You are on page 1of 259

Manual of

Total
Mesorectal
Excision

HEBK001-FM.indd 1 11/02/13 6:40 PM


HEBK001-FM.indd 2 11/02/13 6:40 PM
Manual of
Total
Mesorectal
Excision

Edited by
Brendan Moran, MCh, FRCSI, FRCS, Consultant Surgeon Basingstoke
and North Hampshire NHS Trust; Honorary Senior Lecturer,
Southampton University, UK

Richard John Heald, CBE, MCHIR, FRCS, Clinical Director, Pelican


Cancer Foundation, Basingstoke, UK; Honorary Professor of Surgery,
Southampton University, UK; President of the Colostomy Association

HEBK001-FM.indd 3 08/02/13 5:27 PM


CRC Press
Taylor & Francis Group
6000 Broken Sound Parkway NW, Suite 300
Boca Raton, FL 33487-2742

© 2013 by Taylor & Francis Group, LLC


CRC Press is an imprint of Taylor & Francis Group, an Informa business

No claim to original U.S. Government works

Printed on acid-free paper


Version Date: 20130129

International Standard Book Number-13: 978-1-4441-1716-5 (Hardback)

This book contains information obtained from authentic and highly regarded sources. While all reasonable efforts have been made to
publish reliable data and information, neither the author[s] nor the publisher can accept any legal responsibility or liability for any errors
or omissions that may be made. The publishers wish to make clear that any views or opinions expressed in this book by individual editors,
authors or contributors are personal to them and do not necessarily reflect the views/opinions of the publishers. The information or guid -
ance contained in this book is intended for use by medical, scientific or health-care professionals and is provided strictly as a supplement to
the medical or other professional’s own judgement, their knowledge of the patient’s medical history, relevant manufacturer’s instructions
and the appropriate best practice guidelines. Because of the rapid advances in medical science, any information or advice on dosages, pro-
cedures or diagnoses should be independently verified. The reader is strongly urged to consult the drug companies’ printed instructions,
and their websites, before administering any of the drugs recommended in this book. This book does not indicate whether a particular
treatment is appropriate or suitable for a particular individual. Ultimately it is the sole responsibility of the medical professional to make
his or her own professional judgements, so as to advise and treat patients appropriately. The authors and publishers have also attempted
to trace the copyright holders of all material reproduced in this publication and apologize to copyright holders if permission to publish in
this form has not been obtained. If any copyright material has not been acknowledged please write and let us know so we may rectify in
any future reprint.

Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced, transmitted, or utilized in any form by
any electronic, mechanical, or other means, now known or hereafter invented, including photocopying, microfilming, and recording, or in
any information storage or retrieval system, without written permission from the publishers.

For permission to photocopy or use material electronically from this work, please access www.copyright.com (http://www.copyright.
com/) or contact the Copyright Clearance Center, Inc. (CCC), 222 Rosewood Drive, Danvers, MA 01923, 978-750-8400. CCC is a not-for-
profit organization that provides licenses and registration for a variety of users. For organizations that have been granted a photocopy
license by the CCC, a separate system of payment has been arranged.

Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used only for identification and
explanation without intent to infringe.
Visit the Taylor & Francis Web site at
http://www.taylorandfrancis.com

and the CRC Press Web site at


http://www.crcpress.com

HEBK001-FM.indd 4 12/02/13 8:46 PM


Contents

Contributors vii

  1 The evolution of a concept: the total mesorectal excision story 1


R.J. Heald
  2 Anatomy of the rectum, anal canal and pelvic floor 31
Thilo Wedel
  3 Clinical ultrasound 52
Oliver Shihab, Arcot K. Venkatasubramaniam
  4 Magnetic resonance imaging staging of rectal cancer 58
Peter How, Gina Brown
  5 Radiological staging for systemic disease 71
Gina Brown, Chris Hunter
  6 Preoperative radiotherapy and chemoradiotherapy for rectal cancer 87
Rob Glynne-Jones, Mark Harrison
  7 Total mesorectal excision for rectal cancer 103
Brendan Moran
  8 Abdominoperineal excision of the rectum 124
Torbjörn Holm
  9 Laparoscopic surgery 140
Katharine E. Bevan, Tom D. Cecil
10 Robotic total mesorectal excision 154
M. Chadwick, H.S. Tilney, A.M. Gudgeon
11 Local excision and transanal endoscopic microsurgery 171
Wolfgang B. Gaertner, David A. Rothenberger
12 Pathology assessment 191
Philip Quirke, Tim Palmer, Gordon G.A. Hutchins, Nick P. West
13 Assessment and management of recurrence 203
Peter J. Lee, Kirk K.S. Austin, Michael J. Solomon
14 Lateral pelvic side-wall nodal involvement in rectal cancer 222
Hideaki Yano, Brendan Moran
15 Intestinal stoma and the role of defunctioning a low anastomosis after
anterior resection 228
David Mitchell, Kandiah Chandrakumaran, Steven Arnold
16 Quality of life in patients undergoing abdominoperineal excision and anterior
resection for rectal cancer 239
Peter How, Kