectum and those
the CRM [4]. tumours with a worse prognosis, namely, T3c
Figure 7.2 shows the difference between and T3d. Therefore, the distinction between T2
fibrotic T2 lesions and actual T3 lesions. However, and T3 tumours with less than 5-mm mesorectal
as we shall see, distinguishing T2 cancers, which spread – T3a and T3b – becomes irrelevant as
would normally not need preoperative CRT, and these patients will have minimal benefit from
7 How Can We Identify Tumour Penetration? 63

Fig. 7.2 (a) This shows the baseline MRI axial image red arrow indicates an example of fibrosis which may be
before chemoradiotherapy and the same patient following mistaken for tumour thus overstaging. (b) MRI axial
chemoradiotherapy. The low signal represents the fibrosis image of a T3 tumour
that has taken place but the actual tumour remains T2. The

CRT. The accuracy of MRI in delineating the 7.5 CRM Involvement


mesorectal fascia and producing comparable
results to histological analysis has been shown The importance of TME surgery in the reduction in
by the MERCURY Study Group (Table 7.2 and risk of local recurrence has been mentioned above.
Fig. 7.3). Key to a successful resection is obtaining tumour-free
64 M. Chand and G. Brown

circumferential margins. Tumour cells identified at to the mesorectal fascia. Stratifying T3 tumours
the circumferential resection margin is known to con- through the sub-classification described above is
fer poor prognosis in terms of local recurrence and important when deciding preoperative oncological
overall survival. This is related to the depth of inva- therapy. However, identification of those patients in
sion into the mesorectum and proximity of the tumour whom the CRM would be involved is equally impor-
tant in surgical planning. These patients with threat-
ened CRM have been shown to benefit from
Table 7.2 T3 sub-classification
neoadjuvant chemoradiotherapy to downstage the
T-stage Depth of extramural spread tumour and thus allowing for a clear resection
T3a <1.00 mm margin.
T3b 1.01–5.00 mm MRI has been the most useful imaging modal-
T3c 5.01–15.00 mm
ity in identifying the mesorectal fascia. Early
T3d >15.00 mm
studies correlating MRI’s accuracy in identifying

a b

c d

Fig. 7.3 (a–d) Subclassification of T3 tumours – T3a–d


7 How Can We Identify Tumour Penetration? 65

the mesorectal fascia and a subsequent negative nodal status and is independent of known vari-
CRM on histology have led to patients in whom ables such as type of surgery and height of
tumour extends to within 5 mm of the mesorectal tumour. Conversely, patients with more extensive
fascia to be given preoperative CRT [8]. Although tumour spread of 5 mm or more beyond the mus-
detailed staging is important to identify patients cularis propria are at increased risk of local recur-
who would most benefit from preoperative onco- rence and distant failure, and therefore,
logical therapy, it is also important not to offer preoperative identification of such patients will
patients CRT where there is no proven benefit. lead to appropriate intensification of preoperative
The MERCURY Study Group showed that it is therapy aimed at reducing such risks.
possible to predict negative CRM histologically
using a 1 mm cut-off on MRI. Further analysis
using different cut-offs (1, 2 and 5 mm) did not References
show any increase in risk of local recurrence [9].
Thus, a distance of 1 mm from the mesorectal 1. Sobin LH, Wittekind C, International Union against
Cancer (1997) TNM classification of malignant
fascia on MRI is the optimal cut-off to identify tumours, 5th edn. Wiley, New York, p xxiii, 227 p
patients who may have positive resection margins 2. Sauer R et al (2004) Preoperative versus postoperative
following surgery. It is important to state that a chemoradiotherapy for rectal cancer. N Engl J Med
certain proportion of patients who have negative 351(17):1731–1740
3. Brown G et al (1999) Rectal carcinoma: thin-section
CRM will still suffer a local recurrence. This sug- MR imaging for staging in 28 patients. Radiology
gests that there are additional phenotypical fac- 211(1):215–222
tors which remain important in the local 4. Brown G et al (2003) Preoperative assessment of prog-
recurrence and may be able to be identified on nostic factors in rectal cancer using high-resolution
magnetic resonance imaging. Br J Surg 90(3):355–364
pretreatment imaging. 5. Jass JR, Love SB, Northover JM (1987) A new prog-
nostic classification of rectal cancer. Lancet 1(8545):
1303–1306
6. Cawthorn SJ et al (1990) Extent of mesorectal spread
7.6 Summary and involvement of lateral resection margin as prog-
nostic factors after surgery for rectal cancer. Lancet
In summary, preoperative MRI measurement of 335(8697):1055–1059
the extramural depth of spread and assessment of 7. Merkel S et al (2001) The prognostic inhomogeneity in
pT3 rectal carcinomas. Int J Colorectal Dis 16(5):
tumour to the potential CRM enables the selec-
298–304
tion of good prognosis rectal cancer patients. In 8. Beets-Tan RG et al (2001) Accuracy of magnetic reso-
clinical practice, this can account for at least one- nance imaging in prediction of tumour-free resection
third of the patients with rectal cancer. With opti- margin in rectal cancer surgery. Lancet 357(9255):
497–504
mal preoperative selection and good quality TME
9. Taylor FG et al (2011) One millimetre is the safe cut-off
surgery, local recurrence rates of 1.7% are achiev- for magnetic resonance imaging prediction of surgical
able in MRI-defined T3a/b. This is regardless of margin status in rectal cancer. Br J Surg 98(6):872–879
How Can We Identify Mesorectal
Fascia Involvement? 8
Regina G.H. Beets-Tan

Contents
8.1 Introduction ..................................................... 67 8.1 Introduction
8.2 What Is the Mesorectal Fascia? ..................... 67
The circumferential resection margin (CRM) is
8.3 Why Do We Need to Evaluate the lateral or radial resection margin created by the
Involvement of the Mesorectal Fascia
surgeon at the total mesorectal excision. A positive
Preoperatively?................................................ 68
CRM is defined as a closest distance of 1 mm or
8.4 Can EUS Visualize the Mesorectal less between tumor and resection margin, as this
Fascia and Its Involvement? ........................... 68
represents the optimal prognostic cutoff point. The
8.5 How Accurately Can CT Identify importance as a prognostic factor and as a param-
Advanced Tumors, Threatening
or Invading the Mesorectal Fascia? ............... 68
eter of surgical quality has been recognized and
confirmed in the past 20 years [1]. Because the
8.6 Is MRI an Accurate Imaging Method to ideal plane of resection in a total mesorectal exci-
Visualize the Mesorectal Fascia and Its
Involvement?.................................................... 69 sion is just outside the mesorectal fascia, a relevant
question in rectal cancer surgery which is addressed
8.7 Can a Restaging MRI After
Preoperative Chemoradiotherapy
in this chapter is “How can we identify mesorectal
Help in Identifying Tumor Regression fascia involvement?”
from the Mesorectal Fascia? .......................... 69
References ................................................................. 71
8.2 What Is the Mesorectal Fascia?

The mesorectal fascia, anatomically known as


the fascia propria is a continuous fascial sleeve
surrounding the rectum and mesorectum that can
be dissected as a complete sock off a fresh extra-
fascial specimen. It is 154 (±1 standard devia-
tion = 61–391) mm thick, is thinner anteriorly than
posteriorly (P < 0.05), and is composed predomi-
R.G.H. Beets-Tan nantly of collagen. It can be identified surgically
Department of Radiology,
at the pelvic brim as a shiny membrane and lies
Maastricht University Medical Centre,
5800, Maastricht, 6202 AZ, The Netherlands inside the hypogastric nerves and the pelvic plex-
e-mail: r.beets.tan@mumc.nl uses [2].

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 67


DOI 10.1007/978-3-642-25005-7_8, © Springer-Verlag Berlin Heidelberg 2012
68 R.G.H. Beets-Tan

8.3 Why Do We Need to Evaluate hampers the visualization of the ultrathin


Involvement of the Mesorectal mesorectal fascia. Furthermore, because of its
Fascia Preoperatively? limited field of view, evaluation of tumor involve-
ment of anatomical structures outside the scope
A positive circumferential resection margin of the probe is a challenging and often a difficult
(CRM) can be the result of inadequate total task. This is especially true in high rectal tumors
mesorectal excision (TME) surgery or of an that have extended to the dorsal pelvic wall. But
advanced tumor that comes close to or invades when it comes to identifying tumor involvement
the mesorectal fascia. The first problem is one of the anterior mesorectal fascia including the
of surgical technique, whereas the second is a assessment of anterior organ invasion (vagina,
matter of preoperative identification and ade- prostate, or seminal vesicles), EUS is as accurate
quate neoadjuvant treatment of advanced tumors. as MRI.
For centers that use only a long course of chemo-
radiation as a neoadjuvant treatment, the dis-
tance of the tumor to the mesorectal fascia is 8.5 How Accurately Can CT Identify
usually not very important in the preoperative Advanced Tumors,
decision process, as all tumors that extend Threatening or Invading the
beyond the muscular wall are considered candi- Mesorectal Fascia?
dates for a long course of chemoradiation, pro-
viding an opportunity for downsizing. For A Dutch prospective multicenter study investi-
centers that also use a short course of 5 × 5 Gy gated the role of 16-slice CT (MSCT) for the pre-
and immediate surgery, this is different. Although diction of an involved mesorectal fascia in 250
it has been shown that 5 × 5 Gy is a very efficient patients [4]. The results show that MSCT was
and cost-effective way to prevent local recur- especially accurate in high tumors that have a
rences in many patients, it is much less effective large distance of the tumor to the mesorectal fas-
when the tumor comes close to or invades the cia with a sensitivity, specificity, PPV, and NPV
mesorectal fascia [3]. These tumors should be of 70%, 96%, 85%, and 92% for the prediction of
identified and treated with a long course of an involved mesorectal fascia. For low rectal
chemoradiation and a long interval to provide tumors, the accuracy was only moderate. Another
downsizing. study confirmed the high negative predictive
Regardless of the neoadjuvant treatment strat- value for the prediction of an involved mesorectal
egies, it is important for the surgeon to know the fascia and considerable overstaging for CT [5].
exact anatomic relation of the tumor to the The inherent low contrast resolution of CT
mesorectal fascia and the surrounding structures together with the complex tapered anatomy of
to obtain a complete resection, and in advanced the mesorectum with little or no fat between
cases, it can be very valuable to have the images tumor and pelvic structures low down in pelvis
available in the operating room. all contribute to the inaccuracy of CT in this
area (Fig. 8.1). The poor accuracy of CT in low
anteriorly located tumors was confirmed by a
8.4 Can EUS Visualize the study that also demonstrated a high variability
Mesorectal Fascia and Its among observers for prediction of an involved
Involvement? mesorectal fascia [6]. The role of new-genera-
tion multidetector-row CT has been addressed
Endorectal ultrasonography (EUS) is inaccurate in one study that showed a reasonable agree-
in identifying patients with a “threatened or ment between CT and MRI but not for predic-
involved mesorectal fascia,” because the tion of tumor involvement in the mesorectal
mesorectal fascia is not well visualized by EUS. fascia in low tumors. Therefore, CT cannot
The inherent low soft-tissue contrast resolution replace MRI in rectal cancer staging [7].
8 How Can We Identify Mesorectal Fascia Involvement? 69

Fig. 8.1 (a, b) Axial CT and T2 weighted MRI of a patient while on the corresponding MRI the tumor is not adjacent
with rectal cancer. On CT a low rectal tumor mass is visual- to the mesorectal fascia but can be delineated at short dis-
ized invading the anterior mesorectal fascia (white arrows) tance from the anterior mesorectal margin (black arrows)

A subsequent large European multicenter


8.6 Is MRI an Accurate Imaging Mercury study showed an accuracy of 91% with
Method to Visualize the a negative predictive value of 93% for patients
Mesorectal Fascia and Its who underwent immediate surgery [12].
Involvement? Two European centers that use a short course
of radiotherapy as a treatment option report a
Magnetic resonance imaging (MRI) is the method decrease in the number of positive margins after
of first choice to identify the mesorectal fascia the incorporation of MRI in the discussion of all
because of its inherent superior contrast resolu- patients with rectal cancer in multidisciplinary
tion as compared to EUS or CT. The ultrathin meetings [13, 14].
fascia can be sharply delineated within the high-
intensity pelvic fat tissue on T2-weighted MR
images (Fig. 8.2). A standard rectal cancer MR 8.7 Can a Restaging MRI After
scan protocol should therefore include sequences Preoperative
with a bright signal of fat, T2W FSE sequences. Chemoradiotherapy Help in
If fat-suppressed sequences are applied, the con- Identifying Tumor Regression
trast between fascia and surrounding fat tissue from the Mesorectal Fascia?
diminishes and mesorectal fascia involvement
will be difficult to determine. Many expert sin- A restaging MRI, performed 6–8 weeks after
gle-center studies have shown that MRI is highly completion of chemoradiotherapy is useful for
accurate for predicting an involved mesorectal evaluation of response to neoadjuvant treatment.
fascia (see Fig. 8.3a) [2, 8–10]. There is evidence that MRI can predict tumor
The results of a systematic review of all pub- regression of locally advanced primary rectal
lished single-center studies confirm the high per- cancer that has undergone preoperative chemora-
formance of MRI, showing a sensitivity for the diotherapy [15]. MRI after chemoradiation has a
prediction of an involved mesorectal fascia vary- sensitivity of 54%, specificity of 87%, PPV of
ing between 60% and 88% and specificity 44%, and NPV of 91% for predicting invasion of
between 73% and 100% [11]. the MRF [16]. A restaging MRI can therefore
70 R.G.H. Beets-Tan

Fig. 8.2 (a, b) Axial and coronal T2 weighted MRI of a patient with rectal cancer. The mesorectal fascia (black arrows)
can be clearly delineated, enveloping the mesorectum

Fig. 8.3 (a) Axial T2 weighted MRI of a patient with 6 weeks after completion of chemoradiotherapy. The tumor
locally advanced rectal cancer, before chemoradiotherapy. (asterisk) has been downsized and has turned fibrotic but the
The tumor (asterisk) is invading the mesorectal fascia ante- fibrosis is still invading the anterior mesorectal fascia (white
riorly (black arrows). Note a suspected node close to the arrow). In this patient one must be reluctant to predict a free
mesorectal fascia (black arrowhead). (b) Axial T2 weighted anterior margin, because the fibrosis may still contain tumor
MRI of the same patient with locally advanced rectal cancer, and the mesorectal fascia may still be involved
8 How Can We Identify Mesorectal Fascia Involvement? 71

reliably identify those tumors that have regressed 5. Taylor A, Slater A, Mapstone N et al (2007) Staging
from the mesorectal fascia. False positivity still rectal cancer: MRI compared to MDCT. Abdom
Imaging 32(3):323–327
occurs in around 50%. These false positives occur 6. Vliegen R, Dresen R, Beets G et al (2008) The accu-
in tumors that show fibrosis after chemoradiation racy of multi-detector row CT for the assessment of
and where the fibrosis has not retracted from the tumor invasion of the mesorectal fascia in primary
mesorectal fascia. In 50%, the fibrosis may still rectal cancer. Abdom Imaging 33(5):604–610
7. Maizlin ZV, Brown JA, So G, Brown C, Phang TP,
contain tumor (Fig. 8.3b). In order not to under- Walker ML, Kirby JM, Vora P, Tiwari P (2010) Can
treat, it is better to err on the safe side and overes- CT replace MRI in preoperative assessment of the cir-
timate an involved margin. cumferential resection margin in rectal cancer? Dis
Colon Rectum 53(3):308–314
8. Beets-Tan RG, Beets GL, Vliegen RF et al (2001)
Conclusion Accuracy of magnetic resonance imaging in predic-
Rectal cancer involvement of the mesorectal tion of tumour-free resection margin in rectal cancer
fascia can be well anticipated by imaging. MRI surgery. Lancet 357(9255):497–504
is the most reliable technique in all rectal can- 9. Blomqvist L, Machado M, Rubio C et al (2000) Rectal
tumour staging: MR imaging using pelvic phased-
cers, while modern multidetector CT could be array and endorectal coils vs endoscopic ultrasonog-
an alternative mainly in mid and high rectal raphy. Eur Radiol 10(4):653–660
tumors. In low tumors, CT cannot replace MRI. 10. Peschaud F, Cuenod CA, Benoist S et al (2005)
Therefore, MRI remains the preferred (re-) Accuracy of magnetic resonance imaging in rectal
cancer depends on location of the tumor. Dis Colon
staging method in rectal cancer. Although EUS Rectum 48(8):1603–1609
is accurate in assessing tumor ingrowth into the 11. Lahaye MJ, Engelen SM, Nelemans PJ et al (2005)
vagina or prostate, it is unable to identify tumor Imaging for predicting the risk factors – the circum-
involvement in the mesorectal fascia. ferential resection margin and nodal disease – of local
recurrence in rectal cancer: a meta-analysis. Semin
Ultrasound CT MR 26(4):259–268
12. MERCURY Study Group (2006) Diagnostic accuracy
of preoperative magnetic resonance imaging in pre-
dicting curative resection of rectal cancer, prospective
References observational study. BMJ 333(7572):779
13. Beets-Tan RG, Lettinga T, Beets GL (2005) Pre-
1. Nagtegaal ID, Quirke P (2008) What is the role for the operative imaging of rectal cancer and its impact on
circumferential margin in the modern treatment of surgical performance and treatment outcome. Eur J
rectal cancer? J Clin Oncol 26(2):303–312 Surg Oncol 31(6):681–688
2. Bissett IP, Fernando CC, Hough DM et al (2001) 14. Burton S, Brown G, Daniels IR et al (2006) MRI
Identification of the fascia propria by magnetic reso- directed multidisciplinary team preoperative treat-
nance imaging and its relevance to preoperative ment strategy: the way to eliminate positive circum-
assessment of rectal cancer. Dis Colon Rectum ferential margins? Br J Cancer 94(3):351–357
44(2):259–265 15. Vliegen RF, Beets GL, Lammering G et al (2008)
3. Peeters KC, Marijnen CA, Nagtegaal ID et al (2007) Mesorectal fascia invasion after neoadjuvant chemo-
The TME trial after a median follow up of 6 years: therapy and radiation therapy for locally advanced
increased local control but no survival benefit in irra- rectal cancer: accuracy of MR imaging for prediction.
diated patients with resectable rectal carcinoma. Ann Radiology 246(2):454–462
Surg 246(5):693–701 16. Kulkarni T, Gollins S, Maw A et al (2008) Magnetic
4. Wolberink SV, Beets-Tan RG, de Haas-Kock DF et al resonance imaging in rectal cancer downstaged
(2009) Multislice CT as a primary screening tool for using neoadjuvant chemoradiation: accuracy of pre-
the prediction of an involved mesorectal fascia and diction of tumour stage and circumferential resec-
distant metastases in primary rectal cancer: a multi- tion margin status. Colorectal Dis 10(5):479–489,
center study. Dis Colon Rectum 52(5):928–934 Epub 2008 Mar 3
How Can We Identify Nodal
Involvement? 9
Regina G.H. Beets-Tan

Contents 9.1 Introduction


9.1 Introduction ................................................... 73
With the shift from postoperative to preoperative
9.2 How Accurate Is Imaging
for Identifying Nodal Metastases? ............... 73
(chemo)radiotherapy (CRT) for rectal cancer
patients, tumor risk profile assessment, previ-
9.3 Has Modern Imaging Improved Its
ously based on histology of the resection speci-
Performance for Detection of Nodal
Metastases? .................................................... 74 men, is done with preoperative imaging. Imaging
provides information on the T-stage, N-stage, and
9.4 Are There New Developments
in Imaging Technology involvement of the mesorectal fascia. Nodal dis-
That May Improve Nodal Staging? ............. 74 ease is one of the most important risk factors both
for local and distant recurrence and is generally
9.5 Can Restaging MRI After
Preoperative Chemoradiotherapy considered an indication for neoadjuvant therapy.
Identify Patients with Sterilized This chapter will discuss whether we can accu-
Nodes (ypN0)? ............................................... 75 rately identify nodal involvement in patients with
References ................................................................. 75 rectal cancer using imaging methods.

9.2 How Accurate Is Imaging


for Identifying Nodal
Metastases?

For a long time, nodal staging was performed


with endorectal ultrasonography (EUS) and com-
puted tomography (CT). Nodal staging was
mainly based on nodal size measurements.
A node that was larger than 8 mm on imaging
was often considered as malignant. For a part this
is correct because in rectal cancer, lymph nodes
with a diameter of 9 mm or larger are invariably
malignant. For the majority of the cases, this is
R.G.H. Beets-Tan incorrect because around 60% of the metastatic
Department of Radiology,
rectal cancer nodes are smaller than 6 mm [1].
Maastricht University Medical Centre,
5800, Maastricht 6202 AZ, The Netherlands The pooled sensitivity and specificity of EUS
e-mail: r.beets.tan@mumc.nl in a meta-analysis based on 35 studies was around

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 73


DOI 10.1007/978-3-642-25005-7_9, © Springer-Verlag Berlin Heidelberg 2012
74 R.G.H. Beets-Tan

75% [2]. A comparative meta-analysis of CT, These morphological features are well visu-
EUS, and MRI showed that the receiver operating alized in nodes larger than 6 mm, but a reliable
characteristic (ROC) curves for all three modali- delineation of the nodal feature can be difficult
ties were only moderate [3]. The sensitivity of if not impossible in the smaller nodes. The prac-
EUS, CT, and first-generation MRI for primary tical difficulties in nodal staging with the stan-
nodal staging in rectal cancer varies between 55% dard imaging methods are illustrated by a
and 70% and specificity between 75% and 80%. multicenter report in which T3N0 tumors, staged
with ERUS or MRI, were found to be node posi-
tive at histology in 22%, despite preoperative
9.3 Has Modern Imaging Improved chemoradiation [6].
Its Performance for Detection
of Nodal Metastases?
9.4 Are There New Developments
In the comparative meta-analysis, EUS performed in Imaging Technology That
slightly better than CT or MRI, most likely May Improve Nodal Staging?
because of the use of criteria for nodal metastases
other than size alone: round shape, border irregu- MR techniques and sequences are continuously
larity, and heterogeneous texture. improving, and with modern and more powerful
Recently, modern MRI technology was intro- machines, new sequences, and lymph node–
duced generating very high-resolution images. specific MR contrast agents, the accuracy is likely
Similar morphological criteria as with EUS (shape, to improve. An example of a MR contrast agent
border irregularity, and heterogeneous signal) can proven highly accurate for nodal staging both in
now also be evaluated with MRI and show improved urological and rectal cancer is ultrasmall super-
accuracy over size alone (Fig. 9.1) [4, 5]. paramagnetic particles of iron oxide (USPIO)
[7]. Unfortunately, USPIO is not approved by the
Food and Drug Administration (FDA) and there-
fore not marketed.
A new functional MRI technique, promising
in oncology imaging, is diffusion-weighted MR
imaging. A restricted diffusion of the protons in a
tissue is associated with a tumoral tissue. This
restricted diffusion can be translated into a high
signal on MRI. It can also be quantified as a
coefficient, the apparent diffusion coefficient
(ADC). Visual perception of the MR signal of
nodes cannot discriminate between benign and
malignant nodes, because both show a high sig-
nal. The ADC of malignant nodes is generally
slightly lower than that of benign nodes, but with
an AUC of 0.66, it is very difficult to find the
right cutoff value for malignant nodes [8].
Fig. 9.1 Axial T2-weighted MR image of a rectal cancer Thus, at the time of writing, despite the huge
patient. Two nodes are seen in the mesorectum, large and
variety of new imaging technology, the problem
small. Both nodes show morphological features strongly
indicative for malignant nodes (round, irregular border, of nodal staging in patients with rectal cancer
and heterogenous texture) remains.
9 How Can We Identify Nodal Involvement? 75

9.5 Can Restaging MRI take into account the prevalence of nodal
After Preoperative metastases according to the T-stage. If the
Chemoradiotherapy Identify patient’s MRI only shows small nodes, yet
Patients with Sterilized Nodes these nodes show obvious features strongly
(ypN0)? indicative for malignancy (round, irregular
border, and heterogenous texture), these
For patients treated with neoadjuvant CRT, there patients can be considered N+. In all other
is now a trend toward minimally invasive, organ- circumstances, the nodal status on imaging
saving treatment, when the tumor and nodes have remains uncertain.
responded well to CRT. Organ-saving treatment
is only an option when the true node-negative
patients can be accurately selected.
A restaging MRI, performed 6–8 weeks after References
completion of chemoradiotherapy, has proven
1. Wang C, Zhou Z, Wang Z et al (2005) Patterns of neo-
valuable for evaluation of tumor response and plastic foci and lymph node micrometastasis within the
nodal response. mesorectum. Langenbecks Arch Surg 390(4):312–318
In contrast to primary staging, a restaging 2. Puli SR, Reddy JB, Bechtold ML et al (2009) Accuracy
MRI shows a high NPV of around 95% for the of endoscopic ultrasound to diagnose nodal invasion by
rectal cancers: a meta-analysis and systematic review.
detection of nodal disease. In other words, the Ann Surg Oncol 16(5):1255–1265
ypN0 patients can be identified with post-CRT 3. Lahaye MJ, Engelen SM, Nelemans PJ et al (2005)
MRI [7, 9]. Due to CRT, the number of small Imaging for predicting the risk factors – the circumfer-
nodes decreases and so does the number of inter- ential resection margin and nodal disease – of local
recurrence in rectal cancer: a meta-analysis. Semin
pretation errors. After CRT, the prevalence of Ultrasound CT MR 26(4):259–268
node-positive patients is lower. These may 4. Brown G, Richards CJ, Bourne MW et al (2003)
explain why in a restaging setting size criteria Morphologic predictors of lymph node status in rectal
seem to work. cancer with use of high-spatial-resolution MR imaging
with histopathologic comparison. Radiology 227(2):
371–377
Conclusion and Recommendations 5. Kim JH, Beets GL, Kim MJ et al (2004) High-resolution
So far, clinicians have been reluctant to make MR imaging for nodal staging in rectal cancer: are
decisions based on the nodal staging by there any criteria in addition to the size? Eur J Radiol
52(1):78–83
imaging because of its insufficient accuracy. 6. Guillem JG, Diaz-Gonzalez JA, Minsky BD et al
A reliable preoperative staging of nodes (2008) cT3N0 rectal cancer: potential overtreatment
could however benefit patients with rectal with preoperative chemoradiotherapy is warranted. J
cancer. Clin Oncol 26(3):368–373
7. Lahaye MJ, Beets GL, Engelen SM et al (2009)
The most practical strategy to work with in Locally advanced rectal cancer: MR imaging for
daily practice is the following, while always restaging after neoadjuvant radiation therapy with con-
keeping in mind the limitations of imaging: comitant chemotherapy. Part II. What are the criteria
If on rectal cancer MRI, no nodes are visi- to predict involved lymph nodes? Radiology 252(1):
81–91
ble, the patient is considered N0. 8. Lambregts DM, Maas M, Riedl RG et al (2011) Value
Patients with nodes ³8 mm that show two of ADC measurements for nodal staging after chemo-
or more of the morphological features (round, radiation in locally advanced rectal cancer-a per lesion
irregular border, and heterogenous texture) are validation study. Eur Radiol 21(2):265–273
9. Barbaro B, Fiorucci C, Tebala C et al (2009) Locally
considered N+. advanced rectal cancer: MR imaging in prediction of
For patients who have nodes £7 mm, the response after preoperative chemotherapy and radia-
nodal status is less certain and one should tion therapy. Radiology 250(3):730–739
How Can We Identify Pathologic
Complete Responders After 10
Radiochemotherapy?

Brunella Barbaro and Lucia Leccisotti

Contents 10.1 Introduction


10.1 Introduction ................................................ 77
Complete tumor regression (pCR) may develop
10.2 Magnetic Resonance Imaging (MRI) ....... 79
after neoadjuvant chemoradiation therapy (CRT)
10.2.1 Morphologic Assessment of MRI ............. 79 for rectal cancer, and it is essential to select accu-
10.2.2 Diffusion-Weighted Magnetic Resonance
rately true complete responders in the manage-
Imaging (DWI)............................................. 79
ment of these patients [1]. Some authors proposed
10.3 Perfusion Computed Tomography that definition of a complete clinical response
(pCT) and Dynamic Contrast-Enhanced
MR Imaging (DCE-MRI) .......................... 87 (CR) should be based on very strict clinical and
endoscopic criteria. The finding of any residual
10.4 Positron Emission Tomography-
superficial ulceration, irregularity, or nodule
Computed Tomography (PET-CT) ........... 88
10.4.1 Pretreatment FDG PET-CT .......................... 89 should be considered residual tumor, and con-
10.4.2 Early Response Evaluation by versely, complete clinical responders should har-
FDG PET-CT ............................................... 89 bor no more than whitening of the mucosa,
10.4.3 Presurgical FDG PET-CT ............................ 89
telangiectasia with mucosal integrity. Definition
References ................................................................. 92 of a CR should be performed using digital exami-
nation, endoscopy, and biopsy, but these methods
are not infallible.
Response evaluation with diagnostic imaging
has evolved over the past 25 years, and methods
for assessing response following oncologic thera-
pies continue to evolve. In 2000, the Response
Evaluation Criteria in Solid Tumors (RECIST)
guidelines were introduced by the EORTC, the
B. Barbaro (*) US National Cancer Institute, and others. These
Department of Bioimaging and Radiological Sciences
investigators published four categories of
Catholic University, School of Medicine
Via della Mendola, 82, 00135 Rome, responses (complete response [CR], partial
Italy response [PR], stable disease [SD], and progres-
e-mail: bbarbaro@rm.unicatt.it sive disease PD). A CR is defined as the disap-
L. Leccisotti pearance of all target lesions, =/>30% decrease of
Department of Bioimaging and Radiological Sciences, the maximum diameter as PR. PD is defined as a
Catholic University, School of Medicine,
20% increase in of the maximum diameter of the
Largo A. Gemelli, 1, 00168 Rome,
Italy target lesions or the appearance of any new
e-mail: lucialeccisotti@yahoo.it lesions, and SD is defined as changes that do not

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 77


DOI 10.1007/978-3-642-25005-7_10, © Springer-Verlag Berlin Heidelberg 2012
78 B. Barbaro and L. Leccisotti

a b

c d

Fig. 10.1 Rectal cancer in a pCR 59-year-old woman. tion after CRT. The lesion volumes were displayed
(a, b) HR oblique T2-weighted images of tumor before automatically in three-dimensional format and were cal-
(a) and at the end (b) of treatment show the tumor contour culated by summing each of the cross-sectional volumes
by manual tracing with a cursor. (c) Pre- and (d) posttreat- (multiplying cross-sectional area by section thickness) for
ment 3D reconstructions define 90% tumor volume reduc- the entire lesion. Courtesy of [5]

meet the previous three criteria. Volumetric analysis CRT, yielding a sensitivity of 83% and specificity
provides reliable and more reproducible data than of 86% for prediction of pCR. Kang et al. [3]
conventional measurements which may be reported that more than 75% of the tumor volume
affected by the anatomy of the viscera, including reduction ratios after CRT were significantly
the rectum which is a hollow viscus with irregular associated with a high pCR rate in 84 patients
morphology, and the intrinsic irregularity of tumor with rectal cancer. To date, the correlation between
shape (Fig. 10.1). Tumor volume measurement is rectal tumor histopathologic downstaging after
widely used for antitumor therapy response. CRT and tumor volume reduction remains contro-
Recently, Lambrecht et al. [2] demonstrated in 21 versial, especially concerning the hypothesis that
patients with locally advanced rectal cancer that percentage volume reduction may indicate pCR.
ROC analysis showed an AUC of 86% at an opti- The metric for positive response is based only on
mal cutoff value of 77% volume reduction after anatomical and morphological changes which are
10 How Can We Identify Pathologic Complete Responders After Radiochemotherapy? 79

temporally downstream manifestations of under- fibrous tissue present after treatment causes thick-
lying pathophysiological changes which may ening of the rectal wall in most cases; thus, MR
occur earlier. In early 2009, RECIST was updated, imaging cannot readily differentiate yT0 or yT1
as RECIST 1.1, and a major development was the stage tumors from yT2 tumors because it is not
incorporation of fluorodeoxyglucose positron possible to visualize individual rectal wall layers,
emission tomography (FDG-PET) in the guide- and thus, thickened hypointense rectal wall is
lines, albeit in the limited role of assessing pro- classified as yT0–2 lesions at morphologic restag-
gressive disease. This is exciting in the sense that ing (Fig. 10.3). Some treated tumors develop a
this is the first time the working group has incor- “colloid” response with mucin production that
porated a nonanatomical imaging modality in the results in a very high signal intensity on a
assessment of tumor response, a trend that will T2-weighted image, which is potentially confus-
almost certainly continue in the future. ing. When MR imaging is used in posttreatment
More advanced and specific imaging methods restaging, an increase in tumor signal intensity
are needed to characterize the underlying when compared to pretreatment MR findings is
pathophysiological changes induced by specific generally considered indicative of a mucinous
targeting agents. Such methods may be consider- response. This is not to be confused with the highly
ably more likely to offer earlier—and more hyperintense areas seen in pretreatment MR images
specific—information on response to treatment which instead correspond to pretreatment muci-
when compared to changes in longest tumor nous tumors [5]. Irradiated tumors may show a
dimensions. This may be especially true in the mucin component larger than 50% of the whole
case of neoadjuvant treatment of rectal cancer. tumor, reaching 80–90%; thus, it is possible to rec-
pCR is highly predictive of survival in rectal can- ognize in MR images posttreatment mucin differ-
cer and offers an earlier endpoint for preliminary entiation as lake of high increase in the signal
validation of image-based response criteria. intensity (Fig. 10.4). However, MR restaging is
unable to differentiate mucinous response or
fibroinflammatory tissue from residual tumor as
10.2 Magnetic Resonance Imaging persistent intermediate signal intensity. In conclu-
(MRI) sion on standard MRI, a normalized rectal wall
without any detectable wall thickening is consid-
10.2.1 Morphologic Assessment of MRI ered a definite criterion for a CR (Fig. 10.2). A solid
residual mass with intermediate signal intensity on
MR imaging is considered the most accurate tool T2-weighted MRI is considered a definite criterion
for primary staging of the tumor extent; however, it for residual tumor. Hypointense signal intensity
is not accurate in rectal cancer restaging after CRT, that changes indicated fibrosis remains undeter-
especially when there is a fibrotic thickening of the mined; thus, purely anatomic MR imaging is
rectal wall, in distinguishing between ypT0, ypT1, insufficient to reliably assess the true complete
ypT2, or ypT3 tumors [4]. The untreated tumor on responders, and there is considerable enthusiasm
T2-weighted images has intermediate signal inten- for employing functional methods for selection of
sity between the high signal intensity of the fat tis- these patients with high positive predictive values
sue and the low signal intensity of the muscular and consequently to avoid put them at risk for
layer and invades mesorectal fat with nodular pat- undertreatment.
tern growth. After CRT, complete disappearance of
the tumor is classified as CR (Fig. 10.2). Most
tumors develop fibrosis, leading to reduction of T2 10.2.2 Diffusion-Weighted Magnetic
signal and decrease in tumor size. A decrease in Resonance Imaging (DWI)
signal intensity when compared with the pretreat-
ment examination represents response with fibrosis, DW-MR imaging may be an appropriate tool to
that is, replacement of neoplasm by fibrosis. The monitor the effects of treatment in vivo. Advantages
80 B. Barbaro and L. Leccisotti

a b c

d e f

Fig. 10.2 Rectal cancer in a pCR 47-year-old man. (a) intensity at DW image (arrow in b) with b factor 1,000
HR oblique T2-weighted image of tumor before CRT (arrow in d) and as avid tracer FDG uptake in pretreat-
shows a tumor on the right lateral rectal wall protruding ment PET-CT (c). The tumor disappears in posttreatment
into the rectal lumen as moderate hyperintense mass, oblique (arrow in e) HR T2-weighted (arrow in c) MR
invading the rectal wall. It is difficult to determine whether image (d) and in axial DW image (e). No FDG uptake in
the perirectal fat (arrow) is infiltrated or spared; thus, MR the posttreatment PET-CT (f). Note concordance in mor-
pretreatment stage was borderline tumor as stage T2 or phologic MR image (d), DW image (e), and FDG-PET
initial stage T3. The tumor appears as area high in signal CT image (f) in this pCR

Fig. 10.3 A 58-year-old man with pCR low rectal can- mesorectal fat, through the pelvic floor, indicating potential
cer. (a) Axial DW MRIs, with a b value of 1,000 s/mm2 involvement of the left levator ani muscle (white arrow).
showed the tumor as areas of high signal intensity; (b) After treatment, before surgery, the tumor showed
the tumor as low signal intensity areas (opposite to DW shrinkage and the rectal wall was hypointense and thick-
images) was drawn manually in the ADC maps based on ened. The isointense to muscle intensity persisted on the
the corresponding (c) axial T2-weighted MRIs before, left site (white arrow). The patient underwent transanal
during, and after treatment. In the DW-MRIs, the refer- endoscopic microsurgery. In the bottom left corner, pho-
ence signal was the prostate (P), before and during treat- tomicrograph (original magnification, ×20; H&E stain)
ment (black arrows in a); we found no residual after surgery shows marked fibrosis (black arrows) in the
hyperintense area in the corresponding tumor at the end rectal wall. No viable cells were detected in the rectal
of treatment as possible CR (white * in a). In the corre- wall. Final evaluation was ypT0, TRG 1/5. In this patient,
sponding areas on the ADC maps, the ADC value the fibrous tissue represents the site of previous tumor
increased in the tumor from 1 × 10−3 to 1.3 × 10−3 mm2/s and this fibrosis caused thickening of rectal wall; thus,
(30%) during treatment, which indicated a possible early morphologic MR imaging cannot really differentiate T0
response. At the end of treatment, the ADC value of the to T2 stage because visualization of individual rectal
whole tumor was 1.2 × 10−3 mm2/s. (d) Coronal wall layers is not possible, but areas of fibrosis typically
T2-weighted MRIs before and after treatment. Before have a low cellular density, which results in low signal
treatment, the tumor was detected as an intermediate intensity on high b value (b1000); thus, visual diffusion
intensity signal that extended into the surrounding images could hypothesize CR in this patient
10 How Can We Identify Pathologic Complete Responders After Radiochemotherapy? 81

d
82 B. Barbaro and L. Leccisotti

a b

c d

Fig. 10.4 62-year-old man with locally advanced lower (g) shows region of interest (ROI) which is drawn manu-
rectal cancer. (a, b) Pretreatment coronal (a) and HR ally using the corresponding oblique HR T2-weighted
oblique (b) T2-weighted MR images show hyperintense image (f) to identify the residual tumor regions. ADC
mass on the lateral low rectal wall with infiltration of the value increases to 1.7 × 10−3 mm2/s reaching
levator ani muscle corresponding to T4 stage (white arrow 1.9 × 10−3 mm2/s in the area of mucin degeneration seen
in a). (c, d) The pretreatment ADC map (c) corresponding as red area in color ADC map (white * in h), as the endolu-
to HR oblique T2-weighted MR image (b) shows a calcu- minal sonography gel (black *). (i) Photomicrograph
lated ADC tumor of 1 × 10−³ mm2/s which appears preva- (original magnification, × 20; [H-E] stain) shows no tumor
lent green in color map (d). (e, f) Coronal (e) and HR in the rectal wall but only inactive mucin lakes (black
oblique (f) T2-weighted MR images obtained 6 weeks arrows) in persistent areas of high signal intensity: ypT0
after the end of CRT show the residual tumor in the rectal TRG 1/5. Note the heterogeneous response in this patient
wall containing heterogeneous high signal intensity in with a substantial posttreatment ADC value increase
keeping with mucin production as possible response related to mucinous response. Courtesy of [5]
(black arrows in e and f). T0–T2 stage at morphologic
presurgical MR imaging. (g, h) Posttreatment ADC map
10 How Can We Identify Pathologic Complete Responders After Radiochemotherapy? 83

f
e

g h

Fig. 10.4 (continued)

of this technique are that it is completely noninva- only a minimal extension of examination time.
sive, does not require exposure to ionizing radia- However, the limited spatial resolution and the
tion or injection of contrast materials, and does not relatively poor signal-to-noise ratio on high-b-value
cause patient discomfort. Another advantage of DW-MR imaging should be considered a limita-
DW-MR imaging is that it can be easily added to tion, and echo-planar imaging sequences, more-
an MR examination protocol because it requires over, have a propensity to yield distorted images.
84 B. Barbaro and L. Leccisotti

DW imaging derives its image contrast from dif- With increasing tumor cellularity and archi-
ferences in the motion of water molecules between tectural distortion, any increase in the tortuosity
tissues. of the extracellular space will additionally con-
tribute to decreased ADC values. It would, there-
10.2.2.1 Qualitative Assessment of DWI fore, be expected that ADC values would correlate
In tissues with normal cellularity, water protons with tumor cellularity and grade. Inverse correla-
can diffuse relatively freely, which results in a tions between MRI-determined ADC values and
loss of signal on DWI. Conversely, in tissues with cellularity and tumor grade have been noted in
increased cellularity (tumor), the diffusion of xenograft and human tumors. The measured
water is restricted, resulting in remaining high ADC is, therefore, inversely related to the cellu-
signal on DWI. Visual assessment of the relative larity of tumors.
tissue signal attenuation at DWI is being applied
for tumor detection, tumor characterization, and 10.2.2.3 Pretreatment ADC Value
the evaluation of treatment response in patients One of the most intriguing findings associated
with cancer. In many reports, DWI has shown with the use of DWI in cancer patients has been
promise for identification of malignant tumors, that ADC measurements appear to be able to pre-
and recent studies on rectal cancer have indicated dict the response of tumor to chemotherapy and
that DWI also may be useful. High-b-value radiation treatment. Many of the clinical studies
DW-MR imaging has shown sufficient diagnostic evaluating DWI for assessing treatment response
ability for detecting colorectal cancer as reflected have been performed in patients with rectal can-
in its high sensitivity and specificity (however, cer that produced provocative results. Dzik-Jurasz
tissues such as the brain, spinal cord, salivary et al. [6] were among the first to find a strong
glands, testes, prostate, endometrium, bowel negative correlation between the mean pretreat-
mucosa, and lymphatic tissues can also be ment tumor ADC and the percentage shrinkage of
visualized). a rectal tumor after CRT. Recently, Sun et al. [7]
reported a similar observation, and Lambrecht
10.2.2.2 Quantitative Assessment of DWI et al. [8] showed that, in 22 patients, the initial
In vivo, intracellular and extracellular compart- ADC was significantly lower in patients with
ments will have their own unique water diffusion pCR compared to those without pCR after CRT.
constants, which are measured in mm2/s. DWI is This negative association is consistent with the
typically performed using at least two b values known relationship between necrosis which
(e.g., b = 0 s/mm2 and other b values from 0 to causes increases in ADC values and poor response
1,000 s/mm2). By performing DWI using differ- to treatment in cancer. Some authors [9], on the
ent b values, quantitative analysis is possible. other hand, found that the pretreatment mean
This analysis is usually performed at a push of a ADCs in patients with rectal cancer did not pre-
button on the scanner or workstation that results dict treatment response. The effects of high-grade
in the calculation of the apparent diffusion tumors on ADC values are potentially problem-
coefficient (ADC). atic when trying to assign predictive significance
By drawing regions of interests on these maps, to pretherapy diffusion MRI values. It is conceiv-
the ADCs of different tissues can be derived. able that low-grade tumors with little necrosis
Areas of restricted diffusion in highly cellular could have similar mean high ADC values to
areas show low ADC values compared with less necrotic high-grade tumors; conversely, low pre-
cellular areas that return higher ADC values. At therapy ADC value could be an indicator of tumor
this point, it is important to mention that although aggressiveness related to the high cellularity in
areas of restricted diffusion will appear to be higher high-grade tumors, particularly for tumors that
in signal intensity on the DW images, these areas are known to do not readily undergo necrosis.
will appear as low signal intensity areas (opposite Nevertheless, caution should be used when
to DW images) on the ADC map (Fig. 10.3). assigning prognostic significance to a mean tumor
10 How Can We Identify Pathologic Complete Responders After Radiochemotherapy? 85

pretherapy ADC value. Patterson et al. [10] scored values as prediction of clinical outcome for some
strength of evidence using a five point of scale tumors during treatment. Nevertheless, the body
(1–5: weak–moderate–substantial–firm–definite) of evidence suggests that percentage increase of
based on authors’ perceptions of the literature ADC value measurement is a potentially useful
used for their review and scored as moderate (2/5) tool for personalized treatment management and
the value of pretherapy ADC maps as possible should be more widely investigated in large clin-
indicator of outcome of therapy and concluded ical studies in the future (Figs. 10.3 and 10.5).
that, although acknowledging the potential role of
pre-CRT ADC as a quantitative biomarker to dis- 10.2.2.5 Presurgical Diffusion-Weighted
criminate the T-downstaged group from the non- Magnetic Resonance Imaging
downstaged group in neoadjuvant CRT for locally (DWI)
advanced rectal cancer, the role of pre-CRT ADC In 2009, the first study by Kim et al. [11] in 40
seems to be very limited, particularly when patients investigated the value of visual DWI for
focused on the prediction of pCR. predicting a pCR after CRT. Results for two inde-
pendent readers indicated that the addition of
10.2.2.4 Early Response Evaluation DWI to standard rectal MRI significantly
The sensitivity of DW-MRI to changes in tissue improved the diagnostic performance for selec-
microstructure and organization has led to tion of pCR from an AUC of 0.68 and 0.66,
increasing interest in the potential of ADC mea- respectively for standard MRI to an AUC of 0.88
surements as a biomarker for early evaluation of and 0.82 for standard MRI + DWI. Results of a
response to treatment, and these changes pre- larger, multicenter study [12] confirm these previ-
ceded significant reductions in tumor volume. ous findings and showed that the diagnostic per-
This response occurs within days of initiating formance for predicting a pCR after CRT
therapy and appears to be a consequence of cel- improved for standard MRI + DWI as compared
lular damage leading to necrosis and, thus, a uni- to standard MRI only and resulted in a substantial
versal response to therapy. Viable tumor cells reduction in the number of equivocal scores and
restrict the mobility of water, whereas necrotic an improved interobserver agreement. In this
tumor cells allow increased diffusion of water multicenter study with the addition of DWI, sen-
molecules as a result of treatment-induced tumor sitivity for identification of a pCR improved by
regression such as necrosis, decreased cellular- 16–52% for the three readers, and it resulted in
ity, and compromised cell membrane integrity. less overestimation of residual tumor. This is
In a number of clinical and preclinical studies, mainly because on the restaging MRI without
increases in tumor ADC values were noted early DWI, many interpretation difficulties were
during the course of therapy, allowing early pre- observed when the primary tumor bed had become
diction of treatment response. Although there fibrotic as a result of the radiation treatment. In
have been few clinical studies of the effects of these cases, it is difficult to differentiate small
treatment on tumor ADC in rectal cancer pub- areas of residual tumor from mere fibrosis and
lished to date, the results of the existing studies readers tend to overestimate the presence of tumor
are promising. Some authors showed that, at the (Fig. 10.3). Apparently, this is where the func-
end of the second week of CRT, significant tional information from DWI proves beneficial.
increases in tumor ADCs occurred in the down- Areas of fibrosis typically have a low cellular
staged group. More recently, Lambrecht et al. density, which results in low signal intensity on
[2] showed an optimal cutoff point of 50% for high-b-value (b1000) visual diffusion images.
the percentage of ADC change during therapy Conversely, residual tumor areas have a relatively
for prediction of pCR with a sensitivity, high cellular density and show high signal on
specificity, PPV, and NPV of 100%. Patterson DWI, which stands out within the low signal of
et al. [10] scored (1–5) strength of evidence scale the surrounding tissue/fibrosis. This is the reason
as moderate-firm (2–4/5) the increase in ADC why small areas of residual tumor are better
86 B. Barbaro and L. Leccisotti

Fig. 10.5 70-year-old-woman with T3 middle rectal can- SUVmax). At the end of treatment, the comparison between
cer. Axial DW MRIs (a), with a b value of 1,000 s/mm2 DW image (a) and FDG PET-CT image (b) reveals nei-
before, during, and after treatment compared to FDG- ther area of restricted diffusion nor uptake of FDG as pos-
PET-CT images (b) before, during, and after treatment, sible CR. (c) Pretreatment and presurgical HR oblique
show the tumor as areas of high signal intensity in pre- T2-weighted MR images show tumor shrinkage after
treatment DW image and as avid tracer FDG uptake in CRT, yellow arrows in (c) (in the bottom left corner, the
pretreatment PET-CT. At Axial DW MR image, a tumor tumor was drawn manually in the ADC maps). The stage
signal intensity reduction, yellow arrows in (a), which was ypT2 at histology, TRG 2/5. In this patient, both
corresponds to 30% ADC increase and at FDG-PET-CT DW-MRIs and FDG-PET-CT images cannot reliably dis-
image qualitative and semiquantitative metabolic response criminate residual viable tumor cells from fibrosis
in the second week, are well evident (50% decrease in microscopically

depicted on DWI. Nevertheless, interpretation DWI is >90%, indicating that the residual tumors
errors were still observed with DWI resulting in a are accurately detected and the risk for under-
suboptimal sensitivity of 52–64%. When the sig- treatment will be <10%. Although DWI allows
nal of the normal rectal wall is not fully sup- detection of even small (2–5 mm) tumor volumes,
pressed on DWI, high signal at the location of the the challenge will remain the detection of micro-
initial tumor area may erroneously be interpreted scopically small clusters of residual tumor cells,
as residual tumor, resulting in overstaging errors. which are difficult to detect—even at histology—
In addition, some imaging artifacts may occur on and are currently beyond the detection level of any
DWI, particularly around air-tissue interfaces. In available imaging modality, including DWI
this multicenter study, specificity for MRI and (Fig. 10.5).
10 How Can We Identify Pathologic Complete Responders After Radiochemotherapy? 87

In some studies, the diffusion images were not The use of whole-tumor summary statistics
only analyzed visually, but also quantitatively by (mean) has the merit of simplicity but may have
measuring the ADC of the tumors. the potential of overlooking tumor heterogeneity;
Patterson et al. [10] describes the behavior of histogram or voxel-wise analyses are supposed to
tumor after successful therapy and reported that be alternatives for evaluating tumor heterogeneity.
removal of dead and/or dying cells is followed by
tissue compaction, fibrosis, and regeneration of
native tissues. These processes can result in 10.3 Perfusion Computed
decreases in ADC from the elevated levels caused Tomography (pCT) and
by decreased cellularity (Fig. 10.3). In addition, the Dynamic Contrast-Enhanced
presence of residual active disease or repopulation MR Imaging (DCE-MRI)
by resistant cells can also decrease ADC values;
thus, these authors concluded that interpretation of Perfusion computed tomography (pCT) and
changing ADC values must, therefore, be made dynamic contrast-enhanced magnetic resonance
with caution. Preliminary results of many clinical imaging (DCE-MRI) are clinical imaging tech-
studies evaluating quantitative DWI for ADC val- niques that are increasingly applied to noninva-
ues at the end of treatment in relatively small num- sively assess the microvascular status of tumor
bers of patients with rectal cancer reported that a tissue. In clinical cancer research, regression of
reduction in ADC values at the end of CRT which tumor microvasculature is considered an impor-
they ascribed to tumor sloughing (i.e., apoptotic tant early surrogate marker for treatment response,
cell removal) and tissue compaction (i.e., fibrosis) before reductions in tumor volume become appar-
and reductions in blood flow. Conversely, some ent. To date, both pCT and DCE-MRI are increas-
authors more recently reported that the post-CRT ingly used for the prediction and evaluation of
ADCs in the pCR were significantly higher than treatment response, as indicators of tumor angio-
those in the non-pCR. Clinical application of the genesis, and for primary tumor staging. Both
cutoff ADC value of 1.30 × 10−3 mm2/s in the study pCT and DCE-MRI are imaging modalities that
of Kim et al. [9] showed a perfect negative-predictive rely on the dynamic assessment of tracer uptake
value (NPV) useful for proper selection of the non- kinetics, subsequently quantified by means of
CR group but low positive-predictive value (PPV) pharmacokinetic models. These models describe,
of 52%. In these authors’ opinion, the main reason in terms of pharmacokinetic parameters, the
for this poor PPV is the considerable overlap of wash-in and wash-out of contrast agent from the
ADC values between pCR and near-pCR and microvasculature into the surrounding tissues. A
concluded that, although ADC is a promising commonly applied pharmacokinetic two-com-
potential quantitative biomarker, it suffers from the partment model is the extended Kety model,
problem that it cannot reliably discriminate residual which consists of the (transendothelial) volume
viable tumor cells from fibrosis microscopically transfer constant Ktrans, the fractional volume of
(Fig. 10.5). the extravascular–extracellular space (EES), ve,
Careful consideration should be given to the and the fractional blood plasma volume, vp.
role of ADC measurements in the determination Clinically, Ktrans is the most valuable pharmacoki-
of CR in patients with mucinous tumors because netic parameter, which describes the transfer rate
patients in this subset supposedly have higher of the contrast agent from the plasma space to the
ADC values than do patients with ordinary ade- EES and represents a combination of microvas-
nocarcinomas. Furthermore, it is still a challenge cular blood flow, vessel wall permeability, and
to predict disease activity in patients with muci- vessel density (i.e., permeability surface area
nous tumors on conventional MR images, as it is product). The usefulness of DCE-MR and pCT in
difficult to differentiate residual tumors from characterizing rectal tumors and their response to
inactive mucin pools also in mucinous response treatment has been demonstrated in several clini-
in non-mucinous pretreatment tumor. cal and preclinical studies to evaluate tumor
88 B. Barbaro and L. Leccisotti

neoangiogenesis noninvasively in prediction semiquantitatively by measuring the standardized


tumor response. Rectal tumors with higher per- uptake value (SUV). The SUV is defined as the
meability at presentation appear to respond better ratio of the activity within the tissue (KBq/ml)
to CRT than those of lower permeability. and the decay-corrected total activity injected
Recently, Kierkels et al. [13] compared pre- (MBq) divided by the body weight (kg). The SUV
treatment scans with pCT vs. DCE-MRI in rectal is usually expressed as its maximum (SUVmax).
tumors and concluded that DCE-MRI is recom- SUVmax value is the most consistent and less
mended over pCT because of the better signal- dependent on region of interest dimension.
to-noise characteristics, stronger contrast uptake, However, the measurement of the SUVmean most
more complete evaluation of the microcirculatory likely better reflects the behavior of the entire
environment of the tumor, larger tumor coverage, tumor mass which contains viable cells mixed
and the absence of ionizing radiation. with necrosis or fibrosis. FDG PET-CT has sev-
As Patterson et al. [10] supposed, fibrosis is eral recognized applications in rectal cancer
associated with reduced enhancement on DCE- including preoperative evaluation of apparently
MRI, whereas residual active disease causes limited metastatic disease, disease recurrence
marked enhancement on DCE-MRI; thus, as detection, clarification of equivocal lesions at ini-
these authors hypothesize, the combined use of tial staging, and investigation of unexplained ris-
anatomic, DWI, and DCE-MR imaging should ing tumor markers. Recently, the applications of
provide more comprehensive and accurate assess- PET-CT in rectal cancer are emerging in therapy
ment of rectal cancer response to therapy, but to response assessment. A tumor that has significant
our knowledge, no data are in the literature con- reduction in size may still contain viable tumor
cerning DCE-MRI or pCT in evaluating pCR cells, and, conversely, a residual mass may repre-
after CRT in patients with rectal cancer. sent posttherapy changes such as edema, fibrosis,
or necrosis with no viable tumoral cells. Since
metabolic changes precede morphological changes,
10.4 Positron Emission functional imaging using PET should be more
Tomography-Computed sensitive and specific for therapy response assess-
Tomography (PET-CT) ment. A favorable response is often associated to
SUV reduction, in terms of absolute SUV differ-
Integrated PET-CT is a hybrid noninvasive imag- ence (DSUV: SUV1 − SUV2) or response index
ing modality which has recently become an inte- [RI: (SUV1 − SUV2)/SUV1]. A further meta-
gral part of multidisciplinary cancer care. For bolic response parameter is the total lesion glyco-
most oncological indications, PET-CT is per- lysis (TLG) which incorporates the differences in
formed using 2-[18F]-fluoro-2-deoxy-d-glucose tumor activity between baseline and posttherapy
(FDG), a glucose analogue that provides func- scans but also takes into account changes in tumor
tional information on tumor metabolism by dem- size [(SUV2*PETvol2)/(SUV1*PETvol1)*100].
onstrating augmented glycolysis in tumoral cells Regardless of metabolic response evaluation, the
compared to normal cells. However, FDG uptake standardization of the scanning parameters, the
is not tumor specific as benign processes such as time points at which responses are measured
infection and inflammation show increased glu- (during CRT vs. presurgical evaluation), the inter-
cose utilization. Patient preparation includes a pretative criteria (visual vs. semiquantitative
6-h fast with an optimal blood glucose analysis, tumor aggressiveness [SUVmax or
level £ 180 mg/dl. Usually, a patient is injected SUVmean] vs. tumor burden [metabolic volume]),
with 259–370 MBq of FDG, and after a and the reference criteria (tumor regression grade
60 ± 10-min uptake period, a “low-dose” CT fol- [TRG] which is based on the ratio of fibrosis to
lowed by PET acquisition (2–3 min per bed posi- residual cancer vs. post-treatment pathological T
tion) is obtained from the skull base to upper stage [ypT]) are essential to minimize interob-
thigh. PET data can be analyzed qualitatively and server variability.
10 How Can We Identify Pathologic Complete Responders After Radiochemotherapy? 89

10.4.1 Pretreatment FDG PET-CT ment response was correlated with the pathologi-
cal response by the evaluation of the TRG and
FDG PET-CT in initial staging of rectal cancer is ypT stage of the resected specimens. Based on
reserved for high-risk patients (locally advanced TRG evaluation, 13 patients were classified as
disease, equivocal findings on morphological pathological responders (TRG1–2), whereas 17
imaging, and high CEA levels). It is well known patients were classified as pathological nonre-
that 95–100% of known primary rectal cancers sponders (TRG3–5). The RI calculated after the
are visible on FDG PET-CT. False-negative first 2 weeks of CRT provided the best predictor
results may be encountered with mucinous tumors of pathological treatment response, reaching
and very small primary tumor. The limited spatial AUCs of 0.87 and 0.84 for the TRG and the ypT
resolution of this technique (4–6 mm) does not stage, respectively. A cutoff value of 43% for the
allow to define the relationship of the primary reduction of SUVmax resulted in a sensitivity of
lesion to the mesorectal fascia or to accurately 77% and a specificity of 93%. Recently, the same
evaluate local nodal disease (sensitivity 30–35%; authors validated their preliminary results on a
specificity ~85%). Pretreatment SUVmax of the larger patient population (n = 51), confirming the
primary tumor does not seem to differentiate pCR potential of PET-based response prediction early
or near-pCR (TRG1–2) from pathological non- during CRT treatment [16]. Applying a cutoff
responders (TRG3–5). However, pretreatment value of 48%, SUVmax reduction to differentiate
FDG PET-CT is essential as baseline scan to pathological responders (TRG1–2) from nonre-
assess metabolic changes of the primary tumor sponders (TRG3–5) resulted in a specificity and
during and after neoadjuvant treatment. sensitivity of, respectively, 93% and 83%, with
only one of the pathological nonresponders being
false-positively predicted as pathological respond-
10.4.2 Early Response Evaluation ing. However, data deriving from tailored therapy
by FDG PET-CT based on metabolic response during neoadjuvant
CRT using alternative dosing, fractionation, or
In locally advanced rectal cancer, there is mount- agents are still lacking.
ing evidence of the value of FDG PET-CT in the
early prediction of the pathological response after
neoadjuvant treatment. SUV reduction as early as 10.4.3 Presurgical FDG PET-CT
12 days after preoperative CRT has been shown
to predict TRG in several published studies. Presurgical PET-CT images usually show a
Cascini et al. [14] correlated the changes in tumor significant lower FDG uptake in the primary
SUVmax 12 days after starting preoperative CRT tumor of patients with pathological confirmed
and 6–8 weeks after the end of preoperative CRT response compared to nonresponders. A mild and
with the pathological results in 33 patients. The diffuse FDG uptake, similar to background activ-
early median decrease of tumor SUV significantly ity, is typical of patients with pCR or near-com-
differed between TRG1–2 (−62%; range: −44% plete pathological response (TRG1–2) (Figs. 10.2
to −100%) and TRG3–5 (−22%; range: −2% to and 10.5). Most of published studies correlated
−48%) (p < 0.0001). Responders were correctly the metabolic tumor response to TRG score and
identified by decreases in the SUVmean (with a more recently to ypT stage. The reported optimal
decrease of ³52% the accuracy was 100%) and cutoff values of the mean reduction in SUV for
SUVmax (with a decrease ³42% the accuracy was identifying a response to preoperative CRT are
94%). Janssen MH et al. [15] studied 30 patients heterogeneous, ranging from 40% to 70%, and
with locally advanced rectal cancer with sequen- mainly rely on post-hoc analysis. Moreover, the
tial FDG PET-CT imaging at four time points: predictive values of FDG PET-CT response
prior to therapy, at days 8 and 15 during CRT, and (NPV) range between 85% and 95%, while pre-
shortly before surgery. Tumor metabolic treat- dictive values of FDG PET-CT nonresponse
90 B. Barbaro and L. Leccisotti

(PPV) range from 75% to 85%. Capirci et al. [17] Moreover, the “stunning” of tumor cells second-
studied 45 patients with locally advanced rectal ary to a therapy-induced reversible decrease in
cancer at baseline and at 5–6 weeks after the glucose metabolism could mimic a response.
completion of neoadjuvant CRT. The baseline Therefore, FDG PET-CT is not accurate in dif-
SUVmax (median 16.7) was not predictive of path- ferentiating TRG1 from TRG2, in completely
ological response. The RI showed a good correla- assessing pCR or predicting tumor clearance from
tion with the TRG. A cutoff value of 66.2% the mesorectal fascia following neoadjuvant CRT
decrease in SUVmax, identified by ROC analysis, treatment. The sensitivity for assessing response
allowed differentiation of responders (8 TRG1 + 15 in regional nodes is also disappointing. Therefore,
TRG2) from nonresponders (9 TRG3 + 13 TRG4– a negative presurgical FDG PET-CT scan cannot
5) with 80% overall accuracy. Melton et al. [18] be considered as surrogate for pCR, because it is
showed that FDG PET-CT parameters are best for not possible to exclude residual microscopic dis-
assessing tumor downstaging and percentage of ease (Fig. 10.5). On the other hand, false-positive
residual tumor after neoadjuvant treatment of rec- presurgical PET images are usually related to the
tal cancer. pCR or microscopic disease (n = 7, inflammation induced by radiotherapy. Activated
33%) was associated with DSUV (AUC = 0.79, macrophages, neutrophils, fibroblasts, and granu-
p = 0.01), while tumor downstaging (n = 14, 67%) lation tissue induced by radiotherapy are known
was associated with greater DPET volume to avidly consume FDG accounting for a consid-
(AUC = 0.82, p < 0.001) and DSUV (AUC = 0.82, erable accumulation of FDG in noncancerous
p < 0.001). Recently, van Stiphout et al. [19] parts of the tumor (Fig. 10.6). Dynamic and dual
developed predictive models based on clinical time point FDG PET-CT imaging as well as dif-
and FDG PET-CT data for pCR based on a pooled ferent PET tracers (i.e., 18F-FLT) might be able to
multicenter database of 953 patients with locally distinguish malignant from inflammatory tissue.
advanced rectal cancer. The authors showed that The emerging need to accurately identify nonin-
tumor dimension in combination with RI and vasively the pCR after preoperative CRT requires
posttreatment SUVmax are the most predictive a reliable test characterized by very PPV to avoid
variable sets for pCR resulting in very good over- undertreatment of patients who still have viable
all performance AUCs of 0.86. It should be keep tumor cells. Although the sensitivity for pCR
in mind that false-negative presurgical PET with FDG PET-CT is quite high with the provided
images may be related to a significant tumor cutoff values, the specificity is actually too low
downsizing after a long course of CRT which can for clinical use. To date, for neither DSUVmax-
result in an underestimation of the FDG uptake mean nor RI, a threshold level rendering a PPV >
within the tumor due to partial volume effect. 90% has yet been determined.

Fig. 10.6 Rectal cancer in a pCR 47-year-old man. (a) (arrow) as possible CR. Note that in the alternative rectal
HR oblique T2-weighted image of tumor before treatment wall layers in posttreatment DW image, the mucosa is
shows a T3 tumor on the anterior rectal wall protruding homogeneously hyperintense, black *, and the submucosa
into the rectal lumen as moderate hyperintense mass is dark, white *, related to edema. ADC value increases to
(arrow). After treatment, before surgery, the tumor showed 1.6 × 10−3 mm2/s in the presurgical image. FDG-PET CT
shrinkage and appears as hypointense area on the anterior scans before and after treatment (c). Focal FDG uptake
site (arrow) (in the bottom left corner, the residual tumor persists in presurgical FDG-PET-CT image (47% decrease
was drawn manually in the corresponding presurgical in SUVmax at presurgical exam). Histology reveals abun-
ADC map). The rectal wall is thickened and alternative dant fibroinflammation tissue without residual viable
rectal wall layers are well evident (yellow * indicates sub- cells. The stage was ypT0, TRG 1/5. Inflammation caused
mucosa which is thickened and hyperintense to muscle errors in FDG PET-CT imaging and is a common source
intensity). Axial DW-MRIs before treatment and before of inaccuracy in tumor restaging. In this patient, the asso-
surgery (b). The tumor appears as area high in signal ciation between DW imaging and FDG PET-CT was use-
intensity in pretreatment DW image (arrow) with b factor ful for predicting response to avoid misinterpretation of
1,000 and disappears in posttreatment axial DW image high residual FDG uptake as residual tumor
10 How Can We Identify Pathologic Complete Responders After Radiochemotherapy? 91

c
92 B. Barbaro and L. Leccisotti

Conclusion preoperative chemoradiation: the impact of the vol-


ume reduction ratio on the prediction of pathologic
In this time of changing treatments, it clearly complete response. Int J Radiat Oncol Biol Phys
appears that a common standard for large het- 76(4):1018–1025
erogeneous patient groups have to be substi- 4. Barbaro B, Fiorucci C, Tebala C et al (2009) Locally
tuted by more individualized therapies. The advanced rectal cancer: MR imaging in prediction of
response after preoperative chemotherapy and radia-
radiologist’s role in the preoperative multidis- tion therapy. Radiology 250(3):730–739
ciplinary team’s decision-making process has 5. Barbaro B, Vitale R, Leccisotti L et al (2010) Restaging
become critical because the information pro- locally advanced rectal cancer with MR imaging after
vided by detailed imaging of the primary chemoradiation therapy. Radiographics 30(3):716–719
6. Dzik-Jurasz A, Domenig C, George M et al (2002)
tumor guides the team to help achieve better Diffusion MRI for prediction of response of rectal
outcomes for patients with rectal cancer. The cancer to chemoradiation. Lancet 360(9329):307–308
use of functional imaging offers an early indi- 7. Sun YS, Zhang XP, Tang L et al (2010) Locally
cation of tumor response in rectal cancer advanced rectal carcinoma treated with preoperative
chemotherapy and radiation therapy: preliminary
which may ultimately allow the development analysis of diffusion-weighted MR imaging for early
of individualized regimens. The ability to dis- detection of tumor histopathologic downstaging.
tinguish residual neoplastic tissue from scar- Radiology 254(1):170–178
ring after CRT, when restaging the patient 8. Lambrecht M (2010) The use of FDG-PET/CT and
diffusion-weighted magnetic resonance imaging for
before surgery, is the major challenge, but the response prediction before, during and after preopera-
detection of small clusters of residual tumor tive chemoradiotherapy for rectal cancer. Acta Oncol
cells remains a problem. In general, we can 49:956–963
state that the integration of functional and 9. Kim SH, Lee JY, Lee JM, Han JK, Choi BI (2011)
Apparent diffusion coefficient for evaluating tumour
molecular imaging modalities into the treat- response to neoadjuvant chemoradiation therapy for
ment paradigm, together with the numerous locally advanced rectal cancer. Eur Radiol 21(5):987–
potential molecular markers, will provide a 995. doi:10.1007/s00330-010-1989-y
bulk of information on each individual patient. 10. Patterson DM, Padhani AR, Collins DJ (2008)
Technology insight: water diffusion MRI-a potential
A good cooperation between clinics and sta- new biomarker of response to cancer therapy. Nat Clin
tistics will be necessary to mine these data and Pract Oncol 5(4):220–233
provide a predictive model enabling the pre- 11. Kim SH, Lee JM, Hong SH et al (2009) Locally
diction of the response for each individual advanced rectal cancer: added value of diffusion-
weighted MR imaging in the evaluation of tumor
patient allowing to optimize and individualize response to neoadjuvant chemo- and radiation ther-
treatment. apy. Radiology 253(1):116–125
12. Lambregts DM, Vandecaveye V, Barbaro B et al
(2011) Diffusion-weighted MRI for selection of com-
plete responders after chemoradiation for locally
advanced rectal cancer: a multicenter study. Ann Surg
References Oncol 18(8):2224–2231
13. Kierkels RG, Backes WH, Janssen MH et al (2010)
1. Maas M, Nelemans PJ, Valentini V et al (2010) Comparison between perfusion computed tomogra-
Pathologic complete response after chemoradiation phy and dynamic contrast-enhanced magnetic reso-
for rectal cancer is associated with improved long- nance imaging in rectal cancer. Int J Radiat Oncol
term outcome: a pooled analysis of 3105 patients. Biol Phys 77(2):400–408
Lancet Oncol 11(9):835–844 14. Cascini GL, Avallone A, Delrio P et al (2006) 18F-
2. Lambrecht M, Vandecaveye V, De Keyzer F et al FDG PET is an early predictor of pathologic tumor
(2012) Value of diffusion-weighted magnetic reso- response to preoperative radiochemotherapy in locally
nance imaging for prediction and early assessment of advanced rectal cancer. J Nucl Med 47:1241–1248
response to neoadjuvant radiochemotherapy in rectal 15. Janssen MH, Ollers MC, Riedl RG et al (2010)
cancer: preliminary results. Int J Radiat Oncol Biol Accurate prediction of pathological rectal tumor
Phys 82(2):863–870 response after two weeks of preoperative radiochemo-
3. Kang JH, Kim YC, Kim H et al (2010) Tumor volume therapy using (18)F-fluorodeoxyglucose-positron
changes assessed by three-dimensional magnetic emission tomography-computed tomography imag-
resonance volumetry in rectal cancer patients after ing. Int J Radiat Oncol Biol Phys 77(2):392–399
10 How Can We Identify Pathologic Complete Responders After Radiochemotherapy? 93

16. Janssen MH, Ollers MC, van Stiphout RG et al (2012) positron emission tomography and computed tomogra-
PET-based treatment response evaluation in rectal phy for assessing primary rectal cancer response to neo-
cancer: prediction and validation. Int J Radiat Oncol adjuvant therapy. J Gastrointest Surg 11:961–969
Biol Phys 82(2):871–876 19. van Stiphout RG, Lammering G, Buijsen J et al (2011)
17. Capirci C, Rampin L, Erba PA et al (2007) Sequential Development and external validation of a predictive
FDG-PET/CT reliably predicts response of locally model for pathological complete response of rectal
advanced rectal cancer to neo-adjuvant chemo-radiation cancer patients including sequential PET-CT imaging.
therapy. Eur J Nucl Med Mol Imaging 34:1583–1593 Radiother Oncol 98:126–133
18. Melton GB, Lavely WC, Jacene HA et al (2007) Efficacy
of preoperative combined 18-fluorodeoxyglucose
How Can We Identify Local Relapse?
11
Doenja M.J. Lambregts
and Regina G.H. Beets-Tan

Contents 11.3.6 What Are the Limitations in the Imaging


Detection of Local Relapse? ...................... 99
11.3.7 Are There Any New Techniques
11.1 Introduction/Clinical Background .......... 96 That May Help in the Evaluation
of Local Relapse? ....................................... 100
11.2 Detection of Local Relapse During Routine
Surveillance After Rectal Surgery .......... 96 References ............................................................... 102
11.2.1 Why Follow-up and Is It Necessary? ......... 96
11.2.2 Which Tools Should Be Used
for Surveillance and at
What Frequency?........................................ 96
11.2.3 What Is the Role of CEA During
Routine Surveillance?................................. 97
11.2.4 What Is the Role of Clinical Abbreviations
Examination (Endoscopy)
During Routine Surveillance? .................... 97
11.2.5 What Is the Role of Imaging
ASCO American Society of Clinical
During Routine Surveillance? .................... 97 Oncology
CEA Carcinoembryonic Antigen
11.3 Detection of Local Relapse in Case
of an Increased Risk for Recurrence ...... 97 CT Computed Tomography
11.3.1 Which Patients Constitute DCE Dynamic Contrast Enhanced
the ‘Increased Risk’ Group? ....................... 97 DWI Diffusion-Weighted (magnetic
11.3.2 What Is the Role
resonance) Imaging
of Clinical Examination? ............................ 98
11.3.3 What Is the Role of CT? ............................. 98 ESMO European Society of Medical
11.3.4 What Is the Role of PET? ........................... 98 Oncology
11.3.5 What Is the Role of MRI? .......................... 98 FACS (trial) Follow-up After Colorectal
Surgery
GILDA (trial) Gruppo Italiano di Lavoro per
la Diagnosi Anticipata
MRI Magnetic Resonance Imaging
PET/FDG-PET Positron Emission Tomography
/18F-fluorodeoxygenase-Positron
D.M.J. Lambregts (*) • R.G.H. Beets-Tan Emission Tomography
Department of Radiology,
RCT Randomised Controlled Trial
Maastricht University Medical Centre,
P.O. Box 5800, 6202 AZ Maastricht, The Netherlands TME Total Mesorectal Excision
e-mail: d.lambregts@mumc.nl; r.beets.tan@mumc.nl TNM Tumour Node Metastasis

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 95


DOI 10.1007/978-3-642-25005-7_11, © Springer-Verlag Berlin Heidelberg 2012
96 D.M.J. Lambregts and R.G.H. Beets-Tan

11.1 Introduction/Clinical reviews with meta-analyses [3, 5, 8, 10]. The


Background main outcomes of these meta-analyses were that
a more intensive follow-up leads to a 7% improve-
After surgical treatment for colorectal cancer, ment in survival and to the detection of more cur-
approximately 35% of patients develop recur- able recurrences.
rent disease, the majority of which occurring
during the first 3 years after surgery. Twenty-
five to thirty percent of patients present with 11.2.2 Which Tools Should Be Used
metachronous metastases in the liver (20%) and/ for Surveillance and at What
or lungs (5–10%). These distant relapses are an Frequency?
important determinant for patient outcome and
survival. Around 5–15% of patients develop a The frequency of follow-up and routinely used
local relapse during follow-up. These local follow-up tools differs between the published
recurrences have little impact on survival, but studies. As a result, there is no consensus as to
are associated with significant morbidity and how frequently and with which modalities
therefore significantly affect the quality of life. patients should be followed. As distant metasta-
If detected when they are still small, local recur- ses have the most significant impact on patient
rences can be treated with curative salvage sur- outcome, the main focus of surveillance is the
gery, often combined with (neo)adjuvant chemo detection of metastatic disease. Imaging should
and/or radiation treatment. The problem is that therefore not only aim at detecting a local
at the time of detection, many recurrences are relapse but also focus on detecting distant
already in an advanced stage and only a limited relapse by a complete evaluation of the whole
number are curable. body including the most likely sites for distant
metastases. So far, the largest body of evidence
exists for a beneficial role of routine carcinoem-
11.2 Detection of Local Relapse bryonic antigen (CEA) testing and some sort of
During Routine Surveillance liver imaging, although it is not clear with which
After Rectal Surgery imaging modalities (CT, ultrasound) and at what
frequency this should best be performed. Three
11.2.1 Why Follow-up and Is It large European RCTs are still ongoing (the
Necessary? GILDA trial in Italy, the FACS trial in the UK
and the COLOFOL trial performed in Denmark,
‘Routine’ clinical surveillance after treatment Sweden, Poland, Ireland and Uruguay) investi-
serves several goals. The main aim is to detect gating the surveillance of colorectal cancer
recurrent and/or metastatic disease, to identify patients after curative surgery. Hopefully, the
the patients who require salvage or other curative results of these trials will provide more definite
treatment and thereby improve survival. evidence and result in more uniform and evi-
Secondary goals are to manage late post-treat- dence-based recommendations. While awaiting
ment complications, to document treatment out- the outcome of these trials, current guidelines
come and to meet patients’ psychological needs differ between Europe and the United States and
and improve the patient-doctor relationship. are mainly based on common clinical practice
Whether routine clinical follow-up is beneficial and the (conflicting) evidence from earlier stud-
and cost-effective has been the subject of ongo- ies. As it is known that the majority of recur-
ing debates and strong evidence supporting its rences occur in the first 3 years after treatment,
use is lacking. So far, mainly cohort and case– it seems logical to at least perform regular
control studies have been performed, together (6 monthly) follow-up during these first 3 years.
with a limited number of randomised controlled Especially in the high-risk patients (for instance
trials (RCT). The results of these RCTs have been the locally advanced rectal cancers), this may be
compared and combined in four systematic extended over the first 5 years.
11 How Can We Identify Local Relapse? 97

11.2.3 What Is the Role of CEA During than in patients with colon cancer, a more fre-
Routine Surveillance? quent (3–6 monthly) endoscopic evaluation of
the rectum or rectosigmoid seems to be justified.
Together with clinical symptoms, elevated CEA Especially in patients who have not received pre-
levels have been reported to be the most frequent operative treatment or who have undergone a
first clinical indicator of relapse [1]. The colorec- local or subtotal excision instead of total mesorec-
tal cancer surveillance guideline of the American tal excision (TME), more frequent endoscopic
Society of Clinical Oncology (ASCO) recom- evaluation is indicated.
mends 3-monthly CEA testing in patients with
stage II or III disease for at least 3 years after the
diagnosis [2]. Furthermore, it is advised to initi- 11.2.5 What Is the Role of Imaging
ate the CEA surveillance after adjuvant chemo- During Routine Surveillance?
therapy is finished, as CEA levels may be falsely
elevated in patients who are still receiving There is no solid evidence that routine imaging is
fluorouracil-based treatment. A second issue to beneficial during the surveillance of patients after
take into account is the fact that CEA levels are colorectal cancer surgery. The ASCO guidelines
only elevated in approximately 60% of the state that routine imaging is only justified in
patients with colorectal cancer. Therefore, it may patients with an increased risk for recurrence and
be considered to only perform routine CEA test- should mainly consist of annual CT of the chest
ing in those patients whose CEA levels were ini- and abdomen. Especially regular monitoring of
tially elevated at the time of their primary tumour the liver by imaging (CT or ultrasound) has been
diagnosis. shown to contribute to an improved survival.
Equal results were reported for CT and CEA for
the detection of curable recurrent disease. Pelvic
11.2.4 What Is the Role of Clinical imaging may be considered in patients with rectal
Examination (Endoscopy) cancer, in particular in patients who have not been
During Routine Surveillance? treated with radiotherapy. There is no role for rou-
tine surveillance using positron emission tomog-
Both the ASCO guidelines, the guidelines of the raphy (PET), as the benefits do not outweigh the
European Society of Medical Oncology (ESMO) high costs. Although MRI is known to be the pre-
[4], as well as a recent European consensus paper ferred modality for the primary assessment of rec-
[11], recommend a colonoscopy every 3–5 years tal tumours, pelvic surveillance by MRI as part of
after treatment, with the aim to exclude second- the routine follow-up scheme is not justified as it
ary tumours in the remaining colon and in rectal is known that – compared to follow-up using only
cancer patients to detect a local recurrence. For CEA testing – only little additional curable and
patients with an increased familial or inherited resectable recurrences are detected with MRI and
risk, a more intensive colonoscopic surveillance the yield is less than one percent [9].
is required, depending on their risk category
(which is based on the number of first-, second-
and third-degree relatives with colorectal cancer 11.3 Detection of Local Relapse
and/or genetic carriership for familial adenoma- in Case of an Increased Risk
tous polyposis or hereditary non-polyposis col- for Recurrence
orectal cancer). The yield of colonoscopy in the
detection of locally recurrent tumour in normal 11.3.1 Which Patients Constitute
risk patients is, however, very low. Colonoscopy the ‘Increased Risk’ Group?
seldom provides the first signs of recurrence and
sensitivity for detection of local relapse is less It has been suggested by different authors that a
than 50% [6]. Since the prevalence of a local more intensive follow-up should be preserved for
relapse in rectal cancer patients is much higher patients that have an increased risk to develop
98 D.M.J. Lambregts and R.G.H. Beets-Tan

recurrent disease. This includes patients with mass on consecutive CT examinations (Fig. 11.1).
prognostically unfavourable primary tumours In many patients, however, pelvic CT remains
(high TNM stage, poor histologic grade, blood/ inconclusive and patients need to be referred for
lymphatic vessel and perineural invasion, low additional imaging (PET and/or MRI). A draw-
number of nodes investigated at pathology), back of CT is that its limited soft tissue contrast
patients who have not undergone radiation treat- does not allow for an assessment of tumour resec-
ment, patients with elevated CEA levels or abnor- tability once a recurrent tumour is suspected.
mal liver function tests and patients with clinical
symptoms suspicious for recurrent disease.
Recently, a pooled analysis of the individual 11.3.4 What Is the Role of PET?
patient data from five European trials was per-
formed with the aim to develop a predictive model PET is generally applied as the second-line tech-
(based on clinical, treatment and pathologic nique in patients with a (strong) clinical suspicion
patient variables) to identify patient groups that of a local relapse in whom CT findings are equiv-
have an increased risk for local recurrence and/or ocal (Fig. 11.2). A recent meta-analysis that com-
distant metastases and may benefit from more pared the use of PET, CT and combined PET-CT
close follow-up. Although the use of such ‘nomo- for the detection of recurrent disease after col-
grams’ will need to be validated using external orectal cancer surgery found that PET and PET-CT
patient data, it may prove to be a useful support are superior to CT, especially in the specific sub-
tool to modulate the intensity of follow-up based group of patients who have a strong clinical suspi-
on a patient’s individual risk profile [12]. cion for recurrence (based on elevated CEA levels
or clinical symptoms). Hence, it may be defensi-
ble to immediately go for PET in these particular
11.3.2 What Is the Role of Clinical high-risk patients [7], although this is not the case
Examination? in routine practice, because of the invasiveness of
the technique and its high costs. Like CT, PET is
In patients with an increased risk for recurrence of limited value to determine tumour resectability.
(i.e., rectal cancer patients who have not under- Moreover, false-positive PET findings are fre-
gone radiation treatment and/or who have under- quently observed in areas of inflammation, in dis-
gone local excision instead of total mesorectal placed small bowel loops and in the proximity of
excision), routine endoscopic surveillance is the urinary bladder. Conversely, false-negative
indicated. In addition, endoscopic examination findings are known to occur in recurrent tumours
should be considered in patients in whom an of the mucinous tumour type.
endoluminal recurrence is expected based on
clinical symptoms (e.g., faecal blood loss) or
digital rectal examination. 11.3.5 What Is the Role of MRI?

Several studies have shown that MRI is more accu-


11.3.3 What Is the Role of CT? rate than CT for the detection of a local relapse.
Nevertheless, the primary use of MRI for detec-
In patients with an increased risk for a local tion of recurrent disease has rarely been investi-
relapse (mainly patients with elevated CEA lev- gated, and MRI is not widely available. Therefore,
els and/or clinical symptoms), pelvic CT is the MRI is not worldwide adopted in clinical practice.
modality of first choice and is generally com- This is also due to the fact that in patients with a
bined with a CT of the chest and abdomen to suspicion of locally recurrent disease, the simulta-
cover the whole body and simultaneously check neous search for distant metastases requires a more
for a distant relapse. CT is mainly useful to detect consolidated whole-body imaging technique such
a local relapse when there is a growing pelvic as CT. MRI has so far been less suitable than CT
11 How Can We Identify Local Relapse? 99

a b

Fig. 11.1 Two consecutive pelvic CT examinations of a (b) On the CT performed after 14 months, a growing
patient treated with TME for a primary T3N2 rectal pelvic mass (arrows) is identified, suspected for a recur-
tumour. CT examinations were performed 6 (a) and 14 (b) rent tumour. (c) Corresponding PET-CT confirmed the
months after rectal cancer surgery. (a) On the CT per- suspicious finding of a local relapse, which was confirmed
formed after 6 months, there are no signs of a local relapse. to be recurrent adenocarcinoma at biopsy

and PET because of the long acquisition times 11.3.6 What Are the Limitations
required to cover the whole body and because of in the Imaging Detection
its limited use for the evaluation of metastatic lung of Local Relapse?
disease. There are, however, several authors that
have advocated MRI as the technique of first It has been shown that pelvic imaging mainly
choice for the evaluation of local relapse. Due to fails in detecting anastomotic, intraluminal recur-
its excellent soft-tissue contrast, MRI is a valu- rences, which tend to be smaller in size. For the
able tool to assess the extent of a locally recurrent assessment of this type of relapse, endoscopy is
tumour and to evaluate possible tumour growth better suitable. Furthermore, the techniques of
into adjacent organs and tissues. Hence, MRI has first choice (CT and to a lesser extent PET) are
proven most valuable in determining whether a of limited use for the assessment of local tumour
local relapse is still resectable (Fig. 11.3). resectability, whereas MRI – which is more
100 D.M.J. Lambregts and R.G.H. Beets-Tan

a b

Fig. 11.2 CT (a), PET (b) and fused PET-CT (c) images relapse. (b/c) On the corresponding PET examination (b)
of a patient suspected of recurrent tumour. (a) On CT, a and the fused PET-CT images (c), a clear area of increased
mass (arrowheads) is visible, but it is not possible to dif- FDG uptake is identified, which was confirmed to be a
ferentiate between postoperative scar tissue and a local local relapse at biopsy

accurate for the latter purpose – is not helpful for cell proteins and carbohydrates, and other factors
whole-body staging of recurrent disease. So far, such as microvessel density, DNA content and
none of the available imaging techniques thus cell proliferation indices. In the future, such
qualifies as a complete one-stop-shop technique. factors may help to determine prognosis and aid
in establishing a risk-adapted surveillance strat-
egy. However, since multivariate analyses and
11.3.7 Are There Any New Techniques prospective validation studies are not yet avail-
That May Help in the Evaluation able, the use of these markers is currently not
of Local Relapse? recommended. New imaging techniques, com-
bining functional and morphological imaging
Numerous studies are being undertaken investi- information, are also being investigated. The first
gating the clinical value of prognostic and predic- pilot results on techniques such as dynamic con-
tive markers, including molecular markers (e.g. trast-enhanced MRI (DCE-MRI), diffusion-
tumour suppressor genes and growth factors), weighted MRI (DWI) and image fusion techniques
11 How Can We Identify Local Relapse? 101

a b

Fig. 11.3 CT (a), PET (b) and MRI (c) examinations of a feasible to determine the tumour ingrowth into adjacent
patient with a local relapse after rectal cancer surgery. (a) organs or structures. (c) On MRI, it can be appreciated
On CT, a mass is visible, although it is difficult to dis- that the tumour grows into the sacrum (arrowheads), the
criminate between postoperative scar tissue and tumour. iliac vessels on the right side (white arrow) and into the
(b) On PET, a clear uptake of FDG highly suspicious of bladder (black arrow). This example illustrates that MRI
recurrent tumour can be discerned. It is, however, not is the preferred technique to evaluate tumour resectability

combining PET/MRI and DWI/MRI have shown ongoing large trials investigating this issue,
encouraging results, and further large-scale current guidelines are based on common clini-
patient studies are required to establish their cal practice and the evidence provided by pre-
potential clinical value for the evaluation of vious meta-analyses, which used different
recurrent disease. follow-up schedules. These state that routine
follow-up in patients with rectal cancer should
Conclusion at least include regular clinical examination,
To date, there is no strong evidence yet CEA testing and some sort of liver imaging. In
on what should be considered the most opti- patients with an increased risk for local recur-
mal surveillance strategy after colorectal can- rence (based on prognostic factors, elevated
cer surgery. While awaiting the evidence of CEA levels or clinical symptoms), imaging is
102 D.M.J. Lambregts and R.G.H. Beets-Tan

justified and CT is the technique of first choice. 6. Kievit J (2002) Follow-up of patients with colorectal
In case of equivocal CT findings, PET is more cancer: numbers needed to test and treat. Eur J Cancer
38:986–999
beneficial in identifying a local relapse. The 7. Maas M, Rutten IJ, Nelemans PJ, Lambregts DM,
main role of MRI is to establish the resectabil- Cappendijk VC, Beets GL, Beets-Tan RG (2011)
ity of a local tumour recurrence once it has What is the most accurate whole-body imaging
been diagnosed. modality for assessment of local and distant recurrent
disease in colorectal cancer? A meta-analysis: imag-
ing for recurrent colorectal cancer. Eur J Nucl Med
Mol Imaging 38(8):1560–1571
8. Renehan AG, Egger M, Saunders MP, O’Dwyer ST
(2002) Impact on survival of intensive follow up after
References curative resection of colorectal cancer: systematic
review and meta-analysis of randomised trials. BMJ
1. Arriola E, Navarro M, Parés D, Muñoz M, Pareja L, 324:813
Figueras J, Soler G, Martinez M, Majem M, Germa- 9. Titu LV, Breen DJ, Nicholson AA, Hartley J, Monson
Lluch JR (2006) Imaging techniques contribute to JR (2006) Is routine magnetic resonance imaging
increased surgical rescue of relapse in the follow-up justified for the early detection of resectable liver
of colorectal cancer. Dis Colon Rectum 49:478–484 metastases from colorectal cancer? Dis Colon Rectum
2. Desch CE, Benson AB 3rd, Somerfield MR, Flynn PJ, 49:810–815
Krause C, Loprinzi CL, Minsky BD, Pfister DG, 10. Tjandra JJ, Chan MK (2007) Follow-up after curative
Virgo KS, Petrelli NJ, American Society of Clinical resection of colorectal cancer: a meta-analysis. Dis
Oncology (2005) Colorectal cancer surveillance: 2005 Colon Rectum 50:1783–1799
update of an American society of clinical oncology 11. Valentini V, Aristei C, Glimelius B, Minsky BD,
practice guideline. J Clin Oncol 23:8512–8519 Beets-Tan R, Borras JM, Haustermans K, Maingon P,
3. Figueredo A, Rumble RB, Maroun J, Earle CC, Overgaard J, Pahlman L, Quirke P, Schmoll HJ,
Cummings B, McLeod R, Zuraw L, Zwaal C, Sebag-Montefiore D, Taylor I, Van Cutsem E, Van de
Gastrointestinal Cancer Disease Site Group of Cancer Velde C, Cellini N, Latini P, Committee S (2009)
Care Ontario’s Program in Evidence-based Care (2003) Multidisciplinary rectal cancer management: 2nd
Follow-up of patients with curatively resected colorec- European rectal cancer consensus conference
tal cancer: a practice guideline. BMC Cancer 3:26 (EURECA-CC2). Radiother Oncol 92:148–163
4. Glimelius B, Pahlman L, Cervantes A, ESMO 12. Valentini V, van Stiphout RG, Lammering G,
Guidelines Working Group (2010) Rectal cancer: Gambacorta MA, Barba MC, Bebenek M, Bonnetain
ESMO clinical practice guidelines for diagnosis, F, Bosset JF, Bujko K, Cionini L, Gerard JP, Rödel C,
treatment and follow-up. Ann Oncol 21(Suppl 5): Sainato A, Sauer R, Minsky BD, Collette L, Lambin P
v82–v86 (2011) Nomograms for predicting local recurrence,
5. Jeffery GM, Hickey BE, Hider P (2002) Follow-up distant metastases, and overall survival for patients
strategies for patients treated for non-metastatic col- with locally advanced rectal cancer on the basis of
orectal cancer. Cochrane Database Syst Rev (1): European randomized trials. J Clin Oncol 29(23):
CD002200 3163–3172
Part IV
Q&As on Radiotherapy
When Should Preoperative
Short-Course Radiotherapy 12
or Long-Course Chemoradiotherapy
Be Performed?

David Sebag-Montefiore and Robert Glynne-Jones

Contents 12.1 Introduction


12.1 Introduction ................................................ 105
Traditionally, there has been a high local recurrence
12.2 Rationale for the Different Approaches ... 106
rate after radical surgery in rectal cancer, and
12.3 Evidence Supporting the Efficacy of 10–40% of patients require a permanent stoma.
Short-Course Radiotherapy ...................... 106 Both short-course preoperative radiotherapy
12.3.1 Clinical Trials............................................... 106
12.3.2 Population-Based Data ................................ 108 (SCPRT) and long-course preoperative chemoradia-
12.3.3 Meta-analysis ............................................... 108 tion (CRT) are advocated to reduce this risk of local
12.4 Evidence Supporting Long-Course
recurrence and enable a curative resection. The role
Preoperative Chemoradiotherapy ............ 109 of preoperative radiotherapy to facilitate sphincter
12.4.1 Clinical Trials............................................... 109 sparing surgery remains highly contentious.
12.4.2 Meta-analysis ............................................... 109 There is considerable debate regarding the
12.5 Direct Comparison choice of preoperative radiotherapy regimen that
of the Two Approaches .............................. 111 should be used in patients with resectable rectal
12.6 Acute and Late Toxicity cancer, i.e., where staging MRI suggests that a
of SCPRT and CRT ................................... 111 standard conventional total mesorectal excision
12.7 Summary of the Current Data .................. 113 (TME) is considered to offer a curative option.
There is a strong supporting evidence base for
12.8 Preoperative Staging .................................. 113
both short-course radiotherapy (SCRT) and con-
12.9 Selection of Patients for Preoperative current chemoradiotherapy (CRT), and it is note-
Radiotherapy (SCPRT or CRT) ............... 113
worthy that the strongest support for either approach
References ................................................................. 116 is from the countries in which the key phase III tri-
als were performed. For example, short-course
radiotherapy is used more frequently in Scandinavia,
D. Sebag-Montefiore (*)
Department of Clinical Oncology, the Netherlands and the United Kingdom. Long-
University of Leeds, St James’s Institute of Oncology, course chemoradiotherapy is more frequently used
St James’s University Hospital, in southern European countries and the United
Beckett St, LS9 7TF Leeds, UK
States. Radiation oncologists usually favour one or
e-mail: d.sebagmontefiore@leeds.ac.uk
other of these approaches. This debate focuses
R. Glynne-Jonesfi more on a blanket approach and previous experi-
Department of Clinical Oncology, ence with these different schedules, with one or
Mount Vernon Centre for Cancer Treatment,
other of the above aims being predominant, rather
Rickmansworth Rd,
Northwood, Middlesex HA6 2RN, UK than choosing treatment based on the individual
e-mail: rob.glynnejones@nhs.net circumstances/stage.

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 105


DOI 10.1007/978-3-642-25005-7_12, © Springer-Verlag Berlin Heidelberg 2012
106 D. Sebag-Montefiore and R. Glynne-Jones

In contrast, where the MRI suggests the cir- 12.3 Evidence Supporting the
cumferential resection margin (CRM) or, more Efficacy of Short-Course
accurately, the mesorectal fascia (MRF) is poten- Radiotherapy
tially involved, and a standard conventional total
mesorectal excision (TME) will not offer a cura- Short-course radiotherapy was developed in
tive option, there is almost universal agreement Sweden at a time when surgery was the sole treat-
that the patient requires preoperative chemora- ment modality for rectal cancer and there was no
diotherapy with an interval to allow for downsiz- evidence to support the use of adjuvant preopera-
ing/downstaging. tive radiotherapy. In order to test the role of pre-
This chapter assesses the role of radiation operative radiotherapy, a minimal delay to surgical
therapy in rectal cancer, with emphasis on patient resection was essential, and Swedish investigators
selection for preoperative chemoradiotherapy elected to evaluate the use of a high dose per frac-
(CRT) and short-course preoperative radiother- tion, hypofractionated 1-week regimen.
apy (SCPRT) and the outcomes obtained from
each of these strategies.
12.3.1 Clinical Trials

12.2 Rationale for the Different Following initial development, a sequence of ran-
Approaches domised phase III trials were performed compar-
ing SCPRT followed by surgery with surgery
The rationale for SCPRT is based on the short alone. Although earlier studies used 5 Gy per
overall treatment time (OTT), which allows sur- fraction with total doses of 15–20 Gy, subsequent
gery to take place promptly (ideally within trials evaluated 25 Gy in 5F. Between 1980 and
7 days) before the radiation reaction is expressed. 1993, over 2,000 patients were evaluated in the
The radiotherapy dose (5 × 5 Gy) is high in bio- Stockholm and Swedish Rectal Cancer trials. The
logical terms and avoids the potential for accel- Stockholm I trial used parallel opposed fields
erated repopulation which may occur in the latter treating a large volume. This approach was asso-
part of long-course chemoradiation. Other ciated with increased operative mortality and
advantages include a very short period of deliv- substantial late toxicity.
ery, high compliance and low cost. Adjuvant The Swedish rectal cancer trial [1] (Table 12.1)
chemotherapy with systemically active sched- was performed between 1987 and 1990 and ran-
ules (e.g., FOLFOX) can be started with mini- domised 1,168 patients to receive either SCPRT
mal delay if deemed necessary, within a few followed by surgery or surgery alone. The use of a
weeks of diagnosis. The disadvantage is that 3- or 4-field radiotherapy technique and a reduc-
there is insufficient time for substantial downsiz- tion in the superior limit of the target volume to the
ing/downstaging to occur, although this may in mid-L4 vertebral body prevented any increase in
fact be an advantage because it allows accurate operative mortality. Local recurrence was reduced
pathological staging in terms of the nodal status. from 27% to 11% (p < 0.001), and 5-year survival
However, SCPRT may compensate only partially increased from 48% to 58% with surgery alone
for a positive CRM. and SPCRT and surgery, respectively (p = 0.004).
The rationale for long-course chemoradiation A recent report confirms that the benefits shown
is to achieve additive effects both locally and are sustained after 13 years of follow-up.
systemically with a concurrent fluoropyrimidine, The above trials reported local recurrence
thereby inducing downstaging/downsizing rates of >20% with surgery alone and reflect sur-
reducing metastases and in a small group of gical practice at the time the trials were con-
patients achieving tumour sterilisation. Its role ducted. However, surgical techniques improved
in facilitating sphincter sparing procedures is with the adoption of total mesorectal excision
contentious. (TME) described by Heald, resulting in reported
12

Table 12.1 Key trials of short-course radiotherapy and surgery versus surgery alone/selective post-operative (chemo)radiotherapy
Duration PAT 1° end
Trial of trial NOS Randomisation NOS TME point LR METS OS DFS
Swedish 1987–1997 1,168 25 Gy/5# + surgery 553 11% 23% 58%
Rectal No OS 5 years 5 years 5 years Not stated
Cancer Surgery alone 557 27% 24% 48%
Trial
(1997)
Updated 9% 34% 38%
(2005) 13 years 13 years 13 years Not stated
26% 34% 30%
Dutch Trial 1996–1999 1,861 25 Gy/5# + surgery 897 2% 14.8% 82%
CKVO Yes LR 2 years 2 years 2 years Not stated
95–04 Surgery + highly 908 8% 16.8% 81.8%
(2001) selective RT
Updated 5% 25% 48%
(2011) 10 years 10 years 10 years Not stated
When Should Preoperative Short-Course Radiotherapy or Long-Course

11% 28% 49%


CRO7 1997–2005 1,350 25 Gy/5# + surgery 674 5% 19% 80% 78%
(2008) Yes LR 3 years 3 years 3 years 3 years
Surgery + highly 676 11% 21% 79% 72%
selective CRT
TME total mesorectal excision, LR local recurrence, DFS disease-free survival, OS overall survival
107
108 D. Sebag-Montefiore and R. Glynne-Jones

local recurrence rates of 10% or less. A key ques- between 1990 and 2002 that were then divided
tion at this time was whether SCPRT compen- into three time intervals: prior to, during and after
sated for poor surgical technique, and further the Dutch TME trial. The 5-year overall survival
trials were then designed to test the role of SCPRT was 56%, 62% and 65% for the three groups,
combined with TME. respectively. Multivariate analysis demonstrated
The Dutch TME trial [2] and the Medical preoperative radiotherapy had a statistically
Research Council CR07 trial [3] (Table 12.1) significant impact on survival (p < 0.001).
compared a policy of routine SPCRT and imme- A recent publication from Sweden [4] has
diate surgery against initial surgery with a policy evaluated 6,878 patients receiving treatment for
of highly selective post-operative (chemo)radio- rectal cancer between 1995 and 2001 when 41%
therapy restricted to patients with involvement of of patients received SCPRT. Local recurrence at
the circumferential resection margin (the Dutch 5 years was 6% with SPCRT + surgery and 12%
trial used radiotherapy alone and CR07 concur- with surgery alone. The authors elected to com-
rent 5FU chemoradiation). Adjuvant chemother- pare the overall survival of the two groups
apy was not used in the Dutch trial, whereas restricted to patients under the age of 75 and
patients with stage III disease received 5FU che- undergoing anterior resection or APER. The
motherapy in the CR07 trial. Hazard Ratio for overall survival was 0.70 (0.69–
The two trials recruited a combined total of 0.72) in favour of SCPRT. However, the
3,150 patients and have reported very similar justification for this subset analysis of 3,466
results which are summarised in Table 12.1. The patients is limited to the authors stating that the
rate of local recurrence was approximately halved excluded patients have ‘higher comorbidity, their
from 11% with TME to 4–5% with the addition of expected survivals are lower and they are less fre-
SCPRT to TME. There is no evidence of any quently referred for irradiation.’
impact on overall survival for the whole trial pop-
ulation. In the CR07 trial, pathologists assessed
the plane of surgery achieved in the surgical resec- 12.3.3 Meta-analysis
tion specimen (mesorectal, intra-mesorectal and
muscularis propria planes). A higher rate of LR A number of meta-analyses have been performed
was seen in the muscularis propria plane, and a of the phase III clinical trials that tested the addi-
reduction in LR was seen with SCPRT for all three tion of preoperative radiotherapy to surgery
planes. This latter finding confirms that adjuvant alone. The Colorectal Cancer Collaborative
preoperative radiotherapy is of additional benefit Group [5] identified 14 preoperative trials that
even when the surgical technique is optimised. recruited a total of 6,350 patients, with seven tri-
The Dutch group have recently reported 10-year als using a short-course of radiotherapy with
outcome data. Subset analysis of the CRM-ve 5 Gy fractions. A significant reduction was dem-
subset demonstrated a benefit for SCPRT for all onstrated in both local recurrence (p < 0.00001)
rectal subsites. The initial CR07 trial report also and overall recurrence (p < 0.00001). A 2.9%
described this finding. A further subset analysis absolute difference in overall survival was seen
showed a statistically improved overall survival in favour of adjuvant radiotherapy (p = 0.06).
for patients with stage III CRM-ve disease with A significant increase in non-cancer-related
the addition of SCPRT. Long-term follow-up from deaths was also seen for patients who received
the CR07 data is awaited regarding this finding. preoperative radiotherapy. The reasons for this
latter finding remain unclear 10 years after this
finding was reported. Other meta-analyses
12.3.2 Population-Based Data broadly concur with these findings.
There is a strong evidence base supporting
Den Dulk et al. used cancer registry records to the benefit of SCPRT with an approximate
identify 3,179 patients with rectal cancer treated halving in local recurrence both before and
12 When Should Preoperative Short-Course Radiotherapy or Long-Course 109

after the introduction of TME. Its impact on toxicity was also significantly less with the pre-
overall survival is less clear, but the current operative approach.
evidence suggests that if any survival advan- The results of these three trials led to a major
tage is achieved, it is very small and does not shift from selective post-operative CRT-based
exceed 2–3%. Other meta-analyses have reported to imaging-based selection of patients for pre-
similar conclusions. operative CRT. This strategy has been further
supported by the results of the NSABP R03 trial
(Table 12.3) The NSABP R03 used a similar
12.4 Evidence Supporting design to the German trial but only recruited
Long-Course Preoperative 267 of its planned patient target number
Chemoradiotherapy (n = 900), so results should be interpreted with
caution. Five-year loco-regional recurrence was
12.4.1 Clinical Trials 10.7% in each treatment arm (p = 0.693).
A significant improvement of 5-year DFS (65%
This approach was initially developed as post- vs. 53% p = 0.011) and a non-significant
operative adjuvant therapy. In North America, an improvement in 5-year OS (75% vs. 66%
NIH consensus statement in 1990 recommend p = 0.065) were also observed for the preopera-
the use of adjuvant post-operative chemotherapy tive arm.
and concurrent chemoradiotherapy using 5FU for Whilst the trials discussed above selected
all patients with T3/4 or node-positive resected patients with resectable disease, a Scandinavian
rectal cancer. trial of 207 patients with non-resectable rectal
In Europe, two phase III trials were performed cancer conducted between 1996 and 2003 ran-
between 1993 and 2003 that compared long- domised patients to receive CRT with 50 Gy plus
course preoperative radiotherapy (45 Gy in 25 5FU/LV versus long-course radiotherapy alone
fractions) with concurrent 5FU leucovorin added (50 Gy). In this more advanced high-risk group, a
to the same radiotherapy schedule (Table 12.2). statistically significant reduction in local failure,
The FFCD 9203 trial [6] recruited 762 patients time to treatment failure and cancer-specific sur-
and recommended post-operative 5FU/LV for all vival was observed in patients who received pre-
patients, whereas the EORTC 22921 [7] recruited operative CRT.
1,011 patients phase III trials and used a factorial We are not aware of any population-based
design that also compared post-operative CRT analyses of the use of preoperative CRT. This
versus no chemotherapy. approach is however widely used. Concurrent
Both trials demonstrated that the addition of chemotherapy may consist of 5FU either as a
concurrent 5FU/LV was associated with an continuous infusion, in combination with leuco-
acceptable increase in acute toxicity and path- vorin or using an oral fluoropyrimidine such as
ological downstaging. Both trials reported very capecitabine.
similar findings with a significant reduction in
the rate of local recurrence from 15% to
8–10%, but with no difference in disease-free 12.4.2 Meta-analysis
or overall survival. The results are summarised
in Table 12.1. A Cochrane overview identified four trials and
The German rectal cancer group [8] concluded that preoperative CRT enhances path-
(Table 12.3) then directly compared preoperative ological response and improves local control but
versus post-operative 5FU CRT. A total of 823 does not benefit disease-free or overall survival.
patients were randomised between 1995 and A recent pooled analysis of 2,795 patients from
2002. The rate of local recurrence was reduced five European trials showed that preoperative
from 12% with post-operative CRT compared CRT impacted on distant metastases and overall
with 6% with preoperative CRT. Acute and late survival [9].
110

Table 12.2 Key trials of preoperative radiotherapy versus preoperative chemoradiotherapy


Duration of
Trial trial PAT NOS Randomisation NOS TME 1° end point Local recurrence METS Overall survival Disease-free survival
EORTC 1993–2003 1,011 45 Gy vs. 505 No OS 17% vs. 9% 5 years 32% 65% vs. 5 years 54% vs. 5 years
22921 FUFA + 45GY 506 No overall 66% 56%
(2006)
FFCD 9203 1993–2003 762 45 Gy vs. 367 No OS 17% vs. 8% 5 years No data 68% vs. 5 years No data 375
(2006) FUFA + 45GY 375 No 67%

Table 12.3 Key trials of preoperative chemoradiotherapy versus post-operative chemoradiotherapy


Duration of 1° end Disease-free
Trial trial PATNOS Randomisation NOS TME point Local recurrence METS Overall survival survival
NSABP 1993–1999 267 Preoperative 123 No OS 11% vs. 5 years Not stated 74% vs. 5 years 64% vs. 5 years
RO3 (2010) 45 Gy + FUFA 131 11% 66% 53%
vs. Post-
operative
Gy + FUFA
CAA/ARO/ 1995–2002 823 Preoperative 421 Yes OS 6% vs. 13% 5 years 36% vs. 76% vs. 5 years 68% vs. 5 years
AIO-94 50.4 Gy +5FU 402 38% 74% 65%
(2004) vs. Post-
operative
55.8 Gy +5FU
D. Sebag-Montefiore and R. Glynne-Jones
12 When Should Preoperative Short-Course Radiotherapy or Long-Course 111

12.5 Direct Comparison of the Two The main concern for many clinicians is the
Approaches use of 5 Gy fractions with the SCPRT regimen
and the risk of substantial late toxicity. This view
Two phase III trials have reported a direct com- is supported by reports from the Stockholm I and
parison of SCPRT with preoperative CRT II trial that reported a significant increase in
(Table 12.4). The Polish trial [10] randomised venous thromboembolism, pelvic and femoral
312 patients with resectable rectal cancer between neck fractures, small bowel obstruction and post-
1999 and 2002. The trial tested the hypothesis operative fistulae. It is possible that the 2-field
that the use of preoperative CRT with a 4–6-week technique and the large field sizes used may
delay to surgery would increase the rate of sphinc- explain some of these findings.
ter preserving resection compared with SPCRT However, the Dutch and CR07 trials have
and immediate surgery. No difference in the pri- reported an increase in faecal incontinence, uri-
mary end point of the sphincter preservation rate nary and sexual dysfunction and second malig-
was seen. The 5-year rate of local recurrence was nancy when SCPRT is combined with TME.
9% and 14% (p = 0.17) for SCPRT and CRT, The data from the MCR CR07 trial demonstrate
respectively, with no significant difference in that a substantial component of late toxicity is
disease-free and overall survival. attributable to surgery with a smaller but
The Trans Tasman Group (TROG) trial also significant additional impact due to the addition
randomised 326 patients with resectable rectal of SPCRT [11].
cancer between 2001 and 2006 to receive either These findings support the concerns regard-
SPCRT or preoperative CRT to compare the rate ing late radiation toxicity with the use of 5 Gy
of local recurrence. The initial report at ASCO in fraction size. However, it is important to recog-
2010 reported no statistical difference in the pri- nise that the key trials of SCPRT have all
mary end point of local recurrence or secondary reported detailed analyses of late toxicity. This
end pointes of failure-free or overall survival. contrasts with very few publications that report
The two trials discussed above are underpow- the incidence of late complications after preop-
ered, either individually or combined, to detect a erative CRT.
clinically relevant but small difference in LR (i.e. A recent publication by Tiv et al. [12] describes
5%) and therefore do not provide sufficient evi- quality of life outcomes in the EORTC 22921
dence to definitively compare efficacy. trial after a median of 4.6 years. Low quality of
life scores for sexual functioning were reported
and were more severe in men than women. The
12.6 Acute and Late Toxicity addition of chemotherapy to radiotherapy was
of SCPRT and CRT associated with increased diarrhoea and lower
global quality of life. There is no data that com-
As SCPRT and CRT appear broadly comparable in pares the late toxicity of surgery versus preopera-
terms of efficacy despite the dangers of cross trial tive CRT.
comparison, a comparison of acute and late toxic- The Polish and the TROG trials are of con-
ity is of considerable importance. Data from the siderable importance when comparing the late
Polish trial demonstrated 18% acute toxicity with toxicity of SCPRT against preoperative CRT
CRT compared with 3% with SCPRT, respectively due to the randomised comparison of the two
(p < 0.001). The provisional report from the TROG treatment approaches. The Polish trial [10]
trial shows a similar pattern. In contrast, SCPRT is reported no significant difference for severe late
associated with a subacute neuropathy in <1–2% toxicity with rates of 10% and 7% for SPCRT
of patients which is usually reversible. This side and CRT, respectively (p = 0.17). A recent pre-
effect has not been reported after CRT. Late toxic- sentation from the TROG trial did not detect any
ity appears also equivalent in the two trials, although significant quality of life differences between
the details and documentation are limited. the treatment arms.
112

Table 12.4 Key trials of preoperative short-course radiotherapy versus chemoradiotherapy


Duration PAT 1° end
Trial of trial NOS Randomisation NOS TME point Local recurrence METS Overall survival Disease-free survival PCR
Polish 1999–2004 316 25 Gy/5# 155 Yes SPS 11% vs. 4 years No data 67% vs. 4 years 58% vs. 4 years 1%
Trial SCPRT vs. 157 16% 66% 56% 16%
(2006) FUFA
+50.4 Gy
Trans 2001–2006 326 25 Gy/5# No data Yes Local 7% vs. 4% 3 years No data 74% vs. 5 years No data No data
Tasman SCPRT vs. recurrence 70%
Radiation 50.4 Gy + PVI
Oncology 5FU
Group
TROG
01–04
(ASCO
2010)
SPS sphincter preserving surgery
D. Sebag-Montefiore and R. Glynne-Jones
12 When Should Preoperative Short-Course Radiotherapy or Long-Course 113

12.7 Summary of the Current Data well as the distance from the most lateral extent
of gross tumour to the mesorectal fascia, leva-
There is strong evidence that supports the use of tor or anal sphincter depending on the level
both SPCRT and preoperative CRT. Regarding within the rectum. All imaging modalities have
efficacy, both approaches approximately halve limitations in their accuracy of predicting
the rate of local recurrence with no convincing lymph node involvement. However, enlarged
evidence of an impact in disease-free or overall nodes with an irregular border and mixed signal
survival within the individual clinical trials. This characteristics are more likely to be involved
finding probably reflects the observation that the by tumour.
majority of patients within these trials had early In routine clinical practice, the radiologist can
stage disease. identify the imaging T stage and the presence or
Local recurrence reflects both tumour stage absence of any involved nodes using the above
and surgical technique, but the development of criteria, macroscopic extramural vascular inva-
metastases appears relatively independent of sur- sion, and patients where the lateral resection mar-
gery as the clinical trials demonstrate that 25–40% gin is at risk (<1 mm) or involved by tumour.
of patients with resectable disease develop metas-
tases despite increasingly low rates of local recur-
rence. Hence, the consistent improvement in 12.9 Selection of Patients for
local control resulting from SCPRT and CRT in Preoperative Radiotherapy
rectal cancer in the total mesorectal excision era (SCPRT or CRT)
has not translated into a benefit in overall
survival. The local disease extent demonstrated on pelvic
Both approaches increase the rate of late tox- staging investigations results in a spectrum of
icity when added to TME, and although there are disease ranging from the smallest tumour consid-
more publications describing late toxicity after ered on imaging to be T2 or less without any
SCPRT, it is likely that CRT results in similar lev- adverse radiological features through to very
els of toxicity. This conclusion is supported by large and bulky T3/4 tumours.
the data from the Polish trial. It is therefore essen- Many centres will seek to identify patients
tial that we consider both the risks as well as the with early stage disease where preoperative
benefits when considering the use of either radiotherapy can be avoided at the early end of
SCPRT or CRT. the disease spectrum. The Dutch and CR07 trials
both demonstrate a 3% rate of local recurrence
for patients with T1–2 N0 disease. It is impor-
12.8 Preoperative Staging tant to exclude the presence of any adverse risk
factors (including EMVI and unequivocal lymph
High-quality imaging investigations are of crit- node involvement) to safely omit SPCRT or CRT
ical importance to determine whether there is in patients with a primary tumour staged as T2
any evidence of metastatic disease and the local or less.
disease extent within the pelvis. In early stage At the other end of the spectrum, preoperative
disease, transrectal ultrasound has an important CRT is the treatment of choice when the primary
role, although in the majority of patients, high- tumour is staged radiologically as T4b (involve-
resolution pelvic MRI is considered the investi- ment of another organ) or when macroscopic
gation of choice. The prospective multicentre tumour involved or threatens (<1 mm) the
MERCURY trial established that the measured mesorectal fascia levator or sphincter complex.
extramural spread of primary tumour seen on Current evidence supports the use of concur-
pelvic imaging was equivalent to the same mea- rent 5FU/LV, continuous infusion 5FU or an oral
surement in the resection specimen. The mac- fluoropyrimidine. There is limited comparative
roscopic extramural spread can be measured as data regarding the choice of fluoropyrimidine,
114 D. Sebag-Montefiore and R. Glynne-Jones

Table 12.5 Risk of local recurrence according to local disease extent


High A threatened (<1 mm) or breached resection margin or low tumours encroaching
onto the intersphincteric plane or with levator involvement
Moderate Any cT3b or greater in which the potential surgical margin is not threatened or any
suspicious lymph node not threatening the surgical resection margin or the presence
of extramural vascular invasion
Low T1 or cT2 or cT3a and no lymph node involvement

although an initial report at ASCO 2011 from the of patients at higher risk of local recurrence or pre-
NSABP R04 trial suggests similar toxicity for dict which patients would benefit most. We there-
concurrent capecitabine compared with continu- fore rely on known histopathological prognostic
ous infusion 5FU. The addition of biological or factors that can also be identified on pelvic MRI
chemotherapy agents should only be evaluated in prior to surgical resection. This included the mea-
the context of clinical trials and is discussed in sured depth of extramural spread of primary T3
Chapters 19 and 18 respectively. disease, the presence of macroscopic vascular
When preoperative CRT is given, an interval invasion and involvement of lymph nodes using
of 4–12 weeks is used to allow tumour regression the criteria described above.
prior to resection with little research evidence to There is clear evidence that supports the
indicate the best time interval to use. choice of either SCPRT or preoperative CRT for
Some patients are considered unsuitable for this group. The current evidence that has com-
preoperative CRT, on the basis of biological age, pared these two approaches is limited but sug-
performance status or major cardiovascular co- gests that the two approaches are broadly similar
morbidity. In this situation, an alternative option in their ability to lower the risk of local recur-
is to use SCPRT with a delay of 6–12 weeks prior rence and so both approaches are acceptable. The
to surgery. The supporting evidence for this authors believe that both SCPRT and CRT are
approach is limited. Individual centre experience acceptable options.
has been reported from Upsalla in Sweden and The National Institute of Health and Clinical
Leeds in the United Kingdom. This is further Excellence has recently released the pre-publica-
supported (albeit in resectable cancers) by an tion of the 2011 colorectal guidelines for the
interim analysis of the Stockholm IV trial which United Kingdom (http://guidance.nice.org.uk/
compares SCPRT with immediate surgery, CG/Wave16/2). This document describes the
SCPRT with delay and 50 Gy in 25 fractions with identification of three differing levels of risk for
delay to surgery. local recurrence and is shown in Table 12.5.
The remaining patients, where the primary Patients are considered low risk when the pelvic
tumour is staged as T3 but does not involve or MRI demonstrated T3a disease or less without
threaten the margins, represent over 50% of adverse features. The authors support the view
patients with rectal cancer, and it is this group that this patient group should avoid radiotherapy
where the role of SPCRT or CRT is frequently altogether and have surgery alone.
debated. The routine use of either SPCRT or CRT Moderate risk is defined as any patient with
represents substantial overtreatment, and in the imaging stage T3b or greater tumour (>5 mm
opinion of the authors, a selective approach is beyond the muscularis propria) or EMVI or
essential, attempting to balance the benefits in the lymph node involvement without any threat to
reduction of local recurrence against the late tox- the margin. The authors support the view that this
icity associated with SCPRT or CRT. patient group should be considered for either
This underlines the need to identify predictive SPCRT or preoperative CRT. Both approaches
factors to assist the decision-making process. Little have supporting evidence, and it is not possible to
progress has been made in the identification of distinguish any clear efficacy or late toxicity dif-
molecular biomarkers to either assist the selection ferences based on the available data.
12 When Should Preoperative Short-Course Radiotherapy or Long-Course 115

Fig. 12.1 Management of Management of local disease - patients with rectal cancer
local disease (From 2011
NICE guidance)
Patient with
rectal cancer

MRI to assess local recurrence


determined by anticipated resection
margin, tumour and lymph node staging,
unless contraindicated

Patient information and support

Risk of local recurrence

High risk (locally


Low risk Moderate risk
advanced)1

Consider Consider
Chemoradio−
therapy2

SCPRT

Interval before surgery to allow


shrinkage and response
Proceed immediately to

Surgery

See algorithm on “Post−operative care”

Patients are considered high risk if tumour local recurrence and entail a high risk of meta-
breaches or threatens (<1 mm) the mesorectal fas- static disease. In contrast to 5FU-based CRT
cia, levator or intersphincteric plane (Fig. 12.1). with an interval to allow downsizing, SCPRT
allows definitive surgery to take place without
Conclusions delay and allows the use of systemically active
Preoperative radiotherapy has a strong sup- chemotherapy (FOLFOX) within weeks.
porting evidence base demonstrating that the In view of the well-documented long-term
rate of local recurrence can be approximately side effects associated with radiation, it is
halved when combined with TME. In contrast important to identify patients at low risk of
to unresectable/borderline resectable cancers, local recurrence who can avoid radiotherapy
there is no evidence for increased efficacy of altogether. Imaging can clearly identify patients
CRT over SCPRT in moderate-risk resectable whose margins are at risk and require preoper-
cancers. The two strategies appear equally ative CRT.
effective, and the main rationale for choosing The main challenge is the majority of patients
between them in this setting relies on high- with T3 disease where the balance between risk
quality MRI-based staging to identify the and benefit must be carefully considered and
extent of local disease and the presence of key where both SPCRT and preoperative CRT are
prognostic factors, which increase the risk of acceptable treatments. Further research is needed
116 D. Sebag-Montefiore and R. Glynne-Jones

in translational molecular biology, pharmacog- 6. Gerard JP, Conroy T, Bonnetain F, Bouche O, Chapet
enomics and biological imaging to identify O, Closon-Dejardin MT et al (2006) Preoperative
radiotherapy with or without concurrent fluorouracil
patients either at excess risk of radiation-related and leucovorin in T3–4 rectal cancers: results of
toxicity or whose cancers are more likely to FFCD 9203. J Clin Oncol 24(28):4620–4625
benefit from preoperative radiation and/or 7. Bosset JF, Collette L, Calais G, Mineur L, Maingon P,
fluoropyrimidine chemotherapy. Radosevic-Jelic L et al (2006) Chemotherapy with
preoperative radiotherapy in rectal cancer. N Engl J
Med 355(11):1114–1123
8. Sauer R, Becker H, Hohenberger W, Rodel C,
Wittekind C, Fietkau R et al (2004) Preoperative ver-
sus postoperative chemoradiotherapy for rectal can-
References cer. N Engl J Med 351(17):1731–1740
9. Valentini V, van Stiphout RG, Lammering G,
1. Swedish Rectal Cancer Trial (1997) Improved sur- Gambacorta MA, Barba MC, Bebenek M et al (2011)
vival with preoperative radiotherapy in resectable rec- Nomograms for predicting local recurrence, distant
tal cancer. Swedish Rectal Cancer Trial. N Engl J Med metastases, and overall survival for patients with locally
336(14):980–987 advanced rectal cancer on the basis of European ran-
2. Kapiteijn E, Marijnen CA, Nagtegaal ID, Putter H, domized clinical trials. J Clin Oncol 29(23):3163–3172
Steup WH, Wiggers T et al (2001) Preoperative radio- 10. Bujko K, Nowacki MP, Nasierowska-Guttmejer A,
therapy combined with total mesorectal excision for Michalski W, Bebenek M, Kryj M (2006) Long-term
resectable rectal cancer. N Engl J Med 345(9):638–646 results of a randomized trial comparing preoperative
3. Sebag-Montefiore D, Stephens RJ, Steele R, Monson J, short-course radiotherapy with preoperative conven-
Grieve R, Khanna S et al (2009) Preoperative radio- tionally fractionated chemoradiation for rectal cancer.
therapy versus selective postoperative chemoradiother- Br J Surg 93(10):1215–1223
apy in patients with rectal cancer (MRC CR07 and 11. Stephens RJ, Thompson LC, Quirke P, Steele R,
NCIC-CTG C016): a multicentre, randomised trial. Grieve R, Couture J et al (2010) Impact of short-
Lancet 373(9666):811–820 course preoperative radiotherapy for rectal cancer on
4. Tiefenthal M, Nilsson PJ, Johansson R, Glimelius B, patients’ quality of life: data from the medical research
Pahlman L (2011) The effects of short-course preop- council CR07/National Cancer Institute of Canada
erative irradiation on local recurrence rate and sur- Clinical Trials Group C016 randomized clinical trial.
vival in rectal cancer: a population-based nationwide J Clin Oncol 28(27):4233–4239
study. Dis Colon Rectum 54(6):672–680 12. Tiv M, Puyraveau M, Mineur L, Calais G, Maingon P,
5. Colorectal Cancer Collaborative Group (2001) Bardet E et al (2010) Long-term quality of life in
Adjuvant radiotherapy for rectal cancer: a systematic patients with rectal cancer treated with preoperative
overview of 8,507 patients from 22 randomised trials. (chemo)-radiotherapy within a randomized trial.
Lancet 358(9290):1291–1304 Cancer Radiother 14(6–7):530–534
Should We Tailor the Delineation
of Pelvic Structures According 13
to Tumor Presentation?

Maria Antonietta Gambacorta and Vincenzo Valentini

Contents For decades, radiotherapy has played a funda-


13.1 Contouring Guidelines .............................. 118 mental role in promoting local control in rectal
cancer. Local control was further increased with
13.2 Local Recurrences...................................... 118
the addition of total mesorectal excision (TME)
13.3 Guidelines Proposal ................................... 121 to radiotherapy as demonstrated in randomized
13.4 CTV Definition ........................................... 125 trials [1–4].
Despite the current use of 3D techniques to
References ................................................................. 126
plan the radiotherapy treatment, the field
definition to encompass the PTV is still often
driven by the pelvic bone anatomy visible on
simulation films, DRRs, or a combination of con-
ventional technique and CT contouring–based
planning [2–6]. The main reason for this could
result from the differences in training and in
interpretation of CTV contouring between radia-
tion oncologists, which leads to large systematic
error, up to a standard deviation of 1 cm, as dem-
onstrated in some reports [7, 8].
This could be also one of the reasons why,
although the “horseshoe” shape of the PTV rep-
resents an ideal target to be treated with IMRT
resulting in a clear advantage in sparing the small
bowel and bladder, the use of this new technique
is far to be adopted into daily practice [9].
Moreover, IMRT could provide a differentiation
of dose prescription and distribution into the pel-
vic volume; for that reasons, CTV should include
subsites at different risks of local recurrence
according to tumor presentation and location.
Following the introduction of 3D radiotherapy
M.A. Gambacorta (*) • V. Valentini techniques, guidelines for CT contouring of pel-
Department of Radiotherapy,
vic nodes at risk in rectal cancer were published
Università Cattolica Sacro Cuore, Rome, Italy
e-mail: magambacorta@rm.unicatt.it; and resulted in decreased interobserver variabil-
vvalentini@rm.unicatt.it ity in CTV delineation [10–12]. However, patients

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 117


DOI 10.1007/978-3-642-25005-7_13, © Springer-Verlag Berlin Heidelberg 2012
118 M.A. Gambacorta and V. Valentini

with rectal cancer continue to be treated with The presacral space represents the area where
standard pelvic radiotherapy fields irrespective to the majority of local relapses of all rectal cancer
tumor presentation at diagnosis. are concentrated. All studies analyzing rate and
site of local recurrences in rectal cancer described
the majority of them located in the posterior pel-
13.1 Contouring Guidelines vis. Although decreased after the introduction of
TME, the presacral space remains the first area of
After the introduction of conformal radiotherapy, local recurrence in rectal cancer; long-term
recommendations for contouring were published; results from Dutch-TME trial show the presacral
the most followed are essentially two: one from space as the first area of local recurrence in both
Europe and the other from the USA [10, 11]. randomization groups: RT + TME 2% versus
In the European guidelines, provided by Roels TME 3.6% [13]. The mechanism of occurrence
and coworkers, five different pelvic subsites were of presacral recurrence is puzzling: the presacral
identified according to the analysis of the inci- space is the easiest plane of dissection during
dence and site of pelvic relapses in rectal cancer: surgery, is always included in the radiotherapy
• Mesorectal subsite (MS) encompassing the field, is often boosted when long-course radio-
entire mesorectum and the mesorectal fascia therapy is delivered, and develops despite pre-
• Posterior pelvic subsite (PPS) corresponding sacral IORT is delivered. A recent anatomical
to the presacral space analysis did not find lymphatic tissues in the pre-
• Inferior pelvic subsite (IPS) corresponding to sacral space. Different mechanisms for local
the ischio-rectal fossae relapse were supposed other than lymph node
• Lateral pelvic subsite (LPS) encompassing to involvement: tumor cell spillage from positive
the extramesorectal nodes corresponding to margins, although presacral recurrence is com-
the obturator an internal iliac nodes mon even in patients with negative margins, and
• Anterior pelvic subsite (APS) corresponding seedings from tumor cells in the lateral spaces
to all pelvic organs that are located ventrally during surgical manipulation [14].
from the MS The mesorectum with its fascia must always
Anatomical boundaries for CT delineation be included in the CTV delineation.
were defined for each subsite. The mesorectum is the adipose tissue, bound-
The US guidelines, proposed by RTOG, devel- ing the mesorectal fascia, which posteriorly and
oped three elective CTVs for the contouring of rec- laterally surrounds the extraperitoneal rectum. It
tal and anal cancers, based on a consensus process: contains the neural and lymphovascular structures
• CTV A defined as the presacral, perirectal, and and is the first site of lymph node involvement in
internal iliac nodes rectal cancer. Surgical series analyzing mesorec-
• CTV B defined as the external iliac nodes tal lymph node involvement showed at least one
• CTV C defined as the inguinal nodes positive lymph node in this area in 46% of patients
Anatomical boundaries for CT delineation [10]. After the introduction of TME, the inci-
were defined for each CTV. dence of recurrence in this site is very low.
However, the analysis of radiological exams of
patients with local recurrence after TME showed a
13.2 Local Recurrences 50% of presence of perirectal fat below the anasto-
mosis, suggesting a not complete mesorectal exci-
Data reporting the patterns of local recurrence in sion although TME surgery was declared by the
rectal cancer, in the TME era, are scarce and con- surgeon; moreover, the majority of patients having
tradictory, leading to confusion regarding the a local relapse at the anastomosis had residual
definition of radiotherapy treatment volumes. mesorectal fat, suggesting a possible increased
Anyway, the following separated areas at risk rate of anastomotic recurrence due to the residual
could be identified. mesorectal fat left behind after surgery [15].
13 Should We Tailor the Delineation of Pelvic Structures According to Tumor Presentation? 119

This supposition is also supported by the long- laterally to the parietal fascia of the mesorectum
term results of the Dutch-TME trial in which the which is usually not removed with the TME sur-
rate of recurrence was higher in patients with pos- gical procedure [14]. This supports the impor-
itive nodes in the TME arm, when the distal mar- tance of the inclusion of the entire mesorectal
gin was 2 cm or lower; in the RT arm, the rate of fascia (visceral and parietal) in the treatment vol-
local recurrence was low except when the distal umes. Further supporting this is the fact that the
margin was less than 5 mm. Data from pathologi- higher tumor regression achievable with preop-
cal specimens showed a presence of positive erative long-course chemoradiation compared
lymph nodes or tumor deposits in the perirectal fat with short-course radiotherapy could be due to
distal to the tumor between 6.4% and 20.2% and the sterilization of the mesorectal fascia, allow-
usually located within 2–3 cm from the cancer. A ing a TME with clear CRM [18].
higher probability of distal tumor deposits when More controversial is the decision to include
positive mesorectal nodes are present may explain the lymph nodes located in the lateral spaces.
the higher incidence of local relapses in the TME A revision of the sites of local recurrence after
arm of the Dutch trial [13]. For the reasons men- the introduction of TME recommends reducing
tioned above, the mesorectum should always be the volume of treatment due to the higher rate of
included in the CTV of patients with rectal cancer recurrence in the posterior pelvis and the anasto-
treated with preoperative radiotherapy. mosis (overall 68% and as high as 89% when
The involvement of CRM has been demon- excluding cT4 at diagnosis which was significantly
strated to be a highly independent predictor of associated with anterior recurrence) [19]. These
both local control and survival; its positivity is data are consistent with a recently published
well predicted by preoperative MR. The impor- study which demonstrated that radiological evi-
tance of inclusion of the entire mesorectal fascia dence of recurrence in the lateral lymph nodes is
in the CTV, even when negative, is based on a well below 5% [15]. However, these results could
subset of patients from a Japanese surgical study be influenced by classification of the site of local
reported by Moriya et al. [16]. In this trial, relapse on the radiologic images used in that
patients with pathologic positive nodes who study. This finding, together with the observation
underwent lateral lymph node dissection com- that the majority of local recurrences, even after
bined with resection of the autonomic nervous preoperative radiotherapy, are presacral or anas-
system, located along the parietal mesorectal fas- tomotic, supports the idea to limit the radiother-
cia, on the affected site had a significant better apy field to the central areas at higher risk of
DFS compared to patients with positive lateral recurrence, at least in selected cases with both
nodes who underwent lateral dissection with negative mesorectal lymph nodes and CRM [20].
preservation of the autonomic nervous system Conversely, the patterns of local recurrence in the
(53% vs. 27%). These data suggest the presence update from the Dutch-TME trial revealed the
of microscopic residual tumor remaining on presacral space as the first site of relapse in
the parietal fascia following surgery. Recently, both arms, a lower anastomotic recurrence in
Nagtegaal reported increased local recurrence the radiotherapy arm, and lateral recurrences
rates, distant metastases, and a 50% decrease in accounted for about 20% of all local recurrences
survival in patients with close or positive circum- [13]. These data are consistent with those reported
ferential radial margins (CRM) after TME as in surgical series, in which patients with low-
well as the presence of tumor cells in the radial lying tumors have approximately 10–25% [21–
margin far from the macroscopic tumor invasion 25] positive lateral lymph nodes at diagnosis.
[17]. These data are further supported by the Furthermore, as high as 40% of patients with
autopsy data from pelvic dissection in the ten involved extramesorectal nodes developed local
fetuses which confirmed the presence of lym- recurrence. The rate of local recurrence was
phatic tissue around the medial part of the infe- related to the number (³4) and the site (obturator
rior hypogastric plexus, anatomically located area) of positive lateral nodes [21].
120 M.A. Gambacorta and V. Valentini

Bilateral lymph node dissection, although not a


routinely practiced in the Western world, has a
lower risk of local relapse compared to a unilat-
eral dissection (14% vs. 33%) [26]. This finding,
together with the fact that in a unilateral lymph
node dissection the site of local relapse is not
always located in the undissected lateral side
wall, suggests that the presence of lateral nodes
influences the appearance of local recurrence
despite that there is no anatomical correlation
between the local relapse and the lateral lymph b
nodes. In addition, it is possible that the removal
of lateral lymph nodes also removes microscopic
tumor cells which are in transit in the lateral
lymph nodes route, which could otherwise
infiltrate the surgical wound [26]. A recent exam-
ination of ten serially sectioned human fetal pel-
vises provides the anatomic support to this
hypothesis, demonstrating a connection between
the mesorectal and extramesorectal lymph nodes
located below the peritoneal reflection through
the middle rectal vessels and the autonomic ner-
vous system [14].
Fig. 13.1 Autoptic pelvic dissection: (a) anatomy; (b)
These results may be useful with the definition
lateral lymph nodes grouping
of radiotherapy fields. In fact, some studies com-
paring lateral lymph node dissection (LLND) and
preoperative radiotherapy showed overlapping spread of pelvic malignancies. Each lateral pelvic
local recurrence and survival in both groups. The space can be divided into three volumes to group
surgically treated patients had inferior genitouri- the nodes identified by Mangan, according to
nary and sexual function, suggesting a beneficial their proximity to the vessels or the nerves: the
effect of radiotherapy in the sterilization of can- internal iliac space including nodal groups 6–9,
cer cells in the lateral lymph nodes and justifying obturator space including group 5, and the iliac
the irradiation of extramesorectal nodes [27, 28]. space including groups 2–4 [30] (Fig.13.1).
Yu et al. examined the pattern of local recur- Anatomically, the internal iliac nodes collect the
rence and the correlation of the site with the lymphatic drainage of the rectum, whereas the
radiotherapy fields and showed that 45% of obturator nodes and the external iliac nodes drain
relapses were either marginal or out of the field, the lymphatic flow of the anterior pelvic organs.
mostly located (about 60%) beyond the anterior Data from surgical series indicate that rectal can-
and superior border of the field in a site corre- cer located below the peritoneal reflection may
sponding to superior-anterior mesorectum area also drain in the obturator nodes (9%) [31].
and anterior obturator nodes area [29]. The rate of involvement of external iliac nodes
Lateral lymph nodes include different node in rectal cancer is below 5%, and they should be
stations. The nomenclature of the lymph node included in tumors with a massive infiltration of
groups draining the pelvic organs was derived the anterior pelvic organ which drains primarily
from gynecological classification proposed by in the external iliac chain [10]. Moreover, as for
Mangan et al. This system indicates nine major seminoma in which has been seen that the pres-
groups of nodes that provide lymph drainage of ence of positive lymph nodes may cause a rever-
the pelvic organs and may be involved in the sion of the lymphatic flow, the inclusion of
13 Should We Tailor the Delineation of Pelvic Structures According to Tumor Presentation? 121

The inferior pelvic subsite, as described by


Roels, consists of the anal triangle of the
perineum containing the sphincter complex with
the surrounding perianal and perirectal space.
The analysis of the literature showed a rate of
recurrence in this site of 4%, being 8% for tumors
located <6 cm from the anal verge and 11% when
an APR was performed. The authors recommend
its inclusion in the target volume in case of low-
seated tumors (<6 cm from the anal verge) and
when an APR is required [10]. However, there is
no consensus in the literature about this issue
[34]. RTOG guidelines for anal-rectal malignan-
cies recommend to involve this site only when a
tumor infiltration in the ischio-rectal fossa is
Fig. 13.2 MR of a rectal cancer with a massive infiltration present, adding 1–2 cm margin up to bone; in all
of the left internal iliac node
other cases, RTOG guidelines recommend to
limit the CTV to few millimeters beyond the
external iliac nodes should be considered when levator muscles [11]. Guidelines for rectal can-
massive obturator nodes are present at diagnosis cer surgery suggest 1 cm distal margin to be
(Fig. 13.2). adequate for low-seated tumors. The intramural
Concern remains about the preoperative subclinical spread in the bowel rarely occurs
identification of nodal disease. Magnetic reso- beyond 1–1.5 cm below the macroscopic caudal
nance (MR) which uses morphological criteria edge and, when it happens, is usually associated
such as border contour and signal intensity with advanced tumor, with aggressive histology,
appears to be superior in the evaluation of lymph features all predicting a poor prognosis. Surgery
nodes compared to CT which uses only a size cri- with longer distal margin seems not to improve
teria. However, MR is not conclusive in the pre- outcomes. Moreover, levators constitute a barrier
operative identification of nodal disease when the against the cancer spread, no nodes are present in
node size is below 8 mm [32]. For that reason, it the ischio-rectal fossa, and recurrences in this
is difficult to preoperatively identify patients who site were reported only after abdomino-perineal
can benefit from radiotherapy fields extended to resection. From the above considerations, in
the lateral spaces. Preliminary reports of MRI tumors without infiltration of the sphincter com-
diffusion-weighted imaging (DWI) in combina- plex, the inferior CTV should be limited to
tion with T(2)-weighted imaging (T(2)WI) for 1.5 cm from the caudal edge of the macroscopic
the detection of rectal cancer as compared with tumor [35].
T(2)WI alone provide better identification of rec-
tal cancer and local nodes [33]. Ongoing research
will clarify the role of this imaging modality. 13.3 Guidelines Proposal
Data from surgical series suggest that the low
location of the tumor (<4 cm from the anal mar- Based on the above information, we propose to
gin), undifferentiated grade 3 tumors, positive identify seven different areas at risk to be con-
mesenteric and mesorectal nodes, and tumor toured in rectal cancer: mesorectum (M), pre-
stage (T3–T4) all significantly correlate with the sacral space (PS), internal iliac nodes (IIN),
lateral lymph node positivity [24]. These factors obturator nodes (ON), external iliac nodes (EIN),
should be used in the radiotherapy decision mak- sphincter complex (SC) inferior pelvic subsite
ing to define areas at risk to be included in the (IPS) corresponding to the ischio-rectal fossae
pelvic volume. (Table 13.1) (Figs. 13.3 and 13.4) [36].
122

Table 13.1 Guidelines for pelvic lymph node contouring in rectal cancer
Cranial Caudal Anterior Posterior Medial Lateral
Mesorectum Bifurcation of the Insertion of the levator Superior: Anterior limit Superior: Mesorectal Superior: Mesorectal
inferior mesenteric ani muscle into the rectal of superior rectal vessels fascia in front of sacral fascia/internal and
artery (IMA) in sigmoid wall/disappearing of or a virtual line between concavity external Iliac lymph
and superior rectal artery mesorectal fat tissue the anterior aspect of node area
around the rectum internal iliac vessels of
both sides
Inferior: Bladder, Inferior: Levator ani Inferior: Levator ani
prostate/seminal vesicle muscle muscle
in man, vaginal wall/
uterus in woman
Presacral space Sacral promontory Coccyx Superior: 1 cm in front Sacral concavity Lateral border of the
(posterior pelvic of the bone sacrum
subsite) Mid-inferior: Presacral
fascia/posterior
mesorectal fascia
Internal iliac nodes Bifurcation of common Ending of the mesorec- Superior: Behind the Lateral edge of the Mesorectal Superior: Psoas
(lymph node regions) iliac artery into internal tum /appearance of external iliac vessels sacro-iliac joint fascia, pelvic muscle and ileum
and external iliac arteries ischio-rectal fossae Mid-inferior: Behind Pyriform muscle organs Inferior: Internal
(bony reference L5–S1) obturator nerve obturator muscle
Obturator nodes (lymph Caudal border of The entrance of the Posterior aspect of the Posterior aspect of the Mesorectal Internal obturator
node regions) sacro-iliac joint obturator nerve/artery in external iliac vessels obturator nerve fascia, pelvic muscle and ileum
the obturator canal organs
External iliac nodes Bifurcation of common The start of the femoralSuperior-mid: 0.7 cm Posterior aspect of the Mesorectal Psoas muscle, iliac
(lymph node regions) iliac artery into internal vessels from the vessel external iliac vein fascia, pelvic muscle
and external iliac arteries Inferior: Abdominal wall organs
(bony reference L5–S1) muscles
Sphincter complex 1.5 cm around the internal and external anal sphincters
Inferior pelvic subsite Levator ani muscle Skin Obturator muscle A virtual line between Anal canal Obturator muscle and
(ischio-rectal fossae) the posterior profile of gluteus muscle
the gluteus muscle of
both sides
M.A. Gambacorta and V. Valentini
13 Should We Tailor the Delineation of Pelvic Structures According to Tumor Presentation? 123

MF

a b
MF

ONe GMM
PM

EIV
EIA
IM

c d
MF

GMM

IOM

e f

g h

Fig. 13.3 Anatomic atlas for rectal cancer contouring. muscle (c–d); B: bladder (f); IOM: internal obturator mus-
IPM: ileopsoas muscle (a–b); *: superior rectal artery (b); cle (f); GMM: gluteus major muscle (a–h) PS: yellow
MF: Mesorectal fascia (b–f); ONe: obturator nerve (c–d); (a–e); EIN: green (a–e); IIN: blue (a–f); M: brown (b–g);
EIV: external iliac vein (b-e);, EIA: external iliac artery ON: pink (c–f); IPS: dotted blue (g); SC: dotted yellow
(b–e); PM: pyriform muscle (c–d); U: uterus (e); IM: Iliac (h) (Images by the courtesy of TIGER project)

Guidelines have the advantage to standardize the uterus/vagina, prostate) are present; however, in
target volume delineation; however, some uncer- the superior pelvis, its anterior part has not an ana-
tainties still remain and are hard to be solved [8]. tomical boundary. We suggest to enclose all the
The mesorectum is easy to identify in the mid- superior rectal vessels when visible, since several
low pelvis where the pelvic organs (bladder, times, lymph nodes can be visible in this area
124 M.A. Gambacorta and V. Valentini

GMM

IPM *
IPM

a b
MF
GMM
GMM PM
PM ONe

EIV U
U
IM EIA IM

c d
MF

GMM ONe
GMM

U IOM

B
GMM

e f

GMM GMM

g h

Fig. 13.4 CT Atlas for rectal cancer contouring. IPM: (c–d); B: bladder (f) IOM: internal obturator muscle (f);
ileopsoas muscle (a–b); * superior rectal artery (b); MF: GMM: gluteus major muscle (b–h) PS: yellow (a–f); EIN:
Mesorectal fascia (c–f); ONe: obturator nerve (c–f); EIV: green (a–e); IIN: blue (a–f); M: brown (b–g); ON: pink
external iliac vein (c); EIA: external iliac artery (c); PM: (c–f); IPS: dotted blue (g); SC: dotted yellow (h)
pyriform muscle (c–d); U: uterus (c–d); IM: Iliac muscle

(Figs. 13.3b and 13.4b), which corresponds usu- The internal iliac nodes, in the mid-low pelvis,
ally with the anterior limit of the lateral nodes. have not a corresponding visible vessel on CT.
The presacral space is so tiny that it is often We suggest to contour the triangular lymphovas-
not visible on CT scan or overlaps other areas at cular fat area located between the pelvic wall and
risks. We suggest to enclose it in mesorectum. the mesorectum.
13 Should We Tailor the Delineation of Pelvic Structures According to Tumor Presentation? 125

Table 13.2 Target volume delineation according to tumor stage and location
Presacral Internal Obturator External Sphincter Ischio-rectal
space Mesorectum iliac nodes nodes iliac nodes complex fossae
cT3 high (above + + +
the peritoneal
reflection)
cT3 mid-low + + + + + (when anal + (when direct
(at the peritoneal canal tumor
reflection) invasion) infiltration)
Any cT with + + + + + (when anal + (when direct
massive positive canal tumor
internal iliac invasion) infiltration)
nodes
Any cT with + + + + + + (when anal + (when direct
massive positive canal tumor
obturator nodes invasion) infiltration)
cT4 with for + + + + + + (when anal + (when direct
anterior pelvic canal tumor
organ invasion) infiltration)

The obturator nerve used as posterior limit of rectal cancers located at >1.5 cm from the from
the obturator nodes could seem difficult to iden- the anal-rectal ring, the lower border should be
tify; however, although very tiny, it may be fol- limited at the end of the mesorectum; (2) in
lowed, scrolling CT images, as a straight structure tumors located £1.5 cm from the anal-rectal ring,
located behind the external iliac vessels, through the lower border should extend to encompass
the whole pelvis (Fig. 13.4c–f). 1 cm of the anal canal; (3) in tumors extending
The external iliac nodes are the easiest to through the anal canal, all the anal canal and the
identify, thanks to the external iliac arteries and sphincter complex should be included; although
veins well visible on CT scan (Figs. 13.3c and in the literature the margin around the sphincter
13.4c). We suggest to start from this structure for complex is not indicated, we recommend
the contouring of the lateral lymph nodes. 5–10 mm of fat around it; (4) when the rectal
fossa is directly infiltrated by the tumor, both the
ischio-rectal fossae should be enclosed.
13.4 CTV Definition Inguinal nodes are not the target in rectal can-
cer; however, when most of the anal canal or the
The inclusion of lateral nodes in the pelvic target ischio-rectal fossa is infiltrated in both these cases
volume for preoperative radiotherapy should be also, they should be part of the CTV.
personalized according to tumor stage, tumor The introduction of the IMRT for the treat-
site, and lymph node location as follows ment of rectal cancer requires a proper and shared
(Table 13.2): always include the primary tumor, definition of the volumes of interest on CT
the entire mesorectum, presacral space, and inter- images; moreover, a careful center-based evalua-
nal iliac nodes in all T3 tumors. In addition, the tion of the target motion results is fundamental
obturator nodes should be added for mid-low- for the evaluation of the correct ITV to avoid tar-
located T3 tumors (< at or below the peritoneal get missing [37].
reflection) or in high T3 with massive positive
IIN. In T4 tumors, with anterior organ invasion, Conclusions
or in T3 tumors with massive obturator node Although agreement exists on the areas at risk
involvement, the addition of the external iliac for local recurrence to be included in the CTV,
nodes is recommended [11, 34] (Fig. 13.5). however, this is not the case for the contouring
The lower edge of the CTV may be modulated boundaries. In this chapter, guidelines for the
according to tumor extension and invasion: (1) in contouring of the pelvic subsites in rectal
126 M.A. Gambacorta and V. Valentini

a c

b d

Fig. 13.5 (a) CTV_T3 mid-low; (b) PTV_T3; (c) CTV_T4; (d) PTV_T4

cancer are proposed to increase the confor- tal cancer (MRC CR07 and NCIC-CTG C016): a mul-
mity in volumes definition among different ticentre, randomized trial. Lancet 373:811–820
3. Bosset JF, Collette L, Calais G et al (2007)
radiation oncologists, to decrease uncertain- Chemotherapy with preoperative radiotherapy in rec-
ties when more conformal technique of irra- tal cancer. N Engl J Med 355:1114–1123
diation such as IMRT are used, to modulate 4. Gerard JP, Conroy T, Bonnetain F et al (2006)
volume definition according to tumor presen- Radiotherapy with or without concurrent fluorouracil
and leucovorin in T3–4 rectal cancers: results of
tation (stage and location), and to facilitate FFCD 9203. J Clin Oncol 24:4620–4625
training of radiation oncologists in training. 5. Gunderson LL, Russell AH, Llewellyn HJ et al (1985)
Treatment planning for colorectal cancer: radiation
and surgical techniques and value of small-bowel
films. Int J Radiat Oncol Biol Phys 11:1379–1393
6. Widder J, Sedlmayer F, Stanek C, Potter R (2000)
References Quality assurance in preoperative radiotherapy of rec-
tal cancer: evaluation of pre-trial dummy-run.
1. Peeters KC, Marijnen CA, Nagtegaal ID et al (2007) Radiother Oncol 56:341–347
Dutch colorectal cancer group the TME trial after a 7. Nijkamp J, de Haas-Kock DFM, Beukema JC et al
median follow-up of 6 years: increased local control (2011) Target volume delineation variation in radio-
but no survival benefit in irradiated patients with therapy for early stage rectal cancer in the Netherlands.
resectable rectal carcinoma. Ann Surg 246:693–701 Radiother Oncol 102:14–21, Epub 2011 Sep 6
2. Sebag-Montefiore D, Stephens RJ, Steele R et al 8. Fuller CD, Nijkamp J, Duppen JC et al (2010)
(2009) Preoperative radiotherapy versus selective Prospective randomized double-blind pilot study of
postoperative chemoradiotherapy in patients with rec- site-specific consensus atlas implementation for rectal
13 Should We Tailor the Delineation of Pelvic Structures According to Tumor Presentation? 127

cancer target volume delineation in the cooperative preoperative chemo-radiotherapy for the treatment of
group setting. Int J Radiat Oncol Biol Phys advanced middle to lower rectal cancers. Int J
79:481–489 Colorectal Dis 19:188–194
9. De Ridder M, Tournel K, Van Nieuwenhove Y et al 23. Ueno H, Mochizuki H, Hashiguchi Y et al (2007)
(2008) Phase II study of preoperative helical tomo- Potential prognostic benefit of lateral pelvic node dis-
therapy in the preoperative treatment for rectal cancer. section for rectal cancer located below the peritoneal
Int J Radiat Oncol Biol Phys 70:728–734 reflection. Ann Surg 245:80–87
10. Roels S, Duthoy W, Haustermans K et al (2006) 24. Ueno M, Oya M, Azekura K et al (2005) Incidence
Definition and delineation of the clinical target vol- and prognostic significance of lateral lymph node
ume for rectal cancer. Int J Radiat Oncol Biol Phys metastasis in patients with advanced low rectal can-
65:1129–1142 cer. Br J Surg 92:756–763
11. Myerson RJ, Garofalo MC, El Naqa I et al (2009) 25. Kim TH, Jeong SY, Choi DH et al (2008) Lateral
Elective clinical target volumes for conformal therapy lymph node metastasis is a major cause of locore-
in anorectal cancer: a radiation therapy oncology gional recurrence in rectal cancer treated with preop-
group consensus panel contouring atlas. Int J Radiat erative chemoradiotherapy and curative resection.
Oncol Biol Phys 74:824–830 Ann Surg Oncol 15:729–737
12. Arcangeli S, Valentini V, Nori SL et al (2003) 26. Kusters M, van de Velde CJ, Beets-Tan RG et al (2008)
Underlying anatomy for CTV contouring and lym- Patterns of local recurrence in rectal cancer: a single-
phatic drainage in rectal cancer radiation therapy. center experience. Ann Surg Oncol 16:289–296
Rays 28(3):331–336 27. Kusters M, Beets GL, van de Velde CJ et al (2009)
13. Kusters M, Marijnen CAM, van de Velde CJH (2010) A comparison between the treatment of low rectal
Patterns of local recurrence in rectal cancer; a study of cancer in Japan and the Netherlands, focusing on the
the Dutch TME trial. Eur J Surg Oncol 36:470–476 patterns of local recurrence. Ann Surg 249:229–235
14. Kusters M, Wallner C, Lange MM et al (2010) Origin 28. Nagawa H, Muto T, Sunouchi K et al (2001)
of presacral recurrence after rectal cancer treatment. Randomized, controlled trial of lateral node dissec-
Br J Surg 97:1582–1588 tion vs. nerve-preserving resection in patients with
15. Syk E, Torkzad MR, Blomqvist L et al (2008) Local rectal cancer after preoperative radiotherapy. Dis
recurrence in rectal cancer: anatomic localization and Colon Rectum 44:1274–1280
effect on radiation target. Int J Radiat Oncol Biol Phys 29. Yu TK, Bhosale PR, Crane CH et al (2008) Patterns of
72:658–664 locoregional recurrence after surgery and radiother-
16. Moriya Y, Sugihara K, Akasu T et al (1997) Importance apy or chemoradiation for rectal cancer. Int J Radiat
of extended lymphadenectomy with lateral node dis- Oncol Biol Phys 71:1175–1180
section for advanced lower rectal cancer. World J Surg 30. Mangan CE, Rubin SC, Rabin DS et al (1986) Lymph
21:728–732 node nomenclature in the gynecologic oncology.
17. Nagtegaal ID, Quirke P (2008) What is the role for the Gynecol Oncol 23:222–226
circumferential margin in the modern treatment of 31. Steup WH, Moriya Y, van de Velde CJH (2002)
rectal cancer? J Clin Oncol 26:303–312 Patterns of lymphatic spread in rectal cancer. A topo-
18. Bujko K, Nowacki MP, Nasierowska-Guttmejer A graphical analysis on lymph node metastases. Eur J
et al (2006) Long-term results of a randomized trial Cancer 38:911–918
comparing preoperative short-course radiotherapy 32. Koh DM, Chau I, Tait D et al (2008) Evaluating
with preoperative conventionally fractionated chemo- mesorectal lymph nodes in rectal cancer before and after
radiation for rectal cancer. Br J Surg 93(10): neoadjuvant. Int J Radiat Oncol Biol Phys 71:456–461
1215–1223 33. Rao S-X, Zeng M-S, Chen C-Z et al (2008) The value
19. Hruby G, Barton M, Miles S et al (2003) Sites of local of diffusion-weighted imaging in combination with
recurrence after surgery, with or without chemother- T2-weighted imaging for rectal cancer detection. Eur
apy, for rectal cancer: implications for radiotherapy J Radiol 65:299–303
field design. Int J Radiat Oncol Biol Phys 55: 34. Bujko K, Bujko M, Pietrzak L (2007) Clinical target
138–143 volume for rectal cancer: in regard to Roels et al. (Int
20. Nijkamp J, Kusters M, Beets-Tan R et al (2011) J Radiat Oncol Biol Phys 2006;65:1129–1142). Int J
Three-dimensional analysis of recurrences patterns in Radiat Oncol Biol Phys 68:313
rectal cancer: the cranial border in the hypofraction- 35. Nelson H, Petrelli N, Carlin A et al (2001) Guidelines
ated preoperative radiotherapy can be lowered. Int J 2000 for colon and rectal cancer surgery. J Natl Cancer
Radiat Oncol Biol Phys 80(1):103–110 Inst 93:583–592
21. Sato H, Maeda K, Maruta M et al (2006) Who can get 36. Valentini V, Dinapoli N, Nori S et al (2004) An appli-
the beneficial effect from lateral lymph node dissec- cation of visible human database in radiotherapy:
tion for Dukes C rectal carcinoma below the peritoneal tutorial for image guided external radiotherapy
reflection? Dis Colon Rectum 49(10 Suppl):S3–S12 (TIGER). Radiother Oncol 70(2):165–169
22. Koda K, Saito N, Oda K, Takiguchi N et al (2004) 37. Ippolito E, Mertens I, Haustermans K et al (2008)
Evaluation of lateral lymph node dissection with IGRT in rectal cancer. Acta Oncol 47:1317–1324
What Is the Role of IMRT and IGRT
in Rectal Cancer? 14
Jasper Nijkamp, Karin Haustermans,
and Corrie A.M. Marijnen

Contents 14.6 IGRT ........................................................... 138


14.1 Introduction ................................................ 129 14.7 Geometric Uncertainties............................ 138
14.1.1 The Good ..................................................... 130 14.7.1 Set-up Errors ................................................ 138
14.1.2 The Bad ........................................................ 130 14.7.2 Target Volume Shape Variation.................... 138
14.1.3 The Ugly ...................................................... 130 14.7.3 Planning Target Volume Margins
to Account for Geometric Uncertainties ...... 139
14.2 Evolution of Radiotherapy Treatment 14.7.4 Correcting Shape Variation .......................... 141
Technique in Rectal Cancer ...................... 130
14.8 Response Evaluation .................................. 142
14.3 Benefits of IMRT ........................................ 132
14.9 Possibility of Dose Escalation.................... 143
14.4 Prerequisites for IMRT ............................. 133
14.4.1 Target Volume Definition ............................. 133 References ................................................................ 145
14.4.2 Target Volume Delineation .......................... 133
14.5 Measures to Reduce Irradiated
Small Bowel Volume .................................. 134
14.5.1 Full Bladder Protocol ................................... 134
14.5.2 Patient Orientation and Belly Board Use ..... 136
14.5.3 Reduction of CTV........................................ 136
14.1 Introduction

Over the past decades, advances in multi-


modality treatment strategies have contributed
significantly to the improvement of outcome in
rectal cancer patients [1–6]. Decisions about the
J. Nijkamp therapeutic regimen are based on tumour charac-
Department of Radiation Oncology, The Netherlands teristics such as TNM stage and the involvement
Cancer Institute – Antoni van Leeuwenhoek Hospital,
of the mesorectal fascia (MRF), determined with
Amsterdam, The Netherlands
magnetic resonance imaging (MRI) and endo-
K. Haustermans
scopic ultrasonography (EUS) at time of diagno-
Department of Radiation Oncology,
Leuven Cancer Institute, sis and on post-operative pathological evaluation.
University Hospital Gasthuisberg, Based on these characteristics, rectal cancers can
Leuven, Belgium be divided into three groups with respect to their
e-mail: karin.haustermans@uz.kuleuven.ac.be
chances of developing either local and/or distant
C.A.M. Marijnen (*) recurrences: low, intermediate and high risk or
Department of Clinical Oncology,
the ‘good’, the ‘bad’ and the ‘ugly’ [7, 8]. In each
Leiden University Medical Center,
Leiden, The Netherlands group, different risks are at stake which chal-
e-mail: marijnen@lumc.nl lenges the determination of optimal treatment.

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 129


DOI 10.1007/978-3-642-25005-7_14, © Springer-Verlag Berlin Heidelberg 2012
130 J. Nijkamp et al.

14.1.1 The Good 14.1.3 The Ugly

The ‘good’ rectal cancers are T1 or T2, node- ‘Ugly’ tumours have features such as a T4 stage,
negative rectal cancers that show no bad prognos- extensive lymph node involvement and are cor-
tic factors on their MRI image. After TME related to a high risk of local recurrence and dis-
surgery, local recurrence rates in these patients tant metastases. Improvement in treatment can be
are very low and cure rates are high [9, 10]. More achieved by early prediction of response, allow-
experimental treatment options such as transanal ing quick adaptation of treatment such as
endoscopic microsurgery (TEM) with or without intensification of chemotherapy, dose escalation
preoperative chemoradiotherapy (CRT) or or the addition of targeted therapies.
chemoradiotherapy followed by a wait-and-see The above-mentioned approaches call for
policy are becoming more popular [11–15]. The improvement of the current possibilities. First of
major aim of these strategies is rectum preserva- all, we need appropriate imaging to select the
tion with the subsequent improved quality of life patients, to allow for preoperative risk indication.
[16]. The question is whether these approaches Second, we need strategies to select patients well
are safe and feasible with local recurrence rates responding to preoperative treatment, either to
varying between 5% and 28% for T1 patients and have them qualified for organ-saving strategies or
11–45% for T2 patients in the context without to adapt the chemotherapy in non-responding
preoperative treatment [11, 12]. Treatment with patients. We may need a higher dose to the gross
TEM surgery alone should only be considered in tumour volume to improve the complete response
those patients with the lowest risk of local recur- rates of patients selected for organ-saving proce-
rence. Since lymph nodes are not removed with dures. An integrated boost may be a way to
TEM surgery, a negative lymph node status is by accomplish this.
far the most important prognostic factor for the
local recurrence risk in patients treated with TEM
only. At present, a chance of lymph node involve-
ment of 6% is considered acceptable to proceed 14.2 Evolution of Radiotherapy
with local excision only. Treatment Technique in Rectal
Cancer

14.1.2 The Bad In ideal radiotherapy, 3D volumetric imaging


with high soft tissue contrast is used on which the
In general, ‘bad’ or intermediate risk rectal cancer gross tumour volume (GTV) and clinical target
patients are considered to have a large T3N0 tumour volume (CTV) can be delineated. Subsequently, a
or a T1–3N1 tumour and will be treated with pre- safety margin should be added to the CTV, creat-
operative (chemo)radiotherapy followed by TME ing a planning target volume (PTV) [18], which
surgery. Agreement exists that in case of a threat- accounts for geometric uncertainties during treat-
ened or involved mesorectal fascia, preoperative ment. Finally, dose delivery should fully conform
chemoradiotherapy is the treatment of choice. the PTV, assuring coverage of macroscopic and
Since the prognosis of rectal cancer is stage depen- microscopic disease, avoiding dose to the organs
dent, one can question whether T1/2–N1 and T3N0 at risk as much as possible.
patients benefit from preoperative treatment [4]. The planning and delivery technique of radio-
However, patient selection is hindered by imper- therapy has improved greatly over the past 30 years.
fections in staging [17]. In addition, for intermedi- In the early years, the radiotherapy dose was deliv-
ate-risk patients, the choice between preoperative ered using two opposed fields. With this technique,
chemoradiotherapy and short-course (SC) radio- a large square area received 100% of the dose,
therapy is still subject of debate. without any shaping of the dose according to the
14 What Is the Role of IMRT and IGRT in Rectal Cancer? 131

Fig. 14.1 Example of a three-field conformal plan (left) outlined in dark blue, the PTV in aqua and small bowel in
and a seven-field IMRT plan (right) on the same patient in yellow
transversal (top) and sagittal (bottom) view. The CTV is

PTV. An improvement in conforming the dose to Besides having a limited number of beam angles
the PTV was made with the introduction of three- (step and shoot), the IMRT can also be delivered
and four-field box techniques, although the lack in a continuous fashion during which the collima-
of visualization of the actual target volume tor, rotation speed and intensity can be modulated
resulted in field borders defined by bony anatomy (intensity-modulated arc therapy (IMAT)) [21].
on fluoroscopic images. Each beam was rectangle Each of the above-described improvements in
shaped, and the size was defined by the outer bor- radiotherapy delivery resulted in more confor-
ders of the PTV. With the introduction of com- mality to the PTV and, subsequently, lower dose
puted tomography (CT), delineation of the CTV, to the surrounding healthy tissues. Several options
3D treatment planning and the multi-leaf colli- are available to further improve radiotherapy.
mator, each beam could be adapted to the actual The required PTV margin is based on geometri-
shape of the PTV, resulting in conformal RT. cal uncertainties, which describe the differences
Nowadays, we have the ability to deliver the RT between how the radiation was planned and
dose using intensity-modulated radiotherapy the actual treatment situation [18]. With the
(IMRT), where typically seven beams from dif- introduction of image-guided radiotherapy
ferent angles are subdivided into segments, for (IGRT), geometrical uncertainties can be mea-
which the intensity is modulated. With IMRT, the sured and corrected during treatment, allowing
conformality and homogeneity of the dose to the for a reduction of the PTV margin. The acquired
PTV are further improved [19, 20] (Fig. 14.1). IGRT images also enable visualization of the
132 J. Nijkamp et al.

target volume during treatment, for example, 25] and the volume receiving more than 45
with cone beam CT scans, enabling better under- (V45) and 50 Gy (V50) [26, 27], are significantly
standing of the treatment situation. With the reduced using IMRT [6–9]. Recently, Samuelian
introduction of more sophisticated response et al. [28] compared 92 consecutive patients of
monitoring, such as with PET and diffusion- which 61 were treated with conformal RT (CRT)
weighted MRI [22], during treatment, the possi- and 31 were treated with IMRT. In the IMRT
bility of adapting treatment according to group, significantly less patients experienced
radiotherapy response has become possible. acute grade 2 or higher GI toxicity with 32% vs.
These techniques allow for tailoring of the treat- 62% in the CRT group, with differences espe-
ment for each patient, improving the efficacy. cially in diarrhoea (23% vs. 48%) and enteritis
In this chapter, we will address the implications (6% vs. 30%). Although these findings were not
of IMRT and IGRT for rectal cancer treatment as based on randomized phase III evidence, they
well as the possible pitfalls that one may encoun- do indicate that the theoretical dosimetric benefit
ter during introduction of those techniques. of IMRT does translate into a clinical benefit.
IMRT has also shown to be an excellent tech-
nique to deliver a boost to the GTV, simultane-
14.3 Benefits of IMRT ously delivering the standard dose to the CTV [19,
29, 30] without compromising the organ at risk
As mentioned before, CTV for radiotherapy of dose. The boost dose can be used to increase
rectal cancer generally consists of the tumour and down-staging, improve the ability to have an R0
involved lymph nodes (GTV), the mesorectum, resection and increase the percentage of patho-
the internal iliac lymph node regions and the pre- logic complete responders [31]. With 3D confor-
sacral lymph node region [23]. This definition of mal RT, it is almost impossible to deliver a
the CTV results in a horseshoe-shaped target vol- simultaneously integrated boost (SIB) without
ume when visualized in the transversal plane increasing the dose to the organs at risk. More
(Fig. 14.1). The challenge in radiotherapy is to often, the boost dose is delivered in a number of
deliver the dose to the target volume while mini- fractions after the standard dose is delivered, unfa-
mizing the dose to the surrounding organs at risk, vourably increasing the overall treatment time.
such as the small bowel and the bladder. The general advantage of IMRT is a more accu-
In current clinical practice, 3D conformal rate delivery of the dose to the target volume, espe-
three- or four-field dose delivery techniques are cially when the shape of the target differs from a
used most often. For each field, the shape of the sphere-like shape. Different tumour presentations
beam is adapted to the shape of the target volume will generally not result in different shape complex-
using a multi-leaf collimator, while using a uni- ities, and as a consequence, advantages of IMRT
form intensity for the field. With this type of treat- will be applicable to all tumour presentations. IMRT
ment, it is impossible to fully conform to the should therefore ideally be implemented for all rec-
horseshoe shape of the CTV, resulting in delivery tal cancer RT treatments. However, IMRT cannot
of dose to the organs at risk anterior of the CTV, be safely implemented without attention for a num-
especially the small bowel (Fig. 14.1). With IMRT, ber of difficulties that need to be solved.
it is possible to deliver more concave dose distri-
butions, which also have a more uniform dose dis-
tribution. During IMRT, typically seven or nine 14.4 Prerequisites for IMRT
fields from different gantry angles are used with
each field subdivided into different segments with 14.4.1 Target Volume Definition
each of their own intensity (Fig. 14.1).
Important parameters for prediction of acute As more conformality to the target volume is
and late bowel toxicity, such as the volume of reached with IMRT, knowledge and application
small bowel receiving 15 Gy or more (V15) [24, of geometrical uncertainties in the construction
14 What Is the Role of IMRT and IGRT in Rectal Cancer? 133

of a PTV is important to prevent underdosage of CTV and OAR delineation nationwide and estab-
the target volume. lish a learning curve by giving feedback [35].
The first step that needs to be taken for every Multi-modality imaging has been proven to
patient is defining the CTV. In general, the reduce target volume delineation variation in
tumour, involved lymph nodes, the mesorectum other tumour sites [36, 37]. For rectal cancer
and the perirectal, presacral and internal iliac target volume delineation, there is only one study
lymph node regions are advised to be included. In available evaluating the influence of FDG-PET/
rectal cancer RT, several guidelines are available CT imaging [38] (Fig. 14.3). The use of FDG-
as how to interpret these CTV definitions [23, 32, PET resulted in reduced variation for delineation
33]. Roels et al. [23] developed an atlas by first of the GTV. However, tumour cells might also be
selecting required subregions based on literature present outside this GTV and should therefore be
about patterns of local recurrences and subse- included in the CTV. So far, no reliable imaging
quently defining the anatomical borders of these of microscopic tumour cells is available, and
subregions of the CTV. The atlas of Myerson from this perspective, the FDG-PET has no addi-
et al. [32] was developed by an RTOG consensus tional value.
panel. The atlas of Nijkamp et al. [33] was based The value of the MRI in visualizing the
on the guidelines of Roels et al. [23] and was pro- tumour, mesorectal fat and mesorectal fascia is
vided digitally, such that users could approach undisputable. Despite this common knowl-
the atlas similar to approaching a CT scan at edge, delineation on MRI is not routinely done,
delineation. and studies demonstrating its superiority are
not available, except for delineation of the
GTV in low-seated tumours [39] (Fig. 14.4).
14.4.2 Target Volume Delineation Challenges in CTV delineation on MR for
treatment planning are in the image fusion with
Delineation variation of the CTV in rectal can- the planning CT. Problems may occur when
cer is known to be larger than set-up errors [33, large anatomic differences exist between both
34]. Interpretation differences of the CTV scans (Fig. 14.4).
between radiation oncologists can result in
CTV differences up to centimetres, resulting in 14.4.2.1 Small Bowel Toxicity
large systematic errors (up to a standard devia- One of the critical organs in treatment for rectal
tion (SD) of 1 cm) (Fig. 14.2). Delineation vari- cancer is the small bowel. Due to its mobility,
ation is not the same for all regions of the CTV. there is no easily defined maximum tolerated
Close to bony anatomy and muscles, variation dose. However, from literature it can be concluded
is in the order of 0.2–0.4 cm SD, while for the that the occurrence of toxicity is primarily depen-
upper anterior part, close to the bladder and dent on the amount of dose to the small bowel.
small bowel, variation up to 1 cm SD is mea- Baglan et al. [24] showed that a cut-off at
sured [33, 34]. 150 cc of small bowel receiving more than
Measures to reduce delineation variation are 15 Gy divided a group of 40 patients into 0%
the use of guidelines and delineation atlases, the and 50% chance of developing acute grade 3
use of central reviewing experts and addition of diarrhoea. This was later confirmed in a larger
other modality scans such as MR and PET to study by Robertson et al. [25], who showed that
increase the anatomic and functional information. a cut-off point of 120 cc and 15 Gy divided the
Guidelines and a delineation atlas can reduce group into 9% vs. 38% risk of developing grade
the delineation variation significantly down to 3 diarrhoea.
approximately 0.7 cm SD [33]. For late toxicity, Gallagher et al. [26] found
In the ongoing Belgium PROCARE radio- already in 1986 a critical level of 45 Gy to 78 cc
therapy reviewing project, CTV delineations are and 50 Gy to 17 cc of the small bowel predictive
reviewed in a central expert institution to monitor in post-operative RT. Letschert et al. [27] showed
134 J. Nijkamp et al.

Fig. 14.2 Example images of


a patient for which the CTV
was delineated by 11 radiation
oncologists, showing the
difference in delineation
variation between delineation
based on local hospital policy
(left) and delineation using a
consensus guideline and
digital delineation atlas
(right). The top image is the
most caudal common
delineated slice. The middle
image is a mid-rectum slice
just cranial of the bladder. The
bottom image is the sagittal
view. Each observer has a
corresponding colour on all
images

afterwards, at a dose range of 40–50 Gy, that a 14.5 Measures to Reduce Irradiated
small bowel volume increase by a factor of 2 Small Bowel Volume
demands for a total dose reduction of 17% for the
same complication rate. 14.5.1 Full Bladder Protocol
Besides the earlier-described effect of using
IMRT instead of conformal RT, several additional Due to the definition of the CTV up to the bifurca-
measures can be taken to further reduce the irra- tion of the common iliac artery into the internal and
diated small bowel volume. In the section below, external iliac artery, there is a high probability that
an overview of all measures and their possible small bowel will be located anterior of the CTV. In
advantages is given. order to reduce the dose to the small bowel, it is
14 What Is the Role of IMRT and IGRT in Rectal Cancer? 135

a b

Fig. 14.3 Example images acquired using a PET-CT and also in the rectum. In (b), the same patient is shown in
scanner. In (a), a sagittal view of a total body FDG-PET a transversal view, with the CT image on the left and on
image is shown, with clear uptake in the brain, the bladder the right the PET overlaid on the CT image

necessary to push the small bowel out of the high- between increasing bladder volume and decreasing
dose region. Kim et al. [40] showed in a study bowel dose was shown in the context of IMRT.
comparing the small bowel dose using conformal A disadvantage of full bladder instructions is
radiotherapy on full and empty bladder scans of the the large day-to-day variation of the bladder vol-
same individuals a significant reduction in bowel ume [42]. This can potentially lead to large sys-
exposure with increasing bladder volumes. In a tematic differences between planned and delivered
recent study by Nijkamp et al. [41], the relation dose to the small bowel.
136 J. Nijkamp et al.

Fig. 14.4 Sagittal view of a male rectal cancer patient sue contrast, especially useful in the caudal region at the
both on CT (left) and MR (right) after bony anatomy reg- level of the prostate
istration. The MR image has clear superiority in soft tis-

14.5.2 Patient Orientation and Belly combined measures resulted in the least bowel expo-
Board Use sure. When combining a full bladder and a belly
board, patients do however indicate discomfort due
The position of the small bowel can also be to the pressure that is put on the full bladder. In the
influenced by the orientation of the patient dur- study of Nijkamp et al. [41], the clinical relevance of
ing treatment. Supine set-up is generally associ- the significant reduction in bowel exposure using the
ated with more stability during irradiation, easier belly board was questioned. With IMRT and a full
set-up and more patient comfort, while prone bladder protocol, the bowel exposure was on average
treatment, especially on a belly board, is associ- already lower than the cut-off values, predicting
ated with reduced dose to the small bowel [40, acute and late toxicity, independent of patient
41, 43, 44]. orientation.
In many radiotherapy institutes, patients are
treated in prone positioning on a flat table to
reduce small bowel toxicity. However, several 14.5.3 Reduction of CTV
studies failed to show significant differences in
bowel exposure between prone and supine posi- Besides putting effort in changing the patient’s
tioning on a flat table [41, 45]. anatomy or improving the conformality of the
Instead of using a flat table, patients can also be treatment plan, it might also be possible to revise
positioned prone on a belly board, compressing the the currently accepted definition of the clinical
lower abdomen and pushing small bowel away from target volume (CTV) in certain cases. The inter-
the high-dose region. With the belly board, the bowel nationally recognized CTV definition [23] is
exposure is significantly reduced compared to prone based on all available literature about patterns of
or supine positioning on a flat table [40, 41]. This local recurrences to come to a general definition
effect is present both when using conformal RT [40] of the CTV and is irrespective of known risk fac-
and when using IMRT [41]. The study of Kim et al. tors, such as T or N stage. In certain subgroups,
[40] showed relatively more benefit from having a however, a smaller CTV might also be sufficient
full bladder than from the belly board, where the [46–49]. Using a 3D-computed model of all
14 What Is the Role of IMRT and IGRT in Rectal Cancer? 137

Fig. 14.5 Representation of the locations of the local locations are coloured for original tumour distance from
recurrences in the Dutch TME study, with patients treated the anal verge, with 0–5 cm (aqua), 5–10 cm (yellow) and
with a TME only on the left and patients treated with >10 cm (red). The top plane indicates the cranial border
5 × 5 Gy followed by a TME on the right. Recurrence of the used RT treatment fields

Fig. 14.6 Selection of the local recurrences in the Dutch plane indicates the cranial border of the used RT treat-
TME study for patients with node-negative disease, a ment fields. The lower plane indicates the S2-S3 inter-
negative CRM and the tumour <10 cm from the anal verge space, to which the cranial border of the CTV could be
(red dots). Patients in the left image were treated with lowered for these patients
TME only; on the right, RT + TME was given. The top

recurrences in the Dutch TME study, Nijkamp patients received radiotherapy (Fig. 14.5). For
et al. [49] demonstrated that most cranial recur- patients without primary nodal and MRF involve-
rences were located a few centimetres caudal of ment, the most cranial recurrences were all located
the promontory, irrespective of whether the below the level of the S2–S3 interspace (Fig. 14.6).
138 J. Nijkamp et al.

Lowering the cranial CTV border in early rectal conventional fractionated RT, the gain in set-up
cancers may therefore be a very effective way of accuracy should be balanced with the increase in
reducing the irradiated small bowel volume. imaging dose and treatment time, but also with the
other uncertainties.
Set-up errors also occur during treatment frac-
14.6 IGRT tions. These intra-fraction set-up errors are gen-
erally not corrected but need to be quantified to
Apart from the above-mentioned CTV and delinea- be taken into account in the PTV margin. The
tion uncertainties, geometrical uncertainties as magnitude of these errors is dependent on the ori-
inter- and intra-fraction set-up errors and inter- and entation of the patient and the immobilization
intra-fraction target volume variation need to be devices that are used for patient set-up. Prone
compensated for to ensure safe application of IMRT positioning on a flat table is associated with errors
in rectal cancer. All these factors together will up to 0.24 cm systematic and 0.22 cm random in
finally result in a CTV-PTV margin that ensures left-right direction [51], while supine positioning
sufficient coverage of the clinical target volume. is associated with errors £0.1 cm systematic and
The introduction of the cone beam CT has made a random [52].
reliable estimate of these uncertainties possible.

14.7.2 Target Volume Shape Variation


14.7 Geometric Uncertainties
With the rectum located within the CTV, large
14.7.1 Set-up Errors variation in the shape of the CTV occurs due to
changes in rectal filling. Like delineation varia-
Set-up errors are measured at the treatment tion, CTV shape variation is also heterogeneous
machine using electronic portal imaging devices [51, 52, 56]; few data are available concerning the
(EPID) [50], cone beam CT (CBCT) [51, 52] or shape variation of the CTV in rectal cancer
tomotherapy CT [53]. Corrections for set-up errors patients. Nuyttens et al. [56] described the move-
can be applied directly before treatment, by apply- ment of the anterior border of the CTV, ranging
ing a table shift. These online corrections demand from 0.4 cm SD at the level of the anus to 1.0 cm
for daily imaging and come with increased imag- SD at 10 cm from the anus. In their study, advanced
ing dose and treatment time. Online correction rectal cancer patients, mostly post-operative, were
protocols are often used for short-course RT of investigated using repeat CT scanning.
5 × 5 Gy, since the amount of fractions is limited In early stage rectal cancer, Nijkamp et al. [51,
and relatively high doses are given per fraction. 52] described the shape variation of the mesorectal
Corrections can also be applied in an offline set- part of the CTV both for patients treated in prone
ting, using statistical models such as shrinking and supine position on a flat table. For the anterior
action level or no action level protocols [54, 55]. border of the CTV, shape variation was 0.2 cm sys-
The statistical models aim to separate the system- tematic SD at the level of the anus up to 0.8 cm
atic errors, average difference between the plan- systematic SD at the upper anterior border of the
ning CT situation and the treatment, from the mesorectum. No significant differences between
random errors that represent the day-to-day varia- prone and supine were found.
tion. Offline set-up correction protocols require a In a recent repeat CT study, Nijkamp et al.
limited amount of images, and image registration [57] investigated the shape variation of the entire
can be performed at any time, not prolonging the CTV, both for short- and long-course RT. For 33
treatment time. With offline correction protocols, patients receiving short-course radiotherapy
set-up errors can be minimized to approximately (5 × 5 Gy, SCRT), daily repeat CT scans were
0.2–0.3 cm SD [50], while online set-up correc- acquired. For 30 chemoradiotherapy (CRT)
tion results in errors of £0.1 cm SD [51, 52]. For patients, daily repeat CT scans were acquired,
14 What Is the Role of IMRT and IGRT in Rectal Cancer? 139

Fig. 14.7 Male patient in a repeat CT study treated with (brown) and CTV (red) are delineated. On the last picture, all
CRT. Sagittal view of a planning CT scan (top left) and nine CTVs of the repeat CT scans are projected on the planning
repeat CT scans. On each scan, the bladder (yellow), rectum CT scan (with the planning CTV in white for visibility)

followed by weekly scans (Fig. 14.7). On a total 14.7.3 Planning Target Volume
of 482 CT scans, the CTV, bladder and rectum Margins to Account for
were delineated and the CTV shape variation was Geometric Uncertainties
determined. In this preoperative study, the het-
erogeneous shape variation as shown by Nuyttens PTV margins are designed to take geometric
et al. [56] was confirmed (Fig. 14.8). In the uncertainties into account and assure a certain
patients treated with CRT, a negative time trend minimum dose to the CTV for a certain percent-
in rectal volume was present, which translated in age of the patient population [18, 58]. Known
a group mean error of approximately 0.5 cm at margin recipes for calculation of PTV margins
the upper anterior border of the mesorectum are defined for rigid translational geometric
(Fig. 14.8). The maximum systematic error was uncertainties, such as patient set-up and CTV
slightly larger in the SCRT patients compared to position variation. However, as described above,
CRT. Based on these results, it is clear that the one of the major geometric uncertainties in
CTV shape variation is one of the major geomet- rectal cancer RT is shape variation of the CTV,
ric uncertainties in radiotherapy of rectal cancer. which cannot be incorporated in the currently
The shape variation is also complex due to its available PTV margin recipes [18, 58]. In the
heterogeneous presentation in different areas of design of a margin recipe for shape variation of
the CTV. the CTV, the first step is to take the correlation
140 J. Nijkamp et al.

Fig. 14.8 Overview (cm)


of the group mean
0.5
(top), systematic
(middle) and random 0.4
(bottom) shape
variation of the CTV 0.3
in SCRT (left) and 0.2
CRT (right). The
group mean error is 0.1
determined in
0.0
absolute cm, the
systematic and −2.0
random errors
in cm SD −0.2
−0.3

−0.4
−0.5
(cm)

1.0
0.9

0.8

0.7
0.6

0.5

0.4

0.3
0.2
0.1

0.0
(cm)

1.0
0.9

0.8

0.7
0.6

0.5

0.4

0.3
0.2
0.1

0.0
14 What Is the Role of IMRT and IGRT in Rectal Cancer? 141

Fig. 14.9 Locally defined 5 × 5 Gy (n = 33) 25 × 2 Gyv (n = 30)


rolling ball margins for the Rolling ball margins Rolling ball margins
5 × 5 Gy patients (left) and the (cm)
25 × 2 Gy patients (right)
3.2
3.0
2.8
2.6
2.4
2.2
2.0
1.8
1.5
1.3
1.1
0.9
0.7
0.5
0.3
0.1

between the different regions of the CTV into It is important to note that these margins only
account for the systematic error [59, 60]. In the account for shape variation. Other uncertainties,
rigid setting, all movements are correlated, such as set-up and delineated errors, should also be
which is taken into account in the van Herk mar- incorporated in the PTV margin. With online set-
gin recipe by multiplying the systematic error up correction, only intra-fraction stability is impor-
with a factor a = 2.5 [18]. In the deformable set- tant for patient set-up errors. With intra-fraction
ting, it is unclear how, for example, the shape set-up errors being small compared to shape varia-
variation around the sphincter is correlated to tion, influence on CTV coverage is small.
shape variation of the anterior part of the Incorporation of delineation uncertainties is much
mesorectum. To overcome the step of determin- more complex since generally the golden standard
ing the correlation between regions, Nijkamp CTV is not known. Observers who delineate always
et al. [57] used the repeat CT data to estimate too small or always too large will have a different
factor a for shape variation. By varying a effect on cure and toxicity of a patient. Just adding
between 2 and 4 in steps of 0.1, 21 different the derived standard deviations for delineation
PTVs were calculated using: variation to the shape and set-up variation in the
margin concept will probably not solve the prob-
mPTV = α ´ å +0.7 ´ σ + GM lem. This would imply addition of several centime-
tres to PTV margin for delineation variation, while
with the group mean, systematic and random clinical evidence on local control is generally good
errors shown in Fig. 14.8. For each PTV, a dose and not suggesting underdosage. As suggested by
distribution was calculated, and the dose was Weiss and Hess [61], it would be more beneficial to
accumulated over the repeat CT data. With focus on strict delineation guidelines, multi-modal-
a = 3.2, a minimum dose of 95% of the pre- ity information and multidisciplinary discussions
scribed dose could be assured for 90% of the with radiologists to reduce delineation variation.
patients. The subsequent required margins are
shown in Fig. 14.9 for SCRT and CRT sepa-
rately. The provided margin recipe was devel- 14.7.4 Correcting Shape Variation
oped and tested within one data set. To fully
validate the proposed margins, a confirmation Correcting for shape variation errors using IGRT
study is needed. is not as easy as dealing with set-up errors. Due to
142 J. Nijkamp et al.

the changes in the shape of the CTV, a simple local control rates and disease-free survival
adaptation of the table position is not sufficient. regardless of their initial clinical staging [67, 68].
Online shape variation correction would involve Up to now, the gold standard to accurately
on-site treatment planning or choosing the most assess tumour response to preoperative CRT
appropriate plan out of a library of plans for the remains pathological examination of the resec-
shape of that day. Although these strategies seem tion specimen. Standard imaging modalities
promising, they have not been investigated in using morphologic and size-related criteria lack
clinical practice yet. Offline shape variation cor- sufficient accuracy for the differentiation of
rection can be done by means of adaptive radio- responders from non-responders [69]. Functional
therapy (ART). By averaging the shape of the imaging modalities allow to assess treatment-
CTV during the first fractions, a better estimate of induced changes before volumetric changes
the ‘real’ CTV shape can be obtained. This ART become apparent [70, 71].
CTV can be used for replanning, in which smaller In rectal cancer, two imaging techniques show
PTV margins can be used because the shape vari- great promise for response prediction during and
ation error is reduced compared to using the plan- after preoperative treatment, namely, 18fluoro-
ning CTV alone. ART has previously been deoxy glucose positron emission tomography-CT
investigated for prostate cancer patients [62] but (18F-FDG PET-CT, Fig. 14.3) and diffusion-
never for rectal cancer. Like offline set-up correc- weighted MRI (DW-MRI). Changes in 18FDG
tion, ART also focuses on minimizing the sys- uptake, quantified by the standardized uptake
tematic part of the shape variation. Due to the value (SUV), appear to be a valid predictive fac-
large systematic errors in shape variation, up to tor for treatment response. This has been investi-
1.0 cm SD at the upper anterior part of the CTV gated in various studies [22, 64, 72–80]. All these
(Fig. 14.8), ART seems to be promising for rectal studies show that the metabolic response to CRT
cancer RT. The downside of ART is that CTV in rectal cancer with 18FDG-PET-CT, both early
needs to be delineated on the scans made in the and pre-surgical, is correlated to the histo-
beginning of the treatment as well as replanning pathological response in rectal cancer, and this
on the average CTV, which is labour intensive. is despite the differences in study set-up, scan
In summary, the dominant uncertainties that type, pathological and metabolic evaluation.
need to be taken into account in the PTV margin Based on both clinical tumour data and
are shape and delineation variation of the CTV sequential 18FDG-PET information, van Stiphout
and with smaller magnitude the set-up errors. et al. developed a predictive model for pathologi-
Current image-guided RT is very effective in cal complete remission (pCR) based on a pooled
minimization of the set-up errors, but not for the multicentre database of 953 patients [81].
other uncertainties. Extensive research in offline Although further prospective validation is neces-
or online adaptive replanning or plan selection sary, this model could prove to be a valuable
needs to be performed to really utilize IGRT for decision support tool towards a more patient-
rectal cancer. In the meantime, proper training, tailored approach.
guidelines and delineation atlases need to be pro- However, 18FDG-PET has some inherent
vided to minimize the delineation variation shortcomings, such as spatial resolution and
between radiation oncologists. difficulty in differentiation between tumour and
inflammation.
As a stand-alone modality in rectal cancer,
14.8 Response Evaluation DW-MRI can be a powerful tool in response
assessment both before and during preoperative
With the current standard treatment of rectal can- treatment as it allows with a high specificity to
cer, about 10–30% of all patients achieve a com- differentiate persistent tumour from therapy-
plete pathological response [63–66]. They have a related inflammation or necrosis [82–87]. Patients
favourable long-term outcome with excellent with a good response to treatment generally show
14 What Is the Role of IMRT and IGRT in Rectal Cancer? 143

a lower apparent diffusion coefficient (ADC) the response of the patient to radiotherapy [94]. If
prior to treatment, with a higher change in ADC dose escalation is considered, accurate knowl-
during and after CRT. The hypothesis is that a edge of the exact tumour volume is required to
low initial ADC value represents a more restric- maintain an acceptable small bowel and bladder
tive environment with less interstitial oedema, toxicity.
necrosis and a higher cellularity. Necrosis is gen- In rectal cancer, the data on the role of func-
erally associated with an acidic environment and tional imaging in dose painting are rather limited
a low oxygen concentration which influences the [95]. Roels et al. investigated the different bio-
response to radiotherapy and chemotherapy [88]. logical characteristics of rectal cancer by repeated
The combination of 18FDG-PET/CT and imaging with three different PET tracers (18F-
DW-MRI increases the accuracy of the predic- FDG, 18F-FLT and 18F-FMISO), aiming at refining
tion. The combination of the thresholds for the the definition of target volume and at testing the
initial ADC value and the change in SUVmax after possibility of radiation dose boosting. They found
ten fractions of chemo-radiation increases the that the mean 18F-FDG, 18F-FLT and 18F-FMISO-
sensitivity to 100% and the specificity to 94% for PET tumour volumes showed a tendency to
prediction of a pathological complete remission shrink during preoperative CRT. At each time
[22]. However, before these complex imaging point, the mean FDG-PET GTV was significantly
techniques and consequent analyses can be imple- larger than the FMISO-PET GTV, but not
mented in daily practice, considerable effort significantly larger than the mean FLT-PET GTV.
towards standardization of protocols and analysis Mismatch analysis confirmed that FDG, FLT and
is necessary to ensure reproducibility of the results FMISO-PET reflect different biological charac-
and enable widespread implementation [89]. teristics and can be used as a target for dose-esca-
lation RT in rectal cancer. In general, FDG and
FLT GTVs corresponded better than FDG and
14.9 Possibility of Dose Escalation FMISO GTVs, probably due to the non-specific
FMISO uptake in normoxic bowel wall and diffu-
Several studies have shown a dose-effect relation sion of FMISO through the rectal wall. Moreover,
in rectal cancer [90–93], implying that dose esca- the spatial distribution of FMISO varied consid-
lation might be an interesting option. Dose escala- erably during RT, while FDG and FLT uptake
tion generally consists of a boost dose given to the was less variable over time. These findings favour
GTV while maintaining a standardized dose to the the use of FDG and FLT as potential tracers for
CTV, for which IMRT seems a suitable option. TV definition in dose escalation and proof that
With use of IMRT, it is possible to deliver a simul- reimaging is important if these GTVs are used as
taneously integrated boost (SIB) to the GTV, a target for dose escalation.
while giving the standard dose to the CTV [19, 29, A second study by Roels et al. investigated
30]. The advantage of SIB over more local thera- FDG-PET/CT and MRI before the start of treat-
pies like contact therapy or brachytherapy is that ment, after ten fractions of radiation and before
treatment time is not prolonged and treatment surgery [96]. The FDG-PET GTV was automati-
burden is not increased, although dose to the cally delineated with two different segmentation
organs at risk might potentially be larger using algorithms: a modified threshold-based method
the SIB technique. and a gradient-based method. The mean FDG-
The concept of GTV as presently used is an derived GTVs were significantly smaller when the
oversimplification of the reality as it does not gradient-based method was used, and the FDG-
approach the complexity of the tumour biology. PET GTVs obtained in this way correlated best
A more elaborate delineation taking into account with the GTV measured on the pathological speci-
the biological components of tumours should men. Ciernik et al. found that the automated seg-
allow a more tailored and refined dose prescrip- mentation of the FDG signal strongly correlated
tion and dose distribution, which might enhance with the CT-derived GTV and the volume as
144 J. Nijkamp et al.

assessed on pathology in 11 patients with rectal


cancer [97]. He stated that PET-based delineation
can be automated which makes it fast and less
prone to inter-observer variation and usable in
creating a working PTV (wPTV) based on the
PET GTV. There are however some limitations to
this study [98]. First, there was no pathological
correlation of the PET-based GTV. Second, the
limited accuracy of FDG-PET in detecting micro-
scopic lymph node invasion makes it unsuitable
for the automatic definition of a CTV or PTV [99].
Patel et al. also addressed the question concerning
PET and inter-observer variability. They com-
pared the nodal and primary tumour GTV contour
for a hypothetical boost volume on conventional
CT alone and on FDG- and FLT-PET/CT in six Fig. 14.10 Sagittal view of a locally advanced rectal can-
rectal cancer patients [100]. Four radiation oncol- cer patient, with different positions of a pathologically
enlarged lymph node within repeat CT scans. The mea-
ogists delineated primary and nodal GTV on all surement bar denotes 1 cm
scans. The boost TVs based on combined PET/CT
resulted in lower inter-observer variability com-
pared with CT alone, particularly for nodal dis- tumour downstaging and downsizing with ulti-
ease. Again, this is only based on a limited number mately a complete response. For these patients, a
of patients, and less variability does not necessar- boost on the gross tumour volume may have
ily imply more correct delineation. Furthermore, additional value, although no data are available
as stated above, visual delineation has the disad- on improved prognosis with complete responses
vantage that it is less independent and less objec- achieved by higher doses.
tive than gradient-based methods. However, these A word of warning concerns the reproducibil-
results suggest a potential value for the integration ity of the GTV on a daily basis. Although in post-
of both 18F-FDG-PET and 18F-FLT-PET in the operative setting, Nuyttens et al. [56] reported
preoperative treatment of rectal cancer [101]. that clips placed at the anastomosis moved in
Two other studies investigated the possibility of cranial-caudal direction in the order of 1–1.5 cm
automated segmentation of the FDG-PET-CT sig- from day to day. In another study, 2–3 gold mark-
nal [95, 97, 102], which would facilitate the inte- ers were implanted in the tumour region preop-
gration of this imaging technique in clinical eratively, and positional variation was measured
practice. with a 3D deviation of 0.38 cm (1SD = 0.99) with
However, the use of dose escalation in the respect to the treatment fields [103]. Based on
standard treatment of rectal cancer can be ques- these data, it is clear that a substantial GTV mar-
tioned. The recent randomized trials with TME gin is needed to cover GTV motion. Similar to
surgery all show local recurrence rates far below the practice in prostate cancer, the use of
10% in the (chemo-)radiotherapy arm. It is implanted fiducial markers may be attractive for
unlikely to expect a much further reduction in LR online image-guided RT.
rates with dose escalation, since aggressive To get an impression of lymph node move-
tumour biology is most probably responsible for ment within the mesorectum, Nijkamp et al. [57]
these recurrences. Therefore, dose escalation delineated in a pilot study nine enlarged lymph
should be reserved for selected patients. nodes in two locally advanced rectal cancer
In patients suitable for minimal invasive sur- patients (Fig. 14.10). Depending on the location
gery or a ‘wait-and-see’ approach, the most of the lymph node, day-to-day motion was in the
important effect that needs to be achieved is order of 0.2 cm close to the posterior wall of the
14 What Is the Role of IMRT and IGRT in Rectal Cancer? 145

mesorectum, while translations up to 1.5 cm were 11. Tsai BM, Finne CO, Nordenstam JF et al (2010)
found close to the rectum itself. Transanal endoscopic mircrosurgery resection of rec-
tal tumor: outcomes and recommendations. Dis Colon
Second, it is important to realize that organ- Rectum 53:16–23
saving treatment, such as TEM or ‘wait-and-see’ 12. Ramirez JM, Aguilella V, Valencia J et al (2011) Transanal
policies, after preoperative (chemo-)radiotherapy endoscopic microsurgery for rectal cancer long-term
turns the rectum itself into an organ at risk. oncologic results. Int J Colorectal Dis 26:437–443
13. Lezoche E, Guerrieri M, Paganini AM et al (2005)
Tolerance limits for the rectum are extensively Long-term results in patients with T2–3N0 distal rec-
studied in prostate cancer patients, but dose dis- tal cancer undergoing radiotherapy before transanal
tribution in these treatments are substantially endoscopic microsurgery. Br J Surg 92:1546–1552
different compared to rectal cancer [104]. In 14. Nair RM, Siegel EM, Chen DT et al (2008) Long-
term results of transanal excision after neoadjuvant
prostate cancer RT, only a small part of the rec- chemoradiation for T2 and T3 adenocarcinomas of
tum is exposed to a high dose (>60 Gy), while in the rectum. J Gastrointest Surg 12:1797–1806
rectal cancer patients, the whole rectum will be 15. Habr-Gama A, Perez RO, Sao Juliao GP et al (2011)
part of the CTV, receiving a base dose of at least Nonoperative approaches to rectal cancer: a critical
evaluation. Semin Radiat Oncol 21:234–239
45 Gy. Extensive dose escalation studies should 16. Allaix ME, Rebecchi F, Giaccone C et al (2011) Long-
therefore be conducted to validate the tolerance term functional results and quality of life after transa-
limits of adding a GTV boost on top. nal endoscopic microsurgery. Br J Surg. doi: 10.1002/
bjs.7584
17. Brown G, Radcliffe AG, Newcombe RG et al (2003)
Preoperative assessment of prognostic factors in rec-
References tal cancer using high-resolution magnetic resonance
imaging. Br J Surg 90(3):355–364
1. Anon (1997) Improved survival with preoperative 18. van Herk M, Remeijer P, Rasch C et al (2000) The
radiotherapy in resectable rectal cancer. Swedish rec- probability of correct target dosage: dose-population
tal cancer trial. N Engl J Med 336:980–987 histograms for deriving treatment margins in radio-
2. Bosset JF, Collette L, Calais G et al (2006) therapy. Int J Radiat Oncol Biol Phys 47:1121–1135
Chemotherapy with preoperative radiotherapy in rec- 19. Guerrero Urbano MT, Henrys AJ, Adams EJ et al
tal cancer. N Engl J Med 355(11):1114–1123 (2006) Intensity-modulated radiotherapy in patients
3. Gerard JP, Conroy T, Bonnetain F et al (2006) with locally advanced rectal cancer reduces volume of
Preoperative radiotherapy with or without concurrent bowel treated to high dose levels. Int J Radiat Oncol
fluorouracil and leucovorin in T3–4 rectal cancers: Biol Phys 65:907–916
results of FFCD 9203. J Clin Oncol 24(28):4620–4625 20. Arbea L, Ramos LI, Martinez-Monge R et al (2010)
4. van Gijn W, Marijnen CAM, Nagtegaal ID et al (2011) Intensity-modulated radiation therapy (IMRT) vs. 3D
Preoperative radiotherapy combined with total conformal radiotherapy (3DCRT) in locally advanced
mesorectal excision for resectable rectal cancer: rectal cancer (LARC): dosimetric comparison and
12-year follow-up of the multicentre, randomised clinical implications. Radiat Oncol 5
controlled TME trial. Lancet Oncol 12:575–582 21. Duthoy W, De Gersem W, Vergote K et al (2004)
5. Sauer R, Becker H, Hohenberger W et al (2004) Clinical implementation of intensity-modulated arc
Preoperative versus postoperative radiotherapy for therapy (IMAT) for rectal cancer. Int J Radiat Oncol
rectal cancer. N Eng J Med 351:1731–1740 Biol Phys 60:794–806
6. Sebag-Montefiore D, Stephens RJ, Steele R et al 22. Lambrecht M, Deroose C, Roels S et al (2010) The
(2009) Preoperative radiotherapy versus selective use of FDG-PET/CT and diffusion-weighted mag-
postoperative chemoradiotherapy in patients with rec- netic resonance imaging for response prediction
tal cancer (MRC CR07 and NCIC-CTG C016): a mul- before, during and after preoperative chemoradiother-
ticentre, randomised trial. Lancet 373(9666):811–820 apy for rectal cancer. Acta Oncol 49(7):956–963
7. Smith N, Brown G (2008) Preoperative staging in rec- 23. Roels S, Duthoy W, Hausermans K et al (2006)
tal cancer. Acta Oncol 47:20–31 Definition and delineation of the clinical target vol-
8. Blomqvist L, Glimelius B (2008) The ‘good’, the ume for rectal cancer. Int J Radiat Oncol Biol Phys
‘bad’, and the ‘ugly’ rectal cancers. Acta Oncol 65:1129–1142
45:5–8 24. Baglan KL, Frazier RC, Yan D et al (2002) The dose-
9. Heald RJ, Moran BJ, Ryall RD et al (1998) Rectal volume relationship of acute small bowel toxicity
cancer: the Basingstoke experience of total mesorec- from concurrent 5-FU-based chemotherapy and radia-
tal excision, 1978–1997. Arch Surg 133:894–899 tion therapy for rectal cancer. Int J Radiat Oncol Biol
10. Ridgeway PF, Darzi AW (2003) The role of total Phys 52:176–183
mesorectal excision in the management of rectal can- 25. Robertson JM, Lockman D, Yan D et al (2008) The
cer. Cancer Control 10(3):205–211 dose-volume relationship of small bowel irradiation
146 J. Nijkamp et al.

and acute grade 3 diarrhea during chemoradiotherapy 39. O’Neill BD, Salerno G, Thomas K et al (2009) MR
for rectal cancer. Int J Radiat Oncol Biol Phys vs. CT imaging: low rectal cancer tumour delineation
70:413–418 for three-dimensional conformal radiotherapy. Br J
26. Gallagher MJ, Brereton HD, Rostock RA et al (1986) Radiol 978:509–513
A prospective study of treatment techniques to mini- 40. Kim TH, Chie EK, Kim DY et al (2005) Comparison
mize the volume of pelvic small bowel with reduction of the belly board device method and the distended
of acute and late effects associated with pelvic irradia- bladder method for reducing irradiated small bowel
tion. Int J Radiat Oncol Biol Phys 12:1565–1573 volumes in preoperative radiotherapy of rectal cancer
27. Letschert JG, Lebesque JV, de Boer RW et al (1990) patients. Int J Radiat Oncol Biol Phys 62(3):769–775
Dose-volume correlation in radiation-related late 41. Nijkamp J, Doodeman B, Marijnen CA et al (2011)
small-bowel complications: a clinical study. Radiother Bowel exposure in rectal cancer IMRT using prone,
Oncol 18:307–320 supine or a belly board. Radiother Oncol. doi:10.1016/j.
28. Samuelian JM, Callister MD, Ashman JB et al (2011) radonc.2011.05.076
Reduced acute bowel toxicity in patients treated with 42. O’Doherty UM, McNair HA, Norman AR et al (2006)
intensity-modulated radiotherapy for rectal cancer. Variability of bladder filling in patients receiving radi-
Int J Radiat Oncol Biol Phys. doi: 10.1016/j. cal radiotherapy to the prostate. Radiother Oncol
ijrobp.2011.01.051 79:335–340
29. De Ridder M, Tournel K, Van Nieuwenhove Y et al 43. Siddiqui F, Shi C, Papanikolaou N, Fuss M (2008)
(2008) Phase II study of preoperative helical Image-guidance protocol comparison: supine and
tomotherapy for rectal cancer. Int J Radiat Oncol Biol prone set-up accuracy for pelvic radiation therapy.
Phys 70:728–734 Acta Oncol 47:1344–1350
30. Seierstad T, Hole KH, Saelen E et al (2009) MR-guided 44. Bayley AJ, Catton NC, Haycocks T et al (2004) A ran-
simultaneous integrated boost in preoperative radiother- domized trial of supine vs. prone positioning in patients
apy of locally advanced rectal cancer following neoad- undergoing escalated dose conformal radiotherapy for
juvant chemotherapy. Radiother Oncol 93:279–284 prostate cancer. Radiother Oncol 70:37–44
31. Valentini V, Beets-Tan R, Borras JM et al (2008) 45. Drzymala M, Hawkins MA, Henrys AJ et al (2009)
Evidence and research in rectal cancer. Radiother The effect of treatment position, prone or supine, on
Oncol 87:449–474 dose-volume histograms for pelvic radiotherapy in
32. Myerson RJ, Garofalo MC, Naqa IE et al (2009) patients with rectal cancer. Br J Radiol 82:321–327
Elective clinical target volumes for conformal therapy 46. Syk E, Torkzad MR, Blomqvist L et al (2006)
in anorectal cancer: an radiation therapy oncology Radiological findings do not support lateral residual
group consensus panel contouring atlas. Int J Radiat tumour as a major cause of local recurrence of rectal
Oncol Biol Phys 74(3):824–830 cancer. Br J Surg 93:113–119
33. Nijkamp J, de Haas-Kock DFM, Beukema JC et al 47. Syk E, Torkzad MR, Blomqvist L et al (2008) Local
(2011) Target volume delineation variation in recurrence in rectal cancer: anatomic localization and
radiotherapy for early stage rectal cancer in the effect on radiation target. Int J Radiat Oncol Biol Phys
Netherlands. Radiother Oncol. doi:10.1016/j. 72(3):658–664
radonc.2011.08.011 48. Chien CR, Chen SW, Chen WT (2009) Radiation
34. Fuller CD, Nijkamp J, Duppen JC et al (2010) fields of neoadjuvant concurrent chemoradiotherapy
Prospective randomized double-blind pilot study of site- for rectal cancer: in response to Yu et al. (Int J Radat
specific consensus atlas implementation for rectal can- Oncol Biol Phys 2008;71:1175–1180). Int J Radiat
cer target volume delineation in the cooperative group Oncol Biol Phys 73:639
setting. Int J Radiat Oncol Biol Phys 79:481–489 49. Nijkamp J, Kusters M, Beets-Tan RG et al (2011)
35. Hortobagyi E, Lambrecht M, Verstraete J et al (2011) Three-dimensional analysis of recurrence patterns in
Improving care of rectal cancer in Belgium by stan- rectal cancer: the cranial border in hypofractionated
dardizing CTV delineation. Radiother Oncol preoperative radiotherapy can be lowered. Int J Radiat
99(1):S67 (173) Oncol Biol Phys 80:103–110
36. Rasch C, Steenbakkers R, van Herk M (2005) Target 50. Robertson JM, Campbell JP, Yan D (2009) Generic
definition in prostate, head, and neck. Semin Radiat planning target margin for rectal cancer treatment
Oncol 15:136–145 setup variation. Int J Radiat Oncol Biol Phys
37. Steenbakkers R, Duppen J, Fitton I et al (2006) 74:1470–1475
Reduction of observer variation using matched CT-PET 51. Nijkamp J, de Jong R, Sonke JJ et al (2009) Target
for lung cancer delineation: a three-dimensional analy- volume shape variation during hypo-fractionated pre-
sis. Int J Radiat Oncol Biol Phys 64:435–448 operative irradiation of rectal cancer patients.
38. Krengli M, Cannillo B, Turri L et al (2010) Target vol- Radiother Oncol 92:202–209
ume delineation for preoperative radiotherapy of rec- 52. Nijkamp J, de Jong R, Sonke JJ et al (2009) Target
tal cancer: inter-observer variability and potential volume shape variation during irradiation of rectal
impact of FDG-PET/CT imaging. Technol Cancer cancer patients in supine position; comparison with
Res Treat 4:393–398 prone position. Radiother Oncol 93:285–292
14 What Is the Role of IMRT and IGRT in Rectal Cancer? 147

53. Tournel K, de Ridder M, Engels B et al (2008) 67. Maas M, Nelemans PJ, Valentini V et al (2010) Long-
Assessment of intrafractional movement and internal term outcome in patients with a pathological complete
motion in radiotherapy of rectal cancer using mega- response after chemoradiation for rectal cancer: a
voltage computer tomography. Int J Radiat Oncol Biol pooled analysis of individual patient data. Lancet
Phys 71:934–939 Oncol 11:835–844
54. Bel A, van Herk M, Bartelink H, Lebesque JV (1993) 68. Vecchio FM, Valentini V, Minsky BD et al (2005) The
A verification procedure to improve patient set-up relationship of pathologic tumor regression grade
accuracy using portal images. Radiother Oncol (TRG) and outcomes after preoperative therapy in
29:253–260 rectal cancer. Int J Radiat Oncol Biol Phys 62:
55. de Boer HC, Heijmen BJ (2007) eNAL: an extension 752–760
of the NAL setup correction protocol for effective use 69. Barbaro B, Fiorucci C, Tebala C et al (2009) Locally
of weekly follow-up measurements. Int J Radiat advanced rectal cancer: MR imaging in prediction of
Oncol Biol Phys 67:1586–1595 response after preoperative chemotherapy and radia-
56. Nuyttens J, Robertson J, Yan D et al (2002) The vari- tion therapy. Radiology 250:730–739
ability of the clinical target volume for rectal cancer 70. Juweid ME, Cheson BD (2006) Positron-emission
due to internal organ motion during adjuvant treat- tomography and assessment of cancer therapy. N Engl
ment. Int J Radiat Oncol Biol Phys 53:497–503 J Med 354:496–507
57. Nijkamp J, Swellengrebel M, Hollmann B et al (2012) 71. Koh DM, Collins DJ (2007) Diffusion-weighted MRI
Repeat CT assessed CTV variation and PTV margins in the body: applications and challenges in oncology.
for short- and long-course pre-operative RT of rectal AJR Am J Roentgenol 188:1622–1635
cancer. Radiother Oncol 102(3):399–405. 72. Capirci C, Rubello D, Chierichetti F et al (2004)
58. Stroom JC, de Boer HC, Huizinga H et al (1999) Restaging after neoadjuvant chemoradiotherapy for
Inclusion of geometrical uncertainties in radiotherapy rectal adenocarcinoma: role of F18-FDG PET. Biomed
treatment planning by means of coverage probability. Pharmacother 58:451–457
Int J Radiat Oncol Biol Phys 43:905–919 73. Capirci C, Rubello D, Pasini F et al (2009) The role of
59. McKenzie A (2004) Defining the PTV and PRV – new dual-time combined 18Fluorodeoxyglucose positron
ideas about old problems. Radiother Oncol ESTRO emission tomography and computed tomography in
73:s203:455 the staging and restaging workup of locally advanced
60. van Kranen SR, van Herk M, Sonke J-J (2008) Margin rectal cancer, treated with preoperative chemoradia-
design for deforming and differential moving target tion therapy and radical surgery. Int J Radiat Oncol
volumes. Radiother Oncol ESTRO 88:s154:446 Biol Phys 74:1461–1469
61. Weiss E, Hess CF (2003) The impact of gross tumor vol- 74. Cascini GL, Avallone A, Delrio P et al (2006) 18F-
ume and clinical target volume definition on the total FDG PET is an early predictor of pathologic tumor
accuracy in radiotherapy. Strahlenther Onkol 179:21–30 response to preoperative radiochemotherapy in locally
62. Yan D, Lockman D, Brabbins D et al (2000) An off- advanced rectal cancer. J Nucl Med 47:1241–1248
line strategy for constructing a patient-specific 75. Janssen MH, Ollers MC, Riedl RG et al (2010)
planning target volume in adaptive treatment pro- Accurate prediction of pathological rectal tumor
cess for prostate cancer. Int J Radiat Oncol Biol response after two weeks of preoperative radiochemo-
Phys 48:289–302 therapy using 18F-fluorodeoxyglucose-positron emis-
63. Capirci C, Rubello C, Chierichetti F et al (2006) sion tomography-computed tomography imaging. Int
Long-term prognostic value of 18F-FDG PET in J Radiat Oncol Biol Phys 77:392–399
patients with locally advanced rectal cancer previ- 76. Janssen MH, Ollers MC, van Stiphout RG et al (2010)
ously treated with neoadjuvant radiochemotherapy. Evaluation of early metabolic responses in rectal can-
AJR Am J Roentgenol 187:W202–W208 cer during combined radiochemotherapy or radiother-
64. Capirci C, Rampin L, Erba PA et al (2007) Sequential apy alone: sequential FDG-PET-CT findings.
FDG-PET/CT reliably predicts response of locally Radiother Oncol 94:151–155
advanced rectal cancer to neo-adjuvant chemo-radia- 77. Mak D, Joon DL, Chao M et al (2010) The use of PET
tion therapy. Eur J Nucl Med Mol Imaging 34: in assessing tumor response after neoadjuvant chemo-
1583–1593 radiation for rectal cancer. Radiother Oncol 97:
65. Valentini V, Coco C, Cellini N et al (1999) Preoperative 205–211
chemoradiation with cisplatin and 5-fluorouracil for 78. Rosenberg R, Herrmann K, Gertler R et al (2009) The
extraperitoneal T3 rectal cancer: acute toxicity, tumor predictive value of metabolic response to preoperative
response, sphincter preservation. Int J Radiat Oncol radiochemotherapy in locally advanced rectal cancer
Biol Phys 45:1175–1184 measured by PET/CT. Int J Colorectal Dis 24:191–200
66. Vliegen RF, Beets-Tan RG, Vanhauten B et al (2008) 79. Yoon MS, Ahn SJ, Nah BS et al (2011) The metabolic
Can an FDG-PET/CT predict tumor clearance of the response using 18F-fluorodeoxyglucose-positron emis-
mesorectal fascia after preoperative chemoradiation sion tomography/computed tomography and the
of locally advanced rectal cancer? Strahlenther Onkol change in the carcinoembryonic antigen level for pre-
184:457–464 dicting response to pre-operative chemoradiotherapy
148 J. Nijkamp et al.

in patients with rectal cancer. Radiother Oncol 91. Tepper JE, Wang AZ (2010) Improving local control
98:134–138 in rectal cancer: radiation sensitizers or radiation dose.
80. Janssen MH, Ollers MC, van Stiphout RG et al (2011) J Clin Oncol 28:1623–1632
PET-based treatment response evaluation in rectal 92. Wiltshire KL, Ward IG, Swallow C et al (2006)
cancer: prediction and validation. Int J Radiat Oncol Preoperative radiation with concurrent chemotherapy
Biol Phys. doi: 10.1016/j.ijrobp.2010.11.038 for resectable rectal cancer: effect of dose escalation
81. van Stiphout RG, Lammering G, Buijsen J et al (2011) on pathologic complete response, local recurrence-
Development and external validation of a predictive free survival, disease-free survival, and overall sur-
model for pathological complete response of rectal vival. Int J Radiat Oncol Biol Phys 64:709–716
cancer patients including sequential PET-CT imaging. 93. Gerard J-P, Chapet O, Nemoz C et al (2004) Improved
Radiother Oncol 98:126–133 sphincter preservation in low rectal cancer with high-
82. Vandecaveye V, de Keyzer F, Nuyts S et al (2007) dose preoperative radiotherapy: the Lyon R96–02 ran-
Detection of head and neck squamous cell carcinoma domized trial. J Clin Oncol 22:2404–2409
with diffusion weighted MRI after (chemo)radiother- 94. Yang Y, Xing L (2005) Towards biologically confor-
apy: correlation between radiologic and histopathologic mal radiation therapy (BCRT): selective IMRT dose
findings. Int J Radiat Oncol Biol Phys 67:960–971 escalation under the guidance of spatial biology distri-
83. Abdel Razek AA, Kandeel AY, Soliman N et al (2007) bution. Med Phys 32:1473–1484
Role of diffusion-weighted echo-planar MR imaging 95. Lambrecht M, Haustermans K (2010) Clinical evi-
in differentiation of residual or recurrent head and dence on PET-CT for radiation therapy planning in
neck tumors and posttreatment changes. AJNR Am J gastro-intestinal tumors. Radiother Oncol 96:339–346
Neuroradiol 28:1146–1152 96. Roels S, Slagmolen P, Nuyts J et al (2009) Biological
84. Dzik-Jurasz A, Domenig C, George M et al (2002) image-guided radiotherapy in rectal cancer: chal-
Diffusion MRI for prediction of response of rectal lenges and pitfalls. Int J Radiat Oncol Biol Phys
cancer to chemoradiation. Lancet 360:307–308 75:782–790
85. Lambrecht M, Vandecaveye V, de Keyzer F et al 97. Ciernik IF, Huser M, Burger C et al (2005) Automated
(2011) Value of diffusion-weighted magnetic reso- functional image-guided radiation treatment plan-
nance imaging for prediction and early assessment of ning for rectal cancer. Int J Radiat Oncol Biol Phys
response to neoadjuvant radiochemotherapy in rectal 62:893–900
cancer: preliminary results. Int J Radiat Oncol Biol 98. Roels S, Haustermans K, Gregoire V, In regard to
Phys. doi:10.1016/j.ijrobp.2010.12.063 Ciernik et al (2006) Automated functional image-
86. Sun YS, Zhang XP, Tang L et al (2010) Locally guided radiation treatment planning for rectal can-
advanced rectal carcinoma treated with preoperative cer. Int J Radiat Oncol Biol Phys 64:1611–1615
chemotherapy and radiation therapy: preliminary 99. International Commission on Radiation Units and
analysis of diffusion-weighted MR imaging for early Measurements ICRU Report 62 (1999) Prescribing,
detection of tumor histopathologic downstaging. recording, and reporting photon beam therapy
Radiology 254:170–178 (Supplement to ICRU report 50). ICRU, Bethesda
87. Lambregts DM, Vandecaveye V, Barbaro B et al 100. Patel DA, Chang ST, Goodman KA (2007) Impact of
(2011) Diffusion-weighted MRI for selection of integrated PET/CT on variability of target volume
complete responders after chemoradiation for locally delineation in rectal cancer. Technol Cancer Res
advanced rectal cancer: a multicenter study. Ann Surg Treat 6:31–36
Oncol 18:2224–2231 101. Muijs CT, Beukema JC, Widder J et al (2011)
18
88. Majno G, Joris I (1995) Apoptosis, oncosis, and F-FLT-PET for detection of rectal cancer. Radiother
necrosis. An overview of cell death. Am J Pathol Oncol 98:357–359
146:3–15 102. Buijsen J, van den Bogaard J, Janssen MH et al
89. Padhani AR, Liu G, Koh DM et al (2009) Diffusion- (2011) FDG-PET provides the best correlation with
weighted magnetic resonance imaging as a cancer the tumor specimen compared to MRI and CT in rec-
biomarker: consensus and recommendations. tal cancer. Radiother Oncol 98:270–276
Neoplasia 11:102–125 103. Vorwerk H, Liersch T, Rothe H et al (2009) Gold
90. Viani GA, Stefano EJ, Soares FV, Afonso SL (2011) markers for tumor localization and target volume
Evaluation of biologic effective dose and schedule of delineation in radiotherapy for rectal cancer.
fractionation for preoperative radiotherapy for rectal Strahlenther Onkol 185:127–133
cancer: meta-analyses and meta-regression. Int J 104. Cummings BJ (2007) Is there a limit to dose escala-
Radiat Oncol Biol Phys 80:985–991 tion for rectal cancer? Clin Oncol 19:730–737
What Are the Dose-Volume
Constraints to Reduce Late Toxicity? 15
Krzysztof Bujko

Contents 15.1 Introduction


15.1 Introduction .............................................. 149
Two recent randomized trials using modern
15.2 Cranial Border of the CTV ..................... 150
surgery (total mesorectal excision [TME]) have
15.3 Caudal Border of the CTV...................... 151 shown local recurrence risk reduction by approx-
References ............................................................... 153 imately 50% with preoperative radiation com-
pared to surgery alone [25, 28]. This benefit,
however, has not translated into improvement of
survivals. This lack of survival benefit and
significant late toxicity caused by radiation has
launched a debate on appropriate indications for
preoperative radiation. There are two main stand-
points regarding this issue. Advocates of using
preoperative radiation claim that all patients who
have clinically diagnosed T3 cancer require this
treatment [16], whereas others limit indications
for preoperative radiation for cT3 tumours with
deep infiltration into the mesorectum (>5 mm) [3].
Some authors do not recommend preoperative
radiation even for patients with ‘good prognosis’
nodal disease [27]. The latter authors point out
that with TME, the risk of local recurrence has
been diminished in specialized centres to less
than 10% and that postradiation toxicity is sub-
stantial which has a negative impact on the qual-
ity of life. This confers that only less than 5% of
irradiated patients (radiation reduces the pelvic
recurrence risk by half) benefit from this treat-
ment, and remaining 95% of patients are ‘unnec-
essarily’ exposed to the risk of radiation toxicity.
K. Bujko Thus, a risk/benefit ratio analysis may favour
Department of Radiotherapy,
omitting preoperative radiation. On the other
Maria Sklodowska-Curie Memorial Cancer Centre,
W.K. Roentgena 5, 02 781 Warsaw, Poland hand, advocates of the preoperative radiation
e-mail: bujko@coi.waw.pl approach point out that in the total population,

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 149


DOI 10.1007/978-3-642-25005-7_15, © Springer-Verlag Berlin Heidelberg 2012
150 K. Bujko

the local recurrence rate after TME is higher non-cancer deaths after preoperative radiation
than observed in the specialized centres and was also due to an increase in deaths attributed
amounts 20% of pathologically staged T3 dis- to vascular and infective causes [5]. This meta-
ease [6]. In addition, with radiotherapy technique analysis also showed that the favourable effect of
modifications, the risk of late toxicity might be radiation still dominated, as at 5 years, there
reduced. Taking both of the above arguments into were 3.3% more of non-cancer deaths and 8.3%
account, a risk/benefit ratio analysis may favour fewer rectal cancer deaths in the irradiated
the use of preoperative radiation for all cT3 can- patients compared to those treated with surgery
cers. Thus, a reduction of the risk of late postra- alone. The above results suggest that preopera-
diation toxicity is a key issue in the current debate tive radiation has a potential for improving over-
on the indications for preoperative radiotherapy. all survival which is counterbalanced by the
In rectal cancer preoperative radiotherapy, two excess of non-cancer deaths.
guidelines showing anatomical borders of clini- Small bowel late toxicity has been reduced
cal target volume (CTV) are available [15, 22]. after replacing postoperative radiation with pre-
The objective of the current article is to provide operative radiation [8, 23]. This is because small
readers with a proposal of modifying these guide- bowel fills up the dead space left behind in a pos-
lines in order to diminish the risk of postradiation terior pelvis after removal of mesorectum and
late side effects. It should be stressed that this rectum. Thus, in order to provide large dose to
proposal is not accepted as a routine procedure the tumour bed, irradiation of large volume of
by all investigators and is still a matter of debate. small bowel cannot be avoided in the postopera-
tive setting. In contrast, in the preoperative set-
ting, when the mesorectum and rectum are still in
15.2 Cranial Border of the CTV situ, the amount of irradiated small bowel is much
smaller. Despite this, in the Swedish randomized
The recent update of the Dutch TME trial, which trial, severe late small bowel side effect, namely,
compared preoperative radiation and TME with small bowel obstruction, has occurred more often
TME alone, demonstrated no difference in over- after preoperative radiation and surgery than sur-
all survival [28]. However, when patients were gery alone – 13.9% versus 5.5% at 14 years [2].
operated with negative circumferential margin, It might be reasonable to assume that a reduc-
higher cancer-specific survival and 10% gain in tion of irradiated volume may result both in a
overall survival at 10 years in stage III cancer decreased risk of postradiation, second malig-
were reported in the preoperative radiation nancy and in the reduction of early and late small
group. Of note, slightly more non-cancer deaths, bowel toxicity. Traditionally, the sacral promon-
mostly due to secondary malignancy, were tory has been the anatomical landmark for cranial
reported in the radiotherapy group. Similar border of CTV [15, 22]. The appropriateness of
observations were recorded in Sweden. The data this rule has been questioned by two recent arti-
from two trials demonstrated that, after a long cles on location of in-pelvic recurrences after
follow-up, 7% of patients had second cancers TME. Syk et al. [26] identified a total of 155
[1]. More patients treated with radiotherapy and patients with local recurrence from a population-
surgery developed a second cancer as compared based cohort of 2,315 patients. The site of recur-
to those treated with surgery alone; the relative rence was observed in the lower half of the pelvis
risk was 1.85 with 95% confidence interval in more than two-thirds of all patients. All recur-
between 1.23 and 2.78. However, the favourable rences were situated below S1–S2 interspace.
effect of radiation still dominated, as 20.3% of Nijkamp et al. [18] analysed the site of recurrence
the irradiated patients got either local recurrence in 94 patients treated within the frame of the
or a secondary cancer, compared with 30.7% of Dutch TME trial. Only 3% of recurrences were
the patients treated with surgery alone (relative situated above the S2–S3 interspace and addi-
risk, 0.55; 95% confidence interval 0.44–0.70). tional 2% at the level of S2–S3 interspace.
The meta-analysis showed that the excess of However, if patients with negative circumferential
15 What Are the Dose-Volume Constraints to Reduce Late Toxicity? 151

resection margin were considered, only in 2% of tinence in the irradiated patients was observed
these patients, recurrence was observed above or only at low level of severity; severe incontinence
at S2–S3 interspace. The corresponding figure for rate was similar in both treatment-assigned groups.
the patients with positive circumferential resec- In the Dutch TME trial, at 24 months post treat-
tion margin was 12%. Interestingly, in the patents ment, 67% of male patients receiving preoperative
with positive circumferential resection margin, irradiation and who were previously sexually
the recurrence did not always appear at the same active, were still active after treatment compared
level as the primary tumour. With a cranially to 76% of patients from the surgery alone group,
reduced CTV to the S2–S3 interspace, over 60% p = 0.06 [14]. For female patients, these figures
of reduction of absolute small bowel exposure at were 72% and 90%, respectively, p = 0.01. It should
dose levels of 15–35 Gy could be achieved with be stressed that the main cause of faecal inconti-
three-field conventional radiotherapy, increasing nence and male sexual dysfunction is surgery,
to 80% when IMRT was used [18]. Both of the rather than radiotherapy [24].
above reports suggested that the cranial border of Excluding the anal canal, or part of it, from the
CTV can be lowered to the S2–S3 interspace in CTV is a strategy that prevents radiation-induced
patients in whom pelvic MRI predicts negative faecal incontinence [12, 29]. Lange et al. [12]
circumferential resection margin (Fig. 15.1). It reported incontinence in 93% of patients in whom
should be stressed that this guideline should be sphincter was included in the preoperative radia-
used selectively. For patients with the primary- tion fields compared to 65% of patients in whom
tumour extension in the presacral region above sphincter was not irradiated, p = 0.059. In addi-
the S2–S3 interspace or with enlarged lymph tion, one can assume that the exclusion of inferior
nodes abutting mesorectal fascia at this level, part of the vagina may prevent vaginal dryness
CTV should be extended cranially. Nijkamp and pain during intercourse. It is also worth to
et al. [18] recommended CTV reduction to the note that scattered cumulative radiation dose at
S2–S3 interspace only in patients without the testicles was reported between 0.7 and 8.4 Gy,
expected nodal or circumferential resection mar- with a mean of 3.56 Gy [10]. Such doses may
gin involvement. result in a permanent infertility and in a risk of
Apart from lowering of caudal border of the hypogonadism in substantial proportion of pati-
CTV, belly-board and distended urinary bladder ents [10, 30]. The increase of the distance between
result in a reduction of small bowel radiation lower field margin and testicles exponentially
exposure [11, 19] (Fig. 15.1). decreased the amount of this dose [10]. Another
way of reducing sexual function impairment is
to avoid irradiation of the penile bulb. Several
15.3 Caudal Border of the CTV studies reported that penile bulb dose-volume
parameters correlated with the risk of erectile dys-
Anorectal and sexual function impairments caused function [21]. For example, erectile dysfunction
by surgery and radiation are the most important was observed in 0%, 80% or 100% of patients
adverse side effects as they affect large proportion with a D70 (i.e. minimum dose received by 70%
of patients and are permanent as well as they inter- volume of the penile bulb) of 0–40, 40–70 and
fere with patients’ daily activity. These adverse >70 Gy, respectively [7]. In addition, excluding
side effects have been reported in details on large the perineal skin and distal part of ischiorectal
number of patients treated within the frame of the fossa is a strategy that prevents wound healing
Dutch TME trial and the MRC CR07 trial [14, 20, delay after abdomino-perineal resection and pain-
24]. Faecal incontinence was reported in 62% of ful acute perineal skin reaction. Marijnen et al.
patients having preoperative radiation compared to [13] reported that 31% of 174 patients irradiated
38% of those treated with surgery alone, p < 0.001 preoperatively with perineum included into the
in the Dutch TME trial [20] and 53.2% versus fields had perineal wound complications in com-
37.3% in the MRC CR07 trial, p = 0.007, respec- parison with 18% of 40 patients in whom the
tively [24]. In the latter trial, the increase of incon- perineum was not irradiated.
152 K. Bujko

All of the above considerations suggest that


the sphincters’ complex, the caudal part of the
vagina, the penile bulb and the perineal skin
should be avoided in CTV delineation (Fig. 15.1),
provided these regions are not invaded by a distal
cancer extension.
When considering the anatomical boundaries
of the CTV, it is worth to refer to the rules and to
the rationales for the boundaries of surgical resec-
tion [4, 17]. This is because there is no reason to
assume that the anatomical boundaries of the CTV
should differ from the anatomical boundaries of
resection of the mesorectal tissue and the distal
bowel margin. Pathologic examinations of rectal
cancer postoperative specimen have demonstrated
that mesorectal cancer deposits are found up to
4 cm caudally from a lower pole of the primary

Fig. 15.1 Clinical target volume (CTV) contoured at the


middle sagittal plane. White thick arrow; lower border of
mesorectum (level of the puborectal muscle) and upper
border of the anal canal. Black thick arrow; S2–S3 inter-
face. White thin arrow; lower border of tumour. In all
patients, there was no upwards tumour extension or threat-
ened circumferential resection margin. Thus, the upper
border of CTV terminates at S2–S3 interface sparing
small bowel. (Panel a) Lower border of tumour was
located 8 cm from the anal verge and 5 cm from the ano-
rectal ring. The lower border of CTV terminates 4 cm cau-
dally from the lower border of tumour and does not
include the distal part of mesorectum. The entire anal
canal and the distal part of vagina are spared. The black
line perpendicular to the surface of the sacral bone and the
thin black arrow shows the possibility of further reduction
of the CTV in order to spare small bowel. (Panel b) Lower
border of tumour was located 2.5 cm from the anal verge
and invaded 0.5 cm of the upper part of the anal canal. The
patient was scheduled for abdomino-perineal resection.
The lower border of CTV terminates at 1.5 cm caudally
from the lower border of tumour to account for distal
intramural spread. The perineal skin is spared in order to
avoid the acute skin toxicity and the perineal wound heal-
ing delay. Sparing of the perineum increases the distance
between lower border of fields and the testicles, diminish-
ing scatter dose at the testicles. Distended urinary bladder
reduces the amount of small bowel within radiation beams.
Belly-board is used in this patient, although, admittedly,
its value is unproven when CTV terminates at S2–S3
interface and the urinary bladder is distended. (Panel c)
Lower border of tumour was located 5 cm from the anal
verge and 2 cm from the anorectal ring. The lower border
of CTV terminates at the anorectal ring and includes the
entire mesorectum. Note that even if a margin for the
planning target volume would be added, still the distal
part of anal canal and the distal part of vagina are spared
15 What Are the Dose-Volume Constraints to Reduce Late Toxicity? 153

gross cancer [9]. For this reason, it is recommended 4. Bujko K, Bujko M, Pietrzak L (2007) Clinical target
that mesorectum with mesorectal visceral fascia volume for rectal cancer: in regard to Roels et al. (Int
J Radiat Oncol Biol Phys 2006;65:1129–1142). Int J
should be resected at least 4 cm caudally from the Radiat Oncol Biol Phys 68:313 (letter)
tumour. This confers that for tumours in the upper 5. Colorectal Cancer Collaborative Group (2001)
rectum, 4 cm of mesorectal clearance distal to the Adjuvant radiotherapy for rectal cancer: a systematic
tumour is sufficient, and for tumours located in the overview of 8,507 patients from 22 randomised trials.
Lancet 358(9290):1291–1304
lower rectum, total mesorectum is removed down 6. Eriksen MT, Wibe A, Haffner J et al (2007) Prognostic
to the pelvic floor. Intramural subclinical spread groups in 1,676 patients with T3 rectal cancer treated
within a bowel wall may occur caudally from gross without preoperative radiotherapy. Dis Colon Rectum
primary cancer. This spread has been found rarely 50:156–167
7. Fisch BM, Pickett B, Weinberg V et al (2001) Dose of
beyond 1–1.5 cm from gross tumour. If distal radiation received by the bulb of the penis correlates
spread occurs beyond 1–1.5 cm, the prognosis is with risk of impotence after three-dimensional
poor due to a high risk of occult distant metastases. conformal radiotherapy for prostate cancer. Urology
For this reason, 1 cm of distal bowel surgical clear- 57:955–959
8. Frykholm GJ, Glimelius B, Pahlman L (1993)
ance is regarded as sufficient in patients undergo- Preoperative or postoperative irradiation in adenocar-
ing anterior resection for low tumours [17]. It cinoma of the rectum: final treatment results of a ran-
should be pointed out that even in low-lying domized trial and evaluation of late secondary effects.
tumours, if levators and external sphincter are not Dis Colon Rectum 36:564–572
9. Heald RJ, Husband EM, Ryall RD (1982) The
invaded, anterior resection is still possible as the mesorectum in rectal cancer surgery – the clue to pel-
ischiorectal fossa is not at risk of tumour recur- vic recurrence? Br J Surg 69:613–616
rence. It seems that levators constitute an effective 10. Hermann RM, Henkel K, Christiansen H et al (2005)
barrier against downwards cancer spread. There Testicular dose and hormonal changes after radiother-
apy of rectal cancer. Radiother Oncol 75:83–88
are no regional lymph nodes in the ischiorectal 11. Kim TH, Kim DY, Cho YH et al (2005) Comparative
fossa. In the literature, recurrences in the ischi- analysis of the effect of belly board and bladder dis-
orectal fossa or in the perineum were reported only tention in preoperative radiotherapy in rectal cancer.
in the patients who have undergone abdomino- Strahlenther Onkol 181:601–605
12. Lange MM, den Dulk M, Bossema ER et al (2007)
perineal resection. It can be assumed that these Cooperative Clinical Investigators of the Dutch Total
recurrences likely originate from contamination of Mesorectal Excision Trial. Risk factors for faecal
cancer cells during surgery because no such cases incontinence after rectal cancer treatment. Br J Surg
were reported after anterior resection. 94:1278–1284
13. Marijnen CA, Kapiteijn E, van de Velde CJ (2002)
The above surgical rules can be applied in the Acute side effects and complications after short-term
CTV construction. The examples are depicted in preoperative radiotherapy combined with total mesorec-
the figure. It should be noted that the rules for tal excision in primary rectal cancer: report of a multi-
CTV lower border location presented in this center randomized trial. J Clin Oncol 20:817–825
14. Marijnen CAM, van de Velde CJ (2005) Impact of
figure are different to those recommended by short-term preoperative radiotherapy on health-related
other investigators [22]. quality of life and sexual functioning in primary rectal
cancer: report of multicenter randomized trial. J Clin
Oncol 23:1847–1858
15. Myerson RJ, Garofalo MC, El Naqa I et al (2009)
References Elective clinical target volumes for conformal therapy
in anorectal cancer: a radiation therapy oncology
1. Birgisson H, Påhlman L, Gunnarsson U et al (2005) group consensus panel contouring atlas. Int J Radiat
Occurrence of second cancers in patients treated with Oncol Biol Phys 74:824–830
radiotherapy for rectal cancer. J Clin Oncol 23: 16. NCCN Guidelines (2011) Rectal cancer, Version
6126–6131 1.2012. http://www.nccn.org. Accessed 26 Sept 2011
2. Birgisson H, Påhlman L, Gunnarsson U et al (2008) 17. Nelson H, Petrelli N, Carlin A et al (2001) National
Late gastrointestinal disorders after rectal cancer sur- Cancer Institute Expert Panel. Guidelines 2000 for
gery with and without preoperative radiation therapy. colon and rectal cancer surgery. J Natl Cancer Inst
Br J Surg 95:206–213 93:583–596
3. Blomqvist L, Glimelius B (2008) The ‘good’, the 18. Nijkamp J, Kusters M, Beets-Tan RG et al (2011)
‘bad’, and the ‘ugly’ rectal cancers. Acta Oncol Three-dimensional analysis of recurrence patterns in
47:5–8 rectal cancer: the cranial border in hypofractionated
154 K. Bujko

preoperative radiotherapy can be lowered. Int J Radiat 25. Sebag-Montefiore D, Stephens RJ, Steele R et al
Oncol Biol Phys 80:103–110 (2009) Preoperative radiotherapy versus selective
19. Nijkamp J, Doodeman B, Marijnen C et al (2012) Bowel postoperative chemoradiotherapy in patients with rec-
exposure in rectal cancer IMRT using prone, supine, or tal cancer (MRC CR07 and NCIC-CTG C016): a mul-
a belly board. Radiother Oncol 102(1):22–29 ticentre, randomised trial. Lancet 373:811–820
20. Peeters KCMJ, van de Velde CJ, Leer JWH (2005) 26. Syk E, Torkzad MR, Blomqvist L et al (2008) Local
Late side effects of short-course preoperative radio- recurrence in rectal cancer: anatomic localization and
therapy combined with total mesorectal excision for effect on radiation target. Int J Radiat Oncol Biol Phys
rectal cancer: increased bowel dysfunction in irradi- 72:658–664
ated patients - A Dutch Colorectal Cancer Group 27. Taylor FG, Quirke P, Heald RJ et al (2011) Preoperative
Study. J Clin Oncol 23:6199–6206 high-resolution magnetic resonance imaging can
21. Roach M 3rd, Nam J, Gagliardi G et al (2010) identify good prognosis stage I, II, and III rectal can-
Radiation dose-volume effects and the penile bulb. Int cer best managed by surgery alone: a prospective,
J Radiat Oncol Biol Phys 76(3 Suppl):S130–S134, multicenter, European study. Ann Surg 253:711–719
Review 28. van Gijn W, Marijnen CA, Nagtegaal ID et al (2011)
22. Roels S, Duthoy W, Haustermans K et al (2006) Preoperative radiotherapy combined with total
Definition and delineation of the clinical target vol- mesorectal excision for resectable rectal cancer:
ume for rectal cancer. Int J Radiat Oncol Biol Phys 12-year follow-up of the multicentre, randomised
65:1129–1142 controlled TME trial. Lancet Oncol 12:575–582
23. Sauer R, Becker H, Hohenberger W (2004) 29. Vordermark D, Schwab M, Ness-Dourdoumas R et al
Preoperative versus postoperative chemoradiotherapy (2003) Association of anorectal dose-volume histo-
for rectal cancer. N Engl J Med 351:1731–1740 grams and impaired fecal continence after 3D confor-
24. Stephens RJ, Thompson LC, Quirke P et al (2010) mal radiotherapy for carcinoma of the prostate.
Impact of short-course preoperative radiotherapy for Radiother Oncol 69:209–214
rectal cancer on patients’ quality of life: data from the 30. Yau I, Vuong T, Garant A et al (2009) Risk of hypogo-
Medical Research Council CR07/National Cancer nadism from scatter radiation during pelvic radiation
Institute of Canada Clinical Trials Group C016 ran- in male patients with rectal cancer. Int J Radiat Oncol
domized clinical trial. J Clin Oncol 28:4233–4239 Biol Phys 74:1481–1486
What Is the Contribution of
Intraoperative Radiotherapy 16
(IORT) in Tailoring Local Therapy
in Primary or Recurrent Rectal
Cancer?
Felipe A. Calvo

Contents 16.1 Introduction


16.1 Introduction ................................................ 155
The available evidences favor the use of neoad-
16.2 Locally Advanced Primary
Rectal Cancer ............................................. 155
juvant chemoradiation to maximize local con-
16.2.1 Pelvic Recurrence After Surgery Alone: trol in cT3–4 or N+ rectal cancer disease stages.
A Topographic Focus ................................... 155 Meta-analysis considering preoperative or
16.2.2 Presacral Recurrence: A Basic postoperative radiotherapy alone versus chemo-
Anatomical Study for Event
Explanation .................................................. 156
radiation strategies has shown significant
16.2.3 Topography of Pelvic Recurrence After improvement in local control by the use of
Combined Surgery and Radiotherapy .......... 156 preoperative chemoradiation. Surgery is the
16.3 Intraoperative Radiotherapy (IORT): decisive component of rectal cancer cures and
Topographic Results After a Limited local control achievement, but even in the con-
Intra-pelvic Radiation Boost ..................... 156 text of expert proven total mesorectal excision
16.3.1 Systematic Review Literature ...................... 156
(TME) performed, the local recurrence rate for
16.3.2 Randomized Trial......................................... 156
16.3.3 Single-Institution IORT Expert Data: surgery alone is in the range of 11–21% in
Topographic Failure Assessment ................. 157 unselected tumor stages [1]. In lymph node
16.3.4 Multi-institutional Pooled positive patients, local recurrence has been
Analysis Studies........................................... 157
reported superior in expert surgical groups:
16.4 Recurrent Rectal Cancer: Isolated 33% after unilateral lymph node dissection and
Pelvic Relapse ............................................. 158 14% for bilateral [2]. TME surgery and neoad-
16.4.1 Mayo Clinic 25 Years Experience ............... 158
16.4.2 Catharina Hospital and Leiden juvant chemoradiation are standards for daily
University Medical Center 1994–2008 practice.
Experience ................................................... 159
References ................................................................. 160
16.2 Locally Advanced Primary
Rectal Cancer

16.2.1 Pelvic Recurrence After Surgery


Alone: A Topographic Focus

F.A. Calvo The surgery alone scenario of quality practice and


Department of Oncology,
excellence describes [1] a local recurrence rate
Hospital General Universitario Gregorio Marañon,
Madrid, Spain related to T stage (12.2% T3, 21.4% T4),
e-mail: fcalvo.hgugm@salud.madrid.org N status (16% N+, 3.6% N−), and circumferential

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 155


DOI 10.1007/978-3-642-25005-7_16, © Springer-Verlag Berlin Heidelberg 2012
156 F.A. Calvo

margin involvement (20% yes, 6.2% no). This recurrences at 43 sites: 28 (65%) were in-field, 7
general pattern of local relapse can be further (16%) marginal, and 8 (19%) out-field radiother-
analyzed in terms of topographic distribution apy recurrences. The total rate of presacral
within the pelvic anatomy. After total mesorectal in-field recurrences was 41%, and the low pelvic
excision (TME), the presacral subsite is the domi- region was dominant for both pre- (60%) or post-
nant involved pelvic area with an incidence of operative (43%) irradiation. Pathologic N1–2 sta-
3.6% of all recurrences and 35% of the local tus was predictive of in-field locoregional
recurrences observed. Other sites of recurrences recurrence in multivariate analysis, while T4,
identified are lateral (18%), anterior (18%), anas- downstaging and pN status were significant in
tomosis (24%), and perineum (5%). univariant evaluation.

16.2.2 Presacral Recurrence: A Basic 16.3 Intraoperative Radiotherapy


Anatomical Study for Event (IORT): Topographic Results
Explanation After a Limited Intra-pelvic
Radiation Boost
The potential origin of local recurrence after
rectal cancer treatment in the presacral space 16.3.1 Systematic Review Literature
has been elegantly studied by Kusters and
coworkers [3]. The examination of ten human A 2011 systematic review identified 15 studies
fetal pelvises allowed to analyze the development selected out of 283 fully analyzed publications
of lymphatic structures of the human pelvis, in and 30 candidates to quality criteria evaluation
particular the lateral lymph node distribution and papers that describe results of 5-year local con-
its relationship with the mesorectum. Three- trol rates over 80% and survival close to 65% in
dimensional reconstruction of histological sections combined treatment containing an IORT compo-
showed that lateral lymph node tissues comprise nent in primary locally advanced rectal cancer.
a major proportion of the pelvic tissue volume. For localized recurrences, 5-year overall survival
There were no lymph nodes located in the pre- was 30%. The addition of IORT to conventional
sacral area. Connections between the mesorectal treatment, in the author’s criteria, reduces the
and extramesorectal lymph node system were incidence of local relapse within the radiation
found in all fetal pelvises. The authors hypothesized area over 10%. The IORT in terms of toxicity
that mobilizing the rectum lymph fluid and tumor concludes that it is a safe technique [5].
cells flow into the lateral lymphatic system,
which is left behind after TME and ultimately
the fluid leaks and is collected in the presacral 16.3.2 Randomized Trial
seroma.
A French multi-institutional randomized trial
performed with 142 patients, T3–4 or N+, M0
16.2.3 Topography of Pelvic stages, has not demonstrated an improvement in
Recurrence After Combined local control and disease-free survival in the
Surgery and Radiotherapy context of 40-Gy preoperative irradiation with
or without 18-Gy IORT pelvic boost [6]. The
The anatomical involvement of recurrences after 5-year local control rates were 91.8% versus
adjuvant (15%) or neoadjuvant (85%) radiother- 92.8%, and postoperative complications inci-
apy (5%) or chemoradiation (95%) describes an dence were 29.6% (IORT) versus 19.1% (p = 0.15).
estimated 5-year locoregional control of 91% [4]. Topography of pelvic recurrences and technical
In the MD Anderson experience (554 patients, characteristics of IORT boosting were not
1989–2001 period), 36 patients had locoregional reported.
16 What Is the Contribution of Intraoperative Radiotherapy (IORT) 157

16.3.3 Single-Institution IORT Expert A prognostic index to guide a tailored deci-


Data: Topographic Failure sion in locally advanced rectal cancer after multi-
Assessment modality treatment (neoadjuvant chemoradiation)
and IORT was proposed by Calvo and Diaz-
Single-institution IORT expert experiences have Gonzalez [9]. Based on gender and downstaging
been reported with a meticulous analysis of ana- response (mic versus mac categories), survival
tomical involvement of pelvic recurrences after varied from 100% (more favorable index) to 37%
combined modality therapy including IORT. (unfavorable). This institution has described, in
At the University of Heidelberg, Roeder et al. [7] an update involving 241 patients and 15 years
observed 17 (7%) local failures in 243 patients period [10], a 3% presacral recurrence in the con-
treated with a component of IOeRT (electron text of a 12% presacral abnormalities findings
IORT, 88 after neoadjuvant chemoradiation). detected by follow-up imaging studies, in which
Local control was correlated positively with the 9% (21 out of 28 patients) had nonmalignant pre-
absence of nodal involvement and complete sacral masses (Fig. 16.1).
resection. IOeRT boosted the presacral space
adjuvantly after surgical resection (10–15 Gy),
and 7 patients developed an in-field presacral 16.3.4 Multi-institutional Pooled
relapse (2.8%). Preoperative chemoradiation Analysis Studies
(3/17) had less pelvic recurrences than postop-
erative (10/17). T4 stage had the highest risk of The European pooled analysis study of IORT
relapse in retrovesical/retroprostatic site (3 out containing multimodality treatment included 605
of 5 recurrences), while node-positive specimens patients treated in four institutions with preoper-
had the highest risk for presacral recurrence ative radiotherapy (64% chemoradiation), sur-
(4 out of 7). gery with IOeRT, and adjuvant chemotherapy
Catharina Hospital and the University of (42%) [1]. Local recurrence was seen in 61
Leiden Medical Center have analyzed [8] the patients (12% at 5 years). The risk factors associ-
patterns of local recurrence following IORT ated with local recurrence were no downstaging,
containing combined modality therapy in the nodal metastasis, margin involvement, and no
context of a strategy not-a-fixed pelvic target for adjuvant chemotherapy. In patients who received
IORT boosting in 290 patients treated from 1994 adjuvant chemotherapy, the local recurrence rate
to 2006. After 5 years, 34 patients (13.2%) was 5.5% versus 12% in patients who did not
developed local recurrences: 47% presacral (5% (p = 0.026). Adjuvant chemotherapy improved
of the total cohort), which was the most com- local recurrences rate from 21.6% to 12.3%
mon site selecting only R0 patients (3.6%, 8/34 (p = 0.03) in patients with tumors in which no
local recurrences). In 53% of procedures, the downstaging had occurred after preoperative
IORT target was lateralized and in 16% was treatment and in positive margin specimens,
ventral. In-field IORT recurrences were consid- 23.8% versus 53.4% (p = 0.03). No information
ered 17 out of 34: 11 had a lateral or ventral was available in terms of topography of pelvic
positioning element. Presacral local recurrence recurrences.
had a significant correlation (uni- and multivari- The observation of the contribution of adju-
ate analysis) with cancer involved circumferen- vant chemotherapy to local control promotion
tial margin or tumor distance more than 5 cm was interpreted, speculating with the possibility
from the anal verge. On the other hand, local that high-dose local irradiation, in a single frac-
recurrence was not influenced (univariate) but tion as it is delivered by IOeRT, may sensitize the
lymph node or margin positivity and no down- surgical and the IORT boosted target for subse-
staging, but in multivariate analysis, margin quent adjuvant chemotherapy. This is a pioneer-
positive (25 patients, IORT dose-escalated 12.5– ing clinical and therapeutic observation of the
17.5 Gy) was significant. long-term proposed “seed and soil” biomedical
158 F.A. Calvo

Fig. 16.1 Three-dimensional virtual treatment planning of an IORT procedure with an electron beam positioned over
a limited area of the presacral space

paradigm. The large and mature nature of this 16.4 Recurrent Rectal Cancer:
cohort of patients allowed the authors to generate Isolated Pelvic Relapse
a prognostic model based on the calculation of an
individualized prognostic index formula. Overall 16.4.1 Mayo Clinic 25 Years Experience
survival of rectal cancer patients was able to be
grouped in four different prognostic categories Haddock and coworkers [11] have reported
using the following formula: results in 607 patients with recurrent colorectal
cancer that received an IOeRT (electrons) com-
Age over 70 years Yes:0.60;no:0 ponent of the treatment (70% rectal cancer, 46%
Male gender Yes:0.36;no:0 previous radiotherapy, 41% in-field relapses).
Any downstaging Yes:0;no:0.30 Survival at 5 and 10 years were 30% and 16%,
Lymph node positivity Yes:0.86;no:0 respectively. Local relapse at 5 years was observed
Margin positivity Yes:0.70;no:0
in 28% of patients. Survival was influenced by
Adjuvant Yes:0;no:0.64
chemotherapy
resection status, period of IORT (before or after
1997), and prior chemotherapy. Disease relapse
Survival ranged from 90% for category with in the central IORT boosted region at 10 years
prognostic index 0–0.5 or 20% for values over was 18% versus 12% regarding the fact of previ-
2.5 (Fig. 16.2). ous radiotherapy or not and 12% versus 16%
16 What Is the Contribution of Intraoperative Radiotherapy (IORT) 159

Fig. 16.2 ISIORT-European Survival functions


pooled analysis prognostic
model for survival generated 1.0
in patients treated with a
component of IORT using the
index formula described [10]
0.8

Overall survival
0.6

0.4

Prognostic index
0.2 0−0.5
0.5−2
2−2.5
2.5>
0.0

0.0 10.0 20.0 30.0 40.0 50.0 60.0


Months after surgery

regarding margin involvement. Neuropathy was resection had a significantly worse prognosis in
observed in 94 patients and was more common posterolateral recurrent subsite. Authors suggest a
with IORT doses exceeding 12.5 Gy. classification of subsite of pelvic recurrence as a
predictor for radical resection and consequently
promotion of improved survival.
16.4.2 Catharina Hospital and Leiden
University Medical Center Conclusions
1994–2008 Experience 1. IORT is a feasible, tolerable, and efficient
radiation-boosting technique that can be
Kusters, Rutten, and colleagues [12] described explored in tailored treatment for rectal
that radicality of resection and survival is influenced cancer patients (Table 16.1).
by subsite of locally recurrent rectal cancer after 2. Recommendations available to guide tai-
multimodality treatment including a component of loring IORT in primary disease in terms of
IORT. In 170 patients (30% not receiving IORT), local tumor control promotion are:
22% had a presacral recurrence, 34% lateralized • The unfavorable nature of nodal and
disease, 22% anastomotic, 23% anterior, and 2.6% margin positivity together with no
perineal. Local re-recurrence occurred in 54 downstaging effects. IORT is an alter-
patients (46%) after 5 years: 52% in the presacral native for further dose-escalation and
subsite, 20% posterolaterally, 17% anterolaterally, target volume redesign to improve local
and 11% other sites. Anastomotic recurrences had control under these conditions.
the highest cancer-specific survival (58%) • The tendency of T4 stage to recur in
(significantly better than the combined group anterior pelvic structures, although
34%). Presacral recurrences resulted in 19% IORT has also been reported to promote
cancer-specific survival, significantly worse than high local control rates in this disease
other recurrences in group (48%, p = 0.03). Radical category [14].
160 F.A. Calvo

Table 16.1 IOeRT 5-year results in primary and recurrent rectal cancer: local control, in-field recurrence, and factors
associated with local relapse
In-field IORT Adverse factors for local
Author/year (reference) Pts Local control recurrence relapse
Dubois 2011a [6]; 72 91% na na
Primary
Roeder 2007 [7]; 243 93% 2.8% presacral Postoperative
Primary chemoradiation (10/17)
Node positive
(4/7 presacral)
Kusters 2009 [8]; 290 86% 5.9% Margin positive
Primary 64% (lateral/ventral)
Calvo 2011 [8, 10]; 241 92% 3% presacral Node positive
Primary
Kusters 2010 [13]; 605 88% na No downstaging
Primary Nodal metastasis
Margin involvement
No adjuvant
chemotherapy
Haddock 2011 [11]; 607 77% (3 year) 12% R1/R2 26% vs. RO 15%
Recurrent Previous RT 31% vs.
17%
Kusters 2009 [12]; 170 54% 14.9% R1/R2 54% vs. R0 9%
Recurrent (57% presacral)
na not available, pts patients
a
Randomized trial

• The excellent local results obtained in cancer (previous radiotherapy, micro-/


more favorable disease risk factors macroscopic residue, previous chemo-
which might reconsider the need for therapy) and should be considered a
radiation treatment intensification. valuable treatment component to be
• The use of adjuvant chemotherapy further optimized (improved target
should be recommended after IORT definition, pharmacological radiation
treatment. modulation, etc.).
3. Tailored IORT may be further defined by
establishing correlations between biologi-
cal equivalent dose (BED) calculations, References
topographic patterns of recurrence, and
prognostic features for local effects. This 1. Kusters M, Marijnen CA, Van de Velde CJ et al (2010)
Patterns of local recurrence in rectal cancer; a study of
information is not available at present.
the Dutch TME trial. Eur J Surg Oncol 36:470–476
4. Recommendations available to guide tailor- 2. Kusters M, van de Velde CJ, Beets-Tan RG et al (2009)
ing IORT in localized recurrent disease in Patterns of local recurrence in rectal cancer: a single-
terms of local tumor control promotion are: center experience. Ann Surg Oncol 16:289–296
3. Kusters M, Wallner C, Lange MM et al (2010) Origin
• The surgical effort should be stressed
of presacral local recurrence after rectal cancer treat-
due to the excellent in-field control rates ment. Br J Surg 97:1582–1587
reported in completely resected tumor 4. Yu TK, Bhosale PR, Crane CH, Yyier RB et al (2008)
bed areas. Patterns of locoregional recurrence after surgery and
radiotherapy or chemoradiation for rectal cancer. Int J
• IORT reaches over 50% local control in
Radiat Oncol Biol Phys 71:1175–1180
the context of adverse features for local 5. Cantero-Muñoz P, Urién MA, Ruano-Ravina A (2011)
treatment, such as resistant/persistent Efficacy and safety of intraoperative radiotherapy in
16 What Is the Contribution of Intraoperative Radiotherapy (IORT) 161

colorectal cancer: a systematic review. Cancer Lett presacral radiation boost for rectal cancer. Radiother
306:121–133 Oncol 99(suppl 1):S18
6. Dubois JB, Bussieres E, Richaud P et al (2011) Intra- 11. Haddock MG, Miller RC, Nelson H et al (2011)
operative radiotherapy of rectal cancer: results of the Combined modality therapy including intraoperative
French multi-institutional randomized study. Radiother electron irradiation for locally recurrent colorectal
Oncol 98:298–303 cancer. Int J Radiat Oncol Biol Phys 79:143–150
7. Roeder F, Treiber M, Oertel S et al (2007) Patterns of 12. Kusters M, Dresen RC, Martin H et al (2009)
failure and local control after intraoperative electron Radicality of resection and survival after multimodal-
boost radiotherapy to the presacral space in combina- ity treatment is influenced by subsite of locally
tion with total mesorectal excision in patients with recurrent rectal cancer. Int J Radiat Oncol Biol Phys
locally advanced rectal cancer. Int J Radiat Oncol Biol 75:1444–1449
Phys 67:1381 13. Kusters M, Valentini V, Calvo FA et al (2010) Results
8. Kusters M, Holman FA, Martijn H et al (2009) Patterns of European pooled analysis of IORT-containing mul-
of local recurrence in locally advanced rectal cancer timodality treatment for locally advanced rectal cancer:
after intra-operative radiotherapy containing multimo- adjuvant chemotherapy prevents local recurrence rather
dality treatment. Radiother Oncol 92:221–225 than distant metastases. Ann Oncol 21:1279–1284
9. Díaz-Gonzalez JA, Calvo FA et al (2006) Prognostic 14. Valentini V, Coco C, Rizzo G et al (2009) Outcomes
factors for disease-free survival in patients with T3–4 of clinical T4M0 extra-peritoneal rectal cancer treated
or N + rectal cancer treated with preoperative chemo- with preoperative radiochemotherapy and surgery: a
radiation therapy, surgery, and intraoperative irradia- prospective evaluation of a single institutional experi-
tion. Int J Radiat Oncol Biol Phys 64:1122–1128 ence. Surgery 145:486–494
10. Calvo F, Serrano J, Lozano MA et al (2011) Presacral
evolutive events after post-neoadjuvant intraoperative
What Is the Contribution
of Brachytherapy in Tailoring 17
Local Therapy?

Jean-Pierre Gérard, Te Vuong, Jean-Michel


Hannoun-Lévi, and Arthur Sun Myint

Contents 17.1 Definition


17.1 Definition..................................................... 163
Brachytherapy is a generic term to describe radi-
17.2 Techniques of Brachytherapy ................... 164
ation techniques using short (less than 6 cm)
17.2.1 Iridium 192 Brachytherapy .......................... 164
17.2.2 Contact X-Ray 50 kV (CXRT) .................... 165 focus skin distance (FSD). It is by opposition
17.2.3 Intraoperative Brachytherapy....................... 166 with “teletherapy,” usually performed with linear
17.3 Clinical Results........................................... 166 accelerators and a long FSD (80 cm or more)
17.3.1 Endoluminal HDR 192 Iridium ................... 166 often called external beam radiotherapy (EBRT).
17.3.2 Interstitial Iridium Implant........................... 167 The dose distribution in brachytherapy depends
17.3.3 Contact X-Ray 50 kV .................................. 167 for a major part on the inverse square law which
17.4 Contribution of Brachytherapy implies that with a short FSD (1–6 cm), the radia-
in Tailoring Treatment............................... 167 tion dose fall off is very rapid (percentage depth
17.5 Perspective .................................................. 168 dose close to 50% at 0.5 cm from the “surface
17.5.1 In Fit Operable Patient ................................. 168 dose”). This rapid fall off (and dose inhomogene-
17.5.2 Inoperable Elderly Patient ........................... 168
ity) is mainly independent of the beam energy
References ................................................................. 169 (50 kV, 0.3 MV, or 1.2 MV). Broadly speaking,
brachytherapy can be performed with two gen-
eral approaches: radionuclide brachytherapy usu-
ally with iridium 192 or with iodine 125
permanent implant (usually called curietherapy
in Europe) or X-ray brachytherapy (contact ther-
J.-P. Gérard (*) • J.-M.Hannoun-Lévi
apy). Such brachytherapy techniques can be used
Department of Oncology,
Antoine Lacassagne Cancer Center, in rectal cancer mainly for accessible tumors with
33 Avenue Valombrose, Nice Cedex 2 06189 France moderate volume (not exceeding 40 cc).
Université de Nice, UNSA, Tailoring Local Treatment for Rectal Cancer.
Nice Cedex 2, France Surgery is the main treatment of rectal cancer. In
e-mail: jean-pierre.gerard@nice.fnclcc.fr this context, brachytherapy can be used in two
Te Vuong situations to tailor the local treatment with the
Department of Radiation Oncology, main following objectives:
Jewish General Hospital, McGill University,
• In association with surgery
Montreal, Canada
– To increase local control for locally advance
A.S. Myint
tumors
Department of Radiation Oncology,
Clatterbridge Centre for Oncology, – To increase the chance of conservative
Wirral, UK treatment for distal tumors

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 163


DOI 10.1007/978-3-642-25005-7_17, © Springer-Verlag Berlin Heidelberg 2012
164 J.-P. Gérard et al.

20 mm

5 mm

Fig. 17.1 Endoluminal HDR 192 iridium brachytherapy. the positioning of the applicator through the anus, and the
This figure is showing the rectal brachytherapy applicator, dose distribution in a sagittal plane
the loading of five channels in front of the rectal tumor,

• Alone 17.2.1 Iridium 192 Brachytherapy


– To cure early tumors (to palliate more
advanced lesions) 17.2.1.1 Endoluminal
It is well admitted that brachytherapy (except • The renaissance of this technique was pioneered
when used alone) has very little impact on in the 1990s by Sun Myint in Clatterbridge [11]
survival. and Vuong in Montreal [12]. It uses a high-dose-
rate (HDR) 192 source stepping in a dedicated
applicator. It is performed without general anes-
thesia. In this technique, the iridium sources are
17.2 Techniques of Brachytherapy at the periphery of the applicator (Fig. 17.1).
• Another applicator was designed in Vejle
These techniques have been used for many years. (Denmark). It uses also HDR 192, but the source
Radium interstitial or endoluminal treatments is in the middle of the applicator to increase the
were initiated in 1906 and contact X-ray 50 kV percentage depth dose. A lead protection can
in 1936. shield the normal rectal mucosa [6].
17 What Is the Contribution of Brachytherapy in Tailoring Local Therapy? 165

m1: 5.00 Gy 41.1 cm^3


Volume [cm^3]
60.0 M1

50.0

40.0

30.0

20.0

10.0

0
0 2.00 4.00 6.00 8.00 10.0
Dose [Gy]

Fig. 17.2 HDR 192 iridium implant for distal supra-anal implant through a transverse axial plane, the 3D recon-
rectal cancer. This figure is showing the perineal template struction of the implant, and the dose-volume histogram
fixed to the patient perineum and connected to the remote for the tumor volume
iridium unit, the CT scan slices showing the needle

17.2.1.2 Interstitial 17.2.2 Contact X-Ray 50 kV (CXRT)


• The technique was initiated in Lyon with iridium
192 low-dose rate in the early 1970s to replace The technique was popularized in the 1950s by
radium needle implant [9]. It is performed under Papillon using the Philips RT50™ machine [8]. It is
general anesthesia. It uses perineal template and an ambulatory treatment. It necessitates a good
4–6 needles implanted through the perineum. It clinical practice of rigid proctoscopy. The dose is
can be used for supra-anal rectal cancer up to delivered through a rectal applicator of 3 cm in
5 cm from the anal verge. The spacing between diameter. A dose between 15 and 40 Gy is delivered
needle is 1 cm. It is most of the time used in under vision control in 1–3 min. Total dose depend-
association with contact X-ray and/or EBRT. ing on the clinical situation is between 50 Gy/3
The dose delivered (Paris System) is between 15 Fr/3 weeks for postexcision CXRT to 90–130 Gy in
and 30 Gy. This interstitial implant can be per- 4 or 5 fractions over 5–8 weeks. Since 2009, a new
formed in the same way but with HDR I92 Iridium CXRT machine (Papillon 50™) has been designed
delivering 36–38 Gy in 10 fractions over 5 days in to replace the Philips RT50™. This new machine
case of iridium implant alone or 12–14 Gy in 3 allows direct vision of the tumor during treatment
fractions over 2 days in case of iridium implant and the use of 3 cm applicator but also smaller one
combined with EBRT or CRXT (Fig. 17.2). with 2.5 and 2.2 cm in diameter (Fig. 17.3).
166 J.-P. Gérard et al.

Fig. 17.3 Contact X-ray 50 kV. Male patient of 74 years Xeloda 1,600 mg/m2 on radiation day). Treatment deliv-
referred by a surgeon for a distal rectal adenocarcinoma ered with the Papillon 50 machine which allows direct
staged T2N0. Patient treated in October 2010 with CXRT viewing of the tumor during irradiation. It is possible to
(110 Gy in 4 fractions) and chemoradiotherapy CAP50 record the progressive shrinkage of the tumor until clinical
regimen (EBRT 50 Gy/25 F/5 weeks and concurrent complete response. Patient alive and well in May 2011

17.2.3 Intraoperative Brachytherapy suggests that this aggressive but well-tolerated


approach should be further investigated.
When patients present with T4 tumors, advanced
nodal metastases, or local recurrence in the pel-
vis, it is difficult to deliver radiation dose above 17.3 Clinical Results
55 or 60 Gy. In case of nonradical surgery (R2
resection), local control is exceptional. To increase 17.3.1 Endoluminal HDR 192 Iridium
safely the dose to gross residual (or R1 resection),
intraoperative radiotherapy (IORT) has been used 17.3.1.1 Preoperative Treatment
in various institutions. It is usually performed This technique can be used as an alternative to
with electrons but can also be done with iridium EBRT for T3–4 Nx M0 tumors. It is not possible to
192 [5] or in some anecdotal cases, with CXRT. treat circumferential stenosing lesions. Between
Although the relative role of IORT cannot be 1998 and 2008, Vuong has treated 285 patients
determined with certainty as there exist no ran- delivering a dose of 26 Gy in 4 fractions at the
domized trial, very encouraging local control rate periphery of the lesion. The immediate tolerance
17 What Is the Contribution of Brachytherapy in Tailoring Local Therapy? 167

was good in 95% of cases. Surgery was performed 17.3.3.2 CXRT Alone (or with EBRT) After
4–6 weeks after (anterior resection: 60%). The rate Local Excision for Malignant
of sterilized operative specimen (ypCR) was 27%. Polyp (T1N0)
At 5 years, the rate of local relapse was 5% [12]. In this adjuvant situation, a local control was
In Vejle, the endoluminal brachytherapy was achieved in 90% of cases in different institutions
used after preoperative EBRT radiochemotherapy treating between 20 and 60 patients [2].
(RT-CT). Out of 58 patients operated, the rate of
ypCR was 27%. Toxicity was low [6]. In 17.3.3.3 CXRT and EBRT for T2 Early T3N0
Clatterbridge, out of 29 patients treated with pre- in Elderly Unfit Patients
operative chemotherapy and a boost of 10 Gy These patients usually inoperable or elderly were
with HDR brachytherapy, 9 (31%) achieved a treated with curative intent. According to institu-
ypCR [11]. tion, the association was initiated with CXRT fol-
lowed by EBRT (± chemotherapy) or with EBRT
17.3.1.2 Combined with EBRT Without followed by CXRT. In T2 tumors, a 5-year local
Surgery in Unfit Patients control was achieved in 70–80% of patients and
Between 2005 and 2010, 41 patients were treated in 45–55% for T3 lesions [1,10].
in Montreal, McGill University (median age
82 years) without surgery. EBRT delivered 17.3.3.4 The Lyon R96.02 Randomized
40–45 Gy/4–5 weeks followed by HDR endolu- Trial
minal brachytherapy (30 Gy/3 F). With a follow- Between 1996 and 2001, 88 patients (T2: 19 pts
up of 14 months, a local control was achieved in – early T3: 69 pts) with a distal rectal cancer were
76% (31/41). Tolerance was good with G3 rectal randomized between preoperative EBRT
bleeding in seven patients and rectal stenosis in (39 Gy/13 F/3 weeks) or the same EBRT with a
three patients [12]. CXRT boost of 90 Gy/3 Fr. The main end point
was sphincter-saving treatment.
The clinical complete response (cCR) was 2%
17.3.2 Interstitial Iridium Implant in the EBRT alone group and 29% in the CXRT
group. After 10 years of follow-up, the colos-
In very low rectal cancer close to the anal canal, tomy-free rate is 40% with EBRT alone and 84%
iridium implant either with low- or high-dose rate in the CXRT group. Out of 45 pts in the CXRT
combined with EBRT (± chemotherapy) and group, 9 were able to preserve their whole rectum
CXRT is able to achieve 50% of long-term local (and anus) either with RT alone (seven patients)
control inoperable patients presenting with early or after transanal local excision (TLE) in two
T3N0 tumors [3]. patients [4].

17.3.3 Contact X-Ray 50 kV 17.4 Contribution of Brachytherapy


in Tailoring Treatment
17.3.3.1 CXRT Alone for T1N0
More than 1,300 patients with early rectal tumors • The advantages of brachytherapy in general
and strict selection were treated in different insti- over teletherapy with external beam radiother-
tutions in France, the UK, and North America apy are the following: excellent geographical
[3]. Local control was achieved in 80–90% of accuracy of dose distribution either image-
cases with no toxicity and good bowel function guided or eye-guided, no impact of tumor or
except in 10–20% of cases moderate rectal bleed- patient movements, and as the target volume is
ing due to radiation telangiectasia. small, the dose can be very high with a short
168 J.-P. Gérard et al.

treatment time. The clinical consequence is a 17.5.1 In Fit Operable Patient


high rate of clinical or pathological response
and a low toxicity. All these characteristics • Malignant polyp or T1N0 with first-line local
allow safe hypofractionation which is more excision: postoperative “adjuvant” irradiation
convenient for the patient than long course of with brachytherapy (± EBRT ± concurrent
5–7 weeks of EBRT. chemotherapy) should keep safely the rate of
• The endoluminal HDR iridium appears as an local recurrence below 10%. It can be per-
alternative for operable patient to preoperative formed with CXRT or interstitial HDR iridium
EBRT especially when given with the short implant for juxtarenal tumors.
course “nordic” 25 Gy/5 F regimen. • T1N0 <3 cm diameter
• It can also be used as a boost in T4 tumors to • Brachytherapy preferably with CXRT should
escalate safely the dose. be able to cure 90% of these tumors.
• For inoperable patient, when the tumor is too • T2N0 <4 cm diameter
large to be treated with CXRT, it is an efficient • A combination of chemoradiation with CXRT
way to escalate the dose in association with should achieve cCR in 80%–90% of cases and
EBRT and to achieve long-term control and can be followed by either local excision or
cure with acceptable toxicity. watch and wait close follow-up. Salvage radi-
• Interstitial iridium implant cal surgery should be proposed according to
• It is well adapted to irradiate the rectal tumor clinical and pathological findings. The rate of
close to or extending into the anal canal local relapse in the pelvis should be kept below
where it is better adapted than endoluminal 10%. Such an approach is still in the field of
brachytherapy or CXRT. clinical research.
CXRT is the more accurate technique to • Early “good” T3a in the distal rectum
irradiate under eye guidance accessible rectal • Achieving cCR with a combination of chemo-
tumors not exceeding 4 cm in diameter. It can be radiation and brachytherapy should increase
used either alone or after local excision for safely the chance of sphincter-saving surgery
T1N0. It can be used in combination with EBRT and, if possible, organ preservation. A new
(± chemotherapy) for inoperable patients with randomized trial to duplicate the results of the
T202 early T3N0 lesions. It is the only tech- Lyon R96.02 trial would be of great interest.
nique whose benefit was proven by a random-
ized trial.
17.5.2 Inoperable Elderly Patient

17.5 Perspective Brachytherapy should play an important role to


cure some of these patients and sometimes to pal-
It an era of treatment individualization for rectal liate large radioresistant lesions. In case of tumor
cancer, brachytherapy should play an important exceeding 50% of circumference, endoluminal
and increasing role mainly to improve the chance brachytherapy usually combined with short-
of conservative treatment either in operable or course EBRT should play a predominant role.
inoperable patients. To be able to run such important trials, radia-
Prospective clinical trials phase II and if pos- tion oncologists must be trained to master clini-
sible phase III such as the CONTEM trials using cal examination of rectal tumors and especially
CXRT should demonstrate the role of brachyther- rigid proctoscopy. Proctoscopy should allow to
apy in the following situations after careful selec- tatoo the tumor site, to take biopsy for response
tion of the patients [7]. evaluation, and to implant fiducial marker.
17 What Is the Contribution of Brachytherapy in Tailoring Local Therapy? 169

A quality assurance program of all these different 5. Goes RN, Beart RW Jr, Simons AJ et al (1997) Use of
brachytherapy techniques is needed. A regular brachytherapy in management of locally recurrent
rectal cancer. Dis Colon Rectum 40:1177–1179
cooperation with the other specialists is manda- 6. Jakobsen A, Mortensen JP, Bisgaard C et al (2006)
tory. Finally, informed patients should agree to Preoperative chemoradiation of locally advanced T3
participate in these clinical trials or observational rectal cancer combined with an endorectal boost. Int J
studies. Radiat Oncol Biol Phys 64(2):461–465
7. Lindegaard J, Gerard JP, Sun Myint A et al (2007)
Whither papillon? Future directions for contact radio-
Disclosure Jean-Pierre GERARD is the medical advisor therapy in rectal cancer. Clin Oncol (R Coll Radiol)
of the Ariane Medical systems company (UK). 19(9):738–741
8. Papillon J (1975) Intracavitary irradiation of early
rectal cancer for cure. A series of 186 cases. Cancer
References 36(2):696–701
9. Papillon J, Montbarbon JF, Gerard JP et al (1989)
Interstitial curietherapy in the conservative treatment
1. Aumock A, Birnbaum EH, Fleshman JW et al (2001) of anal and rectal cancers. Int J Radiat Oncol Biol
Treatment of rectal adenocarcinoma with endocavi- Phys 17(6):1161–1169
tary and external beam radiotherapy: results for 199 10. Sun Myint A, Grieve RJ, McDonald AC (2007)
patients with localized tumors. Int J Radiat Oncol Biol Combined modality treatment of early rectal cancer:
Phys 51(2):363–370 the UK experience. Clin Oncol (R Coll Radiol)
2. Gérard JP, Chapet O, Romestaing P et al (2000) Local 19(9):674–681
excision and adjuvant radiotherapy for rectal adeno- 11. Sun Myint A, Mukhopadhyay T, Ramani VS et al
carcinoma T1–2N0. Gastroenterol Clin Biol (2010) Can increasing the dose of radiation by HDR
24(4):430–435 brachytherapy boost following preoperative chemora-
3. Gerard JP, Romestaing P, Chapet O (2003) Radio- diotherapy for advanced rectal cancer improve surgi-
therapy alone in the curative treatment of rectal carci- cal outcome. Colorectal Dis 12(suppl 2):30–36
noma. Lancet Oncol 4(3):158–166 12. Vuong T, Richard C, Niazi T (2010) High dose rate
4. Gerard JP, Chapet O, Nemoz C et al (2004) Improved endorectal brachytherapy for curable rectal cancer.
sphincter preservation in low rectal cancer with Semin Colon Rectal Surg 21:115–119
high-dose preoperative radiotherapy: the lyon R96–02
randomized trial. J Clin Oncol 22(12):2404–2409
Part V
Q&As on Chemotherapy
Should Oxaliplatin Be Added
to Preoperative Chemoradiation? 18
Carlo Aschele

Contents 18.1 Disease Background


18.1 Disease Background ...................................... 173
Despite major improvements during the last
18.2 Rationale for Incorporating
Oxaliplatin in the Preoperative
20 years, the management of patients with locally
Treatment of Rectal Cancer ......................... 174 advanced rectal cancer is far from satisfactory
and mortality rates remain high. In particular,
18.3 Phase I–II Studies ......................................... 174
local recurrences have been substantially reduced
18.4 Phase III Studies ........................................... 175 with the combination of accurate surgery, pre- or
References ................................................................. 179 postoperative chemoradiation, and sequential
adjuvant chemotherapy but remain a major prob-
lem in subsets of patients with more advanced/
aggressive disease [2,3]. Distant metastases
develop in approximately 30–40% of rectal can-
cer patients. In contrast with local recurrence,
this figure has remained substantially unchanged
over the last 20 years with only minor improve-
ments achieved with postoperative fluorouracil-
based chemotherapy [2,3]. Now that local
recurrence rates are much lower due to optimized
local treatment, distant recurrence has thus
become the major form of failure in these patients.
In addition, more than 20% of clinically resect-
able patients are found to actually have a positive
circumferential resection margin after surgery
with a significantly increased risk of local recur-
rence and distant metastases and reduced 5-year
C. Aschele survival rates. Of note, this negative prognostic
Department of Medical Oncology,
effect is particularly marked for patients with a
Ospedale Sant’Andrea e del Felettino,
ASL 5 – Liguria, La Spezia, Italy positive circumferential resection margin after
e-mail: carlo.aschele@asl5.liguria.it neoadjuvant treatment [13].

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 173


DOI 10.1007/978-3-642-25005-7_18, © Springer-Verlag Berlin Heidelberg 2012
174 C. Aschele

18.2 Rationale for Incorporating 18.3 Phase I–II Studies


Oxaliplatin in the Preoperative
Treatment of Rectal Cancer Week 1 and 5 schedules of oxaliplatin in combi-
nation with 5-fluorouracil and radiation have
5-Fluorouracil has been the mainstay of both been initially evaluated in a series of phase I
chemoradiation regimens and adjuvant chemo- studies of preoperative chemoradiotherapy in
therapy programs for rectal cancer. Potentiation the setting of locally advanced rectal cancer
of these regimens by incorporation of new drugs [9,10,17]. These studies demonstrated the feasi-
active in colorectal cancer may improve the con- bility of adding oxaliplatin to 5-fluorouracil and
trol of distant metastases. standard preoperative pelvic radiation with a
Oxaliplatin is a diaminocyclohexane (DACH) – high delivered dose intensity, acceptably low
platinum compound, active in several solid tumor gastrointestinal toxicity, neutropenia or early
types, including some cisplatin/carboplatin postoperative complications. The optimal dose
refractory neoplasm as colorectal cancer. The on this 4-week schedule in combination with
main mechanism of action, like cisplatin, is medi- infusional 5-fluorouracil and radiation therapy
ated through the formation of DNA adducts, but was defined at 130 mg/m2 [9]. These initial stud-
DACH-platinum adducts are bulkier and more ies also provided clinical evidence supporting
hydrophobic than cisplatin adducts. the preclinical rationale for the incorporation of
The improvement in disease free and overall oxaliplatin in chemoradiation programs for rec-
survival achieved when oxaliplatin was added to tal cancer with rates of pathological complete
5-fluorouracil as adjuvant treatment for com- responses (pCRs) that have generally been
pletely resected intraperitoneal colon cancer [1] above 25% [9,10,17].
suggests that incorporating this agent in the treat- A weekly schedule of oxaliplatin administra-
ment programs for rectal cancer may improve the tion concomitant to continuous infusion
control of micrometastases at distant sites. 5-fluorouracil and preoperative chemoradiation
Oxaliplatin also increases the antitumor activ- has been developed by our group in Italy [2–4].
ity of 5-fluorouracil in metastatic colorectal can- Weekly administration of oxaliplatin in combina-
cer [8] and has been shown to be a radiation tion with pelvic radiotherapy may present several
sensitizer in preclinical models with optimal advantages compared to administration every
efficacy when it is given concomitantly with radi- 3–4 weeks. This schedule may in fact result in
ation, presumably by inhibiting postradiation reduced acute toxicity, thanks to dose fraction-
DNA repair [6]. Its combination with ation, and improved control of side effects, thanks
5-fluorouracil and radiation in the preoperative to better modulation of drug delivery. The high-
setting may thus improve also tumor shrinkage dose density of a weekly schedule of administra-
before surgery. This may be particularly impor- tion may also be optimal to maximize the
tant in low-lying tumors when sphincter preser- inhibition of sublethal, radiation-induced DNA
vation is attempted and in patients with bulky T3, damage repair (advocated as the main mechanism
T4, and tethered tumors where R0 resections are for the radiosensitizing effect of oxaliplatin) [7]
difficult to achieve unless the tumor is adequately (Table 18.1).
shrunk before the operation. Results from the pivotal phase I–II study of
Oxaliplatin has other pharmacological proper- this regimen have shown that the recommended
ties that are favorable for its incorporation in the weekly dose of oxaliplatin (given for six times on
preoperative treatment of rectal cancer. In partic- the first day of each week of treatment at escalat-
ular, the toxicity profile with a low incidence of ing doses from 25 up to 60 mg/m2) in combina-
gastrointestinal toxicity makes this agent a good tion with pelvic radiotherapy (50.4 Gy – 28
drug for combination with pelvic radiotherapy fractions) and infused 5-fluorouracil (adminis-
and concurrent infused 5-fluorouracil. tered for the entire duration of radiotherapy at
18 Should Oxaliplatin Be Added to Preoperative Chemoradiation? 175

Table 18.1 Phase I–II studies Number Number Grades III–IV


of oxaliplatin and pelvic Regimen of studies of patients pCR (%) toxicity (%)
radiation therapy
FU + OXA 15 450 7–36 6–39
CAPE/UFT + OXA 14 616 6–29 6–33
RTX + OXA 4 160 14 7–23
Abbreviations: pCR pathologic complete response, FU fluorouracil, OXA
oxaliplatin, CAPE capecitabine, UFT uracil/tegafur, RTX raltitrexed

225 mg/m2/day) is 60 mg/m2 given weekly ×6 Of note, all of these studies have used a
[4]. The feasibility of this regimen and the rec- weekly schedule for oxaliplatin administration
ommended dose of weekly oxaliplatin previously building on the chemoradiation regimen origi-
established in our study (60 mg/m2/week) were nally generated by our group. Only our study
confirmed in CALGB 89901 [16]. (the Italian STAR-01 trial), however, maintained
At the recommended dose, this regimen allows the six oxaliplatin courses as in the original regi-
therefore to deliver a total dose of oxaliplatin men. Both the NASBP and the French study had
(360 mg/m2 of over a 6-week time concomitant in fact five oxaliplatin administrations while
to radiation) substantially higher compared to oxaliplatin was administered on days 1, 8, 22,
other oxaliplatin-based chemoradiation programs and 29 in the German study that prescribed a
[9,10,17]. In addition, this oxaliplatin dose is poten- chemotherapy break during the third week of
tially active also systemically with a dose intensity radiation.
approximately 50% higher compared to the regi- As to control arms, the STAR-01, NSABP,
mens commonly used in the treatment of metastatic and French trials used exactly the same FP dose
colorectal cancer [8]. This schedule has therefore and schedule as in the experimental arm, while a
an added advantage over a traditional week 1 and 5 shorter infusion with higher 5-fluorouracil daily
chemotherapy program in that it incorporates a doses (mimicking the chemoradiation regimens
dose and schedule of both oxaliplatin and used in the treatment of anal cancer) was used in
5-fluorouracil known to be systemically active. the control arm of the German study.
This may be particularly important to optimize Radiotherapy intensification was pursued
both local control, with an enhanced chemoradia- in the experimental arm in the French study
tion regimen, and the control of (micro)metastases (45 Gy in the control arm vs. 50 Gy in the experi-
at distant sites with the concomitant administration mental arm) while the planned radiotherapy dose
of systemically active combination chemotherapy. was 50.4 Gy for both arms in the other three
studies.
Data on safety, surgery, and response to preop-
18.4 Phase III Studies erative chemoradiation have been reported for all
of these studies [5,11,14,15]. These results
There are four completed phase III randomized confirm that weekly oxaliplatin can be added to
clinical trials assessing the value of oxaliplatin FP-based preoperative chemoradiation even
combined with fluoropyrimidines (FPs) and pre- though toxicity is increased. Unexpected adverse
operative external-beam pelvic radiation in locally events were not reported, and there was no excess
advanced rectal cancer (Table 18.2). Infused treatment mortality in the oxaliplatin arms; over
5-fluorouracil was the FP backbone in the STAR- 95% of patients were operated as planned within
01 and CAO/ARO/AIO studies, while weekly 6–8 weeks from the end of chemoradiation inde-
oxaliplatin was combined with capecitabine in pendent of the preoperative treatment they had
the French ACCORD trial and either capecitabine received, and surgical mortality was also low and
or infused 5-fluorouracil in the NSABP study that similar with or without oxaliplatin. Treatment
entailed a 2 × 2 factorial design. compliance was reduced with an apparent
176 C. Aschele

Table 18.2 Phase III randomized studies of FP-based preoperative chemoradiation ± oxaliplatin
Primary Postoperative
Study n endpoint Status Control arm Experimental arm treatment
ACCORD 598 pCR Reported Cape 1,600 mg/m2/
Cape 1,600 mg/m2/ Center choice
0405 (primary day d1–5 weekly day d1–5 weekly ×5;
endpoint) ×5; RT 45 Gy OXA 50 mg/m2 d1
weekly ×5; RT 50 Gy
STAR-01 747 OS Reported CI FU 225 mg/m2/ CI FU 225 mg/m2/ FU/LV
(secondary day d1–35; RT day d1–35; OXA (both arms)
endpoint) 50.4 Gy 60 mg/m2 d1 weekly
×6; RT 50.4 Gy
NSABP R-04 1,608 LR Reported CI FU 225 mg/m2/ CI FU 225 mg/m2/ Center choice
(secondary day or Cape day or Cape
endpoint) 1,650 mg/m2/d d1–5 1,650 mg/m2/d d1–5
weekly ×5; RT weekly ×5; OXA
46 Gy 50 mg/m2 d1 weekly
(+5.4–10.8 Gy) ×5; RT 46 Gy
(+5.4–10.8 Gy)
CAO/ARO/ 1,265 DFS Reported CI FU 1,000 mg/ CI FU 250 mg/m2 FU 500 mg/m2
AIO-04 (secondary m2/day d1–5, d1–14, 22–35; OXA day ×5 q 4 week
endpoint) 29–33; RT 50.4 Gy 50 mg/m2 d1, 8, 22, (4 cycles) vs.
29; RT 50.4 Gy mFOLFOX-6
(8 cycles)
PETACC-6 634 (1,090) DFS Accruing Cape 1,650 mg/m2/ Cape 1,650 mg/m2/ Cape 2,000 mg/
day d1–33; RT day d1–33; OXA m2/day d1–14 q
45 Gy (±5.4 Gy) 50 mg/m2 d1 weekly 3 week ± OXA
×5; RT 45 Gy 130 mg mg/m2
(±5.4 Gy) d1 q 3 week
(6 cycles)
Abbreviations: pCR pathologic complete response, OS overall survival, LR local relapse, DFS disease-free survival,
Cape capecitabine, RT pelvic radiotherapy, OXA oxaliplatin, CI continuous infusion, FU fluorouracil, mFOLFOX-6
modified FOLFOX-6

relationship between the oxaliplatin dose deliv- be less pronounced in the German study, possibly
ered concomitant to radiation and the proportion because of the lower cumulative dose of oxalipla-
of patients receiving a reduced radiotherapy dose tin administered along with radiation. The higher
(6% in the German study where 200 mg/m2 of rate of severe toxicity in the control arm of this
oxaliplatin were delivered concomitant to radia- study (22% compared to less than 10% in the
tion, 13% in the French study with 250 mg/m2, Italian and French studies) provides an alterna-
and 16% in the STAR-01 study with 360 mg/m2 tive explanation for the less evident increment
of oxaliplatin added to 5-fluorouracil and radia- observed with the addition of oxaliplatin.
tion). However, even in the STAR study that Despite the promising phase II data (Table 18.1),
entailed the highest added oxaliplatin dose, more these randomized studies indicate that adding
than 90% of the patients in the oxaliplatin arm oxaliplatin to preoperative 5-fluorouracil and
received at least 45 Gy and more than 80% had at pelvic radiation does not improve primary tumor
least 5 weekly administrations of oxaliplatin response. pCR rates were identical, or nearly
(Tables 18.3 and 18.4). identical, in the experimental compared to the
The addition of oxaliplatin resulted in more control arms in the STAR-01 and NSABP-R04
toxicity with grade III–IV events reported in studies and numerically, but not significantly,
more than 20% of patients and rates of grade increased in the French study (that also entailed
III–IV diarrhea between 12% and 15% in all the intensified radiotherapy in the oxaliplatin-
four studies. This increase in toxicity appears to containing arm). A 3.7% absolute difference in
18 Should Oxaliplatin Be Added to Preoperative Chemoradiation? 177

Table 18.3 Treatment compliance and safety of preoperative chemoradiation ± oxaliplatin in randomized
phase III studies
STAR-01 ACCORD 0405 NSABP R-04 CAO/ARO/AIO-04
Study arm, n Study arm, n Study arm, n Study arm, n
Ctr OXA Ctr OXA Ctr OXA Ctr OXA
379 352 293 291 622 631 624 613
RT full dose, % 92 84 100 87 nr nr 95 94
OXA full dose, % – 66a – nr – nr – 85c
Grade III–IV toxicity, %
Overall 8 24b 11 25b nr nr 22 23
Diarrhea 4 15b 3 13b 7 15b 8 12
Neuropathy (³G2) 0 8b 0.4 6b b
nr nr <1 2c
Abbreviations: ctr control arm, OXA oxaliplatin-containing arm, RT radiotherapy, CT chemotherapy
a
83% received at least 5 weekly courses
b
Statistically significant
c
Chemoradiation full dose

Table 18.4 Response to preoperative chemoradiation ± oxaliplatin in randomized phase III studies
STAR-01 ACCORD NSABP R-04 CAO/ARO/AIO-04
Study arm, n Study arm, n Study arm, n Study arm, n
Ctr OXA Ctr OXA Ctr OXA Ctr OXA
379 352 293 291 622 631 624 613
ypCR, % 16 16 14 19 19 21 13 17*
ypT0, % 18 19 14 20 nr nr 13 18
ypN+, % 26 29 30 28 nr nr 30 28
R0 resection, % 94 97 nr nr nr nr 92 90
CRM + (£1 mm), % 7 4 13 8 nr nr 6 5
SSS, % 78 79 75 75 64 60 71 69
Abbreviations: ctr control arm, OXA oxaliplatin-containing arm, ypCR pathological complete response, CRM circum-
ferential resection margin, SSS sphincter-saving surgery
*p = 0.045 (unplanned exploratory analysis)

favor of the oxaliplatin arm that reached statis- not have TME surgery, and at least 90% had an
tical significance in an exploratory unplanned R0 resection. This may have contributed to lower
analysis was reported in the German study. Other the pCR rate compared to initial phase II studies.
measures of response to preoperative chemoradi- The quality of surgery and pathology indeed
ation (including tumor regression grade, the pro- affect the ability to detect residual tumor after
portion of patients with residual tumor confined preoperative chemoradiation and is therefore
to the muscular layer of the rectal wall, the rates inversely correlated with the pCR rate.
of node-positive disease found at surgery, and The lower pCR rate observed with OXA in
the incidence of circumferential resection margin these randomized studies probably reflects also
positivity) were also similarly distributed among the better control for the multiple factors affect-
patients treated with or without oxaliplatin in ing response to preoperative chemoradiation
the four studies ruling out any clinically relevant and the larger sample size compared to phase II
effect on primary tumor response. assessment. This underlines the crucial impor-
Although surgery details are not available for tance of properly designed, adequately powered
NSABP-R04, in the three European trials, the randomized multidisciplinary studies with stan-
median number of examined lymph nodes was dardized and optimized radiological staging,
between 12 and 15, less than 10% of patients did radiotherapy, TME surgery, and pathologic
178 C. Aschele

evaluation to assess the effect of new treatment long-term outcome in a phase III trial are pre-
strategies in rectal cancer. dicted comparing pCR rates between treatment
This lack of improvement in tumor response arms. The mechanisms of cytotoxicity and radio-
does not appear to be explained by a lower adher- sensitization may in fact not be the same, and
ence to the treatment program due to the increased local tumor regression does not necessarily pre-
toxicity in the oxaliplatin arms. In the STAR-01 dict responsiveness of disseminated tumor cells
study, where all measures of compliance were toward the chemotherapy component of combined
significantly reduced in the group randomized to modality treatment programs.
receive oxaliplatin compared to the control group, Of interest, in these studies, extrapelvic metas-
pCR rates were similar between the two arms also tases were found at surgery less frequently in
in exploratory analyses restricted to patients that patients treated with oxaliplatin compared to those
completed all the planned radiation and chemo- receiving single-agent FP and radiation, although
therapy as assigned [5]. In addition, even in the statistical significance was reached only in the
study that had the highest treatment compliance in STAR-01 trial.
the oxaliplatin arm, with 85% of patients receiving Further follow-up is thus required to establish
full-dose chemoradiation [14], a marginal, clini- if the early introduction of oxaliplatin in the man-
cally not relevant, although statistically significant, agement of locally advanced rectal cancer can
4% increase in the pCR rate was observed. improve longer-term outcomes such as disease-
These results indicate therefore that oxalipla- free and overall survival. At the present time,
tin is not a clinically effective radiation sensitizer however, oxaliplatin has no role in the neoadju-
in rectal cancer (or at least that oxaliplatin does vant radiochemotherapy of this disease and should
not increase radiosensitization achieved with not be added to preoperative chemoradiation.
5-fluorouracil or capecitabine administration Single-agent FPs thus remain the standard che-
concomitant to radiation). motherapy in this setting. Given this lack of effect
Of note, the pCR rates in the control arms of on primary tumor response, even future favorable
these four studies appear to be higher than those results in long-term efficacy should be weighed
reported in less recent studies, probably thanks to against the increased toxicity observed when
optimized radiotherapy techniques and higher oxaliplatin has been added to preoperative chemo-
radiation doses. In particular, pCR rates in the radiation. The control of metastases at distant
control arm were above 15% in the ACCORD- sites could in fact be improved, also adding oxali-
PRODIGE, STAR-01, and NSABP R-04 trials all platin in the postoperative setting at a lower toxic-
involving continuous administration of the FP ity cost as successfully done in colon cancer.
(protracted 5-fluorouracil infusion or oral capecit- Alternative strategies are thus required to
abine) for all the duration of radiotherapy. It is improve primary tumor response to preoperative
thus conceivable that optimal radiotherapy with treatment in patients with locally advanced rectal
optimal radiosensitization by FPs already maxi- cancer. Induction chemotherapy and image-
mizes primary tumor response with little or no guided radiotherapy techniques are actively
room for further improvement with additional investigated at this end. Preclinical data also sug-
radiation sensitizers. gest that the synergism between oxaliplatin,
Primary tumor response was the main endpoint 5-fluorouracil, and radiation is restricted to cell
only for the French study, while the other studies lines with specific gene expression profiles [12].
were designed to test the impact of oxaliplatin on Future studies of novel therapies in rectal cancer
long-term efficacy endpoints (overall survival for should thus include molecular profiling to indi-
the STAR-01 study, local relapse in NSABP R04, vidualize therapy protocols and identify patients
and disease-free survival for the German trial). that may achieve a good response even with
The lack of effect on primary tumor response does standard treatment and those demonstrating
not contradict this hypothesis. In rectal cancer, aggressive biology that could benefit from a more
there is in fact no evidence that differences in intensified approach.
18 Should Oxaliplatin Be Added to Preoperative Chemoradiation? 179

References 10. Gérard JP, Chapet O, Nemoz C et al (2003)


Preoperative concurrent chemoradiotherapy in locally
advanced rectal cancer with high-dose radiation and
1. Andre T, Boni C, Mounedji-Boudiaf L et al (2004)
oxaliplatin-containing regimen: the Lyon R0–04
Oxaliplatin, fluorouracil, and leucovorin as adjuvant
phase II trial. J Clin Oncol 21:119–1124
treatment for colon cancer. N Engl J Med 350:
11. Gérard JP, Azria D, Gourgou-Bourgade S et al (2010)
2343–2351
Comparison of two neoadjuvant chemoradiotherapy
2. Aschele C, Lonardi S (2007) Addition of weekly
regimens for locally advanced rectal cancer: results of
oxaliplatin to standard preoperative chemoradiation
the phase III trial ACCORD 12/0405-prodige 2. J Clin
for locally advanced rectal cancer. J Clin Oncol
Oncol 28:1638–1644
25:602–603
12. Magné N, Fischel JL, Formento P et al (2003)
3. Aschele C, Lonardi S (2007) Multidisciplinary treat-
Oxaliplatin-5-fluorouracil and ionizing radiation.
ment of rectal cancer: medical oncology. Ann Oncol
Importance of the sequence and influence of p53 status.
18:1908–1915
Oncology 64:280–287
4. Aschele C, Friso ML, Pucciarelli S et al (2005) A phase
13. Nagtegaal ID, Quirke P (2008) What is the role for the
I-II study of weekly oxaliplatin, 5-fluorouracil continu-
circumferential margin in the modern treatment of
ous infusion and preoperative radiotherapy in locally
rectal cancer? J Clin Oncol 26:303–312
advanced rectal cancer. Ann Oncol 16:1140–1146
14. Rodel C, Becker H, Fietkau R et al (2011)
5. Aschele C, Cionini L, Lonardi S et al (2011) Primary
Preoperative chemoradiotherapy and postoperative
tumor response to preoperative chemoradiation with
chemotherapy with 5-fluorouracil and oxaliplatin
or without oxaliplatin in locally advanced rectal can-
versus 5-fluorouracil alone in locally advanced rectal
cer: pathologic results of the STAR-01 randomized
cancer: first results of the German CAO/ARO/AIO-
phase III trial. J Clin Oncol 29:2773–2780
04 randomized phase III trial. J Clin Oncol 29(suppl;
6. Blackstock AW, Hess S, Chaney S et al (1999)
abstract LBA3505)
Oxaliplatin: in vitro evidence of its radiation sensitiz-
15. Roh MS, Yothers GA, O’Connell MJ et al (2011) The
ing activity. Pre-clinical observations relevant to clini-
impact of capecitabine and oxaliplatin in the preop-
cal trials. Int J Radiat Oncol Biol Phys 45:253–254
erative multimodality treatment in patients with carci-
7. Cividalli A, Ceciarelli F, Livdi E et al (2002)
noma of the rectum: NSABP R-04. J Clin Oncol
Radiosensitization by oxaliplatin in a mouse adeno-
29(suppl; abstract 3503)
carcinoma: influence of treatment schedule. Int J Radiat
16. Ryan DP, Niedzwiecki D, Hollis D et al (2006) Phase
Oncol Biol Phys 52:1092–1098
I/II study of preoperative oxaliplatin, fluorouracil, and
8. de Gramont A, Figer A, Seymour M et al (2000)
external-beam radiation therapy in patients with
Leucovorin and fluorouracil with or without oxalipla-
locally advanced rectal cancer: cancer and leukemia
tin as first-line treatment in advanced colorectal cancer.
group B 89901. J Clin Oncol 24:2557–2562
J Clin Oncol 18:2938–2947
17. Sebag-Montefiore D, Glynne-Jones R, Falk S et al
9. Freyer G, Bossard N, Romestaing P et al (2001)
(2001) Preoperative radiation and oxaliplatin in com-
Addition of oxaliplatin to continuous fluorouracil
bination with 5-fluorouracil (5-FU) and low-dose leu-
l-folinic acid and concomitant radiotherapy in rectal
covorin (LV) in locally advanced rectal cancer. Proc
cancer: the Lyon R97–03 phase I trial. J Clin Oncol 19:
Am Soc Clin Oncol 21:146a, (abstract 580)
2433–2438
Should Biologic Targeted Agents
Be Combined with Preoperative 19
Chemoradiation in Rectal Cancer?

Pieter-Jan Cuyle and Eric Van Cutsem

Contents 19.1 Introduction


19.1 Introduction ................................................ 181
Over the last decades, the clinical management of
19.2 EGFR Inhibitors in Rectal Cancer........... 182
locally advanced rectal cancer (LARC) has
19.3 VEGF Inhibitors in Rectal Cancer........... 186 changed substantially. Improvements in preoper-
19.4 Combined EGFR and VEGF Inhibition ative staging techniques with magnetic resonance
in Rectal Cancer ......................................... 188 imaging (MRI) and endoscopic ultrasonography
References ................................................................. 189 (EUS), the use of neoadjuvant therapy, improved
and innovative surgical approach, and standard-
ized recommendations for histopathological
analysis (tumor regression grading (TRG) and
circumferential resection margin (CRM) involve-
ment) have gradually improved local control rates
and the outcome of patients with rectal cancer.
Preoperative radiotherapy with concomitant infu-
sional 5-fluorouracil (5FU) or oral capecitabine,
followed by total mesorectal excision (TME), has
become the standard of care for patients with T3/
T4 or lymph node positive rectal cancer in most
Western countries, reducing local failure rates to
<10%. The pathological complete response
(pCR), defined by an absence of residual tumor
in the resected specimen after preoperative ther-
apy, is reported between 0% and 31% in recent
phase II/III trials with these regimens. Despite
these advancements, the rate of distant metasta-
ses remains high: around 30–40%. Therefore, the
challenge remains to optimize further rectal can-
cer treatment. With the increased understanding
of molecular pathways that drive colorectal can-
P.-J. Cuyle • E. Van Cutsem (*) cer and the ongoing development of biological
Department of Digestive Oncology,
agents targeting these pathways, recent trials
University Hospital Gasthuisberg,
Leuven 3000, Belgium have investigated the role of targeted biologic
e-mail: eric.vancutsem@uzleuven.be agents in combination with cytotoxic drugs in

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 181


DOI 10.1007/978-3-642-25005-7_19, © Springer-Verlag Berlin Heidelberg 2012
182 P.-J. Cuyle and E. Van Cutsem

preoperative chemoradiation regimens. By doing cancers, and overexpression is regarded as a


so, it is the hope to further improve local control, negative prognostic factor [1, 2]. Moreover, a ret-
resectability, and sphincter preservation rates but rospective analysis has shown inferior results for
more importantly to decrease the risk of distant disease-free survival and pathological complete
recurrence and ultimately to improve overall sur- response (pCR) rates in patients with EGFR-
vival. This manuscript discusses the integration expressing rectal cancer who were treated with
of epidermal growth factor receptor (EGFR) inhi- neoadjuvant radiotherapy [1]. The interaction of
bition and vascular endothelial growth factor ionizing radiation with different levels of the
(VEGF) inhibition in neoadjuvant treatment EGFR pathway has been studied in preclinical
strategies for locally advanced rectal cancer. models, suggesting a synergistic effect for EGFR
inhibition with radiation therapy [3, 4]. Based on
their results, these authors suggested the use of
19.2 EGFR Inhibitors in Rectal EGFR inhibitors as radiosensitizers in clinical
Cancer practice. A landmark phase III study in locally
advanced head and neck squamous cell carci-
The epidermal growth factor receptor (EGFR, noma combining cetuximab with standard high-
HER1, or ErbB1) is a glycoprotein receptor, com- dose radiotherapy showed improved locoregional
prising of an extracellular ligand-binding domain, control and reduced mortality, without increasing
a transmembrane region, and an intracytoplas- the number of adverse events [5]. The clinically
matic domain with tyrosine kinase activity. Ligand significant activity of anti-EGFR targeting agents
binding of the extracellular domain results in in metastatic colorectal cancer has also been
homodimerization or heterodimerization with demonstrated by a number of phase III clinical
other members of the EGFR family (HER2, HER3, trials [6].
HER4) and subsequent initiation of downstream Based on these data, several phase I and II
signaling pathways by autophosphorylation. These trials were undertaken, incorporating the use of
downstream signaling cascades include the mito- EGFR inhibitors in preoperative chemoradi-
gen-activated protein kinase (MAPK) pathway ation for LARC, as summarized in Table 19.1.
and the phosphatidylinositol-3-kinase (PI3K) Pathological complete response (pCR) was the
pathway. EGFR regulates cellular growth, differ- primary endpoint in the majority of reports. The
entiation, and survival, and abnormal EGFR acti- eligibility criteria for inclusion, including initial
vation can result in uncontrolled cell proliferation, tumor stage, varied considerably between stud-
which makes this receptor an attractive target for ies. Also, a wide variation in drug regimen, dose,
cancer treatment. Several EGFR inhibitors are treatment duration, and sequence was noticed
available for clinical use. Cetuximab (Erbitux; between study protocols. The number of reported
ImClone/Bristol-Myers Squibb/Merck Serono) patients was often small and different methods of
and panitumumab (Vectibix; Amgen) are mono- assessing tumor response rates were used, mak-
clonal antibodies directed against the extracellular ing it difficult to compare results or draw firm
domain of the receptor, inhibiting ligand binding. conclusions. Cetuximab was integrated in 13 tri-
Gefitinib (Iressa; AstraZeneca) and erlotinib als [7–19], whereas panitumumab and gefitinib
(Tarceva; Genentech/OSI Pharmaceuticals) are each were used in two trials [20–23]. The reported
small-molecule tyrosine kinase inhibitors (TKI) pCR ranged from 0% to 31%, 11% to 21%, and
targeting the intracellular tyrosine kinase domain 0% to 30%, respectively. A loading dose of cetux-
of the receptor, preventing ATP binding and imab or panitumumab was often administered 1
autophosphorylation. or 2 weeks prior to the start of chemoradiation.
Several preclinical and clinical studies have The rate of (National Cancer Institute CTC) grade
provided a strong rationale for integrating EGFR 3 or 4 (G3/4) adverse events was within the
inhibitors in the preoperative treatment of LARC. expected range, with G3/4 diarrhea being reported
EGFR is overexpressed in 60–80% of colorectal in 5–30% and G3/4 skin rash in 2–15% of patients
19
Table 19.1 Published papers and abstracts reporting the use of EGFR inhibitors in preoperative chemoradiation in rectal cancer
Authors Chemoradiotherapy (CRT) Interval
[Ref.] Phase N Pre-CRT Target 5FU cap oxa iri RT (Gy) Post-CRT (weeks) pCR (%)
Cabebe et al. I 23 cet × × 50.4 cet NR 17
[ 7]
Chung et al. I 20 cet × 50.4 cet 1–3 12
[ 8]
Hofheinz I 20 cet × × 50.4 4–5 26
et al. [9]
Machiels I/II 40 cet cet × 45 6–8 5
et al. [10]
Mai et al. [11] I/II 47 cet × × 50.4 NR NR
Rödel et al. I/II 58 cet cet × × 50.4 6 9
[12]
Bertolini et al. II 40 cet cet × 50.4 6–8 8
[13]
Eisterer et al. II 28 cet × 45 4–6 0
[14]
Kim et al. II 39 cet cet × × 50.4 4–8 23
[15]
Horisberger II 50 cet × × 50.4 4–6 8
et al. [16]
McCollum II 67 cet cet × 50.4 cet 6–8 31
et al. [17]
Velenik et al. II 37 cap/cet cet × 45 4–6 8
[18]
Dewdney II 46 cap/oxa/cet cet × 54 6 11
et al. [19]
(continued)
Should Biologic Targeted Agents Be Combined with Preoperative Chemoradiation in Rectal Cancer?
183
Table 19.1 (continued)
184

Authors Chemoradiotherapy (CRT) Interval


[Ref.] Phase N Pre-CRT Target 5FU cap oxa iri RT (Gy) Post-CRT (weeks) pCR (%)
Czito et al. I 6 gef × 50.4 6–8 0
[20]
Valentini et al. I/II 41 gef × 50.4 7–8 30
[21]
Pinto et al. II 60 pan pan × × 50.4 7–8 21
[22]
Helbling et al. II 40 pan pan × 45 pan 3–5 11
[23]
N number of patients entering study, pre-CRT agents given before start of radiotherapy, target targeted agent used, 5FU 5-fluorouracil, cap capecitabine; oxa, oxaliplatin, iri
irinotecan, RT(Gy) radiotherapy dose, post-CRT agents given after completion of radiotherapy, interval therapy-free interval between the end of preoperative therapy and surgery
pCR pathological complete response, cet cetuximab, gef gefitinib, pan panitumumab, NR not reported
P.-J. Cuyle and E. Van Cutsem
19 Should Biologic Targeted Agents Be Combined with Preoperative Chemoradiation in Rectal Cancer? 185

receiving a cetuximab-containing regimen. Both radiotherapy with concomitant EGFR inhibitors,


trials integrating panitumumab reported a high may be strategies worth exploring [3, 4].
incidence of G3/4 diarrhea, reaching up to 39% On top of modifications in the drug schedule,
of patients when combined with oxaliplatin, 5FU, adequate patient selection appears to be crucial to
and radiation [22]. The association of gefitinib to improve the treatment outcome in rectal cancer.
preoperative chemoradiation had a comparable Currently, no predictive or prognostic biomarkers
toxicity profile; however, G3 hepatic toxicity was that impact the individual treatment strategy
encountered in 26% of treated patients [21]. or outcome in preoperative treatment for rectal
In summary, results of these phase I/II studies cancer are universally accepted. EGFR protein
confirm that combination of EGFR inhibitors expression evaluated by immunohistochemistry
with neoadjuvant chemoradiation for rectal can- (IHC) is not predictive for response to cetuximab
cer is feasible with acceptable toxicity. However, in metastatic colorectal cancer, nor in locally
the rates of pCR achieved with this combination advanced rectal cancer [26]. The EGFR gene
are disappointingly low. copy number (GCN) determined by FISH
A number of possible explanations have been has been proposed as a more reliable marker.
proposed why these phase I/II trials are unable to However, in metastatic colorectal cancer, only a
reproduce the benefit seen with EGFR-directed fraction of patients with an increased EGFR GCN
therapy in concomitant radiotherapy for head and responds [3]. In LARC, a significant relationship
neck cancer or in metastatic colorectal cancer. between EGFR high GCN and high pathological
Accelerated repopulation of tumor cells surviv- TRG was observed in patients treated with cetux-
ing irradiation is an adaptive mechanism produc- imab and 5FU-based chemoradiation [26]. The
ing radioresistance and can be suppressed by best probability to predict a high TRG in this
EGFR inhibitors [2, 4]. This phenomenon, how- treatment group was achieved when the combina-
ever, seems less important in the treatment of rec- tion of a high GCN and KRAS wild type status
tal cancer, compared to head and neck cancer, was present [26]. The same authors found no
since radiation doses tend to be lower, concomi- such relation for EGFR GCN or KRAS mutation
tant 5FU or capecitabine is usually administered, status in patients who were treated with standard
and surgical resection generally follows treat- chemoradiation without cetuximab [27]. KRAS
ment [24]. Nyati et al. have clearly illustrated mutation is found in 30–40% of patients with
possible interactions between EGFR inhibitors LARC, but no correlation between mutation sta-
and cytotoxic drugs, including 5FU, oxaliplatin, tus and response to cetuximab-based chemoradi-
and irinotecan [4]. The G1 cell cycle arrest, ation could be established in two separate studies
caused by EGFR inhibitors through upregulation [26, 28]. However, these results could be due to
of cyclin-dependent kinase inhibitors p27 and the small number of patients treated in these tri-
p21, may in fact antagonize the cytotoxic activity als. A recent retrospective pooled analysis by
of other cell-cycle-dependent chemotherapeutic Grimminger et al. suggested that pretreatment
agents. A well-considered sequence in the admin- intratumoral EGFR and VEGF mRNA expres-
istration of different therapeutic agents and radio- sion levels, as well as KRAS mutation status, are
therapy may be the key to optimize synergy and predictive markers of pathological response in
produce better outcome results. Morelli and col- these patients [29]. Mutations in downstream
leagues have studied the antiproliferative activity effectors of the EGFR pathway, such as BRAF
of cytotoxic agents and EGFR inhibitors in vitro and PIK3CA, are rare in patients with KRAS
and have shown that a synergistic activity was wild-type rectal cancer, and their predictive value
only obtained when chemotherapy administra- remains unclear [30]. Erben et al. found no cor-
tion preceded EGFR blocking treatment [25]. relation between mRNA expression of platelet-
The administration of EGFR antagonists as a derived growth factor receptor-b (PDGFR-b) or
maintenance after completion of chemoradia- c-kit and tumor regression rate after cetuximab-
tion or induction chemotherapy, followed by based chemoradiotherapy [31].
186 P.-J. Cuyle and E. Van Cutsem

19.3 VEGF Inhibitors in Rectal Again, substantial differences were noted between
Cancer studies concerning the inclusion criteria and the
applied drug regimen, dose, treatment duration,
Neoangiogenesis is crucial for tumor growth and sequence. The rate of a pathological com-
and malignant progression. In the majority of plete response, although difficult to compare,
cancers, tumor vessels appear to be abnormal tends to be somewhat higher with bevacizumab-
in structure and function, leading to a hostile based regimens than seen with EGFR inhibitor
microenvironment characterized by hypoxia, low containing regimens, ranging from 13% to 36%
pH, and high interstitial fluid pressure [32]. On of patients.
the one hand, the escape of tumor cells through The reported incidence of grade 3–4 diarrhea
leaky vessels is facilitated, and on the other hand, ranged between 20% and 40% in most trials.
transport and distribution of cytotoxics and oxy- Other more typical bevacizumab-related adverse
gen seem to be impaired. Blockade of VEGF sig- events, such as hypertension, proteinuria, mucosal
naling by pharmacological agents can transiently bleeding, and arterial thrombosis, were reported
repair these vascular abnormalities, thus improv- at rates similar to those seen in phase III trials of
ing oxygenation and lowering interstitial fluid bevacizumab in metastatic colorectal cancer.
pressure. This process is referred to as “vascular Postoperative complication rates, including pel-
normalization” [32]. Improving oxygenation can vic infection, delayed wound healing, anasto-
sensitize tumors to the cytotoxic effects of irra- motic leak, and fistulae, were relatively high in a
diation, while a decrease in interstitial fluid pres- number of trials, ranging between 36% and 63%
sure improves cytotoxic drug delivery. Extensive of cases [37, 39, 43–45]. On the other hand, a
preclinical studies in human tumor xenograft recent report of Liang et al. investigated the tech-
models have demonstrated tumor suppression nical feasibility of laparoscopic TME for low rec-
ability of VEGF blockade [2] as well as its radio- tal cancer after preoperative treatment with the
sensitizing ability [33]. Willett and colleagues combination bevacizumab-oxaliplatin-5FU and
have assessed the effect of a single administration fractionated radiotherapy [46]. Although the
of bevacizumab (Avastin; Roche/Genentech), a interval to surgery after the last dose of bevaci-
recombinant humanized monoclonal antibody to zumab was only 2–4 weeks, a R0 resection was
VEGF, on vascular biology in human rectal can- achieved in all 28 treated patients at a postopera-
cer [34]. This study indicates that, even at low tive complication rate of “only” 21.4%. Long-term
doses, bevacizumab alone can decrease perfu- results after bevacizumab-containing chemora-
sion, microvascular density, and interstitial fluid diation were only available for the study by
pressure in a solid tumor. Addition of bevaci- Willett et al. providing a 5-year actuarial overall
zumab to conventional chemotherapy in meta- survival rate of 95.0%, a disease-free survival of
static colorectal cancer has resulted in significant 68.9%, and a local control rate of 91.7% [47].
and clinically meaningful survival benefit in a In summary, results of these phase I/II trials
number of phase III trials [1]. indicate that integration of VEGF inhibition, in
The potential benefit of VEGF blockade was this case bevacizumab, into preoperative chemo-
further explored in a number of phase I and II radiation regimens is feasible at a manageable
studies, summarized in Table 19.2. The reported toxicity rate but does not appear to dramatically
trials have integrated bevacizumab into preopera- enhance pathological downstaging rates when
tive 5FU- or capecitabine-based chemoradiation compared to more conventional preoperative reg-
regimens for locally advanced rectal cancer [35– imens. There are however no data available from
46]. Five of them also used oxaliplatin as a sec- larger studies whether the addition of bevaci-
ond cytotoxic agent during treatment [35, 39, zumab decreases the rate of metastases in rectal
44–46]. Surgical intervention was usually delayed cancer, and some concerns may of course be
until at least 4–6 weeks after the last dose of bev- raised in view of the lack of benefit of bevaci-
acizumab to prevent postoperative complications. zumab in improving the outcome of stage III
19

Table 19.2 Published papers and abstracts reporting use of VEGF inhibitors in preoperative chemoradiation in rectal cancer
Authors Chemoradiotherapy (CRT) Interval
[Ref.] Phase N Pre-CRT Target FU cap oxa iri RT (Gy) Post-CRT (weeks) pCR (%)
Czito et al. I 11 bev × × 50.4 6–8 18
[35]
Blaszkowski I/II 21 bev/erl × 50.4 6–9 47
et al. [36]
Willett et al. I/II 32 bev bev × 50.4 7–10 16
[37]
Crane et al. II 25 bev × 50.4 6–10 32
[38]
Kennecke II 42 bev bev × × 50.4 7–9 18
et al. [39]
Nogué et al. II 47 cap/oxa/bev bev × 50.4 6–8 36
[40]
Resch et al. II 8 bev × 45 6–8 25
[41]
Spigel et al. II 35 bev × 50.4 2–8 29
[42]
Velenik et al. II 61 bev bev × 50.4 6–8 13
[43]
Dipetrillo II 26 FU/oxa/bev bev × × 50.4 4–8 20
et al. [44]
Hoehler et al. II 70 bev × × 50.4 NR 17
[45]
Liang et al. II 28 bev × × 45 FU/oxa/bev 2–4 25
[46]
N number of patients entering study, pre-CRT agents given before start of radiotherapy, target targeted agent used, FU 5-fluorouracil, cap capecitabine, oxa oxaliplatin, iri irino-
tecan, RT(Gy) radiotherapy dose, post-CRT agents given after completion of radiotherapy, interval therapy-free interval between the end of preoperative therapy and surgery,
pCR pathological complete response, bev bevacizumab, erl erlotinib, NR not reported
Should Biologic Targeted Agents Be Combined with Preoperative Chemoradiation in Rectal Cancer?
187
188 P.-J. Cuyle and E. Van Cutsem

colon cancer [48]. Moreover, the high incidence combination in treatment of metastatic colorectal
of postoperative complications has raised some cancer demonstrated an inferior median progres-
concerns about the safety of this approach. sion-free survival at the cost of an unfavorable
So far, no molecular biomarkers reliably pre- toxicity profile [2]. A small phase I/II study by
dicting response to VEGF inhibition therapy are Blaszkowski et al. investigated the combination of
available in clinical practice. Pretreatment VEGF bevacizumab and erlotinib in combination with
concentrations in tumoral tissue and circulation infusional 5FU and pelvic radiation in patients
failed to correlate with outcome to therapy with with LARC [36]. This combination appeared to
VEGF blocking agents [49], although some be well tolerated, and a pCR of 47% was achieved
recent studies suggested a potential predictive in 15 patients undergoing surgery so far.
role of serum VEGF-A, as determined with
newer assays, in some cancers including pancre- Conclusions
atic and gastric cancer [50, 51]. Willett and col- Improving survival in locally advanced rectal
leagues have put considerable effort in identifying cancer, while minimizing treatment-related
the kinetic changes in tumor cell proliferation comorbidity, remains challenging. Integration
and apoptosis indices in response to anti-VEGF of targeted therapy in preoperative chemora-
treatment. They found that bevacizumab admin- diation regimens is supported by a strong the-
istration increases levels of plasma VEGF and oretical background, promising preclinical
placental-derived growth factor (PlGF) and data, and encouraging experiences with these
decreases levels of circulating endothelial cells agents in metastatic colorectal cancer and
(CECs) and progenitor cells (CPCs) [37, 52]. locally advanced head and neck cancer. Phase
This group also looked at soluble VEGF recep- I results have demonstrated that this approach
tor 1 (sVEGFR-1), which is an antiangiogenic is feasible with an acceptable toxicity.
factor released by endothelial cells, playing an However, results from phase II studies, incor-
important role in the process of vascular normal- porating EGFR and VEGF targeting therapy,
ization [49]. They noticed a striking association have been unimpressive, showing no clear
of pretreatment plasma sVEGFR-1 concentra- response benefit by adding these agents to
tions and both primary tumor response and tox- standard therapy. No randomized phase III
icity after bevacizumab-based chemoradiation. studies are currently available. Consequently,
However, further evaluation of these markers is the use of these agents in the preoperative
needed in larger studies to confirm their predic- treatment for rectal cancer remains experi-
tive and/or prognostic value. We are coordinat- mental. The use of the pathological complete
ing at the moment in our center a multicenter response as an endpoint remains controversial
randomized phase 2 study evaluating the combi- since it is largely influenced by the initial size
nations of bevacizumab/capecitabine or bevaci- of the tumor, the interval between radiother-
zumab/capecitabine/oxaliplatin plus radiotherapy apy and surgery, and the quality of histopatho-
in locally advanced rectal cancer, with a focus on logical examination. The correlation between
marker analysis and translational research (AXE- pathological complete response and long-term
BEAM study). outcome has been variable in previous trials.
A better understanding of the mechanisms
responsible for the disappointing results in
19.4 Combined EGFR and VEGF these phase II trials is necessary before pro-
Inhibition in Rectal Cancer ceeding to phase III. Future strategies should
focus on accurate identification of biomarker-
Despite evidence from preclinical data for a syn- defined subpopulations with high-risk rectal
ergistic effect of EGFR and VEGF blockade, tumors that will benefit most from a certain
results from phase III trials implementing this treatment approach.
19 Should Biologic Targeted Agents Be Combined with Preoperative Chemoradiation in Rectal Cancer? 189

References Int J Radiat Oncol Biol Phys 73(2):466–472, Epub


2008 Nov 10
14. Eisterer WM, De Vries A, Oefner D et al (2009)
1. Czito BG, Willett CG (2011) Beyond 5-fluorouracil:
Neoadjuvant chemoradiation therapy with capecit-
the emerging role of newer chemotherapeutics and
abine (X) plus cetuximab (C), and external beam
targeted agents with radiation therapy. Semin Radiat
radiotherapy (RT) in locally advanced rectal cancer
Oncol 21(3):203–211
(LARC): ABCSG trial R03. J Clin Oncol 27:15s,
2. Wadlow RC, Ryan DP (2010) The role of targeted
(suppl; abstract 4109)
agents in preoperative chemoradiation for rectal can-
15. Kim SY, Hong YS, Kim DY et al (2011) Preoperative
cer. Cancer 116(15):3537–3548
chemoradiation with cetuximab, irinotecan, and capecit-
3. Debucquoy A, Machiels JP, McBride WH et al (2010)
abine in patients with locally advanced resectable rectal
Integration of epidermal growth factor receptor inhib-
cancer: a multicenter phase II study. Int J Radiat Oncol
itors with preoperative chemoradiation. Clin Cancer
Biol Phys 81(3):677–683, Epub 2010 Oct 1
Res 16(10):2709–2714, Epub 2010 Apr 28
16. Horisberger K, Treschl A, Mai S et al (2009)
4. Nyati MK, Morgan MA, Feng FY et al (2006) Integration
Cetuximab in combination with capecitabine, irinote-
of EGFR inhibitors with radiochemotherapy. Nat Rev
can, and radiotherapy for patients with locally
Cancer 6(11):876–885, Epub 2006 Oct 12
advanced rectal cancer: results of a phase II MARGIT
5. Bonner JA, Harari PM, Giralt J et al (2006)
trial. Int J Radiat Oncol Biol Phys 74(5):1487–1493,
Radiotherapy plus cetuximab for squamous-cell car-
Epub 2009 Jan 7
cinoma of the head and neck. N Engl J Med 354(6):
17. McCollum AD, Kocs DM, Chada P et al (2010) A
567–578
randomized phase II trial of preoperative chemoradio-
6. Prenen H, Tejpar S, Van Cutsem E (2010) New strate-
therapy with or without cetuximab in locally advanced
gies for treatment of KRAS mutant metastatic col-
adenocarcinoma of the rectum. J Clin Oncol 28:15s,
orectal cancer. Clin Cancer Res 16(11):2921–2926,
(suppl; abstract 3635)
Epub 2010 May 11
18. Velenik V, Ocvirk J, Oblak I et al (2010) A phase II
7. Cabebe EC, Kuo T, Koong A et al (2008) Phase I trial of
study of cetuximab, capecitabine and radiotherapy in
preoperative cetuximab in combination with oxaliplatin,
neoadjuvant treatment of patients with locally
capecitabine, and radiation therapy for locally advanced
advanced resectable rectal cancer. Eur J Surg Oncol
rectal cancer. J Clin Oncol (Meeting Abstracts)
36(3):244–250, Epub 2009 Dec 29
26(15 suppl; abstract 15019)
19. Dewdney A, Capdevila J, Glimelius B et al (2011)
8. Chung K, Minsky B, Schrag D et al (2006) Phase I
EXPERT-C: a randomized, phase II European multi-
trial of preoperative cetuximab with concurrent con-
center trial of neoadjuvant capecitabine plus oxalipla-
tinuous infusion 5-fluorouracil and pelvic radiation in
tin chemotherapy (CAPOX) and chemoradiation
patients with local-regionally advanced rectal cancer.
(CRT) with or without cetuximab followed by total
J Clin Oncol 24(18 suppl):256; (abstract #3560)
mesorectal excision (TME) in patients with MRI-
9. Hofheinz R, Horisberge K, Woernle C et al (2006)
defined, high-risk rectal cancer. J Clin Oncol 29
Phase I trial of cetuximab in combination with capecit-
(suppl; abstract 3513)
abine, weekly irinotecan and radio therapy as neoad-
20. Czito BG, Willett CG, Bendell JC et al (2006)
juvant therapy for rectal cancer. Int J Radiat Oncol
Increased toxicity with gefitinib, capecitabine, and
Biol Phys 66:1384–1390
radiation therapy in pancreatic and rectal cancer:
10. Machiels J, Sempoux C, Scalliet P et al (2007) Phase
phase I trial results. J Clin Oncol 24(4):656–662
I/II study of preoperative cetuximab, capecitabine,
21. Valentini V, De Paoli A, Gambacorta MA et al (2008)
and external beam radiotherapy in patients with rectal
Infusional 5-fluorouracil and ZD1839 (gefitinib-
cancer. Ann Oncol 18:738–744
iressa) in combination with preoperative radiotherapy
11. Mai SK, Hoffheinz R, Treschl A et al (2008)
in patients with locally advanced rectal cancer: a
Correlation of minimal tumor dose and histopatho-
phase I and II trial (1839IL/0092). Int J Radiat Oncol
logical regression of rectal cancer after neoadjuvant
Biol Phys 72(3):644–649, Epub 2008 Apr 18
combined radio-chemo-immunotherapy – results of a
22. Pinto C, Di Fabio F, Maiello E et al (2011) Phase II
prospective phase I/II study (cetuximab capiri-RT).
study of panitumumab, oxaliplatin, 5-fluorouracil,
Int J Radiat Oncol Biol Phys 72:S263
and concurrent radiotherapy as preoperative treatment
12. Rödel C, Arnold D, Hipp M et al (2008) Phase I-II
in high-risk locally advanced rectal cancer patients
trial of cetuximab, capecitabine, oxaliplatin, and
(StarPan/STAR-02 study). Ann Oncol 22(11):2424–
radiotherapy as preoperative treatment in rectal can-
2430, Epub 2011 Mar 8
cer. Int J Radiat Oncol Biol Phys 70(4):1081–1086,
23. Helbling D, Bodoky G, Gautschi O et al (2011)
Epub 2007 Sep 19
Neoadjuvant chemoradiation (CRT) with or without
13. Bertolini F, Chiara S, Bengala C et al (2009)
panitumumab (pan) in patients with K-ras-unmutated,
Neoadjuvant treatment with single-agent cetuximab
locally advanced rectal cancer (LARC): a randomized
followed by 5-FU, cetuximab, and pelvic radiother-
multicenter phase II trial (SAKK 41/07). J Clin Oncol
apy: a phase II study in locally advanced rectal cancer.
29 (suppl; abstract 3546)
190 P.-J. Cuyle and E. Van Cutsem

24. Glynne-Jones R, Mawdsley S, Harrison M (2010) beam radiation therapy (RT) in locally advanced rec-
Cetuximab and chemoradiation for rectal cancer–is tal cancer (LARC). J Clin Oncol 27:15s, (suppl;
the water getting muddy? Acta Oncol 49(3):278–286 abstract 4106)
25. Morelli MP, Cascone T, Troiani T et al (2005) 37. Willett CG, Duda DG, di Tomaso E et al (2009)
Sequence-dependent antiproliferative effects of cyto- Efficacy, safety, and biomarkers of neoadjuvant beva-
toxic drugs and epidermal growth factor receptor cizumab, radiation therapy, and fluorouracil in rectal
inhibitors. Ann Oncol 16(Suppl 4):vi61–vi68 cancer: a multidisciplinary phase II study. J Clin
26. Bengala C, Bettelli S, Bertolini F et al (2009) Oncol 27(18):3020–3026, Epub 2009 May 26
Epidermal growth factor receptor gene copy number, 38. Crane CH, Eng C, Feig BW et al (2010) Phase II trial
K-ras mutation and pathological response to preoper- of neoadjuvant bevacizumab, capecitabine, and radio-
ative cetuximab, 5-FU and radiation therapy in locally therapy for locally advanced rectal cancer. Int J Radiat
advanced rectal cancer. Ann Oncol 20(3):469–474, Oncol Biol Phys 76(3):824–830, Epub 2009 May 21
Epub 2008 Dec 18 39. Kennecke H, Berry S, Wong R et al (2012) Pre-
27. Bengala C, Bettelli S, Bertolini F et al (2010) operative bevacizumab, capecitabine, oxaliplatin and
Prognostic role of EGFR gene copy number and KRAS radiation among patients with locally advanced or low
mutation in patients with locally advanced rectal can- rectal cancer: a phase II trial. Eur J Cancer 48:37–45
cer treated with preoperative chemoradiotherapy. Br J 40. Nogué M, Salud A, Vicente P et al (2011) Addition of
Cancer 103(7):1019–1024, Epub 2010 Sep 14 bevacizumab to XELOX induction therapy plus con-
28. Debucquoy A, Haustermans K, Daemen A et al (2009) comitant capecitabine-based chemoradiotherapy in
Molecular response to cetuximab and efficacy of pre- magnetic resonance imaging-defined poor-prognosis
operative cetuximab-based chemoradiation in rectal locally advanced rectal cancer: the AVACROSS study.
cancer. J Clin Oncol 27(17):2751–2757, Epub 2009 Oncologist 16(5):614–620, Epub 2011 Apr 5
Mar 30 41. Resch G, De Vries A, Ofner D et al (2012) Preoperative
29. Grimminger PP, Danenberg P, Dellas K et al (2011) treatment with capecitabine, bevacizumab and radio-
Biomarkers for cetuximab-based neoadjuvant radio- therapy for primary locally advanced rectal cancer – a
chemotherapy in locally advanced rectal cancer. Clin two stage phase II clinical trial. Radiother Oncol
Cancer Res 17(10):3469–3477, Epub 2011 May 10 102:10, Epub 2011 Jul 7
30. Koeberle D, Bougel S, Benhattar J et al (2011) 42. Spigel DR, Bendell JC, McCleod M et al (2012) Phase
Prospective analysis of KRAS, BRAF, and PIK3CA II study of bevacizumab and chemoradiation in the
mutational status and EGFR copy number in patients preoperative or adjuvant treatment of patients with
(pts) with locally advanced rectal cancer: A transla- stage II/III rectal cancer. Clin Colorectal Cancer
tional substudy of a clinical trial (SAKK 41/07) evalu- 11:45–52, Epub 2011 Aug 15
ating the effect of neoadjuvant chemoradiation (CRT) 43. Velenik V, Ocvirk J, Music M et al (2011) Neoadjuvant
with or without panitumumab. J Clin Oncol 29 (suppl; capecitabine, radiotherapy, and bevacizumab (CRAB)
abstract 3549) in locally advanced rectal cancer: results of an open-
31. Erben P, Horisberger K, Muessle B et al (2008) mRNA label phase II study. Radiat Oncol 6:105
expression of platelet-derived growth factor receptor- 44. Dipetrillo T, Pricolo V, Lagares-Garcia J et al (2012)
beta and C-KIT: correlation with pathologic response Neoadjuvant bevacizumab, oxaliplatin, 5-fluorouracil,
to cetuximab-based chemoradiotherapy in patients and radiation for rectal cancer. Int J Radiat Oncol Biol
with rectal cancer. Int J Radiat Oncol Biol Phys Phys 82:124–129, Epub 2010 Oct 13
72(5):1544–1550 45. Hoehler T, Dellas K, Riesenbeck D et al (2011)
32. Carmeliet P, Jain RK (2011) Principles and mechanisms Preoperative chemoradiation (CRT) with concurrent
of vessel normalization for cancer and other angiogenic capecitabine (Cap), oxaliplatin (Ox), and bevaci-
diseases. Nat Rev Drug Discov 10(6):417–427 zumab (Bev) in patients with locally advanced rectal
33. Willett CG, Kozin SV, Duda DG et al (2006) Combined cancer (RC): effects on pathological complete
vascular endothelial growth factor-targeted therapy response (pCR) and surgical complications. J Clin
and radiotherapy for rectal cancer: theory and clinical Oncol 29(suppl; abstract 3630)
practice. Semin Oncol 33(5 Suppl 10):S35–S40 46. Liang JT, Lai HS, Cheng KW (2011) Technical feasi-
34. Willett CG, Boucher Y, di Tomaso E et al (2004) bility of laparoscopic total mesorectal excision for
Direct evidence that the VEGF-specific antibody bev- patients with low rectal cancer after concurrent radia-
acizumab has antivascular effects in human rectal tion and chemotherapy with bevacizumab plus
cancer. Nat Med 10(2):145–147, Epub 2004 Jan 25 FOLFOX. Surg Endosc 25(1):305–308, Epub 2010
35. Czito BG, Bendell JC, Willett CG et al (2007) Jun 5
Bevacizumab, oxaliplatin, and capecitabine with radi- 47. Willett CG, Duda DG, Ancukiewicz M et al (2010) A
ation therapy in rectal cancer: phase I trial results. Int safety and survival analysis of neoadjuvant bevaci-
J Radiat Oncol Biol Phys 68(2):472–478 zumab with standard chemoradiation in a phase I/II
36. Blaszkowsky LS, Hong TS, Zhu AX et al (2009) A study compared with standard chemoradiation in
phase I/II study of bevacizumab (beva), erlotinib (erl), locally advanced rectal cancer. Oncologist 15(8):845–
and 5-fluorouracil (5-FU) with concurrent external 851, Epub 2010 Jul 28
19 Should Biologic Targeted Agents Be Combined with Preoperative Chemoradiation in Rectal Cancer? 191

48. Van Cutsem E, Lambrechts D, Prenen H, Jain RK, (GE) in patients (pts) with metastatic pancreatic cancer
Carmeliet P (2011) Lessons from the adjuvant bevaci- (mPC). Eur J Cancer 47:S95–S96 (803)
zumab trial on colon cancer: what next? J Clin Oncol 51. Jayson G, De Haas S, Delmar P et al (2011) Evaluation
29(1):1–4, Epub 2010 Nov 29 of plasma VEGFA as a potential predictive pan-
49. Duda DG, Willett CG, Ancukiewicz M et al (2010) tumour biomarker for bevacizumab. Eur J Cancer
Plasma soluble VEGFR-1 is a potential dual bio- 47:S96 (804)
marker of response and toxicity for bevacizumab with 52. Willett CG, Boucher Y, Duda DG et al (2005)
chemoradiation in locally advanced rectal cancer. Surrogate markers for antiangiogenic therapy and
Oncologist 15(6):577–583, Epub 2010 May 18 dose-limiting toxicities for bevacizumab with radia-
50. Van Cutsem E, Jayson G, Dive C et al (2011) Analysis of tion and chemotherapy: continued experience of a
blood plasma factors in the AVITA phase III randomized phase I trial in rectal cancer patients. J Clin Oncol
study of bevacizumab (bev) with gemcitabine-Erlotinib 23(31):8136–8139
Should Upfront Chemotherapy
Precede Preoperative 20
Chemoradiation and Surgery?

Carlos Fernandez-Martos and Xabier Garcia


de Albeniz

Contents 20.1 Introduction


20.1 Introduction ................................................ 193
In North America and in many European countries,
20.2 Adjuvant Chemotherapy
in Colon Versus Rectal Cancer ................. 194
preoperative 5FU and radiation followed by total
mesorectal excision (TME) and postoperative
20.3 Induction Chemotherapy Followed
adjuvant 5FU is a standard treatment for locally
by Concomitant Chemoradiotherapy
and Surgery ................................................ 196 advanced rectal (stage II and III) cancer, resulting
in a 5-year cumulative rate of local relapse rate of
20.4 Appendix: Evidence to Proceed to
a Phase III Trial ......................................... 200 less than 10% and an incidence of distant metas-
20.4.1 Question to Answer ..................................... 200 tases of about 35% [5, 40].
20.4.2 Study Design ................................................ 200 The phase III studies published in the last
20.4.3 Biomarkers ................................................... 200 decade include patients with stage I, II and III
20.4.4 Randomization ............................................. 201
20.4.5 Sample Size.................................................. 201 [29] and clinically staged II and III [5, 20, 40].
The most important conclusion is that preopera-
References ................................................................. 201
tive treatment either with long-term radiotherapy
(RT) and chemotherapy (CT) or short-course RT
has a major impact on local control. However,
there has been no impact on DFS or OS. Since
survival is mainly determined by distant metasta-
ses, efforts should be directed towards preventing
systemic disease.
Comparing DFS in recent landmark trials in
adjuvant therapy in stage II and III colon cancer
versus stage II and III rectal cancer, the results
indicate a less favourable outcome for the latter
C. Fernandez-Martos (*)
Medical Oncology Department, group (Fig. 20.1).
Fundacion Instituto Valenciano de Oncologia, Although currently we do not have a definitive
Calle Gregorio Gea 31, Valencia 46009, Spain answer for this fact, the differences may be
e-mail: cfmartos@fivo.org
explained, in part, by the highest incidence of
X. Garcia de Albeniz local relapse and the limited and inconstant
Channing Laboratory, Cubicle 346F,
efficacy of adjuvant postoperative chemotherapy
Brigham and Women’s Hospital,
181 Longwood Avenue, Boston, MA 02115, USA in patients undergoing preoperative radio(chemo)
e-mail: xgarciad@hsph.harvard.edu therapy [8]. Furthermore, rectal and colon cancers

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 193


DOI 10.1007/978-3-642-25005-7_20, © Springer-Verlag Berlin Heidelberg 2012
194 C. Fernandez-Martos and X.G. de Albeniz

90
84.7% 83.7%
81.9%
79.9%
80 5FU/LV FOLFOX
5FU/LV
Control

70 68%
66.4% 65%
Postop
58.9% FOLFOX Preop FU/RT 59.4%
60 FU/RT FU 58.2%
Postop 55.5% Preop
5F/LV FU/RT Preop
FU 52.2% FU/RT
Preop Postop
RT Or RT
50 Postop
FU
Preop NO
FU FU/RT PostopFU
Or RT
NO
40 PostopFU

30

20

10

0
QUASAR MOSAIC MOSAIC CAO/ARO/AIO FFCD 9203 EORTC 22921
Stage II Stage II Stage III 94 Stage II/III Stage II/III Stage II/III

Fig. 20.1 5-year disease-free survival for landmark clini- EORTC 22921 [5] (Adapted from Ther Adv Med Oncol
cal trials in colon. Quasar [24]; MOSAIC [1] and rectal [3]. © Sage Publications, INC. London:SAGE)
cancer trials; CAO/ARO/AIO [40]; FFCD 9203 [20];

may be genetically different. Different levels of was statistically significantly increased in the
microsatellite instability and mutations in BRAF subset of patients with positive regional lymph
and NRAS have been described [19, 28, 39] and nodes but not in patients with N0 disease when
can be of importance for the efficacy in the adju- compared to patients not receiving chemotherapy
vant setting in these two different anatomic sites. [21]. Although results of a subset analysis of data
from the MOSAIC trial did not show a significant
DFS benefit of FOLFOX over 5FU/LV for patients
20.2 Adjuvant Chemotherapy with stage II disease at a follow-up of 6 years, a
in Colon Versus Rectal Cancer trend was observed for improved DFS in high-risk
stage II patients (i.e. disease characterized by at
In colon cancer, adjuvant chemotherapy is rec- least one of the following: T4 tumour, tumour per-
ommended in stage III because, as demonstrated foration, bowel obstruction, poorly differentiated
in the MOSAIC trial, DFS at 5 years was tumour, venous invasion, £ lymph nodes exam-
significantly better in the FOLFOX arm (66.4%) ined) receiving FOLFOX compared to infusional
when compared with those receiving 5FU/leu- 5FU/LV, suggesting that this patient population
covorin (58.9%) P = 0.005. Overall survival at may benefit from treatment with FOLFOX. No
6-year follow-up was also significantly increased benefit, however, of FOLFOX over 5FU/LV was
(72.9% vs. 68.7%; HR = 0.80; P = 0.023) [1]. seen for patients with low-risk stage II disease in
The impact of adjuvant chemotherapy for the MOSAIC trial.
patients with stage II disease is much less clear. Although, to date, there has been no definitive
An analysis of pooled data from seven random- randomized study to identify the optimal post-
ized trials indicated that overall survival of operative adjuvant treatment and the optimal
patients with resected early stage colon cancer duration of therapy in rectal cancer [43], the
treated with 5FU-based adjuvant chemotherapy general recommendation is to give adjuvant
20 Should Upfront Chemotherapy Precede Preoperative Chemoradiation and Surgery? 195

chemotherapy to patients with stage II and III By contrast, the adherence to the planned treatment
[22, 35]. is higher in colon cancer trials. In the MOSAIC
European Organization for Research and trial, 98.6% of patients received at least one cycle
Treatment of Cancer (EORTC) 22921 was a ran- of fluorouracil/leucovorin and oxaliplatin and
domized trial to evaluate the role of concurrent 98.9% received at least one cycle of fluorouracil/
chemoradiation in the neoadjuvant setting, along leucovorin and 74.7% of patients in the group given
with the role of adjuvant chemotherapy in patients fluorouracil plus oxaliplatin and 86.5% in the FL
with resectable stage T3/T4 rectal cancer [5]. group received the planned 12 cycles [2]. This fact
Patients (n = 1,011) were randomized to one of could explain, at least in part, the lack of a clear
four treatment arms: benefit to DFS or OS in the EORTC trial.
1. Preoperative radiation (45 Gy over 5 weeks) Second, this trial included clinically staged
alone T3/T4 patients [6], 35% of whom were recruited
2. Preoperative radiation plus two 5-day courses after a clinical examination that included some
of chemotherapy (5FU at 350 mg/m2/day and subjective criteria (i.e. clinical T3 had to have
leucovorin at 20 mg/m2/day in week 1 and 5 of one of the following at digital rectal examination:
radiation therapy) a circular extension, a lateral or posterior fixity
3. Preoperative radiotherapy plus four postoper- and an anterior adhesion to the prostate or inva-
ative courses of chemotherapy sion of the rectovaginal wall), and 60% were
4. Preoperative radiotherapy and chemotherapy ultrasound-staged T3, and as the German study
plus postoperative chemotherapy demonstrated, about 20% could be stage I [40].
The adjuvant chemotherapy used in this study Therefore, in this study, the methods used for
was 5FU (350 mg/m2/day) and leucovorin (20 mg/ staging could have determined that the true popu-
m2/day) given on days 1–5, and this regimen was lation would be made up of stages I, II and III.
given every 3 weeks for a total of four courses. Third, the survival curves for patients who
At the time of the initial analysis, which was received postoperative adjuvant chemotherapy
reported after a median follow-up of 5.4 years, begin to diverge after 2 years for DFS and after
adjuvant chemotherapy did not appear to confer 4 years for OS when compared to those who did
benefit in terms of DFS or OS. There are, how- not receive adjuvant chemotherapy, suggesting
ever, three important points to note. The first is that further follow-up is required to determine
that 26.9% of patients randomized to receive whether these differences will continue.
adjuvant chemotherapy actually never started the An updated analysis was subsequently per-
treatment and only 43% received 95% of the formed to determine the long-term results of the
planned dose of chemotherapy. Data from other EORTC 22921 study [14]. The goal of this
modern phase II and phase III [18, 37, 40, 41] exploratory multivariate analysis was to investi-
trials are similar, showing that applying chemo- gate whether it was possible to identify a sub-
therapy (5FU or capecitabine/oxaliplatin) after group of patients who would benefit most from
preoperative chemoradiation and surgery is prob- adjuvant chemotherapy in the long term. Adjuvant
lematic and often not feasible with adequate dos- chemotherapy did not improve DFS or OS for all
ing. More recently, a National Comprehensive patients with resectable T3/T4 rectal cancer.
Cancer Network (NCCN) analysis carried out on A subset analysis, however, revealed that patients
810 stage II and III patients from eight specialty whose disease responded to neoadjuvant therapy,
cancer centres shows that 20% of patients treated specifically those patients whose tumours were
with neoadjuvant chemoradiation did not receive downstaged to ypT0–T2 compared with stages
any adjuvant CTX [30]. The most frequent ypT3–T4 with preoperative therapy, experienced
causes are the patient’s decision to abandon a survival benefit from adjuvant chemotherapy.
treatment because of the fatigue following the Exploring the same issue, more recently a
aggressive therapy schedule, toxicity, and physi- large retrospective analysis has been reported.
cian’s recommendation due to comorbid illness. Two thousand seven hundred twenty-four
196 C. Fernandez-Martos and X.G. de Albeniz

patients were included. Forty-one percent under- clear (R0) resections despite nearly a third having
went adjuvant chemotherapy, which consisted had tumours that were initially threatening or
mostly of 5FU-based chemotherapy. The aim of involving the circumferential resection margin
that analysis was to determine whether patients (CRM). However, only one pathological com-
with pathological complete response, ypT1–T2 plete response was achieved.
or ypT3–T4 tumour after CRT have different More recently, investigators from the same
benefits of adjuvant chemotherapy for DFS. The group have published the largest phase II trial
HR with 95% CI for DFS for adjuvant chemo- (first published in 2006 with 77 patients and
therapy was 0.94 (0.50–1.78) for patients with updated 2010 with 105 patients and median fol-
pCR, 0.61 (0.40–0.92) for patients with ypT1–T2 low-up of 55 months) which examined the use of
tumours and 0.97 (0.75–1.25) for patients with four cycles of induction, this time with capecit-
ypT3–T4 tumours. ypT1–T2N0 benefit most abine plus oxaliplatin (CAPOX) followed by
from adjuvant CT, and for ypT3-T4 patients, pN- CRT with capecitabine [11, 13]. This trial
stage did not alter benefit of adjuvant chemo [4]. included poor-risk rectal cancer patients as
defined by magnetic resonance imaging (MRI).
Eligible patients must have tumours within 1 mm
20.3 Induction Chemotherapy of mesorectal fascia (i.e. circumferential resec-
Followed by Concomitant tion margin threatened), T3 tumours at or below
Chemoradiotherapy levators, tumours extending 5 mm into perirectal
and Surgery fat, T4 tumours and T1–4N2 tumours. There was
a clinically significant occurrence of cardiac/
Treatment of high-risk patients with fluoro- thromboembolic (CTE) toxicity during neoadju-
pyrimidines and concurrent radiotherapy without vant oxaliplatin/capecitabine, which led to three
the addition of adequate doses and cycles of CT mortalities. This fact prompted a protocol amend-
appears to be less than optimal. Strategies to ment, and more stringent criteria were added to
improve adherence to the systemic component of exclude entry of patients with a previous history
the treatment are necessary. of stable angina, arrhythmia, acute coronary syn-
One approach to address this issue is to deliver drome (even if controlled by drugs) or myocar-
induction CT before preoperative CRT. Induction dial infarction within the past 12 months. After
CT may be associated with better treatment com- amendment of the protocol, only one further fatal
pliance and may enable full systemic doses of CT pulmonary embolism occurred.
to be delivered. Other theoretical advantages of High radiological response rates to preopera-
induction CT include the possibility of shrinking tive treatment were recorded (96% of assessable
or downstaging a locally advanced tumour, thereby patients), 20% pCR and no disease progression
facilitating more effective local treatment and early during preoperative treatment. Three- and 5-year
treatment of micrometastasis. Disadvantages failure-free survival (ITT population) was 68%
include the delay in surgery and the reduced and 64%, respectively; 3- and 5-year overall sur-
efficacy of subsequent radiotherapy with selection vival was 83% and 75%, respectively. Clinical
of radiotherapy-resistant clones [23]. results of these and other phase II trials are given
The first study published with this strategy in Table 20.1.
was from the UK, in which 36T3/T4 and N×/+ On the basis of these encouraging results, we
rectal cancer patients were treated with 12 weeks designed a phase II, randomized trial to compare
of induction protracted venous infusion of this approach with conventional preoperative CRT
fluorouracil and mitomycin C before standard followed by surgery and postoperative adjuvant
chemoradiation and surgery [12]. A high rate of CT [17] (Fig. 20.2). A total of 108 patients with
tumour downstaging was recorded without any locally advanced rectal (LAR) cancer as defined
progressors during neoadjuvant chemotherapy, by magnetic resonance imaging (MRI) of the pel-
and 82% of patients achieved microscopically vis were randomly assigned to preoperative CRT
Table 20.1 Phase II studies with induction chemotherapy followed by chemoradiotherapy (CRT) with or without adjuvant chemotherapy in rectal cancer patients
20

Series n Inclusion criteria Treatment Toxicity pCR DFS


Chau et al. [12] 36 MRI or CT + DRE T3/ Induction CT PVI 5FU + MMC 12 weeks Induction CT: G3/4: 25% 2.7%
T4 N×/+ M0
CRT PVI 5FU during RT 50.4–54 Gy CRT: G3/4 28% (skin)
Surgery: 4–6 weeks after CRT Surgery: 1 death (anastomotic leak with multiorgan
failure)
Adjuvant 12 weeks. PVI 5FU, MMC
Chau et al. [11] 105 MRI: Induction CT: CAPOX 4 cycles Induction CT: 5 deaths 20%
(updated
T3 £1 mm to CRM CRT: Starting week 13, Cape during RT 54 Gy 1 neutropenic sepsis
Chua [13])
T3 at/below levators Surgery: 6 weeks after CRT 2 pulmonary embolus
T3 ³5 mm into fat; T4; Adjuvant Cape, 4 cycles 1 myocardial infarction
N2; M0
1 cardiac failure; G3–5 diarrhea 10%
CRT: G3–5: 42% skin
Surgery: no deaths within 30 days
Calvo et al. [9] 52 US T3/T4 or N+ M0 Induction CT: FOLFOX −4, 2 cycles Induction CT: G3: 6% N.R.
CRT: starting day 30, Tegafur during RT 45–50.4 Gy CRT: G3–4: 33% ypT0: 29%
Surgery: 4–6 weeks after CRT + IORTBoost Surgery: 31% postoperative complications
(10–15 Gy)
Adjuvant CT: left to the treating oncologist’s discretion
Koeberle 60 MRI or US T3/T4 Induction CT: CAPOX 1 cycle Induction CT: G3 17% diarrhea, 3% G4 infection, 1/60 23%
et al. [31] N×/+ M0 died 19 days after start of CT due to neutropenic sepsis
CRT: starting day 22, Cape and Oxa during RT 45 Gy CRT: G3 17% diarrhea
Surgery: 5–6 weeks after CRT Surgery: not given
Should Upfront Chemotherapy Precede Preoperative Chemoradiation and Surgery?

Adjuvan CT: left to the treating oncologist’s discretion


Gunnlaugsson 49 “Nonresectable” Induction CT: CAPOX 2 cycles Induction CT: G3/4: 11% diarrhea 13%
et al. [25] colorectal carcinoma
T4N × M0–1
41 pts rectal CRT: starting week 7, Cape and Oxa during RT 50.4 Gy CRT: G3/4: 24% diarrhea
Surgery: performed, if feasible 1 pt died due to myocardial infarction
Adapted from Rödel et al. [36]
197

5-FU 5-Fluorouracil, MMC Mitomycin C, pCR pathological complete response, CT chemotherapy, RT radiotherapy, MRI Magnetical Resonance Image, DRE Digital Rectal Examination,
US Ultrasonography, FOLFOX 5FU Leucovorin and Oxaliplatin, CAPOX Capecitabine and Oxaliplatin, N.R. Not reported
198 C. Fernandez-Martos and X.G. de Albeniz

Resectable rectal adenocarcinoma


RMI defined locally advanced rectal
cancer <12 cm anal verge

Stratification: center

Randomization
Cape: 825 mg/m2 × 2 Cape : 2,000 mg/m2 14 days. 1 week rest
5 days/ week-5 weeks Oxa: 130 mg/m2 day 1
Concomitant with RT 4 cycles
+
Oxa: 50 mg/m2 weekly × 5 Cape: 825 mg/m2 x2
Concomitant with RT 5 days/ week-5 weeks
+ Concomitant with RT
Pelvic RT : 5,040 cGys +
5 − 6 weeks Oxa: 50 mg/m2 weekly x 5
Surgery Concomitant with RT
+
Pelvic RT : 5,040 cGys
Cape : 2,000 mg/m2 14 days. 1 week rest 5 − 6 weeks
Oxa 130 mg/m2 day 1
Surgery
4 cycles

Fig. 20.2 Schema for randomized, Grupo Cáncer de Recto (GCR) 3, phase II study. Cape capecitabine, RT radiation
therapy, Oxa oxaliplatin

with capecitabine, oxaliplatin and concurrent Furthermore, 25% of patients in the adjuvant arm
radiation followed by surgery and four cycles of A did not begin treatment, and 51% received all
postoperative adjuvant capecitabine and oxalipla- four cycles, whereas 100% of patients in the
tin (CAPOX) or induction CAPOX followed by induction arm began treatment, and 92% received
CRT and surgery. MRI criteria for LAR cancer all four cycles. These differences reached statisti-
were as follows: tumours extending to within cal significance.
2 mm of, or beyond, the mesorectal fascia (i.e. an The updated results with a median follow-up
involved or threatened circumferential resection time of 39.3 months and 36 months DFS rates
margin); lower third (£6 cm from the anal verge) were 68% (95% CI, 53–80%) for adjuvant arm
cT3 tumours; resectable cT4 tumours and any and 70% (95% CI, 55–80%) in the induction arm
cT3N+. The primary endpoint was pathologic (P = 0.97) (Fig. 20.3). Analyzing the subset of
complete response rate (pCR). On an intention- patients at higher risk (i.e. patients with cT4, cT3
to-treat basis, the pCR for the standard strategy distal third or CRM + tumours), the 36 months
(adjuvant CT) was 13.5% (95% CI, 5.6–25.8%) DFS was 62% for the adjuvant arm versus 70%
and for the induction strategy 14.3% (95% CI, for the induction arm. Although the numbers are
6.4–26.2%) respectively. There were no statisti- small, this insignificant difference may indicate a
cally significant differences in other endpoints, trend towards some benefit of the induction strat-
including downstaging, tumour regression and egy in the higher-risk population [16]. Further
R0 resection. evidence that induction chemotherapy can con-
The most compelling results of this study tribute to better control of distant relapse comes
concern the secondary endpoints. In the course from the recently published NSABP R-03 phase
of CRT, there were no differences between the III trial [38]. This is the first trial to document a
arms in the number of patients with grades 3–4 significant improvement in disease-free survival
toxicity. However, during adjuvant/induction in patients undergoing neoadjuvant chemoradio-
CT, significantly more patients in adjuvant arm therapy therapy. Patients enrolled in R-03 received
than in the induction arm had grades 3–4 toxic- 6 weeks of fluorouracil (500 mg/m2) and leuco-
ity (54% vs. 19%; P = 0.0004, respectively). vorin (500 mg/m2) before the start of radiotherapy
20 Should Upfront Chemotherapy Precede Preoperative Chemoradiation and Surgery? 199

1.00

0.75 69%
Survival probability

68%
0.50

Adjuvant arm
0.25 Induction arm
Subjects Event Censored Median (CL 95%)
Brazo A 52 29% (15) 71% (37) NA (NA, NA)
Brazo B 56 29% (16) 71% (40) NA (NA, NA)
Log−Rank 0.9704
0.00
0 6 12 18 24 30 36 42 48 54
Time from consent date (months)
Number of patients at risk
Brazo A 52 47 44 41 40 36 27 18 2 0
Brazo B 56 54 46 41 38 28 16 5 0

Fig. 20.3 3-year failure-free survival for all eligible patients in GCR 3 trial

(cycle 1). Two courses of chemotherapy (cycles 2 5FU and radiation increases the pathological
and 3) were administered during the first and fifth complete response (pCR) rate over radiother-
week of radiotherapy and four courses (cycles apy alone and improve locoregional control
4–7) postoperatively. Perhaps a 6-week course of but to date have not demonstrated an improve-
induction chemotherapy and a greater total cumu- ment in survival outcomes. The phase III stud-
lative dose of FU are more effective in control- ies published in the last decade include a
ling micrometastatic disease and improving clinically staged population of heterogeneous
disease-free survival. risk. Added to this, a frequently occurring fact
is that there is a poor adherence to the adjuvant
Conclusion treatment with chemotherapy after surgery. In
Several conclusions can be drawn based on the this scenario, no randomized study has shown
studies discussed above. In colon cancer, the that delivery of adjuvant chemotherapy in
recommended adjuvant treatment is based on patients undergoing preoperative radio(chemo)
trials with an appropriate selection of patients therapy improves outcome as compared with
with pathological stages II and III, and one observation. Emerging evidence from several
important observation is that the benefit in phase II trials and, recently, randomized phase
terms of DFS is more evident in high-risk II trials indicates that induction chemotherapy
groups. Moreover, the patients are included in for rectal cancer patients is feasible, does not
clinical trials following surgery, including only compromise pCR, or R0 resection rates, is
patients who are well recovered after surgery less toxic and enables chemotherapy to be
and with a good performance status to receive delivered in adequate dose and intensity.
complementary treatment, allowing in this way Nonetheless, while these data are statistically
a high compliance with the planned treatment. and potentially clinically meaningful, these
On the other hand, in cancer of the rectum, pre- remarkable findings are regarded as hypothesis
operative combined modality treatment with generating. Given the fact that the predominant
200 C. Fernandez-Martos and X.G. de Albeniz

site of failure after combined modality treat- 20.4.2.2 Secondary Endpoint


ment in rectal cancer is systemic rather than Pathologic complete response, overall survival,
local, we think that introducing a systemically toxicity, genetic signature validation, tumour
active combination chemotherapy prior to regression grade and rate of local relapse.
CRT, in a high-risk population selected by
MRI, may well be the next step in phase III 20.4.2.3 Study Population
testing in order to improve DFS (see proposal Further progress in the management of locally
for phase III trial in Appendix). advanced rectal cancer will require the ability to
define which patients may or may not benefit from
preoperative treatment and to develop new treat-
ment strategies for microscopic distant disease.
20.4 Appendix: Evidence to Proceed The extent of extramural tumour spread (EMS),
to a Phase III Trial circumferential resection margin (CRM) and nodal
status as well as extramural venous invasion
The GCR-3 trial is relevant due to the following (EMVI) have been identified as powerful predic-
points [17]: tive factors for local recurrence, distant metastases
• It addresses a relevant clinical question. A and overall survival in patients with rectal cancer
priori, better compliance with the adjuvant [26, 27, 33, 34]. Although currently a reliable ass-
regimen can be translated in a better control of essment of the pretherapeutic mesorectal regional
micrometastatic disease. lymph node status is not possible by the present
• Less toxicity also translates into a better qual- imaging modalities [32], thin-slice high-resolution
ity of life for the patient and lower expenses. MRI can identify accurately the extent of EMS,
• It uses a legitimate endpoint for phase II the EMVI and the status of CRM [7, 42].
trials in rectal cancer, pathological complete
response. Main eligibility criteria:
• It is a randomized phase II trial with an appro- 1. Patient with rectal adenocarcinoma: Distal
priate control arm. border of tumour below the sacral promontory
• It opens the door to explore a new strategy of 2. Any T4 resectable
treatment delivery that may improve patient care. 3. Any distal (less or equal to 5 cm to anal verge)
T3
4. T3 middle third tumours threatening or affect-
20.4.1 Question to Answer ing the mesorectal fascia (distance from
tumour to fascia £ than 2 mm)
With this background, the question to answer 5. T3 middle third and one or both of the
would be: ‘Does the new regimen (adjuvant che- following:
motherapy administered prior to CRT and TME) (a) T3 >5 mm EMS
improve the disease-free survival of patients with (b) Presence of EMVI
resectable rectal cancer?’

20.4.3 Biomarkers
20.4.2 Study Design
A recent publication [10] showed that a 13-gene
20.4.2.1 Primary Endpoint signature could predict, with 86% accuracy, rec-
Superiority in disease-free survival (DFS), tal cancer patients that will respond to CRT. This
defined as the time from date of randomization study was retrospective and validation in a differ-
until disease progression, relapse or death from ent population and prospectively is mandatory to
any cause. establish the clinical utility to such a signature.
20 Should Upfront Chemotherapy Precede Preoperative Chemoradiation and Surgery? 201

Therefore, we considered our study an ideal sce- is 62%. The hazard ratio comparing the DFS in
nario for such a validation. the induction arm to the control arm is 0.78. This
is equivalent, based on design assumptions, to a
3-year DFS rate in the control arm of 62% versus
20.4.4 Randomization 70% in the induction arm. Under these assump-
tions, a total of 1,028 patients would be needed.
A blocked randomization with blocks of size 4
and 6 is proposed with the following stratification
factors: References
• Institution
• Circumferential margin threatened or involved 1. André T, Boni C, Navarro M (2009) Improved overall
versus neither survival with oxaliplatin, fluorouracil, and leucovorin
as adjuvant treatment in stage II or III colon cancer in
the MOSAIC trial. J Clin Oncol 27(19):3109–3116
2. André T, Boni C, Mounedji-Boudiaf L et al (2004)
20.4.5 Sample Size Oxaliplatin, fluorouracil, and leucovorin as adjuvant
treatment for colon cancer. N Engl J Med 350:
2343–2351
In order to ensure an appropriate power, an adap-
3. Assed D, Coelho S, de Freitas D et al (2010)
tive design will be used as recommended by the Combination therapy in high-risk stage II or stage
FDA [15]. The strategy to maintain power is to III colon cancer: current practice and future prospects.
adjust the sample size if the overall event rate is Ther Adv Med Oncol 2(4):261–272
4. Beets G, Maas M, Nelemans P et al (2011) Evaluation
lower than initially anticipated, the variability of
of response after chemoradiation for rectal cancer as a
the main outcome is greater than expected or the predictive factor for the benefit of adjuvant chemo-
adherence is low. In the case of ‘time-to-event’ therapy: a pooled analysis of 2,724 individual patients.
main outcomes, this adaptive design is straight- J Clin Oncol 29(suppl 4; abstract 361)
5. Bosset JF, Collette L, Calais G et al (2006)
forward. As the power depends on the number of
Chemotherapy with preoperative radiotherapy in rec-
events rather than on the number of patients tal cancer. N Engl J Med 355:1114–1123
recruited, either the length of follow-up, the num- 6. Bosset JF, Calais J, Mineur L et al (2005) Enhanced
ber of recruited patients or both can be increased tumorocidal effect of chemotherapy with preoperative
radiotherapy for rectal cancer: preliminary results –
to achieve the desired power (event-driven trials).
EORTC 22921. J Clin Oncol 23:5620–5627
Given that a time-to-event analysis is used, 7. Brown G, Radcliffe AG, Newcombe RG et al (2003)
instead of planning a specific sample size, patient Preoperative assessment of prognostic factors in rec-
enrolment will be continued until the a priori tal cancer using high-resolution magnetic resonance
imaging. Br J Surg 90:355–364
specified number of events occur. Two interim
8. Bujko K, Glynne-Jones R, Bujko M (2010) Does
analyses (a total number of looks of three) will be adjuvant fluoropyrimidine-based chemotherapy pro-
performed to assess both efficacy and futility. vide a benefit for patients with resected rectal cancer
who have already received neoadjuvant radiochemo-
therapy? A systematic review of randomized trials.
Assumptions:
Ann Oncol 21(9):1743–1750
Bilateral alpha level: 0.05 9. Calvo FA, Serrano FJ, Diaz-Gonzalez JA et al (2006)
Power of 80% Improved incidence of pTO downstaged surgical spec-
Assigned fraction of treatment: 0.5 imens in locally advanced rectal cancer (LARC) treated
with induction oxaliplatin plus 5-fluorouracil and pre-
Planned number of looks (equally spaced): 3
operative chemoradiation. Ann Oncol 17:1103–1110
Alpha and beta spending function: Lan- 10. Casado E, Moreno V, Sanchez JJ et al (2011) A com-
DeMets bined strategy of SAGE and quantitative PCR pro-
Subject accrual per month: 20 vides a 13-gene signature that predicts preoperative
chemoradiotherapy response and outcome in rectal
Length of patients’ accrual: 60 months
cancer. Clin Cancer Res 17:4145–4154
Based on preliminary data and literature review, 11. Chau I, Brown G, Cunningham D et al (2006)
the expected 3-year DFS rate in the control arm Neoadjuvant capecitabine and oxaliplatin followed by
202 C. Fernandez-Martos and X.G. de Albeniz

synchronous chemoradiation and total mesorectal sis, treatment and follow-up. Ann Oncol 21(Suppl
excision in magnetic resonance imaging-defined poor- 5):v82–v86
risk rectal cancer. J Clin Oncol 24:668–674 23. Glynne-Jones R, Grainger J, Harrison J et al (2006)
12. Chau I, Allen M, Cunningham D et al (2003) Neoadjuvant chemotherapy prior to preoperative
Neoadjuvant systemic fluorouracil and mitomycin C chemoradiation or radiation in rectal cancer: should
prior to synchronous chemoradiation is an effective we be more cautious? Br J Cancer 94:363–371
strategy in locally advanced rectal cancer. Br J Cancer 24. Gray R, Barnwell J, McConkey C et al (2007)
88:1017–1024 Adjuvant chemotherapy versus observation in patients
13. Chua YJ, Barbachano Y, Cunningham D et al (2010) with colorectal cancer: a randomised study. Lancet
Neoadjuvant capecitabine and oxaliplatin before 370:2020–2029
chemoradiotherapy and total mesorectal excision in 25. Gunnlaugsson A, Anderson H, Fernebro E et al (2009)
MRI-defined poor-risk rectal cancer: a phase 2 trial. Multicentre phase II trial of capecitabine and oxalipl-
Lancet Oncol 11:241–248 atin in combination with radiotherapy for unresect-
14. Collette L, Bosset JF, den Dulk M et al (2007) Patients able colorectal cancer: the CORGI-L study. Eur J
with curative resection of cT3–4 rectal cancer after Cancer 45:807–813
preoperative radiotherapy or radiochemotherapy: does 26. Hermanek P, Merkel S, Fietkau R et al (2010) Regional
anybody benefit from adjuvant fluorouracil-based lymph node metastasis and locoregional recurrence of
chemotherapy? A trial of the European organization rectal carcinoma in the era of TNM surgery.
for research and treatment of cancer radiation oncol- Implications for treatment decisions. Int J Colorectal
ogy group. J Clin Oncol 25:4379–4386 Dis 25:359–368
15. Cook T, DeMets DL (2010) Review of draft FDA 27. Horn A, Dahl O, Morild I (1991) Venous and neural
adaptive design guidance. J Biopharm Stat 20: invasion as predictors of recurrence in rectal adeno-
1132–1142 carcinoma. Dis Colon Rectum 34:798–804
16. Fernandez-Martos C, Pericay C, Salud A et al (2011) 28. Kalady MF, Sanchez JA, Manilich E et al (2009)
Three-year outcome of GCR-3: a phase II randomized Divergent oncogenic changes influence survival dif-
trial comparing conventional preoperative chemora- ferences between colon and rectal adenocarcinomas.
diation (CRT) followed by surgery and postoperative Dis Colon Rectum 52:1039–1045
adjuvant chemotherapy (CT) with induction CT fol- 29. Kapiteijn E, Marijnen CA, Nagtegaal ID et al (2001)
lowed by CRT and surgery in locally advanced rectal Preoperative radiotherapy combined with total
cancer. J Clin Oncol 29(suppl; abstract 3552) mesorectal excision for resectable rectal cancer. N
17. Fernandez-Martos C, Pericay C, Aparicio J et al Engl J Med 345:638–646
(2010) Phase II, randomized study of concomitant 30. Khrizman P, Niland JC, Veer A et al (2011)
chemoradiotherapy followed by surgery and adjuvant Postoperative adjuvant chemotherapy (CTX) use in
capecitabine plus oxaliplatin (CAPOX) compared patients (Pts) with stage II/III rectal cancer treated
with induction CAPOX followed by concomitant with neoadjuvant therapy: a National Comprehensive
chemoradiotherapy and surgery in magnetic reso- Cancer Network (NCCN) analysis. J Clin Oncol
nance imaging-defined, locally advanced rectal can- 29(suppl; abstract 3515)
cer: Grupo cancer de recto 3 study. J Clin Oncol 28: 31. Koeberle D, Burkhard R, von Moos R et al (2008)
859–865 Phase II study of capecitabine and oxaliplatin given
18. Fernandez-Martos C, Aparicio J, Bosch C et al (2004) prior to and concurrently with preoperative pelvic
Preoperative uracil, tegafur, and concomitant radio- radiotherapy in patients with locally advanced rectal
therapy in operable rectal cancer: a phase II multi- cancer. Br J Cancer 98:1204–1209
center study with 3 years’ follow-up. J Clin Oncol 32. Lahaye MJ, Engelen SME, Nelemans PJ et al (2005)
22:3016–3022 Imaging for predicting the risk factors – the circum-
19. Fransen K, Klintenas M, Osterstrom A et al (2004) ferential resection margin and nodal disease – of local
Mutation analysis of the BRAF, ARAF, and RAF-1 genes recurrence in rectal cancer: a meta-analysis. Semin
in human colorectal adenocarcinomas. Carcinogenesis Ultrasound CT MRI 26:259–268
25:527–533 33. Merkel S, Mansmann U, Siassi M et al (2001) The
20. Gerard JP, Conroy T, Bonnetain F et al (2006) prognostic inhomogeneity in pT3 rectal carcinomas.
Preoperative radiotherapy with or without concur- Int J Colorectal Dis 16:298–304
rent fluorouracil and leucovorin in T3–4 rectal can- 34. Nagtegaal I, Quirke P (2008) What is the role for the
cers. Results of FFCD 9203. J Clin Oncol 24: circumferential margin in the modern treatment of
4620–4625 rectal cancer? J Clin Oncol 26:303–312
21. Gill S, Loprinzi C, Sargent D et al (2004) Pooled anal- 35. NCCN Clinical Practice Guidelines in Oncology
ysis of fluorouracil-based adjuvant therapy for stage II (2011) Rectal Cancer. Version 4.2011. http://NCCN.org
and III colon cancer: who benefits and by how much? Accessed Dec 2011
J Clin Oncol 22(10):1797–1806 36. Rödel C, Arnold D, Becker H et al (2010) Induction
22. Glimelius B, Pahlman L, Cervantes A (2010) Rectal chemotherapy before chemoradiotherapy and sur-
cancer: ESMO clinical practice guidelines for diagno- gery for locally advanced rectal cancer. Is it time for
20 Should Upfront Chemotherapy Precede Preoperative Chemoradiation and Surgery? 203

a randomized phase III trial? Strahlenther Onkol 41. Sebag-Montefiore D, Rutten H, Rullier E et al (2009)
186:658–664 Three-year survival results of CORE (Capecitabine,
37. Rödel C, Liersch T, Hermann R et al (2007) Multicenter Oxaliplatin, Radiotherapy, and Excision) study after
phase II trial of chemoradiation with oxaliplatin for postoperative chemotherapy in patients with locally
rectal cancer. J Clin Oncol 25:668–674 advanced rectal adenocarcinoma. ASCO GI Meeting.
38. Roh M, Colangelo L, O’Connell M et al (2009) Abstracts 447
Preoperative multimodality therapy improves disease- 42. Smith N, Barbachano Y, Norman A et al (2008)
free survival in patients with carcinoma of the rectum: Prognostic significance of magnetic resonance imag-
NSABP R-03. J Clin Oncol 27:5124–5130 ing-detected extramural vascular invasion in rectal
39. Russo A, Borger D, Ryan D et al (2011) Mutational cancer. Br J Surg 95:229–236
analysis and clinical correlation of 185 consecutive 43. Valentini V, Grimelius B (2010) Rectal cancer radio-
metastatic colorectal patients: similarities and differ- therapy: Towards European consensus. Acta Oncol
ences between colon and rectal cancers patients. 49:1206–1216
J Clin Oncol 29(suppl; asbtract 3536)
40. Sauer R, Becker H, Hohenberger W et al (2004)
Preoperative versus postoperative chemoradiotherapy
for rectal cancer. N Engl J Med 351:1731–1740
How to Achieve Long-Term
Survival in Patients 21
with Metastatic Rectal Cancer?

Alexander Stein and Hans-Joachim Schmoll

Contents 21.6 Controversies About Preoperative


Treatment.................................................... 212
21.1 Introduction ................................................ 205 21.6.1 Complete Clinical Remission During
21.2 Management of Patients Preoperative Chemotherapy ......................... 212
with Metastatic Rectal Cancer.................. 206 21.6.2 Influence of Chemotherapy
on Perioperative Morbidity,
21.3 Selection of Patients Undergoing Mortality and Prognosis ............................... 212
Resection for Metastatic Disease .............. 206
21.7 Management of Limited Peritoneal
21.4 Management of Patients Disease with Cytoreductive Surgery
with Initially R0 Resectable (CRS) and Hyperthermic Intraperitoneal
Liver+/−Lung Metastases .......................... 207 Chemotherapy (HIPEC)............................ 213
21.4.1 Perioperative Treatment of Patients
with R0 Resectable Liver Metastases .......... 209 References ................................................................. 214
21.4.2 Postoperative Treatment of Patients
with R0 Resectable Liver+/−Lung
Metastases .................................................... 209
21.4.3 Peri- Versus Postoperative Treatment
of Patients with R0 Resectable
Liver+/−Lung Metastases ............................ 210
21.4.4 Local Treatment/Hepatic Artery Infusion
(HAI) After Resection of CLM ................... 211 21.1 Introduction
21.5 Management of Patients with Initially
Colorectal cancer (CRC) is the most frequently
Unresectable Metastases............................ 211
21.5.1 Current Developments ................................. 212 diagnosed cancer in Europe and one of the lead-
ing causes of cancer death worldwide [20, 29].
About 20–25% of patients with CRC present
with metastatic disease at time of diagnosis. The
majority of patients with metastatic rectal cancer
will have liver+/−lung metastases. All patients
A. Stein with metastatic rectal cancer should be discussed
University Cancer Center Hamburg, and managed within a multidisciplinary team to
University Medical Center Hamburg-Eppendorf, ensure selection of patients, which can be treated
Hamburg, Germany
with curative intent combining systemic (chemo-
H.-J. Schmoll (*) therapy) and local (surgery, radiotherapy) treat-
Department for Oncology/Haematology,
ment, although achievement of long-term survival
Martin-Luther-University Halle-Wittenberg,
Halle, Germany strongly depends on the biology and therefore
e-mail: hans-joachim.schmoll@medizin.uni-halle.de course of the metastatic disease.

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 205


DOI 10.1007/978-3-642-25005-7_21, © Springer-Verlag Berlin Heidelberg 2012
206 A. Stein and H.-J. Schmoll

Resection of liver metastases from colorectal 21.2 Management of Patients


cancer (CLM) is a potentially curative treatment, with Metastatic Rectal Cancer
and reported 5-year overall survival rates
(5-yOSR) range from 20% to 40% [11, 66]. Prognosis of patients with metastatic disease
Further follow-up after resection of liver metasta- depends mainly on the course of metastatic dis-
ses from colorectal cancer revealed a rate of long- ease; thus, primary treatment aim is to achieve
term survival and finally cure for every fifth resectability of metastatic disease. In case of
patient even in the presence of poor prognostic either initial or secondary resectability, local
factors [1, 22, 58, 59]. Retrospective series treatment according to stage for rectal primary
revealed profound improvements in outcome of should be performed followed by resection of all
patients with colorectal cancer in the last 20 years sites of disease. The choice of treatment sequence,
due to advancements in chemotherapy and dra- chemotherapy, radio(chemo)therapy and surgery
matic increase in patients undergoing liver resec- depends on characteristics of primary tumour
tion [35, 42]. (local symptoms and stage) and metastatic dis-
Moreover, curative surgery can be per- ease (initially or potentially resectable after che-
formed in CLM with extrahepatic disease. motherapy). Possible treatment algorithms are
Resection of lung metastases of colorectal can- summarized in Figs. 21.1 and 21.2 for synchro-
cer could result in a 5-yOSR of 45% in a nous metastatic disease including management of
selected group of patients [28]. Several series primary tumour and in Fig. 21.3 for metachro-
revealed promising 5-yOSR of 30–40% for the nous disease.
resection of both liver and lung metastases
counted after the last organ resection [40, 45,
47]. Retrospective series of 186 patients with 21.3 Selection of Patients
CLM and extrahepatic disease (lung, lymph Undergoing Resection
node metastases and peritoneal metastases) for Metastatic Disease
receiving perioperative chemotherapy and
resection demonstrated a 5-yOSR after first Risk factors for poor outcome after resection of
pulmonary resection for patients with liver and CLM were identified already 25 years ago, e.g.,
lung metastases (n = 88) of 37%, although one- presence of positive portal lymph nodes, extrahe-
third of patients had bilateral pulmonary dis- patic metastases, four or more hepatic metastases
ease. In case of other sites of extrahepatic or primary tumour stage Dukes’ C [26]. The scor-
disease, 5-yOSR was much lower (e.g., 18% ing system for CLM described by Fong and col-
for lymph node metastases) [14]. Recent retro- leagues includes five risk factors: node-positive
spective analysis of 112 patients undergoing primary, disease-free interval <12 months, > one
resection of both liver and lung metastases lesion, size >5 cm and CEA > 200 ng/m. Patients
demonstrated a 5-yOSR of 50%, which was with five risk factors had a 5-yOSR of 14% with a
even better than 5-yOSR after resection of median of 22 months compared to a 5-ySR of
CLM (40% in 1,148 patients) [9]. 44% and a median of 51 months in the presence of
Furthermore, multimodality treatment includ- one risk factor [22]. However, the ‘Fong score’
ing cytoreductive surgery and perioperative intra- does not include contemporary variables like pre-
peritoneal and systemic chemotherapy resulted in operative chemotherapy. Recently, the interna-
a 5-yOSR of 27% in patients with localized perito- tional liver metastases survey, including data of
neal carcinomatosis in a retrospective series of 523 about 4,500 patients with preoperatively treated
patients (35% with synchronous disease) [16]. CLM, confirmed the poor prognosis of patients
The following text refers to metastatic disease with lymph node positive primary, abnormal CEA
with liver and/or lung or limited peritoneal levels and number of CLM, although cut-off value
carcinomatosis. was three in the current analysis. Furthermore,
21 How to Achieve Long-Term Survival in Patients with Metastatic Rectal Cancer? 207

Synchronous rectal cancer with


intact primary and R0/R1
resectable metastases

<T3 NO ≥ T3 or N+

or
3 months Preop RT (5x5) 3 months
preop FOLFOX or CRT preop FOLFOX

3 months Preop RT (5x5)


preop FOLFOX or CRT

Resection of primary and met

Postop CRT or short course RT if T3, CRM+,


N+, perforation

6months postop
3 months postop FOLFOX
FOLFOX

Fig. 21.1 Treatment algorithm for resectable synchronous metastatic rectal cancer

more than one line and six cycles (3 months) of disease. Although number (more than 4) and size
preoperative treatment, incomplete macroscopic (more than 5 cm) have an impact on prognosis
resection (R2), concomitant extrahepatic metasta- [69], patients could still be deemed resectable
ses and progression during preoperative chemo- with these characteristics. The ‘European Expert
therapy were associated with poor prognosis [1]. Panel’ recently stated that the resection should
Whereas scoring for survival is broadly have the potential to be complete and macroscopi-
accepted, criteria for resectability of CLM are cally curative [49].
difficult to define. Although several approaches
were published, no consensus could be reached
yet [43, 49, 52, 62, 69]. In the EORTC 40983 trial, 21.4 Management of Patients with
patients with up to four potentially resectable liver Initially R0 Resectable
metastases and no evidence of extrahepatic dis- Liver+/−Lung Metastases
ease were included [48]. Current clinical trials
include patients depending on resectability deter- The role of perioperative treatment in resectable
mined by a surgeon with hepatic surgery exper- liver+/−lung metastases, especially the point
tise. Limitations for liver surgery are remnant in time (pre- vs. postoperative), mode (systemic
liver of less than 30%, unfavourable location, co- vs. local) and intensity (monotherapy vs. combi-
morbidities excluding major surgery, aggressive nation), is still a matter of debate. Although
tumour biology and/or presence of extrahepatic postoperative therapy after resection of liver
208 A. Stein and H.-J. Schmoll

Synchronous unresectable rectal


cancer with intact primary

Upfront chemotherapy

Resectability of mets achieved?

Yes No

Continue/change chemotherapy
< T3 N0 ≥ T3 or N+

Resectability of mets achieved?


Preop CRT or
short course RT

Yes No
Resection of primary
+ resection of metastases
Avoid radical and mutilating surgery
RCTx or short course RT for locally
advanced tumors
Resume initial treatment for a total
of 6 months

Fig. 21.2 Treatment algorithm for unresectable synchronous metastatic rectal cancer

Metachronous liver/lung
metastases

Clearly R0 resectable Borderline R0 resectable or


initially unresectable

Single, <2 cm FOLFOX Intensive chemotherapy


liver met 3 months preop 3-4(-6) months

Resection No resection

R0/1: FOLFOX R0/1: FOLFOX R0/1: complete periop. Escalate/change


6 months postop 3 months postop chemotherapy for total chemotherapy, than
of 6 months resection, if possible

Fig. 21.3 Treatment algorithm for metachronous metastatic rectal cancer


21 How to Achieve Long-Term Survival in Patients with Metastatic Rectal Cancer? 209

Table 21.1 EORTC trial 40983 of perioperative chemotherapy versus observation in patients with resectable colorec-
tal liver metastases: results by group of analysis
Perioperative Surgery Absolute difference for 3-year
Patients chemotherapy alone progression-free survival rate HR p-value
All 182 182 +7.3% (28.1–35.4%) 0.79 (0.62–1.02) 0.058
Eligible 171 171 +8.1% (28.1–36.2%) 0.77 (0.60–1.00) 0.041
Resected 151 152 +9.2% (33.2–42.4%) 0.73 (0.55–0.97) 0.025
HR hazard ratio, p-value significance level

metastases of colorectal origin CLM has become toxicities occurred in the chemotherapy arm.
an accepted standard of care in many parts of Nearly 80% of patients completed the preopera-
the world, data on the benefit are limited. There tive part. After resection, 76% of patients were
are only a few randomized trials comparing pre- receiving postoperative treatment, with 52% of
and/or postoperative intravenous chemotherapy patients (n = 80) completing six cycles.
or hepatic artery infusion with or without intra- During preoperative chemotherapy, main
venous chemotherapy to surgery alone. grade 3/4 toxicities were nausea and vomiting
Moreover, in adjuvant systemic chemotherapy, 4%, diarrhoea 8%, stomatitis 7%, sensory neu-
none of the conducted clinical trials achieved ropathy 2% and neutropenia 18% (febrile 2%).
the recruitment goal, leading to solely prema- Postoperative reversible complications were
ture data. significantly higher (25% vs. 16%, p = 0.04) in
the chemotherapy group. Particularly intra-
abdominal infections, biliary fistulas with an out-
21.4.1 Perioperative Treatment of put of more than 100 ml/day for more than
Patients with R0 Resectable 10 days and hepatic failure with a bilirubin eleva-
Liver Metastases tion grade 3/4 for more than 3 days occurred
more often in the perioperative group. There was
The EORTC 40983 trial accrued 364 patients to no impact on postoperative death rate (1% in both
be randomized between surgery alone and che- arms). During postoperative chemotherapy, tox-
motherapy with 5-fluorouracil, leucovorin and icities were similar, with a higher rate of grade 3
oxaliplatin (FOLFOX4 regimen), administered sensory neuropathy (10%) and other neurological
3 months pre- and postoperatively [48]. Patients toxicities (12%) due to the higher cumulative
had to be technically resectable (as stated by CT dose of oxaliplatin.
scan) with a maximum of 4 CLM and no prior According to those data, this approach is
treatment with chemotherapy. Despite favourable widely considered as a standard for patients with
patients`characteristics, with 50 % bearing only resectable CLM.
one CLM and surgery at “high-quality” institu-
tions the 3-year progression-free survival rate
was relatively poor. A significant increase in pro- 21.4.2 Postoperative Treatment of
gression-free survival rate at 3 years from 33.2% Patients with R0 Resectable
to 42.4% was shown in the group of patients Liver+/−Lung Metastases
(n = 171) receiving chemotherapy and resection
of CLM (HR 0.73, 95% CI 0.55–0.97). In the Despite being the standard of care in patients
randomized population (‘intent-to-treat analy- with stage III disease, data for adjuvant treatment
ses’), the difference was not statistically after resection of stage IV disease are still limited.
significant due to ineligibility of 6% of patients Two randomized phase III trials have compared
(HR 0.79, 95% CI 0.62–1.02) but still favouring adjuvant systemic chemotherapy with 5FU/FA
the chemotherapy arm (Table 21.1). No unusual after resection of liver or lung metastases to
210 A. Stein and H.-J. Schmoll

Table 21.2 Postoperative treatment after curative resection of liver+/−lung metastases


Postoperative Absolute difference for
Trial Pat. (n) R0 treatment 5yOSR or median OS HR (95% CI) p-value
Langer 129 nr 5FU/FA +10 months (43 vs. 53) 1.30 (0.71–2.36) 0.39
et al. [36]
Portier 171 171 5FU/FA +16 months (46.4 vs. 0.73 (0.48–1.10) 0.13
et al. [51] 62.1)
Ychou 321 321 LV5FU vs. FOLFIRI +1% (71.6% vs. 72.7%) 1.09 (0.72–1.64) 0.69
et al. [70] (3yOSR)
Voest and 79 79 CAPOX (6 m) +18% (52% vs. 70%) nr 0.074
Snoeren [64] vs. CAPOX (2-year DFS rate)
(6 m) + beva (12 m)
(n) number of patients, R0 (n) number of R0 resections, OS overall survival, OSR overall survival rate, HR hazard ratio, CI
confidence interval, p-value significance level, 5FU 5-fluorouracil, FA folinic acid, HAI: nr not reported, LV5FU leuco-
vorin and 5FU, FOLFIRI FA, 5FU and irinotecan, CAPOX capecitabine and oxaliplatin, beva bevacizumab, m months

surgery alone, but both closed prematurely due to However, the lack of a clear benefit might also be
slow accrual. A small but significant improve- related to the relatively poor efficacy, as shown in
ment in disease-free survival rate (DFSR) was the adjuvant treatment of stage III colorectal dis-
demonstrated in the FFCD trial at the time of clo- ease with FOLFIRI [57, 61, 71]. Recently, results
sure, with a 5-year DFSR of 26.7% vs. 33.5% of the prematurely closed Dutch HEPATICA trial
(p = 0.028), favouring the group with adjuvant were presented, demonstrating a DFS benefit
treatment after resection of CLM [51]. Although after 2 years of 18% (70% vs. 52%) in favour of
the 5-year overall survival rate (OSR) increased 12 months bevacizumab with 6 months chemo-
from 41.1% to 51.1%, this difference was not therapy with capecitabine and oxaliplatin com-
significant (p = 0.13). The ENG (EORTC/NCI- pared to the same chemotherapy alone after R0
CTG/GIVIO) trial, still not fully published, dis- resection of CLM in 79 patients [64]. Further
played a non-significant trend towards a prolonged follow-up might reveal whether this benefit is
DFS with a median of 39 vs. 20 months (p = 0.35) only transient, as recently seen with bevacizumab
and an increased overall survival (OS) with a after resection of stage II or III colon cancer
median of 53 vs. 43 months (p = 0.39) in patients [2, 13]. However, it is questionable whether fur-
with liver (n = 90) or lung (n = 13) metastases ther results regarding adjuvant chemotherapy will
[36]. However, a combined analysis of both trials be available, as most current postoperative trials
(n = 278) showed a non-significant prolongation (ADHOC, NSABP C-09 and HEPATICA) were
of the median disease-free survival (DFS) from closed due to insufficient accrual (Table 21.2).
18.8 to 27.9 months (p = 0.058) and the OS from
47.3 to 62.2 months (p = 0.095) [41]. Both trials
were using a non-contemporary 5FU bolus regi- 21.4.3 Peri- Versus Postoperative
men, which is more toxic and possibly not active Treatment of Patients with R0
enough to reveal a significant increase in this lim- Resectable Liver+/−Lung
ited number of patients. Postoperative treatment Metastases
with 5FU plus irinotecan in a prematurely
stopped, randomized phase III trial revealed no Currently, available data seem to favour peri-
additional benefit over 5FU alone in terms of OS operative chemotherapy in clearly R0 resect-
(3-year survival rate of 73% vs. 72%) [70]. able liver metastases, although OS data for the
Regarding median disease-free survival (DFS), perioperative approach are still lacking. In case
there was a non-significant increase of 3 months final analyses of the EORTC 40983 trial cannot
(21.6 vs. 24.7 months, HR 0.90, 95% CI 0.67– demonstrate a significant OS benefit, both
1.2, p = 0.47). The gain in DFS is possibly smaller approaches (peri- and postoperative) would be
as to be detected with 150 patients per group. of similar evidence. However, preoperative
21 How to Achieve Long-Term Survival in Patients with Metastatic Rectal Cancer? 211

treatment offers the option of individual assess- (HR 1.0, 95% CI 0.84–1.21; p = 0.96) [46, 67].
ment of tumour biology and response to chemo- Recently, a small phase II trial, evaluating the
therapy and does therefore help to better select addition of bevacizumab to HAI and systemic
patients for surgery and postoperative treatment; chemotherapy with 5FU/FA and either oxalipla-
e.g., patients with progressive disease during tin or irinotecan, demonstrated increased biliary
preoperative chemotherapy will have no benefit toxicity (p = 0.02) without any difference in RFS
from liver surgery in case of extrahepatic spread or OS [31]. Regarding these results, application
of disease, and even if still resectable, continua- of HAI may not be recommended as a standard
tion of same chemotherapy postoperatively of care.
would not be meaningful. Furthermore, the peri-
operative approach enables administration of
chemotherapy to a higher rate of patients, as 21.5 Management of Patients
postoperative treatment will not be feasible in with Initially Unresectable
up to 20% as demonstrated in the EORTC 40983 Metastases
trial, with 115 of 151 resected patients starting
postoperative FOLFOX and only 80 completing The group of patients with potentially resectable
6 cycles [48]. metastases warrants a highly active, neoadjuvant
treatment aiming on conversion into resectabil-
ity, with tumour shrinkage as an important goal.
21.4.4 Local Treatment/Hepatic Artery Regarding the reported RECIST overall response
Infusion (HAI) After Resection rates (ORR) displayed in clinical trials in the
of CLM first-line treatment for metastatic colorectal can-
cer, either a chemo-doublet in combination with
Several trials were comparing postoperative a monoclonal antibody (e.g., cetuximab, panitu-
hepatic artery infusion combined with or without mumab or bevacizumab) or a three-drug chemo-
systemic treatment to surgery alone after resec- therapy (e.g., FOLFOXIRI) are possible options
tion of CLM with diverging results [37, 55, 65]. [8, 15, 19, 27, 39, 56, 60]. However, the avail-
The systematic review by Nelson et al. of 592 able large trials have included all patients with
patients revealed a non-significant survival metastatic CRC, and the bene fi t con fi ned to
advantage in favour of the control (non-HAI) CLM and ‘conversion rates’ cannot be assessed
group of 8.9% (HR 1.089, 95% CI 0.887–1.334), (Table 21.3).
which is in contrast to a current meta-analysis The CELIM included ‘potentially resectable’
demonstrating a RFS benefit (HR 0.78, 95% CI CLM only. With RECIST response rates of
0.65–0.95; p = 0.01), although OS did not differ 70% resulting from treatment with oxaliplatin- or

Table 21.3 Efficacy of first-line chemotherapy in unselected patients with metastatic colorectal cancer
R0 rate
Trial Treatment Pat. (n) RR (%) RR (‘liver only’) R0 rate all ‘liver only’
Van Cutsem et al. [60] FOLFIRI + cet 316* 57.3* 70.6* (n = 68) 5.1 13.2
Bokemeyer et al. [8] FOLFOX + cet 82* 57* 76* (n = 25) 7.3 16
Maughan et al. [39] 5FU/cape + oxali + cet 362* 64* nr nr nr
Douillard and Siena [15] FOLFOX + pan 593 57* nr nr 28
Hurwitz and IFL + beva 402 45 nr <2% nr
Fehrenbacher [27]
Saltz et al. [56] 5FU/cape + oxali + beva 699 47 nr 6.3 12.3
Falcone et al. [19] FOLFOXIRI 122 66 nr 15 36
*KRAS wildtype, (n) number of patients, R0 (n) number of R0-resections, 5FU 5-fluorouracil, cape capecitabine, oxali
oxaliplatin, irino irinotecan, beva bevacizumab, cet cetuximab, pan panitumumab, nr not reported
212 A. Stein and H.-J. Schmoll

irinotecan-based chemotherapy in combination 21.6 Controversies About


with cetuximab in the KRAS wild-type population, Preoperative Treatment
a conversion rate of 28% could be observed, leading
to R0 resection in 36% of patients [21]. In 45 CLM One of the major drawbacks in neoadjuvant treat-
patients with poor risk features (more than 4 CLM, ment is the conversion into irresectability in case
diameter >5 cm, synchronous disease), treatment of progression during chemotherapy. In the
with capecitabine, oxaliplatin and bevacizumab EORTC 40983 trial, 12 (7%) patients experi-
showed an ORR of 77%, leading to conversion to enced progressive disease during preoperative
resectability in 40% (12 of 30 initially unresectable chemotherapy leading to irresectability in eight
patients) in the BOXER trial [68]. patients, half of them presenting with new lesions
Although tumour shrinkage is important in [48]. However, switch to irresectability reveals
order to achieve technical resectability, and aggressive tumour biology and predicts a worse
RECIST response rates are likely to correlate outcome even in case of resection [1].
well with the percentage of resected or ‘con-
verted’ patients, it has to be kept in mind that
prognosis of patients may (also) be determined 21.6.1 Complete Clinical Remission
by the effect of treatment on tumour tissue. During Preoperative
Interestingly, good correlations of pathohistolog- Chemotherapy
ical response to preoperative chemotherapy,
which seem to be increased with bevacizumab- The issue of complete radiological response has to
containing combinations, with overall survival be kept in mind during neoadjuvant treatment, as
were reported from a cohort of patients after complete clinical response does not mean complete
resection of CLM [7, 33, 54]. Another issue of pathological response [5, 23]. Therefore, patients
defining the ‘optimal regimen’ is that in antian- should be closely monitored during preoperative
giogenic drugs, response to treatment may be chemotherapy and ideally resected before complete
rather displayed in change of radiographic mor- remission [50]. If an anatomical resection can be
phology than in tumour shrinkage only [12]. performed, complete response is not a major prob-
lem because resection will be based on initial sites
of liver metastases. In case of complete response on
21.5.1 Current Developments CT and no option for anatomical resection, differ-
ent imaging methods might be used (MRI, PET
Recent single-arm phase II trials in metastatic scan, contrast-enhanced US) or resection might be
colorectal cancer with four-drug regimens com- delayed until relapse occurs.
bining either EGFR antibodies or bevacizumab
with fluoropyrimidines, oxaliplatin and irinote-
can demonstrated an ORR of 77–82% with a 21.6.2 Influence of Chemotherapy on
complete response rate of about 12% [4, 10, 38]. Perioperative Morbidity,
In regard to a secondary R0 resection rate up to Mortality and Prognosis
26% in an unselected patient population, this
seems to be a promising approach for potentially Chemotherapy can result in severe damage of
resectable patients. Although preliminary efficacy liver tissue. Whereas oxaliplatin-based combina-
of four-drug regimens seems to be similar for tions are associated with increased risk of vascu-
EGFR antibodies and bevacizumab, toxicity lar lesions, irinotecan-containing regimens have
profile favours bevacizumab-containing regi- been associated with increased risk of steatohep-
mens, with rate of grade 3–4 diarrhoea of 52% atitis, especially in obese patients with BMI over
with the ERBIRINOX regimen (cetuximab and 25 kg/m2 [3, 6, 63]. The clinical impact of che-
FOLFIRINOX) compared to 14–18% with beva- motherapy-associated histological changes seems
cizumab and FOLFOXIRI [4, 18, 38]. to favour oxaliplatin in the neoadjuvant setting.
21 How to Achieve Long-Term Survival in Patients with Metastatic Rectal Cancer? 213

Increased risk of perioperative bleeding, postop- tumour dissemination (peritoneal cancer index,
erative complication rate and poorer functional PCI < 20), limited small bowel disease and no
reserve was found to be associated with vascular extra-abdominal metastasis. Furthermore localiza-
lesions. Although influence on morbidity is still tion, histology and lymph node status of the pri-
controversial, mortality was not affected [44, 63]. mary tumour as well as response to systemic
In the EORTC 40983 trial, the perioperative treat- chemotherapy should be considered.
ment resulted in a similar postoperative death rate
as compared to surgery alone, with a significantly Conclusion and Recommendations
higher rate of reversible postoperative complica- Multidisciplinary management of patients
tions (p = 0.04). Higher risk of complications and with metastatic rectal cancer is crucial in order
an increased 90-day mortality (1.6% vs. 14.7%, to select patients amenable for a curative
odds ratio (OR) 10.5) was reported for irinotecan- approach. In regard to long-term survival
associated steatohepatitis [34, 63]. The addition achievable in liver+/−lung metastases or even
of bevacizumab to neoadjuvant oxaliplatin-based in localized peritoneal carcinomatosis, patients
treatment is feasible and does not limit the ability eligible for a curative intent approach should
of liver regeneration [25, 53]. Duration of preop- be transferred to specialized centres for hepatic
erative treatment seems to have major impact on or peritoneal surgery.
perioperative morbidity and prognosis and should Patients with synchronous metastatic rec-
therefore be as short as possible in case of tal cancer should be treated with primary
initially unresectable disease [1, 3, 30, 32]. chemotherapy, followed by secondary local
Duration of perioperative chemotherapy in case treatment including surgical resection if fea-
of initially R0 resectable CLM should be either sible according to extent of disease, response
6 months postoperatively or 3 months pre- and to chemotherapy and co-morbidity with the
postoperatively. aim of long-term survival. Achievement of
resectability of metastatic disease is the main
treatment aim; therefore, the primary tumour
21.7 Management of Limited is less relevant. Only in case of symptomatic
Peritoneal Disease rectal primary tumour local measures (e.g.,
with Cytoreductive insertion of a stent or stoma) and in specific
Surgery (CRS) and circumstances palliative surgical resection
Hyperthermic Intraperitoneal should be performed.
Chemotherapy (HIPEC) For synchronous R0 resectable liver+/−lung
metastases, treatment approaches are as follows:
Peritoneal carcinomatosis as single lesion in • For clearly R0 resectable metastatic dis-
advanced colorectal cancer represents a special ease, irrespective of primary tumour, peri-
biologic entity with poor prognosis under systemic operative chemotherapy (3 months pre- and
chemotherapy alone. Current data including one postoperative FOLFOX) should be applied
randomized controlled trial and numerous pro- analogue to the EORTC 40983 trial [48].
spective and retrospective studies suggest a role of • In locally advanced primary tumours (³T3
CRS and HIPEC within the multimodal treatment or N+): upfront chemotherapy with
regimen and may improve PFS as well as OS for FOLFOX for 3 months and local treatment
selected patients with peritoneal carcinomatosis according to stage (or reverse sequence)
[16, 17, 24]. The procedure can be performed with followed by resection of the primary (staged
acceptable morbidity and low mortality in special- or synchronous) followed by postoperative
ized centres. Nevertheless, preoperative patient FOLFOX for 3 months should be applied.
selection is crucial for the success of the combined • In early primary tumours (<T3 N0): resec-
treatment concept. Main selection criteria are good tion of primary and metastases followed by
general health status, limited intraperitoneal postoperative treatment with FOLFOX for a
214 A. Stein and H.-J. Schmoll

total of 6 months might be considered, and if 4. Assenat E, Desseigne F et al (2011) Cetuximab plus
necessary (e.g., CRM+ or N+) postoperative FOLFIRINOX (ERBIRINOX) as first-line treatment
for unresectable metastatic colorectal cancer: a phase
local treatment according to stage. II trial. Oncologist 16(11):1557–1564
For patients with rectal cancer and synchro- 5. Benoist S, Brouquet A et al (2006) Complete
nous, potentially resectable metastatic disease response of colorectal liver metastases after chemo-
after chemotherapy, the most active available therapy: does it mean cure? J Clin Oncol 24(24):
3939–3945
induction treatment should be chosen. If metas- 6. Benoist S, Nordlinger B (2009) The role of preopera-
tases become resectable, local treatment accord- tive chemotherapy in patients with resectable colorectal
ing to stage for primary followed by resection liver metastases. Ann Surg Oncol 16(9):2385–2390
of primary and metastases should be performed, 7. Blazer DG 3rd, Kishi Y et al (2008) Pathologic
response to preoperative chemotherapy: a new out-
followed by postoperative continuation of the come end point after resection of hepatic colorectal
same regimen for a total of 6 months (including metastases. J Clin Oncol 26(33):5344–5351
preoperative). If metastases remain unresect- 8. Bokemeyer C, Bondarenko I et al (2011) Efficacy
able, treatment should be continued or switched, according to biomarker status of cetuximab plus
FOLFOX-4 as first-line treatment for metastatic col-
depending on quality of response. orectal cancer: the OPUS study. Ann Oncol 22(7):
In case of metachronous disease, similar 1535–1546
treatment approaches should be applied. 9. Brouquet A, Vauthey JN et al (2011) Improved sur-
• Cleary, R0 resectable liver+/−lung metasta- vival after resection of liver and lung colorectal metas-
tases compared with liver-only metastases: a study of
ses should be treated with perioperative che- 112 patients with limited lung metastatic disease.
motherapy (3 months pre- and postoperative J Am Coll Surg 213(1):62–69; discussion 69–71
FOLFOX) and resection. In case of single 10. Bruera G, Santomaggio A et al (2010) “Poker” asso-
liver metastases with a size up to 2 cm, pri- ciation of weekly alternating 5-fluorouracil, irinote-
can, bevacizumab and oxaliplatin (FIr-B/FOx) in first
mary resection followed by postoperative line treatment of metastatic colorectal cancer: a phase
chemotherapy should be considered. II study. BMC Cancer 10:567
• For potentially resectable metastatic dis- 11. Choti MA, Sitzmann JV et al (2002) Trends in long-
ease after chemotherapy, most active induc- term survival following liver resection for hepatic col-
orectal metastases. Ann Surg 235(6):759–766
tion chemotherapy followed by secondary 12. Chun YS, Vauthey JN et al (2009) Association of
resection, if feasible, should be performed. computed tomography morphologic criteria with
pathologic response and survival in patients treated
Acknowledgments The manuscript was prepared with- with bevacizumab for colorectal liver metastases.
out any funding or contribution of persons not mentioned JAMA 302(21):2338–2344
in the authors’ section. 13. De Gramont A, Van Cutsem E (2011) AVANT: Results
from a randomized, three-arm multinational phase III
Conflict of Interest AS honoraria: Roche, Merck study to investigate bevacizumab with either XELOX
Serono. HJS: consultant or advisory role: Roche hono- or FOLFOX4 versus FOLFOX4 alone as adjuvant
raria: Roche; research support: Roche and Merck Serono. treatment for colon cancer. J Clin Oncol 29(suppl 4):
abstr 362
14. de Haas RJ, Wicherts DA et al (2008) Resection of
colorectal liver metastases with extrahepatic disease.
References Dig Surg 25(6):461–466
15. Douillard JY, Siena S (2011) Final results from
1. Adam R, Barroso E (2011) Impact of the type and PRIME: randomized phase III study of panitumumab
modalities of preoperative chemotherapy on the out- (pmab) with FOLFOX4 for first-line metastatic col-
come of liver resection for colorectal metastases. orectal cancer (mCRC). J Clin Oncol 29(suppl): abstr
J Clin Oncol 29(suppl): abstr 3519 3510
2. Allegra CJ, Yothers G et al (2011) Phase III trial 16. Elias D, Gilly F et al (2010) Peritoneal colorectal car-
assessing bevacizumab in stages II and III carcinoma cinomatosis treated with surgery and perioperative
of the colon: results of NSABP protocol C-08. J Clin intraperitoneal chemotherapy: retrospective analysis
Oncol 29(1):11–16 of 523 patients from a multicentric French study.
3. Aloia T, Sebagh M et al (2006) Liver histology and J Clin Oncol 28(1):63–68
surgical outcomes after preoperative chemotherapy 17. Esquivel J, Sticca R et al (2011) Cytoreductive sur-
with fluorouracil plus oxaliplatin in colorectal cancer gery and hyperthermic intraperitoneal chemotherapy
liver metastases. J Clin Oncol 24(31):4983–4990 in the management of peritoneal surface malignancies
21 How to Achieve Long-Term Survival in Patients with Metastatic Rectal Cancer? 215

of colonic origin: a consensus statement. Ann Surg in patients with resected hepatic metastases from col-
Oncol 18(Suppl 3):334–335 orectal cancer. J Clin Oncol 29(7):884–889
18. Falcone A, Loupakis F (2010) FOLFOXIRI plus bev- 32. Kishi Y, Zorzi D et al (2010) Extended preoperative
acizumab (BV) versus FOLFIRI plus BV as first-line chemotherapy does Not improve pathologic response
treatment of metastatic colorectal cancer (MCRC): and increases postoperative liver insufficiency after
preliminary safety results of the phase III randomized hepatic resection for colorectal liver metastases. Ann
TRIBE study by the Gruppo Oncologico Nord-Ovest Surg Oncol 17(11):2870–2876
(GONO). J Clin Oncol 28(15 s): abstr 3543 33. Klinger M, Tamandl D et al (2010) Bevacizumab
19. Falcone A, Ricci S et al (2007) Phase III trial of infu- improves pathological response of colorectal cancer
sional fluorouracil, leucovorin, oxaliplatin, and irino- liver metastases treated with XELOX/FOLFOX. Ann
tecan (FOLFOXIRI) compared with infusional Surg Oncol 17(8):2059–2065
fluorouracil, leucovorin, and irinotecan (FOLFIRI) as 34. Kooby DA, Fong Y et al (2003) Impact of steatosis on
first-line treatment for metastatic colorectal cancer: perioperative outcome following hepatic resection.
the Gruppo Oncologico Nord Ovest. J Clin Oncol J Gastrointest Surg 7(8):1034–1044
25(13):1670–1676 35. Kopetz S, Chang GJ et al (2009) Improved survival in
20. Ferlay J, Shin HR et al (2010) Estimates of worldwide metastatic colorectal cancer is associated with adop-
burden of cancer in 2008: GLOBOCAN 2008. Int J tion of hepatic resection and improved chemotherapy.
Cancer 127(12):2893–2917 J Clin Oncol 27(22):3677–3683
21. Folprecht G, Gruenberger T et al (2010) Tumour 36. Langer B, Bleiberg H, Labianca R, et al (2002)
response and secondary resectability of colorectal Fluorouracil (FU) plus l-leucovorin (l-LV) versus
liver metastases following neoadjuvant chemotherapy observation after potentially curative resection of liver
with cetuximab: the CELIM randomised phase 2 trial. or lung metastases from colorectal cancer (CRC):
Lancet Oncol 11(1):38–47 results of the ENG (EORTC/NCIC CTG/GIVIO) ran-
22. Fong Y, Fortner J et al (1999) Clinical score for domized trial. Proc Am Soc Clin Oncol 21: (abstr 592)
predicting recurrence after hepatic resection for met- 37. Lygidakis NJ, Ziras N et al (1995) Resection versus
astatic colorectal cancer: analysis of 1001 consecu- resection combined with adjuvant pre- and post-oper-
tive cases. Ann Surg 230(3):309–318; discussion ative chemotherapy–immunotherapy for metastatic
318–321 colorectal liver cancer. A new look at an old problem.
23. Gaujoux S, Goere D et al (2011) Complete radiologi- Hepatogastroenterology 42(2):155–161
cal response of colorectal liver metastases after che- 38. Masi G, Loupakis F et al (2010) Bevacizumab with
motherapy: what can we expect? Dig Surg 28(2): FOLFOXIRI (irinotecan, oxaliplatin, fluorouracil,
114–120 and folinate) as first-line treatment for metastatic col-
24. Gava VG, Reidy DL (2010) Surgical cytoreduction in orectal cancer: a phase 2 trial. Lancet Oncol 11(9):
patients with colorectal peritoneal carcinomatosis 845–852
treated with contemporary chemotherapy. J Clin 39. Maughan TS, Adams RA et al (2011) Addition of
Oncol 28(7 s): abstr 3546 cetuximab to oxaliplatin-based first-line combination
25. Gruenberger B, Tamandl D et al (2008) Bevacizumab, chemotherapy for treatment of advanced colorectal
capecitabine, and oxaliplatin as neoadjuvant therapy cancer: results of the randomised phase 3 MRC COIN
for patients with potentially curable metastatic col- trial. Lancet 377(9783):2103–2114
orectal cancer. J Clin Oncol 26(11):1830–1835 40. Miller G, Biernacki P et al (2007) Outcomes after
26. Hughes KS, Simon R et al (1986) Resection of the resection of synchronous or metachronous hepatic
liver for colorectal carcinoma metastases: a multi- and pulmonary colorectal metastases. J Am Coll Surg
institutional study of patterns of recurrence. Surgery 205(2):231–238
100(2):278–284 41. Mitry E, Fields AL et al (2008) Adjuvant chemother-
27. Hurwitz H, Fehrenbacher L et al (2004) Bevacizumab apy after potentially curative resection of metastases
plus irinotecan, fluorouracil, and leucovorin for meta- from colorectal cancer: a pooled analysis of two ran-
static colorectal cancer. N Engl J Med 350(23): domized trials. J Clin Oncol 26(30):4906–4911
2335–2342 42. Morris EJ, Forman D et al (2010) Surgical manage-
28. Inoue M, Ohta M et al (2004) Benefits of surgery for ment and outcomes of colorectal cancer liver metasta-
patients with pulmonary metastases from colorectal ses. Br J Surg 97(7):1110–1118
carcinoma. Ann Thorac Surg 78(1):238–244 43. Nagashima I, Takada T et al (2006) Proposal of a new
29. Jemal A, Bray F et al (2011) Global cancer statistics. and simple staging system of colorectal liver metasta-
CA Cancer J Clin 61(2):69–90 sis. World J Gastroenterol 12(43):6961–6965
30. Karoui M, Penna C et al (2006) Influence of pre- 44. Nakano H, Oussoultzoglou E et al (2008) Sinusoidal
operative chemotherapy on the risk of major hepa- injury increases morbidity after major hepatectomy in
tectomy for colorectal liver metastases. Ann Surg patients with colorectal liver metastases receiving pre-
243(1):1–7 operative chemotherapy. Ann Surg 247(1):118–124
31. Kemeny NE, Jarnagin WR et al (2011) Randomized 45. Neeff H, Horth W et al (2009) Outcome after resec-
phase II trial of adjuvant hepatic arterial infusion and tion of hepatic and pulmonary metastases of colorec-
systemic chemotherapy with or without bevacizumab tal cancer. J Gastrointest Surg 13(10):1813–1820
216 A. Stein and H.-J. Schmoll

46. Nelson R, Freels S (2006) Hepatic artery adjuvant metastases defines cure. J Clin Oncol 25(29):
chemotherapy for patients having resection or abla- 4575–4580
tion of colorectal cancer metastatic to the liver. 60. Van Cutsem E, Kohne CH et al (2011) Cetuximab
Cochrane Database Syst Rev(4): CD003770 plus irinotecan, fluorouracil, and leucovorin as first-
47. Nojiri K, Tanaka K et al (2011) Efficacy of surgery for line treatment for metastatic colorectal cancer:
lung metastases from colorectal cancer synchronous updated analysis of overall survival according to
to or following that for liver metastases. Anticancer tumor KRAS and BRAF mutation status. J Clin Oncol
Res 31(3):1049–1054 29(15):2011–2019
48. Nordlinger B, Sorbye H et al (2008) Perioperative che- 61. Van Cutsem E, Labianca R et al (2009) Randomized
motherapy with FOLFOX4 and surgery versus surgery phase III trial comparing biweekly infusional
alone for resectable liver metastases from colorectal fluorouracil/leucovorin alone or with irinotecan in the
cancer (EORTC intergroup trial 40983): a randomised adjuvant treatment of stage III colon cancer:
controlled trial. Lancet 371(9617):1007–1016 PETACC-3. J Clin Oncol 27(19):3117–3125
49. Nordlinger B, Van Cutsem E et al (2009) Combination 62. Van Cutsem E, Nordlinger B et al (2006) Towards a
of surgery and chemotherapy and the role of targeted pan-European consensus on the treatment of patients
agents in the treatment of patients with colorectal liver with colorectal liver metastases. Eur J Cancer
metastases: recommendations from an expert panel. 42(14):2212–2221
Ann Oncol 20(6):985–992 63. Vauthey JN, Pawlik TM et al (2006) Chemotherapy
50. Nordlinger B, Van Cutsem E et al (2007) Does chemo- regimen predicts steatohepatitis and an increase in
therapy prior to liver resection increase the potential 90-day mortality after surgery for hepatic colorectal
for cure in patients with metastatic colorectal cancer? metastases. J Clin Oncol 24(13):2065–2072
A report from the European Colorectal Metastases 64. Voest EE, Snoeren N (2011) A randomized two-arm
Treatment Group. Eur J Cancer 43(14):2037–2045 phase III study to investigate bevacizumab in combi-
51. Portier G, Elias D et al (2006) Multicenter random- nation with capecitabine plus oxaliplatin (CAPOX)
ized trial of adjuvant fluorouracil and folinic acid versus CAPOX alone in post radical resection of
compared with surgery alone after resection of col- patients with liver metastases of colorectal cancer.
orectal liver metastases: FFCD ACHBTH AURC J Clin Oncol 29(suppl): abstr 3565
9002 trial. J Clin Oncol 24(31):4976–4982 65. Wagman LD, Kemeny MM et al (1990) A prospec-
52. Poston GJ, Figueras J et al (2008) Urgent need for a tive, randomized evaluation of the treatment of col-
new staging system in advanced colorectal cancer. orectal cancer metastatic to the liver. J Clin Oncol
J Clin Oncol 26(29):4828–4833 8(11):1885–1893
53. Ribero D, Wang H et al (2007) Bevacizumab improves 66. Weber SM, Jarnagin WR et al (2000) Survival after
pathologic response and protects against hepatic resection of multiple hepatic colorectal metastases.
injury in patients treated with oxaliplatin-based che- Ann Surg Oncol 7(9):643–650
motherapy for colorectal liver metastases. Cancer 67. Wieser M, Sauerland S et al (2010) Peri-operative
110(12):2761–2767 chemotherapy for the treatment of resectable liver
54. Rubbia-Brandt L, Giostra E et al (2007) Importance metastases from colorectal cancer: a systematic review
of histological tumor response assessment in predict- and meta-analysis of randomized trials. BMC Cancer
ing the outcome in patients with colorectal liver 10(1):309
metastases treated with neo-adjuvant chemotherapy 68. Wong R, Cunningham D et al (2011) A multicentre
followed by liver surgery. Ann Oncol 18(2):299–304 study of capecitabine, oxaliplatin plus bevacizumab
55. Rudroff C, Altendorf-Hoffmann A et al (1999) as perioperative treatment of patients with poor-risk
Prospective randomised trial on adjuvant hepatic- colorectal liver-only metastases not selected for
artery infusion chemotherapy after R0 resection of upfront resection. Ann Oncol 22(9):2042–2048
colorectal liver metastases. Langenbecks Arch Surg 69. Yamaguchi T, Mori T et al (2008) A new classification
384(3):243–249 system for liver metastases from colorectal cancer in
56. Saltz LB, Clarke S et al (2008) Bevacizumab in combi- Japanese multicenter analysis. Hepatogastroenterology
nation with oxaliplatin-based chemotherapy as first-line 55(81):173–178
therapy in metastatic colorectal cancer: a randomized 70. Ychou M, Hohenberger W et al (2009) A randomized
phase III study. J Clin Oncol 26(12):2013–2019 phase III study comparing adjuvant 5-fluorouracil/
57. Saltz LB, Niedzwiecki D et al (2007) Irinotecan folinic acid with FOLFIRI in patients following com-
fluorouracil plus leucovorin is not superior to plete resection of liver metastases from colorectal
fluorouracil plus leucovorin alone as adjuvant treat- cancer. Ann Oncol 20(12):1964–1970
ment for stage III colon cancer: results of CALGB 71. Ychou M, Raoul JL et al (2009) A phase III ran-
89803. J Clin Oncol 25(23):3456–3461 domised trial of LV5FU2 + irinotecan versus LV5FU2
58. Scheele J, Stang R et al (1995) Resection of colorectal alone in adjuvant high-risk colon cancer (FNCLCC
liver metastases. World J Surg 19(1):59–71 Accord02/FFCD9802). Ann Oncol 20(4):674–680
59. Tomlinson JS, Jarnagin WR et al (2007) Actual
10-year survival after resection of colorectal liver
Will Adjuvant Chemotherapy
Improve Outcome After 22
Preoperative Chemoradiation?

Bengt Glimelius and Peter Nygren

Contents 22.1 Introduction


22.1 Introduction ................................................ 217
In colon cancer, there is high scientific evidence
22.2 Retrieval of Published Data ...................... 218
for significant gains in both disease-free survival
22.3 Summary of Published Data ..................... 218 (DFS) and overall survival (OS) from adjuvant
22.3.1 Adjuvant Chemotherapy Following biochemically modulated 5-fluorouracil (5-FU)
Surgery Alone .............................................. 218
22.3.2 Adjuvant Chemotherapy Following or capecitabine [15, 20, 44]. Large randomized
Surgery and Postoperative RT or CRT ......... 219 trials show that addition of oxaliplatin to 5-FU/
22.3.3 Adjuvant Chemotherapy Following leucovorin or capecitabine further improves DFS
Preoperative RT and Surgery ....................... 219 and possibly OS [4, 30, 35]. The relative reduc-
22.3.4 Adjuvant Chemotherapy Following
Preoperative CRT ......................................... 219 tion in recurrences by fluoropyrimidines is in the
22.3.5 Adjuvant Chemotherapy Before order of 30–35% and further 15–20% by the
and After CRT and Surgery ......................... 220 addition of oxaliplatin, totally resulting in a rela-
22.4 Discussion ................................................... 220 tive reduction of 40–45%. Based on these results,
adjuvant systemic chemotherapy is standard
References ................................................................. 223
treatment in stage III and in stage II if the risk of
recurrence is high.
As opposed to colon cancer, traditionally, many
recurrences have occurred loco-regionally in rectal
cancer, and although systemic recurrences were
also common, the loco-regional recurrences domi-
nated the clinical situation for many patients due
to their severe symptoms prior to death. With
improved surgery, based on the total mesorectal
B. Glimelius (*) excision (TME) concept and increasing use of
Department of Radiology, Oncology and Radiation preoperative radiotherapy (RT) and chemoradio-
Science, University of Uppsala, Akademiska sjukhuset, therapy (CRT), the risk of a local recurrence has
SE-751 85, Uppsala, Sweden
diminished substantially, from about 30–40%
Department of Oncology and Pathology, down to less than 10% considering the entire pop-
Karolinska Institutet, Stockholm, Sweden
ulation with a new rectal primary [42].
e-mail: bengt.glimelius@onkologi.uu.se
In rectal cancer, the scientific evidence for
P. Nygren
convincing gains in DFS and OS from adjuvant
Department of Radiology, Oncology and Radiation
Science, University of Uppsala, Akademiska sjukhuset, chemotherapy is much less than in colon cancer.
SE-751 85, Uppsala, Sweden The large adjuvant trials in this field have included

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 217


DOI 10.1007/978-3-642-25005-7_22, © Springer-Verlag Berlin Heidelberg 2012
218 B. Glimelius and P. Nygren

comparatively few patients with rectal cancer or 22.3 Summary of Published Data
have focused on colon cancer alone. Most patients
with rectal cancer receive loco-regional treatment The published data on the role of adjuvant chemo-
with RT or CRT, either after or, currently more therapy in rectal cancer derive from several
common, before surgery, whereas this is rarely decades during which the loco-regional treatment
done in colon cancer. This more complex treat- has undergone considerable changes. The data are
ment scenario has made the effect of systemic divided based on whether surgery was the only
adjuvant chemotherapy in rectal cancer more treatment or was accompanied by RT or CRT and
difficult to study, and the evolvement over time of whether such treatment was given before or after
the local treatment strategies has added further to the surgery. Very few studies about the value of
this complexity. adjuvant chemotherapy for rectal cancer have
The low level of scientific evidence for a been performed after preoperative CRT followed
benefit from adjuvant chemotherapy in rectal by TME, i.e. the focus of this review.
cancer is acknowledged in authoritative treatment
guidelines and by expert groups that still often
conclude that such treatment should be consid- 22.3.1 Adjuvant Chemotherapy
ered based on the principles used for colon can- Following Surgery Alone
cer [25, 40, 51]. In a recent systematic review, it
was concluded that adjuvant chemotherapy after This treatment setting, in most cases not rele-
surgery for rectal cancer following RT or CRT is vant today to the treatment of rectal cancer in
not ‘evidence-based’ [7]. In this review, where the Western world, except for the early stages,
the key findings from the pivotal clinical trials has mostly been studied in Japan, and the
performed in this field are summarized, we dis- fluoropyrimidine used was mostly uracil/tegafur
cuss how to interpret the current state of knowl- administered for 1–2 years, sometimes also
edge to be able to answer the question posed in together with mitomycin C. Several Japanese tri-
the title and related questions. als, including more than 5,000 patients, showed
statistically significant benefit from adjuvant che-
motherapy in DFS and/or OS with risk reductions
22.2 Retrieval of Published Data in the 15–40% range [31, 32, 45, 46]. Similar
observations were also observed in two western
Although the recent systematic review by Bujko world trials, including the QUASAR trial which
et al. concentrated on its role following CRT, the explored the value of adjuvant folinic-acid-
literature search covered all treatment settings modulated 5-FU in patients with colon cancer or
and was performed through August 2009 [7]. rectal cancer where the benefit of adjuvant chemo-
The current review is based on the literature therapy was considered uncertain [43].
retrieved by Bujko et al., updated with the addi- Of 3,239 patients in the QUASAR trial, 984
tional relevant data published from August 2009 had rectal cancer. In this group, a survival benefit
through May 2011. The PubMed and Cochrane from adjuvant chemotherapy was shown with a
databases were searched using the keywords 5-year OS of 78% compared with 74% in the
‘rectal cancer and adjuvant chemotherapy’. In control group (HR 0.77; p = 0.05). Subgroup
addition, abstracts and conference reports from analysis with patients divided into those with sur-
the scientific meetings of the American Society gery alone (n = 549) or pre- (n = 198) or postop-
of Clinical Oncology and the European Society erative (n = 201) RT was presented separately
of Medical Oncology and smaller focused meet- [27]. This analysis showed no significant hetero-
ings 2009–2011 relevant to the field were geneity between the groups, which separately
retrieved. We limited our report to data from tri- were too small to show statistically significant
als in which treatment allocation was randomized differences, but all showed point estimates indi-
and to patients operated on with curative intent. cating benefit from adjuvant chemotherapy.
22 Will Adjuvant Chemotherapy Improve Outcome After Preoperative Chemoradiation? 219

A joint analysis of Nordic trials showed no for DFS 0.87 (0.72–1.04). However, in patients
statistically significant benefit in OS at 5 years in given adjuvant chemotherapy, the local recur-
stage II/III rectal cancer (n = 691) from 4 to rence rate was reduced from 17% to 9%
12 months of various types of 5-FU-based che- (p = 0.002). OS and DFS were not reported sep-
motherapy [26]. A separate analysis of the arately for the RT and CRT groups. In the
Norwegian study where 5-FU + levamisole was QUASAR trial, 198 patients had preoperative
used for 12 months did similarly not reveal any RT and were then randomized to adjuvant che-
benefit in rectal cancer after surgery alone [13]. motherapy or not [27, 43]. In this subgroup,
An OS gain was seen in colon cancer stage III. In there was an obvious trend to benefit from adju-
addition, smaller studies showed no benefit in vant chemotherapy with hazard ratios for OS
DFS or OS [24, 28, 29]. and DFS close to 0.5. These differences were
not statistically significant due to the small
sample size.
22.3.2 Adjuvant Chemotherapy Early interim data from the ongoing PROCTOR/
Following Surgery and SCRIPT trial in which patients following short-
Postoperative RT or CRT course RT and immediate surgery are randomized
to 6 months folinic-acid-modulated 5-FU (n = 177)
Four moderately sized trials in this treatment set- or capecitabine (n = 183) or observation only and
ting showed no benefit in DFS or OS [9, 22, 37, joined with data (n = 107) from the Chronicle trial,
49]. In these trials, the adjuvant chemotherapy with randomization after preoperative CRT to
was 5-FU for 6–12 months. This treatment set- observation or adjuvant capecitabine and oxalipl-
ting was also part of the QUASAR trial (see atin, have been presented at a conference [53].
above), with 201 patients randomized [27, 43]. Based on these 470 patients, there were no signs
The hazard ratios indicated benefit from adjuvant of benefit in OS but numerically a minor advan-
chemotherapy for OS and DFS in this subgroup tage in terms of local recurrence and rate of dis-
of patients, although the differences were not sta- tance metastasis. The follow-up time of the
tistically significant. patients was not presented, but likely short since
some patients were recently included. The
PROCTOR/SCRIPT is still ongoing and has been
22.3.3 Adjuvant Chemotherapy expanded to also include patients having preop-
Following Preoperative erative long-course CRT.
RT and Surgery

This treatment setting was investigated as part 22.3.4 Adjuvant Chemotherapy


of the 2 × 2 factorially designed EORTC 22921 Following Preoperative CRT
trial, in a subgroup of patients in the QUASAR
trial and preliminary data have also been pre- This setting was investigated as part of the
sented from three trials still ongoing or finished EORTC 22921 trial as presented above, and the
early. In the EORTC 22921 trial, a total of 1,011 addition of adjuvant chemotherapy to preopera-
patients were randomized to preoperative RT or tive CRT had no effect on local recurrence [6]. In
CRT and then to postoperative adjuvant chemo- a similar Italian trial, still only reported as an
therapy or observation only [6]. The adjuvant abstract [11], 635 patients had preoperative CRT
chemotherapy was 5-FU, but only for 3 months. and were then randomized to 4.5 months of adju-
Adjuvant chemotherapy tended to provide vant folinic-acid-modulated 5-FU or observation
benefit in DFS and OS following preoperative alone. OS was essentially identical, and there was
RT or CRT, but the differences were not statisti- also no difference in local recurrence rate. The
cally significant, and hazard ratio for OS was results of the small Chronicle trial (see above)
0.85 (95% confidence interval 0.68–1.04) and have not been separately reported.
220 B. Glimelius and P. Nygren

22.3.5 Adjuvant Chemotherapy Before are positive. The overall data for this setting
and After CRT and Surgery support the notion that rectal cancer, in corre-
spondence with colon cancer, is possible to tar-
This setting could be considered experimental get with adjuvant chemotherapy. From a clinical
and is currently being studied, and no mature data practice point of view, however, this has limited
are available. Phase 2 trial data show high relevance since probably very few patients today
response rates in the rectal cancer primary from with more advanced rectal cancer go to surgery
chemotherapy with 5-FU/capecitabine and oxali- without preoperative (C)RT.
platin and better tolerance and compliance from In addition to being interesting from a princi-
adjuvant chemotherapy starting before CRT and ple point of view, this knowledge is of use in a
surgery vs. CRT, surgery and adjuvant chemo- situation in which a patient has been operated
therapy [10, 18]. without prior (C)RT based on favourable clinical
More relevant to the current issue of adjuvant and radiology findings at staging, but with unex-
effects is the randomized phase 2 Expert-C trial pected high-risk features in the histopathological
(n = 164) in which all patients with high-risk report. In this situation, adjuvant chemotherapy
operable RC were treated with 3 months of may be reasonable to consider as an alternative to
capecitabine combined with oxaliplatin (Capox), postoperative CRT since adjuvant chemotherapy
then had CRT with capecitabine followed by may provide both improved local and systemic
surgery and finally three more months of postop- control. If the high-risk features indicate a com-
erative Capox. The patients were randomized to parably high risk of local failure (e.g. crm+), it
addition of the EGFR antibody cetuximab to may be difficult for many to accept removal of
the pre- and postoperative chemotherapy and the the (C)RT component used for decades. If the
CRT or not. Efficacy data were presented at the features rather indicate high risk of failing sys-
2011 ASCO Conference [17]. Interestingly, in temically (e.g. extramural vascular invasion with
addition to significantly higher local tumour clear crm), adjuvant chemotherapy only maybe
response, addition of cetuximab also provided a considered acceptable. It is acknowledged,
statistically significant benefit in OS (hazard ratio though, that these data are not strong and are also
0.27; p = 0.035) and a trend towards better DFS unexpected from a pharmacological point of
(hazard ratio 0.81; p = 0.668) in the subset of view. A beneficial effect from adjuvant chemo-
patients with KRAS wild-type tumours. therapy on local recurrences is also supported by
population-based data [36] and in an analysis of
data from five European clinical trials [52].
22.4 Discussion The overall picture gets complicated when
(neo)adjuvant (C)RT has been used. For such
The level of scientific evidence in favour of therapy given postoperatively, most data show
benefit from adjuvant chemotherapy in rectal that addition of adjuvant chemotherapy pro-
cancer after curative surgery is considerably vides no major benefit, notably with the data
weaker than that in colon cancer. Not only are the from the QUASAR trial as an exception.
overall efficacy data far from convincing but they However, in practice, this situation is less rele-
often derive from old trials using adjuvant che- vant to discuss since there is now little support
motherapy schedules not in current use, and little for (C)RT after surgery [40, 51]. Most data
data are from treatment settings in which optimal indicate that when preoperative (C)RT has been
surgery and preoperative short-course RT or CRT given, the benefit from adding adjuvant chemo-
have been applied. therapy is likely to be very small or absent,
Adding 5-FU-based adjuvant chemotherapy again with the QUASAR trial as an exception.
to surgery alone seems to provide meaningful Although the Chronicle/PROCTOR/SCRIPT
benefit in terms of OS and DFS and perhaps also data are early and based on an interim analysis
local recurrence rate, although not all study data only, the lack of indication of a benefit from
22 Will Adjuvant Chemotherapy Improve Outcome After Preoperative Chemoradiation? 221

adjuvant chemotherapy is troublesome, given may be important in putative tumour ‘stem cells’,
the use in these trials of up-to-date neoadjuvant responsible for establishment and growth of
(C)RT, high-quality surgery and adjuvant che- metastasis. Furthermore, the pattern of metastatic
motherapy of current standard (5FU/leucovorin, spread differs between colon cancer and rectal
capecitabine or capecitabine/oxaliplatin). cancer, and theoretically, ‘stem cells’ deposited
By comparison with the efficacy of adjuvant in e.g., the lungs may be less drug sensitive than
chemotherapy in colon cancer and in rectal can- those in the liver or lymph nodes due to differ-
cer when adjunctive (C)RT has not been given, it ences in the interaction between tumour cells and
might be concluded that by adding (C)RT, which stroma [33, 38]. However, the finding of an adju-
has an obvious effect on small tumour deposits as vant effect in rectal cancer following surgery only
indicated by reduced local recurrence rate from does not support this possibility.
its addition to surgery, the effect from adjuvant Yet, another explanation to the small or absent
chemotherapy is in some way reduced. In the effect from adjuvant chemotherapy in rectal can-
case of CRT, the chemotherapy added to RT cer treated with preoperative (C)RT would be tim-
could theoretically have an adjuvant systemic ing of adjuvant chemotherapy, i.e. that adjuvant
effect although this is unlikely considering the chemotherapy is starting later than in colon can-
lack of benefit in OS and DFS from preoperative cer due to the time spent on the (C)RT and waiting
CRT compared with RT alone [6, 8] and the low for surgery and/or postoperative complications
total systemic exposure to chemotherapy in this leading to postponed start of adjuvant chemother-
setting compared with conventional adjuvant apy compared with the situation in colon cancer.
chemotherapy. This might be relevant since in colon cancer, the
Theoretically, the finding of an attenuated efficacy of adjuvant chemotherapy is attenuated
effect from adjuvant chemotherapy when given by time of start after surgery [5, 16]. However, the
after (C)RT could be explained if the untreated indication of a lack of effect from adjuvant che-
primary rectal cancer is an important source of motherapy also in the PROCTOR/SCRIPT study,
tumour cells that preoperatively spread and later in which the delay to adjuvant chemotherapy was
developed into metastases. However, in that case, likely small, argues against a major effect from
(C)RT should provide some benefit over no (C) timing. Another and related explanation could be
RT in terms of OS and development of metasta- the mostly lower compliance with the adjuvant
sis, which has not been observed, unless short- chemotherapy in rectal cancer compared with
course RT was given in the pre-TME era [21, 48]. colon cancer. In this context, presentation of study
On the other hand, the very complex view of the data both based on intention to treat and per pro-
primary tumour, metastasis and microenviron- tocol would be valuable. Although Swedish popu-
ment ecology emerging [38] may open for yet lation data show that OS after short-course RT,
unknown distant interactions between the pri- TME and adjuvant chemotherapy is significantly
mary and the metastases that speculatively could better than if adjuvant chemotherapy is not given
depend on the local treatment. [50], patient selection could have influenced the
Another possibility is a lower sensitivity of outcome since treatment allocation was not by
rectal cancer compared with colon cancer in the randomization.
adjuvant setting based on differences in tumour The relevance of the timing effect could be
biology. Similar activity of chemotherapy in approached by starting with the systemic rather
terms of tumour response rates and benefit in PFS than the local treatment in rectal cancer. The
and OS as well as the spectrum of drug activity in apparently favourable findings in the Expert-C
the metastatic setting argue against this possibil- trials indicate that this approach may be
ity. However, colon cancer differs from rectal beneficial [10, 17]. This approach seems fea-
cancer in several aspects relevant to tumour biol- sible and well tolerated [10, 18] and will be
ogy [1, 23, 39, 47], and although such differences investigated in the Nordic/Dutch RAPIDO
may not materialize in the metastatic setting, they trial, in which patients with poor prognosis
222 B. Glimelius and P. Nygren

rectal cancer will be randomized to conven- cancer are regarded as one disease entity essen-
tional CRT followed by surgery and optional tially separated only by an anatomical border.
adjuvant chemotherapy or short-course RT fol- Consequently, the role of adjuvant chemotherapy
lowed immediately by neoadjuvant chemother- in rectal cancer following (C)RT and surgery is
apy and finally surgery. conveniently and pragmatically extrapolated
Subgroup analyses of the EORTC 22921 trial from colon cancer and from the old, mainly US
have indicated that patients with tumour down- studies in rectal cancer where adjuvant chemo-
staging to pT0–2 compared with those with therapy with adjuvant chemoradiotherapy was
tumours remaining as pT3–4 following (C)RT had tested [41], without too much consideration of
better prognosis but they also showed benefit the evidence basis for this approach. The NCCN
from adjuvant chemotherapy [12]. However, this guidelines clearly state the adjuvant chemother-
effect from adjuvant chemotherapy seemed apy should be given to those at risk for recurrence
restricted to preoperative RT but not to CRT and [40]. In a recent exploration of the use of adju-
was, surprisingly, not related to the nodal status vant chemotherapy in a large US population
after treatment. These findings contrast to other registry, suboptimal use was noticed. It was con-
subgroup analysis on this issue [19] and the gen- cluded that even at specialty cancer centres, a
eral observations of better effect from adjuvant sizeable minority of rectal cancer patients treated
chemotherapy in colon cancer in more advanced with curative intent neoadjuvant CRT do not
disease [4, 35]. Additional methodological prob- complete postoperative chemotherapy. Strategies
lems with the EORTC trial subgroup analysis to foster adherence to the third and final compo-
were discussed by Bujko et al. [7]. Thus, although nent of curative intent treatment were considered
response to neoadjuvant (C)RT provides strong necessary [34]. However, for those with a more
prognostic information in rectal cancer, it cannot, puristic view on the scientific basis of cancer
based on current knowledge, be used to predict treatment, associated with considerable costs
the efficacy of adjuvant chemotherapy. both in terms of health-care funding and individ-
The notion that rectal cancer may successfully ual patient suffering from adverse effects, this
be targeted in the adjuvant setting, as shown in the approach is clearly disturbing.
early trials adding adjuvant chemotherapy to sur- In routine care, any treatment should be given
gery only, but that a substantial effect is somehow if it is proven that it is sufficiently effective and
lost or at least severely attenuated when combined that the negative effects and costs do not counter-
with preoperative (C)RT points to the use in adju- balance this. Since many patients with rectal can-
vant chemotherapy of drugs more active against cer recur, there is a need for adjuvant therapies
subclinical disease than 5-FU or its analogues that decrease the risk. After surgery alone, the
alone. The addition of oxaliplatin to 5-FU data indicate that adjuvant chemotherapy may
improves the adjuvant effect in colon cancer [4, have sufficient effects, but this is presently not
30, 35] and needs to be studied also in rectal can- the case after CRT. The answer to the question
cer. Unfortunately, effects in the metastatic situa- ‘will adjuvant therapy’ posed in the title is ‘well,
tion cannot be extrapolated to the adjuvant setting it should’. But, does it? No, we do not know.
as illustrated by the recent negative trial data on Should it be given? Basically not. Will it be
the VEGF and EGFR antibodies in colon cancer given? Well, in many, but not all health-care sys-
[2, 3, 14]. Thus, there is a need for preclinical tems it will, and even strongly recommended in
models of subclinical disease relevant to colon official guidelines. When the use was quality
and rectal cancer to optimize the selection of can- controlled, the doctors were almost ‘banned not
didate drugs to be included in clinical trials on the to have followed the guidelines’ [34]. In every
adjuvant effects in these tumour types. health-care system, doctors and patients must
In many, perhaps even most, centres treating weigh pros and cons in every clinical situation.
rectal cancer, adjuvant chemotherapy in this Considering the weak scientific evidence, with
tumour type is a non-issue; colon cancer and rectal comparably small gains in the positive trials and
22 Will Adjuvant Chemotherapy Improve Outcome After Preoperative Chemoradiation? 223

lack of signs of gain in other trials, it may actually adjuvant postoperative RT vs. postoperative RT plus
be very sound that not all patients get treatment, 5-FU and levamisole in patients with TNM stage II-III
resectable rectal cancer. J Surg Oncol 75:80–88
even if recommended. 10. Chua YJ, Barbachano Y, Cunningham D et al (2010)
The lack of convincing data showing a benefit Neoadjuvant capecitabine and oxaliplatin before
from adjuvant chemotherapy in rectal cancer chemoradiotherapy and total mesorectal excision in
argues for participation in well-designed clinical MRI-defined poor-risk rectal cancer: a phase 2 trial.
Lancet Oncol 11:241–248
trials in currently used treatment settings. 11. Cionini L, Manfredi B, Sainato A et al (2001)
Unfortunately, the trials ongoing in this field are Randomized study of postoperative chemotherapy
not designed to answer the fundamental question after preoperative chemoradiation in locally advanced
whether adjuvant chemotherapy provides any rectal cancer. Preliminary results. Eur J Cancer 37:
S300 (Abstr)
benefit over no adjuvant systemic treatment at all, 12. Collette L, Bosset JF, den Dulk M et al (2007) Patients
since no non-treatment control arms are included. with curative resection of cT3–4 rectal cancer after
In this light, the PROCTOR/SCRIPT trial is preoperative radiotherapy or radiochemotherapy: does
important to finalize. anybody benefit from adjuvant fluorouracil-based
chemotherapy? J Clin Oncol 25:4379–4386
13. Dahl O, Fluge O, Carlsen E et al (2009) Final results of
a randomised phase III study on adjuvant chemother-
References apy with 5 FU and levamisol in colon and rectum can-
cer stage II and III by the Norwegian Gastrointestinal
1. Aamodt R, Jonsdottir K, Andersen SN et al (2009) Cancer Group. Acta Oncol 48:368–376
Differences in protein expression and gene 14. De Gramont A, Van Cutsem E, Tabernero J et al
amplification of cyclins between colon and rectal ade- (2011) AVANT: results from a randomized three-arm
nocarcinomas. Gastroenterol Res Pract 2009:285830 multinational phase III study to investigate bevaci-
2. Alberts SR, Sargent DJ, Smyrk TC et al (2010) zumab with either XELOX or FOLFOX4 versus
Adjuvant mFOLFOX6 with or without cetuximab in FOLFOX alone as adjuvant treatment for colon can-
KRAS wild-type patients with resected stage III colon cer. J Clin Oncol 29(suppl 4):A 362
cancer: results from NCCTG intergroup phase III trial 15. de Gramont A, Hubbard J, Shi Q et al (2010)
N0147. J Clin Oncol 28:A3507 Association between disease-free survival and overall
3. Allegra CJ, Yothers G, O’Connell MJ et al (2011) survival when survival is prolonged after recurrence
Phase III trial assessing bevacizumab in stages II and in patients receiving cytotoxic adjuvant therapy for
III carcinoma of the colon: results of NSABP protocol colon cancer: simulations based on the 20,800 patient
C-08. J Clin Oncol 29:11–16 ACCENT data set. J Clin Oncol 28:460–465
4. Andre T, Boni C, Navarro M et al (2009) Improved 16. Des Guetz G, Nicolas P, Perret GY et al (2010) Does
overall survival with oxaliplatin, fluorouracil, and leu- delaying adjuvant chemotherapy after curative sur-
covorin as adjuvant treatment in stage II or III colon gery for colorectal cancer impair survival? A meta-
cancer in the MOSAIC trial. J Clin Oncol 27: analysis. Eur J Cancer 46:1049–1055
3109–3116 17. Dewdney A, Capdevila J, Glimelius B et al (2011)
5. Biagi JJ, Raphael M, King WD et al (2011) The Expert-C: a randomized phase II European multicenter
impact of time to adjuvant chemotherapy on survival trial of neoadjuvant chemotherapy (CAPOX) and
in colorectal cancer: a systematic review and meta- chemoradiation (CRT) with or without cetuximab fol-
analysis. J Clin Oncol 29(suppl 4):A364 lowed by total mesorectal excision (TME) in patients
6. Bosset JF, Collette L, Calais G et al (2006) with MRI-defined, high-risk rectal cancer. Proc Am
Chemotherapy with preoperative radiotherapy in rec- Soc Clin Oncol; J Clin Oncol 29:suppl abstract 3513
tal cancer. N Engl J Med 355:1114–1123 18. Fernandez-Martos C, Pericay C, Aparicio J et al
7. Bujko K, Glynne-Jones R, Bujko M (2010) Does (2010) Phase II, randomized study of concomitant
adjuvant fluoropyrimidine-based chemotherapy pro- chemoradiotherapy followed by surgery and adjuvant
vide a benefit for patients with resected rectal cancer capecitabine plus oxaliplatin (CAPOX) compared
who have already received neoadjuvant radio(chemo) with induction CAPOX followed by concomitant
therapy? A Systematic Review of Randomized Trials. chemoradiotherapy and surgery in magnetic reso-
Ann Oncol 21:1743–1750 nance imaging-defined, locally advanced rectal can-
8. Bujko K, Nowacki MP, Nasierowska-Guttmejer A cer: Grupo cancer de recto 3 study. J Clin Oncol
et al (2006) Long-term results of a randomised trial 28:859–865
comparing preoperative short-course radiotherapy vs 19. Fietkau R, Barten M, Klautke G et al (2006)
preoperative conventionally fractionated chemoradia- Postoperative chemotherapy may not be necessary for
tion for rectal cancer. Br J Surg 93:1215–1223 patients with ypN0-category after neoadjuvant chemo-
9. Cafiero F, Gipponi M, Peressini A et al (2000) radiotherapy of rectal cancer. Dis Colon Rectum
Preliminary analysis of a randomized clinical trial of 49:1284–1292
224 B. Glimelius and P. Nygren

20. Figueredo A, Coombes, ME, Mukherjee S (2008) Cancer Network (NCCN) analysis. J Clin Oncol
Adjuvant therapy for completely resected stage II 29:suppl; abstr 3515
colon cancer. Cochrane Database Syst Rev (3): 35. Kuebler JP, Wieand HS, O’Connell MJ et al (2007)
CD005390 Oxaliplatin combined with weekly bolus fluorouracil
21. Folkesson J, Birgisson H, Påhlman L et al (2005) and leucovorin as surgical adjuvant chemotherapy for
Swedish Rectal Cancer Trial: long lasting benefits stage II and III colon cancer: results from NSABP
from radiotherapy on survival and local recurrence C-07. J Clin Oncol 25:2198–2204
rate. J Clin Oncol 23:5644–5650 36. Kusters M, Valentini V, Calvo FA et al (2010) Results
22. Fountzilas G, Zisiadis A, Dafni U et al (1999) of European pooled analysis of IORT-containing mul-
Postoperative radiation and concomitant bolus timodality treatment for locally advanced rectal can-
fluorouracil with or without additional chemotherapy cer: adjuvant chemotherapy prevents local recurrence
with fluorouracil and high-dose leucovorin in patients rather than distant metastases. Ann Oncol
with high-risk rectal cancer: a randomized phase III 21:1279–1284
study conducted by the Hellenic Cooperative 37. Mansour EG, Lefkopoulou M, Johnson R et al (1991)
Oncology Group. Ann Oncol 10:671–676 A comparison of postoperative adjuvant chemother-
23. Fransen K, Klintenas M, Osterstrom A et al (2004) apy, radiotherapy or combination therapy in poten-
Mutation analysis of the BRAF, ARAF and RAF-1 tially curable resectable rectal carcinoma. An ECOG
genes in human colorectal adenocarcinomas. study Est 4276. Proc Am Soc Clin Oncol 10:154
Carcinogenesis 25:527–533 abstr
24. Gastrointestinal Tumor Study Group (1985) Prolongation 38. McAllister SS, Weinberg RA (2010) Tumor-host
of the disease-free interval in surgically treated rectal interactions: a far-reaching relationship. J Clin Oncol
carcinoma. N Engl J Med 312:1465–1472 28:4022–4028
25. Glimelius B (2010) Adjuvant chemotherapy in rectal 39. Minoo P, Zlobec I, Peterson M et al (2010)
cancer – an issue or a non-issue? Ann Oncol 21: Characterization of rectal, proximal and distal colon
1739–1741 cancers based on clinicopathological, molecular and
26. Glimelius B, Dahl O, Cedermark B et al (2005) protein profiles. Int J Oncol 37:707–718
Adjuvant chemotherapy in colorectal cancer: a joint 40. National Comprehensive Cancer Network (2011)
analysis of randomised trials by the Nordic Rectal cancer. NCCN clinical practice guidelines in
Gastrointestinal Tumour Adjuvant Therapy Group. oncology Version 3
Acta Oncol 44:904–912 41. NCI (1991) Clinical announcement. Adjuvant therapy
27. Gray R, McConkey C (2008) Adjuvant chemotherapy of rectal cancer. National Cancer Institute, Bethesda
for rectal cancer: authors’ reply. Lancet 371:1503 42. Pahlman L, Bohe M, Cedermark B et al (2007) The
28. Hafstrom L, Rudenstam CM, Domellof L (1985) A Swedish rectal cancer registry. Br J Surg 94:
randomized trial of oral 5-fluorouracil versus placebo 1285–1292
as adjuvant therapy in colorectal cancer Dukes’ B and 43. Quasar Collaborative Group, Gray R, Barnwell J et al
C: results after 5 years observation time. Br J Surg (2007) Adjuvant chemotherapy versus observation in
72:138–141 patients with colorectal cancer: a randomised study.
29. Hafström L, Domellöf L, Rudenstam CM et al (1990) Lancet 370:2020–2029
Adjuvant chemotherapy with 5-fluorouracil, vincris- 44. Ragnhammar P, Brorsson B, Nygren P et al (2001) A
tine and CCNU for patients with Dukes’ C colorectal prospective study of the use of chemotherapy in
cancer. The Swedish Gastrointestinal Tumour Sweden and assessment of the use in relation to
Adjuvant Therapy Group. Br J Surg 77:1345–1348 scientific evidence. Acta Oncol 40:391–411
30. Haller DG, Tabernero J, Maroun J et al (2011) 45. Sakamoto J, Hamada C, Kodaira S et al (1999)
Capecitabine plus oxaliplatin compared with Adjuvant therapy with oral fluoropyrimidines as main
fluorouracil and folinic acid as adjuvant therapy for chemotherapeutic agents after curative resection for
stage III colon cancer. J Clin Oncol 29:1465–1471 colorectal cancer: individual patient data meta-analy-
31. Hamaguchi T, Shirao K, Moriya Y et al (2011) Final sis of randomized trials. Jpn J Clin Oncol 29:78–86
results of randomized trials by the national surgical 46. Sakamoto J, Hamada C, Yoshida S et al (2007) An
adjuvant study of colorectal cancer (NSAS-CC). individual patient data meta-analysis of adjuvant ther-
Cancer Chemother Pharmacol 67:587–596 apy with uracil-tegafur (UFT) in patients with cura-
32. Hojo K, Kajitani T (1986) Adjuvant chemotherapy of tively resected rectal cancer. Br J Cancer
colorectal cancer – results of prospective randomized 96:1170–1177
trials. Gan To Kagaku Ryoho 13:3063–3073 47. Slattery ML, Wolff E, Hoffman MD et al (2011)
33. Joyce JA, Pollard JW (2009) Microenvironmental MicroRNAs and colon and rectal cancer: differential
regulation of metastasis. Nat Rev Cancer 9:239–252 expression by tumor location and subtype. Genes
34. Khrizman P, Niland JC, ter Veer A et al (2011) Chromosomes Cancer 50:196–206
Postoperative adjuvant chemotherapy (CTX) use in 48. Swedish Rectal Cancer Trial (1997) Improved sur-
patients (Pts) with stage II/III rectal cancer treated vival with preoperative radiotherapy in resectable rec-
with neoadjuvant therapy: a National Comprehensive tal cancer. N Engl J Med 336:980–987
22 Will Adjuvant Chemotherapy Improve Outcome After Preoperative Chemoradiation? 225

49. Taal BG, Van Tinteren H, Zoetmulder FA (2001) 52. Valentini V, van Stiphout RG, Lammering G et al
Adjuvant 5FU plus levamisole in colonic or rectal (2011) Nomograms for predicting local recurrence,
cancer: improved survival in stage II and III. Br J distant metastases, and overall survival for patients
Cancer 85:1437–1443 with locally advanced rectal cancer on the basis of
50. Tiefenthal M, Nilsson PJ, Johansson R et al (2011) European randomized clinical trials. J Clin Oncol
The effects of short-course preoperative irradiation on 29:3163–3172
local recurrence rate and survival in rectal cancer: a 53. van de Velde CJH (2010) Chronicle/PROCTOR/
population-based nationwide study. Dis Colon Rectum SCRIPT trials. European Multidisciplinary Colorectal
54:672–680 Cancer Congress, Nice, 28–30 Mar, 2010, Oral
51. Valentini V, Glimelius B (2010) Rectal cancer radio- presentation
therapy: towards European consensus. Acta Oncol
49:1206–1216
Part VI
Q&As on Surgery
How to Evaluate the Quality
of Surgery? Suggestions 23
for Critical Reading of Surgical
and Pathological Reports

Lars Påhlman

Contents 23.1 Introduction


23.1 Introduction ................................................ 229
The differences in outcome between surgeons are
23.2 Volume ........................................................ 230
sometimes great and cannot only be explained by
23.3 The Pathologist’s Evaluation stage of disease and patients’ co-morbidity but
of Surgical Quality ..................................... 230
merely of the fact that the quality in the surgical
23.4 How to Access Quality of Surgery? .......... 230 technique differs among surgeons [1].
References ................................................................. 231 In rectal cancer, this phenomenon has been
discussed extensively. In the early 1980s, when
preoperative radiotherapy was introduced to
reduce the often unacceptable high local recur-
rence rate, some centres reported results regard-
ing local recurrences better than standard surgery
combined with radiotherapy [2–4]. All these
reports pinpointed the importance of good surgi-
cal quality, and the TME technique (total mesorec-
tal excision), i.e., dissection in the embryological
plane, should be the gold standard [2]. As a con-
sequence, several countries started training pro-
grammes, and in Scandinavia, not only the surgical
technique changed but also a concentration to
fewer centres has been carried out [5–7].
With a careful examination of the specimen, it
is possible for the surgeon but also by the pathol-
ogists to grade the quality of surgery. If the sur-
geon has followed the embryological plane,
which has been shown to be a barrier for local
spread, no tears in the mesorectal fascia can be
seen.
L. Påhlman This chapter will describe the importance of
Department of Surgical Sciences,
surgical standardization and quality registration
Uppsala University,
SE 751 85, Uppsala, Sweden but also a structured evaluation of the specimen
e-mail: lars.pahlman@surgsci.uu.se by the pathologists.

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 229


DOI 10.1007/978-3-642-25005-7_23, © Springer-Verlag Berlin Heidelberg 2012
230 L. Påhlman

23.2 Volume When applying such a grading on the whole


material in the MRC CR07 trial, on radiotherapy
One important question has been raised in the in rectal cancer, the quality of the specimen pre-
surgical community and also among health-care dict not only local recurrence rate but also sur-
providers and that is the volume for units or indi- vival [9]. For a more specific way of treating the
vidual surgeons. Rectal cancer is a rather com- specimen, see the chapter by Nigel Scott.
mon disease with an incidence in Europe of Therefore, although not yet recommended, all
approximately 15–20 new cases per 100,000 pathological reports should ideally have photo
inhabitants. Therefore, a unit taking care of documentation of the specimen but also a judge-
500,000 up to 1 million inhabitants will have ment of the quality of the specimen. Based upon
around 100 rectal cancers a year, which is prob- this grading, a constructive feedback can be given
ably a sufficient volume to require good knowl- to the surgeon at the postoperative MDT confer-
edge and treatment in all level, including surgery, ence. Moreover, by combining this grading with
nursing, follow-up, etc. This is, however, not the the preoperative MRI stage, important end point
case in the majority of countries in Europe. At for a positive outcome, like the circumferential
several hospitals, not more than 10–20 rectal resection margin, can be better evaluated.
cancers are treated by more than one surgeon
giving the numbers rather small. There are data
from literature supporting that volume is impor- 23.4 How to Access Quality
tant, but it is difficult to interpret the literature, of Surgery?
mainly due to so many confounding factors like
co-morbidity, stage of disease, social depriva- The shape and quality of the specimen is of
tion, etc. [8]. utmost importance and can be evaluated by the
pathologist as described above. However, there
are several other parameters that will differ and
23.3 The Pathologist’s Evaluation also are indications on differences in quality.
of Surgical Quality Based upon data emanated from quality assur-
ance registries in the Scandinavian countries
The pathologist has the ultimate position to eval- during the last 15 years, it is possible to evalu-
uate the surgical quality. If the specimens are sent ate the ‘overall’ outcome. Those registries are
fresh to the pathology laboratory, it is possible to national based with participation from all hos-
grade the quality of the specimen but also have pitals. With more or less transparent reports
photo documentation in the files. Different termi- and honest discussions among surgeons, the
nology has been proposed to classify the speci- improvement has been dramatic in the coun-
men where three groups have been proposed tries involved [5–7].
including a perfect specimen, rather well and a Data collected in these registries are surgical
poor specimen. Professor Quirke, a pathologist procedure, level of vessel ligation, type of recon-
from Leeds, has proposed that the three planes struction and outcome in terms of complications
should be classified as follows [9]: to surgery but also cancer-related end points like
(a) Mesorectal plane, indicating that the whole local control and survival. Moreover, the diag-
mesorectal fascia is intact nostic process preoperatively is registered
(b) Intra-mesorectal plane, with minor tears in together with the follow-up routines and the use
the mesorectal fascia of neo-adjuvant and/or adjuvant use of chemo-
(c) Muscularis propria plane, which is a speci- therapy and radiotherapy. In some, registries also
men where lots of mesorectum is left behind register co-morbidity and health-related parame-
and the muscle tube of the bowel can be seen ters like smoking, BMI, etc. Also, prospective
macroscopically registration of the preoperative tumour stage
23 How to Evaluate the Quality of Surgery? 231

based upon imaging as well as postoperative risk of damage of the nerves should be minimal.
tumour stage based upon pathological examina- However, if the tumour is growing outside the
tion exists. normal plane of dissection, the risk of nerve dam-
The registries have annual reports with data age is obvious. By registering such outcome
divided upon hospitals but also showing chang- together with the knowledge of tumour stage, it is
ing’s and trends in each country. Most of the reg- possible to increase the quality.
isters have been enlarged based upon the need for
new items to be register and studied. Once a new Conclusion
parameter is involved in the registration, i.e., Quality in surgery has become an important
lymph node retrieval, the first year registration topic in rectal cancer treatment. Lots of data
often shows unaccepted differences between support that provided surgery is done in an
units, but with transparent reports and discus- optimal way, one can reduce the use of radio-
sions among the surgical communities in the therapy and perhaps also reduce the use of
country, the next years’ registration will often chemotherapy. To have an immediate feed-
show an increase to the better, and within some back to the surgeon whether or not the surgi-
years it reaches acceptable levels. Subsequently, cal procedure has been done in an optimal
a quality register not only guarantees good assur- way, photo documentation of the specimen is
ance that the level of care is acceptable but also essential and grading by the pathologist of the
acts as a vehicle for quick introduction of new macroscopic view of the specimen is also
treatment standards. crucial. With a quality assurance programme,
The experience from the Scandinavian countries the whole treatment of rectal cancer can be
has led to the same project in many European coun- evaluated.
tries. In an effort to have similar programmes,
ECCO has sponsored together with European
Society of Surgical Oncology (ESSO) and Euro-
pean Society of Coloproctology (ESCP) a collabo- References
ration with all countries with quality registration,
the EURECCA project (European Registration of 1. Mc Ardle CS, Hole D (1901) Impact of variability
among surgeons on postoperative morbidity and mor-
Cancer Care) [10]. Countries involved so far, as
tality and ultimate survival. BMJ 302:1501–1505
national registries or regional, are Belgium, 2. Heald RJ, Husband EM, Ryall RDH (1982) The
Denmark, Germany, Italy, the Netherlands, Norway, mesorectum in rectal cancer surgery: the clue to pel-
Spain, Sweden and the UK. vic recurrence. Br J Surg 69:613–616
3. Enker WE, Thaler HT, Cranor ML, Polyak T (1995)
Other important end points to be evaluated are
Total mesorectal excision in the operative treatment of
specific complications which can be results of carcinoma of the rectum. J Am Coll Surg 18:
non-optimal surgical technique. Well-known 335–346
complications or merely dysfunctions are sexual 4. Moriya Y, Hojo K, Sawada T, Koyama Y (1989)
Significance of lateral lymph node dissection for
impairment, urinary dysfunctions and bowel
advanced rectal carcinoma at or below the peritoneal
problems [11, 12]. In all, this is a whole spectrum reflection. Dis Colon Rectum 32:307–315
of quality of life. To add this in a quality registra- 5. Wibe A, Møller B, Norstein J, Carlsen E, Wiig JN,
tion, prospective validated questionnaires have to Heald RJ, Langmark F, Myrvold HE, Søreide O,
Norwegian Rectal Cancer Group (2002) A national
be used.
strategic change in treatment policy for rectal cancer –
Regarding the sexual problems and also uri- implementation of total mesorectal excision as routine
nary impairments, modern more precise surgery treatment in Norway. A national audit. Dis Colon
will prevent such complications in the majority of Rectum 45:857–866
6. Påhlman L, Bohe M, Cedermark B, Dahlberg M,
the cases. Today, with good preoperative staging,
Lindmark G, Sjödahl R, Öjerskog B, Damber L,
surgeons should know if the normal plane of dis- Johansson R (2007) The Swedish rectal cancer regis-
section could be followed. If that is the case, the try. Br J Surg 94:1285–1292
232 L. Påhlman

7. Bülow S, Harling H, Iversen LH, Ladelund S, Danish CR07 and NCIC-CTGCO16 randomised clinical trial.
Colorectal Cancer Group (2009) Survival after rectal Lancet 373:821–828
cancer has improved considerably in Denmark. Ugeskr 10. van Gijn W, van de Velde CJH, and on behalf of the mem-
Laeger 171:2735–2738 bers of the EURECCA Consortium (2010) Improving
8. Kressner M, Bohe M, Cedermark B, Dahlberg M, quality of cancer care through surgical audit. Eur J Surg
Damber L, Lindmark G, Öjerskog B, Sjödahl R, Oncol 36:S23–S26
Johansson R, Påhlman L (2009) The impact of hospi- 11. Dahlberg M, Glimelius B, Graf W, Påhlman L (1998)
tal volume on surgical outcome for rectal cancer – a Preoperative irradiation for rectal cancer affects the
survey of the Swedish Rectal Cancer Register. Dis functional results after colorectal anastomosis –
Colon Rectum 52:1542–1549 results from the Swedish Rectal Cancer Trial. Dis
9. Quirke P, Steele R, Monson J, Grieve R, Khanna S, Colon Rectum 41:543–551
O’Callghan C, Sun Myint A, Bessell E, Thompson 12. Marijnen CA, van de Velde CJ, Putter H et al (2005)
LC, Parmar M, Stephens RJ, Sebag-Montefiore D Impact of short-term preoperative radiotherapy on
(2009) Effect of the plane of surgery achieved on local health-related quality of life and sexual functioning in
recurrence in patients operated with operable rectal primary recta caner: report of a multicentre trial.
cancer: a prospective study using data from the MRC J Clin Oncol 23:1847–1858
How Is Nerve-Sparing Surgery
Well Performed? 24
Zoran Krivokapic and Ivan Dimitrijevic

Contents 24.1 Introduction


24.1 Introduction ................................................ 233
Major goals of rectal cancer treatment are
24.2 The Importance of Pelvic
Vegetative Nerve Preservation .................. 233 improving of overall survival rates by minimiz-
ing the percentage of local and distant recurrence.
24.3 Physiological Considerations .................... 235
Efforts are also made in attempt to further reduce
24.4 Anatomical Consideration in the Context the percentage of permanent stomas as well as to
of Function Preservation ........................... 235 improve the life quality of patients treated for this
24.4.1 Pelvic Vegetative Nerve Structures .............. 235
24.4.2 Anatomy of Denonvilliers’ Fascia ............... 237 disease. Until around 20 years ago, there was a
24.4.3 The ‘Lateral Ligaments’ of the Rectum ....... 237 lot of space for improvement of surgical results.
24.4.4 Pudendal and Levator Ani Nerve ................. 238 The percentage of permanent stomas in around
24.5 Important Points of Nerve 55%, 5-year survival of around 50%, local recur-
Preserving TME ......................................... 238 rence in 30–40%, and distant recurrence in more
24.5.1 Mobilization of the Left Colon than 60% made situation with rectal cancer treat-
and Ligation of the Inferior
Mesenteric Artery ........................................ 238
ment very upsetting. After the introduction of
24.5.2 Division of the Sigmoid Colon .................... 239 total mesorectal excision (TME) in 1982, by Bill
24.5.3 High Posterior and Lateral Dissection ......... 239 Heald, significant improvement was noted in all
24.5.4 Distal Posterior and Lateral Dissection ....... 240 fields of the surgical treatment of rectal cancer.
24.5.5 Anterior Dissection ...................................... 241
24.5.6 Mobilization of the Most Caudal
Five-year local recurrence rate was reduced to
Portion of the Mesorectum .......................... 243 only 5%, and 5-year overall survival was 80%,
24.5.7 Laparoscopic TME in the Context of Nerve according to Mr. Heald’s results [18].
Identification and Preservation .................... 245
References ................................................................. 245
24.2 The Importance of Pelvic
Vegetative Nerve Preservation

Having in mind the importance of life quality of


patients operated for rectal cancer, systematic
identification and preservation of pelvic vegeta-
Z. Krivokapic • I. Dimitrijevic (*) tive nerves was applied in wide surgical practice
Colorectal Department, Institute for Digestive Diseases, simultaneously with the introduction of TME.
Medical School of Belgrade University,
High percentage of postoperative sexual and uri-
First Surgical Clinic, Clinical Center of Serbia,
Belgrade, Serbia nary dysfunctions was well-known phenomenon
e-mail: scpy@beotel.yu in the early years of modern rectal cancer surgery.

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 233


DOI 10.1007/978-3-642-25005-7_24, © Springer-Verlag Berlin Heidelberg 2012
234 Z. Krivokapic and I. Dimitrijevic

Goligher, Bernstein and Weinstein [2, 63, 67] with sphincter-saving procedures [13, 27, 31,
published the papers where significant percent- 41, 59, 66]. These results are in a way obvious
age of patients operated for rectal cancer had since the abdominoperineal resection (APR) is
sexual and urinary dysfunction. In one meta- more mutilant in its nature, and presence of a
analysis that dealt with quality of life of patients stoma has a significant psychological influence
operated for rectal cancer in the second half of on these individuals. Mr. Heald stressed that
the twentieth century, sexual dysfunction was injury to the neurovascular bundle of Walsh is
observed in 39% and urinary dysfunction likely to happen during the perineal act due to
occurred in 27% of patients [4]. Fortunately, with poor visibility and the fact that statistically more
the introduction and popularization of the TME, advanced tumours are found in this group of
the things changed for better [16]. Besides men- patients [15].
tioned oncological benefits, identification and As it is clearly apparent, the sex of patient
preservation of vegetative nerves has become significantly affects the percentage of nerve
easier [18]. By abiding the main principles of identification and preservation as well as patients’
TME technique, i.e. sharp dissection under the constitution (obese individuals, narrow ‘male’
direct control of vision, we have almost no bleed- pelvis) [56]. Neo-adjuvant therapy can also be a
ing, and by following the correct dissection plane, factor that influences nerve identification and
there is minimal risk of serious vegetative nerve preservation. Nevertheless, literature data are
damage. Additionally, we obtain adequate resec- inconsistent due to different inclusion criteria,
tion margins, especially important circumferen- and research methods and this subject deserve to
tial resection margin, good specimen and ability be further investigated [27, 32]. Tumour stage
to perform high percentage of sphincter-preserv- and localization as well as operation type also
ing operations. have significant influence on the degree of
Sympathetic vegetative nerve structures are identification and preservation of vegetative
generally situated more proximally and are easier nerves [27, 31]. The factor that significantly
to identify and preserve. According to literature affects nerve preservation and identification is
data, by conducting proper TME, identification surgeons experience and training in this field.
of these structures is possible in 80–94% [13] Learning curve showed to be independent factor
in males and in even 96% [21] in females. The that predisposed the degree of nerve preservation
situation with identification and preservation of and identification [27].
parasympathetic structures differs. They are Nevertheless, the good postoperative results
placed deep in the pelvis, in delicate fascial lay- published by highly specialized rectal surgeons
ers of closely situated pelvic organs and are much have not been achieved in larger studies [38, 43].
more difficult to identify and preserve. There is Concerning sexual function, age showed to be
a lot of discrepancy in literature data on this a very, if not the most, important predictive fac-
subject. The percentage of preservation and tor after rectal cancer operations [19, 29, 59].
identification of these structures varies between Patients over 60 years of age have a significantly
53% and 96% [21, 39, 44]. higher risk of loss of sexual function postopera-
In almost all studies published on this sub- tively than younger patients [13]. The difference
ject, the operation type proved to be a significant in the upper age limit between a number of stud-
factor of influence. In cases where surgeons pre- ies can explain various rates of sexual dysfunc-
formed the transection of mesorectum, situation tion reported.
was very good as presented by some [42]. On Some investigators, in order to further
the other hand, when performing more challeng- improve and facilitate the nerve preservation,
ing procedure, TME, the results tend to be attempted to employ neurostimulation and mea-
worse. Unfavourable functional results were surement of penile tumescence or intravesical
noted in patients where abdominoperineal resec- pressure. Using these methods, the percentage
tion (APR) was performed, compared to those of identification of pelvic vegetative nerves rises
24 How Is Nerve-Sparing Surgery Well Performed? 235

from standard 70–80% to almost 100% [7, 58]. engorgement of the clitoris and labia. Disruption
The initial results are promising but deserve fur- of the autonomic nerves in females may cause
ther research. dyspareunia and vaginal dryness. Reviewing the
Concerning all postoperative functional deficits literature, we see that postoperative sexual dys-
and recovery rates in the follow-up period, we can function is present in the range between 3% and
conclude that if preoperative function is intact, the 57% [19, 49]. Dyspareunia is the most common
likelihood of complete recovery is fair. symptom after rectal cancer surgery and some-
times even before the operation [13]. Disturbances
of the pelvic anatomy can explain some of the
24.3 Physiological Considerations female sexual problems. APR results in higher
incidence of dyspareunia and is almost always
The sympathetic and parasympathetic nervous present if the posterior wall of the vagina is
systems act as an integral system in the coordina- removed.
tion of urinary and sexual function. Sacral para- Urinary bladder is also innervated with both
sympathetic fibres carry the impulses that mediate components of the autonomic system. The base
dilatation of the vascular inflow to the cavernous of the bladder is supplied by sympathetic
bodies which results in erection. The neuronally impulses, and the detrusor muscle is innervated
triggered vasodilatation responsible for erection by parasympathetic fibres. These two systems
is mediated by nitric-oxide-induced relaxation of together with centres in the spinal cord and pons
the cavernosal sinusoids. The damage to these coordinate the cycle of filling and empting of the
structures results in impotence, the most severe bladder. Damage to the parasympathetic fibres
sexual deficit in men. causes non-coordinated action of the detrusor
On the other hand, sympathetic fibres are and overactive sympathetic stimulation of the
responsible for emission of semen from the semi- bladder base. As a result, we have the most com-
nal vesicles into the urethra. Sympathetic fibres mon consequence of the damaged vegetative
also innervate the vasa, prostate, urethra and the innervation, flaccid bladder with high intravesi-
internal urethral sphincter. Coordinated sympa- cal pressure. Patients in this situation experience
thetic activation of these structures, together with urinary retention. Infrequently, due to the isolated
somatically innervated bulbospongiosus and sympathetic denervation or a lesion of levator ani
ischiocavernosus muscles, results in ejaculation. nerve, which is a somatic structure, patients can
The sacral segment of spinal cord is responsible experience urinary incontinence [28, 62]. Urinary
for the coordination of the sexual response. dysfunction after rectal cancer operations is pres-
Damage to these structures most commonly ent in 4–28% of the cases [27, 37, 48]. In males,
causes the absence of ejaculation or a case of a urinary disturbances are rather frequent, occur-
retrograde one due to denervation of internal ure- ring in around 40%. On the other hand, urinary
thral sphincter. Male sexual dysfunction is pres- incontinence is infrequently reported [27, 37].
ent in around 10–35% of cases after rectal cancer
operations [13, 34, 39]. Enker reported that spon-
taneous postoperative erection was possible in 24.4 Anatomical Consideration
85% of the patients [9], and similar results were in the Context of Function
achieved by other specialized surgeons [14, 48]. Preservation
In women, parasympathetic impulses trigger
the release of vasoactive intestinal polypeptide 24.4.1 Pelvic Vegetative Nerve
from nerve endings in the vaginal wall that results Structures
in intensive vaginal transudation of fluid which
together with secretion of Bartholin glands allows 24.4.1.1 Sympathetic Innervation
good lubrication of vagina. As in males, parasym- The sympathetic impulses to the pelvis are gener-
pathetic impulses are responsible for vascular ated in the intermediolateral grey matter of the
236 Z. Krivokapic and I. Dimitrijevic

contact with mesorectal fascia. Individual dif-


ferences in the size of these structures are rather
great; sometimes, they are thick voluminous
structures, but in some cases, they are filament
like, barely visible.
Distally hypogastric nerves join parasympa-
thetic errigent nerves to form inferior hypogastric
(pelvic) plexus.

24.4.1.2 Parasympathetic Innervation


The parasympathetic nerve supply to the pelvis
derives from sacral parasympathetic ganglia
located in the S2–S4 segments of the spinal cord.
Preganglionic fibres pass through second, third
and fourth sacral foramina, going anterolaterally,
just close to piriform muscle towards the hypo-
gastric nerves piercing the pelvis sidewall facia,
and after joining with these sympathetic struc-
tures, they form inferior hypogastric (pelvic)
plexus [28].
Fig.24.1 Schematic presentation of pelvic autonomic There are certain variations in the type and
system
number of errigent nerves that participate in for-
mation of pelvic plexus. They most frequently
spinal cord in the level of T12 to L2(3) (Fig. 24.1). are from S3 and S4 (60%), sometimes from S2
The preganglionic fibres come out of the spinal (40%) and rarely from S5 roots of the spinal
cord through spinal nerve and through white cord [40].
ramus communicans into the sympathetic para-
vertebral chain. The lumbar splanchnic nerves 24.4.1.3 The Pelvic Plexus
are predominantly preganglionic sympathetic The pelvic plexus is the major centre for coordi-
structures that join preaortic plexus that covers nation of the vegetative innervation of the pelvis.
the full length of anterior side of the abdominal It is placed between thick pelvic sidewall fascia
aorta. The logical continuation of this structure is and much thinner mesorectal fascia, which makes
inferior mesenteric plexus, localized opposite the it in some individuals very difficult to properly
inferior mesenteric artery. These nerve fibres visualize.
form two filaments that lead to the anterior sur- This plexus in males is described as a rectan-
face of the aorta that join together in the level of gular structure, around 5 cm long and 3 cm wide,
sacral promontory where superior hypogastric with its midpoint at the tip of the seminal vesicles
plexus is formed [6, 40]. Many of these pregan- [62]. It is located in the sagittal plane on either
glionic fibres make synapses at this point. Besides side of the rectum and pierced by a number of
these, this plexus contains ascending parasympa- vessels that supply rectum, bladder, seminal ves-
thetic filaments that innervate descending colon icles and prostate [53].
as well as afferent fibres from the pelvic organs. On the other hand, in females, pelvic plexus, by
Distally, superior hypogastric plexus divides some authors, is described as a triangle, with a
into two filament-like structures, around 4–7- posterior base and an anterior inferior top which is
mm wide called hypogastric nerves. These at the ureter’s point of contact where it perforates
nerves lead inferolaterally medial to the ureter, the posterior layer of the broad ligament [40].
beneath the peritoneum. They are also placed Besides its main tributaries, hypogastric and
inside the internal iliac vessels and in close errigent nerves, in 30% of the cases, sympathetic
24 How Is Nerve-Sparing Surgery Well Performed? 237

afferences exist, most commonly from a single fascia can sometimes be more obvious in patients
branch of the second, third or fourth sacral gan- thatunderwentneo-adjuvanttreatment.Nevertheless,
glion [40]. in most of the cases, this layer is thicker than the
mesorectal fascia [5].
24.4.1.4 Efferences of the Pelvic Plexus Histologically, it is constituted of collagen,
In females, the vaginal branches distally come out elastic and smooth muscle fibres. As mentioned,
of the top of the pelvic plexus through branches these longitudinal smooth muscle fibres are
leading to the bladder, vagina and the rectum. These accompanied by delicate nerve filaments [1, 55].
branches come from two stalks just beneath the Prostate and seminal vesicles in males and vagina
point where ureter crosses uterine artery. One trunk in females are situated anteriorly [1, 35].
that innervates bladder is distally, down to the ure- Posteriorly, mesorectal fascia, thin mesorectum
ter, divided into further two branches – lateral and and rectal wall are found. This structure is contin-
medial trigonal. The other trunk innervates vagina ued to the perineal body caudally [8].
and goes along with uterine artery. This branch, at On the matter of adherence of this structure to
the lateral edge of vagina, is divided into further the surrounding organs, there is no current con-
two fibre groups. Some of these fibres perforate the sensus. Sex differences are present, and generally
posterior vaginal wall and can be found in the rec- in females, it is less adherent to the anterior organs
tovaginal septum [40]. opposed to males, especially distally. Some
In males, the most prominent and important authors state that adherence is generally stronger
efferent of the pelvic plexus is the urogenital neu- to the anterior organs, but others have opposite
rovascular bundle. From its origin, at the most dis- opinion [5, 23].
tal part of the pelvic plexus, it goes posterolaterally According to Bill Heald and a number of other
to the prostate and continues laterally on the both authors [17, 45] Denonvilliers’ fascia is a part of
sides of the urethra. Approximate thickness of this the mesorectum anteriorly, thus suggesting that
structure is around 10–12 mm. It contains ganglia during the dissection, this structure should be left
as well as nerve fibres together with connective tis- on the mesorectal specimen. According to them,
sue. This structure is situated between two layers a distinct plane of dissection exists between sem-
of the lateral pelvic fascia (levator and prostatic inal vesicles and Denonvilliers’ fascia. Applying
fascia). The neurovascular bundle branches this approach mentioned, authors managed to
through parietal pelvic fascia and medially through maintain good sexual and urinary function post-
Denonvilliers’ fascia to the prostate and urinary operatively, which suggests that we can avoid
bladder. The rest of the fibres (cavernous nerves) damage to vegetative nerve structures if we fol-
pass around the base of the prostate base and con- low the so-called mesorectal plane [15, 36].
verge in a ‘pincer-shaped’ formation, close to ure- Others state that this structure has two leaves,
thra. Distally, these nerves pierce through pelvic suggesting that appropriate dissection plane
floor muscles and course to genital organs [6, 54]. should be through the Denonvilliers’ fascia [47].
Nevertheless, this structure is far from being well- Some more recent histological studies show
defined. Some authors [30] found distinct neuro- that this fascia is more adherent to the anterior
vascular bundle in only 48% of the cases. In the organs, and the reason for leaving it on the rectal
remaining 52%, nerve fibres were spread all along side is more oncologic than anatomic one, in cases
the lateral side of the prostate. of low anteriorly placed rectal carcinomas [35].

24.4.2 Anatomy of Denonvilliers’ Fascia 24.4.3 The ‘Lateral Ligaments’


of the Rectum
During the rectal mobilization, this structure is
sometimes easily visible as a membrane, but some- These ligaments have been described in some
times, it can be very thin and fragile. Denonvilliers’ papers and textbooks as a structure that arises
238 Z. Krivokapic and I. Dimitrijevic

from the endopelvic fascia of the pelvic sidewall sacral plexus, separately from pudendal nerve.
that goes anteromedially as it approaches to the The levator ani nerve approaches levator ani mus-
rectum. Miles, Goligher and some other older cle from within the pelvis on the superior surface
authors suggested ‘clamping’ or ‘hooking on the of the pelvic floor. Its position makes this struc-
finger’ of this structure [11]. ture amenable to accidental damage during TME.
Also, it has been suggested that these liga- This nerve gives substantial innervation to levator
ments contain fatty tissue, nerves and vessels muscle as mentioned, and its injury can explain
(middle rectal artery), which present oncological certain proportion of postoperative faecal and uri-
danger for the rectal cancer patients; thus, these nary incontinence [61].
should be clamped near the lateral pelvic side-
wall [11]. This approach proved to be devastating
in the terms of preservation of pelvic vegetative 24.5 Important Points of Nerve
nerves, since according to recent studies, impor- Preserving TME
tant pelvic ganglia are contained in this area,
especially in its lateral third [6]. The basic concept of the TME procedure is to
With the advances in this field of surgery, follow embryologically defined layers. According
especially since the introduction of TME in 1982, to Mr. Bill Heald, the founder of this method, ‘the
by Bill Heald [18], situation changed. Heald and corollary to the perfect specimen and cure is
others state that there is no such structure and the perfect preservation of the layers surrounding
sharp dissection just close to mesorectal fascia is the mesorectum which, are formed by the auto-
adequate and oncologically sufficient. Resent nomic nerves and plexuses’ [15].
anatomical studies supported this opinion [25]. During the nowadays standard operation for
In addition, studies have shown that middle rectal rectal carcinoma of the middle and lower third
artery exists in only 25–50% of cases and its of rectum (TME), there are a number of points
diameter is rarely greater than 2 mm [25, 52], where a surgeon can easily damage some of the
which makes diathermy perfectly safe method in mentioned vegetative nerve structures. We will
transection of this structure. go through these steps and try to ascertain
mechanisms of vegetative nerve injury and their
prevention.
24.4.4 Pudendal and Levator Ani Nerve

Not only autonomic nerves play an important 24.5.1 Mobilization of the Left Colon
role in sexual and urinary function. Two somatic and Ligation of the Inferior
nerves are responsible for certain aspect of these Mesenteric Artery
functions. Those are pudendal nerve and levator
ani nerve. Proper mobilization of the left colon involves
In the infralevatory compartment, pudendal finding the right plane between two leaves of
nerve arises from S2–S4 roots of the sacral plexus Toldt’s fascia, leaving retroperitoneal surface
and runs through ischioanal fossa to the genital with gonadal vessels, ureter and preaortic sym-
area giving sensory and motor branches, i.e., dor- pathetic fibres intact. This is done by dividing
sal penis or clitoris nerve. In males, pudendal the white (Toldt’s) line of the sigmoid colon
nerve gives motoric impulses for ischiocaverno- from the peritoneum of the lateral abdominal
sus and bulbospongiosus muscle, important for wall (Fig. 24.2). By gently lifting upwards meso-
rigid phase of erection [6]. The damage to puden- colon together with so-called pedicle package,
dal nerve can cause faecal incontinence. Relatively, we reveal a correct dissection plane (Fig. 24.3).
recent papers have re-emphasized another somatic Performing this, we can easily identify and pre-
nerve that innervates levator muscles. That is leva- serve already mentioned two preaortic sympa-
tor ani nerve that arises from S3–S4 roots of the thetic trunks going around inferior mesenteric
24 How Is Nerve-Sparing Surgery Well Performed? 239

Fig.24.2 The division of the Toldt’s white line of the


sigmoid colon

Fig. 24.4 Ligated inferior mesenteric artery and vein

inferior mesenteric artery as described, but the


vein can be divided nearby.

24.5.2 Division of the Sigmoid Colon

Sigmoid colon along with its mesentery should


be divided on the safe distance proximally to the
Fig. 24.3 Mobilized left colon with visible ‘pedicle tumour. This step may appear insignificant, but
package’, superior rectal artery, vein and para-aortic plexus its importance is in the optimal mobility of the
specimen which provides clear and direct visual-
artery. Additionally, this plane of dissection is ization of the plane of dissection around mesorec-
continued distally with the extrafascial plane of tal fascia where important vegetative nerve
dissection of mesorectum, leaving its fascia structures are situated.
intact [15, 28].
With completely mobilized and lifted left
colon, it is easy to identify the proximal end of 24.5.3 High Posterior and Lateral
the pedicle package. It is advisable to separately Dissection
ligate inferior mesenteric vessels (Fig. 24.4).
Ligating of inferior mesenteric artery can be The traction of the transected sigmoid colon
safely done 1–2 cm distally to the aorta in order ‘towards the ceiling’ reveals shiny posterior side of
to avoid conflict with preaortic sympathetic mesorectal fascia. Visualizing the fascia usually
nerves. On the other hand, the vein is usually precedes identification of vegetative nerve struc-
divided close to the lower edge of pancreas. tures (Fig. 24.5). Superior hypogastric plexus is
When performing APE, it is satisfactory to ligate situated on the sacral promontory, and hypogastric
240 Z. Krivokapic and I. Dimitrijevic

Fig. 24.5 Posterior surface of mesorectal fascia and areolar,


avascular tissue preceding superior hypogastric plexus Fig. 24.7 Recto-sacral (Waldeyer’s) fascia partially
transected

24.5.4 Distal Posterior and Lateral


Dissection

Further distal posterior dissection is an important


step of the TME. It is mandatory to follow the
right plane as one proceeds downwards. At a
strict posterior plane, there are no important veg-
etative nerve structures, but sharp dissection of
the areolar, avascular tissue together with the
transection of the recto-sacral (Waldeyer’s) fascia
gives a surgeon correct plane for lateral and even-
tually anterior dissection (Fig. 24.7). Also, by
mobilizing the posterior aspect of the mesorec-
tum, proper traction and counter-traction can be
achieved in order to perform correct visualization
Fig. 24.6 Superior hypogastric plexus and hypogastric of the lateral vegetative nerve structures, which is
nerves (With permission of Bill Heald) one of the main advantages of open surgery to
laparoscopic approach [12, 15, 24, 50, 51].
nerves, by leaving it, form a ‘wishbone-like’ struc- Lateral dissection is one of the most important
ture (Fig. 24.6). Accidental damage to these struc- phases of the nerve-preserving TME. It involves
tures is avoided by traction and counter-traction strict following of the plane obtained posteriorly,
together with sharp dissection keeping the nerves with further ‘peeling off’ of the hypogastric
in view all the time. The hypogastric nerves are nerves to the level of the ‘lateral ligaments’
placed just beneath the level of the peritoneal (Fig. 24.8). Already mentioned errigent nerves
reflection, and by incising the peritoneum more lat- are during the lateral dissection seen as ‘errigent
erally than optimal, surgeon can easily transect pillars’ [15]. These structures are placed laterally
them. Also, hypogastric nerves are relatively adher- outside the parietal fascia which they penetrate,
ent to the mesorectal fascia, and by traction of the going towards the pelvic plexus in posteroante-
specimen to the left, nerves can be ‘tented up’ and rior direction. Though we already mentioned that
accidentally injured. To avoid this, one must care- there are most frequently three errigent nerves,
fully ‘peel off’ the nerves from mesorectal fascia. intraoperatively, we usually identify one or some-
Bilateral sacrifice of hypogastric nerves almost cer- times two of these nerve structures (most fre-
tainly leads to ejaculatory failure [37, 56]. quently from S3 component), making a nerve
24 How Is Nerve-Sparing Surgery Well Performed? 241

and small blood vessels penetrate mesorectal


fascia. Once more, it is important to stress that
main constituents of the so-called lateral liga-
ment are vegetative nerve fibres and ganglia,
whereas middle rectal artery is found only in
around 28% in anatomical studies [33, 52, 57],
bilaterally present in only one-third of the cases.
The diameter of this artery is only around 1.3 mm
and gradually deceasing during the course to
mesorectal fascia, thus becoming barely visible
at the point where it pierces it. Therefore, it is
practically impossible to identify lateral liga-
ments during the correct TME procedure. Also,
Fig. 24.8 Proximal lateral dissection with visible
hypogastric nerves adherent to mesorectal fascia
it is perfectly safe to follow the mesorectal plane
with absolutely no need to clamp or ligate any
vascular structures [33, 52, 57].
If one tends to proceed with the dissection
more laterally, injury to the pelvic plexus may
occur, especially if one applies too much traction
to the rectum, displacing the plexus superiorly
and medially [46]. At this point, if the pelvic
plexus is damaged, both sympathetic and para-
sympathetic functional deficits may occur.
On the other hand, if a surgeon tries to stay far
away from the pelvic plexus and ‘cones in’ the
mesorectum, a need for ligation of the vessels is
frequently possible. These vessels are actually a
Fig. 24.9 ‘Lateral ligament’ of the rectum, i.e. inferior
hypogastric plexus with visible ‘errigent pillar’ (With
part of the mesorectal envelope and should be left
permission of Bill Heald) intact. Otherwise, an oncological quality of the
operation is diminished by leaving a part of the
mesorectum within the pelvis [15].
root from the front of the S3 part of the main
sacral plexus (Fig. 24.9).
The traction to the rectum is most likely 24.5.5 Anterior Dissection
responsible for pillar-like appearance of errigent
nerves, which makes them easily recognizable. This phase of the TME is probably the most
This may be an explanation why it is easier to demanding in terms of vegetative nerve preserva-
identify these nerves during open surgery, despite tion. As mentioned, the dissection is conducted
magnification that is available during laparo- in a very thin area between rectum and seminal
scopic approach that lacks in sufficient traction to vesicles and prostate or vagina where anterior
the rectum. mesorectum, mesorectal fascia and Denonvilliers’
Further anteriorly, the dissection plane is fascia are situated (Fig. 24.10). There is still no
extended to the vesicles with the hypogastric absolute consensus in terms where to proceed
plexus neural plate placed with its centre on the with the dissection plane in this area [35].
tips of the seminal vesicles. At this point, one According to Bill Heald, dissection anteriorly
has to be extremely careful. Namely, between and anterolaterally should follow the plane
mesorectal fascia and pelvic plexus, an area of obtained during posterior and lateral phase.
adherence exists, in the place where nerve fibres Proximally, peritoneum should be incised in a way
242 Z. Krivokapic and I. Dimitrijevic

Fig. 24.12 The correct plane of anterior dissection with


Fig. 24.10 The area of anterior dissection after two thirds visible Denonvilliers’ fascia posteriorly and seminal vesi-
of posterolateral dissection performed cles anteriorly

Fig. 24.11 Proper incision of the peritoneum reflection at Fig. 24.13 Specimen after performed TME with lower
the beginning of anterior dissection edge of Denonvilliers’ fascia transected in the level of the
lower third of prostate

to encompass peritoneal reflection that should transversely (Fig. 24.13). This is a crucial step in
remain on the specimen (Fig. 24.11). Entering this the preservation of delicate neurovascular bundles
plane enables us to clearly visualize anterior that present distal continuation of the inferior
organs, seminal vesicles and vagina. Good retrac- hypogastric plexus and practically invisible cav-
tion of these structures in most cases reveals avas- ernous nerves that pass anterolaterally to the rec-
cular, areolar tissue and relatively straightforward tum on the way through urogenital diaphragm
dissection plane distally which leaves shiny, before entering cavernous bodies [10, 16]. Once
smooth anterior side of the specimen (Fig. 24.12). again, in order to properly preserve neurovascular
This smooth surface on the specimen is actually bundles of Walsh, it is mandatory to be aware of its
Denonvilliers’ fascia. As one proceeds distally, correct anatomical position, especially in males
comes to a point where Denonvilliers’ fascia (Fig. 24.14). From the distal part of the pelvic
becomes adherent to the posterior capsule of plexus, it travels caudally, under the pelvic floor
the prostate. Here, fascia should be divided fascia. The neurovascular bundle pierces this
24 How Is Nerve-Sparing Surgery Well Performed? 243

malignant tissue. Another possible approach is


‘close rectal plane’, which lies immediately on
the rectal musculature. This is not an anatomical
plane, and the dissection through the tissue of
anterior mesorectum is often bloody and difficult
since numerous blood vessels cross this plane
before entering the rectal wall [5]. In the mind of
most colorectal surgeons today, when we are
talking about malignant disease, there is little
doubt that this approach is not safe. Still, some
Fig. 24.14 View on the posterior side of prostate, with [35, 36] advocate that in cases of posteriorly
present Denonvilliers’ fascia transected in the lower third
placed non-advanced tumours, we can use this
and intact neurovascular bundles posterolaterally, after
performed ‘face down’ APR (With permission of Bill approach since the risk of damaging vegetative
Heald) nerves is minimal and oncological safety is satis-
factory since there is little risk of tumour spread-
ing in this area of mesorectum. Then again, there
fascia, and part of the nerves goes through is data that if we, even in cases of benign disease,
Denonvilliers’ fascia towards prostate and urinary use close rectal dissection plane, we do not
bladder. The rest of the fibres courses around the improve vegetative nerve preservation, compared
prostate base, then through pelvic diaphragm to TME [34].
towards genital organs.
Having in mind these anatomical consider-
ations, it is obvious that the dissection plane 24.5.6 Mobilization of the Most Caudal
described above has several advantages. First, the Portion of the Mesorectum
plane one follows is avascular, thus visualization
is optimal; second, from oncological point of The anatomical relations of the most distal
view leaving the Denonvilliers’ fascia on the mesorectum and surrounding organs are often very
specimen, the risk for oncologically insufficient difficult to visualize and comprehend because
operation is minimal; and third, by transecting the traction, counter-traction and upward traction
the rectogenital fascia at the mentioned level, we distorts actual situation. However, a clear percep-
avoid collision with often invisible vegetative tion of the cone-like pelvic floor together with
structures [20]. globular bilobed mesorectum is necessary in
Nevertheless, different opinions considering order to preserve delicate nerve structures placed
the right plane in the anterior dissection exist. around it.
Some advocate taking the entire Denonvilliers’ It is well established that there is higher per-
fascia [45], using as some would say ‘extrame- centage of sexual and urinary dysfunction after
sorectal plane’ [35, 36]. This approach is onco- APR compared to sphincter-saving procedures
logically safe, but risk of the damaging vegetative [13, 37, 63, 67]. This can be explained by the
nerves is too high. On the other hand, some advo- damage to the neurovascular bundles and cavern-
cate ‘mesorectal’ plane, which is immediately ous nerves during the perineal act of the proce-
outside the mesorectal fascia, practically the con- dure. It is often difficult to enter the correct plane,
tinuation of the plane obtained during posterior especially anteriorly where all delicate structures
and lateral dissection thus leaving entire are placed. Additional difficulty presents an effort
Denonvilliers’ fascia on the anterior organs [35, not to ‘cone in’ the specimen and start the perineal
36]. This plane is however difficult to follow and phase early enough. In order to avoid this, some
not always readily visible. Additionally, if we authors advocate ‘face down’ position and plac-
have a low, anteriorly placed tumour, oncologi- ing the clips on the most distal vegetative nerve
cally it might not be safe to go so close to the structures visible during the abdominal phase of
244 Z. Krivokapic and I. Dimitrijevic

Fig. 24.15 Specimen after


correctly performed TME with
coloured mesorectal fascia and
cross sections to inspect
specimen quality

the operation. Removing of the coccyx which is a [60]. This is a smooth muscle situated in the
standard part of this procedure enables the sur- levator hiatus, in the place where Denonvilliers’
geon to properly visualize neurovascular bundles fascia ends. It is connected to the external anal
and posterior part of the prostate, much easier sphincter, as well as with muscle layer of the rec-
than during standard abdominoperineal proce- tum. Cavernous nerves pierce this structure, and
dure [3, 22, 64, 65]. it is surrounded with tortuous anorectal veins. If
Additionally, in cases of very low rectal carci- we tend to dissect through this structure during
nomas when performing APR or intersphincteric the perineal act of APR, we can transect cavern-
resection, it is rather difficult to find a correct ous nerves and mentioned veins can be damaged.
plane or one can encounter venous bleeding in Afterwards, in an attempt to control the bleeding,
the area of anorectum passing levator hiatus. we can also do the damage to vegetative nerves
Some anatomical studies showed that this can be during the mass ligation of the anterior structures
explained by existence of rectourethral muscle (Fig. 24.15).
24 How Is Nerve-Sparing Surgery Well Performed? 245

24.5.7 Laparoscopic TME in the Context even with expert rectal surgeons. Thus, in
of Nerve Identification and words of Mr. Bill Heald, ‘specimen-oriented
Preservation surgery’ may be the clue. If we obtain a perfect
specimen, with no defects of extra tissue on the
In recent years, laparoscopic approach is gaining mesorectal fascia, it can be expected, with a
ground in this field of surgery. It would be logical certain amount of certainty, that functional
that better magnification facilitates the visualiza- result of the operation will be favourable. A
tion and identification of nerve structures within careful TME with proper reconstruction is a
the pelvis. A number of studies addressed the complex procedure, and with careful nerve
issue of postoperative sexual and urinary func- preservation takes 3–5 h according to the
tion after laparoscopic TME. A few authors patient’s and tumour characteristics; a conven-
achieved excellent results, of only 6% urinary tional APE was often completed in 1 h.
and sexual dysfunction [26]. Still, majority state
that postoperative urinary and sexual function
was worse compared to open technique. Quah
and co-workers noted significant percentage of References
sexual and urinary deficits especially in patients
1. Aigner F, Zbar AP et al (2004) The rectogenital sep-
with low and bulky rectal tumours [50]. To the tum: morphology, function, and clinical relevance.
similar conclusion came Rullier in a study that Dis Colon Rectum 47(2):131–140
dealt only with distally placed rectal carcinomas 2. Bernstein WC, Bernstein EF (1966) Sexual dysfunc-
[51]. This can be explained by insufficient trac- tion following radical surgery for cancer of the rec-
tum. Dis Colon Rectum 9(5):328–332
tion and counter-traction in the areas where this 3. Bevan KE, Moran BJ (2009) Optimizing rectal cancer
manoeuvre helps us to differentiate minute nerve surgery by total mesorectal excision and “cylindrical”
structures placed between thicker fascial struc- extralevator techniques for abdominoperineal exci-
tures and organs. Also, present laparoscopic sion. Curr Colorectal Cancer Rep 5(4):219–223
4. Camilleri-Brennan J, Steele RJC (1998) Quality of
instruments do not allow adequate manipulation life after treatment for rectal cancer. Br J Surg 85(8):
in the distal parts of the pelvis. Mentioned trend 1036–1043
confirms multicentre randomized trial that com- 5. Church JM, Raudkivi PJ et al (1987) The surgical anat-
pared short-term results of open versus laparo- omy of the rectum - a review with particular relevance
to the hazards of rectal mobilisation. Int J Colorectal
scopic-assisted surgery for colorectal cancer. Dis 2(3):158–166
Severe change in sexual function was noted in 6. Clausen N, Wolloscheck T et al (2008) How to opti-
41% of patients in the laparoscopic group com- mize autonomic nerve preservation in total mesorectal
pared to 26% of patients in the open group. These excision: clinical topography and morphology of pelvic
nerves and fasciae. World J Surg 32(8):1768–1775
data are to be taken with caution, because groups 7. Da Silva GM, Zmora O et al (2004) The efficacy of a
were not well matched due to higher number of nerve stimulator (Cavermap®) to enhance autonomic
TME procedures in laparoscopic group caused nerve identification and confirm nerve preservation
by inability to correctly locate the tumour and during total mesorectal excision. Dis Colon Rectum
47(12):2032–2038
perform transection. Additionally, conversions 8. Denonvilliers C (1836) Anatomie du perinée. Bull
took place due to excessive bleeding that pre- Soc Anat 3rd Series Paris 11: 105–106
vented visualization of nerve structures [12]. 9. Enker WE, Thaler HT et al (1995) Total mesorectal
excision in the operative treatment of carcinoma of
the rectum. J Am Coll Surg 181(4):335–346
Conclusion 10. Fazio VW, Fletcher J et al (1980) Prospective study of
Functional deficits after rectal cancer surgery the effect of resection of the rectum on male sexual
are still present in significant percentage of function. World J Surg 4(2):149–152
cases. The way to improve functional results is 11. Goligher J (1960) Colon, rectum, and anus – surgical.
Med Annu 78:40–53
to advance surgical training and knowledge in 12. Guillou PJ, Quirke P et al (2005) Short-term end-
this area. Exact identification of all pelvic veg- points of conventional versus laparoscopic-assisted
etative nerve structures is not always possible surgery in patients with colorectal cancer (MRC
246 Z. Krivokapic and I. Dimitrijevic

CLASICC trial): multicentre, randomised controlled 30. Kiyoshima K, Yokomizo A et al (2004) Anatomical
trial. Lancet 365(9472):1718–1726 features of periprostatic tissue and its surroundings: a
13. Havenga K, Enker WE et al (1996) Male and female histological analysis of 79 radical retropubic prostate-
sexual and urinary function after total mesorectal ctomy specimens. Jpn J Clin Oncol 34(8):463–468
excision with autonomic nerve preservation for carci- 31. Kneist W, Junginger T (2007) Intraoperative electrostim-
noma of the rectum. J Am Coll Surg 182(6):495–502 ulation objectifies the assessment of functional nerve
14. Havenga K, Maas CP et al (2000) Avoiding long-term preservation after mesorectal excision. Int J Colorectal
disturbance to bladder and sexual function in pelvic Dis 22(6):675–682
surgery, particularly with rectal cancer. Semin Surg 32. Lange MM, Maas CP et al (2008) Urinary dysfunc-
Oncol 18(3):235–243 tion after rectal cancer treatment is mainly caused by
15. Heald B (2008) Autonomic nerve preservation in rec- surgery. Br J Surg 95(8):1020–1028
tal cancer surgery – the forgotten part of the TME 33. Lin M, Chen W et al (2010) The anatomy of lateral
message a practical “workshop” description for sur- ligament of the rectum and its role in total mesorectal
geons. Acta Chir Iugosl 55(3):11–16 excision. World J Surg 34(3):594–598
16. Heald RJ (1988) The ‘Holy Plane’ of rectal surgery. 34. Lindsey I, George BD et al (2001) Impotence after
J R Soc Med 81(9):503–508 mesorectal and close rectal dissection for inflammatory
17. Heald RJ, Moran BJ (1998) Embryology and anatomy bowel disease. Dis Colon Rectum 44(6):831–835
of the rectum. Semin Surg Oncol 15(2):66–71 35. Lindsey I, Guy RJ et al (2000) Anatomy of denon-
18. Heald RJ, Moran BJ et al (1998) Rectal cancer: the viliers’ fascia and pelvic nerves, impotence, and impli-
Basingstoke experience of total mesorectal excision, cations for the colorectal surgeon. Br J Surg 87(10):
1978–1997. Arch Surg 133(8):894–899 1288–1299
19. Hendren SK, O’Connor BI et al (2005) Prevalence of 36. Lindsey I, Warren BF et al (2005) Denonvilliers’ fas-
male and female sexual dysfunction is high follow- cia lies anterior to the fascia propria and rectal dissec-
ing surgery for rectal cancer. Ann Surg 242(2): tion plane in total mesorectal excision. Dis Colon
212–223 Rectum 48(1):37–42
20. Hill GL, Rafique M (1998) Extrafascial excision of 37. Maas CP, Moriya Y et al (1998) Radical and nerve-
the rectum for rectal cancer. Br J Surg 85(6):809–812 preserving surgery for rectal cancer in the Netherlands:
21. Hojo K, Vernava AM III et al (1991) Preservation of a prospective study on morbidity and functional out-
urine voiding and sexual function after rectal cancer come. Br J Surg 85(1):92–97
surgery. Dis Colon Rectum 34(7):532–539 38. Maas CP, Moriya Y et al (2000) A prospective study on
22. Holm T, Ljung A et al (2007) Extended abdomino- radical and nerve-preserving surgery for rectal cancer
perineal resection with gluteus maximus flap recon- in the Netherlands. Eur J Surg Oncol 26(8):751–757
struction of the pelvic floor for rectal cancer. Br J Surg 39. Masui H, Ike H et al (1996) Male sexual function after
94(2):232–238 autonomic nerve-preserving operation for rectal can-
23. Huland H, Noldus J (1999) An easy and safe approach cer. Dis Colon Rectum 39(10):1140–1145
to separating Denonvilliers’ fascia from rectum dur- 40. Mauroy B, Demondion X et al (2007) The female
ing radical retropubic prostatectomy. J Urol 161(5): inferior hypogastric (= pelvic) plexus: anatomical and
1533–1534 radiological description of the plexus and its affer-
24. Jayne DG, Guillou PJ et al (2007) Randomized trial of ences – applications to pelvic surgery. Surg Radiol
laparoscopic-assisted resection of colorectal carci- Anat 29(1):55–66
noma: 3-year results of the UK MRC CLASICC Trial 41. McArdle CS, McMillan DC et al (2003) Male gender
Group. J Clin Oncol 25(21):3061–3068 adversely affects survival following surgery for col-
25. Jones OM, Smeulders N et al (1999) Lateral ligaments orectal cancer. Br J Surg 90(6):711–715
of the rectum: an anatomical study. Br J Surg 42. McDonald PJ, Heald RJ (1983) A survey of postop-
86(4):487–489 erative function after rectal anastomosis with circular
26. Jones OM, Stevenson ARL et al (2009) Preservation stapling devices. Br J Surg 70(12):727–729
of sexual and bladder function after laparoscopic rec- 43. Moriya Y (2006) Function preservation in rectal can-
tal surgery. Colorectal Dis 11(5):489–495 cer surgery. Int J Clin Oncol 11(5):339–343
27. Junginger T, Kneist W et al (2003) Influence of 44. Moriya Y, Hojo K et al (1989) Significance of lateral
identification and preservation of pelvic autonomic node dissection for advanced rectal carcinoma at or
nerves in rectal cancer surgery on bladder dysfunction below the peritoneal reflection. Dis Colon Rectum
after total mesorectal excision. Dis Colon Rectum 32(4):307–315
46(5):621–628 45. Moriya Y, Sugihara K et al (1995) Nerve-sparing sur-
28. Keating JP (2004) Sexual function after rectal exci- gery with lateral node dissection for advanced lower
sion. ANZ J Surg 74(4):248–259 rectal cancer. Eur J Cancer Part A: Gen Top 31(7–8):
29. Kim NK, Aahn TW et al (2002) Assessment of sexual 1229–1232
and voiding function after total mesorectal excision 46. Mundy AR (1982) An anatomical explanation for
with pelvic autonomic nerve preservation in males with bladder dysfunction following rectal and uterine sur-
rectal cancer. Dis Colon Rectum 45(9):1178–1185 gery. Br J Urol 54(5):501–504
24 How Is Nerve-Sparing Surgery Well Performed? 247

47. Nano M, Levi AC et al (1998) Observations on surgical 58. Tawk RG, Ondra SL et al (2002) Intraoperative
anatomy for rectal cancer surgery. Hepatogastroenter- parasympathetic nerve stimulation with tumescence
ology 45(21):717–726 monitoring during total mesorectal excision for rec-
48. Nesbakken A, Nygaard K et al (2000) Bladder and tal cancer. J Am Coll Surg 195(4):506–512
sexual dysfunction after mesorectal excision for rectal 59. Tekkis PP, Cornish JA et al (2009) Measuring sexual
cancer. Br J Surg 87(2):206–210 and urinary outcomes in women after rectal cancer
49. Platell CFE, Thompson PJ et al (2004) Sexual health excision. Dis Colon Rectum 52(1):46–54
in women following pelvic surgery for rectal cancer. 60. Uchimoto K, Murakami G et al (2007) Rectourethralis
Br J Surg 91(4):465–468 muscle and pitfalls of anterior perineal dissection in
50. Quah HM, Jayne DG et al (2002) Bladder and sexual abdominoperineal resection and intersphincteric
dysfunction following laparoscopically assisted and resection for rectal cancer. Anat Sci Int 82(1):8–15
conventional open mesorectal resection for cancer. Br 61. Wallner C, Lange MM et al (2008) Causes of fecal
J Surg 89(12):1551–1556 and urinary incontinence after total mesorectal exci-
51. Rullier E, Sa Cunha A et al (2003) Laparoscopic sion for rectal cancer based on cadaveric surgery: a
intersphincteric resection with coloplasty and coloa- study from the cooperative clinical investigators of the
nal anastomosis for mid and low rectal cancer. Br J Dutch total mesorectal excision trial. J Clin Oncol
Surg 90(4):445–451 26(27):4466–4472
52. Sato K, Sato T (1991) The vascular and neuronal 62. Walsh PC, Retik AB, Vaughan ED, Wein AJ (eds) (2003)
composition of the lateral ligament of the rectum and Campbell’s urology. Elsevier Science, Philadelphia
the rectosacral fascia. Surg Radiol Anat 13(1):17–22 63. Weinstein M, Roberts M (1977) Sexual potency fol-
53. Schlegel PN, Walsh PC (1987) Neuroanatomical lowing surgery for rectal carcinoma. A follow-up of
approach to radical cystoprostatectomy with preserva- 44 patients. Ann Surg 185(3):295–300
tion of sexual function. J Urol 138(6):1402–1406 64. West NP, Anderin C et al (2010) Multicentre experi-
54. Schwalenberg T, Neuhaus J et al (2010) Neuroanatomy ence with extralevator abdominoperineal excision for
of the male pelvis in respect to radical prostatectomy low rectal cancer. Br J Surg 97(4):588–599
including three-dimensional visualization. BJU Int 65. West NP, Finan PJ et al (2008) Evidence of the onco-
105(1):21–27 logic superiority of cylindrical abdominoperineal
55. Silver PH (1956) The role of the peritoneum in the excision for low rectal cancer. J Clin Oncol 26(21):
formation of the septum recto-vesicle. J Anat 90(4): 3517–3522
538–546 66. Wichmann MW, Muller C et al (2001) Gender differ-
56. Sugihara K, Moriya Y et al (1996) Pelvic autonomic ences in long-term survival of patients with colorectal
nerve preservation for patients with rectal carcinoma: cancer. Br J Surg 88(8):1092–1098
oncologic and functional outcome. Cancer 78(9): 67. Williams DI, Watson PC et al (1951) Discussion on
1871–1880 urological complications of excision of the rectum.
57. Takahashi T, Ueno M et al (2000) Lateral ligament: its Proc R Soc Med 44(9):819–828
anatomy and clinical importance. Semin Surg Oncol
19(4):386–395
Is Laparoscopic Rectal Surgery
the Gold Standard? 25
David Jayne and Laeeq Khan

Contents 25.1 Introduction


25.1 Introduction ................................................ 249
The last two decades have seen a gradual but
25.2 Evolution of Laparoscopic
Colorectal Surgery ..................................... 250
shifting enthusiasm for laparoscopic general sur-
gery. A better appreciation of the laparoscopic
25.3 Future Developments in Laparoscopic
approach with adaptation of open surgical tech-
Rectal Cancer Surgery ............................... 255
niques and improvements in instrumentation has
References ............................................................... 255 helped this transition. A realization of the patient
benefits that can be gained from laparoscopic sur-
gery in benign disease has increased interest in its
application in cancer surgery. Colorectal surgery,
by its very nature, is challenging for the laparo-
scopic approach, involving multi-quadrant dis-
section, manipulation of bulky tissues and a
dependency on assistants for retraction. In rectal
cancer, the situation is made more difficult by the
confines of the bony pelvis, the proximity of vital
structures and the precision demanded to achieve
complete tumour resection with an intact
mesorectum and preservation of pelvic auto-
nomic nerves. For colon cancer, there has been a
D. Jayne (*)
John Goligher Colorectal Unit, general recognition for some time that laparo-
University of Leeds, scopic surgery can produce oncological outcomes
Leeds, UK at least equivalent to open surgery, but with the
Level 7 Clinical Sciences Building, added advantages of quicker patient recovery and
St James’s University Hospital, shorter hospital stay. In this chapter, we will
Leeds LS9 7TF, UK
explore the evolution of laparoscopic colorectal
e-mail: d.g.jayne@leeds.ac.uk
surgery, with particular emphasis on rectal can-
L. Khan
cer, and ask whether laparoscopic rectal cancer
John Goligher Colorectal Unit,
Leeds Teaching Hospitals NHS Trust, surgery has acquired the same acceptance as
Leeds, UK colon cancer to become the new ‘gold standard’.

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 249


DOI 10.1007/978-3-642-25005-7_25, © Springer-Verlag Berlin Heidelberg, 2012
250 D. Jayne and L. Khan

25.2 Evolution of Laparoscopic and readmission rates. There was no difference in


Colorectal Surgery survival outcomes, with local recurrence rates of
16% and 18% and overall 3-year survival rates of
Laparoscopic techniques were introduced into 86% and 85% for the laparoscopic-assisted and
colorectal practice in the late 1980s, and it open groups, respectively. Importantly, recurrence
soon became apparent that the benefits seen in rates in surgical wounds were 1% for both the
other general surgical procedures (cholecystec- laparoscopic-assisted and open groups, indicating
tomy, fundoplication, hernia repair, etc.) could that abdominal wound metastasis was not peculiar
also be achieved following colectomy. Several to the laparoscopic approach.
studies reported improved outcomes compared The first randomized control trial to address
to conventional open surgery with reduced the issue of rectal cancer was the UK MRC-
post-operative pain, less analgesic requirement, CLASICC (Conventional versus Laparoscopic-
shorter hospital stay, better cosmesis and quicker Assisted Surgery for Colorectal Cancer) trial [3].
return to normal function. A desire to replicate Recruitment commenced in 1996 and, over the
these outcomes in cancer patients led to the next 6 years, accumulated a total of 794 patients
uptake of laparoscopic techniques for colon and, from 32 surgeons across the UK, randomized on
subsequently, rectal cancer. Initial clinical enthu- a 2:1 basis in favour of the laparoscopic approach.
siasm, supported to some extent by commercial Of the 794 patients, 381 had rectal cancer, with
drivers, was tempered by unease expressed by 252 randomized to laparoscopic-assisted surgery
many that the widespread adoption of hitherto either by anterior resection or abdominoperineal
untested laparoscopic techniques might risk excision. Uniquely at that time, the CLASICC
inferior oncological outcomes. This view found trial included a centralized review of pathology
support with the reporting of high rates of port- that included assessment of the quality of surgi-
site metastases and led to calls for laparoscopic cal plane. The early results from CLASICC were
cancer surgery to be abandoned. The response reported in 2005 and showed no significant dif-
from national bodies at that time was reserved, ference in the short-term end points, which
with the National Institute for Clinical Excellence included morbidity, mortality and pathological
in the UK advising ‘For colorectal cancer, open surrogate markers for oncological outcome. Time
rather than laparoscopic resection should be the to resumption of normal diet and time to first
preferred surgical procedure’, and ‘Laparoscopic bowel movement were no different between
surgery should only be undertaken for colorectal laparoscopic-assisted and open arms, but length
cancer as part of a randomised controlled clini- of hospital stay was 2 days shorter in the laparo-
cal trial’ [1]. scopic group, and for successful laparoscopic
In response, several groups took the initiative resections, hospital stay was 3 days shorter than
to undertake randomized controlled trials to for converted cases. Pathological analysis was
address the safety and efficacy of laparoscopic generally encouraging with high-quality surgery
surgery for colorectal cancer. The first group to observed in both laparoscopic-assisted and open
report their findings was the Clinical Outcomes arms, similar longitudinal resection margins and
of Surgical Therapy Study Group (COST), lymph node yields comparable to the best results
who published the results of a US multi-centre reported in other large series (median lymph node
randomized controlled trial (RCT) comparing yield: 12 (IQR: 8–17) lap vs. 13.5 (IQR: 8–19)
laparoscopic and open surgery for colon cancer open). However, the data did raise two important
in 2004 [2]. In their study of 872 patients recruited concerns. In patients undergoing laparoscopic-
from 48 centres and followed up for a median of assisted anterior resection, there was a higher rate
4.4 years, they showed benefits for laparoscopic- of circumferential resection margin involvement
assisted surgery, including less analgesic require- (CRM positivity: 12% lap vs. 6% open, 95% CI:
ment, shorter hospital stay (lap, 5 days; open, −2.1%, 14.4%, p = 0.19), although this was not
6 days; p < 0.001) and similar morbidity, mortality statistically significant. The other issue was a
25 Is Laparoscopic Rectal Surgery the Gold Standard? 251

1.0

0.9

0.8

0.7
Proportion surviving

0.6 Open

0.5 Laparoscopic

Conversion
0.4

0.3

0.2
P–value for Longronk test = 0.0017
0.1
P–value for Wilcoxon test = 0.0004
0.0
0 6 12 18 24 30 36

Number at risk Time from randomisation (months)


Open 280 255 234 210 195 178 163
Lap 343 323 307 290 266 244 217
Con 144 120 110 98 92 88 79

Fig. 25.1 Three-year overall survival by actual procedure: open, laparoscopic and converted. A significant difference
is observed with worse outcomes for the converted patients

high conversion rate of 34% for laparoscopic- laparoscopic operation, a fi nding that makes
assisted surgery. This was a reflection of both a intuitive sense.
learning curve effect, as demonstrated by a fall Disappointingly, CLASICC failed to show
in conversion rates throughout the study period, any difference in quality of life between laparo-
but also the increased technical difficulty associ- scopic-assisted and open surgery, a feature that
ated with rectal cancer as compared to colon has also been reported in other trials. The reason
cancer surgery. The main reasons for conversion for this is unclear as the earlier recovery demon-
were excessive tumour fixity or uncertainty of strated by shorter hospital stays is a consistent
tumour clearance (41% of conversions), obesity finding. It is perhaps related to the quality of life
(26%), anatomical uncertainty (21%) and inac- instruments used, which may be insensitive to
cessibility of tumour (20%). Importantly, conver- subtle differences in physical and psychological
sion appeared to have a negative influence on well-being, or to the timing of the investigation,
early post-operative morbidity and mortality, missing early benefits which occur within the
with a significant difference in those patients who first 2 weeks following surgery. One area of con-
were converted and those who completed a lap- cern, specifically related to laparoscopic rectal
aroscopic resection (Fig. 25.1). Three factors cancer surgery, has been the reporting of impaired
were subsequently found to be independent pre- post-operative bladder and sexual function [5].
dictors for conversion to open surgery, namely, Although this is a recognized complication fol-
body mass index, male sex and extent of tumour lowing open surgery, with reported rates of blad-
spread from the muscularis mucosa [4]. Thus, the der and sexual dysfunction between 0–15% and
obese male patient with a locally advanced rectal 10–35%, respectively, the incidence of sexual
cancer is much less likely to undergo a successful dysfunction following laparoscopic surgery in
252 D. Jayne and L. Khan

male patients in particular seems to be increased cancer [8]. Significant benefits in favour of the
(overall sexual function: difference lap vs. open laparoscopic approach were found for time to
−11.18; 95% CI: −10.94, −0.74; p = 0.063). stomal function, first bowel movement, feeding
Adequate training in laparoscopic total mesorec- solids and length of hospital stay. Additional
tal excision with attention to preservation of the benefit was found for those undergoing laparo-
pelvic autonomic nerves is therefore important if scopic abdominoperineal resection in terms of
optimal functional outcomes are to be achieved. decreased post-operative analgesic requirements
In 2006, NICE updated its guidance for lap- and less wound infections. No difference was
aroscopic colorectal cancer surgery, stating ‘lap- found between the laparoscopic and open groups
aroscopic (including laparoscopically assisted) in extent of oncological clearance. In 2008,
resection is recommended as an alternative to Anderson et al. reported a meta-analysis focusing
open resection for individuals with colorectal can- on the oncological outcomes following laparo-
cer in whom both laparoscopic and open surgery scopic rectal cancer resection [9]. Over 3,000
are considered suitable’ [6]. This statement was patients from 24 studies were compared for dif-
qualified by the caveat that ‘laparoscopic colorec- ferences in oncological outcome between laparo-
tal surgery should be performed only by surgeons scopic and open surgery. At 3 years, no significant
who have completed appropriate training in the difference was seen between the two treatment
technique and who perform this procedure often groups: radial margin positivity was 5% (lap)
enough to maintain competence’. The NICE state- versus 8% (open), overall survival was 76% (lap)
ment failed to distinguish between laparoscopic versus 69% (open) and local recurrence was 7%
surgery for colon and rectal cancer, with the two (lap) versus 8% (open). The authors concluded
entities being merged under the same ‘colorectal’ that there was no oncological difference between
umbrella. This was taken by many surgeons as a laparoscopic and open resections for primary rec-
‘green light’ to progress with laparoscopic rectal tal cancer. This conclusion accords with the
cancer surgery and indeed was informally embod- recently published long-term results from the
ied into the 2006 Department of Health initiative CLASICC trial [10]. Initial concerns regarding
to promote laparoscopic colorectal cancer surgery the higher rate of circumferential margin involve-
in the UK [7]. ment following laparoscopic anterior resection
A summary of the published data on laparo- failed to translate into a difference in local recur-
scopic rectal cancer surgery up to 2006 is shown rence, overall survival or disease-free survival.
in Table 25.1 and includes a total of 44 studies. The most recent evidence to emerge is that
The conversion rates range from 0% to 33% with from the COLOR II study. This multi-centre study
only two studies reporting rates greater than 20%. involving 30 centres across the world and recruit-
Three studies reported a significant reduction in ing 1,103 rectal cancer patients over the period
morbidity with the laparoscopic approach, with 2004–2010 has recently presented its initial results
11 studies showing no significant difference; no at the European Society of Surgical Oncology
study reported an increase in morbidity from (ESSO) conference in Stockholm in 2011 (unpub-
laparoscopic surgery. The duration of surgery lished). Patients undergoing laparoscopic rectal
was consistently longer with the laparoscopic cancer resection had less blood loss but longer
approach, with no obvious differences in short- operations than those undergoing open surgery.
term survival measures. Conversion to open operation was still observed
Two meta-analyses specifically addressing in 16.4% of cases. There was no difference in the
laparoscopic rectal cancer surgery have attempted circumferential or longitudinal resection margins
to collate the available evidence. In 2006, Aziz or the number of retrieved lymph nodes. However,
et al. reported their meta-analysis, including 20 laparoscopic surgery had the advantage of earlier
studies and 2,071 patients with 44% undergoing recovery of bowel function, less analgesic require-
laparoscopic and 56% open surgery for rectal ment and shorter hospital stay.
Table 25.1 Published series for laparoscopic rectal cancer surgery
25

Procedure 5-year Duration of Lymph nodes Hospital stay Local


n (%) Conversion survival (%) Morbidity (%) surgery (min) removed (days) recurrence (%) Follow-up
Study author Year Lap Open LAR APR rate (%) Lap Open Lap Open Lap Open Lap Open Lap Open Lap Open (months)
Barlehner 2005 194 – 92 8 1 88 – 20 – 174 – 25 – – – 4 – 46
Bretagnol 2005 144 – 100 0 14 – – 34 – – – 10 – – – 1.4 – 18
Dulucq 2005 218 – 100 0 12 67 – 25.6 – 138 – 25 – 6 – 6.8 – 57
Yamamoto 2005 31 – 100 0 0 – – – – 267 – – – 8 – – – –
Delgado 2004 220 – 80 19 20 – – 26 – 179 – 14 – 6.8 – 5.4 – 18
Law 2004 102 – 86.7 13.3 3 75 – 27 – 202 – 8 – 11.9 – – – 36
Leung 2004 203 200 100 0 23.2 75.3 78.3 19.7 22.5 190* 144* 11.1 12.1 8.2* 8.7* – – 49.2
Wu 2004 18 18 61 39 0 – – 5.6* 27.8* 189* 146* 7.8 8.2 – – – – –
Zhou 2004 82 89 100 0 – – – 6.1* 12.4* 120* 106* – – 8.1* 13.3* – – –
Anthube 2003 101 334 77 23 11 – – 11 25 218 218 12* 22* 14 20 – – –
Feliciotti 2003 81 43 74 26 12.3 62.5 60.6 – – – – 10.3 9.8 – 20.8 20.8 – 43.8
Morino 2003 100 – 100 0 12 63 – 36 – 250 – 12.8 – 16.6 – 4.2 – 45.7
Is Laparoscopic Rectal Surgery the Gold Standard?

Rullier 2003 32 – 100 0 9 – – 31 – 420 – 10 – 9 – 0 – –


Tsang 2003 44 – 100 0 0 – – 38.6 – 180 – – – 8 – 4.5 – 15
Baker 2002 28 61 – 100 8 – – – – – – 6 5 13* 18* 33 33 36
Chen 2002 8 – 100 0 – – – 25 – 210 – – – 13 – 0 – 14
Lezoche 2002 42 26 71 29 19 – – – – – – – – – 24.1 25 – 49.4
Pietrabissa 2002 16 – 100 0 0 – – 19 – 233 – – – 6.6 – – – –
Poulin 2002 80 – 65 35 18.25 72.1 – 40 – 200 – – – 6.5 – 3.75 – 31
Reis Neto 2002 32 – 100 0 3.1 – – – – – – 12.3 – 5.12 – 3.12 – 14
Scheibach 2002 308 – 60.8 39.2 6.1 – – 37.6 – 208 – 13 – – – 6.6 – 24.8
Yamamoto 2002 70 – 94.3 5.7 2.9 92.1 – 18.6 – – – 14.3 – 8 – 2.8 – 23
Chung 2001 5 – 100 0 – – – – – 208 – – – 10.6 – 0 – 13
Hartley 2001 21 22 71 29 33 – – 28.5 18 180* 125* 6 7 13.5 15 5 4.5 38
Hu 2001 20 25 100 0 0 – – 0 8 227* 146* – – 18.3 18 – – 8
Pasupathy 2001 11 22 100 0 – – – 18 9 97.5 90 – – 6.5 6 – – 12
Vithiananthan 2001 27 17 100 0 – – – 26* 28* – – 12 11 6.1* 11.1* – – –
Leung 2000 25 34 0 100 8 – – 48 62 215* 166* 10 12 16* 25.5* – – 28.3
Kockerling 2000 119 – 0 100 3.4 – – 34.4 – 226 – – – 21.6 – 9.5 – 16
253

(continued)
Table 25.1 (continued)
254

Procedure 5-year Duration of Lymph nodes Hospital stay Local


n (%) Conversion survival (%) Morbidity (%) surgery (min) removed (days) recurrence (%) Follow-up
Study author Year Lap Open LAR APR rate (%) Lap Open Lap Open Lap Open Lap Open Lap Open Lap Open (months)
Schiedeck 2000 259 – 60.6 39.4 6.6 – – 26.4 – 235 – 12.1 – 14.4 – 1.9 – 26
Wantanabe 2000 7 – 100 0 – – – – – 280– – – – – – – – –
450
Weiser 2000 21 – – – – – – – – 300 – – – – – 0 – –
Fleshmann 1999 42 152 100 0 21 54 60 76 50 234 209 9.7 7.9 7.4* 11.9* 21.4 – 55
Idani 1999 5 – 100 0 – – – 20 – 177 – – – 16.2 – 0 – 3
Schwander 1999 32 32 59.3 40.7 0 – – 31.3 31.3 281* 209* 13 13 15.3* 21.9* 3.1 0 32
Bokey 1997 10 – 2 8 – – – – – 342 – 10 – 16 – 0 – 29
Bruch 1997 20 – 30 70 10 – – – – 315 – 12 – 16 – 5 – 46
Goh 1997 20 20 100 100 0 – – – 90 73 20 19 5 5.5 – – 38
Ramos 1997 18 18 0 100 10 – – 44 66 229 208 11.1 7.8 7.4* 12.9* 6 17 20
Seow–Choen 1997 16 11 0 100 – – – – – – – 10 10 6.5* 8* 0 0 12 vs. 33
Wu 1997 14 – 0 100 14 – – 42 – 240 – – – – – 7 – 17
Darzi 1995 12 16 0 100 0 – – 33 56 195 104 9.5 6 11 17.5 – – –
Larach 1993 4 – 0 100 20 – – 40 – 323 – 4.75 – 12.3 – – – –
Tate 1993 11 14 100 0 – – – 45 29 205* 123* 10 13 12.3 14.3 – – –
Lap laparoscopic, LAR laparoscopic anterior resection, APR abdominoperineal resection
Data not reported marked with ‘–’. Values marked with ‘*’ were reported as being statistically significant (p £ 0.05)
D. Jayne and L. Khan
25 Is Laparoscopic Rectal Surgery the Gold Standard? 255

Two other trials of laparoscopic rectal cancer they can be clinically justified in terms of
surgery are currently still in progress: a US trial cost-effectiveness given the capital cost of the
of laparoscopic-assisted versus open resection robotic system, remains to be seen. But, there is
for rectal cancer (ClinicalTrials.gov Identifier: an inevitability that new technologies and tech-
NCT00726622) and the Australasian A La Cart niques will continue to be incorporated into
trial (www.australiancancertrials.gov.au). The laparoscopic practice, enabling current bound-
results of these trials are eagerly awaited. aries to be surpassed with benefits to both
patients and health-care providers.

Conclusion
25.3 Future Developments Is laparoscopic surgery the current gold stan-
in Laparoscopic Rectal dard? Based on the current available evidence,
Cancer Surgery it is still probably a little too premature to make
such a bold statement. A gold standard is a
The last two decades have seen much improve- level that can be attained by the majority. It
ment in laparoscopic techniques and technol- cannot be judged on the results of personal
ogy. The use of modified approaches, such as series from enthusiastic centres of excellence,
medial-to-lateral dissection, combined with bet- but rather we must await the results of the large
ter instrumentation has facilitated the perfor- randomized clinical trials that are still yet to
mance of laparoscopic surgery, making it safer report. But there is no reason to believe that
and encouraging wider adoption. It is likely that they will show any real difference from that
this trend will continue, driven by increased already observed, i.e. the laparoscopic approach
patient demand, pressure from commercial for rectal cancer produces oncological results
sources and greater acceptance by health-care similar to open surgery, but with the advantage
providers. Current estimates of penetration for of better short-term outcomes. If this proves to
laparoscopic colorectal surgery are around be the case, then it will be safe to conclude that
30% in the UK and Australasia in 2011, so there a new gold standard for rectal cancer surgery
remains the potential for considerable growth, has been established.
particularly in rectal cancer. However, there
is probably a ceiling beyond which laparo-
scopic surgery in its current form cannot extend.
This is governed by the inherent limitations of References
laparoscopy, namely, the fulcrum effect of the
1. National Centre for Clinical Excellence (2000)
ports, two-dimensional imaging and limited Technology Appraisal Guidance No 17: Guidance on
instrumentation. The introduction of robotics the use of laparoscopic surgery for colorectal cancer.
into surgical practice offers to overcome at least www.nice.org.uk. Accessed March 2012
some of these limitations. Currently, there is 2. The Clinical Outcomes of Surgical Therapy Study
Group (2004) A comparison of laparoscopically
only one commercially available robotic system, assisted and open colectomy for colon cancer. N Engl
the da Vinci® surgical system (Intuitive Surgical, J Med 350:2050–2059
Sunnyvale, CA). Several personal series and one 3. Guillou PJ, Quirke P, Thorpe H, Walker J, Jayne DG,
small randomized controlled trial testify to the Smith AMH, Heath RM, Brown JM, for the MRC
CLASICC trial group (2005) Short-term end-points
safety and feasibility of robotic-assisted rectal of conventional versus laparoscopic-assisted surgery
cancer surgery, with initial indications of short- in patients with colorectal cancer (MRC CLASICC
term benefit with low conversion rates to open trial): multicentre, randomised controlled trial. Lancet
surgery and reduced rates of circumferential 365:1718–1726
4. Thorpe H, Jayne DG, Guillou PJ, Quirke P, Copeland J,
resection margin involvement. Whether these Brown JM, for the Medical Research Council Con-
indicators bear out in future long-term studies ventional versus Laparoscopic-Assisted Surgery in
and randomized controlled trials, and whether Colorectal Cancer Trial Group (2008) Patient factors
256 D. Jayne and L. Khan

influencing conversion from laparoscopically assisted 8. Aziz O, Constantinides V, Tekkis PP, Athanasiou T,
to open surgery for colorectal cancer. Br J Surg 95: Purkayatha S, Paraskeva P, Darzi AW, Heriot AG
199–205 (2006) Laparoscopic versus open surgery for rectal
5. Jayne DG, Brown JM, Thorpe H, Walker J, Quirke P, cancer: a meta-analysis. Ann Surg Oncol 13:413–424
Guiloou PJ (2005) Bladder and sexual function fol- 9. Anderson C, Uman G, Pigazzi A (2008) Oncological
lowing resection for rectal cancer in a randomized outcomes of laparoscopic surgery for rectal cancer: a
clinical trial of laparoscopic versus open technique. systematic review and meta-analysis of the literature.
Br J Surg 92:1124–1132 J Cancer Surg 34:1135–1142
6. (2006) NICE Technology Appraisal Guidance 10. Jayne DG, Thorpe H, Copeland J, Quirke P, Brown
105: Laparoscopic surgery for colorectal cancer. JM, Guillou PJ (2010) Five year follow-up of the
www.nice.org.uk/TA105. Accessed March 2012 Medical Research Council CLASICC trial of laparo-
7. LAPCO: National Training Programme in Laparo- scopically assisted versus open surgery for colorectal
scopic Colorectal Surgery. www.lapco.nhs.uk. Acces- cancer. Br J Surg 97:1638–1645
sed March 2012
Is a Diverting Stoma Always
Necessary for a Low Anterior 26
Resection of a Rectal Cancer?

Geerard L. Beets

Contents 26.1 Background: Anastomotic


26.1 Background: Leakage
Anastomotic Leakage ................................... 257
26.2 Rationale and Evidence
The main surgical complication of a low anterior
for Diverting Stoma ...................................... 258 resection of the rectum with a low anastomosis is
pelvic sepsis. Pelvic sepsis is most often due to
26.3 Routine or Selective
Diverting Stoma? .......................................... 258 poor anastomotic healing and can have different
manifestations: anastomotic leakage, presacral
26.4 Ileostomy or Colostomy? .............................. 258
abscess, rectovaginal fistula, etc. The symptoms
References ................................................................. 259 can occur as early as a couple of days after the
operation or as late as several weeks after dis-
charge, and the severity can range from minor to
life threatening. The reported incidence of pelvic
sepsis is variable, ranging from 2% to as high as
39%. A large part of this variability is most likely
due to a lack of standardized definition, a lack of
standardized reporting and inclusion of different
types of patients and operations. The newly
formed International Study Group of Rectal
Cancer recently proposed a simple and practical
grading system that includes abscesses near the
anastomosis and that is focused on the conse-
quences for the patient [1]. Grade A is basically
an asymptomatic leak, Grade B a leak or abscess
that can be managed without relaparotomy and
Grade C one that requires a relaparotomy. The
reported mortality after a leakage/pelvic sepsis
ranges from 2% to 24%, with the variability most
likely related to the previously mentioned lack of
standardization in definition. Additional to the
G.L. Beets immediate morbidity and mortality, there are
Department of Surgery,
also long-term functional consequences for the
Maastricht University Medical Centre,
Maastricht, The Netherlands patient. Many patients will suffer from poor
e-mail: g.beets@mumc.nl anorectal function due to ongoing low-grade

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 257


DOI 10.1007/978-3-642-25005-7_26, © Springer-Verlag Berlin Heidelberg 2012
258 G.L. Beets

infection or pelvic fibrosis, and a substantial num- reversal and has its own morbidity in the form of
ber of patients will end up with some form of stoma-related complications and the decreased
definitive colostomy. Some older reports also sug- quality of life that is inherently associated with a
gested a worse long-term oncological outcome, stoma. A valid question is, therefore, whether all
with a higher rate of local recurrence. A recent patients should be given a diverting stoma or only
study on pooled data from five large randomized those at high risk for an anastomotic problem.
trials on rectal cancer showed a 5-year overall sur- Many papers have described risk factors for anas-
vival rate of 66% in patients who had an anasto- tomotic leakage, and the most important risk fac-
motic leak, compared to 74% in patients who had tor without question is the height of the
an uneventful anastomotic healing [2]. This 8% anastomosis: the lower the anastomosis, the
long-term survival difference was, however, not higher the chance of leakage. When the entire
due to a higher relapse rate but to a higher mortal- mesorectum is removed in a total mesorectal
ity in the postoperative period. excision, the anastomosis is always very low and,
therefore, at increased risk for leakage. Other risk
factors that are often mentioned in reports are
26.2 Rationale and Evidence peripheral vascular disease, male patients, meta-
for Diverting Stoma static or advanced disease, smoking, low albu-
min, diabetes, high age, blood transfusion and
A diverting loop ileostomy or colostomy has long technical mishaps during the operation. It is
been used by surgeons with the intention to both unclear whether or not neoadjuvant therapy
reduce the incidence and the severity of anasto- increases the risk for anastomotic problems. In
motic leakage. By decreasing the faecal load at the large randomized trials, the rate of anasto-
the site of the leakage, it is hoped that less bacte- motic leakage is generally the same in patients
rial contamination results in a less severe clinical with and without neoadjuvant therapy, while
course or even in a complete subclinical leakage. multivariate analyses of large cohort series some-
A recent Cochrane review of six randomized tri- times suggest an increased risk. In practice, it
als evaluating a routine diverting ileostomy or remains difficult to predict which group of
colostomy versus no diverting stoma after low patients is at risk, and given the evidence on the
anterior resection for rectal cancer shows a two- routine use of a diverting stoma in the random-
thirds reduction of clinical leak rate from 19.6% ized trials, it seems prudent to use a routine rather
to 6.3% and a three-fourths reduction in urgent than a selective policy.
reoperation rate from 16.1% to 3.9% [3]. The
reduction in mortality was not significant. Two
earlier reviews that included also nonrandomized 26.4 Ileostomy or Colostomy?
series basically had come to the same conclusion,
with the additional finding that in the nonran- An additional question is which diverting stoma
domized series, the mortality was significantly is to be preferred, ileostomy or colostomy. This is
lower in the patients who were given a diverting a topic for discussion among colorectal surgeons,
stoma [4, 5]. and strong personal opinions are often expressed.
It is more a matter of personal choice than a
choice based on evidence. There is a Cochrane
26.3 Routine or Selective Diverting review of five randomized trials and a meta-anal-
Stoma? ysis that adds seven observational studies. In the
Cochrane review, the only significant finding is a
Although a temporary diverting stoma dimin- higher rate of stoma prolapse with a colostomy
ishes the incidence and severity of anastomotic [6]. The other meta-analysis confirms this and
leakage, it does require a second procedure for also found an increased risk of infectious
26 Is a Diverting Stoma Always Necessary for a Low Anterior Resection of a Rectal Cancer? 259

complications with the colostomy [7]. The ileos- International Study Group of Rectal Cancer. Surgery
tomy, on the other hand, was more associated 147(3):339–351
2. den Dulk M, Marijnen CA, Collette L et al (2009)
with dehydration and late intestinal obstruction Multicentre analysis of oncological and survival
after closure. There is, therefore, no compelling outcomes following anastomotic leakage after rectal
evidence to prefer one to the other. cancer surgery. Br J Surg 96(9):1066–1075
3. Montedori A, Cirocchi R, Farinella E, Sciannameo F,
Abraha I (2010) Covering ileo- or colostomy in
Conclusion anterior resection for rectal carcinoma. Cochrane
A diverting stoma decreases both the incidence Database Syst Rev 12(5):CD006878
and the consequences of anastomotic failure 4. Huser N, Michalski CW, Erkan M et al (2008)
after a low anterior resection for rectal cancer. Systematic review and meta-analysis of the role of
defunctioning stoma in low rectal cancer surgery. Ann
The available evidence supports a routine Surg 248(1):52–60
diverting stoma for all patients rather than a 5. Tan WS, Tang CL, Shi L, Eu KW (2009) Meta-
selective approach. There is no solid evidence analysis of defunctioning stomas in low anterior
favouring either ileostomy or colostomy. resection for rectal cancer. Br J Surg 96(5):462–472
6. Guenaga KF, Lustosa SA, Saad SS, Saconato H, Matos
D (2007) Ileostomy or colostomy for temporary
decompression of colorectal anastomosis. Cochrane
References Database Syst Rev (1):CD004647
7. Rondelli F, Reboldi P, Rulli A et al (2009) Loop ileo-
1. Rahbari NN, Weitz J, Hohenberger W et al (2010) stomy versus loop colostomy for fecal diversion after
Definition and grading of anastomotic leakage follow- colorectal or coloanal anastomosis: a meta-analysis.
ing anterior resection of the rectum: a proposal by the Int J Colorectal Dis 24(5):479–488
Will Extralevator Abdominoperineal
Excision Become the New Gold 27
Standard?

Ingrid Martijnse, Nicholas West, Phil Quirke,


Richard Heald, Cornelius J.H. van de Velde,
and Harm Rutten

Contents 27.6 How Should Surgery in the Supine


Position Be Performed? ............................ 265
27.1 Does a Patient After Abdominoperineal
Excision Have a Worse Prognosis 27.7 What Are the Advantages and Pitfalls
Than After (Low) of Performing an Extralevator APE
Anterior Resection? .................................. 261 in the Prone Position? ............................... 266

27.2 How Does Preoperative Imaging Result 27.8 How Should Surgery in the Prone
in Better Surgery?..................................... 262 Position Be Performed? ............................ 267

27.3 Is Selective Use of Preoperative 27.9 Which Surgical Technique Should Be


Radio(−Chemo)Therapy Effective Performed? ................................................ 267
in Patients with Low Rectal Cancer? ...... 262 27.10 What Role Do Pathologists Play
27.4 How to Decide on Which Surgical in the Treatment of Rectal
Procedure to Perform? ............................. 263 Cancer Surgery? ....................................... 269

27.5 What Are the Advantages and 27.11 Is a New Standard for
Limitations of Performing an Extralevator Abdominoperineal Excision
APE in the Supine Position? .................... 264 Necessary? ................................................. 270

References ............................................................... 271

I. Martijnse
Colorectal Surgery, Catharina Hospital Eindhoven,
The Netherlands
N. West • P. Quirke
Pathology and Tumour Biology, Leeds Institute of
27.1 Does a Patient After
Molecular Medicine, University of Leeds, Abdominoperineal Excision
United Kingdom Have a Worse Prognosis Than
R. Heald After (Low) Anterior Resection?
Pelican Cancer Foundation, Basingstoke,
United Kingdom The introduction of TME surgery led to the real-
C.J.H. van de Velde ization that the quality of surgery significantly
Colorectal Surgery, Leiden University Medical Center, influences the prognosis of rectal cancer patients
Leiden, The Netherlands
and has resulted in a huge improvement in sur-
H. Rutten (*) gery and its outcome [1]. On a population-based
Colorectal Surgery, Catharina Hospital Eindhoven,
level, an improved survival of more than 10%
Michelangelolaan 2 5623EJ Eindhoven,
The Netherlands was realized in countries adopting TME as the
e-mail: harm.rutten@cze.nl standard surgical technique [2, 3].

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 261


DOI 10.1007/978-3-642-25005-7_27, © Springer-Verlag Berlin Heidelberg, 2012
262 I. Martijnse et al.

However, patients with low rectal cancer did [19, 20] In the MERCURY study, MRI could pre-
not seem to benefit from this improvement [4, 5]. dict extramural tumor spread to within 0.5 mm
After more than 20 years of widespread TME [21]. Even the current multislice CT does not
surgery, the difference between patients undergo- have the spatial resolution that is needed in low
ing APE or AR still persists [6–8]. It has been rectal cancer staging [22, 23]. Even though some
demonstrated that it is not the tumor biology question the use of MRI in low rectal cancer [24],
which heralds a worse outcome, but an increased most physicians agree that MRI is superior to
number of positive circumferential resection define the surgical planes for resection both in
margins (CRM) and iatrogenic perforations, both mid and low rectal cancer [25–27]. In modern
of which are avoidable and confer a poor progno- rectal cancer surgery, it is essential that the sur-
sis [9–13]. The latest analysis of the Dutch TME geon knows whether the tumor infiltrates through
study shows that when radical surgery is per- the muscularis propria in the anal canal to avoid
formed, APE patients share the same prognosis an intersphincteric resection, which would lead
as AR patients [14]. to an involved or threatened circumferential
Besides the change in surgical approach, the margin. In locally advanced rectal cancer in the
introduction of TME surgery has been associated lower rectum, MRI imaging to evaluate the rela-
with improved preoperative staging, proper selec- tion of the tumor to the levator muscles is equally
tion of preoperative treatment, and feedback on important for the surgical planning, and the com-
the quality of surgery by pathologists. parison of MRI before and after neoadjuvant
treatment may lead to different surgical options
[28]. A recent manuscript demonstrated that MRI
27.2 How Does Preoperative was able to predict long-term outcome in low
Imaging Result in Better rectal tumors, proving its validity in staging in
Surgery? rectal cancer [29].

At the beginning of the TME era during which


the anatomical planes became increasingly 27.3 Is Selective Use of Preoperative
important for surgery, preoperative MRI was not Radio(−Chemo)Therapy
widely used for local staging. Current guidelines Effective in Patients with Low
are based on patients often operated upon with Rectal Cancer?
only a rectal examination and endoscopy. Older
CT scans could not differentiate between the cru- In the Dutch TME study, preoperative short
cial anatomical layers and the mesorectal fat in course radiotherapy did not seem effective in
the lower pelvis and therefore did not help the reducing local recurrence rates in patients under-
visualization of the margins of the tumor [15, going APE. Preoperative radiotherapy did not
16]. Ultrasound, which was often used, discrimi- compensate for positive CRM, which were found
nates between the bowel layers and helps to diag- in 30% of low rectal cancer patients [30].
nose the earlier T stages, but performs poorly in However, the latest update did show that patients
the identification of the mesorectal fascia [17, with negative margins after APE responded well
18]. Furthermore, ultrasound is not reproducible to preoperative short course irradiation [14].
and does not provide the surgeon with a roadmap Similar findings apply to the MRC CR07 study
for surgery in anything other than pT1 cases, nor which showed higher positive CRM rates after
does it help to identify patients that require pre- APE than AR and subsequent worse outcome
operative treatment. The introduction of MRI as [31]. Reassuring are the recent findings from a
the principal local staging imaging technique Swedish population-based study, which also sug-
made it possible to identify the mesorectal fascia gested an effect of preoperative radiotherapy on
and the relation of the tumor to this fascia. lower rectal cancers and APE patients [32].
Patients at risk for an irradical resection can be Better local staging helps to identify patients
identified, and treatment plans could be adjusted. at risk for CRM. Long-course preoperative
27 Will Extralevator Abdominoperineal Excision Become the New Gold Standard? 263

Fig. 27.1 Cylindrical


extralevator specimen from
William Ernest Miles and
two important steps in the left
lateral semiprone perineal
phase: above, wide approach
with cutting of the coccyx;
below, everted specimen
and dissection of the ventral
side [41]

radio(−chemo)therapy may downstage and comparable to the discussion in the 1930s. In


downsize the tumor, which in combination with those days, Miles was a proponent of a one-stage
a better delineation of the tumor in the lower pel- abdominoperineal resection. Starting with the
vis facilitates a better surgical resection [33–40]. abdominal phase, the pelvic colon was mobilized
Most national guidelines have therefore included and a stoma created. After closure of the abdo-
(radio-)chemotherapy in the preoperative treat- men, the patient was turned into a right-sided
ment plan of rectal cancer. In a recent Dutch semiprone position. A generous incision was
audit, the number of CRM + margins in 2009 and made encircling the anus and extending up to the
2010 was reduced to 14% for tumors between 0 sacrococcygeal joint. The coccyx was then
and 5 cm from the anal verge (paper submitted). transected and the levator muscle cut near its lat-
Therefore, selective use of preoperative treat- eral insertion. This access allowed eversion of the
ment can influence T stage and circumferential rectosigmoid and dissection of the remaining con-
resection margins which are the most important nections between the internal genital organs and
prognostic variables in low rectal cancer and sub- the rectum (Fig. 27.1). The ischiorectal fat was
sequently result in improved outcome. removed en bloc with the specimen. Dealing
mostly with advanced cases, he had noticed tumor
deposits in the ischiorectal fat and, as we know
27.4 How to Decide on Which now, mistook these for lymph nodes [41]. Gabriel
Surgical Procedure to Perform? was a proponent of the perineal first approach.
This approach also included a wide resection dur-
Being well informed after local imaging and pos- ing the perineal phase as was custom in those days
sibly after downstaging the tumor, the surgical for a perineal-only approach [42]. The patient was
resection remains the most important prognostic placed in prone position and turned for the abdom-
factor with at least a 1 mm margin. Currently there inal phase [43, 44]. However, despite the efforts
is a debate on which surgical technique is the best, of these pioneers of wide perineal dissection, the
264 I. Martijnse et al.

Lloyd-Davies method of operating patients with


the legs in stirrups in supine position which
allowed a simultaneous perineal and abdominal
access by two teams became the most popular
technique and was used worldwide [45].
In those days, every procedure started as an
exploratory laparotomy with no preoperative infor-
mation on local tumor infiltration or metastatic dis-
ease in contrast to the modern colorectal surgeon.
The resection is now based on the anatomical rela-
tionship of the tumor to its surroundings. The basis
for a successful resection is a detailed understand-
ing of the basic pelvic anatomy, not only to achieve
a radical resection, but also to avoid unnecessary
damage to the patient [46, 47].
In TME surgery, the plane of dissection fol-
lows the mesorectal fascia, which envelopes the
mesorectum and terminates at the pelvic floor,
where visceral and parietal fascia blend and con- Fig. 27.2 Illustrating that at different levels of the rectum,
tinue in the intersphincteric plane in close contact small extensions of tumor into the mesorectum have different
consequences for the resection. The red arrow indicates the
with the internal anal sphincter. For low rectal extralevator route, necessary to avoid breaching of the plane
cancer, an intersphincteric resection is only safe between distal mesorectum and parietal fascia
in early stage rectal cancer. Due to the reduction
in mesorectal fat at the height of the levators and
its disappearance in the sphincter area, smaller tumors with fixation to pelvic structures and where
degrees of tumor spread will result in increased the pelvic floor muscles are often infiltrated by
numbers of positive margins (Fig. 27.2). Fortu- tumor, this bimodal approach can be very useful.
nately, the levator muscles act like a bowl in A disadvantage of this technique would be the
which the distal rectum and mesorectum com- limited surgical space and reportedly difficult expo-
fortably rest. Therefore, distal tumors extending sure of the anatomical structures. However, with
into and beyond the mesorectal fascia or inter- the correct positioning of the patient and placing
sphincteric plane can be removed en bloc with the legs in movable stirrups with individual drap-
the external sphincter-levator muscle complex ing, a wide range of movement and good exposure
via an extralevator approach [48, 49]. of the perineum is obtainable. Lloyd-Davies origi-
nally described the position [45]. This is also the
position when APE surgery is compared to AR.
27.5 What Are the Advantages The perineal phase is commenced after finishing
and Limitations of Performing the abdominal dissection. One reason for explain-
an Extralevator APE ing the inferior results is that the dissection along
in the Supine Position? the mesorectal fascia is carried too far down, that is,
up to the level where mesorectal and parietal fascia
Performing APE in the classical supine position is blend. The protecting levator muscles are dissected
preferred by several surgeons since it avoids the from the distal mesorectum exposing the tumor
unpopular and time-consuming turning of the resulting in typical waist formation at the tumor
patient during the operation. Furthermore, the onco- site with consequently a higher chance on positive
logical advantage of the supine position is that the margins. Waist formation was clearly illustrated by
tumor can be assessed from both the abdominal and Salerno by comparing the pathology specimens
perineal incision. Especially in locally advanced and the preoperative MRI. Tumors near the
27 Will Extralevator Abdominoperineal Excision Become the New Gold Standard? 265

no suspicion of intra-abdominal metastases. It is


important to realize that this phase is the most
important one. Good visualization, focused and
standardized dissection, and knowledge of the
pelvic anatomy are essential to achieve radical
surgery.
The legs are lifted and the perineal stage
commences. An incision is made around the
anus at the transition of anal to perineal skin.
The external sphincter is covered by the deep
perineal fascia, which is a continuation of the
fascia covering the levator ani muscles. On the
lateral sides, this fascia can easily be followed
until the lateral attachment of the levator ani to
the arcuate line. At this lateral level, the levator
ani can be cut, exposing the mesorectum
(Fig. 27.4). Dorsally, the anococcygeal ligament
must be transected. At this point, a U-shaped
incision is used to transect the levator ani mus-
cle. Dissection of the anterior aspect of the
external sphincter must not be started until this
Fig. 27.3 Illustrating how digital hooking of the levator point of the procedure is reached. The interfas-
may lead to waist formation of the specimen [51] cial presacral space can be opened up as far as
the level of S3. The perineal body in male and
junction of the mesorectal and visceral fascia were female is the central tendinous anchor of the
at highest risk for an incomplete resection [25, 50]. anterior perineal muscles. Posterior of the
The perineal dissection technique, as demonstrated perineal body, the urogenital diaphragm muscles
in Fig. 27.3, further leads to waist formation. Many and the anterior levator muscles blend with the
surgeons were taught to hook their finger behind extension of the mesorectal fascia and outer lon-
the levator and then transect the levator from poste- gitudinal layer of the muscularis propria of the
rior to anterior. However, by hooking the finger as rectum and constitute the anterior part of the
shown, the levator is incised too medially and is anal sphincter. This segment lacks an inter-
bluntly mobilized from the tumor leading to tumor sphincteric dissection plane. Dissecting from
exposure and positive margins [51]. the deep anterior levator ani muscles toward the
These pitfalls can be avoided. Several authors more superficial perineal body, first the pub-
have shown that they can avoid a high positive orectalis is cut at the level of the prostate or cer-
CRM rate and demonstrate that quality of sur- vix where Denonvilliers’ fascia is identified and
gery, or rather the lack of quality, does not need removed with the specimen (Fig. 27.4). The dis-
to be a prognostic variable [52–56]. section continues by transecting the muscle
attachments to the longitudinal layer of the mus-
cularis propria. Pushing the anteriorly fixed rec-
27.6 How Should Surgery tal specimen down facilitates this dissection.
in the Supine Position This results in tension on these muscle bundles
Be Performed? and avoids a false route leading into the rectum.
Unlike laterodorsally, there is no true dissection
Surgery can commence with either the abdomi- plane on the anterior side. The anterior part of
nal or the perineal phase; however, it is advised the external sphincter is tightly connected to the
to start with the perineal phase first when there is perineal body and the urogenital diaphragm by
266 I. Martijnse et al.

Fig. 27.4 Perineal phase in supine position

intertwining muscular bundles anchoring the can start. The focus on the perineal dissection
anus in the perineum. At this point, the abdomi- and its standardization by beginning perineal dis-
nal phase can commence. section laterally and dorsally leaving the levator
Mobilization of the rectosigmoid and lympho- muscle on the specimen and then transecting the
vascular pedicle occurs according to the princi- anterior attachments in the described technique
ples of TME. During dissection, the mesorectal helps to avoid iatrogenic tumor perforations and
fascia is left intact and care is taken to avoid dam- involved margins.
age to the hypogastric nerve. On the anterior side
when the peritoneal flap is opened and
Denonvillier’s fascia is followed until the vesi- 27.7 What Are the Advantages
cles, sufficient dissection has been performed. and Pitfalls of Performing
On the posterior side, the correct plane is easy to an Extralevator APE
find. The final phase is a sharp dissection of the in the Prone Position?
remaining attachments through the abdominal
phase and removal of the specimen. Extralevator excision in the prone position is
When the surgeon does decide to start with the becoming increasingly popular. It is believed
abdominal phase prior to perineal dissection, the that this technique is easier to learn and that in
dissection must be limited. To avoid coning in this position superior exposure of the anatomy
toward the tumor, dissection should stop above can be achieved. Furthermore, it has been
the levator muscles after which the perineal phase hypothesized that the blood loss is less due to the
27 Will Extralevator Abdominoperineal Excision Become the New Gold Standard? 267

higher position of the venous plexus. However, lenging anterior side where the specimen remains
there is very limited evidence to support either attached. By rolling the specimen from side to
the prone or the supine approach. They both have side and applying gentle traction on the speci-
their advantages and disadvantages, and the men, the dissection continues along the correct
experience of the surgeon on either technique plane. From the perineal body onward, the con-
will determine the outcome. Standardization of necting muscles are divided. The most challeng-
technique is considered to be the most important ing point during the dissection is the 2 cm after
factor for radical surgery. cutting these connecting muscles before reaching
The main limitation of this technique is that a the prostate. This is where the ureter is in very
bimodal approach is not possible. In advanced close proximity to the plane of dissection, and
tumors, it is impossible to assess and dissect from care must be taken to avoid taking a false route.
both the abdominal and the perineal phase. The hypogastric nerves are identified and spared.
In front of the prostate, the plane of the fascia of
Denonvillier should be followed until the vesicles
27.8 How Should Surgery are reached. The specimen can now be removed.
in the Prone Position Excision of the ischiorectal fat is only neces-
Be Performed? sary in exceptional cases, for example, when
tumor infiltrates through the external sphincter-
Surgery always commences with the abdominal levator muscle funnel or when the tumor has per-
phase with either a conventional laparotomy or a forated or fistulated. Furthermore, when the anal
laparoscopic approach. Inspection of the abdo- skin is involved, a more radical excision should
men is performed, and the sigmoid and the rec- be considered. More ischioanal fat should then be
tum are mobilized along the TME planes. Once removed to achieve radical excision of the tumor,
the hypogastric nerves and the vesicles have been but the surgical technique remains the same.
identified on the anterior side, dissection from the
abdominal phase must be stopped. The incisions
are sutured and the patient is turned into prone 27.9 Which Surgical Technique
position. Care must be taken in positioning the Should Be Performed?
patient to avoid pressure ulcers.
Normal surgical preparation with disinfection
and covering with sterile drapes is performed. Recently the Cleveland Clinic published a man-
A rhombus-like incision is made around the anal uscript comparing the supine and prone posi-
skin and extended proximally above the sacro- tion. The type of surgery chosen was based on
coccygeal joint (Fig. 27.5). With diathermy, dis- the preference of the surgeon, which resulted in
section is performed on the lateral sides, and two comparable groups with similar patient
limited dissection is performed on the anterior characteristics. CRM involvement rates were
(perineal body) and the posterior side. Posteriorly, 14.6% and 9%, and the iatrogenic rectal perfo-
dissection is performed behind the os coccygis in ration rates were 2.4% and 4.6% for supine APE
order to remove this “en bloc” with the rectal and prone APE, respectively, and no difference
specimen to create extra space. Lateral dissection in oncological outcome was noticed [54].
takes place along the levator ani muscle until the Recently a paper from a Dutch referral center
mesorectum is reached. On the posterior side, the for advanced rectal cancer showed that stan-
dissection plane is relatively easy to find, and this dardization of the supine APE technique resulted
is followed until the mesorectum is encountered. in a perforation rate of 0.7% and CRM + (<1 mm)
The specimen is then everted from the perineal of 14%, keeping in mind that two-thirds of their
wound. Care must be taken to avoid tearing the patients had T4 tumors [56]. Excellent results
specimen by pulling with too much force. This have also been demonstrated with the prone
creates more space and a better view on the chal- APE technique [48, 57].
268 I. Martijnse et al.

Fig. 27.5 Perineal phase in the prone position

An increased number of radical resections are vs. standard APE were 4.1% vs. 10.4% (relative
achieved by the en bloc removal of the distal rec- risk reduction 60.6%, p = 0.004) and 9.6% vs.
tum and levator muscles, therefore highlighting 15.4% (relative risk reduction 37.7%, p = 0.022),
the need of an extralevator excision more than respectively. The local recurrence rate was 6.6%
the positioning of the patient. A recent review vs. 11.9% (relative risk reduction 44.5%, p < 0.001)
of more extended perineal resections showed for the two groups [58].
that the rate of inadvertent bowel perforation These results set a new standard for APE.
and the rate of CRM involvement for extended Colorectal surgeons have to know their own results
27 Will Extralevator Abdominoperineal Excision Become the New Gold Standard? 269

Table 27.1 Three-point grading system for assessment of the plane of mesorectal dissection in TME/anterior resection
specimens for rectal cancer
Grade Short description Long description
Mesorectal plane Good surgery Intact smooth mesorectal surface with only minor irregularities. Any
defects must be no deeper than 5 mm. No coning of the specimen
distally. Smooth CRM on slicing
Intramesorectal plane Moderate surgery Moderate bulk to mesorectum but irregularity of the mesorectal
surface. Moderate distal coning. Muscularis propria not visible with
the exception of levator insertion. Moderate irregularity of CRM
Muscularis propria plane Poor surgery Little bulk to mesorectum with defects down onto the muscularis
propria and/or very irregular CRM. Includes infraperitoneal
perforations

and should strive that the quality of surgery is no tion rates. It helps to improve the quality of the
longer a prognostic factor. specimen produced by giving an objective feed-
Secondary endpoints such as nerve preserva- back to the surgeon.
tion and other postoperative complications will Within the Dutch TME and the UK CR07
eventually make the difference besides the ability trial, 57% and 52% of the anterior resection spec-
to teach and implement the principle of extraleva- imens were graded as being in the mesorectal
tor resection. plane, 19% and 34% were intramesorectal, and
24% and 13% were muscularis propria, respec-
tively [31, 62]. The CR07 trial showed that
27.10 What Role Do Pathologists Play through pathological feedback, CRM involvement
in the Treatment of Rectal rates fell to as low as 8%.
Cancer Surgery? For abdominoperineal excision, an addi-
tional grading system for the quality of the dis-
Pathologists have played an essential role in the section around the sphincters has been
improvement in rectal cancer outcomes. Besides developed. Besides grading the mesorectum,
tissue diagnosis and staging, the pathologist plays the plane of dissection around the anal sphinc-
a central role in the multidisciplinary approach. ters should also be graded as shown in
They give feedback on preoperative radiology Table 27.2. The levator ani muscle is left
and the quality of surgery. Furthermore, an evalu- attached to the mesorectum during extralevator
ation of the effectiveness of the administered excision that creates a more cylindrical speci-
neoadjuvant therapy can be made. This facilitates men. The extra tissue around the tumor results
internal auditing as well as a continued education in an increased radical resection rate and less
and improvement of all members of the MDT. perforations of the tumor (see Fig. 27.6). The
The involvement of the CRM has proven to be assessment of the plane of APE surgery is based
the most important prognostic factor for local on similar principles like the anterior resec-
recurrence within the pelvis. Pathologic feedback tions. Any defects must be no deeper than
during the era of TME has resulted in a significant 5 mm, and there should be no waisting of the
increase of radical resections [59–61]. specimen. The APE specimens should receive
A three-point grading system of the macro- two grades, namely one grade for the sphincter
scopic specimen was developed for the MRC and another for the mesorectum.
CLASSIC and CR07 trials and has been shown to In order for the pathologist to evaluate and audit
predict CRM involvement [31]. The recom- the quality of the MDT, their reports must be stan-
mended mesorectal grading system is described dardized. The specimen should be examined and
in Table 27.1. graded accordingly before fixation and slicing.
This grading system reports the plane of Photographs of the specimen provide a permanent
dissection besides the CRM status and perfora- record of the surgical quality. However, the macro-
270 I. Martijnse et al.

Table 27.2 Sphincter/levator grading according to the plane of surgery as assessed at the time of pathological dissec-
tion noting the presence and extent of any defects below the mesorectum in the sphincter/levator muscle complex
Grade Short description Long description
Extralevator plane Good surgery The specimen has a cylindrical shape due to the presence of levator
muscle removed en bloc with the mesorectum and sphincters. Any
defects must be no deeper than 5 mm. No waisting of the
specimen. Smooth CRM on slicing
Sphincteric plane Moderate surgery The specimen is waisting, and the CRM in this region is formed by
the surface of the sphincter muscles which have been removed intact
Intramuscular/submucosal Poor surgery The specimen is waisted and includes deviations into the sphincter
plane/perforation muscle complex, submucosa, and complete perforations

a b c

Fig. 27.6 Three APE specimens including an extralevator APE (a), a standard APE with classic surgical waist (b), and
a standard APE with a large anterior perforation (c)

scopic evaluation of this assessment of the quality 27.11 Is a New Standard


of surgery is rarely performed. The grading system for Abdominoperineal
is essential to improve surgery and to evaluate the Excision Necessary?
preoperative radiology and the effectiveness of
(neo)adjuvant treatment in relation to the predicted A recent update of the Dutch Surgical Colorectal
circumferential tumour margin. Audit of 2009 and 2010, covering more than 90%
The identification of the importance and of APE surgery in the Netherlands, demonstrated
frequency of CRM involvement, the importance that 14% of patients had a positive CRM (<1 mm)
of planes of surgery, and the identification of the which was significantly higher than after AR
major issues involved in APE for low rectal can- (11%) (data presented at ECCO Stockholm).
cer enables surgeons to evaluate the quality of This percentage of margin involvement has
surgical dissection and radiologists to audit the significantly improved when compared to the
accuracy of preoperative staging. It can be con- 30% of positive CRM in APE patients in the
cluded that surgery has developed throughout the Dutch TME study, which accrued patients
years by looking at the grades in previous studies, between 1995 and 2000 [62]. From these recent
but there is still room for improvement. figures, it may be concluded that the awareness of
27 Will Extralevator Abdominoperineal Excision Become the New Gold Standard? 271

a poor outcome has led to a decrease in margin References


involvement and iatrogenic perforations, both
important indicators of surgical quality. 1. Heald RJ (1988) The “holy plane” of rectal surgery.
Besides the increased awareness for radical J R Soc Med 81(9):503–508
2. den Dulk M, Krijnen P, Marijnen CA et al (2008)
surgery, preoperative planning with MRI, MDT Improved overall survival for patients with rectal
meetings, and the selective use of neoadjuvant cancer since 1990: the effects of TME surgery and
treatment is common practice. pre-operative radiotherapy. Eur J Cancer 44(12):1710–
With improved imaging, the area at risk has 1716, Oxford, England:1990
3. Elferink MAG, van Steenbergen LN, Krijnen P et al
been made visible which has resulted in patient (2010) Marked improvements in survival of patients
tailored treatment protocols. When evaluating all with rectal cancer in the Netherlands following
these changes in the modern treatment of low changes in therapy, 1989–2006. Eur J Cancer 46(8):
rectal cancer, the question whether a drastic 1421–1429
4. den Dulk M, Marijnen CA, Putter H et al (2007) Risk
change in surgical technique is necessary could factors for adverse outcome in patients with rectal
be fighting a ghost from the past. Unfortunately, cancer treated with an abdominoperineal resection in
the excellent recent reviews of How and Stelzner the total mesorectal excision trial. Ann Surg 246(1):
still do not give a clear answer since most studies 83–90
5. den Dulk M, Putter H, Collette L et al (2009) The
are a little bit dated and without the present stan- abdominoperineal resection itself is associated with
dard of staging, neoadjuvant treatment, and feed- an adverse outcome: the European experience based
back from the pathologist [6, 58]. on a pooled analysis of five European randomised
Several papers support the more conventional clinical trials on rectal cancer. Eur J Cancer 45(7):
1175–1183, Oxford, England: 1990
approach, among them Messenger who states 6. How P, Shihab O, Tekkis P et al (2011) A systematic
that a meticulous executed conventional tech- review of cancer related patient outcomes after ante-
nique, taking care not to breach the interface rior resection and abdominoperineal excision for rec-
between tumor and levators also yields excellent tal cancer in the total mesorectal excision era. Surg
Oncol 20:e149–e155
results [55]. 7. Anderin C, Martling A, Hellborg H, Holm T (2010) A
Even more, the number of patients requiring population-based study on outcome in relation to the
APE has diminished over the years, resulting in type of resection in low rectal cancer. Dis Colon
the selection of more advanced tumors that Rectum 53(5):753–760
8. Marr R, Birbeck K, Garvican J et al (2005) The mod-
undergo an APE compared to AR. Most impor- ern abdominoperineal excision. Ann Surg 242(1):
tantly, extralevator excision whether in supine or 74–82
prone position is not the debate; the real issue is 9. Bülow S, Christensen I, Iversen L, Harling H (2011)
how to identify those patients who require preop- Intraoperative perforation is an important predictor of
local recurrence and impaired survival after abdomi-
erative treatment and subsequently a more noperineal excision for rectal cancer. Colorectal Dis
extended resection [7]. 13:1256–1264
No rectal cancer patient should be allowed to 10. Eriksen MT, Wibe A, Syse A, Haffner J, Wiig JN
go to surgery without being discussed in a MDT (2004) Inadvertent perforation during rectal cancer
resection in Norway. Br J Surg 91(2):210–216
where preoperative treatment, MRI TNM stage, 11. Gosens MJEM, Klaassen RA, Tan-Go I et al (2007)
but also the exact anatomical landmarks which Circumferential margin involvement is the crucial
dictate type and extent of surgery, have been dis- prognostic factor after multimodality treatment in
cussed. The extralevator resection principle patients with locally advanced rectal carcinoma. Clin
Cancer Res 13(22):6617–6623
should be part of the standard surgical armamen- 12. Lindsey I (2011) Abdominoperineal excision rate: a
tarium. Finally, the role of the pathologist is to marker of quality in rectal cancer surgery. Colorectal
give continuous feedback on the decisions made Dis 13(7):731
during that MDT and how well the procedure 13. Nagtegaal ID, Quirke P (2008) What is the role for the
circumferential margin in the modern treatment of
was executed. In the end, tumor perforations and rectal cancer? J Clin Oncol 26(2):303–312
involved radial margins should become very rare 14. van Gijn W, Marijnen CA, Nagtegaal ID et al (2011)
and cease to be a prognostic factor. Preoperative radiotherapy combined with total mesorectal
272 I. Martijnse et al.

excision for resectable rectal cancer: 12-year follow-up 29. Shihab OC, Taylor F, Salerno G et al (2011) MRI pre-
of the multicentre, randomised controlled TME trial. dictive factors for long-term outcomes of low rectal
Lancet Oncol 12(6):575–582 tumours. Ann Surg Oncol 18:3278–3284
15. Wolberink SVRC, Beets-Tan RGH, de Haas-Kock 30. Marijnen CAM, Nagtegaal ID, Kapiteijn E et al (2003)
DFM et al (2009) Multislice CT as a primary screen- Radiotherapy does not compensate for positive
ing tool for the prediction of an involved mesorec- resection margins in rectal cancer patients: report of a
tal fascia and distant metastases in primary rectal multicenter randomized trial. Int J Radiat Oncol Biol
cancer: a multicenter study. Dis Colon Rectum 52(5): Phys 55(5):1311–1320
928–934 31. Quirke P, Steele R, Monson J, Grieve R, Khanna S,
16. Beets GL, Borstlap ACW, Oei TK et al (2000) Couture J, O’Callaghan C, Myint AS, Besselli E,
Abdominal imaging preoperative assessment of local Thompsonj LC, Parmarj M, Stephens RJ, Sebag-
tumor extent in advanced rectal cancer: CT or high- Montefioreb D, on behalf of the MRC CR07/NCIC-
resolution MRI ? Abdom Imaging 541:533–541 CTG CO16 Trial Investigators; NCRI Colorectal
17. Halefoglu A-M (2008) Endorectal ultrasonography ver- Cancer Study Group (2009) Effect of the plane of
sus phased-array magnetic resonance imaging for pre- surgery achieved on local recurrence in patients with
operative staging of rectal cancer. World J Gastroenterol operable rectal cancer: a prospective study using data
14(22):3504 from the MRC CR07 and NCIC-CTG CO16 ran-
18. Kav T (2010) How useful is rectal endosonography in domised clinical trial. Lancet 373(9666):821–828
the staging of rectal cancer? World J Gastroenterol 32. Tiefenthal M, Nilsson PJ, Johansson R, Glimelius B,
16(6):691 Påhlman L (2011) The effects of short-course
19. Beets-Tan RG, Beets GL, Vliegen RF et al (2001) preoperative irradiation on local recurrence rate and
Accuracy of magnetic resonance imaging in predic- survival in rectal cancer: a population-based nation-
tion of tumour-free resection margin in rectal cancer wide study. Dis Colon Rectum 54(6):672–680
surgery. Lancet 357(9255):497–504 33. Valentini V, Coco C, Picciocchi A et al (2002) Does
20. Beets-Tan RGH, Beets GL (2011) Local staging of downstaging predict improved outcome after preop-
rectal cancer: a review of imaging. J Magnetic Reson erative chemoradiation for extraperitoneal locally
Imaging 33(5):1012–1019 advanced rectal cancer? A long-term analysis of 165
21. Magnetic T, Imaging R, Euro RC (2007) Extramural patients. Int J Radiat Oncol Biol Phys 53(3):664–674
depth of tumor invasion at thin-section MR in patients 34. Janjan NA, Abbruzzese J, Pazdur R et al (1999)
with rectal cancer: results of the MERCURY study 1 Prognostic implications of response to preoperative
purpose: methods: results: conclusion. Radiology 243(1): infusional chemoradiation in locally advanced rectal
132–139 cancer. Radiother Oncol 51(2):153–160
22. Vliegen R, Dresen R, Beets G et al (2008) The accu- 35. Mohiuddin M, Hayne M, Regine WF et al (2000)
racy of multi-detector row CT for the assessment of Prognostic significance of postchemoradiation stage
tumor invasion of the mesorectal fascia in primary following preoperative chemotherapy and radiation
rectal cancer. Abdom Imaging 33(5):604–610 for advanced/recurrent rectal cancers. Int J Radiat
23. Maizlin ZV, Brown JA, So G et al (2010) Can CT Oncol Biol Phys 48(4):1075–1080
replace MRI in preoperative assessment of the cir- 36. Janjan NA, Khoo VS, Abbruzzese J et al (1999) Tumor
cumferential resection margin in rectal cancer? Dis downstaging and sphincter preservation with preop-
Colon Rectum 53(3):308–314 erative chemoradiation in locally advanced rectal can-
24. Dent OF, Chapuis PH, Haboubi N, Bokey L cer: the M.D. Anderson Cancer Center experience.
(2011) Magnetic resonance imaging cannot predict Int J Radiat Oncol Biol Phys 44(5):1027–1038
histological tumour involvement of a circumferential 37. Patel PA (2011) Evolution of 5-fluorouracil-based
surgical margin in rectal cancer. Colorectal Dis chemoradiation in the management of rectal cancer.
13:974–981 Anticancer Drugs 22:311–316
25. Salerno GV, Daniels IR, Moran BJ et al (2009) 38. Theodoropoulos G, Wise WE, Padmanabhan A et al
Magnetic resonance imaging prediction of an involved (2002) T-level downstaging and complete pathologic
surgical resection margin in low rectal cancer. Dis response after preoperative chemoradiation for
Colon Rectum 52(4):632–639 advanced rectal cancer result in decreased recurrence
26. Shihab OC, Heald RJ, Rullier E et al (2009) Defining and improved disease-free survival. Dis Colon Rectum
the surgical planes on MRI improves surgery for 45(7):895–903
cancer of the low rectum. Lancet Oncol 10(12): 39. Huerta S (2011) Radiosensitizing agents for the
1207–1211 management of rectal cancer. Anticancer Drugs 22:
27. Shihab OC, Moran BJ, Heald RJ, Quirke P, Brown G 305–307
(2009) MRI staging of low rectal cancer. Eur Radiol 40. Martijnse IS, Dudink RL, Kusters M et al (2012) T3+
19(3):643–650 and T4 rectal cancer patients seem to benefit from the
28. Hermanek P, Hohenberger W, Fietkau R, Rödel C addition of oxaliplatin to the neoadjuvant chemoradi-
(2011) Individualized magnetic resonance imaging- ation regimen. Ann Surg Oncol 19:392–401
based neoadjuvant chemoradiation for middle and 41. Miles WE (1944) Rectal surgery, 2nd edn. Cassell,
lower rectal carcinoma. Colorectal Dis 13(1):39–47 London
27 Will Extralevator Abdominoperineal Excision Become the New Gold Standard? 273

42. Lynch JM, Hamilton GJ (1939) Facts and fallacies in abdominoperineal resection for rectal cancer results
the treatment of rectal cancer. Am J Surg 43(1): in comparable oncologic outcomes. Dis Colon Rectum
39–44 54(8):939–946
43. Gabriel WB (1950) Perineo-abdominal excision of 55. Messenger DE, Cohen Z, Kirsch R et al (2011)
the rectum in one stage. Am J Surg 79(1):76–84, Favorable pathologic and long-term outcomes from
illust; Disc, 94 the conventional approach to abdominoperineal resec-
44. Gabriel WB (1932) Principles and practice of rectal tion. Dis Colon Rectum 54(7):793–802
surgery. Lewis & Co, London 56. Martijnse IS, Dudink RL, West NP, Wasowicz D,
45. Lloyd-Davies OV (1939) Lithotomy-trendelenburg Nieuwenhuijzen GA, van Lijnschoten I, Martijn H,
position. Lancet 7:74–76 Lemmens VE, van de Velde CJ, Nagtegaal ID, Quirke P,
46. Stelzner S, Holm T, Moran BJ et al (2011) Deep pel- Rutten HJ (2011) Focus on the extralevator perineal
vic anatomy revisited for a description of crucial steps dissection in supine position for low rectal cancer has
in extralevator abdominoperineal excision for rectal led to better quality of surgery and oncological
cancer. Dis Colon Rectum 54(8):947–957 outcome. Ann Surg Oncol 19(3):786–793
47. Wallner C, Lange MM, Bonsing BA et al (2008) 57. West NP, Anderin C, Smith KJE, Holm T, Quirke P
Causes of fecal and urinary incontinence after total (2010) Multicentre experience with extralevator
mesorectal excision for rectal cancer based on cadav- abdominoperineal excision for low rectal cancer.
eric surgery: a study from the cooperative clinical Br J Surg 97(4):588–599
investigators of the Dutch total mesorectal excision 58. Stelzner S, Koehler C, Stelzer J, Sims A, Witzigmann H
trial. J Clin Oncol 26(27):4466–4472 (2011) Extended abdominoperineal excision vs. stan-
48. West NP, Finan PJ, Anderin C et al (2008) Evidence dard abdominoperineal excision in rectal cancer – a sys-
of the oncologic superiority of cylindrical abdomino- tematic overview. Int J Colorectal Dis 26:1227–1240
perineal excision for low rectal cancer. J Clin Oncol 59. Kapiteijn E, Marijnen CAM, Nagtegaal ID et al (2001)
26(21):3517–3522 Preoperative radiotherapy combined with total mesorec-
49. Holm T, Ljung A, Häggmark T, Jurell G, Lagergren J tal excision for resectable rectal cancer. N Engl J Med
(2007) Extended abdominoperineal resection with 345(9):638–646
gluteus maximus flap reconstruction of the pelvic 60. Folkesson J, Birgisson H, Pahlman L et al (2005)
floor for rectal cancer. Br J Surg 94(2):232–238 Swedish rectal cancer trial: long lasting benefits from
50. Salerno G, Chandler I, Wotherspoon A et al (2008) radiotherapy on survival and local recurrence rate.
Sites of surgical wasting in the abdominoperineal J Clin Oncol 23(24):5644–5650
specimen. Br J Surg 95(9):1147–1154 61. Sebag-Montefiore D, Stephens RJ, Steele R et al
51. Corman ML (1984) Colon and rectal surgery. (2009) Preoperative radiotherapy versus selective
Lippincott, London postoperative chemoradiotherapy in patients with
52. Silberfein EJ, Kattepogu KM, Hu C-Y et al (2010) rectal cancer (MRC CR07 and NCIC-CTG C016): a
Long-term survival and recurrence outcomes follow- multicentre, randomised trial. Lancet 373(9666):
ing surgery for distal rectal cancer. Ann Surg Oncol 811–820
17(11):2863–2869 62. Nagtegaal ID, van de Velde CJH, Marijnen CA, van
53. Chuwa EWL, Seow-Choen F (2006) Outcomes for Krieken JHJM, Quirke P (2005) Low rectal cancer: a
abdominoperineal resections are not worse than those call for a change of approach in abdominoperineal
of anterior resections. Dis Colon Rectum 49(1):41–49 resection. J Clin Oncol 23(36):9257–9264
54. de Campos-Lobato LF, Stocchi L, Dietz DW et al
(2011) Prone or lithotomy positioning during an
Which Patients Do Benefit from
Extended Resections in Case of 28
Locally Advanced Rectal Cancer?

Ralph L. Dudink, Miranda Kusters, and Harm Rutten

Contents 28.1 Case Presentation


28.1 Case Presentation .......................................... 275
A male patient, 69 years of age, consulted his
28.2 What Is the Role of Neo-adjuvant
(Chemo-)radiation in the Treatment
general physician for rectal bleeding and an altered
of Primary Unresectable Patients? .............. 276 defecation pattern. At digital rectal examination,
a fixed tumour was found, and, subsequently, the
28.3 Which Extended Resections
Are Necessary to Achieve patient was referred for further evaluation.
a Radical Resection? ..................................... 278 At the start of a complete workup, the colonos-
28.4 Abdominosacral Resection ........................... 278 copy confirms the suspected malignancy of the
rectum at 7 and 8 cm in length, covering the com-
28.5 Results ............................................................ 280
plete circumference. Pathology examination of
28.6 Results After Pelvic Exenteration ................ 281 the biopsies taken reveals an adenocarcinoma.
28.7 Is an Extra Intra-operative Boost A chest and abdominal CT scan do not show
of Irradiation Effective to Reduce signs of distant metastasis. An MRI of the pelvis
the Chance of Local Recurrence? ................ 283 demonstrates a large tumour of 10 cm invading
28.8 Is There a Role for the Use the mesorectal fascia and infiltrating into the
of the Cellsaver in Locally prostate and bladder (see Fig. 28.1).
Advanced and Locally
This patient with a T4 tumour infiltrating
Recurrent Rectal Cancer Surgery? ............. 284
through the mesorectal fascia into the surround-
28.9 Which Patients Do Benefit ing structures requires an extended resection out-
from Extended Resections
in Case of Advanced Cancer? ...................... 285 side the involved mesorectal fascia. The subject
of this chapter are those patients in whom the nor-
References ................................................................. 287
mal anatomical relations are disturbed by tumour
growth. In those patients, often deemed unresect-
able for cure at presentation, a tailored multidisci-
plinary approach is necessary. Several key topics
regarding such treatment will be discussed.
First, the question will be raised if the expec-
tations from neo-adjuvant chemoradiotherapy are
different in these patients when compared to less
locally advanced cases. Another topic will be if
R.L. Dudink • M. Kusters • H. Rutten (*)
extended resections including the sacrum or the
Department of Colorectal Surgery, Catharina Hospital,
Eindhoven, The Netherlands pelvic organs are worthwhile in offering the
e-mail: harm.rutten@cze.nl patient a chance for cure. The third question will

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 275


DOI 10.1007/978-3-642-25005-7_28, © Springer-Verlag Berlin Heidelberg 2012
276 R.L. Dudink et al.

a b

Fig. 28.1 (a) Sagittal T2-weighted MRI slice showing a T4 (b) Transverse T2-weighted MRI slice showing the same
locally advanced rectal carcinoma which invades the blad- tumour, which invades the bladder (black arrowhead). The
der (black arrowhead) and the prostate (grey arrowhead). mesorectal fascia is invaded as well (white arrowhead)

deal with the usefulness of an intra-operative multicentre studies, have shown that the addition
radiation boost to further increase the therapeutic of oxaliplatin enhanced the downstaging effect of
ratio of the radiotherapy component. And last, the 5-FU-based neo-adjuvant treatment scheme
some thoughts about the use of the cellsaver to [10–16]. However, this increased downstaging
deal with the blood loss, which may be excessive did not always result in a better oncological
in these extended procedures, and its effect on outcome [15, 16].
oncological outcome. Nonetheless, the question is not: does neo-adju-
vant CRT work, but does intensified CRT work?
The Catharina Hospital Eindhoven is a national
28.2 What Is the Role of Neo- referral centre for primary unresectable T4 tumours.
adjuvant (Chemo-)radiation During the period of 1994–2010, a total of 504
in the Treatment of Primary consecutive patients without metastatic disease
Unresectable Patients? received four different neo-adjuvant treatment
schemes. In the early years, long-course radio-
To improve downstaging and downsizing of the therapy was the standard of treatment (RTH).
tumour, neo-adjuvant treatment became the stan- However, in 1998, the EORTC 22921 study intro-
dard of treatment in locally advanced rectal cancer. duced neo-adjuvant chemoradiation, consisting
Several studies have shown that neo-adjuvant long- of a bolus injection of 350 mg/m2 5-FU and leu-
course treatment schemes can effectively downstage covorin 20 mg/m2 (5-FU bolus) [1]. In 2003, the
and downsize tumours, which subsequently results CORE study commenced. It introduced two new
in a better oncological outcome [1–3]. However, principles: (1) continuous administration of 5-FU
there is no uniform definition of which scheme in the form of 825 mg/m2 capecitabine twice daily
should be used. The addition of a 5-FU-based radi- and (2) the addition of a second radiation sensi-
ation sensitizer resulted in increased downstaging, tizer in the form of 50 mg/m2 oxaliplatin once
compared to radiotherapy alone [1, 4, 5]. Since the weekly (CORE) [17]. In 2005, however, Dutch
introduction of 5-FU agents, bolus injections have guidelines changed and long-course radiotherapy
been replaced by continuous 5-FU administration with capecitabine twice daily only became the
[6–9]. Several studies, including two randomized standard of treatment (CAP). Therefore, selection
28 Which Patients Do Benefit from Extended Resections in Case of Locally Advanced Rectal Cancer? 277

1.0 Multivariable cox regression 0.10


Multivariable cox regression local
Cancer specific survival curve
recurrence rate

0.08
0.9

Local recurrence rate


Cum survival

0.06
Radicality of resection
0.8 R0
R1
0.04
Type of neo-adjuvant
0.7 treatment
RTH 0.02
5FUBolus
CAP
CORE 0.00
0.6

0 12 24 36 46 60 72 84 96 0 20 40 60 80
Months Months

Fig. 28.2 The left figure shows cancer-specific survival for the different neo-adjuvant treatment schemes after multi-
variate Cox regression analysis. The right figure shows the importance of radical resection

of the different neo-adjuvant treatment schemes ratios were 3.78, 2.73, 1.34 and 1.00 (p = 0.001)
was based on hospital policy rather than tumour and for downstaging 0.31, 0.44, 0.30 and 1.00
characteristics. All patients received 45–50.5 Gy (p < 0.0001). Toxicity rates between the CAP and
preoperative radiotherapy in 25 fractions and the CORE regimens did not differ significantly.
minimum waiting period was 8 weeks. In multivariate Cox regression analysis, the
The RTH, 5-FU bolus, CAP and CORE neo- CORE regimen remained significantly better for
adjuvant treatment schemes were administered to cancer-specific survival [18] (Fig. 28.2).
106, 137, 106 and 155 patients, respectively. R1 In previous papers, it has been shown that nega-
resections (tumour cells at the resection margin) tive circumferential resection margins remain of
occurred in 24.5%, 13.1%, 9.4% and 6.5% and paramount importance [19, 20]. Radical resec-
CRM+ (defined as tumour within 1 mm of the tion is a strong prognostic outcome parameter
circumferential resection margin) in 31.1%, [21–23]. Downstaging and downsizing before
21.9%, 14.2% and 8.4% after the different preop- surgery are essential to avoid positive resec-
erative treatments. No response to neo-adjuvant tion margins. Furthermore, pathologic complete
treatment was found in 57%, 44%, 49% and 27%, response (ypCR) is an important prognostic fac-
respectively. Pathologic complete response rates tor. Pathologic complete response rates vary in
(pCR) were 4.7%, 13.1%, 7.5% and 14.2%. In literature, ranging from 0% to 40% for combined
these challenging patients, intensification of the capecitabine and oxaliplatin regimens [13]. Any
neo-adjuvant treatment did lead to a more pro- response to neo-adjuvant treatment may also be an
nounced effect on downstaging. This could also important factor [3, 24–27].
be turned into a better oncological outcome. The interim results of two recent studies show
After multivariate regression analysis for R1, contradictory results [28, 29]. The NSABP04
CRM+ and any downstaging, the different neo- found no increase in pathologic complete response
adjuvant treatment schemes had a highly significant and an increase in toxicity when oxaliplatin was
impact on oncological outcome. Odds ratios for added. The CAO/ARO/AIO04, however, did find
having R1 resection for the RTH, 5-FU bolus, an increased pathologic complete response rate,
CAP and CORE group were 3.74, 1.94, 1.14 and without higher toxicity. The ACCORD study
1.00 (p = 0.003). For CRM+ resection, the odds showed an increase in radicality of resection and
278 R.L. Dudink et al.

tumour response but could not translate this into truly unresectable. Intraperitoneally metastasized
better oncological outcome [16]. However, the tumours have always been an exclusion criterion
Italian randomized STAR-01 phase III trial did for curative surgery for a long time, but nowa-
find a significant reduction of distant metastases days, there are curative options with HIPEC
when oxaliplatin was added [15]. For a definitive (hyperthermic intraperitoneal chemotherapy).
answer, the long-term results of these trials should The location of the tumour influences the
be awaited. However, these trials were multi- radicality of the resection and the consequent
institutional and the definition of locally advanced local recurrence rate. In both primary and locally
did not refer to unresectable T4 patients. In pri- recurrent rectal cancer, it has been shown that the
mary unresectable patients, intensified RCT may more dorsally the tumour is located, the higher
lead to better downstaging and hence increase the the irradical resection rate and local (re)recur-
chance for a radical resection [18]. rence rate is [30–34]. In these patients, often the
only chance for a radical resection is a sacral
resection.
28.3 Which Extended Resections Central and anterior tumours might require
Are Necessary to Achieve resection of the bladder and the prostate or semi-
a Radical Resection? nal vesicles in males and the vagina and uterus in
females. Lateral and dorsolateral tumours are
The main goal in curative surgery for both locally sometimes fixed to the pelvic sidewalls and can
advanced and locally recurrent rectal cancer is cause major bleeding during surgery.
achieving a radical resection. Radicality of the
resection is the most important factor which
influences local control and oncological outcome 28.4 Abdominosacral Resection
in general.
Only a thorough preoperative staging can A locally advanced dorsal rectal carcinoma and
provide the road map for a potential surgical especially the dorsal recurrent rectal carcinomas
intervention. A CT thorax/abdomen is neces- constitute a specific problem. When the dorsal
sary to exclude distant metastatic disease, as resection margin is threatened or involved, it is
most patients with distant metastases will be no not possible to achieve a radical resection with-
candidate for an extended resection (maybe with out an en bloc resection of the pelvic floor mus-
the exception of those with limited resectable cles, os coccyx or partial resection of the sacrum
metastases). An MRI scan of the pelvis is manda- (see Fig. 28.3).
tory, as delineation of tumour infiltration into the The sacral approach in prone position was
soft perirectal tissues is a prerequisite for surgi- originally described more than 100 years ago [35].
cal planning. Furthermore, the MRI is the best The combined abdominal and sacral approach
modality for local restaging to evaluate response was known as abdominosacral extirpation of the
to neo-adjuvant treatment, which may lead to rectum [35]. In its posterior approach, a much
less extended surgery. A PET scan may be help- wider resection is possible. The anterior approach
ful, especially in locally recurrent cases to dif- follows the rules of TME. Oncological outcome,
ferentiate between benign and malignant lesions. as described in literature, is disappointing. Five-
Preoperative discussion in a multidisciplinary year overall survival rates vary between 15% and
team with experience in the treatment of locally 30%, and local control rate varies between 15%
advanced and locally recurrent rectal cancer is and 40% [36–38].
essential to reach optimal treatment plans. Indications for abdominosacral resection
Tumours which invade the sacrum or the sacral (ASR) typically include invasion of the sacrum or
neural roots at the level of S2 or higher or tumours coccyx, tumour growth into the pelvic floor mus-
which extend more than a few centimetres beyond cles or lateral pelvic sidewalls and the need for a
the infrapiriform foramen will often be considered wider dorsal resection in order to achieve a radical
28 Which Patients Do Benefit from Extended Resections in Case of Locally Advanced Rectal Cancer? 279

a b

Fig. 28.3 (a) Sagittal T2-weighted MRI slice showing a pri- T2-weighted MRI slice showing the same tumour. The
mary locally advanced rectal carcinoma which invades the mesorectal fascia is clearly visible (black arrowheads) and is
sacrum and spinal canal (black arrowheads). (b) Transverse invaded by the tumour (white arrowhead)

a b c

Fig. 28.4 (a) Sagittal view of a wide field en bloc resec- sion [3]). (c) The dotted line shows the horseshoe-shaped
tion, indicated by the dotted line, of tumours with primar- circumferential margin at risk for irradical resection
ily latero-dorsal spread (Reproduced with permission [3]) (Reproduced with permission [3])
(b) Posterior line of transection (Reproduced with permis-

resection margin (Fig. 28.4) [39]. Double J-ureter sacral resections can also be performed in litho-
catheters can be helpful to identify and subse- tomy position. It is very helpful to identify the
quently avoid damage to the ureters. An ASR level of sacral transection by full thickness cut
starts with the abdominal phase. If the tumour through the sacrum with the osteotome (chisel)
growth only extends dorsally without threatening during the abdominal phase. The level of transec-
the ventral plane, TME rules can be followed, e.g. tion should be distal to the dura mater, which proj-
the ventral dissection plane will be guided by ects as far as mid S2 [36, 39].
Denonvilliers’ fascia. If the tumour invades ven- Before the patient is turned in prone position,
tral structures, e.g. the prostate in the male or the the abdominal phase can be completed by closure
uterus in the female, a more extended resection in of the abdomen and placement of the stoma.
the ventral plane is also necessary. The resection of A midline incision is made across the sacrum
the mid sacrum S2–S4 can be best done in prone from L5 to the perineum. The incision is contin-
position, as the venous plexus collapses in prone ued around the anus or scar after abdomino-
position and blood loss will be much less. Distal perineal extirpation (APE). The gluteus muscles
280 R.L. Dudink et al.

insertion from the sacrum, the sacrospinous and Table 28.1 Operative details of patients undergoing
sacrotuberous ligaments are transected near the sacral resection for T4 primary rectal cancer (n = 25) or
locally recurrent rectal cancer (n = 70).
sacrum. The levator ani muscles are in direct view
after this step and can be transected near their lat- T4-LARC LRRC
eral insertion at the obturator internus. The osteot- No. of patients No. of patients
Level of transection
omy performed during the abdominal phase may
S2 1 10
now be palpated. The sacral resection is com-
S3 5 33
pleted by a cut with the osteotome in the dorsal
S4 1 15
cortical wall in a V shape, in order to avoid dam- More distally 18 12
age to the sacral roots. The surgical specimen can Bladder resection
now be removed en bloc. An omentoplasty, which Total with ileal 20 9
has been mobilized during the abdominal phase conduit
can be used to fill the presacral space before clo- Partial 17 1
sure. In most cases, when no skin is lost, wound Urinary bladder flap 3 2
closure is primary. However, in case of a large right
defect, a myocutaneous rectus abdominis flap Urinary bladder flap 10 2
left
(VRAM) is rotated in the perineal wound [39].
Uterus extirpation
Other authors have advocated the use of (bio-) Without adnexes 0 1
meshes to create a new pelvic floor [40–42]. In With adnexes 40 16
our experience, prosthetic material was never nec- Vagina
essary for closure of the perineo-sacral defect. Posterior wall w/o 26 14
reconstruction
With reconstruction 10 5
28.5 Results Prostate
Capsule 29 6
From 1994 until 2010, we identified a group of Prostatectomy 3 6
369 patients who underwent surgery for T4 pri- Seminal vesicles
mary rectal cancer and 301 patients who under- Right only 13 0
Left only 8 0
went surgery for locally recurrent rectal cancer.
Both 20 3
For the analysis of the results, patients with peri-
Reconstruction
toneal carcinomatosis or distant metastasis were
VRAM w/o skin 11 4
excluded, resulting in a total of 336 patients with
VRAM with 14 7
T4-LARC (188 male, 147 female; mean age abdominal skin
60 years) and 212 patients with LRRC (126 male,
86 female; mean age 62 years) who were treated
with curative intent. achieved in 18 patients (72%), and R1 resection
Twenty-five patients with T4-LARC under- occurred in 7 patients (28%).
went sacral resection. Sacral resection was per- Post-operative complications typically incl-
formed by transection at either S2 (n = 1), S3 uded wound healing problems, superficial and
(n = 5), S4 (n = 1) or more distally (n = 18). All deep wound infections with or without wound
patients had preoperative long course of radio- dehiscence and abscesses (see Table 28.2).
therapy (23%) or radiochemotherapy (77%). Five-year cancer-specific survival is 68%.
Operative details are summarized in Table 28.1. Overall survival is 56%. Twenty-one percent of
Mean blood loss was 5,225 ml (200–18,000). patients develop local recurrence within 5 years
Packing of the small pelvis because of excessive of follow-up. Five-year metastasis-free survival
bleeding was performed in two patients (8%). is 69%, and relapse-free survival is 61%.
Intra-operative radiotherapy was administered in Seventy patients with LRRC underwent sacral
all but three patients. A radical resection was resection. In these patients, preoperative treatment
28 Which Patients Do Benefit from Extended Resections in Case of Locally Advanced Rectal Cancer? 281

Table 28.2 Post-operative complications in patients sacral resection (p = 0.191). There is no significant
undergoing sacral resection for T4 primary rectal cancer difference in the level of resection. Five-year
(n = 25) or locally recurrent rectal cancer (n = 70)
overall survival rates are 29% and 40%, respec-
T4-LARC LRRC tively (p = 0.217). Five-year local recurrence rates
No. of patients No. of patients are 46% and 43%, respectively (p = 0.379).
Wound infection 1 8
Metastasis-free survival rates are 47% and 50%,
Abdominal 1 8
infection/presacral
respectively (p = 0.442). Relapse-free survival
abscess rates are 24% and 39%, respectively (p = 0.025).
Abdominal 0 1 Ferenschild et al. report local control rates of
bleeding 88% for locally advanced rectal cancer and only
Wound dehiscence 2 1 10% for patients with recurrent rectal cancer,
Persisting ileus 1 3 with radicality of resection and lymph node posi-
Enterocutaneous 0 1 tivity being the only significant outcome factors
fistula
[36]. Three- and five-year overall survival rates
Abdominal sepsis 0 3
were reported at 46% and 30%, respectively [36].
Urinary leakage 0 1
Ureter obstruction 0 1
Typically, reported 5-year survival rates vary
Urinary retention 0 15 between rates of 18% and 37% [36]. Radicality
Urinary tract 0 2 of resection remains the most important prognos-
infection tic variable [36, 39, 43].
Severe pneumonia 4 1
DVT/pulmonary 0 1
embolism 28.6 Results After Pelvic
CVA 0 1 Exenteration
Death 0 1

In the T4 locally advanced rectal cancer patients,


consisted of no treatment (15%), reirradiation with treated at our institute, 20 complete bladder
or without RCT 65% and full course of radiation resections with ileal conduit, 17 partial bladder
with or without RCT in 20%. Twenty-five patients resections and 13 urinary bladder flap procedures
had a presacral recurrence (36%), 11 patients had were performed.
a posterolateral recurrence (16%), 7 had an anas- Forty patients underwent excision of the uterus
tomotic recurrence (10%), 1 had a perineal recur- and adnexes as part of the en bloc resection of a T4
rence and the remaining patients had a recurrence tumour. Twenty-six patients underwent resection
located anteriorly. Sacral resection was performed of the posterior vaginal wall without reconstruc-
by transection at either S2 (n = 10), S3 (n = 33), S4 tion, a further ten resections required subsequent
(n = 15) or more distally (n = 12). reconstruction. The prostate capsule was resected
Mean operation time was 420 min (240–660), in 29 patients and radical prostatectomy was per-
mean blood loss was 6,868 ml (500–20,000). formed three times. Seminal vesicles were either
Packing of the small pelvis because of excessive resected on the right side (n = 13), left side (n = 8)
bleeding was performed in 15 patients (21%). or bilaterally (n = 20). Reconstruction of the
Intra-operative radiotherapy was administered in perineum or vagina using a vertical rectus abdo-
all but four patients. A radical resection was minis myocutaneous flap (VRAM) with or with-
achieved in 31 patients (44%), R1 and R2 rates out abdominal skin was performed in 11 and 14
were 28 (40%) and 11 (16%), respectively. patients, respectively.
The post-operative complications are summa- Mean blood loss was 3,261 ml (50–25,000).
rized in Table 28.2. Intra-operative radiotherapy was administered in
Five-year cancer-specific survival rate for 328 of 336 patients. Radical resection was achieved
patients who underwent a sacral resection is 34% in 282 patients (84%), and R1 resection occurred
compared to 48% in patients who did not undergo in 52 patients (16%). Packing of the small pelvic
282 R.L. Dudink et al.

Table 28.3 Post-operative complications in patients prostatectomy. Furthermore, the right seminal
with T4 primary rectal cancer (n = 336) or locally recur- vesicle was resected in 3 patients, the left semi-
rent rectal cancer (n = 212)
nal vesicle was resected in 5 patients and both
T4-LARC LRRC were resected in 27 patients. Mean operation
No. of patients No. of patients time was 380 min (170–660), and mean blood
Wound infection 29 19
loss was 6,607 ml (420–34,000). Intra-operative
Abdominal infection/ 26 23
presacral abscess
radiotherapy was administered in all but 13
Abdominal bleeding 6 3 patients. A radical resection was achieved in 124
Wound dehiscence 8 1 patients (59%), and R1 and R2 rates were 63
Persisting ileus 2 4 (30%) and 25 (11%), respectively. Packing of the
Enterocutaneous 2 1 small pelvic because of excessive bleeding was
fistula performed in 36 (17%) of patients. A hemostatic
Abdominal sepsis 6 6 balloon was placed in two patients (1%). Post-
Anastomotic leakage 10 3 operative complications are summarized in
Urinary leakage 1 3 Table 28.3.
Ureter obstruction 0 1 Five-year cancer-specific survival is 43%, and
Urinary retention 5 29
10-year cancer-specific survival for these patients
Urinary tract 4 4
infection
with locally recurrent rectal cancer is 34%.
Urosepsis 1 1 Overall survival rates are 36% and 20%, respec-
Severe pneumonia 10 3 tively. Local recurrence rates after 5 and 10 years
DVT/pulmonary 1 3 are 43% and 51%, respectively. Five-year metas-
embolism tasis-free survival is 49% and 44% after 10-year
CVA 1 1 follow-up. Relapse-free survival rates after 5-year
Death 2 2 and 10-year follow-up are 34% and 24%, respec-
tively. Radicality of resection is the most impor-
tant prognostic variable, in which there is no
because of excessive bleeding was performed in significant difference between R1 and R2 resec-
20 (6%) patients. A hemostatic balloon was placed tions (see Fig. 28.5).
in three patients (1%). Post-operative complica- A recent study, describing pelvic exentera-
tions are summarized in Table 28.3. tion for locally advanced and recurrent rectal
In these patients, 5-year cancer-specific sur- cancer, confirms that radicality of resection was
vival is 66% and overall survival is 53%. Local the only significant outcome factor in multivari-
recurrence and metastasis-free survival rates at ate analysis. An excellent 2-year overall sur-
5-year follow-up are 19% and 67%. Relapse-free vival rate of 80% for primary and 75% for
survival is 58%. recurrent tumours was reported. Nine percent
In the locally recurrent rectal cancer patients developed local recurrence [44]. Ferenschild
group, 26 underwent total bladder resection with et al. report 5-year local control rates for pri-
ileal conduit, four partial bladder resections were mary and recurrent rectal cancer at 89% and
performed and urinary bladder flap was per- 38%, respectively [45]. Five-year overall sur-
formed on either the right side (n = 7) or left side vival rates were reported at 66% and 8%, respec-
(n = 10). Extirpation of the uterus was performed tively. Completeness of resection was identified
either with (n = 38) or without extirpation of the as one of the most important prognostic factors
adnexa (n = 2). Partial extirpation of the vagina [45]. Harris et al. report a 3-year local recur-
was performed in 33 patients, 11 patients under- rence rate of 13% [40]. Surprisingly, there was
went extirpation of the vagina with subsequent no significant association between local recur-
vertical rectus abdominis muscle reconstruction. rence and circumferential margin involvement.
Resection of the prostate capsule was performed The authors report 5-year overall survival rates
in 12 patients and 16 patients underwent total of 48% and 33% for radical and irradical
28 Which Patients Do Benefit from Extended Resections in Case of Locally Advanced Rectal Cancer? 283

Fig. 28.5 Local recurrence Local recurrence


rates according to radicality of
resection in recurrent rectal 1.0
cancer

0.8

Local recurrence rate


0.6

0.4

0.2

0.0

0 20 40 60 80 100
Months
Radicality of resection

R0 R0-censored
R1 R1-censored
R2 R2-censored

resected patients, respectively [40]. Wiig et al. (EBRT) and is feasible without increased toxicity
also emphasized the importance of a complete [47–49]. The theory behind IORT is that it
resection in salvage surgery for recurrent rectal reduced local recurrence rates by sterilizing
cancer. A 5-year survival for R0 patients at 53% microscopic residual tumour particles in a specific
compared to only 19% for R1 patients [46] was area. IORT is only applied on locally advanced
noticed. None of the R2 resected patients sur- rectal cancer (cT3+/T4) and in recurrent rectal
vived for 4 years (Fig. 28.5). cancer, as there is risk for remaining tumour
cells.
Historical studies suggest that IORT might
28.7 Is an Extra Intra-operative improve local control and survival [47, 50, 51].
Boost of Irradiation Effective Some studies suggest that it can only improve
to Reduce the Chance of Local local recurrence rates [52, 53], but some authors
Recurrence? find no benefit [54, 55], and some have even
abandoned IORT [56].
Intra-operative radiotherapy (IORT) is an entity The only way to quantify the effect is by
which is only practiced in a few specialized cen- analyzing the sites of local recurrence between
tres. It is administered as a boost of 10–15 Gy centres with a different policy in IORT applica-
after preoperative external beam radiotherapy tion. Most institutions using IORT-containing
284 R.L. Dudink et al.

multimodality treatment regimens deliver the boost surgery in high-volume referral centres special-
to the presacral space because this is the area con- ized in multimodality treatment is essential.
sidered most at risk [51] and because other areas Thus, we recommend the application of IORT
are difficult to cover with the applicator [57]. In in locally advanced rectal cancer, and data sug-
two studies reporting on patterns of local recur- gest that a boost to the area considered most at
rence after IORT, 59–67% of the local relapse risk is most effective in preventing local recur-
showed to develop outside the presacral IORT rences from developing.
radiation field [51, 57].
In our institution, the delivery of the boost is
to the area mostly considered at risk on the 28.8 Is There a Role for
basis of radiological and intra-operative the Use of the Cellsaver
findings. In a previous study, we questioned in Locally Advanced and
whether this approach leads to less outfield Locally Recurrent Rectal
local recurrences than the mentioned 59–67% Cancer Surgery?
in centres where IORT is only applied to the
presacral area. As mentioned before, blood loss may be consid-
From 1994 and 2006, 290 primary locally erable in locally advanced and recurrent rectal
advanced patients received intra-operative irradi- cancer because dissections planes often need to
ation. After 5 years 34 of the 290 patients devel- be extra-anatomically. Hence, allogeneic blood
oped local recurrence (13.2%). The subsites of transfusion is necessary in many cases.
the local recurrences were identified on MRI or Preservation techniques for the patient’s own
CT scan. Relating the patterns of local recurrence blood, or intra-operative cell salvage, has become
to the IORT-target, 47% of the local recurrences widely accepted and utilized in many braches of
developed outside the IORT-field [30]. This is surgery when substantial blood loss can occur.
less than in the few studies reporting on this sub- However, autologous blood transfusion has not
ject, in which the boost of IORT was given only been adopted widely in cancer surgery due to the
on the presacral area. fear of reintroducing viable tumour cells shed
Consequently, it might be suggested that an from the surgical site into the circulation. Based
IORT-boost specifically to the area at risk is more on this theoretical risk, the American Medical
effective in the prevention of local recurrence, Association Council on Scientific Affairs recom-
possibly because the area that causes tumour spill mended against autologous blood transfusion in
is sterilized. Also very suggestive that there is cancer surgery in 1986 [58].
effect of IORT is that 62% of the patients with an On the basis of these concerns, one could
irradical resection in this study [30] do not readily choose for allogeneic blood transfusion.
develop local recurrence. However, fresh autologous blood is superior to
Delivery of IORT to any specific area is tech- allogeneic blood in a number of areas. In the past,
nically very feasible. Normally, it can be deliv- a couple of studies suggested a relationship
ered through an abdominal access. However, the between perioperative blood transfusion and a
ventral area can be irradiated more adequately higher risk of recurrence of colorectal cancer
through the perineal wound. The only situation [59]. It is hypothesized that allogeneic blood
when IORT cannot be delivered is when there is transfusion has an immunosuppressive effect due
major blood loss during the operation and pack- to transfused allogeneic passenger leukocytes
ing is necessary. [59–61]. Furthermore, a dose-dependent relation-
IORT equipment is expensive and the logis- ship is suggested [59]. Allogeneic blood transfu-
tics are complex. Furthermore, non-metastasized sion is also associated with a higher risk of
locally advanced rectal carcinoma and local post-operative bacterial infections [61, 62].
recurrence are not very common. Therefore, for Furthermore, autologous blood has a higher
optimal treatment of these advanced cancers, 2,3-diphosphoglycerate (2,3-DPG) concentration
28 Which Patients Do Benefit from Extended Resections in Case of Locally Advanced Rectal Cancer? 285

Table 28.4 Five-year oncological outcome for the use of the cellsaver
Cancer-specific survival (%) Metastasis-free survival (%) Local recurrence rate (%)
Cellsaver No cellsaver p-value Cellsaver No cellsaver p-value Cellsaver No cellsaver p-value
All 74 57 0.001 70 61 0.036 12 22 0.007
patients
Q1 78 69 0.363 59 72 0.743 9 13 0.227
Q2 87 62 0.054 81 66 0.337 5 17 0.047
Q3 71 50 0.012 70 57 0.085 13 30 0.014
Q4 63 38 0.027 66 35 0.005 22 42 0.151

and thus lower oxygen binding affinity, resulting seems to be a survival benefit in patients with the
in better tissue oxygenation [63]. Experimental highest blood loss. The metastasis-free and local
studies have demonstrated that transfusion with recurrence-free survival rates show that the intro-
stored allogeneic blood results in less tissue oxy- duction of the cellsaver did not compromise onco-
gen consumption when compared to fresh autolo- logical outcome and is safe to use in these kinds of
gous blood [64]. patients (see Table 28.4 and Fig. 28.6).
Given these advantages, the cellsaver was The use of the cellsaver during oncological
used to collect, filter, wash and return the patient’s procedures remains controversial. However,
erythrocytes in order to maintain the circulating approximately 20–45% of patients with colorec-
red cell volume. After collection, the heparinized tal cancer develop metastases, which suggests
blood is being centrifugated, washed with a 0.9% that tumour cell dissemination is an early event in
saline solution and concentrated to hematocrit colorectal cancer. One study detected circulating
levels of 60–65%. The intra-operative red blood tumour cells in 41% of colorectal cancer patients
cell salvage (ICS) device was used with a leuco- in the preoperative stage [65]. Furthermore, it is
cyte depletion filter, which was replaced after suggested that surgical manipulation enhances
every two processed bowls. In case of contamina- the dissemination of malignant cells [65, 66].
tion by faeces or pus, ICS blood was not returned However, it has been estimated that only 0.01–
to the patient. If blood loss was excessive, plasma 0.000001% of circulating tumour cells have the
volume was replaced by moderate use of fresh potential to form metastatic lesions [58]. The
frozen plasma. most widely used technique to remove tumour
From 1994 until August 2010, data on 546 cells from the patient’s blood is the leucocyte
patients who have been treated for locally depletion filter. Several studies were not able to
advanced (n = 444) or recurrent (n = 102) rectal detect viable tumour cells after filtering the blood
cancer was collected prospectively. Four quartiles [67–69]. One study evaluated the effect of the use
representing the volume of blood loss were cre- of the cellsaver on local recurrence after radical
ated: Q1 less than or equal to 1,250 ml (n = 153), retropubic prostatectomy for prostate cancer.
Q2 1,251 ml up to 2,500 ml (n = 140), Q3 2,501 ml There was no statistical significant difference
up to 5,000 ml (n = 138), Q4 5,001 ml or more found for local recurrence rate [70].
(n = 115).
In these patients, mean blood loss was 3,697 ml,
3,110 ml for locally advanced rectal cancer 28.9 Which Patients Do Benefit from
patients and 6,209 ml for recurrent rectal cancer Extended Resections in Case
patients. Autologous blood was returned in 315 of Advanced Cancer?
patients (58%). Cancer-specific 5-year survival
for all patients and per quartile blood loss volume So, are extended resections including the sacrum
was higher when the cellsaver was used, compared or the pelvic organs worthwhile in offering the
to those without cellsaving (see Table 28.4). There patient a chance for cure? It depends on different
286 R.L. Dudink et al.

Fig. 28.6 (a) Five-year a Cancer specific survival


cancer-specific survival for Locally advanced rectal cancer
locally advanced rectal cancer,
stratified for the use of the 1.0
cellsaver (p = 0.043).
(b) Five-year cancer-specific
survival for locally recurrent
rectal cancer, stratified for the use 0.8
of the cellsaver (p = 0.061)

Cum survival 0.6

0.4

Cellsaver used
0.2
No
Yes
No−censored
Yes−censored
0.0

0 10 20 30 40 50 60
Months

b Cancer specific survival


Locally recurrent rectal cancer

1.0

0.8
Cum survival

0.6

0.4

Cellsaver used
0.2
No
Yes
No−censored
Yes−censored
0.0

0 10 20 30 40 50 60
Months
28 Which Patients Do Benefit from Extended Resections in Case of Locally Advanced Rectal Cancer? 287

important factors in the tailored treatment of References


patients presenting with extended tumour growth.
Achieving a radical resection should be the pri- 1. Bosset JF, Calais G, Mineur L et al (2005) Enhanced
mary goal when treating these patients, as sup- tumorocidal effect of chemotherapy with preoperative
radiotherapy for rectal cancer: preliminary results –
ported by numerous authors [36, 37, 39, 40, EORTC 22921. J Clin Oncol 23:5620–5627
42–46, 71–75]. Radical resection offers the only 2. Frykholm GJ, Pahlman L, Glimelius B (2001)
chance of cure. In case of a tumour invading sur- Combined chemo- and radiotherapy vs. radiotherapy
rounding structures or organs, en bloc resection alone in the treatment of primary, nonresectable ade-
nocarcinoma of the rectum. Int J Radiat Oncol Biol
is necessary to avoid tumour dissemination or Phys 50:427–434
positive margins [40]. In these patients, multivis- 3. Valentini V, Coco C, Picciocchi A et al (2002)
ceral resection offers the only possibility for cure Does downstaging predict improved outcome after
[40]. Lateral lymph node involvement, fixation to preoperative chemoradiation for extraperitoneal
locally advanced rectal cancer? A long-term anal-
the bony pelvis and sciatic nerve involvement are ysis of 165 patients. Int J Radiat Oncol Biol Phys
poor outcome parameters and challenge the sur- 53:664–674
geon [71, 76]. 4. Gerard JP, Conroy T, Bonnetain F et al (2006)
Therefore, to achieve a radical resection, Preoperative radiotherapy with or without concurrent
fluorouracil and leucovorin in T3–4 rectal cancers:
proper patient selection is of paramount impor- results of FFCD 9203. J Clin Oncol 24:4620–4625
tance [36, 37, 40, 42, 44, 71, 72, 74, 75, 77]. 5. Sauer R, Fietkau R, Wittekind C et al (2003) Adjuvant
Hence, accurate preoperative staging, including vs. neoadjuvant radiochemotherapy for locally
CT thorax/abdomen, MRI of the small pelvis and advanced rectal cancer: the German trial CAO/ARO/
AIO-94. Colorectal Dis 5:406–445
possibly PET, is required to evaluate if curative 6. Chan AK, Wong AO, Jenken DA (2010) Preoperative
surgery is feasible. Contraindications for resec- capecitabine and pelvic radiation in locally advanced
tion are distant metastasis, extensive sidewall rectal cancer – is it equivalent to 5-FU infusion plus
involvement, sacral invasion above S2, encase- leucovorin and radiotherapy? Int J Radiat Oncol Biol
Phys 76:1413–1419
ment of external iliac vessels, extensions of 7. Dunst J, Debus J, Rudat V et al (2008) Neoadjuvant
tumour through the sciatic notch and poor perfor- capecitabine combined with standard radiotherapy in
mance status [42, 71]. patients with locally advanced rectal cancer: mature
Because of the complexity of extended sur- results of a phase II trial. Strahlenther Onkol 184:
450–456
gery, these procedures should be undertaken in 8. Minsky BD, Roedel C, Valentini V (2010) Combined
specialized centres committed to the multimodal- modality therapy for rectal cancer. Cancer J 16:253–261
ity treatment of these patients. Therefore, central- 9. Saif MW, Hashmi S, Zelterman D et al (2008)
ization should be advocated [43, 44, 72]. Capecitabine vs. continuous infusion 5-FU in neoad-
juvant treatment of rectal cancer. A retrospective
Only in very few patients, there may be a place review. Int J Colorectal Dis 23:139–145
for these extended procedures in a palliative set- 10. Chau I, Brown G, Cunningham D et al (2006)
ting. Only if other measures have failed and life Neoadjuvant capecitabine and oxaliplatin followed by
expectancy is still moderate, an extended resec- synchronous chemoradiation and total mesorectal
excision in magnetic resonance imaging-defined poor-
tion to alleviate the symptoms of progressive risk rectal cancer. J Clin Oncol 24:668–674
local disease, which may have led to uncontrol- 11. Fakih MG, Bullarddunn K, Yang GY et al (2008)
lable tenesmus, severe pelvic pain or other qual- Phase II study of weekly intravenous oxaliplatin com-
ity-of-life-limiting symptoms, may be considered. bined with oral daily capecitabine and radiotherapy
with biologic correlates in neoadjuvant treatment of
Especially in metastasized patients, who respond rectal adenocarcinoma. Int J Radiat Oncol Biol Phys
well to palliative chemotherapy, the response of 72:650–657
the metastases may last longer than the postpone- 12. Marks JH, Valsdottir EB, Rather AA et al (2010)
ment of local progression of the primary to severe Fewer than 12 lymph nodes can be expected in a surgi-
cal specimen after high-dose chemoradiation therapy
symptoms [37, 42]. for rectal cancer. Dis Colon Rectum 53:1023–1029
288 R.L. Dudink et al.

13. Ofner D, Devries AF, Schaberl-Moser R et al (2011) infusional chemoradiation in locally advanced rectal
Preoperative oxaliplatin, capecitabine, and external cancer. Radiother Oncol 51:153–160
beam radiotherapy in patients with newly diagnosed, 25. Mohiuddin M, Hayne M, Regine WF et al (2000)
primary operable, cT(3)NxM0, low rectal cancer: a Prognostic significance of postchemoradiation stage
phase II study. Strahlenther Onkol 187:100–107 following preoperative chemotherapy and radiation
14. Sebag-Montefiore D, Glynne-Jones R, Falk S et al for advanced/recurrent rectal cancers. Int J Radiat
(2005) A phase I/II study of oxaliplatin when added to Oncol Biol Phys 48:1075–1080
5-fluorouracil and leucovorin and pelvic radiation in 26. Reerink O, Verschueren RC, Szabo BG et al (2003)
locally advanced rectal cancer: a colorectal clinical A favourable pathological stage after neoadjuvant
oncology group (CCOG) study. Br J Cancer 93: radiochemotherapy in patients with initially irresect-
993–998 able rectal cancer correlates with a favourable progno-
15. Aschele C, Pinto C, Cordio S et al (2009) Preoperative sis. Eur J Cancer 39:192–195
fluorouracil (FU)-based chemoradiation with and 27. Theodoropoulos G, Wise WE, Padmanabhan A et al
without weekly oxaliplatin in locally advanced rectal (2002) T-level downstaging and complete pathologic
cancer (STAR)-01 randomized phase III trial. J Clin response after preoperative chemoradiation for
Oncol 27:18s/CRA4008 advanced rectal cancer result in decreased recurrence
16. Gerard JP, Azria D, Gourgou-Bourgade S et al (2010) and improved disease-free survival. Dis Colon Rectum
Comparison of two neoadjuvant chemoradiotherapy 45:895–903
regimens for locally advanced rectal cancer: results of 28. Roh MS, Yothers GA, O’Connell MJ et al (2011) The
the phase III trial ACCORD 12/0405-prodige 2. J Clin impact of capecitabine and oxaliplatin in the preop-
Oncol 28:1638–1644 erative multimodality treatment in patients with carci-
17. Rutten H, Sebag-Montefiore D, Glynne-Jones R et al noma of the rectum: NSABP R-04. J Clin Oncol
(2006) Capecitabine, oxaliplatin, radiotherapy, and 29(suppl; abstract 3503)
excision (CORE) in patients with MRI-defined locally 29. Roedel C, Becker H, Fietkau R et al (2011)
advanced rectal adenocarcinoma: results of an inter- Preoperative chemoradiotherapy and postoperative
national multicenter phase II study. J Clin Oncol chemotherapy with 5-fluorouracil and oxaliplatin ver-
24:18s, abstract 3528 sus 5-fluorouracil alone in locally advanced rectal
18. Martijnse IS, Dudink RL, Kusters M et al (2012) T3+ cancer: first results of the German CAO/ARO/AIO-04
and T4 rectal cancer patients seem to benefit from the randomized phase III trial. J Clin Oncol 29(suppl;
addition of oxaliplatin to the neoadjuvant chemoradi- abstract LBA3505)
ation regimen. Ann Surg Oncol 19(2):392–401, Epub 30. Kusters M, Holman FA, Martijn H, Nieuwenhuijzen
2011 Jul 27 GA, Creemers GJ, Daniels-Gooszen AW et al (2009)
19. Dresen RC, Gosens MJ, Martijn H et al (2008) Radical Patterns of local recurrence in locally advanced rectal
resection after IORT-containing multimodality treat- cancer after intra-operative radiotherapy containing
ment is the most important determinant for outcome multimodality treatment. Radiother Oncol 92(2):
in patients treated for locally recurrent rectal cancer. 221–225
Ann Surg Oncol 15:1937–1947 31. Park JK, Kim YW, Hur H, Kim NK, Min BS, Sohn
20. Gosens MJ, Klaassen RA, Tan-Go I et al (2007) SK et al (2008) Prognostic factors affecting oncologic
Circumferential margin involvement is the crucial outcomes in patients with locally recurrent rectal can-
prognostic factor after multimodality treatment in cer: impact of patterns of pelvic recurrence on cura-
patients with locally advanced rectal carcinoma. Clin tive resection. Langenbecks Arch Surg 394:71–77,
Cancer Res 13:6617–6623 Epub 2008 July 29
21. Quirke P, Durdey P, Dixon MF MF et al (1986) Local 32. Salo JC, Paty PB, Guillem J, Minsky BD, Harrison
recurrence of rectal adenocarcinoma due to inade- LB, Cohen AM (1999) Surgical salvage of recurrent
quate surgical resection. Histopathological study of rectal carcinoma after curative resection: a 10-year
lateral tumour spread and surgical excision. Lancet 2: experience. Ann Surg Oncol 6(2):171–177
996–999 33. Suzuki K, Dozois RR, Devine RM, Nelson H, Weaver
22. Quirke P, Steele R, Monson J et al (2009) Effect of the AL, Gunderson LL et al (1996) Curative reoperations
plane of surgery achieved on local recurrence in for locally recurrent rectal cancer. Dis Colon Rectum
patients with operable rectal cancer: a prospective 39(7):730–736
study using data from the MRC CR07 and NCIC- 34. Kusters M, Dresen RC, Martijn H, Nieuwenhuijzen
CTG CO16 randomised clinical trial. Lancet 373: GA, van de Velde CJ et al (2009) Radicality of resec-
821–828 tion and survival after multimodality treatment is
23. Quirke P, Dixon MF (1988) The prediction of local influenced by the subsite of locally recurrent rectal
recurrence in rectal adenocarcinoma by histopatho- cancer. IJROB 75:1444–1449
logical examination. Int J Colorectal Dis 3:127–131 35. Bebenek M (2009) Abdominosacral amputation of
24. Janjan NA, Abbruzzese J, Pazdur R et al (1999) the rectum for low rectal cancers: ten years of experi-
Prognostic implications of response to preoperative ence. Ann Surg Oncol 16(8):2211–2217
28 Which Patients Do Benefit from Extended Resections in Case of Locally Advanced Rectal Cancer? 289

36. Ferenschild FT, Vermaas M, Verhoef C, Dwarkasing 49. Gunderson LL (1996) Past, present, and future of
RS, Eggermont AM, de Wilt JH (2009) Abdominosacral intraoperative irradiation for colorectal cancer. Int J
resection for locally advanced and recurrent rectal Radiat Oncol Biol Phys 34(3):741–744
cancer. Br J Surg 96(11):1341–1347 50. Gunderson LL, Nelson H, Martenson JA, Cha S,
37. Melton GB, Paty PB, Boland PJ, Healey JH, Savatta Haddock MG, Devine RM et al (1997) Locally
SG, Casas-Ganem JE, Guillem JG, Weiser MR, Cohen advanced primary colorectal cancer: IOERT and EBRT
AM, Minsky BD, Wong WD, Temple LK (2006) +/−5-FU. Front Radiat Ther Oncol 31:204–208
Sacral resection for recurrent rectal cancer: analysis 51. Calvo FA, Gomez-Espi M, az-Gonzalez JA, Alvarado
of morbidity and treatment results. Dis Colon Rectum A, Cantalapiedra R, Marcos P et al (2002)
49(8):1099–1107 Intraoperative presacral electron boost following pre-
38. Heriot AG, Byrne CM, Lee P, Dobbs B, Tilney H, operative chemoradiation in T3–4Nx rectal cancer:
Solomon MJ, Mackay J, Frizelle F (2008) Extended initial local effects and clinical outcome analysis.
radical resection: the choice for locally recurrent rec- Radiother Oncol 62(2):201–206
tal cancer. Dis Colon Rectum 51(3):284–291 52. Treiber M, Lehnert T, Oertel S et al (2004)
39. Mannaerts GH, Rutten HJ, Martijn H, Groen GJ, Intraoperative radiotherapy – special focus: recurrent
Hanssens PE, Wiggers T (2001) Abdominosacral rectal carcinoma. Front Radiat Ther Oncol 38:
resection for primary irresectable and locally recur- 52–56
rent rectal cancer. Dis Colon Rectum 44(6):806–814 53. Vermaas M, Ferenschildt FT, Nuyttens JJ et al (2005)
40. Harris DA, Davies M, Lucas MG, Drew P, Carr ND, Preoperative radiotherapy improves outcome in recur-
Beynon J, Swansea Pelvic Oncology Group (2011) rent rectal cancer. Dis Colon Rectum 48:918–928
Multivisceral resection for primary locally advanced 54. Palmer G, Martling A, Cedermark B, Holm T (2007)
rectal carcinoma. Br J Surg 98(4):582–588 A population based study on the management and
41. Heger U, Koch M, Büchler MW, Weitz J (2010) Totale outcome in patients with locally recurrent rectal can-
beckenexenteration. Chirurg 81(10):897–901 cer. Ann Surg Oncol 14:447–454
42. Mirnezami AH, Sagar PM (2010) Surgery for recur- 55. Heriot AG, Byrne CM, Lee P et al (2008) Extended
rent rectal cancer: technical notes and management of radical resection: the choice for locally recurrent rec-
complications. Tech Coloproctol 14(3):209–216 tal cancer. Dis Colon Rectum 51:136–147
43. Bebenek M, Pudełko M, Cisarz K, Balcerzak A, 56. Wiig JN, Tveit KM, Poulsen JP, Olsen DR, Giercksky
Tupikowski W, Wojciechowski L, Stankowska A, KE (2002) Preoperative irradiation and surgery for
Tarkowski R, Szulc R (2007) Therapeutic results in recurrent rectal cancer. Will intraoperative radiother-
low-rectal cancer patients treated with abdominosacral apy (IORT) be of additional benefit? A prospective
resection are similar to those obtained by means of study. Radiother Oncol 62:2007–2013
anterior resection in mid- and upper-rectal cancer 57. Roeder F, Treiber M, Oertel S, Dinkel J, Timke C,
cases. Eur J Surg Oncol 33(3):320–323 Funk A et al (2007) Patterns of failure and local con-
44. Zoucas E, Frederiksen S, Lydrup ML, Månsson W, trol after intraoperative electron boost radiotherapy
Gustafson P, Alberius P (2010) Pelvic exenteration for to the presacral space in combination with total
advanced and recurrent malignancy. World J Surg mesorectal excision in patients with locally advanced
34(9):2177–2184 rectal cancer. Int J Radiat Oncol Biol Phys 67(5):
45. Ferenschild FT, Vermaas M, Verhoef C, Ansink AC, 1381–1388
Kirkels WJ, Eggermont AM, de Wilt JH (2009) Total 58. Waters JH, Donnenberg AD (2009) Blood salvage
pelvic exenteration for primary and recurrent malig- and cancer surgery: should we do it? Transfusion
nancies. World J Surg 33(7):1502–1508 49(10):2016–2018
46. Wiig JN, Larsen SG, Dueland S, Flatmark K, 59. Amato A, Pescatori M (2006) Perioperative blood
Giercksky KE (2011) Salvage surgery for locally transfusions for the recurrence of colorectal cancer.
recurrent rectal cancer: total mesorectal excision dur- Cochrane Database Syst Rev 1:CD005033
ing the primary operation does not influence the out- 60. Burrows L, Tartter P (1982) Effect of blood transfu-
come. Colorectal Dis 13(5):506–511 sions on colonic malignancy recurrent rate. Lancet
47. Mannaerts GH, Martijn H, Crommelin MA, Dries W, 2(8299):662
van Driel OJ Repelaer, Rutten HJ (2000) Feasibility 61. Bordin JO, Blajchman MA (1995) Immunosuppressive
and first results of multimodality treatment, combin- effects of allogeneic blood transfusions: implications
ing EBRT, extensive surgery, and IOERT in locally for the patient with a malignancy. Hematol Oncol Clin
advanced primary rectal cancer. Int J Radiat Oncol North Am 9(1):205–218
Biol Phys 47(2):425–433 62. Mynster T, Christensen IJ, Moesgaard F, Nielsen HJ
48. Azinovic I, Calvo FA, Puebla F, Aristu J, Martinez- (2000) Effects of the combination of blood transfu-
Monge R (2001) Long-term normal tissue effects of sion and postoperative infectious complications on
intraoperative electron radiation therapy (IOERT): prognosis after surgery for colorectal cancer. Danish
late sequelae, tumor recurrence, and second malig- RANX05 colorectal cancer study group. Br J Surg
nancies. Int J Radiat Oncol Biol Phys 49(2):597–604 87(11):1553–1562
290 R.L. Dudink et al.

63. Apstein CS, Dennis RC, Briggs L, Vogel WM, Frazer J, 70. Davis M, Sofer M, Gomez-Marin O, Bruck D,
Valeri CR (1985) Effect of erythrocyte storage and Soloway MS (2003) The use of cell salvage during
oxyhemoglobin affinity changes on cardiac function. radical retropubic prostatectomy: does it influence
Am J Physiol 248(4 Pt 2):H508–H515 cancer recurrence? BJU Int 91(6):474–476
64. Gonzalez AM, Yazici I, Kusza K, Siemionow M 71. Austin KKS, Solomon MJ (2009) Pelvic exenteration
(2007) Effects of fresh versus banked blood transfu- with en bloc iliac vessel resection for lateral pelvic
sions on microcirculatory hemodynamics and tissue wall involvement. Dis Colon Rectum 52:1223–1233
oxygenation in the rat cremaster model. Surgery 72. Madoff RD (2006) Extended resections for advanced
141(5):630–639 rectal cancer. Br J Surg 93(11):1311–1312
65. Weitz J, Kienle P, Lacroix J, Willeke F, Benner A, 73. Moriya Y, Akasu T, Fujita S, Yamamoto S (2005)
Lehnert T, Herfarth C, von Knebel Doeberitz M Total pelvic exenteration with distal sacrectomy for
(1998) Dissemination of tumor cells in patients under- fixed recurrent rectal cancer. Surg Oncol Clin N Am
going surgery for colorectal cancer. Clin Cancer Res 14(2):225–238
4(2):343–348 74. Yamada K, Ishizawa T, Niwa K, Chuman Y, Aikou T
66. Yamaguchi K, Takagi Y, Aoki S, Futamura M, Saji S (2002) Pelvic exenteration and sacral resection for
(2000) Significant detection of circulating cancer cells locally advanced primary and recurrent rectal cancer.
in the blood by reverse transcriptase-polymerase chain Dis Colon Rectum 45(8):1078–1084
reaction during colorectal cancer resection. Ann Surg 75. Law WL, Chu KW, Choi HK (2000) Total pelvic
232(1):58–65 exenteration for locally advanced rectal cancer. J Am
67. Catling S, Williams S, Freites O, Rees M, Davies C, Coll Surg 190(1):78–83
Hopkins L (2008) Use of a leucocyte filter to remove 76. Ishiguro S, Akasu T, Fujita S, Yamamoto S, Kusters M,
tumour cells from intra-operative cell salvage blood. Moriya Y (2009) Pelvic exenteration for clinical T4 rectal
Anaesthesia 63(12):1332–1338 cancer: oncologic outcome in 93 patients at a single insti-
68. Miller GV, Ramsden CW, Primrose JN (1991) tution over a 30-year period. Surgery 145(2):189–195
Autologous transfusion: an alternative to transfusion 77. Sanfilippo NJ, Crane CH, Skibber J, Feig B,
with banked blood during surgery for cancer. Br J Abbruzzese JL, Curley S, Vauthey JN, Ellis LM, Hoff
Surg 78(6):713–715 P, Wolff RA, Brown TD, Cleary K, Wong A, Phan T,
69. Edelman MJ, Potter P, Mahaffey KG, Frink R, Leidich Janjan NA (2001) T4 rectal cancer treated with preop-
RB (1996) The potential for reintroduction of tumor erative chemoradiation to the posterior pelvis fol-
cells during intraoperative blood salvage: reduction of lowed by multivisceral resection: patterns of failure
risk with use of the RC-400 leukocyte depletion filter. and limitations of treatment. Int J Radiat Oncol Biol
Urology 47(2):179–181 Phys 51(1):176–183
Can Standard Surgical Procedure
Reliably Be Avoided in Major 29
Responders After Radio(chemo)
therapy?

Claudio Coco and Gianluca Rizzo

Contents Abbreviations

29.1 Introduction ................................................... 291 cCR Clinical complete response


CT Chemotherapy
29.2 Role of Total Mesorectal
Excision After Pathological
DFS Disease-free survival
Complete Response ....................................... 292 DWI Diffusion-weighted imaging
LE Local excision
29.3 Wait and See Policy ....................................... 293
MRI Magnetic resonance imaging
29.4 Definition and Value OS Overall survival
of Clinical Complete Response .................... 295
pCR Pathological complete response
29.5 Role of Local Excision After PET Positron emission tomography
Major Clinical Response
RCT Radiochemotherapy
to Neoadjuvant Treatment ........................... 296
TEM Transanal endoscopic microsurgery
References ................................................................. 300 TME Total mesorectal excision

29.1 Introduction

Based on the results of large randomized con-


trolled trials, the management of locally advanced
rectal cancer actually consists of neoadjuvant
radio(chemo)therapy (RCT) followed by radical
surgical rectal resection with total mesorectal
excision (TME). This approach has shown its
efficacy in reducing local recurrence rate, by
downstaging both the tumor and associated
C. Coco (*) • G. Rizzo lymph nodes and in increasing the likelihood of
Department of General Surgery, sphincter preservation [28, 55]. The degree of
Catholic University of Sacred Heart, tumor downstaging varies between patients and
Rome, Italy
can range from no response at all to complete
Complesso Integrato Columbus, sterilization of all tumor cells. This last condition
U.O.C. Chirurgia Generale 2,
is called pathological complete response (pCR).
Via Moscati 31-33, Rome 00168, Italy
e-mail: coco_claudio@rm.unicatt.it; During the last 10 years, there has been an
gianluca.rizzo1979@libero.it increasing proportion of patients who achieved a

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 291


DOI 10.1007/978-3-642-25005-7_29, © Springer-Verlag Berlin Heidelberg 2012
292 C. Coco and G. Rizzo

pCR. As it is well known from many studies survival, overall survival, and cancer-specific
[3, 5, 15, 16, 19, 25, 27, 35, 36, 40, 41, 49, 55, survival were 98.8%, 84.7%, 91.6%, and 95.6%,
61], the rate of pCR after neoadjuvant RCT in respectively. Yeo and the Korean Radiation
rectal cancer ranges between 8% and 35% Oncology Group in 2010 reported clinical data of
(Table 29.1). Pathological complete response has 304 ypT0N0 rectal cancer patients following pre-
a very important prognostic value and is associ- operative RCT and curative radical resections
ated with very good results in terms of local con- between 1993 and 2007 [63]. After a median
trol, disease-free survival (DFS), and overall follow-up of 43 months, 5-year DFS was 88.5%
survival (OS) [9, 14, 20, 52, 57, 58] (Table 29.2). and OS was 94.8%. In a recent pooled analysis
These good results have been confirmed in a large published in Lancet Oncology [37], Maas et al.
multicenter study published in 2008 by Capirci identified 27 articles, based on 17 different data-
and the Gastro-Intestinal Working Group of the sets, for long-term outcome of patients with and
Italian Association of Radiation Oncology. This without pCR after RCT and TME. Four hundred
study collected 566 patients with pCR (pT0N0) eighty-four of 3,105 (15.6%) included patients
after neoadjuvant therapy and surgery with TME had a pCR. After a median follow-up for all
for locally advanced rectal cancer with no evi- patients of 48 months, 5-year crude DFS was
dence of metastases at the time of diagnosis [12]. 83.3% for patients with pCR (with a disease
After a median follow-up of 45.6 months, locore- recurrence occurred in 61 patients) and 65.6% for
gional recurrence occurred in 7 patients (1.2%) those without pCR (p < 0.0001). The adjusted
and distant metastases in 49 patients (8.9%). hazard ratio for pCR for failure was 0.54 (95%
Overall, 5-year rates of local control, disease-free confidence interval: 0.40–0.73), indicating that
patients with pCR had a significantly increased
probability of disease-free survival. Even after
Table 29.1 Rate of pCR after RCT for extraperitoneal
rectal cancer adjustment for other relevant prognostic factors,
pCR still had a significant impact.
Authors Patients % pCR
Luna-Pérez et al. [36] 120 25
Hiotis et al. [25] 488 10
Mehta et al. [40, 41] 62 35 29.2 Role of Total Mesorectal
Sauer et al. [55] 405 8 Excision After Pathological
Wiltshire et al. [61] 135 21 Complete Response
Bosset et al. [5] 572 13
Gambacorta et al. [19] 102 27
Hughes et al. [27] 143 18 Total mesorectal excision was introduced by
Pucciarelli et al. [49] 235 24 Heald and colleagues [24] in 1982, and this tech-
Coco et al. [15, 16] 272 21 nique is based on the concept that an adequate en
Beddy et al. [3] 126 21 bloc clearance of the rectal mesentery, including
Lindebjerg et al. [35] 135 19 its blood supply and lymphatic drainage, would

Table 29.2 Oncologic outcome in patients who obtained pCR after RCT
Median FU
Authors Patients pCR (%) (months) 5-year LC 5-year DFS 5-year OS
Theodoropoulos et al. [58] 88 16 (18) 33 100 100 100
Garcia-Aguilar et al. [20] 168 21 (13) 37 100 95 95
Rödel et al. [52] 344 40 (12) 41 100 86 NS
Chan et al. [14] 120 32 (27) – 100 97 97
Stipa et al. [57] 200 60 (30) 39 96 NS 90
Bujko et al. [9] 131 22 (17) 48 95 91 NS
FU follow-up, LC local control, DFS disease-free survival, OS overall survival, NS not stated
29 Can Standard Surgical Procedure Reliably Be Avoided in Major Responders After Radio(chemo)therapy? 293

minimize possible disease relapse. Early experi- term RT: sensation of incomplete evacuation is
ence by Heald et al. (on 100 cases) documented reported in a range varying from 35% to 69% and
very good results in terms of local recurrence rate urgency between 16% and 42%; incontinence to
also without the benefit of adjuvant RT [24]. flatus, soiling, and incontinence to solid stools
Actually, TME is considered the gold standard of are reported, respectively, in a range varying from
treatment for extraperitoneal rectal cancers in 25% to 68%, from 20% to 50%, and from 7% to
Europe and in the USA. 14%, with almost 25% of cases which need to
While generally the value of TME is out of wear a pad [7, 18, 38].
doubt, there is much more concern about its role There are evidences in literature that TME
in pathological complete responders patients. It compromises urinary function (from 10% to
raises the question of what can be the contribu- 70%) and sexual function (from 13% to 70%)
tion of a TME in improving results in this specific too, even if nerve-sparing techniques or laparo-
group of patients and if it is reasonable to imag- scopic approach are employed. About urinary
ine to improve the prognosis by removing a functional outcome, the Dutch Colorectal Cancer
healed organ. Study Group [47] on 306 patients reported a rate
On the other hand, what is definitely known is of 39% of urinary incontinence with 57% of the
that TME is associated with a significant postop- patients wore pads because of urinary inconti-
erative morbidity and mortality rate, respectively nence. Moreover, 70% of patients needed to uri-
between 20% and 35% and between 1% and 4% nate again within 2 h, 53% referred urgency, and
[48, 51]. The most frequent postoperative com- 47% complained an incomplete bladder empty-
plication after TME is anastomotic leakage. Leak ing. Concerning sexual disorders after TME,
rates following TME are reported in the range of Sartori et al. [54] reported a reduced sexual desire
15–20% compared with a 5% or less after colonic and impaired spontaneous erectile function in
and intraperitoneal rectal anastomoses [48]. almost half the cases, respectively in 47.1% and
The anterior resection syndrome, including 41.1%; about one-quarter (23.5%) of patients
dysfunction as incontinence, urgency, incomplete still had not the possibility of penetration.
rectal evacuation, inability to defer defecation, Keeping in mind all these considerations about
and clustering of bowel movements, is consid- the possible complications after a TME, there is
ered a consequence of TME and low rectal anas- more than a suspect that this operation in pCR
tomoses. Loss of the physiological rectal reservoir patients (Fig. 29.1) could be an overtreatment. At
and alteration of anus-rectal physiology are the same time, the real problem nowadays is how
responsible for compromised anorectal function. can we identify patients with a pCR before a
In 2007, our institution published functional gas- standard surgical resection and how reliable is
trointestinal results [15, 16] of 100 patients our diagnosis of clinical complete response
treated with neoadjuvant long-term RCT and (cCR).
anterior resection with TME, from 1996 to 2003.
After 1 year from surgery, sensation of incom-
plete evacuation was reported in 58% of cases, 29.3 Wait and See Policy
necessity to return to the bathroom <15 min in
37%, and inability to evacuate completely Long-term results of “wait and see” approach
<15 min in 35%; urgency was referred in 31% of have been firstly reported by Habr-Gama and
cases. Incontinence to flatus was reported in 46% colleagues in 2004 [22] on selected group of
of cases, soiling in 19%, and incontinence to patients with radiological and clinical evidence
solid stools in 5% with 14% of patients needed of of CR after neoadjuvant RCT. Two hundred
wearing pad. In 29% of cases, these functional sixty-five patients with distal resectable rec-
gastrointestinal disorders affected normal social tal cancer were treated with preoperative RCT
and physical lifestyle. These poor functional (RT at a dose of 50.4 Gy in 1.8 Gy fractions
results after TME are also present after short- for six consecutive weeks plus concomitant CT
294 C. Coco and G. Rizzo

an endoluminal relapse, both of whom were


successfully treated; three patients (4.2%) devel-
oped metastatic disease. This series was updated
in 2005 and in 2006 extending to 360 patients
treated and 99 patients included in the observa-
tion group because classified as having achieved
cCR (MRI was included as staging tool) [23].
Local recurrence has occurred in three additional
patients (five in total, 5%) in the observation
group, all amenable to salvage surgery, and none
of whom have developed further recurrence.
Few other studies have assessed nonoperative
observational treatment after RCT. Other units
from the same city as Habr-Gama have reported
contrasting results. In the experience published
by Rossi et al. [53], in 1998, 16 patients with low
infiltrative rectal tumors (with initial indications
for abdominoperineal resection) were selected
prospectively to be treated with RCT (5,040 cGy
RT dose and CT during the first 3 and last 3 days
Fig. 29.1 Clinical complete response after radiochemo- of RT, using 425 mg/m2/day of 5-fluorouracil and
therapy for rectal cancer: no macroscopic evidence of 20 mg/m2/day of folinic acid) as an attempt to
disease is visible on specimen after total mesorectal
excision preserve sphincter function. Six patients (37.5%)
achieved a cCR and were followed in close sur-
veillance by monthly proctoscopy. Five of six
consisted of fluorouracil, 425 mg/m2/day, and (83.3%) patients failed locally after 1–10 months,
folinic acid, 20 mg/m2/day, on the first 3 days and and only a single patient maintained local control
last 3 days of RT) from 1991 to 2002. Patients at 34 months. Nakagawa et al. [43] reported on
were assessed at 8 weeks after completion of 51 patients with locally advanced extraperitoneal
RCT, and patients without any abnormality dur- rectal cancer treated with RCT (external beam
ing tumor response assessment were considered radiation plus 5-fluorouracil plus folinic acid).
to have cCR; these patients were referred to Ten (19.2%) patients showed cCR. Eight of ten
monthly follow-up visits for repeat physical and (80%) recurred locally within 3.7–8.8 months,
digital rectal examination, proctoscopy, biopsies and six succeeded in achieving salvage surgery.
(when feasible), and serum CEA levels (abdomi- Only two patients survived without cancer at 37
nal and pelvic scans and chest radiographs were and 58 months. In a recent report from UK by
repeated every 6 months during the first year). Hughes et al. [26], 10 of 58 patients unfit for sur-
Patients with sustained complete tumor regres- gery were identified as having had a cCR at clini-
sion for at least 12 months were considered stage cal reassessment 6–8 weeks after RCT. The
0, cCR, and were included in observation group. reasons for not proceeding with surgery in these
Seventy-one patients (26.8%) were deemed to ten patients were patient choice in three patients
have achieved cCR; all other patients proceeded and comorbidity in seven patients. Six of these
to surgery (resection group). After a median ten patients (60%) subsequently developed
follow-up of almost 5 years (57.3 months, range intrapelvic recurrent disease with a median time
12–156 months), OS and DFS was 88% and to local progression of 20 months. Local relapse
83% in the resection group and 100% and 92% preceded the development of metastatic disease
in the observation group, respectively; only two or occurred simultaneously. No patients under-
patients (2.8%) in the observation group had went salvage resection.
29 Can Standard Surgical Procedure Reliably Be Avoided in Major Responders After Radio(chemo)therapy? 295

The excitement born by Habr-Gama’s results The pathologic complete response rate among
and the necessity of a systematic prospective trial clinical complete responders was 25% with 75%
induced the Royal Marsden Hospital (Sutton, of cCR patients which had persistent foci of
Surrey, UK) and Pelican Cancer Foundation tumor that were not detectable on preoperative
(Basingstoke, Hampshire, UK) to start a pilot examination. Moreover, 55 patients (59.1%)
study investigating a nonoperative approach for preoperatively defined as cCR were affected by a
complete responders patients [45]. pT ³2 cancer and 16 (17.2%) had positive nodes
at pathological staging. On the contrary, 27 of
395 patients clinically classified as not com-
29.4 Definition and Value plete responders were ypT0N0 at pathological
of Clinical Complete Response examination.
In the study of Kim and colleagues in 2001
[29], all patients underwent sigmoidoscopy and a
At present, the criticisms existing about the digital rectal examination to evaluate clinical
“wait and see” approach are mainly due to the response after RCT and endorectal ultrasound
difficulties in identifying with certainty pCR was done after RCT in 12 of the 26 patients. In
patients. We must assume that what appears to the study of Onaitis et al. [46], clinical restaging
be a cCR is equal to a pCR, but this is not true after treatment consisted of proctoscopic exami-
in many times and we still not have nowadays a nation and often computed tomography scan. In
uniform accepted definition of cCR. Schell’s experience [56], digital rectal examina-
Several studies have investigated how cCR tion, endoscopy, computed tomography scan, and
is correlated with pCR. In Table 29.3 are endorectal magnetic resonance or endorectal
reported some who evaluated the ratio between ultrasound were utilized to define cCR.
cCR and pCR [25, 29, 46, 56, 64]. The ratio In last years, new diagnostic techniques con-
varies between 25% and 77%. This large vari- curred to refine the definition of cCR. Diffusion-
ability depends on the fact that the definition of weighted imaging (DWI) has shown promising
clinical complete response as “no evidence of results in the assessment of treatment response to
disease” is often vague and not homogeneous. neoadjuvant therapy. A recent multicenter com-
The largest, published in 2002, was a retro- parative study [30, 31] evaluated the accuracy of
spective review of the clinical and pathologic DWI in addiction to standard rectal MRI for
characteristics of 488 patients, from the Memorial selection of complete responders (only for ypT0)
Sloan-Kettering prospective colorectal database, after RCT. A total of 120 patients with locally
who received preoperative chemoradiation fol- advanced rectal cancer from three university hos-
lowed by resection for primary rectal cancer [25]. pitals (Maastricht University Medical Center,
Definition of cCR was based on the absence of Catholic University of Sacred Heart in Rome,
detectable tumor on preoperative digital rectal and University Hospital of Leuven) underwent
examination and proctoscopy; pCR was defined RCT followed by a restaging MRI, consisting of
as the absence of cancer cells in the resected standard T2W-MRI and DWI. Three independent
specimen. A cCR was achieved in 93 patients readers first scored the standard MRI only for the
(19%) and pCR rate among all patients was 10%. likelihood of a CR, after which DWI images were

Table 29.3 Ratio between Authors Patients cCR (%) Ratio pCR/cCR (%)
cCR and pCR
Kim et al. [29] 95 22/95 (23%) 17/22 (77%)
Onaitis et al. [46] 141 30/141 (21%) 18/30 (60%)
Hiotis et al. [25] 488 93/488 (19%) 23/93 (25%)
Schell et al. [56] 74 11/74 (15%) 8/11 (73%)
Zmora et al. [64] 109 47/109 (43%) 15/47 (33%)
296 C. Coco and G. Rizzo

added and the scoring was repeated. Histology surgeons (58%) would not consider conservative
was the standard reference. A pCR occurred in 25 management of patients with a complete response
patients (20.8%). The accuracy for three readers and even more (69%) expressed that they would
improved from 76%, 68%, and 58%, using only never discuss nonoperative management in
MRI, to 80%, 80%, and 78%, respectively, after patients with rectal cancer who are fit for curative
addiction of DWI (p-value: 0.39, 0.02, and 0.002). surgery.
Positive predictive value increased, after adding In conclusion, cCR is an evolving concept,
DWI, from a range of 0–56% to 62–81%, and but, at the current time, even using the latest avail-
negative predictive value increased from 79–85% able imaging diagnostic techniques, we have
to 88–90%. In this study, authors did not evaluate always a 15–20% of error in identifying pCR. In
lymph nodes. Standard MRI accuracy for lymph these patients with a major clinical response, we
node post-RCT is high and, in our institution, is are in a dilemma. Should we wait and see or
reported as 86.8% on the basis of morphologic should we perform a TME? The first option seems
criteria [1]. The only study [30, 31] specifically too little but at the same time the second one
focused on DWI for staging of rectal cancer seems too much.
nodes after RCT already showed good results for
standard MRI only (negative predictive value:
94–95%) and reported no clear benefit after addi- 29.5 Role of Local Excision After
tion of DWI (negative predictive value: Major Clinical Response
92–93%). to Neoadjuvant Treatment
Positron emission tomography (PET) demon-
strated high accuracy defining the response to Historically, local excision was considered in
RCT. Nearly the same multicenter study group rectal cancer patients as an alternative to radical
has recently developed predictive models for resection if patients were unfit for major surgery
pathologic complete response based on clinical or refused a permanent stoma. When used alone,
data (including age, gender, clinical tumor and local excision of rectal tumors was associated
nodal stage, and tumor location and tumor length with a high recurrence rate, but the key for an
based on MRI or endoscopy if MRI was unavail- appropriate use of local excision for rectal cancer
able) and on PET-based data (maximal tumor is the patient selection [2]. So, during the last
diameter, gross tumor volume, and maximal and 25 years, local excision as curative procedure
mean SUV values within the gross tumor vol- was accepted only for highly selected rectal can-
ume) [60]. Sequential PET-CT data in combina- cer patients who presented with a T1 (sm1 or
tion with clinical variables seem to significantly sm2) nonmucinous adenocarcinoma, with small
increase the performance of prediction models size (less than 4 cm), exophytic, mobile, moder-
for pathologic complete response with an accu- ately or well differentiated histology, and without
racy of 86 ± 5%. vascular, lymphatic, or perineural invasion [2].
Further complicating the situation is the fact Traditional local excision (Fig. 29.2), usually
that there is not a standard interval of time from performed using transanal surgical techniques
the end of RCT when the diagnosis of cCR should (Parks, Mason, Francillon, etc.), is performed
be done. This can vary from 4 weeks up to under general or regional anesthesia; the tumor
1 year. should be removed with a 1-cm margin of normal
In 2010, a questionnaire was sent to members mucosa around the tumor; the resection should
of the Association of Coloproctology of Great be a full-thickness excision, and the perirectal fat
Britain and Ireland regarding investigations, clin- should be seen clearly. Tumor fragmentation
ical management, pathological assessment, and should be avoided. The excised specimen should
oncological outcome in rectal cancer patients be oriented and pinned before fixation. Short-
with a complete response to neoadjuvant RCT term postoperative complications (including uri-
[62]. A total of 122 consultants responded: most nary retention, bleeding, and pelvic sepsis) are
29 Can Standard Surgical Procedure Reliably Be Avoided in Major Responders After Radio(chemo)therapy? 297

Fig. 29.2 Transanal


endoscopic microsurgery
equipment

infrequent, and most patients are discharged from Local excision of rectal cancers after RCT,
the hospital within 48 h of the operation. The especially using TEM, is increasingly being
introduction of transanal endoscopic microsur- described in the literature. The first docu-
gery (TEM) by Buess in 1983 [6] was an impor- mentation was in 1990 in a study conducted
tant technical advance in the local excision of in Comprehensive Rectal Cancer Center of
rectal cancer. For the TEM technique (Fig. 29.2), Philadelphia [39]. Fourteen patients, unable to
a large operating proctoscope with a stereoscopic tolerate radical surgery or with completely disap-
telescope is used, specially designed instruments peared tumor after irradiation, were treated with
are introduced through the proctoscope, and the preoperative RT (for a total dose of 45 Gy) and
rectum is insufflated with CO2, which maintains full-thickness local excision by transanal, trans-
exposure. This method, compared with the tradi- sphincteric, and transsacral techniques. Follow-up
tional transanal approach using Park’s retractor, observation ranged from 24 to 48 months with a
permits good exposure of the operative field with median of 31 months. Local recurrence devel-
stereoscopic magnified endoscopic vision that oped in three patients (21%), and 3-year actuarial
allows extremely precise dissection and permits local recurrence rate was 23%. Actuarial Kaplan-
a complete full-thickness excision to be per- Meier survival at 3 years was 61%. The Italian
formed with an appropriate margin (ablation with Lezoche’s group has the largest number of pub-
1 cm of surrounding free margin and with the lications on the subject. In 2002, this group of
largest amount of adjacent perirectal fat) [10]. study evaluated the results of preoperative high-
Moreover, TEM enables extension of local exci- dose RT (50.4 Gy) and TEM in 35 patients with
sion to tumors located in the mid-upper rectum pre-RT T2N0 rectal cancer [32]. The tumors were
that are not amenable to removal by standard responsive (50% or more reduction of the diam-
transanal techniques [10]. TEM is technically eter) to preoperative RT in 82.8% of cases. Low
difficult and requires a significant investment responsiveness or unresponsive patients were
in equipment and training for competent per- submitted to radical surgery with TME. After a
formance [2]. median follow-up of 38 months, only one local
298 C. Coco and G. Rizzo

Table 29.4 Rate of lymph Nodal metastases (%)


node metastasis in relation Authors Patients pCR ypT1 ypT2 ypT3 ypT4
to pathological tumor stage
after RCT Read et al. [50] 644 2 4 23 47 48
Bujko et al. [8] 316 5 8 26 55 64
Guillem et al. [21] 180 3 7 20 36 36
Mignanelli et al. [42] 242 3 11 29 37 ns

recurrence (2.85%) was noted and the probabil- in node-positive patients. In the first [11], 47
ity of surviving at 96 months was 83%. A larger patients underwent by traditional local excision
publication [33] from the same group included for a relatively small (median diameter: 4 cm),
data from a 15-year experience of the technique low (median distance from anal verge: 4 cm) rec-
on 151 patients with N0 rectal cancer (88 T2 and tal cancer. After long-term RCT (RT doses of 45,
63 T3) who underwent to TEM after preoperative 50.4, or 52.5 Gy with concurrent 5-fluorouracil-
high-dose RT. The definitive histology was 34 based CT), 49% of patients obtained a pCR, and
(22.5%) pT0, 23 (15.2%) pT1, 68 (45.0%) pT2, in 36%, only a microscopic disease was observed.
and 25 (16.3%) pT3. No pT0 patients did not go At a median follow-up of 63 months, five patients
to radical surgery with TME. For the patients (10.6%) developed local recurrence. None of the
with pre-RT T2, the probability of local failure LE patients who developed local recurrence have
was 6% and 5% for T3 patients. The probability died of rectal cancer. Thirteen patients (27.7%)
of metastasis was 3% for pre-RCT T2 patients were pre-RCT N1; of these, 3 (23.1%) developed
and 4% for T3 staging. The rectal cancer-specific a local recurrence versus 2 (5.9%) of the 34
survival rate at the end of the follow-up period patients with pre-RCT N0 disease; this difference
was 90% for pT2 and 77% for pT3 patients. In was relevant but not statistically significant
2008 results were published from a randomized (Fisher exact test, p = 0.1213), probably due to
study [34] that was performed to compare the the small number of N1 patients observed. In
oncologic results obtained by TEM and those contrast, the second study [44] did not find that
by laparoscopic TME resection in the treatment nodal positivity before RCT was associated with
of T2N0, G1–2 rectal cancer after neoadjuvant increased rates of local recurrence but, after a
therapy (high-dose 50,4 Gy RT combined with median follow-up of 64 months, the rate of local
continuous infusion of 5-flurouracil). The study recurrence was high in both groups: 15% in node-
enrolled 70 patients: of these, 35 were random- negative and 18% in node-positive patients. The
ized to TEM and 35 to TME. At pathological major criticism of this technique remains the
exam, there were 11 (32%) pT0 in TEM group impossibility to radically remove all the mesorec-
and 10 (29%) in TME group. After a median tal fat and, consequently, we have no direct path-
follow-up of 84 months, two local recurrences ological information about mesorectal lymph
(5.7%) were observed after TEM and one (2.8%) nodes status. Even when a tumor that has under-
after TME. Distant metastases (2.8%) occurred gone significant pT downstaging after RCT is
in one case each after TEM and TME. The prob- resected with clear margins, lymph node may
ability of survival for rectal cancer was the same still be involved [8, 21, 42, 50] (Table 29.4).
(94%) for TEM and TME. The main reason for performing local excision
As nodal positivity is an accepted adverse (in cCR patients) instead of radical surgery with
prognostic factor after RCT, the majority of stud- TME is to preserve the anatomical and functional
ies have assessed the value of TEM after RCT integrity of the rectum, avoiding a permanent
primarily in patients who were node negative colostomy and some of the bowel, bladder, and
before treatment on the assumption that they sexual dysfunction secondary to radical surgery
would remain so after RCT. Only two studies and improving quality of life. Although it seems
[11, 44] investigated the role of TEM after RCT reasonable, few studies have assessed this con-
29 Can Standard Surgical Procedure Reliably Be Avoided in Major Responders After Radio(chemo)therapy? 299

cept formally. Cataldo PA and colleagues [13] Table 29.5 Rate of lymph node metastasis in ypT0
prospectively evaluated functional outcome on patients
39 patients who underwent TEM (without pre- or Authors # ypT0 # pN+ % N+
postsurgical RCT) by a completed preoperative Crane et al. (2003) [17] 84 1 1.2
and postoperative (6 weeks) survey including Read et al. [50] 42 1 2.3
fecal incontinence severity index, fecal inconti- Bedrosian et al. [4] 45 4 8.8
nence quality of life, number of bowel move- Pucciarelli et al. [49] 56 1 1.8
Tulchinsky et al. [59] 17 1 5.8
ments per 24 h, and ability to defer defecation.
Coco et al. [15] 56 1 1.8
There was no change in any of these parameters
Maas et al. [37] 509 26 5.1
when preoperative and postoperative data were
Total 809 35 4.3
compared. To date, no published data exist about
quality of life after TEM for rectal cancer previ-
ously treated with RCT. In our institution from thickness excision of the rectal wall disk pre-
2000 to 2008, 21 patients underwent TEM after viously containing the rectal cancer can assess
RCT for rectal cancer and no needed further radi- indeed the pathologic response of the primary
cal surgery. After 1 year, all patients were asked tumor (ypT) with an accuracy of nearly 99%.
by a questionnaire on evacuation and continence. This result in assessing pathological response
No patients referred more than three daily bowel of T can be reached with a very low morbidity
movements, sensation of incomplete evacuation, rate and with good functional results. We still
necessity to return in bathroom, or inability to have the problem to have no direct pathologi-
completely evacuate within 15 min. Only one cal information about mesorectal lymph nodes,
patient (4.8%) refers urgency. If compared with but we can have data on this aspect in a round-
an historical series of 100 patients who under- about way. In 2007, our group have published
went radical surgery with TME [15, 16], these an analysis of 272 consecutive rectal cancer
symptoms were statistically less frequent. The [15] submitted to neoadjuvant RCT and surgery
median evacuation score was 24.5 (score varied with TME, to prospectively evaluate the correla-
from 0 to 28; the highest value corresponds to tion between pathological response of primary
better function), significantly lower than patients tumor and mesorectal lymph node involvement.
who underwent TME (16.1; p-value: 0.004). A pCR was recorded in 56 patients (20.6%),
Continence score in TEM patients results to be and lymph node metastases were found in 72
significantly lower than TME series (18.6 vs. patients (26.5%). When a pCR of primary tumor
13.7; p < 0.001; score varied from 0 to 20; the (pT0, TRG1) occurred, the rate of lymph node
highest value corresponds to better function): involvement has been less than 2% (1.8% in our
only two patients (9.5%) referred incontinence to series). Despite at univariate analysis also clini-
flatus after TEM; no patients reported soiling, cal pretreatment N-stage was statistically corre-
incontinence to solid stools, necessity of a pad, or lated with the risk of pathological lymph node
lifestyle limitations after RCT and TEM. metastasis, at multivariate analysis, the best
Local excision after neoadjuvant treatment predictors of pathologic lymph node involve-
of rectal cancer can be seen under two different ment were only ypT stage (p = 0.0013) and TRG
points of view. It can be considered as a minimal (p = 0.0011). Table 29.5 reports a collective
treatment of residual disease or it can be consid- review of recent papers [4, 15–17, 37, 49, 59]
ered just a diagnostic tool. If we consider local published on this topic. If we sum all the series,
excision and especially TEM, not as a therapeu- we can see that on 809 pT0 patients, only 35
tic choice but as the most effective diagnostic (4.3%) had a lymph node involvement. So we
tools we have today to confirm a pathological can consider local excision after a major clini-
complete response after neoadjuvant RCT, we cal response as the best diagnostic tool to patho-
have probably in our hands the most safe way to logically confirm the complete response of the
avoid unnecessary radical resections. The full- primary tumor after RCT and to estimate with
300 C. Coco and G. Rizzo

a very high accuracy (near 96%) the mesorectal erative chemoradiation for locally advanced rectal
nodal involvement. If pathological examination carcinoma. J Gastrointest Surg 8:56–62
5. Bosset JF, Calais G, Daban A et al (2004) Preoperative
of surgical specimen after TEM reveals the pres- chemoradiotherapy versus preoperative radiotherapy
ence of residual neoplastic diseases, patients in rectal cancer patients: assessment of acute toxicity
should be advised to undergo a standard surgical and treatment compliance. Report of the 22921 ran-
resection with total mesorectal excision because domised trial conducted by the EORTC radiotherapy
group. Eur J Cancer 40:219–224
of the higher risk of lymph node involvement. 6. Buess G, Theiss R, Hutterer F et al (1983) Transanal
endoscopic surgery of the rectum – testing a new
Conclusions method in animal experiments. Leber Magen Darm
Pathological complete response after RCT is 13:73–77
7. Bujko K, Nowacki MP, Oledzki J et al (2001)
increasingly reported and identifies a group of Sphincter preservation after short-term preoperative
patients with favorable prognosis in which radiotherapy for low rectal cancer – presentation of
major surgery is probably useless. On the own data and a literature review. Acta Oncol 40:
other hand, a radical resection with TME has a 593–601
8. Bujko K, Nowacki MP, Nasierowska-Guttmejer A
high rate of short- and long-term morbidity. et al (2005) Prediction of mesorectal nodal metastases
Major clinical response is an evolving concept after chemoradiation for rectal cancer: results of a
not easy to define and not truly reliable in pre- randomised trial: implication for subsequent local
dicting pCR. “Wait and see” policy appears excision. Radiother Oncol 76:234–240
9. Bujko K, Kolodziejczyk M, Nasierowska-Guttmejer
today still controversial, and reported results A et al (2010) Tumor regression grading in patients
are contrasting. with residual rectal cancer after preoperative chemo-
If a major or complete clinical response radiation. Radiother Oncol 95:298–302
after a neoadjuvant treatment occurs, full- 10. Burghardt J, Buess G (2005) Transanal endoscopic
microsurgery (TEM): a new technique and develop-
thickness local excision, rather by TEM, ment during a time period of 20 years. Surg Technol
should be considered as the best diagnostic Int 14:131–137
tool we have today to confirm the tumor 11. Callender GG, Das P, Rodriguez-Bigas MA et al
response. The accuracy of the pathological (2010) Local excision after preoperative chemoradia-
tion results in an equivalent outcome to total mesorec-
definition of tumor response after a local exci- tal excision in selected patients with T3 rectal cancer.
sion is near 99% and, in ypT0 patients, is pos- Ann Surg Oncol 17:441–447
sible to predict the absence of lymph node 12. Capirci C, Valentini V, Cionini L et al (2008)
involvement with an accuracy of nearly 96%. Prognostic value of pathologic complete response
after neoadjuvant therapy in locally advanced rectal
Clinical trials to confirm validity of an organ- cancer: long-term analysis of 566 ypCR patients. Int J
sparing approach after local excision diagno- Radiat Oncol Biol Phys 72:99–107
sis of ypT0 are required. 13. Cataldo PA, O’Brien S, Osler T (2005) Transanal
endoscopic microsurgery: a prospective evaluation of
functional results. Dis Colon Rectum 48:1366–1371
14. Chan AK, Wong A, Jenken D et al (2005) Post-
References treatment TNM staging is a prognostic indicator of
survival and recurrence in tethered or fixed rectal car-
1. Barbaro B, Fiorucci C, Tebala C et al (2009) Locally cinoma after preoperative chemotherapy and radio-
advanced rectal cancer: MR imaging in prediction of therapy. Int J Radiat Oncol Biol Phys 61:665–677
response after preoperative chemotherapy and radia- 15. Coco C, Manno A, Mattana C et al (2007) The role of
tion therapy. Radiology 250:730–739 local excision in rectal cancer after complete response
2. Baxter NN, Garcia-Aguilar J (2007) Organ preserva- to neoadjuvant treatment. Surg Oncol 16(Suppl 1):
tion for rectal cancer. J Clin Oncol 25:1014–1020 S101–S104
3. Beddy D, Hyland JM, Winter DC et al (2008) A 16. Coco C, Valentini V, Manno A et al (2007) Functional
simplified tumor regression grade correlates with sur- results after radiochemotherapy and total mesorectal
vival in locally advanced rectal carcinoma treated excision for rectal cancer. Int J Colorectal Dis 22:
with neoadjuvant chemoradiotherapy. Ann Surg 903–910
Oncol 15:3471–3477 17. Crane CH, Skibber J (2003) Preoperative chemoradia-
4. Bedrosian I, Rodriguez-Bigas MA, Feig B et al (2004) tion for locally advanced rectal cancer: rationale,
Predicting the node-negative mesorectum after preop- technique, and results of treatment. Semin Surg Oncol
2:265–270
29 Can Standard Surgical Procedure Reliably Be Avoided in Major Responders After Radio(chemo)therapy? 301

18. Dahlberg M, Glimelius B, Graf W et al (1998) rectal cancer: a multicenter study. Ann Surg Oncol
Preoperative irradiation affects functional results after 18:2224–2231. doi:10.1245/s10434–011–1607–5
surgery for rectal cancer: results from a randomized 32. Lezoche E, Guerrieri M, Paganini AM et al (2002)
study. Dis Colon Rectum 41:543–549 Long-term results of patients with pT2 rectal cancer
19. Gambacorta MA, Valentini V, Coco C et al (2004) treated with radiotherapy and transanal endoscopic
Chemoradiation with raltitrexed and oxaliplatin in microsurgical excision. World J Surg 26:1170–1174
preoperative treatment of stage II-III resectable rectal 33. Lezoche E, Guerrieri M, Paganini AM et al (2005)
cancer: phase I and II studies. Int J Radiat Oncol Biol Long-term results in patients with T2–3N0 distal rec-
Phys 60:139–148 tal cancer undergoing radiotherapy before transanal
20. Garcia-Aguilar J, Hernandez de Anda E, Sirivongs P endoscopic microsurgery. Br J Surg 92:1546–1552
et al (2003) A pathologic complete response to preop- 34. Lezoche G, Baldarelli M, Guerrieri M et al (2008) A
erative chemoradiation is associated with lower local prospective randomized study with a 5-year mini-
recurrence and improved survival in rectal cancer mum follow-up evaluation of transanal endoscopic
patients treated by mesorectal excision. Dis Colon microsurgery versus laparoscopic total mesorectal
Rectum 46:298–304 excision after neoadjuvant therapy. Surg Endosc
21. Guillem JG, Dıaz-Gonzalez JA, Minsky BD et al 22:352–358
(2008) cT3 N0 rectal cancer: potential overtreatment 35. Lindebjerg J, Spindler KL, Ploen J et al (2009) The
with preoperative chemoradiotherapy is warranted. prognostic value of lymph node metastases and tumor
J Clin Oncol 26:368–373 regression grade in rectal cancer patients treated with
22. Habr-Gama A, Perez RO, Nadalin W et al (2004) long-course preoperative chemoradiotherapy.
Operative versus nonoperative treatment for stage 0 Colorectal Dis 11:264–269
distal rectal cancer following chemoradiation therapy: 36. Luna-Pérez P, Rodríguez-Ramírez S et al (2001)
long-term results. Ann Surg 240:711–717 Preoperative chemoradiation therapy and anal sphinc-
23. Habr-Gama A (2006) Assessment and management of ter preservation with locally advanced rectal adeno-
the complete clinical response of rectal cancer to carcinoma. World J Surg 25:1006–1011
chemoradiotherapy. Colorectal Dis 8(Suppl 3):21–24 37. Maas M, Nelemans PJ, Valentini V et al (2010) Long-
24. Heald RJ, Husband EM, Ryall RD (1982) The term outcome in patients with a pathological complete
mesorectum in rectal cancer surgery – the clue to pel- response after chemoradiation for rectal cancer: a
vic recurrence? Br J Surg 69:613–616 pooled analysis of individual patient data. Lancet
25. Hiotis SP, Weber SM, Cohen AM et al (2002) Assessing Oncol 11:835–844
the predictive value of clinical complete response to 38. Machado M, Nygren J, Goldman S et al (2003) Similar
neoadjuvant therapy for rectal cancer: an analysis of outcome after colonic pouch and side-to-end anasto-
488 patients. J Am Coll Surg 194:131–135 mosis in low anterior resection for rectal cancer: a
26. Hughes R, Harrison M, Glynne-Jones R (2010) Could prospective randomized trial. Ann Surg 238:214–220
a wait and see policy be justified in T3/4 rectal cancers 39. Marks G, Mohiuddin MM, Masoni L et al (1990)
after chemo-radiotherapy? Acta Oncol 49:378–381 High-dose preoperative radiation and full-thickness
27. Hughes R, Glynne-Jones R, Grainger J et al (2006) local excision. A new option for patients with select
Can pathological complete response in the primary cancers of the rectum. Dis Colon Rectum 33:
tumor following pre-operative pelvic chemoradiother- 735–739
apy for T3–T4 rectal cancer predict for sterilisation of 40. Mehta VK, Poen J, Ford J et al (2001) Radio-
pelvic lymph nodes, a low risk of local recurrence and therapy, concomitant protracted-venous-infusion
the appropriateness of local excision? Int J Colorectal 5-fluorouracil, and surgery for ultrasound-staged T3
Dis 21:11–17 or T4 rectal cancer. Dis Colon Rectum 44:52–58
28. Kapiteijn E, Marijnen CA, Nagtegaal ID et al (2001) 41. Mehta VK, Cho C, Ford JM et al (2003) Phase II trial
Preoperative radiotherapy combined with total of preoperative 3D conformal radiotherapy, protracted
mesorectal excision for resectable rectal cancer. N venous infusion 5-fluorouracil, and weekly CPT-11,
Engl J Med 345:638–646 followed by surgery for ultrasound-staged T3 rectal
29. Kim TH, Chang HJ, Kim DY et al (2001) Pathologic cancer. Int J Radiat Oncol Biol Phys 55:132–137
nodal classification is the most discriminating prog- 42. Mignanelli ED, de Campos-Lobato LF, Stocchi L et al
nostic factor for disease-free survival in rectal cancer (2010) Downstaging after chemoradiotherapy for
patients treated with preoperative chemoradiotherapy locally advanced rectal cancer: is there more (tumor)
and curative resection. Int J Radiat Oncol Biol Phys than meets the eye? Dis Colon Rectum 53:251–256
77:1158–1165 43. Nakagawa WT, Rossi BM, de O’Ferreira F et al
30. Lambregts DM, Maas M, Riedl RG et al (2011) Value (2002) Chemoradiation instead of surgery to treat mid
of ADC measurements for nodal staging after chemo- and low rectal tumors: is it safe? Ann Surg Oncol
radiation in locally advanced rectal cancer – a per 9:568–573
lesion validation study. Eur Radiol 21:265–273 44. Nair RM, Siegel EM, Chen DT et al (2008) Long-
31. Lambregts DM, Vandecaveye V, Barbaro B et al (2011) term results of transanal excision after neoadjuvant
Diffusion-weighted MRI for selection of complete chemoradiation for T2 and T3 adenocarcinomas of
responders after chemoradiation for locally advanced the rectum. J Gastrointest Surg 12:1797–1805
302 C. Coco and G. Rizzo

45. O’Neill BD, Brown G, Heald RJ et al (2007) Non- 57. Stipa F, Chessin DB, Shia J et al (2006) A pathologic
operative treatment after neoadjuvant chemoradio- complete response of rectal cancer to preoperative
therapy for rectal cancer. Lancet Oncol 8:625–633 combined-modality therapy results in improved onco-
46. Onaitis MW, Noone RB, Fields R et al (2001) logical outcome compared with those who achieve no
Complete response to neoadjuvant chemoradiation downstaging on the basis of preoperative endorectal
for rectal cancer does not influence survival. Ann Surg ultrasonography. Ann Surg Oncol 13:1047–1053
Oncol 8:801–806 58. Theodoropoulos G, Wise WE, Padmanabhan A et al
47. Peeters KC, van de Velde CJ, Leer JW et al (2005) (2002) T-level downstaging and complete pathologic
Late side effects of short-course preoperative radio- response after preoperative chemoradiation for
therapy combined with total mesorectal excision for advanced rectal cancer result in decreased recurrence
rectal cancer: increased bowel dysfunction in irradi- and improved disease-free survival. Dis Colon Rectum
ated patients – a Dutch colorectal cancer group study. 45:895–903
J Clin Oncol 23:6199–6206 59. Tulchinsky H, Rabau M, Shacham-Shemueli E et al
48. Pinsk I, Phang PT (2007) Total mesorectal excision (2006) Can rectal cancers with pathologic T0 after
and management of rectal cancer. Expert Rev neoadjuvant chemoradiation (ypT0) be treated by
Anticancer Ther 7:1395–13403 transanal excision alone? Ann Surg Oncol 13:
49. Pucciarelli S, Urso E, DeSalvo GL et al (2006) 347–352
5-fluorouracil and weekly oxaliplatin combined with 60. van Stiphout RG, Lammering G, Buijsen J et al (2011)
radiotherapy for locally advanced rectal cancer: surgi- Development and external validation of a predictive
cal complications and long-term results. Arch Med model for pathological complete response of rectal
Res 37:860–865 cancer patients including sequential PET-CT imaging.
50. Read TE, Andujar JE, Caushaj PF et al (2004) Radiother Oncol 98:126–133
Neoadjuvant therapy for rectal cancer: histologic 61. Wiltshire KL, Ward IG, Swallow C et al (2006)
response of the primary tumor predicts nodal status. Preoperative radiation with concurrent chemotherapy
Dis Colon Rectum 47:825–831 for resectable rectal cancer: effect of dose escalation
51. Ridgway PF, Darzi AW (2003) The role of total on pathologic complete response, local recurrence-
mesorectal excision in the management of rectal can- free survival, disease-free survival, and overall sur-
cer. Cancer Control 10:205–211 vival. Int J Radiat Oncol Biol Phys 64:709–716
52. Rödel C, Martus P, Papadoupolos T et al (2005) 62. Wynn GR, Bhasin N, Macklin CP et al (2010)
Prognostic significance of tumor regression after pre- Complete clinical response to neoadjuvant chemora-
operative chemoradiotherapy for rectal cancer. J Clin diotherapy in patients with rectal cancer: opinions of
Oncol 23:8688–8696 British and Irish specialists. Colorectal Dis 12:
53. Rossi BM, Nakagawa WT, Novaes PE et al (1998) 327–333
Radiation and chemotherapy instead of surgery for 63. Yeo SG, Kim DY, Kim TH et al (2010) Pathologic
low infiltrative rectal adenocarcinoma: a prospective complete response of primary tumor following preop-
trial. Ann Surg Oncol 5:113–118 erative chemoradiotherapy for locally advanced rectal
54. Sartori CA, Sartori A, Vigna S et al (2011) Urinary cancer: long-term outcomes and prognostic
and sexual disorders after laparoscopic TME for rec- significance of pathologic nodal status (KROG
tal cancer in males. J Gastrointest Surg 15:637–643 09–01). Ann Surg 252:998–1004
55. Sauer R, Becker H, Hohenberger W et al (2004) 64. Zmora O, Dasilva GM, Gurland B et al (2004) Does
Preoperative versus postoperative chemoradiotherapy rectal wall tumor eradication with preoperative
for rectal cancer. N Engl J Med 351:1731–1740 chemoradiation permit a change in the operative strat-
56. Schell SR, Zlotecki RA, Mendenhall WM et al (2002) egy? Dis Colon Rectum 47:1607–1612
Transanal excision of locally advanced rectal cancers
downstaged using neoadjuvant chemoradiotherapy.
J Am Coll Surg 194:584–590
Part VII
Q&As on Pathology
What Is the Correct Procedure
for Handling the Surgical Specimen? 30
Nigel Scott

Contents 30.1 Introduction


30.1 Introduction .............................................. 305
In spite of recent developments in the preoperative
30.2 Specimen Dissection and Sampling ........ 306
30.2.1 Specimen Dissection and Macroscopic
imaging of rectal cancer using endoluminal ultra-
Assessment................................................... 306 sound, CT scans, magnetic resonance imaging
30.2.2 Sampling for Microscopic Examination ...... 307 (MRI) and PET-CT, pathological examination of
30.3 Microscopic Examination and Key the operative specimen remains a key part of the
Features in the Pathology Report ........... 308 management of the rectal cancer patient. The
30.3.1 Histological Typing and Grading ................. 309 pathologist’s report allows the patient to be placed
30.3.2 Direct Tumour Spread and pT Stage............ 309
in a prognostic category, indicates the likelihood
30.3.3 Circumferential Resection Margin
Involvement.................................................. 310 of tumour recurrence and determines the need for
30.3.4 Mesorectal Grade ......................................... 311 post-operative adjuvant therapy. A good macro-
30.3.5 APR Grading................................................ 311 scopic description, especially when supplemented
30.3.6 Local Peritoneal Involvement ...................... 312
by high-quality digital images, facilitates audit of
30.3.7 Vascular Invasion ......................................... 312
30.3.8 Lymph Node Metastasis and pN Stage ........ 314 the quality of radiology and surgery. Following
30.3.9 Examination of the Specimen After neoadjuvant treatment, pathological examination
Neoadjuvant Radiotherapy........................... 315 provides an immediate indication of how sensi-
References ............................................................... 316 tive the tumour is to radio- and chemotherapy.
Finally, the accurate recording of a minimum
standardised pathological data set is vital to
stratification and interpretation of clinical trials,
comparison of outcomes between different cen-
tres and health-care systems, evaluation of the
impact of population-based interventions such as
bowel cancer screening and the conduct of epide-
miological studies.
In this chapter, I intend to outline what I con-
sider the most important elements of rectal can-
cer reporting. I do not intend to cover biopsy
N. Scott reporting or evaluation of polypectomy and
Department of Histopathology, local excision specimens but will describe in
Leeds Teaching Hospitals NHS Trust,
St James University Hospital,
some detail the examination of rectal cancer
Leeds, UK after radical surgery. Inevitably, this is written
e-mail: nigel.scott@leedsth.nhs.uk from the perspective of a UK pathologist trained

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 305


DOI 10.1007/978-3-642-25005-7_30, © Springer-Verlag Berlin Heidelberg 2012
306 N. Scott

under the guidance of the Royal College of polyps, ulcers or colitis; and an assessment of the
Pathologists. Since 1998, the Royal College of completeness of mesorectal excision. The latter
Pathologists minimum data set has provided a is particularly important when trying to gauge
detailed, evidence-based set of instructions for the quality of surgery and in recent studies has
colorectal cancer reporting, widely available on been shown to be an important prognostic factor
the college website [54]. This was last revised in for both loco-regional recurrence and survival.
2007 and is now the standard against which all Mesorectal grading is described in more detail
UK pathology reports are judged. The introduc- below. Additionally, in the case of APR speci-
tion of a reporting proforma to accompany or mens, distance from tumour to dentate line is
replace free text reports has also had a major recorded. Although not widely used at present, a
impact on the completeness of pathological new grading system has recently been proposed
reporting in the UK as demonstrated by numer- to reflect the extent of surgical dissection at the
ous audits [7]. Proformas are increasingly help- level of the anal canal and ano-rectal junction.
ful in the construction of large population This has not yet entered general practice in the
databases by cancer registries, and we would UK but is described briefly in section 30.3.5 for
strongly encourage their use. completeness.
Tumour dimensions may be difficult to mea-
sure accurately when the tumour-bearing bowel
30.2 Specimen Dissection is left unopened; however, tumour size is not an
and Sampling important prognostic factor. Moreover, length
and width can usually be estimated adequately
30.2.1 Specimen Dissection from the tumour slices after dissection.
and Macroscopic Assessment The mesorectal surface (circumferential resec-
tion margin, CRM) is painted with India ink
Ideally, the resection specimen should be received (or an appropriate alternative), and the tumour-
fresh and unopened. Digital pictures are taken of bearing segment is sliced in the transverse plane
the front and back to demonstrate the quality of at 3–5-mm intervals. Slices through the tumour
mesorectal excision and any significant perito- and surrounding mesorectum are laid out in order
neal pathology, e.g., tumour perforation. The rec- on a white board, with proximal and distal ends
tum is opened from both ends along the anterior identified, and photographs taken (Fig. 30.1). It is
margin to within 5–10 mm of the tumour. It may at this stage that tumour dimensions and macro-
not always be possible to see the tumour clearly, scopic type can be recorded. Slices where tumour
especially after chemoradiotherapy, but if the invades closest to the CRM (either by direct
bowel is cut a few millimetres at a time and a spread or within lymph nodes or vessels) are pho-
finger inserted into the lumen to palpate the tographed individually (Fig. 30.2).
tumour edge, it is usually possible to avoid cut- Following a systematic examination of the
ting into the lesion. The opened rectum is then tumour slices, the maximum distance of tumour
pinned to a cork board and immersed in formalin invasion outside muscularis propria is recorded
for 48–72 h. If the tumour is more than 1 or 2 cm as well as the closest point of approach to the
long, a formalin-soaked paper wick can be inked circumferential margin. The location of
threaded through the lumen to facilitate fixation. cancer in the bowel wall is described as involving
Prior to dissection, the specimen is examined anterior, left, right and posterior quadrants or
macroscopically and its external appearance with reference to a clock face (as viewed from
described. This includes the type of operation per- below), e.g., ‘tumour located between 2 and 7
formed, e.g., abdominoperineal excision or ante- o’clock’. It should also be recorded where the
rior resection; length of bowel resected; location CRM is threatened, e.g., ‘tumour invades to
of tumour with respect to distal resection margin within 2 mm of the CRM at 3 o’clock’.
and peritoneal reflection; any peritoneal abnor- The mesorectum proximal and distal to the
malities, e.g., tumour perforation; coincidental luminal cancer is sliced in a similar way to look
30 What Is the Correct Procedure for Handling the Surgical Specimen? 307

Anterior

Proximal

Fig. 30.1 Digital image of slices through tumour and mesorectum for presentation at a multidisciplinary team meeting

30.2.2 Sampling for Microscopic


Examination

The distal resection margin of the specimen is


examined histologically only if macroscopic
tumour lies within 3 cm. Previous audits indicate
this margin is rarely invaded (<1% of all resec-
tions). The only exception to this rule is where on
histology the carcinoma shows a highly infiltrative
growth pattern, extensive lymphatic channel
invasion or is of signet ring cell type.
At least four blocks of tumour are sampled to
determine tumour type, grade and direct spread
(pT stage). These blocks must include the perito-
neum if tumour extends above the peritoneal
Fig. 30.2 Slice through tumour and mesorectum show- reflection, the area of deepest invasion through
ing direct tumour spread to the CRM at 12 o’clock and the bowel wall and closest point to the circumfer-
satellite tumour deposits within 1 mm of the CRM at ential margin. Ideally, one or more large (whole
3 o’clock
mount) sections including the whole circumfer-
ence of the rectum and surrounding mesorectum
for lymph nodes and other mesorectal tumour should be processed for histology. These large
deposits, but these slices are not routinely photo- sections are particularly useful for correlation
graphed unless the CRM is threatened. with MRI images at the multidisciplinary team
308 N. Scott

The distance between tumour and CRM is


measured using the vernier scale on the micro-
scope or an ocular micrometer. In the case of
involved lymph nodes adjacent to the CRM, the
measurement is made from the edge of the intra-
nodal tumour deposit rather than the lymph node
capsule.
Once blocks have been taken from the primary
tumour, peritoneum and CRM, a careful search
for lymph nodes (and other tumour deposits) is
made by inspection and manual palpation of the
perirectal fat. The vascular pedicle containing the
inferior mesenteric and superior rectal artery
should be identified first and sliced at 2–3-mm
Fig. 30.3 Whole mount H&E stained section showing
pT3 tumour well clear of the inked CRM intervals from its cranial tip to identify the lymph
node closest to the high vascular tie. This is the
C2 lymph node as defined in the Dukes staging
(MDT) meeting (Fig. 30.3). Sections through the system and, although not included in recent edi-
tumour are usually taken at right angles to the tions of the TNM classification, is nevertheless a
bowel wall to demonstrate mural penetration; marker of poor prognosis if involved. Most lymph
however, a recent study suggests that one or more nodes will be found in the posterior and lateral
additional blocks cut tangentially across the outer quadrants of the mesorectum at the level of the
surface of muscularis propria may increase the tumour and immediately above [11]. Nodes are
detection of vascular invasion [44]. rarely found in the anterior mesorectum. Tumour
At least two blocks of tissue should be sam- deposits may occasionally be found in the distal
pled where tumour invades close to the perito- mesorectal fat in which case they are usually
neum. Peritoneal involvement may be suspected associated with extensive proximal lymphatic
macroscopically from areas of serosal pallor, metastasis and a high likelihood of tumour recur-
retraction or puckering but, in a minority of cases, rence [40].
may only be detected microscopically. Conversely,
peritumoural fibrosis and inflammation can often
simulate peritoneal invasion, and microscopic 30.3 Microscopic Examination
examination is always necessary to correctly and Key Features in the
classify the lesion. Pathology Report
Where direct tumour spread or macroscopic
tumour nodules lie within 3 mm of the circumfer- Microscopic examination of tissue samples will,
ential margin, these areas should be sampled for in many instances, confirm the macroscopic
histology. CRM involvement (CRMI) is defined assessments described above. In a proportion of
as tumour invasion 1 mm or less from the cases, however, conclusions from the macro-
mesorectal surgical margin. It is important to scopic assessment may have to be modified as a
note this is not the same as the TNM classification result of histology, e.g., tumour involvement of
of an R1 or R2 resection where tumour is actually peritoneum versus subperitoneal fibrosis. Finally,
present, microscopically or macroscopically, at a number of important pathological features can
the margin, i.e., 0 mm clearance. It is also impor- only be diagnosed by looking down the micro-
tant to note that the peritoneal surface is NOT a scope, e.g., tumour grade, small vessel invasion,
surgical margin, and tumour involving the serosal small lymph node metastases, etc. The accurate
surface should not be confused with CRM assessment of these parameters relies upon ade-
involvement or an R1 resection. quate sampling of the gross specimen. It cannot
30 What Is the Correct Procedure for Handling the Surgical Specimen? 309

be emphasised too much that good histology will Grade is mainly determined by the extent of gland
never compensate for inadequate macroscopic formation. In the WHO grading system, tumours
examination and an intelligent purposeful are divided into low- and high-grade categories,
approach to selection of tissue blocks. It may take the latter corresponding to poorly differentiated
30–45 minutes for the proper dissection of a rec- carcinomas in a three-tier system. In the UK, the
tal cancer excision specimen, but the extra effort Royal College of Pathologists has proposed that
taken will be more than paid back by the quality colorectal adenocarcinomas are graded accord-
of the report. ing to the dominant pattern of differentiation,
The list of pathological prognostic factors whereas WHO advises that grade is based on the
which could be included in a report is large. Core least differentiated component [13, 54]. Both
data items of key importance to the correct man- have been shown to correlate with tumour stage
agement of patients, quality control of diagnosis and prognosis.
and surgery and prediction of disease recurrence
are more limited however, and include tumour
site, tumour size, relationship to anal canal and 30.3.2 Direct Tumour Spread
peritoneal reflection, distance to nearest longitu- and pT Stage
dinal resection margin, mesorectal grade, tumour
type, tumour grade, presence of vascular invasion, All pathological reports on rectal cancer must
pT stage, pN stage, total number of lymph nodes describe the maximum extent of direct tumour
harvested, pR stage and CRM involvement. spread through the bowel wall and provide
While some of these features have already the correct TNM stage. Use of Dukes stage is
been mentioned in the preceding section, a more optional. In the UK, carcinoma is only diagnosed
detailed description is given below including evi- when neoplastic cells invade through muscularis
dence from clinical and pathological studies mucosae into submucosa. Neoplastic glandular
which supports their inclusion as important data proliferations confined to the mucosa are, by
items in the pathologist’s report. common convention, regarded as dysplasia. The
only exception to this rule occurs following neo-
adjuvant therapy where, very occasionally, small
30.3.1 Histological Typing and Grading islands of adenocarcinoma are left behind in the
mucosa after extensive tumour regression in the
Four blocks of tumour are sufficient to type and remainder of the specimen. In this circumstance,
grade most rectal carcinomas. Typing is per- the TNM category pTis can be used. While
formed according to the fourth edition of the intramucosal adenocarcinoma clearly exists, it
WHO Classification of Tumours of the Digestive is doubtful whether this stage of carcinogenesis
System [13]. Over 90% are adenocarcinomas and can be consistently recognised using morpho-
most are either well or moderately well differen- logical criteria alone (except where the tumour is
tiated. Rarer types of carcinoma encountered in poorly differentiated and invades as single cells),
the rectum include signet ring cell carcinoma, and the risk of lymph node metastasis in such
adenosquamous carcinoma, squamous cell carci- early disease is widely regarded as negligible if
noma, neuroendocrine carcinoma and undiffer- it exists at all [16]. pT stage is described in the
entiated carcinoma. Mucinous adenocarcinomas seventh edition of the TNM system as: pT0 – no
are defined as tumours in which over 50% of the evidence of primary tumour, pTis – carcinoma in
tumour volume is made up of extracellular mucin situ: intraepithelial or invasion of lamina propria,
pools and can be either low or high grade. This pT1 – adenocarcinoma confined to the submu-
type is more commonly seen in the colon. cosa, pT2 – invasion of muscularis propria, pT3
Tumour grade reflects how well differentiated invasion through muscularis propria into perirec-
the carcinoma is i.e., the degree to which it tal fat, pT4a – tumour perforation of the visceral
resembles normal rectal (glandular) epithelium. peritoneum and pT4b – direct tumour invasion
310 N. Scott

into other organs or structures [43]. The latter and threatened CRM [46]. Routinely measuring
includes invasion of the levator ani muscle in extramural invasion is therefore, not only a record
advanced low rectal cancers. Other organs com- of a valuable prognostic factor but can be used to
monly invaded by aggressive rectal tumours and audit the quality of magnetic resonance imaging.
included in en bloc excisions or pelvic exentera-
tions include the urinary bladder, seminal vesi-
cles, prostate gland, vagina and uterus. Invasion 30.3.3 Circumferential Resection
of these structures should always be confirmed Margin Involvement
by microscopic examination. Occasionally, only
fibro-inflammatory changes are seen in the adhe- In 1986 Quirke and colleagues from Leeds, UK,
sion between a rectal cancer and excised uterus described the results of the first systematic exam-
or bladder. The prognosis in these patients is bet- ination of the circumferential or radial resection
ter than if carcinoma were present, and unless margin of the rectum [29]. In this seminal paper
there is convincing evidence of previous tumour they showed a strong relationship between CRM
perforation at this site, the final pT stage should involvement (CRMI) and local recurrence in the
be pT1–3 rather than pT4. Rectal perforation is pelvis. Eleven out of 13 patients (85%) with CRMI
seen in 10–20% of operations and may be either eventually developed a loco-regional recurrence
spontaneous or iatrogenic. Surgical perforation compared with 3% of those with clear margins.
is particularly likely to occur during an APR Prior to this publication most pathologists’ atten-
procedure. In an analysis of the Dutch TME- tion had been concentrated on the distal mural
radiotherapy trial, perforation was documented resection margin. Subsequent studies from Leeds
in 13.7% of APR specimens and 2.5% of ante- and elsewhere, including large population-based
rior resections, although not all of these occurred series and data from randomised controlled trials,
through the tumour and therefore, would not all confirmed the prognostic significance of tumour
qualify as pT4 [27]. Both iatrogenic and spontane- at the CRM, defined as carcinoma 1 mm or less
ous perforations are associated with an increased from the resection line, the most recent publica-
risk of tumour recurrence however, and must be tions reporting local recurrence in approximately
documented in the pathology report. 20% of CRM-positive cancers compared with
In addition to pT stage, it is now common 10% if the margin was uninvolved. As well as
practice in the UK to measure the maximum being one of the best pathological predictors of
extent of direct tumour spread outside the muscu- local recurrence, CRM positivity is also predic-
laris propria in millimetres. This is usually per- tive of distant metastasis, disease-free survival
formed at the time of macroscopic examination and overall survival. In a series of 141 patients
and then confirmed microscopically. Where the undergoing curative surgery for rectal cancer in
muscularis propria is destroyed by carcinoma, an Leeds between 1985 and 1990, CRMI was the
estimate is made by extrapolating the line of the strongest independent prognostic factor together
muscle wall from adjacent bowel. Several studies with lymph node status [2]. Reported series
show that advanced pT3 tumours which have where preoperative radiotherapy or chemoradio-
invaded further outside the bowel wall have a therapy has been used likewise show CRMI to be
higher risk of local recurrence and poorer survival an important prognostic factor, even after neoad-
than more superficially invasive lesions [9, 21]. juvant therapy [25].
This is found to be true for both node-positive and An analysis of the relationship between sur-
node-negative groups. In 2007, the Mercury study vival, recurrence rates and tumour distance from
group found that MRI scans were highly accurate the CRM has generally supported the validity of
in measuring this parameter preoperatively [20]. the 1 mm definition with the exception of a single
Subsequently, this has led to the proposition that study from the Dutch TME-radiotherapy trial
patients could be selected for preoperative radio- which suggested that a 2-mm cut-off was more
therapy on the basis of MRI determination of effective in defining a poor prognostic group [24, 28].
extent of extramural spread, vascular invasion It is clear, however, that within the group of
30 What Is the Correct Procedure for Handling the Surgical Specimen? 311

tumours 1 mm or less from the surgical margin, 30.3.4 Mesorectal Grade


those which are actually present at the specimen
edge i.e., 0 mm (TNM category R1), have a par- The mesorectum is an anatomically defined enve-
ticularly high risk of recurrence. For this reason, lope of fat surrounding the rectum and separated
it has been suggested that the TNM residual from surrounding structures by a thin mesorectal
tumour classification be expanded to accommo- fascia. It contains the main vascular supply and
date CRMI and include two categories of R1: lymphatic drainage of the rectum; consequently
‘R1-dir’ where distance from carcinoma to the most rectal tumours and involved lymph nodes
edge is 0 mm and ‘R1 < 1 mm’ where clearance is will be contained within it. Since it is now widely
>0 mm but <1 mm [55]. believed that many local recurrences can be pre-
A national audit conducted in the UK between vented by careful removal of the mesorectum,
1999 and 2002, including data from 39 differ- the pathologist should pay particular attention to the
ent surgical units, found the CRM to be involved quality of mesorectal excision i.e., whether the
in between 0% and 33% of surgical resections; mesorectum is removed intact or not. Quirke has
overall rate 12.5% [47]. In the Dutch TME- described a grading system which categorises
radiotherapy trial and recently reported MRC the rectal cancer specimen according to whether
CRO-7 trial in which total mesorectal excision the surgeon has dissected outside the mesorectal
was the normal practice, the margin was found fascia (mesorectal plane excision) or cut into the
to be involved in 18.3% and 12% of unirradiated mesorectum, leaving mesorectal fat behind in the
patients respectively [24, 28, 35]. Not surpris- pelvis: intra-mesorectal and muscularis propria
ingly, CRMI was more common in advanced- plane excision [31]. Examples of complete and
stage tumours. In addition, margin involvement incomplete TME are shown in Fig. 30.4. This
is more common when the tumour is situated low mesorectal grading system has been evaluated
down in the rectum and an APR is performed in the Dutch and MRC CRO-7 trials of short-
and when the tumour is located anteriorly. Short- course radiotherapy [24, 28, 32]. In both studies,
course radiotherapy with immediate surgery has mesorectal grade was found to be an indepen-
no impact on CRMI, but chemoradiotherapy and dent predictor of local recurrence. In the Dutch
delayed surgery cause tumour regression; conse- study, there was in addition a significant asso-
quently the CRM is less often positive [8]. ciation between plane of surgery and survival in
The CRM can be involved by direct tumour patients with an uninvolved CRM. Trial data and
spread, vascular invasion, perineural spread or single-centre series indicate that deep defects in
lymph node metastasis. CRMI due to nodal dis- the mesorectum exposing muscularis propria are
ease is uncommon according to a recent publica- seen in 13–32% of operations, and that incom-
tion from the Mercury group where it occurred in plete mesorectal excisions are more common
<2% of all cancers and accounted for one in ten after abdominoperineal resection and where the
of all positive margins [39]. When assessing CRM is positive [14, 15, 24, 28, 32]. It is impor-
lymph nodes close to the circumferential margin, tant in the case of APR specimens however, not
it is important for the pathologist to be aware that to confuse normal anatomical thinning of the
the distance measured is that from the edge of the mesorectum as one approaches the pelvic floor
intranodal tumour deposit rather than the distance (ano-rectal junction) with ‘coning’ by the surgeon
from the lymph node capsule. Two studies have who may inadvertently cut into distal mesorectal
looked at the relationship between mode of CRMI fat as he dissects deep into the pelvis.
and local recurrence. Both have reported that
recurrence rates in patients with CRMI due to
nodal involvement are identical to those with a 30.3.5 APR Grading
negative margin [6, 24, 28]. Until this is corrobo-
rated by larger studies however, tumour within a There is currently some variation between sur-
lymph node <1 mm from the CRM is still geons in how an abdominoperineal resection is
classified as CRMI. performed [52]. While some operators dissect
312 N. Scott

following: (1) intramuscular/submucosal plane


a
where perforation has occurred or the surgeon has
dissected into muscularis propria, (2) sphincteric
plane where the surgeon has dissected around the
sphincter muscle tube or (3) extralevator plane
where the levator muscle is removed en bloc and
the specimen has a cylindrical appearance [27].
It remains to be seen whether this classification
will have the same prognostic significance as
mesorectal grading, but it will undoubtedly assist
b in a more systematic audit of a difficult surgical
procedure.

30.3.6 Local Peritoneal Involvement

Rectal tumours located at or above the level of


the anterior peritoneal reflection may ulcerate the
peritoneal surface and are classified as TNM
stage pT4a. Few studies have specifically
addressed this pathological feature which occurs
in 5–27% of resected cancers, but there is grow-
Fig. 30.4 (a) Abdominoperineal resection specimen
ing evidence that microscopic peritoneal involve-
with ‘mesorectal plane excision’: mesorectum is ment is not only associated with an increased risk
bulky with smooth surface and no obvious defects. (b) of loco-regional failure but is also predictive of
Abdominoperineal resection specimen with ‘muscularis overall survival [36]. In a recent publication,
propria plane excision’: mesorectum is irregular with tat-
tered surface and multiple defects exposing muscular wall
Mitchard et al. suggested that between 27% and
50% of local recurrences might be attributable to
peritoneal seeding [22]. Peritoneal involvement
in the UK and USA is diagnosed according to
down and around the mesorectal fascia to the pel- descriptions provided by Shepherd et al. where
vic floor and ano-rectal junction, others perform tumour cells must be seen at the peritoneal sur-
a wider perineal resection, excising the levator face associated with inflammation or mesothelial
muscle which is then left covering the distal part ulceration [37]. We have found the Shepherd
of the surgical specimen (extralevator abdomi- classification to be highly reproducible between
noperineal excision). For the pathologist the first experienced GI pathologists (kappa values rang-
procedure results in an APR specimen with an ing between 0.74 and 0.89) and would strongly
hourglass appearance and narrow waist at the recommend its use. In our study of over 100 cases
inferior limit of the mesorectum. If the tumour of pT3/pT4 colorectal cancer, at least 3 out of 4
occupies this area, even very superficial inva- pathologists agreed on the presence of LPI in
sion through the muscularis propria or internal more than 90% of examinations [17].
anal sphincer will result in a positive CRM. If,
however, an extralevator excision is performed,
the specimen has a cylindrical shape and CRMI 30.3.7 Vascular Invasion
is reduced. To compliment mesorectal grading
and facilitate the audit of surgery for low rectal Multiple studies have shown that vascular inva-
cancer, Nagtegaal et al. proposed that APR speci- sion is a significant prognostic factor in rectal
mens should be classified by the pathologist as the cancer and is correlated with haematogenous
30 What Is the Correct Procedure for Handling the Surgical Specimen? 313

Fig. 30.5 (a) H&E stained


section (×200) of extramural a
vein invasion: this lesion
might easily be missed as
vessel wall is difficult to
identify and lumen is
obliterated by carcinoma.
(b) Elastin Van Gieson
stained section (×200) of
same area as (a): vein wall is
highlighted by black staining
elastic fibres

metastasis to the liver [23, 45]. In a minority of increase the rate with which invasion is seen.
specimens, invasion of large veins can be recog- Routine use of an elastin stain to highlight the
nised macroscopically in tumour slices, but in vein wall has been evaluated in several studies
most cases the diagnosis is made microscopi- and not only increases detection rate but
cally. The frequency with which vascular improves reproducibility of diagnosis between
involvement is seen varies according to tumour different pathologists [1, 50]. Elastin stains are
stage and grade and the method of detection. particularly helpful where a small vein is com-
Reported rates vary between less than 20% and pletely occluded by the invading tumour and its
60% or more. Increasing the number of tumour wall partially destroyed (Fig. 30.5). Even with-
blocks examined and taking sections parallel out these techniques however, extramural venous
rather than perpendicular to the bowel wall may invasion (EMVI) should be seen in over 25% of
314 N. Scott

cases and must always be reported. A recent nodes containing metastases will be large and
large population-based study showed that the easily found; 45–50% of involved nodes are
5-year survival of patients with stage II disease <5 mm in diameter [3]!
(Dukes stage B) and EMVI was significantly Most pathologists will identify nodes by a
worse than that seen in patients with a single combination of thin slices through the mesorec-
metastatic lymph node [23]. EMVI therefore, tal and mesocolic fat, visual inspection and
helps to identify a group of patients with ‘high- palpation. Fat clearance techniques and lymph
risk’ Dukes B cancers who may benefit from node enhancement solutions e.g., GEWF, can
adjuvant chemotherapy. enhance the lymph node yield but have not yet
been widely adopted, presumably owing to cost,
the greater time required and perceived difficulty
30.3.8 Lymph Node Metastasis of incorporating such techniques into a busy cut-
and pN Stage up schedule. There is also little evidence that
these methods increase the frequency with which
Rectal adenocarcinomas are metastatic to regional patients are classified as having nodal metasta-
lymph nodes contained within the surgical speci- ses (pN1/pN2) despite substantially improving
men (mesorectal, superior rectal artery and inferior overall lymph node yield. Similarly, while there
mesenteric artery nodes) in 35–50% of patients. are one or two studies suggesting that more posi-
Lateral pelvic lymph nodes (also regarded as tive nodes can be found by cassetting individual
regional nodes in the TNM classification) may lymph nodes separately, processing the entire
be involved by tumour in as many as 10–25% of node for microscopic examination, serial slicing
patients with pT3/pT4 cancers lying distal to the larger nodes and examining smaller nodes at three
peritoneal reflection, but it is unusual in European or more levels, the evidence is still insufficient
and North American surgical practice for the sur- to make clear recommendations on this subject
geon to perform a lateral pelvic lymph node dis- [49]. For the time being most pathologists will
section, and these nodes are rarely seen by the embed only a fraction of larger nodes and exam-
Western pathologist. ine a single section at a single level.
It is essential that time and effort are spent on Macroscopic (and microscopic) tumour nod-
finding as many lymph nodes as possible in the ules may occasionally be found in the mesorectal
resection specimen. The total number of lymph fat unassociated with residual lymph node struc-
nodes found and the number containing metasta- tures. These have been variously called satellite
ses should always be reported. Although pNX is tumour deposits and mesorectal microfoci [12,
applied only to cases where no lymph nodes at all 33]. In some cases there is convincing evidence
can be found, the UICC guidelines advise that in that these deposits have arisen as a result of
most cases 12 or more nodes should be identified. perineural or vascular spread; in other instances,
Evidence from several studies indicates that the it is surmised that they represent lymph nodes
frequency of lymph node positivity increases outgrown by metastatic tumour deposits. While
with the number of lymph nodes found and that these lesions are clearly associated with more
patients staged as node negative (TNM stage II) advanced cancers and reduced survival, incorpo-
on the basis of a small lymph node harvest (the rating them into the TNM staging system has
exact figure varies between studies) have a caused considerable controversy. Older iterations
significantly worse survival outcome than those have regarded them as lymph node deposits
where larger numbers of negative nodes are found if 3 mm in size (fifth edition) or, alternatively,
[23, 48]. While there are other potential explana- when they possess the rounded outline of a node,
tions for this relationship, it seems very likely irrespective of size (sixth edition) [41, 42]. The
that failure to sample an adequate number of current seventh edition removes these
nodes in the surgical specimen risks understaging qualifications and places them in a distinct cate-
the patient’s disease. The pathologist needs to be gory of lymph node metastasis i.e., pN1c [43]. In
aware that it cannot be taken for granted that the UK, the Royal College of Pathologists has
30 What Is the Correct Procedure for Handling the Surgical Specimen? 315

advised retaining the fifth edition of the TNM complete regression has not occurred, the patholo-
system for routine reporting, but it is question- gist needs to sample the remaining cancer widely
able whether all pathologists are able or willing enough to allow accurate assessment of lesser
to apply these rules consistently. A recently pub- degrees of tumour response and provide reliable
lished pathological review of colorectal cancers staging. In our experience regression can vary quite
from the UK and Sweden showed that reporting markedly between different parts of the same
of these lesions was most reproducible using cri- tumour, a fact which may be missed if only limited
teria described in the fifth edition and that the sampling is performed. While there is no univer-
result of successive changes to the classification sally agreed protocol for sampling tumours follow-
has been to increase the proportion of pN1/pN2 ing neoadjuvant treatment, Quirke has suggested
cases in these particular series from 34% using that where no residual tumour is visible macro-
the fifth edition to 43% using the seventh [26]. scopically, the whole tumour site should be sliced
at 5–8-mm intervals and processed for microscopy
[30]. If tumour cells are still not seen after com-
30.3.9 Examination of the Specimen pletely embedding the lesion, each block is to be
After Neoadjuvant Radiotherapy examined at three levels. Where macroscopic
tumour persists, at least four blocks should be sam-
Over the past 10–15 years it has become common- pled including one large (whole mount) block.
place for surgery to be performed after a period of It is also widely recognised that identification
preoperative radiotherapy with or without con- of lymph nodes is more difficult following preop-
comitant chemotherapy. This usually takes the erative radiotherapy and lymph node harvests are
form of either short-course radiotherapy adminis- significantly reduced. It is our experience, how-
tered over 5 days followed by resection a week ever, that it is still possible to find more than 12
later or chemoradiotherapy given over several nodes in most cases if a careful examination is
weeks with a delay of 6–8 weeks before surgery. performed. Interestingly, a recent study suggests
After short-course radiotherapy and early surgery that the relationship between low lymph node
the pathologist will see acute radiation proctitis in numbers and poorer survival in stage II cancers
the peritumoural mucosa but little or no evidence described for patients treated by surgery alone
of tumour regression. Results from the Dutch does not hold true after radiotherapy [34]. This
TME-radiotherapy trial found a modest increase in needs to be validated however.
the frequency of poorly differentiated mucinous Reporting of T and N stage CRMI, histologi-
tumours following this regimen but no evidence of cal type and grade, peritoneal involvement and
downstaging [19]. In contrast, chemoradiotherapy vascular invasion is identical to that for the
with delayed surgery will produce complete untreated specimen except that the prefix ‘y’ is
tumour disappearance (ypT0 ypN0) in 10–20% of used to denote that staging has been performed
cases and lesser degrees of regression in many of after preoperative radiotherapy, e.g., ypT2 ypN1.
the others. In these specimens it may be difficult to Tumour regression grade (TRG) should also be
identify the tumour macroscopically or reliably reported, although its exact role in patient man-
differentiate residual carcinoma from post-radio- agement is still debated. There is good evidence
therapy fibrosis. While there is some correlation that TRGs are prognostic, but it is not clear if
between macroscopic appearances and degree of they offer any additional information that is not
regression, in up to a third of cases where there is a provided by TNM stage and CRM status. Despite
good clinical response i.e., minor mucosal abnor- this many oncologists will request tumour regres-
mality or ulcer scar, residual malignant cells can sion grading as a measure of the tumour’s radio
still be demonstrated microscopically [5]. In these and chemosensitivity and the likelihood of
cases it is important that the pathologist performs a response to future chemotherapy. The most com-
sufficiently thorough examination to confirm com- monly employed TRG is that developed by
plete tumour regression which has been shown Mandard et al. for oesophageal cancer [18].
to have a very good prognosis [51, 53]. Where Dworak described a similar system for colorectal
316 N. Scott

Table 30.1 Tumour regression grading: comparison of three commonly used classifications
Mandard et al. [18] TRG 1 TRG 2 TRG 3 TRG 4 TRG 5
No residual Rare residual Fibrosis Residual cancer Absence of
cancer cancer cells outgrowing outgrowing regressive changes
residual cancer fibrosis
Royal College of TRG 1 TRG 2 TRG 3
Pathologists [54]
No residual Minimal residual No marked regression
tumour cells, tumour. Occasional
mucus lakes microscopic tumour
only foci identified with
difficulty
Beddy et al. [4] TRG 1 TRG 2 TRG 3
No cancer cells, single cells or Residual cancer Fibrosis outgrown by cancer or no
small groups of cancer cells outgrown by fibrosis with extensive residual
fibrosis cancer

cancer, and more recently, Beddy et al. and the meetings that we understand which pieces of
Royal College of Pathologists have proposed a information are important for clinical decision
simpler classification with three grades instead of making and where the uncertainties lie. The
five [4, 10]. These have the advantage of being recent emphasis on quality control of diag-
more reproducible but still retaining prognostic nosis and treatment and the introduction of
significance. Three of these TRG classifications national guidelines with explicit recommenda-
are described and compared in Table 30.1. tions for surgical and pathological audit have
Regressed primary tumours and lymph created a new role for pathologists who must
node metastases may leave behind areas of now describe and record features which not
inflammation, fibrosis, calcification and pools of only have a prognostic function, and guide the
mucin. These are not to be regarded as evidence selection of adjuvant treatments, but are also a
of residual tumour unless tumour cells are actu- measure of the quality of surgery performed.
ally seen and do not count towards ypT stage, If the pathologist is to achieve these aims
ypN stage or CRM involvement. It may be neces- there must be a standardised, evidence-based
sary however, to examine multiple levels through approach to examining the rectal cancer speci-
an area of mucin accumulation before one can be men and an agreed minimum data set which is
sure that no malignant cells are present, and very collected on every case. In this chapter, I have
occasionally immunohistochemistry is required set out to describe what I believe to be the most
to distinguish between tumour cells and mac- important elements of rectal cancer reporting
rophages. In a series of 108 patients undergo- and the evidence, as it currently stands, for
ing long-course chemoradiation at the Memorial their inclusion in the pathologist’s report.
Sloan-Kettering Cancer Centre, acellular mucin
pools were found in 15% of irradiated cancers
[38]. Follow-up confirmed that the presence of
these pools had no significant impact on survival. References
Conclusion 1. Abdulkader M, Abdulla K, Rakha E, Kaye P (2006)
The effective management of rectal cancer Routine elastic staining assists detection of vascular
cannot be achieved without the collaboration invasion in colorectal cancer. Histopathology
49:487–492
of pathologists, radiologists, surgeons and 2. Adam IJ, Mohamdee MO, Martin IG et al (1994) Role
oncologists cooperating in a multidisciplinary of circumferential margin involvement in the local
team. It is only by working together and regular recurrence of rectal cancer. Lancet 344:707–711
30 What Is the Correct Procedure for Handling the Surgical Specimen? 317

3. Andreola S, Leo E, Belli F et al (1996) Manual dissec- 17. Littleford SE, Baird A, Rotimi O, Verbeke C, Scott N
tion of adenocarcinoma of the lower third of the rec- (2009) Interobserver variation in the reporting of local
tum specimens for detection of lymph node metastases peritoneal involvement and extramural venous inva-
smaller than 5 mm. Cancer 77:607–612 sion in colon cancer. Histopathology 55:407–413
4. Beddy D, Hyland JMP, Winter DC et al (2008) A 18. Mandard A, Dalibard F, Mandard J et al (1994)
simplified tumor regression grade correlates with sur- Pathologic assessment of tumor regression after pre-
vival in locally advanced rectal carcinoma treated operative chemoradiotherapy of esophageal carci-
with neoadjuvant chemoradiotherapy. Ann Surg noma. Cancer 73:2680–2686
Oncol 15:3471–3477 19. Marijnen CAM, Nagtegaal ID, Kranenbarg EK et al
5. Bedrosian I, Rodriguez-Bigas MA, Feig B et al (2004) (2001) No downstaging after short-term preoperative
Predicting the node-negative mesorectum after preop- radiotherapy in rectal cancer patients. J Clin Oncol
erative chemoradiation for locally advanced rectal 19:1976–1984
carcinoma. J Gastrointest Surg 8:56–63 20. Mercury Study Group (2007) Extramural depth of
6. Birbeck KF, Macklin CP, Tiffin NJ et al (2002) Rates tumour invasion at thin-section MR in patients with
of circumferential resection margin involvement vary rectal cancer: results of the MERCURY Study.
between surgeons and predict outcomes in rectal can- Radiology 243:132–139
cer surgery. Ann Surg 235:449–457 21. Merkel S, Mansmann U, Siassi M, Papadopoulos T,
7. Branston LK, Greening S, Newcombe RG et al Hohenberger W, Hermanek P (2001) The prognostic
(2002) The implementation of guidelines and com- inhomogeneity in pT3 rectal carcinomas. Int J
puterised forms improves the completeness of can- Colorectal Dis 16:298–304
cer pathology reporting. The CROPS project: a 22. Mitchard JR, Love SB, Baxter KJ, Shepherd NA
randomised controlled trial in pathology. Eur J (2010) How important is peritoneal involvement in
Cancer 38:743–744 rectal cancer ? A prospective study of 331 cases.
8. Bujko K, Nowacki MP, Nasierowska-Guttmejer A Histopathology 57:671–679
et al (2006) Long term results of a randomized trial 23. Morris EJA, Maughan NJ, Forman D, Quirke P (2007)
comparing preoperative short-course radiotherapy Who to treat with adjuvant therapy in dukes B/stage II
with preoperative conventionally fractionated chemo- colorectal cancer? The need for high quality pathol-
radiation for rectal cancer. Br J Surg 93:1215–1223 ogy. Gut 56:1419–1425
9. Cawthorn SJ, Parums DV, Gibbs NM et al (1990) 24. Nagtegaal ID, Marijnen CAM, Kranenbarg EK et al
Extent of mesorectal spread and involvement of lat- (2002) Circumferential margin involvement is still an
eral resection margin as prognostic factors after sur- important predictor of local recurrence in rectal carci-
gery for rectal cancer. Lancet 335:1055–1059 noma: not one millimetre but two millimetres is the
10. Dworak O, Keilholz L, Hoffman A (1997) Pathological limit. Am J Surg Pathol 26:350–357
features of rectal cancer after preoperative radiochemo- 25. Nagtegaal ID, Quirke P (2008) What is the role for the
therapy. Int J Colorectal Dis 12:19–23 circumferential margin in the modern treatment of
11. Engelen SME, Beets-Tan RGH, Lahaye MJ, Kessels rectal cancer. J Clin Oncol 26:303–312
AGH, Beets GL (2008) Location of involved mesorec- 26. Nagtegaal ID, Tot T, Jayne DG et al (2011) Lymph
tal and extramesorectal lymph nodes in patients with nodes, tumor deposits and TNM: are we getting better
primary rectal cancer: preoperative assessment with ? J Clin Oncol 29:2487–2492
MR imaging. Eur J Surg Oncol 34:776–781 27. Nagtegaal ID, Van de Velde CJH, Marijnen CAM,
12. Goldstein NS, Turner JR (2000) Pericolonic tumor Van Krieken JHJM, Quirke P (2005) Low rectal can-
deposits in patients with T3N + M0 colon adenocarci- cer: a call for a change of approach in abdomino-
nomas. Cancer 88:2228–2238 perineal resection. J Clin Oncol 23:9257–9264
13. Hamilton SR, Bosman FT, Boffetta P et al (2010) 28. Nagtegaal ID, Van de Velde CJH, Van der Worp E
Carcinoma of the colon and rectum. In: Bosman FT, et al (2002) Macroscopic evaluation of rectal cancer
Carneiro F, Hruban RH, Theise ND (eds) WHO resection specimen: clinical significance of the pathol-
classification of tumours of the digestive system. ogist in quality control. J Clin Oncol 20:1729–1734
IARC, Lyon 29. Quirke P, Durdey P, Dixon MF, Williams NS (1986)
14. Jeyarajah S, Sutton CD, Miller AS, Hemingway D Local recurrence of rectal adenocarcinoma due to
(2007) Factors that influence the adequacy of total inadequate surgical resection. Histopathological study
mesorectal excision for rectal cancer. Colorectal Dis of lateral tumour spread and surgical excision. Lancet
9:808–815 2(8514):996–998
15. Leonard D, Penninckx F, Fieuws S et al (2010) Factors 30. Quirke P (2003) CORE study: capecitabine/oxaliplatin,
predicting quality of total mesorectal excision for rec- radiotherapy and excision protocol Study No. C8601.
tal cancer. Ann Surg 252:982–988 Pathological technique. Sanofi-Aventis, Appendix 7.
16. Lewin MR, Fenton H, Burkart AL et al (2007) Poorly Lancet Oncol 4(11):695–702
differentiated colorectal carcinoma with invasion 31. Quirke P, Morris E (2007) Reporting colorectal can-
restricted to lamina propria (intramucosal carcinoma): cer. Histopathology 50:103–112
a follow-up study of 15 cases. Am J Surg Pathol 32. Quirke P, Steele R, Monson J et al (2009) Effect of the
31:1882–1886 plane of surgery achieved on local recurrence in
318 N. Scott

patients with operable rectal cancer: a prospective colorectal carcinoma: the efficacy of various types of
study using data from the MRC CR07 and NCIC- tissue blocks. J Clin Pathol 59:207–210
CTG C016 randomised clinical trial. Lancet 373: 45. Talbot IC, Ritchie S, Leighton MH, Hughes AO,
821–828 Bussey HJR, Morson BC (1980) The clinical
33. Ratto C, Ricci R, Rossi C, Morelli U, Vecchio FM, significance of invasion of veins by rectal cancer. Br J
Doglietto GB (2002) Mesorectal microfoci adversely Surg 67:439–442
affect the prognosis of patients with rectal cancer. Dis 46. Taylor FGM, Quirke P, Heald RJ et al (2011)
Colon Rectum 45:733–743 Preoperative high-resolution magnetic resonance
34. Rullier A, Laurent C, Capdepont M et al (2008) imaging can identify good prognosis stage I, II and III
Lymph nodes after preoperative chemoradiotherapy rectal cancer best managed by surgery alone. Ann
for rectal carcinoma: number, status and impact on Surg 253:711–719
survival. Am J Surg Pathol 32:45–50 47. Tekkis PP, Heriot AG, Smith J, Thompson R, Finan P,
35. Sebag-Montefiore D, Stephens RJ, Steele R et al Stamatakis JD (2005) Comparison of circumferential
(2009) Preoperative radiotherapy versus selective margin involvement between restorative and non-
postoperative chemoradiotherapy in patients with rec- restorative resections for rectal cancer. Colorectal Dis
tal cancer (MRC CR07 and NCIC-CTG C016): a 7:369–374
multi-centre, randomised trial. Lancet 373:811–820 48. Tepper JE, O’Connell MJ, Niedzwiecki D et al (2001)
36. Shepherd NA, Baxter KJ, Love SB (1995) Influence Impact of number of nodes retrieved on outcome in
of local peritoneal involvement on pelvic recurrence patients with rectal cancer. J Clin Oncol 19:157–163
and prognosis in rectal cancer. J Clin Pathol 48: 49. Van Wyk Q, Hosie KB, Balsitis M (2000)
849–855 Histopathological detection of lymph node metastases
37. Shepherd NA, Baxter KJ, Love SB (1997) The prog- from colorectal carcinoma. J Clin Pathol 53:685–687
nostic importance of peritoneal involvement in colonic 50. Vass DG, Ainsworth R, Anderson JH, Murray D,
cancer: a prospective evaluation. Gastroenterology Foulis AK (2004) The value of an elastic tissue stain
112:1096–1102 in detecting venous invasion in colorectal cancer.
38. Shia J, McManus M, Guillem JG et al (2011) J Clin Pathol 57:769–772
Significance of acellular mucin pools in rectal carci- 51. Vecchio FM, Valentini V, Minsky BD et al (2005) The
noma after neoadjuvant chemoradiotherapy. Am J relationship of pathologic tumor regression grade
Surg Pathol 35:127–134 (TRG) and outcomes after preoperative therapy in
39. Shihab OC, Quirke P, Heald RJ, Moran BJ, Brown G rectal cancer. Int J Radiat Oncol Biol Phys 62:
(2010) Magnetic resonance imaging detected lymph 752–760
nodes close to the mesorectal fascia are rarely a cause 52. West NP, Anderin C, Smith KJE, Holm T, Quirke P
of margin involvement after total mesorectal excision. (2010) Multicentre experience with extralevator
Br J Surg 97:1431–1436 abdominoperineal excision for low rectal cancer. Br J
40. Shirouzu K, Isomoto H, Kakegawa T (1995) Distal Surg 97:588–599
spread of rectal cancer and optimal distal margin of 53. Wiig JN, Larsen SG, Dueland S, Giercksky K (2005)
resection for sphincter-preserving surgery. Cancer Clinical outcome in patients with complete pathologic
76:388–392 response (pT0) to preoperative irradiation/chemo-
41. Sobin LH, Wittekind C (eds) (1997) International irradiation operated for locally advanced or locally
union against cancer TNM classification of malignant recurrent rectal cancer. J Surg Oncol 92:70–75
tumors, 5th edn. Wiley, Hoboken 54. Williams GT, Quirke P, Shepherd NA (2007) The
42. Sobin LH, Wittekind C (eds) (2002) International Royal College of Pathologists dataset for colorectal
union against cancer TNM classification of malignant cancer, 2nd edn. The Royal College of Pathologists,
tumors, 6th edn. Wiley, Hoboken London, http://www.rcpath.org=ersol;resources/pdf/
43. Sobin LH, Gospodarowicz M, Wittekind C (eds) (2009) colorectalcancer.pdf
International union against cancer TNM classification 55. Wittekind C, Compton C, Quirke P et al (2009) A uni-
of malignant tumors, 7th edn. Wiley, Hoboken form residual tumor (R) classification. Cancer
44. Sternberg A, Mizrahi A, Amar M, Groisman G (2006) 115:3483–3488
Detection of venous invasion in surgical specimens of
What Is the Prognostic
Value of (y)pT and (y)pN? 31
Nadine Ectors

Contents 31.1 Introduction


31.1 Introduction ................................................... 319
Based on scientific evidence gathered over a couple
31.2 ‘pT’: Pathologic Classification of Primary
Tumour Extent .............................................. 320
of decades, total mesorectal excision (TME) has
been introduced as the standard surgical tech-
31.3 ‘pN’: Pathologic Classification of Lymph
nique in rectal cancer and neoadjuvant chemo-
Node Involvement.......................................... 321
radiation (CRT) has become standard practice
31.4 SLN: Sentinel Lymph Nodes ........................ 322 for advanced rectal cancers. Both therapeuti-
31.5 Isolated Tumour Cells (ITC) and cal approaches, TME and CRT, have generated
Micrometastasis............................................. 323 specific issues related to pathological assessment
31.6 In Between ‘T’ and ‘N’ and Close to ‘V’: of rectal resection specimens (see Part VII). The
Tumour Deposits ........................................... 323 macroscopic and microscopic pathologic assess-
31.7 CRT: y(pT) and y(pN) .................................. 324 ment of the resection specimen provides the
opportunity to achieve the most accurate esti-
References ................................................................. 325
mation of prognosis. In general, rectal cancer is
slow-growing and spreads in a mostly predictable
and gradual fashion. This mode of direct spread
is the basis of anatomical staging. The anatomic
extent of rectal cancer being, still, by far the most
important prognostic factor.
Dr. Cuthbert E. Dukes (1890–1977), a patholo-
gist at St. Mark’s Hospital in London, proposed
in 1929 a classification of rectal cancer based on
findings from pathological examination of the
resected bowel. The classification reflected the
extent of spread of the cancer in view of its prog-
nostic significance. This classification has been
N. Ectors subjected to some revisions and adaptations caus-
Department of Imaging and Pathology, ing some confusion and misinterpretations [30].
University Leuven,
Anyhow, the Dukes’ classification was cited in
Leuven, Belgium
terms of ‘American chaos versus British order’
Activity Center Biobanking, University Hospitals
[31]. The TNM staging system of the American
Leuven, Campus Gasthuisberg, Herestraat 49, 3000
Leuven, Belgium Joint Committee on Cancer (AJCC) and the
e-mail: nadine.ectors@uzleuven.be Union for International Cancer Control (UICC)

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 319


DOI 10.1007/978-3-642-25005-7_31, © Springer-Verlag Berlin Heidelberg 2012
320 N. Ectors

Table 31.1 TNM classification of malignant tumours (seventh edition) – digestive system tumours: colon and
rectum
T Primary tumour
TX Primary tumour cannot be assessed
T0 No evidence of primary tumour
Tis Carcinoma in situ: intraepithelial or invasion of lamina propria
T1 Tumour invades submucosa
T2 Tumour invades muscularis propria
T3 Tumour invades subserosa or into non-peritonealized pericolic or perirectal tissue
T4 Tumour directly invades other organs or structures and/or perforates visceral peritoneum
T4a Tumour perforates visceral peritoneum
T4b Tumour directly invades other organs or structures
N Regional lymph nodesa
NX Regional lymph nodes cannot be assessed
N0 No regional lymph node metastasis
N1 Metastasis in 1–3 regional lymph nodes
N1a Metastasis in one regional lymph node
N1b Metastasis in 2–3 regional lymph nodes
N1c Tumour deposit(s), i.e. satellitesb in the subserosa, or in non-peritonealized pericolic or perirectal soft
tissue without regional lymph node metastasis
N2 Metastasis in four or more regional lymph nodes
N2a Metastasis in 4–6 regional lymph nodes
N2b Metastasis in seven or more regional lymph nodes
M Distant metastasis
M0 No distant metastasis
M1 Distant metastasis
M1a Metastasis confined to one organ (liver, lung, ovary, non-regional lymph node(s))
M1b Metastasis in more than one organ or the peritoneum
Changes in TNM7 compared to TNM6
a
Regional lymph nodes of the rectum: superior, middle and inferior rectal (haemorrhoidal), inferior mesenteric, internal
iliac, mesorectal (paraproctal), lateral sacral, presacral and sacral promontory (Gerota). Metastasis in nodes other than
those listed above (e.g., external iliac or common iliac nodes) is classified as distant metastasis
b
Tumour deposits (satellites), i.e. macroscopic or microscopic nests or nodules, in the pericolorectal adipose tissue’s
lymph drainage area of a primary carcinoma without histological evidence of residual lymph node in the nodule, may
represent discontinuous spread, venous invasion with extravascular spread (V1/2) or a totally replaced lymph node
(N1/2). If such deposits are observed with lesions that would otherwise be classified as T1 or T2, then the T classification
is not changed, but the nodule(s) is recorded as N1c. If a nodule is considered by the pathologist to a be a totally replaced
lymph node (generally having a smooth contour), it should be recorded as a positive lymph node and not as a satellite,
and each nodule should be counted separately as a lymph node in the final pN determination

(first publication in 1940) is nowadays the most been modified over the years, up to the seventh
universal cancer staging system. The T (tumour), N edition, attempting to follow the pace of progress
(node) and M (metastasis) staging system classifies and evidence-based developments in oncology
tumours according to the anatomic extent. Tumour (Table 31.1) [39, 40].
staging according to the TNM system is currently
accepted as the strongest prognostic parameter for
different types of cancers, including rectal cancers. 31.2 ‘pT’: Pathologic Classification
Staging provides clinically applicable prognostic of Primary Tumour Extent
information for individual patient management
as well as standardization allowing interpreta- Tumour extent is classified as Tis, T1, T2, T3 and
tion and comparison of evidence-based data at a T4. Changes and discussions relate to sub-
much broader scale. The TNM classification has classification.
31 What Is the Prognostic Value of (y)pT and (y)pN? 321

Unlike other organs, for colorectal carcino- recently [25]. According to the SEER analyses,
mas, ‘carcinoma in situ’ (pTis: Carcinoma in situ: prognosis for patients with pT4a is (10–20%
intraepithelial or invasion of lamina propria) 5-year survival) better than prognosis for pT4b,
includes lesions limited to the epithelium with an independently of the N category [10].
intact basement membrane as well as invasive For rectal cancer, invasion of the external
lesions extending into the mucosal lamina pro- sphincter is classified as pT3, while invasion into
pria up to but not through the muscularis muco- the levator ani muscle(s) is classified as pT4 [43].
sae (intramucosal carcinoma). The underlying
argument being that the lamina propria of the rec-
tum contains extremely few lymphatics. However, 31.3 ‘pN’: Pathologic Classification
the direct link between the number of lymphatic of Lymph Node Involvement
vessels and the associated risk of lymph node
metastasis has been questioned recently [34]. Invasion in regional lymph nodes is classified as
In pT1 tumours (invasion through the mus- N0, N1 and N2. The risk of metastasis to regional
cularis mucosae into the submucosa but not lymph nodes correlates with the T category and
into the muscularis propria), the frequency of other parameters (e.g. grade).
lymph node metastasis relates to the depth of The number of positive lymph nodes affects
invasion, i.e. the superficial (2%), middle (8%) prognosis for most TN categories of rectal can-
and deep (23%) thirds of the submucosa [12, cer. The categories pN1 (1–3 positive regional
34]. This sub-classification is especially useful LN) and pN2 (³4 positive regional LN) remained
for the assessment of local excisions of early unchanged in TNM7. However, both were sub-
lesions. classified in a and b based on numbers of involved
Tumour extension into the muscularis propria lymph nodes (Table 31.1). Patients staged pN1a
(pT2), into the subserosa or the non-peritoneal- (1 positive regional LN) were shown to have a
ized pericolic or perirectal tissue (pT3) and into 3–10% better 5-year relative and observed sur-
other organs or structures and/or perforating vis- vival compared to pN1b (2–3 positive regional
ceral peritoneum (pT4) has been subject to split- LN), while pN2a (4–6 positive regional LN) have
ting and lumping manoeuvres in, e.g. the TNM a 5–20% better 5-year survival than pN2b (³7
and Japanese staging systems. For example, the positive regional LN) [10]. Nevertheless, some
sub-classification of pT3 was included in the authors persist arguing on the basis and validity
third edition of the TNM supplement (2003) and of the cut-off values for sub-classification.
is included in the Japanese classification [44]. A high-quality, i.e. complete ‘total mesorectal
Data confirming the impact of pT3 sub- excision’ (TME), specimen includes the primary
classification on prognosis of patients with rectal tumour as well as all regional lymph nodes [1,
cancer exist; however, it did not find wide accep- 24, 33]. The accuracy and predictive value of
tance probably because of discussions on the stage II (pT3,T4-N0M0) assignment are directly
basis and validity of the cut-off values for sub- proportional to the quality of the TME, i.e., the
classification and the lack of standardized meth- inclusion of all regional lymph nodes and the
ods of assessment [25]. More recently, TNM7 pathological examination of the specimen, i.e.
introduced differentiation between pT4a and the retrieval and harvest of all regional lymph
pT4b, which was previously also addressed in the nodes. The minimal acceptable harvest has been
third edition of the TNM supplement (2003) [44]. set at 12 lymph nodes [5, 6, 36]. However, ‘lymph
Multivariate analysis having demonstrated that node retrieval in rectal cancer is dependent on
involvement of the serosa by tumour (pT4b) is an many factors – the role of the tumour, the patient,
important independent prognostic factor imply- the surgeon, the radiotherapist, and the patholo-
ing an increased risk of distant metastasis and a gist’, not in the least the pathologist [4, 18].
shorter median survival time [17]. Whether the Although these authors tend to settle for a median
risk incurred in pT4a differs significantly from of at least 8 lymph nodes, a number of studies
pT4b and how they relate has been questioned indicate that examination of increased numbers
322 N. Ectors

of lymph nodes is, in itself, associated with an prognostic significance of apical-node metasta-
increased survival advantage (e.g. 18). Moreover, sis around the inferior mesenteric artery (IMA)
it has been shown in rectal cancer that lymph remains unclear [16]. Therefore, ‘Western’ prac-
node metastasis are often found in small lymph tice does not advocate (systematic) extensions of
nodes (<5 mm diameter) [43]. lymphadenectomies.
More recently, the debate has moved away Metastasis to non-regional lymph nodes, e.g.
from the exact number of lymph nodes towards external iliac or common iliac nodes, is classified
the lymph node ratio (LNR) as an even better as distant metastasis (M1).
indicator of prognosis [2, 3, 8, 15, 24, 30]. If the
LNR can be considered as a prognostic factor, it
may have the advantage to be less dependent on 31.4 SLN: Sentinel Lymph Nodes
the number of retrieved lymph nodes than stage
[7]. Moreover, in a systemic review, it was pointed Sentinel lymph node mapping is the standard
out that marked heterogeneity exists within stage nodal staging technique in patients with breast
III patients and LNR may prove useful to stratify cancer without gross nodal involvement. This
outcome in those patients [2]. Although data look procedure allows to adapt the extent of the resec-
promising, both publications as well as the SEER tion on a case by case basis and thus to avoid
analysis concluded that further evidence is needed unnecessary extensive surgery, i.e. axillary
[2, 7, 10]. Independently of the potential of LNR, lymphadenectomy, to decrease the rate of com-
it is crucial that the pathology report should plications and side effects and to improve quality
clearly indicate the number of positive lymph of life. The concept is based on the existence and
nodes as well as the total number of lymph nodes understanding of an orderly and sequential drain-
examined. age of lymphatic fluid, carrying tumour cells
Since the fifth edition of the TNM (1997), the from the primary tumour through lymphatic ves-
classification of positive lymph nodes according sels and lymph nodes ultimately into the blood
to the location along (named) arteries (introduced circulation. The procedure consists of preopera-
as pN3 in TNM4) has been discarded, this in con- tive injection and tracing of a marker (blue dye,
trast to the actual Japanese classification [35]. No radiocolloids) in order to identify and dissect a
major impact towards patient management is to sentinel lymph node. The lymph node is sub-
be expected. jected to a preoperative extensive pathological
The regional lymph nodes of the rectum are investigation including multiple serial levels with
superior, middle and inferior rectal (haemor- (in a second time) special/ancillary techniques
rhoidal), inferior mesenteric, internal iliac, (immunohistochemistry, qRT-PCR).
mesorectal (paraproctal), lateral sacral, pre- Recently, a number of articles, including sys-
sacral and sacral promontory (Gerota). The ‘total tematic reviews and meta-analysis, have addressed
mesorectal excision’ – which ideally should the issue in rectal cancer [1, 42]. However, the
include all regional lymph nodes – is described underlying surgical motive is not applicable in
in Japanese literature as a ‘limited resection’. rectal cancer: involvement of a sentinel lymph
A Japanese ‘standard resection’ and an ‘extended node will not change the extent of the resection
resection’ include lateral extensions, i.e. respec- on a case by case basis, i.e. the en bloc resection
tively a dissection along the parietal pelvic fascia of the primary rectal cancer including the regional
and the internal iliac artery and a dissection of the lymph nodes; it is the total mesorectal excision,
internal iliac vessels preserving the superior vesi- the cornerstone for locoregional control of rectal
cal artery and obturator nerve [1]. Wide pelvic cancer. Moreover, in rectal cancer, sentinel lymph
lymphadenectomies – which do include lymph nodes have been shown not to be representative
nodes along the internal iliac artery – correlate of involvement of the locoregional lymph nodes
with increased morbidity and a limited, selective, in an important percentage of patients. Finally,
improvement of prognosis [45]. Similarly, the skip metastasis or aberrant metastasis has been
31 What Is the Prognostic Value of (y)pT and (y)pN? 323

noted. Therefore, the drainage of lymphatic fluid ing, implying that this additional staging in other-
in rectal cancer is not sufficiently ‘orderly and wise ‘pN0’ patients may improve identification
sequential’. Theoretically, mapping of the lym- of potential candidates for adjuvant therapy, how-
phatic drainage of an individual rectal cancer ever, up till now without proven benefit [42].
could be taken into consideration when consider- The available data do not provide unequivocal
ing extended lymphadenectomies. Unfortunately, evidence to support classifying micrometastasis
specifically, the lateral lymphatic drainage is as positive lymph nodes, let alone to prescribe
extremely variable. In conclusion, sentinel lymph adjuvant therapy.
node mapping in rectal cancer should be consid-
ered as investigational and does not belong to the
routine clinical practice [13, 20]. 31.6 In Between ‘T’ and ‘N’ and
Close to ‘V’: Tumour Deposits

31.5 Isolated Tumour Cells (ITC) Tumour deposits are defined, in the TNM staging
and Micrometastasis system, as irregular discrete tumour deposits in
perirectal fat away from the leading edge of the
A micrometastasis is defined, in the TNM staging tumour and showing no evidence of residual
system, as a metastasis measuring more than lymph node tissue, but within the lymphatic
0.2 mm and less than or equal to 2.0 mm in diam- drainage of the primary tumour, are considered
eter. Isolated tumour cells (ITC) are defined as peritumoural deposits nodules. The finding of
single tumour cells or small clusters of tumour tumour deposits (satellites or numerous other
cells measuring 0.2 mm in diameter or less. The synonyms) has always confronted pathologists
latter are usually identified by special techniques, with uncertainty concerning the underlying etio-
e.g. immunohistochemistry. pathogenic mechanisms and thus the impossibil-
According to TNM6 and 7, micrometasta- ity to classify them in a prognostic staging
sis should be classified as such (abbreviations: systems, i.e. as T or N factor or neither of them.
mi or mic) and staged as lymph node positive The changes in definitions of tumour deposits
(e.g. pN(mi)), owing to their possible malignant and interpretations in the fifth, sixth and seventh
potentiality [37, 39]. In contrast, isolated tumour editions of the TNM classification clearly dem-
cells (ITC) are considered insignificant and thus onstrate this problem [35, 37, 39]. Unfortunately,
should not be taken into account as lymph node these issues have direct implications concerning
metastasis (pN0). It is explicitly stated that spe- staging and thus treatment of individual patients.
cial techniques (e.g. multiple tissue levels, immu- Tumour deposits were first acknowledged
nohistochemistry, qRT-PCR) to detect ITCs are in TNM5 (1997) [35]. The ‘3-mm rule’ stated
not recommended for the routine examination of that tumour deposits 3 mm or larger in diameter
regional lymph nodes. The pathological report should be regarded as a positive lymph node.
should clearly mention the number of lymph The implementation of the ‘3 mm rule’ was not
nodes involved by micrometastasis and ICTs. evidence-based, acknowledged post-factum, but
The presence of micrometastasis in colorectal had the advantage of being an ‘objective’ crite-
cancer has been previously investigated, but there rium [40]. The next edition of the TNM staging
has been no consensus in relation to their prog- system (TNM6 2002) introduced controversial
nostic significance, which in part is surely due to changes [37]. The ‘3 mm rule’ was withdrawn,
the nature of the studies themselves and other and the definitions of lymph nodes as well as
technical aspects [19]. The prognostic significance vascular invasion were revised. A tumour nodule
of ITCs is still unproven. Some authors of articles in the pericolic/perirectal adipose tissue without
concerning sentinel lymph node mapping do state histological evidence of residual lymph node in
that the main advantage of the sentinel procedure the nodule is classified in the pN category as a
could well be the improvement of the cancer stag- regional lymph node metastasis, as the nodule has
324 N. Ectors

the form and smooth contour of a lymph node. do not know if adjuvant therapy in patients sub-
If the nodule has an irregular contour, it should jected to stage migration, following onto these
be classified in the T category and also coded as interpretations, has a true benefit. Therefore, in
V1 (microscopic venous invasion) or V2 if it was a number of European countries, it was decided
grossly evident, because there is a strong likeli- that the definitions in TNM5 are the most repro-
hood that it represents venous invasion. This ducible and thus should be adhered to, especially
interpretation elicited major concerns on repro- as far as tumoural deposits are concerned [26, 28].
ducibility and thus interobserver variability on the Therefore, it is advisable to indicate the number
one hand and discussion on biological and clini- of TNM version used (e.g. v5, v6, v7) [41].
cal underlying evidence on the other hand. These As a reminder, the objectives for cancer stag-
concerns have been discussed and published in ing were defined by the International Union
great length by Philip Quirke and other European Against Cancer (UICC) TNM Committee almost
pathologists [11, 26]. The paragraph related to the 50 years ago in this order: to aid the clinician in
tumour deposits, and consequently lymph node planning treatment, to give some indication of
and vascular invasion, has been rewritten, but not prognosis, to assist in evaluating the results of
necessarily clarified, in the most recent edition treatment, to facilitate the exchange of informa-
of the TNM staging system (TNM7 2009) [39] tion between treatment centres and, last but
– tumour deposits (satellites), i.e. macroscopic admittedly not least, to contribute to continuing
or microscopic nests or nodules, in the pericol- investigation of human malignancies [9].
orectal adipose tissue’s lymph drainage area of a
primary carcinoma without histological evidence
of residual lymph node in the nodule, may repre- 31.7 CRT: y(pT) and y(pN)
sent discontinuous spread, venous invasion with
extravascular spread (V1/2) or a totally replaced Y(pT) and y(pN) refer to a primary tumour and
lymph node (N1/2). If such deposits are observed lymph nodes that have been previously treated by
with lesions that would otherwise be classified as means of CRT.
T1 or T2, then the T classification is not changed, Neoadjuvant combined treatment in rectal
but the nodule(s) is recorded as N1c. If a nodule cancer is associated with significant tumour
is considered by the pathologist to a be a totally response and downstaging. Minimal residual dis-
replaced lymph node (generally having a smooth ease has been shown to have a better prognosis
contour), it should be recorded as a positive than gross residual disease. Thorough pathologi-
lymph node and not as a satellite and each nodule cal assessment of rectal resection specimens after
should be counted separately as a lymph node in chemoradiation is mandatory (e.g. sampling pro-
the final pN determination. Again, this interpre- tocols, grading systems for tumour response (see
tation elicited major concerns on reproducibility Part VII)) [42] [44].
and thus interobserver variability on the one hand The College of American Pathologists (CAP)
and discussion on biological and clinical under- recommends that (1) tumour regression should
lying evidence on the other hand [21, 27, 40]. In be assessed only in the primary tumour, lymph
the meantime, the literature on tumoural deposits node metastases should not be included in the
has been thoroughly reviewed concluding that, assessment and (2) acellular pools of mucin in
indeed, tumoural deposits have an adverse prog- specimens from patient receiving neoadjuvant
nostic factor [20]. In a comparative study of two therapy are considered to represent completely
independent study populations, it was confirmed eradicated tumour and are not used to assign pT
that tumoural deposits should be taken into stage or counted as positive lymph nodes [43].
account in staging patients with colorectal cancer The latter statement has received scientific sup-
[22]. However, numerous questions remained to port from a recent publication addressing
be addressed as to the definition and reproducibility specifically this issue [32].
and thus interobserver variability before applica- After neoadjuvant chemoradiation and total
tion in the TNM classification. Moreover, we still mesorectal excision, lymph node positivity is
31 What Is the Prognostic Value of (y)pT and (y)pN? 325

associated with significant worse survival time orectal cancer: a systemic review. Ann Surg Oncol
and time to local recurrence. Neoadjuvant treat- 17:2847–2855
3. Chin CC, Wang JY, Yeh CY,et al (2009) Metastatic
ment is a well-documented cause of limited lymph node ratio is a more precise predictor of prog-
lymph node retrieval (<12) [4, 18]. Lymph node nosis than number of lymph node metastases in stage
ratio has been proposed as an alternative prog- III colon cancer. Int J Colorectal Dis 24:1297-1302
nostic parameter and may possibly show to be a 4. Chou JF, Row D, Gonen M et al (2010) Clinical and
pathologic factors that predict lymph node yield from
better independent staging method than absolute surgical specimens in colorectal cancer. Cancer
count especially when limited numbers of lymph 116:2560–2570
nodes have been retrieved [8]. Moreover, 5. Compton CC, Fielding LP, Burgart LJ et al (2000)
specifically in this setting, the absolute number of Prognostic factors in colorectal cancer; College of
American Pathologists consensus statement 1999.
lymph node retrieved may have a less important Arch Pathol Lab Med 124:979–994
impact on prognosis [14]. 6. Compton CC, Greene FL (2004) The staging of col-
The consequent application of the TNM7 in orectal cancer: 2004 and beyond. CA Cancer J Clin
the setting of patients with rectal cancer treated 54:295–308
7. Dekker JWT, Peeters KC, Putter H et al (2010) Metastatic
with neoadjuvant chemoradiation will imply that lymph node ratio in stage III rectal cancer, prognostic
a number of patients with tumour regression and significance in addition to the 7th edition of the TNM
residual tumour foci, interpreted as tumoural classification. Eur J Surg Oncol 36:1180–1186
deposits, will be classified in the node positive 8. Doll D, Gertler R, Maak M et al (2009) Reduced
lymph node yield in rectal carcinoma specimen after
group, rightly or wrongly [22]. neoadjuvant radiochemotherapy has no prognostic
relevance. World J Surg 33:340–347
Conclusions 9. Gospodarowicz MK, Miller D, Groome PA et al
The TNM classification is a worldwide bench- (2004) The process of continuous improvement of the
TNM classification. Cancer 100:1–5
mark for reporting the extent of cancer and is 10. Gunderson LL, Jessup MJ, Sargent DJ et al (2010)
a major prognostic factor in predicting the Revised tumor and node categorization for rectal can-
outcome of patients and thus a valuable aid cer based on surveillance, epidemiology, and end-
in planning treatment of individual patients. results and rectal pooled analysis outcomes. J Clin
Oncol 28:256–263
Mandatory continuous improvement of the 11. Horvath SM, Morgan JM, Williams GT (2006) The
TNM classification should be evidence-based. new (6th edition) TNM classification of colorectal
The prognostic values of ‘T’ and ‘N’ are facts, cancer: a stage too far. Gut 53:A21
no more, no less. However, the prognostic 12. Kikuchi R, Takano M, Takagi K, et al (1995)
Management of early invasive colorectal cancer: risk
values of various subcategories do not always of recurrence and clinical guidelines. Dis Colon
achieve the same standards of evidence- Rectum 38:1286–1295
based. Communication between the oncology 13. Kim JW (2011) The clinical usefulness of the sentinel
community and those involved in the TNM lymph node in rectal cancer: do we believe in it ? J
Korean Soc Coloproctol 27:51–52
classification in order to create a structured 14. Klos CL, Shellito PC, Rattner DW et al (2010) The
process for introducing changes is essential effect of neoadjuvant chemoradiation therapy on the
for success. prognostic value of lymph nodes after rectal cancer
surgery. Am J Surg 200:440–445
15. Kobayashi H, Mochizuki H, Kato T,et al (2011)
Lymph node ratio is a powerful prognostic index in
patients with stage III distal rectal cancer: a Japanese
References multicenter study. Int J Colorectal Dis 26:891-896
16. Lange MM, Buunen M, van de Velde CJ et al (2008)
Level of arterial ligation in rectal cancer surgery: low
1. Bell S, Sasaki J, Sinclair G et al (2009) Understanding tie preferred over high tie. A review. Dis Colon
the anatomy of lymphatic drainage and the use of Rectum 51:1139–1145
blue-dye mapping to determine the extent of lymph- 17. Ludman L, Shepherd NA (2005) Serosal involvement
adenectomy in rectal cancer surgery: unresolved in gastro-intestinal cancer: its assessment and
issues. Colorectal Dis 11:3443–3449 significance. Histopathology 47:123–131
2. Ceelen W, Van Nieuwenhove Y, Pattyn P (2010) 18. Mekenkamp LJM, van Krieken JHJM, Marijnen
Prognostic value of lymph node ratio in stage III col- CAM et al (2009) Lymph node retrieval in rectal can-
326 N. Ectors

cer is dependent on many factors – the role of the noma after neoadjuvant chemoradiotherapy. Am J
tumour, the patient, the surgeon, the radiotherapist, Surg Pathol 35:127–134
and the pathologist. Am J Surg Pathol 33:1547–1553 34. Simunovic M, Smith AJ, Heald RJ (2009) Rectal can-
19. Messerini L, Cianchi F, Cortesini C et al (2006) cer surgery and regional lymph nodes. J Surg Oncol
Incidence and prognostic significance of occult tumor 99:256–259
cells in lymph nodes from patients with stage IIA col- 35. Smith KJE, Jones PF, Burke DA et al (2011) Lymphatic
orectal carcinoma. Hum Pathol 37:1259–1267 vessel distribution in the mucosa and submucosa and
20. Nagtegaal ID, Quirke P (2007) Colorectal tumour potential implications for T1 colorectal tumors. Dis
deposits in the mesorectum and pericolon: a critical Colon Rectum 54:35–40
review. Histopathology 51:141–149 36. Sobin LH, Wittekind CH, International Union Against
21. Nagtegaal ID, Quirke P (2010) Revised staging: is it Cancer (UICC) (1997) TNM classification on malig-
really better, or do we not know? J Clin Oncol nant tumours, 5th edn. Wiley-Liss Publications, New
28(23):e397–e398, author reply e399–400 York
22. Nagtegaal ID, Tot T, Jayne DG et al (2011) Lymph 37. Sobin LH, Greene FL (2001) TNM classification:
nodes, tumor deposits, and TNM: are we getting bet- clarification of number of regional lymph node for
ter? J Clin Oncol 29:2487–2492 N0. Cancer 92:452
23. Penninckx F (2009) About sentinels and airstrikes. 38. Sobin LH, Wittekind CH, International Union Against
Colorectal Dis 11:441–442 Cancer (UICC) (2002) TNM classification on malig-
24. Peschaud F, Benoist S, Julié C, et al (2008) The ratio nant tumours, 6th edn. Wiley-Liss Publications, New
of metastatic to examined lymph nodes is a powerful York
independent prognostic factor in rectal cancer. Ann 39. Sobin LH, Greene FL (2004) Global TNM advisory
Surg 248:1067-1073 group. Cancer 100:1106
25. Pirro N, Sielezneff I, Ouaissi M et al (2009) Que 40. Sobin LH, Gospodarowicz MK, Wittekind C (2009)
savons-nous du drainage lymphatique due rectum ? TNM classification of malignant tumours, 7th edn.
(What do we know about lymphatic drainage of the Wiley-Blackwell, New York
rectum?). Gastroenterol Clin Biol 33:138–146 41. Sobin LH, Compton CA, Gospodarowicz M et al
26. Pollheimer MJ, Kornprat P, Pollheimer VS et al (2010) (2010) “Evidence-based medicine: the time has
Clinical significance of pT- sub-classification in surgi- come to set standards for staging”. Is a radical over-
cal pathology of colorectal cancer. Int J Colorectal Dis haul really needed? J Pathol 221:361–362, Invited
25:187–196 Response
27. Quirke P, Williams GT, Ectors N et al (2007) The 42. Valentini V, Aristei C, Glimelius B et al (2009)
future of the TNM staging system in colorectal can- Multidisciplinary rectal cancer management: 2nd
cer: time for a debate? Lancet Oncol 8:651–657 European rectal cancer consensus conference
28. Quirke P, Cuvelier C, Ensari A et al (2010) Evidence- (EURECA-CC2). Radiother Oncol 92:148–163
based medicine: the time has come to set standards for 43. Van der Pas MHGM, Meijer S, Hoekstra OS et al
staging. J Pathol 221:357–360 (2011) Sentinel-lymph-node procedure in colon and
29. Quirke P, Risio M, Lambert R et al (2011) Quality rectal cancer: a systemic review and meta-analysis.
assurance in pathology in colorectal cancer screening Lancet Oncol 12:540–550
and diagnosis – European recommendations. Virchows 44. Washington MK, Berlin J, Branton P et al (2010)
Arch 458:1–19 Protocol for the examination of specimens from
30. Rosenberg R, Friederichs J, Schuster T,et al (2008) patients with primary carcinoma of the colon and rec-
Prognosis of patients with colorectal cancer is associ- tum. Arch Pathol Lab Med 133:1539–1551
ated with lymph node ratio: a single-center analysis of 45. Wittekind Ch, Green FL, Henson DE et al (eds) (2003)
3,026 patients over a 25-year time period. Ann Surg TNM supplement: a commentary on uniform use, 3rd
248:968-978 edn. Wiley-Liss Inc, New York
31. Sarma DP (1988) Dukes’ classification of rectal can- 46. Yano H, Moran BJ (2008) The incidence of lateral
cer. South Med J 81:407–408 pelvic side-wall nodal involvement in low rectal can-
32. Schubert W (1990) Dukes’ classification: American cer may be similar in Japan and the west. Br J Surg
chaos versus British order. Can J Surg 33:8–11 95:33–49
33. Shia J, McManus M, Guillem JG et al (2011)
Significance of acellular mucin pools in rectal carci-
What Is the Prognostic Value
of CRM Involvement? 32
Iris D. Nagtegaal

Contents Last century, the major problem in the treatment


32.1 What Is the CRM? ........................................ 328 of rectal cancer was the very high local recur-
rence rate after surgery. Local recurrence rates of
32.2 What Causes a Positive CRM? .................... 328
30–50% were common and resulted in a very
32.3 Incidence of CRM Involvement ................... 329 poor quality of life and short survival of patients
32.4 The Prognostic Value with rectal cancer. Treatment of these local recur-
of CRM Involvement..................................... 329 rences is difficult and often mutilating. In 1986,
References ................................................................. 330 the landmark paper by Phil Quirke and others [1]
described that involvement of the radial surgical
margin, or circumferential resection margin
(CRM), was the main cause of local recurrence.
This finding, together with the excellent clinical
results by Bill Heald and others with a new surgi-
cal technique [2], total mesorectal excision
(TME), caused a major change in the treatment
and prognosis of rectal cancer patients.
At the same time, there was a shift from post-
operative radiotherapy toward preoperative radio-
therapy. This also strongly improved the prognosis
of rectal cancer patients and decreased the num-
ber of local recurrences. The combination of
TME with short-term preoperative radiotherapy
decreased the frequency of local recurrences even
further [3].
Despite the improvements in outcome for rec-
tal cancer patients, we still need good prognostic
markers. In addition, in order to be able to imple-
ment evidence-based changes in treatment more
quickly, there is an obvious need for surrogate
endpoints. Moreover, changes in organization of
I.D. Nagtegaal health care require careful monitoring of quality
Department of Pathology,
of treatment. Quality indicators are becoming
Radboud University Nijmegen Medical Center,
Nijmegen, The Netherlands increasingly important. With the changes in treat-
e-mail: i.nagtegaal@pathol.umcn.nl ment, the role of the CRM has been doubted.

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 327


DOI 10.1007/978-3-642-25005-7_32, © Springer-Verlag Berlin Heidelberg 2012
328 I.D. Nagtegaal

In this chapter, I will demonstrate that the CRM Lymph node metastases in the CRM were associ-
is a very good prognostic marker, a surrogate ated with a lower than expected local recurrence
endpoint, as well as a quality indicator. rate; however, only 42 of these patients have been
described [4, 6].

32.1 What Is the CRM?


32.2 What Causes a Positive CRM?
The CRM, also known as radial or lateral resec-
tion margin, is the surgical soft tissue margin cre- Both tumor-related and surgery-related factors
ated by careful cleavage of (ideally) the mesorectal determine the risk of a positive CRM [7]. Very
fascia. This cleavage creates a fat envelope that few studies concern the patient-related factors
encompasses the tumor, the mesorectal lymph that contribute to the risk of CRM positivity, and
nodes, and tumor deposits, resulting in a radical their results are conflicting.
resection (Fig. 32.1). The CRM should be There is an obvious correlation of CRM + with
reported as the shortest distance from the tumor TNM stage. Advanced stage increases the risk on
to the resection margin. For identification pur- CRM involvement, which has been observed by
poses, the margin should be inked during the many authors [7]. Both increasing depth of tumor
pathology workup. A margin of 1 mm or less is invasion and the presence of tumor deposits and
generally considered positive, although a number involved lymph nodes contribute to this correla-
of studies show that also patients with a margin tion. More positive margins are present in tumors
of 2 mm or less are at increased risk [4, 5]. With that have an ulcerative growth pattern, and in
an increasing distance from the CRM, the risk of tumors that show a stenosing growth pattern.
local recurrence decreases, so it is advisable to Bigger tumors more often have a positive CRM.
mention the exact margin for each individual Histologic factors that are associated with
patient in order to determine the risk of this positive CRM are an infiltrating margin, poor dif-
patient. ferentiation, and vascular invasion. No significant
The CRM can be determined by the primary relationship with tumor budding was observed.
tumor or positive lymph nodes, tumor deposits, Moreover, poor differentiation in submucosal
perineural growth, or extramural vascular invasion. transanal biopsies is predictive of CRM involve-
ment (OR 10.8 (95% CI 1.7–67.1)), as is vascular
invasion (OR 16.1 (95% CI 1.9–139.2)) [8]. The
Mesorectal same is true for biopsies with mucinous carcino-
fascia
mas [9].
Circumferential
resection margin There are more positive margins in tumors
located in the lower rectum than in the middle
Tumor and upper rectum. We believe the main cause of
this is the difference in surgical technique applied
Lymph and the different local anatomy. Many studies
node
observed higher CRM + in APR-operated patients
compared to LAR. Perforations are more com-
mon in APR and associated with an increased
CRM+. The mesorectal excision plane is more
often on the muscularis propria, and in the sphinc-
ter area, the plane of resection is often in either
the lumen, submucosa, or in the sphincters.
Fig. 32.1 Histological scan of a rectal carcinoma. The
tumor is growing in the muscularis propria, and the
There is a large variation between surgeons in
mesorectal fat column has been remove completely, thus the percentage of positive CRM [6], but due to
creating a free circumferential resection margin training, there is a marked improvement to be
32 What Is the Prognostic Value of CRM Involvement? 329

gained. By judging the quality of the surgery per- numbers of positive margins, although there was
formed by evaluating the completeness of more downstaging in the radiochemotherapy arm
mesorectal excision, we proved the direct rela- (p < 0.001) [14]. From the frequencies in the non-
tion between CRM + and quality of surgery. In randomized neoadjuvant studies, no conclusions
two large randomized multicenter trials [10–12], can be drawn about the efficiency of regimens to
we demonstrated that if the mesorectum is reduce positive margins, due to the variability in
removed as a whole (i.e., the resection margin is inclusion criteria, treatment schemes, and lack of
on the mesorectal plane), very few positive mar- pathology quality control.
gins are present and local recurrence rates are
very low [10–12]. In contrast, when the plane of
resection is on the muscularis propria, CRM 32.4 The Prognostic Value
involvement is common and local recurrence of CRM Involvement
rates are high. The plane of resection can explain
why, in a few cases (1.1% [6], 2.0% [4]), positive High local recurrence rates ranging from 25% to
CRMs are present in TNM stage I tumors. 50% in the past have markedly reduced in recent
years, due to changes in surgical approach com-
bined with neoadjuvant therapy. The recognition
32.3 Incidence of CRM Involvement of CRM involvement as one of the main causes of
local recurrence has led to the global introduction
Large differences exist between centers with of TME, resulting in fewer positive margins and
regard to the number of CRM + patients, percent- less residual disease. Since predictive values
ages ranging from 1% to 28% in curatively oper- depend strongly on the prevalence of local recur-
ated patients. Various factors should be taken into rence, the role of CRM was expected to be less
account when reviewing these percentages. The prominent after the introduction of the TME pro-
percentage of CRM + patients is dependent on cedure. However, this does not appear to be the
patient selection, performance of preoperative case owing to the variability of the quality of
imaging, preoperative long-course therapy, surgi- TME. Due to the lower local recurrence rates,
cal technique, and skill of the pathologist. both the sensitivity and the positive predictive
Frequencies of CRM involvement in single cen- value of circumferential margin involvement have
ter studies should thus be treated with caution. decreased. However, after TME treatment, local
On the other hand, less variability is present in recurrence can be predicted with a high specificity
the population-based studies, ranging from 8% to (92%) and a high negative predictive value (95%),
13%. In these studies, there is a difference in which are both clinically relevant [15].
patient selection as well, reflected by differences A recent review [7] evaluated the prognostic
in the number of node-positive patients ranging value of the CRM in a meta-analysis of 17,568
from 21% to 40%. patients, described in the literature between 1985
Randomized neoadjuvant trials give insight to and 2006. From these analyses, it became very
mechanisms by which negative CRMs can be clear that CRM is an important predictor of local
obtained. Short-course 5 × 5 Gy does not influence recurrence. In 4,899 patients treated with surgery,
the percentage of positive margins, as could be only a positive resection margin increased the
expected by the lack of downstaging due to this risk of local recurrence (HR 2.0, 95% CI
regimen. When comparing short-course radio- 1.4–2.9).
therapy with long-course chemoradiation, there The addition of neoadjuvant therapy (varying
is a difference, 13% versus 4% involved margins, from short-course radiotherapy to long-course
p = 0.017 [13]. In this study, downstaging is radiochemotherapy depending on national guide-
observed as well, 48% versus 32% TNM III lines and patient selection based on diagnostic
(p = 0.007). The addition of 5FU/folinic acid to imaging) was expected to compensate for poor
long-term radiotherapy does not decrease the surgical performance and to diminish the role of
330 I.D. Nagtegaal

the CRM. However, when comparing neoadju- significant relation with CRM, both in the neoad-
vant studies with non-neoadjuvant studies, a juvant setting as well as in the patients treated
significant difference in hazard ratio is observed: with surgery alone (n = 6,148, HR 1.7, 95% CI
6.3 (95% CI 3.7–16.7) versus 2.0 (95% CI 1.4– 1.3–2.3). A recent study investigated the value of
2.9). In other words, in contrast to the expecta- CRM for survival in a multivariate model and
tions, a positive CRM is a more powerful predictor found that CRM is more important than T-stage.
of local recurrence in patients treated with neoad- In combination with lymph node status, CRM
juvant therapy. One could argue that this might status provides a better prognostic model than the
be due to the fact that patients treated with neoad- current TNM system [17, 18].
juvant therapy are usually patients with locally
advanced tumors. CRM involvement can be seen Conclusions
as tumor resistance to therapy and indicate a lack The circumferential margin has been around
of downstaging. Indeed, when downstaging for over 25 years. Although the treatment of
occurs, the distance of tumor to the CRM rectal cancer has changed significantly over
increases in some but not all studies [14, 16]. those years, achieving large improvements in
However, in patients treated with 5 × 5 Gy, no prognosis, the prognostic value of the CRM is
downstaging occurs, and still the hazard ratios still there. A positive margin after neoadjuvant
are 3.8 and 2.3 (no neoadjuvant therapy, 95% CI therapy implies a major increase in local recur-
3.3–5.6 and 1.9–3.0) versus 10.0 and 5.3 (5 × 5 Gy, rence risk. In addition, CRM + increases the
95% CI 6.7–25.0 and 3.6–10.0). Thus, the effect risk on development of distant metastases and
of CRM involvement was significantly more pro- decreases overall survival, both in univariate
nounced in the patients who were treated with as in multifactorial analyses.
neoadjuvant therapy, without the downstaging. In addition, a free CRM can be used as a sur-
To monitor the effects of neoadjuvant treat- rogate marker for effectiveness of neoadjuvant
ment on the histology of the tumor, various tumor therapy. CRM evaluation, together with grad-
regression grading systems (TRG) have been ing of the quality of surgery, is the most impor-
employed over the years. These pathological tant quality indicator for surgical treatment.
evaluations are based on the relative amount of
tumor cells present and the desmoplastic reac-
tion. The definitions used in the different studies
vary and reproducibility is poor. Despite these References
disadvantages, TRG has been suggested as a sur-
rogate and early outcome parameter for neoadju- 1. Quirke P, Durdey P, Dixon MF, Williams NS (1986)
Local recurrence of rectal adenocarcinoma due to
vant trials. When comparing the value of CRM
inadequate surgical resection. Histopathological study
with TRG [7], four independent studies demon- of lateral tumor spread and surgical excision. Lancet
strate superiority of CRM above TRG in a multi- 2:996–999
variate model. 2. Heald RJ, Ryall RDH (1986) Recurrence and survival
after total mesorectal excision for rectal cancer. Lancet
All studies that included the development of
1:1479–1482
distant metastases as a separate outcome vari- 3. Kapiteijn E, Marijnen CAM, Nagtegaal ID et al (2001)
able (total n = 2,267) show a significant differ- Preoperative radiotherapy combined with total
ence in prognosis between the CRM + and the mesorectal excision for resectable rectal cancer.
N Engl J Med 345(9):638–646
CRM − patients (HR 2.8, 95% CI 1.9–4.3). No dif-
4. Nagtegaal ID, Marijnen CAM, Klein Kranenbarg E,
ference is observed between the patients treated van de Velde CJH, van Krieken JHJM (2002)
with or without neoadjuvant therapy. Circumferential margin is still an important predictor
The relation of CRM involvement and patient of local recurrence in rectal carcinoma: not one mm
but two mm is the limit. Am J Surg Pathol 26(3):
survival is not clear from all studies, probably
350–357
due to lack of statistical power. However, when 5. Bernstein TE, Endreseth BH, Romundstad P, Wibe A
all studies are summarized, there is a clear and (2009) Circumferential resection margin as a
32 What Is the Prognostic Value of CRM Involvement? 331

prognostic factor in rectal cancer. Br J Surg 96(11): therapy. Preliminary results of the MRC CR07 trial.
1348–1357 J Clin Oncol 24(18s):A3512
6. Birbeck KF, Macklin CP, Tiffin NJ et al (2002) Rates 13. Bujko K, Nowacki MP, Nasierowska-Guttmejer A
of circumferential resection margin involvement vary et al (2004) Sphincter preservation following preop-
between surgeons and predict outcomes in rectal can- erative radiotherapy for rectal cancer: report of a ran-
cer surgery. Ann Surg 235(4):449–457 domised trial comparing short-term radiotherapy vs.
7. Nagtegaal ID, Quirke P (2008) What is the role for the conventionally fractionated radiochemotherapy.
circumferential margin in the modern treatment of Radiother Oncol 72(1):15–24
rectal cancer? J Clin Oncol 26(2):303–312 14. Bosset JF, Calais G, Mineur L et al (2005) Enhanced
8. Ueno H, Mochizuki H, Shinto E, Hashiguchi Y, Hase K, tumorocidal effect of chemotherapy with preoperative
Talbot IC (2002) Histologic indices in biopsy speci- radiotherapy for rectal cancer: preliminary results –
mens for estimating the probability of extended local EORTC 22921. J Clin Oncol 23(24):5620–5627
spread in patients with rectal carcinoma. Cancer 15. Wibe A, Rendedal PR, Svensson E et al (2002)
94(11):2882–2891 Prognostic significance of the circumferential resec-
9. Nagtegaal ID, Van Krieken JH (2007) The multidisci- tion margin following total mesorectal excision for
plinary treatment of rectal cancer: pathology. Ann rectal cancer. Br J Surg 89(3):327–334
Oncol 18(9):ix122–ix126 16. Rullier E, Goffre B, Bonnel C, Zerbib F, Caudry M,
10. Nagtegaal ID, van de Velde CJH, van der Worp E, Saric J (2001) Preoperative radiochemotherapy and
Kapiteijn E, Quirke P, van Krieken JHJM (2002) sphincter-saving resection for T3 carcinomas of the
Macroscopic evaluation of rectal cancer resection lower third of the rectum. Ann Surg 234(5):633–640
specimen: clinical significance of the pathologist in 17. Nagtegaal ID, Gosens MJ, Marijnen CA, Rutten HJ,
quality control. J Clin Oncol 20(7):1729–1734 van de Velde CJ, van Krieken JH (2007) Combinations
11. Nagtegaal ID, van de Velde CJH, Marijnen CAM, of tumor and treatment parameters are more discrimi-
van Krieken JH, Quirke P (2005) Low rectal cancer; a native for prognosis than the present TNM system in
pathologists’ call for a change of approach in abdomi- rectal cancer. J Clin Oncol 25(13):1647–1650
noperineal resection. J Clin Oncol 23:9257–9264 18. Gosens MJ, van Krieken JH, Marijnen CA et al (2007)
12. Quirke P, Sebag-Montefiore D, Steele R et al (2006) Improvement of staging by combining tumor and
Local recurrence after rectal cancer resection is treatment parameters: the value for prognostication in
strongly related to the plane of surgical dissection and rectal cancer. Clin Gastroenterol Hepatol 5(8):
is further reduced by preoperative short course radio- 997–1003
What Is the Prognostic
Value of TRG? 33
Fabio M. Vecchio

Contents Abbreviations
33.1 Definition and Grades of TRG ..................... 333
CRT Chemoradiotherapy
33.2 Prognostic Significance of TRG ................... 336
CSS Cancer-specific survival
References ................................................................. 337 DFS Disease-free survival
m-RCRG Modified version of rectal cancer
regression grade
RCPath Royal College of Pathologists
RCRG Rectal cancer regression grade
TRG Tumor regression grade
ypTNM TNM staging after neoadjuvant
therapy

33.1 Definition and Grades of TRG

Tumor response to preoperative chemoradiother-


apy (CRT) varies from the total regression of the
tumor with complete absence of tumor cells to
no regression at all. Many studies have demon-
strated that a good response to preoperative CRT
is a favorable prognostic factor. Therefore, an
accurate evaluation of tumor response is useful
for predicting oncologic outcomes and planning
further treatment. The term tumor regression
grade (TRG) was firstly introduced by Mandard
et al. in 1994 [3]. The authors examined 93
resected specimens from patients affected by
esophageal carcinoma treated with preoperative
CRT. They found that TRG was a significant pre-
F.M. Vecchio dictor of disease-free survival (DFS). In their
Department of Pathology,
study, Mandard et al. defined five grades of TRG,
Università Cattolica S.Cuore,
Largo F.Vito, 1, 00168 Rome, Italy ranging from TRG 1 characterized by complete
e-mail: fmvecchio@rm.unicatt.it regression to TRG 5 with no regression at all.

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 333


DOI 10.1007/978-3-642-25005-7_33, © Springer-Verlag Berlin Heidelberg 2012
334 F.M. Vecchio

Table 33.1 Mandard and Dworak TRG


Mandard TRG Dworak TRG
Definition Score Score Definition
Absence of tumor cells 1 4 No tumor cells (total regression)
Residual cancer cells scattered through the 2 3 Very few (difficult to find) tumor cells in fibrotic
fibrosis tissue
Fibrosis outgrowing residual cancer cells 3 2 Dominant fibrotic changes with few tumor cells or
groups (easy to find)
Residual cancer cells outgrowing fibrosis 4 1 Dominant tumor mass with obvious fibrosis
Absence of regressive changes 5 0 No regression

Tumor regression was assessed by examining the No response (%) Grade response 0
residual cancer cells and scoring both cytologi- 1–33 Grade response 1
cal changes and stromal changes. On the basis of 34–66 Grade response 2
the combination of these findings, tumor regres- 67–95 Grade response 3
sion was classified according to the five grades, 96–99 Grade response 3+
reported in Table 33.1. 100 Grade response 4
The main advantage of Mandard’s TRG was that
regression was assessed only by comparing the pro- In the conclusions of their study, Ruo et al.
portion of residual carcinoma to scarring by a very found that a marked response to preoperative
simple and reproducible method. In fact, this method radiotherapy ± chemo was clinically associated
was promptly applied also to rectal cancer treated with fewer recurrences and good long-term out-
with preoperative CRT. In 1997, Dworak et al. come, but was not an independent predictor of
adopted a very similar grading of regression exam- recurrence-free survival.
ining the surgical specimens of 17 patients with rec- Wheeler et al. quantified the histologic regres-
tal carcinoma after CRT [2]. Like Mandard et al., sion of rectal cancer after irradiation in a study on
they designed five regression grades, but defined 42 patients. They proposed a 3-point rectal can-
and named them in a different way. Dvorak TRG cer regression grade (RCRG).
varies from grade 0 with no regression to grade 4 RCRG 1 was characterized by “good” radio-
with no tumor cells (see Table 33.1, showing the responsiveness with sterilization or only micro-
correspondence between the Mandard and Dworak scopic foci of adenocarcinoma remaining. In
TRG systems). In 17 specimens studied by Dworak, RCRG 2, macroscopic disease was present asso-
no case of total regression was observed after stan- ciated with marked fibrosis. Finally, RCRG 3
dardized pathological workup, i.e., embedding of indicated a “poor” response with abundant mac-
the whole suspicious area in paraffin blocks and step roscopic disease and little or no fibrosis. Wheeler
sectioning, if necessary. They pointed out that the stressed the importance of determining RCRG
demonstration of residual tumor depended mainly by the consideration that as radiotherapy reduced
on the accuracy of the pathological technique. the number of viable cells present in field of
In 2002, two other different regression grad- treatment, it seemed appropriate to use a patho-
ing systems were proposed by Ruo and by logic staging system that measured tumor regres-
Wheeler [6, 10]. Ruo et al. analyzed the long- sion in addition to the traditional TNM staging
term prognostic significance of selected clinico- after neoadjuvant therapy (ypTNM). Tumors
pathologic factors, including the extent of with the same ypT stage may have different
pathologic response in 69 patients with locally TRG because ypTNM staging does not differen-
advanced rectal cancer. They recorded rectal can- tiate tumors with quite different responses to
cer response as percentage histologic response, CRT. In these cases, RCRG may aid prediction
ranging from no evidence of treatment effect of prognosis.
(0%) to a complete response with no viable tumor Rodel et al. in 2005 evaluated prognostic
identified (100%) by a 6-point grading scale: significance of TRG on surgical specimens of
33 What Is the Prognostic Value of TRG? 335

Table 33.2 Dworak and Rodel TRG Table 33.3 Mandard TRG and Ryan and Vecchio
modifications
Dworak TRG Rodel TRG
Definition Score Definition Mandard Ryan Vecchio
No tumor cells (total 4 Complete regression score score score
regression) Absence of tumor cells 1 1 1
Very few (difficult to 3 Good regression Residual cancer cells 2 2
find) tumor cells in >50% of tumor mass scattered through the
fibrotic tissue fibrosis
Dominant fibrotic 2 Moderate regression Fibrosis outgrowing 3 2
changes with few tumor with fibrosis residual cancer cells
cells or groups (easy to ³25–50% of tumor Residual cancer cells 4 3 3
find) mass outgrowing fibrosis
Dominant tumor mass 1 Minor regression Absence of regressive 5
with obvious fibrosis with fibrosis <25% changes
of tumor mass
No regression 0 No regression
Complete pathologic response edition of dataset guidelines for colorectal can-
Intermediate pathologic response cer reporting. The system defined the following
Poor tumor response
categories:
(A) No residual tumor cells and/or mucus lakes
only
385 patients treated with preoperative CRT [5]. (B) Minimal residual tumors, i.e., only occa-
They defined a 5-point TRG derived from Dworak sional microscopic tumor foci, are identified
scale with some modifications (see Table 33.2). with difficulty
Rodel incorporated Dworak TRG 3 and 2 in TRG (C) No marked regression
3 and split Dworak TRG 1 in TRG 2 and 1. TRG In 2009, Bateman et al. proposed a modified
0 and 4 remained unmodified. On this basis, version of Wheeler RCRG (m-RCRG) in which
Rodel grouped TRG by a 3-grade system. They the key change was the inclusion of a more
reported that 5-year DFS after CRT and curative detailed definition of each category (Table 33.4)
resection was 86% for TRG 4 with no tumor cells [1]. In their study performed by reviewing the
(complete pathologic response), 75% for grouped rectal excision specimens from 54 patients,
TRG 2 + 3 with regression ³25% of tumor mass Bateman et al. compared the utility and reproduc-
(intermediate pathologic response), and 63% for ibility of three different TRG scoring system: the
grouped TRG 0 + 1, with regression <25% of Mandard TRG, the modified RCRG, and RCPath
tumor mass (poor tumor regression). Cancer Dataset method. They concluded that all
In 2005, Ryan et al. proposed a modified three scoring systems were usable in a diagnostic
Mandard score with a 3-point TRG in which TRG setting and all with acceptable degrees of interob-
1 grouped the Mandard TRG 1 and TRG 2, TRG server agreement. The authors favored the use of
2 was equivalent to Mandard grade 3, and TRG 3 m-RCRG for the assessment of rectal cancer
grouped the Mandard TRG 4 and TRG 5 (see specimens after neoadjuvant therapy because the
Table 33.3) [7]. In their study based on 60 patients m-RCRG system provided the optimum balance
with a mean follow-up of 22 months, the Authors between applicability and the accurate recording
emphasized that TRG 1 and 2 could be consid- of low, moderate, and high tumor regression.
ered a complete pathologic response and that the They also suggested that regression grade scoring
modified 3-point TRG had the advantage of bet- must be entirely based on the microscopic pres-
ter reproducibility, with similar prognostic ence of residual tumor, rather than including mac-
significance. roscopic features suggestive of residual tumor.
Another method of grading the degree of Finally, in 2010, Vecchio et al. examined the
tumor regression post CRT in rectal cancer was relationship between TRG and 10-year outcomes
the system proposed in 2007 by Royal College of in 502 patients with locally advanced rectal cancer
Pathologists (RCPath) included within the 2nd treated with preoperative RCT [9]. They proposed
336 F.M. Vecchio

Table 33.4 Wheeler RCRG and Bateman m-RCRG multivariate analysis, only ypN (p < 0.001) and
RCRG 1 Sterilization or only microscopic foci of TRG (p = 0.005) significantly predicted improved
adenocarcinoma remaining, with marked DFS. Moreover, TRG predicted the incidence of
fibrosis positive nodes (p < 0.0001). Vecchio et al. con-
m-RCRG 1 The macroscopic features may be varied.
cluded that, given the ability of TRG to predict
Microscopy reveals no tumor epithelium
or scattered foci of malignant epithelium those patients with N+ disease, it might be help-
comprising <5% of the overall area of ful, in combination with other clinicopathologic
abnormality. Mucin pools may be present factors, in selecting patients for a more conserva-
but do not contain malignant epithelium
tive procedure following CRT such as local exci-
RCRG 2 Marked fibrosis, but macroscopic disease
present
sion rather than radical surgery.
m-RCRG 2 The macroscopic features may be varied. Rodel et al. evaluated TRG on surgical speci-
Microscopy reveals a combination of mens of 385 patients with 5-year outcome and
viable tumor epithelium and fibrosis. concluded that complete and intermediate patho-
Malignant epithelium comprises 5–50% logic response suggested improved DFS after
of the overall area of abnormality
preoperative CRT. They found that complete
RCRG 3 Little or no fibrosis with abundant
macroscopic disease regression of the primary tumor was associated
m-RCRG 3 The macroscopic and microscopic with better control of disease in lymph nodes
features may not be significantly different (ypN positive, 10%) and a minor risk to develop
to cases in which neoadjuvant therapy has distant metastases (DFS, 86%). Intermediate
not been given. Over 50% of the area of
tumor regression was associated with intermedi-
abnormality comprises malignant
epithelium. Some fibrosis may be present ate risk of lymph node involvement (ypN posi-
but no more than that commonly seen as tive, 32%) and an intermediate prognosis (DFS,
desmoplastic stroma in cases where no 75%). Patients with tumors showing poor tumor
neoadjuvant therapy has been given
regression also had more advanced ypT catego-
ries, higher incidence of nodal involvement (ypN
a simplification of Mandard score in three grades positive, 42%), and unfavorable outcome (DFS,
(see Table 33.3): 63%). In their study, Rodel et al. did not find any
TRG 1: Complete regression with absence of significant relationship of TRG to specific pre-
tumor cells. treatment characteristics, such as the cT and cN
TRG 2: Incomplete regression with fibrosis categories, and hypothesized that tumor regres-
predominant on cancer cells. sion after preoperative CRT was a multifaceted
TRG 3: Absent or poor regression with resid- phenomenon that appeared to be associated with
ual cancer outgrowing fibrosis. smaller, less aggressive disease and may also cor-
respond to the molecular tumor profiling regulat-
ing treatment response. There is evidence that
33.2 Prognostic Significance of TRG positive lymph nodes, after preoperative CRT
indicating an aggressive potential of the malig-
Apart from the chosen method, tumor regression nant cells associated with resistance toward CRT,
after preoperative RCT has been found to have suggest an unfavorable prognosis irrespective of
prognostic significance [5, 8]. Vecchio et al. any TRG of the primary tumor [5]. In a recent
examined the relationship between TRG and out- paper, Min et al. investigated the clinical impact
come in 144 patients with rectal cancer treated of TRG after preoperative CRT for locally
with preoperative CRT and with a median follow- advanced rectal cancer especially in lymph node
up of 72 months. They quantified TRG in five negative patients [4]. They studied a group of 178
grades according to Mandard score and com- patients who had cT3–T4 tumors and examined
bined TRG 1–2 and TRG 3–5. They found that the prognostic significance of TRG in comparison
TRG was a predictor for local failure, metasta- with histopathologic staging. They found that
ses-free survival, DFS, and overall survival. By TRG was an independent prognostic factor for
33 What Is the Prognostic Value of TRG? 337

the cancer-specific survival (CSS) of the ypN0 3. Mandard AM, Dalibard F, Mandard JC et al (1994)
patients and that TRG had a stronger impact on Pathologic assessment of tumor regression after pre-
operative chemoradiotherapy of esophageal carci-
the CSS of ypN (−) patients (p = 0.002) than on noma. Cancer 73:2680–2686
that of the ypN (+) patients (p = 0.521). 4. Min BS, Kim NK, Pyo JY et al (2011) Clinical impact
An accurate prediction of CSS requires both of tumor regression grade after preoperative chemora-
information on the number of residual cancer diation for locally advanced rectal cancer: subset
analyses in lymph node negative patients. J Korean
cells (by TRG) and on their location in rectal wall Soc Coloproctol 27:31–40
and perirectal tissue (by ypT stage). Therefore, 5. Rodel C, Martus P, Papadoupolos T et al (2005)
TRG determination is the logical complement to Prognostic significance of tumor regression grade
traditional ypTNM staging when considering after preoperative chemoradiotherapy for rectal can-
cer. J Clin Oncol 23:8688–8696
patients for further adjuvant therapy. As TRG 6. Ruo L, Tickoo S, Klimstra DS et al (2002) Long-term
defines the amount of residual cancer cells after prognostic significance of extent of rectal cancer
preoperative CRT, further investigations are man- response to preoperative radiation and chemotherapy.
datory in order to define the quality of neoplastic Ann Surg 236:75–81
7. Ryan R, Gibbons D, Hyland JMP et al (2005)
cells and their behavior, especially in patients Pathologic response following long-course neoadju-
without lymph node metastasis. Identification of vant chemoradiotherapy for locally advanced rectal
factors indicating a more aggressive phenotype cancer. Histopathology 47:141–146
and/or resistance toward CRT may contribute to 8. Vecchio FM, Valentini V, Minsky BD et al (2005) The
relationship of pathologic tumor regression grade
improve the prognostic value of TRG and to (TRG) and outcomes after preoperative therapy in rec-
allow tailoring of treatment. tal cancer. Int J Radiat Oncol Biol Phys 62:752–760
9. Vecchio FM, Barba MC, Gambacorta MA et al (2010)
Pathologic tumor regression grade and 10-year out-
References comes in 502 patients with rectal cancer treated with
preoperative therapy. Mod Pathol 23(Suppl):171
10. Wheeler JM, Warren BF, Mortensen NJ et al (2002)
1. Bateman AC, Jaynes E, Bateman AR (2009) Rectal
Quantification of histologic regression of rectal cancer
cancer staging post neoadjuvant therapy – how should
after irradiation. Dis Colon Rectum 45:1051–1056
the changes be assessed? Histopathology 54:713–721
2. Dworak O, Keilholz L, Hoffmann A (1997) Pathologic
features of rectal cancer after preoperative radiochemo-
therapy. Int J Colorectal Dis 12:19–23
Part VIII
Q&As on Multidisciplinary Team Management
What Are the Recommendations
to Ensure a Successful 34
Multidisciplinary Team
in Rectal Cancer?*

Sujay Shah, Pawan Mathur,


and Robert Glynne-Jones**

34.7 Health-Care Professionals: The Team ..... 344


Contents 34.7.1 Representation and Attendance ................... 344
34.1 Introduction ................................................ 341 34.7.2 Roles of Individuals ..................................... 344
34.7.3 Is There a Role for Training? ....................... 345
34.2 Do MDTs Improve Clinical 34.7.4 The Meetings ............................................... 345
Outcomes? .................................................. 342 34.7.5 Is Videoconferencing a Good Idea? ............. 345
34.3 What Are the Challenges 34.7.6 The Patients’ Perspective ............................. 347
Facing MDTs? ............................................ 343 34.7.7 The Process .................................................. 347
34.7.8 Data Collection ............................................ 347
34.4 What Are the Primary Functions 34.7.9 Feedback ...................................................... 347
of an MDT?................................................. 343
34.8 What Are the Next Steps? ......................... 348
34.5 What Are the Markers
of a Successful MDT? ................................ 343 34.9 What Is the Future of MDT Working? .... 348

34.6 What Recommendations Can Be References ................................................................. 349


Made for Ensuring a Successful
Multidisciplinary Team Meeting? ............ 344

34.1 Introduction

In the UK, multidisciplinary team discussions


started with the formation of breast cancer teams
nearly 20 years ago and have expanded across all
tumour types over the past decade. It was further
promoted by the 1995 Calman-Hine report – ‘A
policy framework for commissioning cancer ser-
vices’ and the ‘Improving Outcomes Guidance’
S. Shah (IOG). A multidisciplinary team is defined as a
Mount Vernon Cancer Centre, Northwood, ‘group of people of different healthcare disci-
Middlesex, UK
plines, which meets together at a given time
P. Mathur (whether physically in one place, or by video or
Mount Vernon Cancer Centre, Northwood,
Middlesex, UK
**
R. Glynne-Jones (*) This chapter is based on literature searches, analysis of
Mount Vernon Cancer Centre, Northwood, UK national peer review data and results from a national
Middlesex, UK survey of UK MDT members. On Behalf of the Barnet
e-mail: rob.glynnejones@nhs.ne Colorectal Cancer Multidisciplinary Team.

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 341


DOI 10.1007/978-3-642-25005-7_34, © Springer-Verlag Berlin Heidelberg 2012
342 S. Shah et al.

teleconferencing) to discuss a given patient and enabled a set of recommendations to be formu-


who are each able to contribute independently to lated to define how an effective MDT would work,
the diagnostic and treatment decisions about the which are available online at www.ncat.nhs.uk.
patient’. Multidisciplinary teams were originally An international survey of 173 colorectal
introduced to counter the apparent shortfalls in surgeons (of whom 123 responded) showed that
cancer care provision in the UK at the time and working in regular MDTs made the use of MRI
aimed to ensure all patients achieved prompt for staging more likely and hence significantly
access to expert advice, up-to-date treatment and influenced the use of neoadjuvant chemoradia-
holistic care from relevant professionals with tion (RR = 5.67, p = 0.03) for a threatened circum-
specialist knowledge and skills. An additional ferential margin on MRI [4]. Also, MDT working
aspiration was to ensure seamless specialist con- improved the quality of pathology reporting
tinuity of care for all patients as well as the offer (RR = 4.85, p = 0.01). These changes were not
of adequate information and support. influenced by caseload. Blazeby [5] provided fur-
Multidisciplinary team-working has now been ther evidence that multidisciplinary team-work-
implemented in cancer care systems throughout ing can make a positive impact on the quality of
much of Europe, the USA and Australia and clinical decision-making, clinical outcomes for
forms a core component of guidelines in all can- patients, patients’ experience of care and also the
cer care services, which is pivotal to many of the impact on the working lives of team members.
national cancer plans in Europe. There is almost In this chapter, we will review the evidence for
universal approval for this strategy, which indeed effective MD team-working and its impact on
some consider crucial [1], despite the fact that outcomes and discuss how the characteristics and
there is little evidence for its effectiveness in structure of the MDT, the team culture, leaders,
improving outcomes [2]. organisation, setting and local environment
The introduction of multidisciplinary teams influence optimal care for the patient.
has been based upon the rationale that as the man-
agement of different diseases becomes more com-
plex, it is important to involve the various key 34.2 Do MDTs Improve Clinical
professional groups in clinical decision-making Outcomes?
for individual patients. Hence, a key objective of
multidisciplinary teams is to ensure that patients Although MDTs are now in widespread use, pro-
are managed by a specialist team. This is particu- vide relatively seamless coordination of care and
larly relevant in rectal cancer, where preoperative are supported in the literature particularly for rec-
radiotherapy and chemoradiation are often deliv- tal cancer [6], there is little evidence to support
ered according to clinical findings and staging their effectiveness. With the overall aim being
(particularly in terms of the MRI) and many dif- improving patient care, very little is known about
ferent surgical options are available with varying the decision-making and the validity of this deci-
levels of radicality and implications for future sion-making [7]. Few have examined whether
QOL in the patient. Since the cancer itself, surgery MDT treatment decisions are actually imple-
and other adjuvant treatments such as radiotherapy mented or investigated the reasons why some
can impact on physical appearance, physical func- decisions changed after the meeting [8]. Indeed,
tioning (bowel, urinary and sexual) and emotional the data we have suggests that MDT discussions
health, the optimal management will necessarily may even fail to reach a decision in 27–52% of
take into account both clinical and psychosocial cases [7].
factors in addition to the prospect of cure. Given However, weak study designs, using retrospec-
the complexity of these decisions, a multidisci- tive before-and-after studies have typically been
plinary approach is essential, if MDTs are to used to evaluate the benefit of multidisciplinary
incorporate modern and changing developments team-working. The findings are often confounded
in all these disciplines into treatment plans [3]. A by changes over time including increased spe-
large survey of MDT members in the UK in 2009 cialisation of the cancer workforce, improved
34 What Are the Recommendations to Ensure a Successful Multidisciplinary Team in Rectal Cancer?* 343

adherence to evidence-based guidelines, better best practice guidelines [12]. Hence, preparation
staging at diagnosis and improved treatments. costs alone come to around £50 million per annum,
In the UK, multidisciplinary team-working is and attendance time will cost a similar amount.
associated with improved 5-year survival in col- Multidisciplinary teams also require good and
orectal cancer [9]. The presence of microscopic timely communication between primary, second-
tumour cells within 1 ml of the circumferential ary and tertiary care; good data collection for the
surgical resection margin (CRM) in the patho- benefit of the individual patient, audit and
logical specimen post surgery is the endpoint research; encouragement of recruitment into clin-
most strongly associated with local recurrence in ical trials; assurance that good practice guidelines
rectal cancer and doubles the risk of developing are adhered to; optimising the use of resources;
distant metastases. Improvements in surgical and hopefully improving the working lives of
technique have dramatically altered the risks of a staff. Hence, MDT meetings pose difficulty for
positive CRM [10]. An increased efficacy for more rural and remote areas particularly when
preoperative CRT followed by surgery after a specialist centres offer regional services – which
delay of 6 weeks to allow downstaging over has driven the use of teleconferencing.
SCPRT and immediate surgery has already been
demonstrated in a randomised trial, where the
involvement of the CRM was reduced from 13% 34.4 What Are the Primary
to 4% [11]. In the study by Burton et al., the Functions of an MDT?
authors showed that in one network, 16/66 (26%)
of patients not discussed by the MDT had a posi- The primary functions of an MDT are generally
tive CRM (£1 mm). In contrast, those discussed agreed to be:
in an MDT preoperatively achieved a negative 1. To discuss all new and recurrent active col-
CRM (>1 mm) in 113/116, i.e. 96% of cases. orectal cancer patients
2. To act as a specialist forum to discuss complex
cases and make clinical decisions
34.3 What Are the Challenges 3. To act as a specialist forum to integrate psy-
Facing MDTs? chosocial, emotional, sexual and financial
issues that may affect the patients
There is wide variation in the knowledge, skills 4. To register patients on relevant cancer regis-
and perspectives of members of multidisciplinary tries and databases
teams – and in particular the chair or MDT lead. 5. To encourage recruitment into relevant and
Multidisciplinary team-working also highlights available clinical trials
the paradox that an evidence base is mandated for 6. To improve practical techniques (imaging,
individual clinical decisions, but not for overall surgical, chemotherapeutic, radiotherapeutic
organisational decisions. In addition, there are and pathological) through review and positive
substantial constraints in terms of time and feedback
resources for a large and varied number of health- 7. To support clinicians in hard-pressed
care professionals (surgeons, physicians, radiation circumstances
and medical oncologists, radiologists, patholo- 8. To promote optimal medical management and
gists, dieticians, clinical nurse specialists and pal- care that is both up-to-date and appropriate
liative care specialists) who are required to attend
such additional meetings to discuss cancer care
and multidisciplinary forum. MDT meetings use 34.5 What Are the Markers
resources. For the 174 colorectal MDTs in the of a Successful MDT?
UK, attendance involves over 100,000 person-
hours each year. In addition, preparation time for There is no such thing as a perfect MDT.
radiologists may take 2 h and pathologists 2.4 h However, there are several markers that can be
prior to MDT meetings in line with Royal College used to gauge the success of MDTs. The
344 S. Shah et al.

‘Improving Outcomes Guidance’ (IOG) defines aspx?rid=136) [15]. The recommendations below
the composition of teams and their working are a combination of the views highlighted in this
practices (e.g. having protocols for referral and report as well as additional views.
treatment). The IOGs provide measurable stan-
dards in many settings, and adherence is assessed
using a detailed peer review process. However, 34.7 Health-Care Professionals:
analysis of data collected in the second round of The Team
peer reviews, conducted between 2004 and 2007,
which reviewed 174 colorectal MDTs, showed 34.7.1 Representation and Attendance
that there was considerable variation in MDT
functioning according to the predefined stan- It is essential that the there is representation
dards (National Cancer Peer Review Programme from members of the all relevant disciplines. In
2004–2007) [13]. The reviewers found a highly the case of rectal cancer, this includes surgeons,
committed workforce, despite acknowledged oncologists, radiologists, pathologists, clini-
gaps in the provision of services. Compliance to cal nurse specialists and MDT coordinators.
the range of expertise in the MDT, i.e. a named Although attendance should be regular, and
core membership was 88%. MDTS adhered least part of the job plan for these individuals, allow-
well with aspects of team-working that require ances should be made for absences and suit-
additional resources, expertise and time such as able cover available. There should be a regular
participating in audits, conducting patient expe- attendance register with details of arrival and
rience surveys and service improvement. This departure from meetings. Additionally, indi-
partly reflects the shortage of some core staff vidual members should strive to be present for
including oncologists [14]. We also have varia- discussion of patients in whose care they have
tion in the provision of clinical nurse specialists, been involved.
who often fail to play as important a role as in Our own colorectal MDT meets for 2.5 h
the day to day clinical setting. weekly. The personnel present includes surgeons,
Finally, the decisions made by MDTs regard- gastroenterologists, clinical oncologists, several
ing patient treatments and subsequent treatment specialist radiologists (gastrointestinal diagnos-
outcomes provide another way of measuring the tic/interventional) with different skills (MRI/CT/
level of success of an MDT. However, factors ultrasound), pathologists, clinical nurse special-
that affect the quality of clinical decisions made ists, stoma nurses, palliative care nurses, doctors
by multidisciplinary cancer teams (MDTs) are in training and MDT coordinators. Intermittently,
poorly understood and rarely investigated [7]. our dietician and anaesthetists also attend. All are
required formally to sign in their attendance.

34.6 What Recommendations Can


Be Made for Ensuring a 34.7.2 Roles of Individuals
Successful Multidisciplinary
Team Meeting? Specific roles within the team are important, i.e.,
who chairs each meeting, who presents each case
and the function of the MDT coordinator. It is
In February 2010, the National Cancer Action important to identify who will act on the deci-
Team in the UK published a blueprint for peer sions proposed, who makes the summary and
review entitled ‘The Characteristics of an Effective who informs the GP and the patient. The roles of
Multidisciplinary Team (MDT)’ and offered rec- the chair and MDT coordinator are vital in ensur-
ommendations regarding membership, attendance, ing smooth running of the meeting, with careful
leadership, team-working and culture, personal attention to each patient’s diagnosis and treat-
development & training (www.ncin.org.uk/view. ment plans.
34 What Are the Recommendations to Ensure a Successful Multidisciplinary Team in Rectal Cancer?* 345

All members of the team should be allowed to the programme to enhance understanding of how
raise their views and initiate discussions into the team members can work together to improve clini-
management of patients. In a study from Lamb cal outcomes.
et al. [16], it was felt that ‘most participants (in Currently, the National Cancer Action Team
the MDT) thought that the discussion was not (NCAT) has a training initiative in some networks for
balanced equally across groups and sometimes MDT lead to address this issue. There are now plans
not open enough’. to run workshops aimed at MDT chairs and partici-
A study of colorectal cancer teams in the UK pants to further improve performances at MDTs.
suggests there is a positive impact of multidisci-
plinary team-working on the working lives of
team members [17, 18]. Providing patient care in 34.7.4 The Meetings
multidisciplinary teams was reported as the pre-
dominant source of job satisfaction by all team It is important that meetings take place in a fixed
members [18]. An observational study of UK place and at a fixed time during core hours to
breast cancer teams also suggests team-working ensure maximal attendance. The environment
has a positive impact on the mental health of can- should be clean, quiet and of a reasonable size to
cer team members [19]. Working in teams may accommodate all team members comfortably.
be beneficial for psychological well-being and to Additionally, suitable equipment should be pres-
avoid burnout [20]. ent to view radiological images, histopathologi-
cal slides and any other test results. Access to
relevant databases should be available to ensure
34.7.3 Is There a Role for Training? prompt data entry (Figs. 34.1 and 34.2). Ideally,
these should all be set up and running prior to the
Educational initiatives should address the start of the meeting.
significant shortfalls in training multidisciplinary An important aspect that is often forgotten is
team members in the core skills required for effec- the prompt availability of IT technical support, in
tive team-working, including communication skills, the event of any malfunctioning of equipment.
team-working, handling complaints and leader-
ship. The Pelican MDT Development Programme
originally provided a national initiative which 34.7.5 Is Videoconferencing
attempted to improve clinical outcomes in col- a Good Idea?
orectal cancer by training all UK MDT colorectal
teams in best clinical practice. The Programme Effective team-working needs a culture that is
was intended to be a unique team-based residential shared within the team, mutual respect and an
training programme, attended by the whole MDT, understanding of each others’ roles. This is the
including surgeons, oncologists, radiologists, oil which lubricates the working of an MDT.
pathologists, clinical nurse specialists and MDT Remote working is necessary with some large
coordinators. The programme focussed on train- teams to accommodate different hospitals and the
ing individual professional groups in imaging, normal working practices of the individuals con-
different potential treatment modalities (chemo- cerned. However, our experience is that techno-
therapy, radiotherapy and surgery), optimising the logical delays, frequent equipment failures and
information obtained from the pathologist and the loss of normal social interactions of face-to-
supportive care. The MDT was encouraged to dis- face meetings do tend to hinder achieving a
cuss and formulate treatment plans, in response to shared team culture.
MDT review of imaging [21] and pathology [22]. It is much more difficult to pick up social cues
Similarly, the programme encouraged reflection which facilitate the activity and development of
on the clinical roles of the individual key team the functioning team [23]. Often, several people
members to emphasise a team-based culture within attempt to speak at once, and then all pause at
346 S. Shah et al.

Figs. 34.1 and 34.2 A regular weekly multidisciplinary team meeting in the UK involving pathologists, surgeons,
radiologists, gastroenterologists, clinical oncologists, colorectal clinical nurse specialists and palliative care nurses

once and finally try to speak again at the same However, newer systems, which make use of
time, which makes for a very staccato decision- wireless Internet connections and virtual servers may
making process. Interactions appear impersonal, provide a suitable solution to these problems and, in
and the group may not fully function. There is so doing, make meetings more efficient, particularly
some evidence that the exchange of information where members have to travel large distances
via virtual teams is less effective than in face- to attend meetings. There are a number of these
to-face meetings [24]. being trialled in various centres across Europe.
34 What Are the Recommendations to Ensure a Successful Multidisciplinary Team in Rectal Cancer?* 347

34.7.6 The Patients’ Perspective inclusion of patients into weekly MDTs, a list
of patients for discussion should be made avail-
Evidence for the benefit of multidisciplinary able to all those involved in advance of the
team-working from the patients’ perspective meeting. This is particularly important for radi-
may be inferred from the results of NHS patient ologists and histopathologists for whom pre-
surveys. However, patients do not attend, and meeting preparation is essential to ensure
their preferences are rarely discussed [7, 8]. The efficient discussions at the meeting itself. There
overall experience of care for cancer patients in should be a minimum set of criteria that is pre-
the UK improved between 2000 and 2004 sented regarding each patient, ideally by the
according to analysis of patient experience sur- individual team member who has made the deci-
veys undertaken by the Picker Institute (Picker sion for inclusion at the meeting. Furthermore,
Institute) [25]. Although many factors are likely any relevant imaging/pathological specimen
to have been responsible (including the impor- should be available.
tance given to patient experience in national Following the meeting, appropriate steps should
policy, stronger leadership at national and local be in place to ensure any recommendations are
levels, the provision of funding, the influence of carried out, and the patient and their GP are
charities and the power of information obtain- informed at the earliest opportunity. If appropriate,
able from the Internet), those cancers where onward referral should be made in a timely man-
team-working was at a more developed stage ner, with all the relevant information enclosed.
(breast, colorectal and lung cancer) showed more
striking improvements than urological cancers
where guidance that included providing care 34.7.8 Data Collection
within multidisciplinary teams was not published
until much later in 2002. A clear database for use in MDTs should be avail-
It is important to remember that the patient is able, which keeps a record of patient demograph-
the central figure in the MDT discussion. With this ics, clinical information as well as the discussion
in mind, it is important to have a good knowledge regarding the patient at the MDT. This is a manda-
of the patient’s comorbid status and their personal tory requirement for MDTs in the UK and provides
preferences with regard to treatment options. access, both prospectively and retrospectively, to
A study by Blazeby [5] showed that 15.1% of treat- data that can be used for research and audit. It also
ment decisions made at upper GI MDT meetings helps identify patients that may be suitable for
were not implemented, with the most common rea- ongoing trials. This process also allows for effec-
sons for this related to lack of information concern- tive follow-up of patients and their outcomes fol-
ing patients’ wishes or comorbid disease. lowing treatment (Fig. 34.3).

34.7.7 The Process 34.7.9 Feedback

It is recommended that MDTs take place weekly. Following meetings, it is important to review pro-
It is important that there are clear guidelines on cesses at regular intervals and assess the deci-
what patients are suitable for discussion at MDT sions made retrospectively. This may reinforce
meetings, as well as at what stage in their diag- the decisions made and highlight the perfor-
nostic/treatment journey they should be dis- mances of individual MDTs. At the same time,
cussed. In general, any new patient with a these may highlight discordance between MDT
diagnosis of colorectal cancer, all patients with recommendations and actual treatment decisions –
a cancer resected and patients with recurrence in this case, it is important to review the available
or metastatic disease are appropriate for discus- data and assess why this discordance arose, and
sion. Following an appropriate cut-off time for try and improve on particular areas.
348 S. Shah et al.

Overall compliance by MDT tumour site (local & specialist teams)

77% 77% 75%


73%
70% 69%
70%

60%

50%

40%

30%

20%

10%

0%

ae
st

gy
ng
ta

G
ea

yn
ec

lo
Lu

er
Br

ro
G
or

pp

U
ol

U
C

Fig. 34.3 Overall compliance to quality measures for MDT in peer review assessment

34.8 What Are the Next Steps? communication of these decisions) may improve
the quality and patient-centredness of decision-
There is evidence that the introduction of multidis- making by multidisciplinary teams.
ciplinary team meetings has led to more rigorous
decision-making processes and has improved out-
comes for patients with cancer as well their experi- 34.9 What Is the Future of MDT
ence of care. Yet, some teams work more effectively Working?
than others. There is also a suggestion that partici-
pation in multidisciplinary teams meetings may It is clear from the evidence presented that MDTs
increase team members’ job satisfaction. are here to stay and now make up a substantial
Randomised trials are unlikely to be able to amount of time and effort for all concerned;
examine the added value in terms of effectiveness indeed, a large proportion of peer review out-
of multidisciplinary teams because they now comes are based around MDT functionality.
form routine clinical practice. However, there is Many MDTs have core members who primarily
evidence for a wide variation in the quality of work at geographically distance sites, and hence
cancer multidisciplinary teams. Therefore, there attending every MDT they are required to is not
is a need to identify dysfunctional teams and possible. Hence, the concept of videoconferenc-
improve their performance. Hence, reliable mea- ing has gathered support. Although some research
surements of MDT functioning and the opportu- may show that this style of working may not be
nity to demonstrate the differences in outcomes effective, it is here to stay. A natural progression
for teams classified as ‘high’ or ‘low’ performers for MDTs therefore is to embrace current and
would be useful. developing technology; the majority of mobile
More formal proformas to standardise all phone companies have ‘health’ sections and all
aspects of multidisciplinary team-working are producing state-of-the-art communication
(such as reporting of diagnostic imaging, sum- portals which facilitate efficient videoconferenc-
marising patient management decisions and the ing functionality. Current technology will allow
34 What Are the Recommendations to Ensure a Successful Multidisciplinary Team in Rectal Cancer?* 349

members to not only log in from their parent sites, 3. Taflampas P, Christodoulakis M, de Bree E, Melissas
but they will be able to communicate effectively, J, Tsiftsis DD (2010) Preoperative decision making
for rectal cancer. Am J Surg 200(3):426–432
see images in higher resolution and include any 4. Augestad KM, Lindsetmo RO, Stulberg J, Reynolds
information that is part of a patient’s electronic H, Senagore A, Champagne B, Heriot AG, Leblanc F,
record. They will even be able to log in from any- Delaney CP, International Rectal Cancer Study Group
where in the world on a secure line. It may even (IRCSG) (2010) International preoperative rectal can-
cer management: staging, neoadjuvant treatment, and
be possible to ‘tap’ into another specialist MDT impact of multidisciplinary teams. World J Surg
to get their opinion in real time. The same soft- 34(11):2689–2700
ware will automatically create electronic atten- 5. Blazeby JM, Wilson L, Metcalfe C, Nicklin J, English
dance sheets, MDT records and communications R, Donovan JL (2006) Analysis of clinical decision-
making in multi-disciplinary cancer teams. Ann Oncol
to the GP, patient and other health-care individu- 17(3):457–460
als. The day of the virtual MDT is here. 6. Obias VJ, Reynolds HL (2007) Multidisciplinary
Core members of the MDT are clearly teams in the management of rectal cancer. Clin Colon
specified, but future MDTs may include individu- Rectal Surg 20(3):143–147
7. Lamb BW, Brown KF, Nagpal K, Vincent C, Green
als who are increasingly part of management JS, Sevdalis N (2011) Quality of care management
decisions such as anaesthetists and perhaps even decisions by multidisciplinary cancer teams: a sys-
patient representatives. Currently, many MDTs tematic review. Ann Surg Oncol 18(8):2116–2125, 7
include operative photographs (this in fact makes 8. Wood JJ, Metcalfe C, Paes A, Sylvester P, Durdey P,
Thomas MG et al (2008) An evaluation of treatment
up an important aspect of rectal cancer surgery), decisions at a colorectal cancer multi-disciplinary
but it may be feasible to include photos of patients team. Colorectal Dis 10(8):769–772
so that clinicians may get an idea of whether par- 9. Morris E, Haward RA, Gilthorpe MS, Craigs C,
ticular treatments may be more efficacious or Forman D (2006) The impact of the Calman-Hine
report on the processes and outcomes of care for
possible than others (e.g. laparoscopic vs. open Yorkshire’s colorectal cancer patients. Br J Cancer
surgery). 95(8):979–985
10. Birbeck KF, Macklin CP, Tiffin NJ et al (2002) Rates
of circumferential resection margin involvement vary
Competing Interests All authors declare that they have
between surgeons and predict outcomes in rectal can-
no conflict of interest.
cer surgery. Ann Surg 235:449–457
11. Bujko K, Nowacki MP, Nasierowska-Guttmejer A
et al (2004) Sphincter preservation following preop-
Conclusions
erative radiotherapy for rectal cancer: report of a ran-
Increasingly, in developed countries, cancer domized trial comparing short-term radiotherapy vs.
care is being delivered by multidisciplinary conventionally fractionated radiochemotherapy.
teams. Assessment, training and support both Radiother Oncol 72:15–24
12. Kane B, Luz S, O’Briain DS, McDermott R (2007)
for the team and the chair and modern facili- Multidisciplinary team meetings and their impact on
ties including videoconferencing, in addition workflow in radiology and pathology departments.
to tackling some of the practical barriers to BMC Med 5:15
effective team-working, will require consider- 13. National Cancer Action Team (2008) National Cancer
Peer Review Programme 2004–2007. National report:
able future investment and resources. an overview of the findings from the Second National
Round of Peer Reviews of Cancer Services in England.
National Cancer Action Team, London
14. Department of Health (2007) Cancer reform strategy.
Department of Health, London
References 15. National Cancer Action Team (2010) The characteris-
tics of an effective multidisciplinary team (MDT).
1. Day F, Pollock C, Brook C, Albus A (2010) www.ncin.org.uk/view.aspx?rid=136. Accessed date:
Multidisciplinary cancer teams. Crucial for popula- 16 August 2011
tion health. BMJ 340:c2125 16. Lamb BW, Wong HW, Vincent C, Green JS, Sevdalis
2. Fennell ML, Das IP, Clauser S, Petrelli N, Sainer A N (2011) Teamwork and team performance in multi-
(2010) The organization of multidisciplinary care teams: disciplinary cancer teams: development and evalua-
modelling internal and external influences on cancer tion of an observational assessment tool. BMJ Qual
care quality. J Natl Cancer Inst Monogr 40:72–80 Saf 20:849–856
350 S. Shah et al.

17. Taylor C (2008) Improving the working lives of UK 21. Burton S, Brown G, Daniels IR, Norman AR, Mason
colorectal cancer teams: an evaluation of the Pelican B, Cunningham D (2006) MRI directed multidisci-
team-based training programme (abstract). National plinary team preoperative treatment strategy: the way
Cancer Research Institute Conference. http://www. to eliminate positive circumferential margins? Br J
ncri.org.uk/ncriconference/2008abstracts/abstracts. Cancer 94:351–357
Accessed date: 16 August 2011 22. West NP, Morris EJ, Rotimi O, Cairns A, Finan PJ,
18. Taylor C, Ramirez AJ (2009) Multidisciplinary team Quirke P (2008) Pathology grading of colon cancer
members’ views about MDT working: results from a surgical resection and its association with survival: a
survey commissioned by the National Cancer Action retrospective. Lancet Oncol 9:857–865
Team. National Cancer Action Team, London. (www. 23. Olver IN, Selva-Nayagam S (2006) Evaluation of a
ncin.org.uk/view.aspx?rid=136). Accessed date: 16 telemedicine link between Darwin and Adelaide to
August 2011 facilitate cancer management. Telemed J 6(2):213–218
19. Haward R, Amir Z, Borrill C, Dawson J, Scully J, 24. Warkentin M, Sayeed L, Hightower R (1997) Virtual
West M et al (2003) Breast cancer teams: the impact teams versus face-to-face teams: an exploratory study
of constitution, new cancer workload, and methods of of a web-based conference system. Decis Sci 28(4):
operation on their effectiveness. Br J Cancer 89(1): 975
15–22 25. Picker Institute Europe (2005) Is the NHS getting bet-
20. Catt S, Fallowfield L, Jenkins V, Langridge C, Cox A ter or worse? An in-depth look at the views of nearly
(2005) The informational roles and psychological a million patients between 1998 and 2004. Picker
health of members of 10 oncology multidisciplinary Institute Europe, Oxford
teams in the UK. Br J Cancer 93(10):1092–1097
What Is the Appropriate Timetable
for Tailored Follow-up? 35
Lars Påhlman

Contents 35.1 Introduction


35.1 Introduction ................................................ 351
The majority of patients with a newly diagnosed
35.2 Who Shall Be Followed? ............................ 352
rectal cancer will be screened for distant meta-
35.3 Distant Metastases ..................................... 352 static disease. The most common places are the
35.4 Metachronous Bowel Tumours ................. 352 liver and/or the lungs. Subsequently these organs
35.5 Local Recurrence ....................................... 353
should be scanned in the perioperative period.
However, dependent upon tumour stage and
35.6 Imaging Technique..................................... 353 despite a curative resection, one will expect recur-
35.7 Tumour Markers ........................................ 353 rences since occult metastases have not been
References ................................................................. 354 found at the time for surgery. For patients with a
stage I disease, almost no recurrences will appear,
but after stage II, approximately 20% of the
patients will recur, and finally, after a tumour in
stage III as much as 40–50% of the patients will
recur. Due to the anatomy and vascular drainage
from the gastrointestinal tract, the majority of
recurrences will be found in the liver but also in a
minority in the lung since the rectum is also
drained via the caval vein. Other places for distant
recurrences are very rare. Moreover, with good
preoperative staging and correct use of radiother-
apy, local recurrences could be avoided in the
majority of patients, and today, in many centres
the local recurrence rate will not exceed 5%.
The aim in tailoring a follow-up programme
must be to find recurrent disease when still a
potential chance for cure exists. Since curative
treatment often includes major surgery, the
patients must be fit for a second major operation.
Therefore, it is essential to take into account the
L. Påhlman
patients’ co-morbidity and the timing when recur-
Department of Surgical Sciences, Uppsala University,
SE 751 85 Uppsala, Sweden rences do recur but also in which organ a recur-
e-mail: lars.pahlman@surgsci.uu.se rence could appear. In this chapter, a proposal for

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 351


DOI 10.1007/978-3-642-25005-7_35, © Springer-Verlag Berlin Heidelberg 2012
352 L. Påhlman

tailored follow-up is based upon how to find and should be followed. After surgery for stage II and
cure the distant metastases, taking care of local III disease, several randomised trials have tested
recurrences and screen for metachronous new the hypothesis of no or mediocre follow-up ver-
primaries in the large bowel. sus a more ambiguous follow-up. Data from
meta-analysis of all these trials indicate that fol-
low-up will increase survival but the timing is not
35.2 Who Shall Be Followed? known [1, 2]. There are programmes with follow-
up every third month or every 6 months or yearly.
The rationale for follow-up and surveillance for Only one randomised trial has addressed the tim-
metastatic disease or synchronous tumours ing of the follow-up. That is a Danish-Swedish
depends upon the health of the patient and the trial, COLOFOL, where stage II and III patients
co-morbidity. If the patients do not tolerate liver are randomised to either a follow-up after 1 and
surgery or lung surgery or is too sick to be oper- 3 years (the standard arm) or every 6 months, the
ated for a new primary, there is no real rationale first 2 years and then it is 3 years follow-up [3]. In
to follow the patient. Therefore, this has to be both arms, a final 5 years’ follow-up is required
taken into account before putting patients in the including a colonoscopy. At each scheduled fol-
follow-up programme. low-up, CT scan over the thorax and liver is rec-
The second decision making must be based ommended and also a carcinoembryonic antigen
upon the stage of the disease. Since patients with (CEA) test. If the scanning is negative but the
stage I disease rarely recur, there is no real ratio- CEA is raising, the patient has to be re-investi-
nale to follow those patients for metastatic dis- gated with other methods like PET-CT. In this
ease. However, they should be scheduled for trial, the recruitment has just been finished (Dec.
surveillance due to metachronous colonic cancer 2010) and the end point is overall survival and
or precancer lesions to the same extent as those possibility to offer curative treatment to those
with stage II and III disease (see below). with metastatic disease. In total, 2,500 patients
Subsequently, only patients with stage II and have been randomised.
III disease should be followed for distant metas- In patients with a stage IV tumour, with radi-
tases. Another group that should be followed is of cal resected metastases, it is obvious that such a
course patients who have been curatively treated tumour has a ‘metastatic capacity’, and therefore,
for stage IV disease with a radical resected pri- those patients might be scheduled to a tighter
mary as well as radically resected metastases to follow-up. However, no evidence exists, but most
the liver and/or lung. centres used to follow those patients every
6 months over a 2-year period.

35.3 Distant Metastases


35.4 Metachronous Bowel Tumours
The aim of the follow-up programme is to find
patients with recurrences, which are possible to In the perioperative workup, the whole bowel must
cure. If there is an extensive spread of occult met- be examined, excluding synchronous adenomas or
astatic disease, there is a risk that each individual cancers. Having done a ‘clean colon’, the question
cell will create metastases, ending up with a com- arises when the next colonoscopy should be done.
pletely incurable disease. However, this is not the One rather large randomised trial has addressed
most common scenario and importantly cannot this question, the Funen trial. Six hundred patients
be predicted in advance. Therefore, all patients were randomised to have a yearly colonoscopy
should be followed, provided they fulfil the crite- over a 10-year period in one arm, and in the other
ria depicted above. The most important question arm patients had a colonoscopy every fifth year. In
regarding tumour biology is whether metastases that trial, there was no difference in survival
will metastasise or not. No data support this, and, between the two arms, neither any difference
therefore, one could argue how often patients regarding uptake of adenomas or metachronous
35 What Is the Appropriate Timetable for Tailored Follow-up? 353

cancer [4]. Although this is the only trial but a scopic follow-up too often. Data do support that,
rather large one, one can justify that colonoscopy provided a ‘clean colon’ has been performed
surveillance is not indicated more often than in perioperatively, the next colonoscopy can be
5-year periods. Therefore, those who survive the postponed to the 5-year follow-up [4].
disease with no metastatic recurrences should be The liver and lungs should preferably be
scheduled for a colonoscopy 5 years after surgery scanned with the same method as was used in the
and then every fifth year until the age of 75 [5]. preoperative scanning, giving an opportunity to
The reason why there is no use to follow the compare and evaluate the findings. There are no
patients longer is that if the bowel is clean from strong data supporting a specific method, but for
adenomas or cancers at 75 years of age, it takes at the liver, contrast-enhanced ultrasound or CT scan
least 15 years to receive other adenomas and can- is available. For the lungs, CT scan or thoracic
cers in that age group, making the follow-up after plain X-ray has been suggested. For detection of a
75 years not cost effective. local recurrence, both endoscopic evaluation and
an imaging technique could be beneficial. The
MRI is probably better than CT scan since fibrosis
35.5 Local Recurrence is better displayed on MRI. Most patients with a
local recurrence experience pain, and, therefore,
Provided a centre has an optimal performance, not if a patient has pain, he/she should automatically
more than 5% of the patients will recur locally. be scheduled for a follow-up with MRI.
Those who recur are the patients with advanced
disease and with a circumferential resection mar-
gin of less than 1 mm [6]. The problem with local 35.7 Tumour Markers
recurrences is that the majority of them are
difficult to treat after TME surgery and only a Several follow-up programmes include testing
minority is possible to resect. Therefore, it is not with tumour markers. The most common used
cost-effective to have everyone in a follow-up marker is CEA. Several series have shown that a
programme, but only patients who are fit and will rising CEA indicates a recurrence, and this
tolerate major surgery can benefit from such a increase often occurs 6 months before a recur-
programme. Probably, a good history is enough rence becomes clinically detectable [2, 8].
together with the scheduled follow-up for distant Therefore, several programmes have included
metastases. Since a local recurrence is rather rare CEA testing in-between the imaging controls,
today, only those with symptoms should be evalu- although no studies have shown any clear advan-
ated with specific imaging. The timing is not tages with such a policy regarding overall sur-
known, but with the modern treatment including vival or cancer-specific survival.
neoadjuvant radiotherapy or chemoradiotherapy
data support that some of the recurrences will be Conclusion
postponed more than 3 years [7]. Therefore, one The available evidence will not support a
could argue to follow patients at risk more after specific timetable for tailored follow-up.
the period of follow-up for distant metastases Tumour biology is probably more important
(after 3 years) including a follow-up for local than the schedule. If there is a massive spread,
recurrences at 4 years and at the 5-year follow-up it will not matter how often a patient is sched-
when the colonoscopy is done. uled for the follow-up to be done since such a
patient is not curable. Therefore, it is probably
enough to see the patient every 6 months or
35.6 Imaging Technique even less. The COLOFOL trial will answer
that question. Regarding colonoscopy, surveil-
The question is which technique should be used. lance data support that only those who have
Most recurrences do recur outside the bowel, and, survived 5 years should be scheduled for a
therefore, there is no real rationale to do endo- follow-up every fifth year to the age of 75–80.
354 L. Påhlman

References surveillance by colonoscopy: effect on the incidence of


colorectal cancer. Telemark Polyp Study I. Scand J
Gastroenterol 34:414–420
1. Renehan AG, Egger M, Saunders MP, O’Dwyer ST
6. Quirke P, Steele R, Monson J, Grieve R, Khanna S,
(2002) Impact on survival of intensive follow up after
O’Callghan C, Sun Myint A, Bessell E, Thompson LC,
curative resection for colorectal cancer: systematic
Parmar M, Stephens RJ, Sebag-Montefiore D (2009)
review and meta-analysis of randomised trials. BMJ
Effect of the plane of surgery achieved on local recur-
324:813–821
rence in patients operated with operable rectal cancer:
2. Jeffery GM, Hickey BE, Hider P (2002) Follow-up
a prospective study using data from the MRC CR07
strategies for patients treated for non-metastatic col-
and NCIC-CTGCO16 randomised clinical trial. Lancet
orectal cancer (Cochrane Review). In: The Cochrane
373:821–828
Library, Issue 4. Update Software, Oxford
7. Khani MH, Smedh K, Kraaz W (2010) Is the circum-
3. Wille-Jørgensen P, Laurberg S, Påhlman L, Carriquiry
ferential resection margin a predictor of local recur-
L, Lundqvist N, Smedh RK, Svanfeldt M, Bengtson A
rence after preoperative radiotherapy and optimal
(2009) An interim analysis of recruitment to the
surgery for rectal carcinom? Colorectal Dis 9:
COLOFOL trial. Colorectal Dis 10:756–758
706–712
4. Jorgensen OD, Kronborg O, Fenger C (1995) A ran-
8. Körner H, Söreide K, Stokkeland PJ, Söreide JA (2007)
domized surveillance study of patients with peduncu-
Diagnostic accuracy of serum-carcinoembryonic anti-
lated and small sessile tubular and tubulovillous
gen in recurrent colorectal cancer: a receiver operating
adenomas. The Funen Adenoma Follow-up Study.
characteristic curve analysis. Ann Surg Oncol
Scand J Gastroenterol 30:686–692
14:417–423
5. Thiis-Evensen E, Hoff GS, Sauar J, Langmark F,
Majak BM, Vatn MH (1999) Population-based
How Should Data Be Shared
and Rapid Learning Health Care 36
Promoted?

Ruud van Stiphout, Erik Roelofs, Andre Dekker,


and Philippe Lambin

Contents 36.1 Clinical Decision Making


36.1 Clinical Decision Making .......................... 355
36.2 Electronic Health Record .......................... 356 In the medical field and inherently also in oncol-
36.3 Data Sharing............................................... 357 ogy, clinical guidelines, protocols, and research
36.3.1 Semantic Interoperability............................. 358 are highly regulated. Nevertheless, both treat-
36.3.2 Current Initiatives ........................................ 359 ments and technologies are surrounded by uncer-
36.4 Prediction Applications ............................. 359 tain factors. Evidence is primarily generated by
36.4.1 Predictive Modeling ..................................... 359
the outcomes of randomized clinical trials. This
36.4.2 Current Models ............................................ 361
36.4.3 Knowledge Dissemination ........................... 362 evidence is however often unavailable, inconclu-
36.5 Toward Rapid Learning sive, valid for only a subgroup of patients, out-
Health-Care System ................................... 363 dated, or of insufficient quality. Therefore, this
References ................................................................. 363 process may not always be suitable to fully base
clinical decisions on. Furthermore, there still
exists a translational gap between scientific dis-
coveries and clinical practice.
A transformation of the way in which evidence
is aggregated and applied may be required.
Substantial investments in health information
technology (HIT), comparative effectiveness
research, health-care quality and value, and per-
sonalized medicine support these efforts.
Personalized medicine, i.e., match individual
patient data with detailed evidence available in
real time, is an emerging trend. In parallel, clini-
cal decision making for a single patient has
become more complex due to the increased
amount of data; data required for decision mak-
ing has exponentially increased from 10 to 10,000
R. van Stiphout () • E. Roelofs • A. Dekker • P. Lambin
Department of Radiation Oncology (MAASTRO), facts per decision in the last two decades, if data
GROW – School for Oncology and Developmental from genetics, proteomics, and diagnostic imag-
Biology, Maastricht University Medical Centre+, ing are taken into account. Human cognitive
Maastricht, The Netherlands
capacity is only able to deal with five facts per
e-mail: ruud.vanstiphout@maastro.nl;
rik.roelofs@maastro.nl; andre.dekker@maastro.nl; decision, i.e., decision by clinical symptoms.
philippe.lambin@maastro.nl In Fig. 36.1, examples of data origins are given

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 355


DOI 10.1007/978-3-642-25005-7_36, © Springer-Verlag Berlin Heidelberg 2012
356 R. van Stiphout et al.

Demographics Imaging Pathology Toxicity Blood biomarkers Genomics Proteomics

Rectal
cancer Treatment
patient
Age, gender (Diffusion)-MRI Tumor staging Gastro-intestinal Biological Proliferation Structure
decision
Medical history (Perfusion)-CT Resection quality Dermatology Growth factors Hypoxia Cellular defense
Social status PET / SPECT Genitor-urinary Tumor load Metabolism
Health status Endoscopy Neuropathy Infalmmation RNA processing
Work history Bone-marrow Metabolism Cycle regulation
Lifestyle Cardiac Angiogenesis
Pain Translation

Fig. 36.1 Treatment decision for rectal cancer patients can be based on various data origins, each complementing each
other in capturing disease characterization and progression

for a typical patient diagnosed with (rectal) can- electronic health record development is most
cer. This increase in decision-making complexity focused on.
requires decision support systems (DSS) and From medical literature of the last two decades
computational support on different levels and (Fig. 36.2), a steep increase is seen of the number
steps in the treatment process [5]. of publications related to clinical decision mak-
The development of a rapid learning health- ing, database infrastructure (represented by ontol-
care system may be very effective to achieve this ogy), and data sharing. While clinical decision
support [1]. In such a system, data is routinely making was important before 1990, research
generated through patient care and clinical involving data ontology and data sharing
research and fed into an ever-growing databank increased only after 2000, in parallel with an
or set of coordinated databases. High-quality data increase of Internet availability and computa-
should be available in real time, simultaneously tional power. Cancer-related publications show a
used to improve clinical care, yield quality mea- proportional trend, except that the number of data
sures, and focus on research. This system intends ontology and data sharing publications stagnates
to improve collaborative care, patient safety, and the last couple of years.
health-care quality and efficiency and supports
medical and clinical research, training, and pub-
lic health. It would also expand the pace and 36.2 Electronic Health Record
magnitude of evidence generation. The steps to
be taken within the rapid learning health-care Currently, electronic health records (EHR) are
system are: the main focus of investments made to transform
1. Collecting data in standardized manner health care. Paper-based records are still the pre-
2. Analyzing captured data ferred method of recording patient information
3. Generating evidence through retrospective for most hospitals and practices in the Western
analyses and prospective studies countries. Medical professionals experience in
4. Implementing new insights in clinical care general that it is easier to record data in paper
5. Evaluating outcomes of changes in care records than in electronic records. Handwritten
6. Generating new hypotheses for investigation paper medical records however can be associated
Many aspects are involved in the develop- with poor legibility, which can contribute to med-
ment of these systems, like setting up and link- ical errors. Standardization in form design, abbre-
ing registries and databases, implementing an viations, and penmanship were improved to
information technology infrastructure, and improve reliability of paper medical records. But
developing applications and interfaces for the shift toward digital records is currently inevi-
patient-centered clinical decision support and table in the ongoing globalization of health care.
policy issues and implications. Each of these Paper records require significant amount of stor-
aspects will be discussed in this chapter. Some age space, and it is very time consuming and
investments have already been made, of which complicated to collate them to a single location to
36 How Should Data Be Shared and Rapid Learning Health Care Promoted? 357

Clinical decision making Data ontology Data sharing


700 500 120
Overall Overall Overall
600 Cancer related Cancer related Cancer related
100
400
Number of publications

500
80
300
400
60
300 200
40
200
100
100 20

0 0 0
1990 1995 2000 2005 2010 1990 1995 2000 2005 2010 1990 1995 2000 2005 2010
Year of publication Year of publication Year of publication

Fig. 36.2 Number of publications over the last two decades related to clinical decision making, data ontology, and data
sharing. Overall and cancer-related publications are reported

be reviewed by a health-care provider. Related records is the first and most crucial requisite to
copying, faxing, and transporting costs are achieve this. It becomes however more challeng-
required for multiple institutional evaluations. ing when data is shared among institutes on a
EHRs on the other hand have increased porta- regional, national, and international level. Data
bility and accessibility and can be instantly updated warehouses and connected medical software have
(within certain legal limitations). Furthermore, it to “understand” what the exchanged data means in
is possible to exchange records between different order to process it. These machine-machine inter-
EHR systems, and data can be used anonymously actions seem straightforward to humans because
for statistical reporting for quality improvement, our brain is continuously and unconsciously pro-
resource management, and public health commu- cessing information in such a manner, but they are
nicable disease surveillance. However, the down- not. What may give insight is the process of
side of the improved accessibility of electronic knowledge generation in decision making
records is that unauthorized persons may be able (Fig. 36.3). This process describes the conversion
to access sensitive medical data. This concern of data to information by putting it in context, gen-
about security is the main reason why resistance is erating knowledge by making sense of the infor-
shown to their widespread adoption. mation, and comparing all the knowledge in the
In current developed EHRs, the quality of data field to find the best path to take in the decision.
is still a concern. For example, issues related to For example, if for a rectal cancer patient
data accuracy, completeness, and comparability cT = 3 is measured (data), one has to know that cT
must be addressed before routine EHR-based means clinical tumor stage, which is part of the
quality of care measurement can be done with TNM classification system, and is measured by
confidence [2]. It is expected that if system capac- imaging and has a value range of 0–4 (informa-
ity grows, users become more comfortable with tion). Subsequently, a physician knows that this
new health IT systems, and if continuous quality information means that the patient has an
improvement principles are applied to the EHR- extended tumor without invasion of any sur-
based data collection-storage-retrieval process, rounding organs but has poorer outcome than
the quality of data in EHRs will improve. lower staged tumors (knowledge). From litera-
ture, it is known that this kind of patients have the
best outcome when receiving long-course chemo-
36.3 Data Sharing radiotherapy (options), thus the patient will
receive this treatment (action).
Standardization of data definition and handling Machines however are only able to deal with
within hospitals and institutes is very important. data and generate information by using terminol-
As stated, proper development of electronic health ogies, definitions, and ontologies. An ontology is
358 R. van Stiphout et al.

Fig. 36.3 Process of


knowledge generation and DATA Analysis Information is data in context
subsequent action for decision
making based on the
Poindexter model Sense Understanding what the
INFORMATION
making information means

Path Deciding what to


KNOWLEDGE
finding do within constraints

OPTIONS Execution

lteration ACTIONS

a standardized representation of knowledge as a There are several standards available to


set of concepts within a domain and the relation- achieve SIO in health care. The most well-known
ships between those concepts. and promoted standard for medical terminologies
is SNOMED CT (Systematized Nomenclature of
Medicine – Clinical Terms). This standard was
36.3.1 Semantic Interoperability created in 2002 and is used in many systems like
EHRs, diagnostic imaging services, knowledge
To standardize data sharing, semantic interoper- databases for clinical decision support, etc. A
ability (SIO) is required. In general, SIO is the standardized system results in common structure
ability of any communicating entity (not only and terminology and a single data source for
computers) to share unambiguous meaning. For review (less redundant data). It also allows the
two computers, this is the ability to exchange use of common tools and techniques, common
information and have that information properly training, and single validation of data. It is how-
interpreted by the receiving system in the same ever also true that not all standards are of equal
sense as intended by the transmitting system. quality. Continuing with bad standards may even-
Four levels of semantic interoperability can be tually result in decrease in quality of care and
distinguished: high costs.
0. None: data cannot properly be transferred to Currently, many data sharing systems do not
the receiver achieve level 2 or 3 semantic interoperability.
1. Technical and syntactical: only the raw data is Realizing full SIO may however not necessarily
transferred without any translation be a consensual goal in every place and any time
2. Partial: some transferred data is converted because of social, cultural, and human factors.
with standardized coding SIO emerges gradually and may remain an
3. Full: sharable context, seamless co-operability incomplete phenomenon, even with the most
Currently, many data intended for care pro- optimistic of assumptions. Concerns about pri-
vision are still shared by remote access to clini- vacy and confidentiality will continue to be a key
cal systems, and for research data spreadsheets limiting factor in interoperability and may impede
are mainly used, which is still level 1 SIO. Some developments that would be technically feasible
systems, as the ones described in Sect. 36.3.2, and beneficial. Anonymization, i.e., the act of
explored and implemented level 2 interopera- permanently and completely removing personal
bility, but level 3 is and may remain still the identifiers from data, should therefore be applied
future. in trans-institutional sharing when possible.
36 How Should Data Be Shared and Rapid Learning Health Care Promoted? 359

36.3.2 Current Initiatives detection, diagnosis, treatment, and preven-


tion. It is founded on the core principles of
Several applications of data sharing for open access, open development, open source,
cancer-related data have been initiated in and federation of resources for either local or
the last decade. shared control.
• EUROCAT (www.eurocat.info): a shared cen- • CancerData (www.cancerdata.org): an open-
tralized database of medical characteristics in source driven medical data repository con-
cancer patients, tumors, and treatments initiated nected to the caGrid. Using some of the caBIG
by several institutes across Europe. The goals tools, images and annotations are offered in
are to be able to better predict the outcome of public data collections in DICOM and AIM1
individual cancer treatments and to be assistive format, respectively. Private data collections
in clinical trial recruitment and analysis. are available for project members after log-
• MISTIR (www.mistir.info): a secured central- ging in. Furthermore, CancerData offers a
ized database of annotated images of cancer platform to exchange data analysis tools and
patients allowing “multicentric planning stud- share information on medical imaging and
ies” in strictly protocolized “in silico (virtual) knowledge engineering in user forums.
clinical trials” [6]. The MISTIR framework
offers high-quality data by applying tools for
quality assurance and solving data incompati- 36.4 Prediction Applications
bility and transfer and storage issues. The treat-
ment planning analysis is performed centrally 36.4.1 Predictive Modeling
to guarantee accurate and uniform results.
• RODA (www.elekta.com): the Radiation When data is sufficiently available and well
Oncology Data Alliance is an initiative with the defined and if the data infrastructure allows for
ultimate goal to create an international radiation data mining, individualized medicine becomes an
oncology registry for collecting and analyzing option. This trend in cancer care involves admin-
data from practices worldwide and correlating istration of the most optimal treatment depending
the data with patient outcomes. An IT infra- on certain prognostic characteristics of the indi-
structure, in which data is collected through vidual patient and the tumor. These prognostic
electronic health records, de-identified, and characteristics for treatment outcome, called fea-
aggregated in near real time to a central data tures, can either be identified from published lit-
repository, is provided by Elekta Software. erature or learned from the current data (feature
• Oncospace (www.hopkinsmedicine.org): a selection). These features will subsequently serve
distributed analytic database for structured as an input for a predictive computational model,
information modeled after the successful which aims at finding the optimal fit of the input
SkyServer program for astronomic data. This data to the treatment outcome. These models
is an initiative of the Johns Hopkins University have a certain accuracy to predict outcome for a
and the University of Dallas. Despite the fact population of patients, and it is important to eval-
that only data for head and neck cancer is uate this accuracy on new data which is not used
shared between those two institutes and ana- during the learning process, preferably an exter-
lyzed, it shows that integration of knowledge nal dataset. Some aspects of this process will be
and expertise from outside the medical domain discussed in the following sections.
may boost optimization of the system.
• caBIG (cabig.nci.nih.gov): the Cancer 36.4.1.1 Data Requirements
Biomedical Informatics Grid was initiated by First of all, large amounts of data are required for
the National Cancer Institute, and its data proper statistics. In clinical trials, sample size cal-
sharing and analyzing infrastructure is focus- culations are performed to determine the minimum
ing on the complete cycle of cancer disease, amount of patients required to detect a certain
360 R. van Stiphout et al.

difference in outcome. In model development, • Binary outcome: two outcome labels, like for
this is not common since there is usually a example treatment response or no response.
restricted amount of suited data available. It is Methods: logistic regression, support vector
more important to increase the ratio between machines.
patients and features to avoid overfitting. This • Ordinal outcome: three or more outcome val-
occurs when a statistical model describes random ues, like for example toxicity grading.
error or noise instead of the underlying relation- Methods: Naïve Bayes, k-means clustering,
ship because the model is too complex. There are Bayesian nets, decision trees. These methods
no strict guidelines for this ratio because of the can also be applied in binary outcome
high variety in data heterogeneity, but as a rule of prediction.
thumb, 5–10 patients per feature are minimally • Time-to-event: follow-up outcomes like local
required. If increasing the number of patients is recurrence status or overall survival with cor-
not possible, reducing the number of features is responding time-to-event. Methods: Cox pro-
necessary. The best method for feature reduction portional hazards (PH) model, Fine and Gray
is selection based on knowledge from literature model (competing risks).
and medical expertise.
The quality of the data may also affect the 36.4.1.3 Performance Evaluation
accuracy of the models. After collection, one may Evaluation of the performance of the model is
be able to deal with wrongly entered data or miss- essential when selecting important features and
ing data. Typos in data are usually found with comparing classification methods. When a pre-
outlier detection, either without prior knowledge diction model is developed, reporting the accu-
or with prelabeled data (normal or abnormal). racy of the model is required for the acceptance
Usually, normal value ranges of prognostic fac- of the model in the target community. In the
tors are known from literature. There are many simplest case of two outcomes, performance is
methods to deal with missing data, but they usually given as sensitivity, i.e., proportion of
mainly involve: actual positive labels which are correctly
• Imputation: missing values are substituted by identified as such, and specificity, i.e., the pro-
the most likely value, calculated by, for exam- portion of negatives which are correctly
ple, the mean or expected maximization identified. When however the discrimination
algorithm. threshold for the classifier is varied, a spectrum
• Partial deletion: patients with too many miss- of sensitivities and specificities is generated.
ing data will be excluded from the analysis. The receiver operating characteristic (ROC)
• Full analysis: the classification method is able curve is the plot of sensitivity vs. 1-specificity.
to deal with missing values. Assumptions and The ROC curve allows selection of the optimal
distortions in the data can be avoided. models because there is always a trade-off
between sensitivity and specificity, and one
36.4.1.2 Model Types might be more important than the other for
In traditional statistics, there is a subset of meth- specific outcomes (Fig. 36.4). A more robust
ods which are used most frequently for cancer- measure for performance is the area-under-the-
related studies. In general, alternative methods curve (AUC) of the ROC curve. A value of 1
are not easily accepted in the medical commu- corresponds to perfect prediction, while an AUC
nity, as for example, methods from machine of 0.5 means random prediction. For time-to-
learning. The method of choice to predict out- event–related data, an equivalent of the AUC,
come is dependent on the type of input and out- the c-statistic, has to be used to deal with cen-
put data. Exploratory modeling studies are sored data.
required to find the optimal model for the avail- It is very important to calculate the performance
able data. When distinguishing outcome types, of the model on new data, which was not used in
the following examples can be considered: the training process to guarantee robustness of the
36 How Should Data Be Shared and Rapid Learning Health Care Promoted? 361

Predicted outcome 1
positive negative
0.9
positive

True positive False negative 0.8


Actual outcome

(TP) (FN)
0.7

0.6

Sensitivity
negative

0.5
False positive True negative
(FP) (TP)
0.4

0.3

TP 0.2
Sensitivity = TP + FN Random performance (AUC = 0.50)
0.1 Moderate performance (AUC = 0.70)
TN Good performance (AUC = 0.85)
Specificity = TN + FP 0
0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1
1-specificity

Fig. 36.4 Explanation of the confusion matrix (left) definitions in the confusion matrix. By using different
showing the definitions of patient numbers when compar- thresholds in the model, a range of sensitivities and
ing predicted with actual outcomes (positive vs. negative). specificities result in the ROC curve (right)
Sensitivity and specificity are calculated using the

model. There are several options available to do biased accuracy because the validation set is
this validation step: independent.
• Random and temporal split: the data is split
into two sets, one for training and one for vali-
dation. Usually, 70–90% of the data is used for 36.4.2 Current Models
training and 10–30% for validation. This split
can be done randomly or based on patient’s For rectal cancer, not many multivariable models
intake date (temporal). are available in literature. An example of a pre-
• Cross-validation: involves partitioning the diction model for follow-up outcome for locally
data into complementary subsets, train the advanced rectal cancer was recently published
model on one subset and validating the analy- [8]. In this study, four European trials (N = 2,242)
sis on the other subset. To reduce variability, were used to train a Cox PH model to predict
multiple rounds of cross-validation are per- local recurrences, distant metastases, and overall
formed using different partitions, and the vali- survival based on clinical and pathological fea-
dation results are averaged over the rounds. tures. The model was externally validation by
Usually five- or ten-fold cross-validation is another European trial (N = 553). The prediction
performed. Leave-one-out (LOO) cross-vali- models were converted to nomograms for easy
dation is the extreme case in which each round and visual interpretation [4]. The prediction
one patient is the validation dataset. This LOO model performed well on the external dataset
method is typically used when dealing with with c-statistics around 0.7. Other examples of
small amounts of data. successful multivariable models have been pub-
• External validation: comparable data from lished [3, 9]. The upcoming trend of public shared
another institute or another trial is used for data also results in published models based on
validation. This method provides least public repositories [7] (Fig. 36.5).
362 R. van Stiphout et al.

Score
−10 −8 −6 −4 −2 0 2 4 6 8 10

pT-stage
0 1 2 3 4

pN-stage
0 1 2

cT
1+2 3 4
Age [years]
25 35 45 55 65 75 85
Adjuvant chemo
Yes No

Surgery procedure
LAR APR
Radiotherapy dose [Gy]
>45 45 <45
Sex
Female Male

Sum of scores
−20 −10 0 10 20 30
Low Medium High
Probability of death
5 10 20 30 40 50 60 70 80 90 95
within 5 years [%]

Training dataset Vaildation dataset


1 1
0.88
0.9 0.9 0.94 0.94
0.8 0.82 0.8 0.83
0.81 0.81
Overall survival [−]

0.7 0.7
0.67
0.6 0.6 0.62
0.59 0.57
0.5 0.5
0.4 0.4
0.4
0.3 0.3
0.2 0.2
Low risk: N = 261 Low risk: N = 50
0.1 Medium risk: N = 669 0.1 Medium risk: N = 160
High risk: N = 1312 High risk: N = 343
0 0
0 20 40 60 80 100 120 0 20 40 60 80 100 120
Time from randomization [months] Time from randomization [months]

Fig. 36.5 Example of a prediction model for rectal can- bility for death within 5 years (bottom scale). The Kaplan-
cer: nomogram to predict overall survival for locally Meier curves show for both the training and the validation
advanced rectal cancer. In the nomogram, the predictor datasets what the overall survival is for the three risk
values for a single patient correspond to a score (upper groups provided by the nomogram
scale). The sum of all the scores corresponds to a proba-

36.4.3 Knowledge Dissemination expected in the health-care community. Clinicians


are the main users of these tools, and they prefer
Knowledge representation is as important as to end up with clinically relevant outcomes mea-
knowledge extraction from data. Bad interpret- sures, such as the predicted probability of a cer-
ability of the results may lead to false conclusions tain event within a given time. Avoiding redundant
and wrong decisions. The initiatives described in statistical results from intermediate steps is there-
Sect. 36.3.2 all have their own interface and pos- fore a prerequisite.
sibilities to mine and analyze data, but permis- The most well-known online website with
sion to use it is often restricted to participating interactive prediction tools is Adjuvant! Online
members only. (http://www.adjuvantonline.com). This website
A more public approach is to make prediction aims to provide decision support for adjuvant ther-
models available on the Internet. When interac- apy (chemotherapy, hormone therapy, etc.) after
tive, peer-reviewed models are provided with surgery for early cancer patients. A prediction
sufficient background information, an increase in website which focuses more on decision support
the acceptability and a raised awareness in favor for radiotherapy was recently set up by MAASTRO
of computer-assisted personalized medicine are Clinic (http://www.predictcancer.org). Also, the
36 How Should Data Be Shared and Rapid Learning Health Care Promoted? 363

Memorial Sloan-Kettering Cancer Center has pre- decision making and care delivery will be
diction tools for different cancer types (http:// increased. Patients are already using the Internet
www.mskcc.org/mskcc/html/5794.cfm). to obtain information about the cancer and treat-
ments, investigate providers and institutions,
and network with other patients. Social net-
36.5 Toward Rapid Learning Health- works can accelerate patient’s access to relevant
Care System information, disseminate clinical trial informa-
tion, expedite recruitment to trials, and thus fos-
As already described in the previous sections, ter research opportunities and data collection
developing a rapid learning health-care system from within patient communities.
requires transformation of IT infrastructure, stan- Some critical hurdles might be encountered
dardization of electronic health records, and seman- while implementing the system. Adding new data
tic interoperability between data sharing systems. collection tasks to the already packed clinicians’
Furthermore, development of accurate prediction schedules may be unrealistic and/or may result in
models and proper knowledge dissemination are poor quality of data. The shift of perspective from
essential for computational clinical decision sup- current care to efforts for future care may be the
port. Linking data systems to regional and national hardest part.
registry-based administrative data also allows for In summary, while research and new discov-
immediate case acquisition and real-time tracking ery are important, the focus needs to be on design-
of care. Technical changes are however not ing new national data policies and HIT systems,
sufficient for a successful implementation of the ensure data interoperability, creating a culture of
system. First of all, several cultural changes should sharing with simultaneous respect for patient pri-
be induced: vacy, and developing methods to make sense of
• Commitment from all levels within and across information and present it to its end users. This
organizations should ultimately lead to the concept of “rapid
• Community participation for infrastructure learning health care.”
development
• Persuade stakeholders to open up information
silos
References
• Federal coordination of intellectual capital
and acknowledgements 1. Abernethy AP, Etheredge LM, Ganz PA et al (2010)
• Leadership and active management Rapid-learning system for cancer care. J Clin Oncol
• Addressing data governance and patient 28(27):4268–4274
2. Chan KS, Fowles JB, Weiner JP (2010) Electronic
privacy
health records and the reliability and validity of quality
• Top research papers and grant funding bodies measures: a review of the literature. Med Care Res Rev
which request open-source data sharing 67(5):503–527
Second, guidelines and quality control consid- 3. Dehing-Oberije C, Aerts H, Yu S (2010) Development
and validation of a prognostic model using blood bio-
erations are very important. These include uni-
marker information for prediction of survival of non-
versally accessible EHR, universally accessible small-cell lung cancer patients treated with combined
patient treatment plans and patient information, a chemotherapy and radiation or radiotherapy alone. Int
common pathology staging system, integrated J Radiat Oncol Biol Phys 81:360–368
4. Iasonos A, Schrag D, Raj GV et al (2008) How to build
clinical/laboratory and pathology modules, a
and interpret a nomogram for cancer prognosis. J Clin
referral system to clinical trials, quality reporting Oncol 26(8):1364–1370
and resource commitment to engage ongoing 5. Lambin P, Petit SF, Aerts HJ et al (2010) The ESTRO
improvement, assessment of practice patterns, Breur Lecture 2009. From population to voxel-based
radiotherapy: exploiting intra-tumour and intra-organ
and signal detection for adverse events.
heterogeneity for advanced treatment of non-small
Thirdly, by promoting patient activation and cell lung cancer. Radiother Oncol 96(2):
empowerment, patient participation in both 145–152
364 R. van Stiphout et al.

6. Roelofs E et al (2010) Design of and technical chal- metastases, and overall survival for patients with locally
lenges involved in a framework for multicentric radio- advanced rectal cancer on the basis of European ran-
therapy treatment planning studies. Radiother Oncol domized clinical trials. J Clin Oncol 29:3163–3172
97(3):567–571 9. van Stiphout RG, Lammering G, Buijsen J et al (2011)
7. Starmans MH, Zips D, Wouters BG (2009) The use of Development and external validation of a predictive
a comprehensive tumour xenograft dataset to validate model for pathological complete response of rectal
gene signatures relevant for radiation response. cancer patients including sequential PET-CT imaging.
Radiother Oncol 92:417–422 Radiother Oncol 98(1):126–133
8. Valentini V, van Stiphout RGPM, Lammering G (2011)
Nomograms for predicting local recurrence, distant
Index

A caudal border of the CTV, 152


Anatomy, 4–5, 32, 35, 38, 61, 68, 78, 117, 120, 131, contouring guidelines, 118
133, 136, 156, 235, 237, 264–266, 328, 351 cranial border of the CTV, 150–151
CTV definition, 125
Diagnosis
B height of tumour, 29, 30
Biochemical and molecular biological factors imaging work-up, 10
for adjuvant and systemic therapy, 45–47 mesorectal fascia, 29, 32, 62, 64,
radiochemotherapy, 42–44 67–71, 262
for surgical decisions, 44–45 T-staging, 32–35
for treatment decisions, 41–47
Brachytherapy
clinical results, 166–167 E
contact x-ray 50kV (CXRT), 165–167 Electronic health record, 356–357, 359, 363
contribution in tailoring treatment, 167–168 Endoanal ultrasound (EAUS), 28–29, 32, 35, 61
elderly patient, 168–169 Epidemiology, 3, 21
endoluminal, 164, 166–168
interstitial, 164, 165, 167, 168
F
Follow-up
C distant metastases (DM), 50, 352
Chemotherapy local recurrence (LR), 50, 51, 280, 282, 297–298,
adjuvant chemotherapy, 15, 41, 45, 46, 50, 52, 97, 352, 353
106, 108, 157, 160, 173, 174, 194–199, 200, 210, metachronous bowel tumours, 352–353
217–223, 314 tumour markers, 353
biologic targeted agents, 181–188
combined EGFR and VEGF, 188
EGFR inhibitors, 182–185 I
metastases, 50, 54, 75, 109, 113, 173–175, 178, 181, Intraoperative radiotherapy (IORT), 118, 155–160,
186, 193, 200, 205–207, 209–214, 219, 221, 292, 166, 280–284
298, 299
oxaliplatin, 173–178, 185, 186, 188, 195, 196, 198,
209–213, 217, 219–222, 276–278 L
upfront chemotherapy, 193–201, 213 Locally advanced primary rectal cancer, 69,
VEGF inhibitors, 186–188 155–156
Clinical decision making, 54, 316, 342–344, 355–357 Local relapse
Computed tomography (CT), 7, 24, 27, 29, 32, CEA surveillance, 97
36–38, 68, 69, 73, 74, 96–102, 117, 118, endoscopy surveillance, 97
121, 124, 125, 131–133, 135, 136, 138, 139, limitations in the imaging detection, 99–100
141, 144, 165, 209, 212, 262, 275, 278, 284, new techniques in the evaluation of local relapse,
287, 295, 305, 344, 352, 353 100–101
role of CT, 98, 99
role of imaging surveillance, 97
D role of MRI, 98–99, 101, 102
Data sharing, 356–359, 363 role of PET, 98, 100
Delineation surveillance and at what frequency, 96

V. Valentini et al. (eds.), Multidisciplinary Management of Rectal Cancer, 365


DOI 10.1007/978-3-642-25005-7, © Springer-Verlag Berlin Heidelberg 2012
366 Index

M EGFR inhibitors, 182, 185


Magnetic resonance imaging (MRI) meta-analysis, 109
diffusion-weighted magnetic resonance imaging oxaliplatin, 174, 175, 177, 178
(DWI), 79–87 selection of patients, 109, 113–115
dynamic contrast-enhanced MR imaging surgical procedure avoidance, 291–300
(DCE-MRI), 87–88 upfront chemotherapy, 193–201
extramural venous invasion (EMVI), 30–32, 200 VEGF inhibitors, 186–188
mesorectal fascia involvement, 69, 70 Preoperative short-course radiotherapy
mesorectal spread, 62–64 acute and late toxicity of SCPRT and CRT, 111
pretreatment ADC value, 84–85 clinical trials, 106–108
qualitative assessment of DWI, 84 meta-analysis, 108–109
Metastases population-based data, 22, 108
extra-hepatic disease, 36–38 selection of patients, 109, 113–115
hyperthermic intraperitoneal chemotherapy Prognostic clinical factors
(HIPEC), 213, 278 distance of the tumor to mesorectal fascia, 24–25
initially unresectable metastases, 211–212 histopathology on pretreatment biopsies, 25–26
limited peritoneal disease, 213 pretreatment laboratory findings, 26
liver, 37, 206, 207, 209, 210, 212–214, 352 sex, age, and body mass index, 21–22
R0 resectable liver +/− lung metastases, 207, tumor size and location, 22–23
209–211

R
N Radiotherapy treatment technique
Nodal metastases, 45, 73–75, 157, 166, 298, 314 benefits of IMRT, 132
Nodal staging, 36, 73–75, 157, 160, 322 correcting shape variation, 141–142
IGRT, 129–145
patient orientation and belly board use, 136
P planning target volume margins, 139–141
Pathology possibility of dose escalation, 143–145
APR grading, 311–312 prerequisites for IMRT, 132–134
circumferential resection margin involvement, to reduce irradiated small bowel volume, 134–138
310–311 reduction of CTV, 136–138
correct procedure for handling the specimen, set-up errors, 133, 138, 141, 142
305–316 target volume shape variation, 138–139
direct tumour spread and pT stage, 309–310 Rapid learning health, 355–363
histological typing and grading, 309 Rectal function, 5–6
key features in the pathology report, 308–316
local peritoneal involvement, 312
lymph node metastases and pN stage, 314–315 S
mesorectal grade, 309, 311, 312 Surgery
prognostic value of (y)pN, 319–325 APR grading, 311–312
prognostic value of (y)pT, 319–325 diverting stoma, 257–259
prognostic value of TRG, 333–337 extended resections, 275–287
specimen after neoadjuvant radiotherapy, 315–316 extralevator abdominoperineal excision,
vascular invasion, 308–315, 323, 324, 328 261–271, 312
Pathophysiology, 6–7 laparoscopic, 30, 186, 240, 241, 245, 249–255,
Positron emission tomography-computed tomography 267, 293, 298, 349
(PET-CT) nerve-sparing surgery, 233–245
early response evaluation FDG PET-CT, 89 quality of surgery, 177, 229–231, 261, 262, 265,
presurgical FDG PET-CT, 89–91 269, 270, 306, 316, 329, 330
pretreatment FDG PET-CT, 89 surgical procedure avoidance, 291–300
Prediction applications, 359–363
Preoperative long-course chemoradiotherapy
acute and late toxicity of SCPRT and CRT, 111 T
adjuvant chemotherapy, 106, 108 Treatment strategy
biologic targeted agents, 114 cM1, non-synchronous, 15
clinical trials, 109, 113, 114 cM1, resectable synchronous metastases, 14
combined EGFR and VEGF, 188 cM1, unresectable synchronous metastases, 14
Index 367

cT1 N0 M0, 11 Tumour heterogeneity


cT2 N0 M0, 11 based on biological evidence, 50
cT3 (MRF−) N0-2 M0, 12 based on clinical observation, 50–52
cT3 (MRF+) N0-2-M0 or cT4 any N M0, 12 surrogate endpoints, 52–55
local recurrence, cM0, 13

S-ar putea să vă placă și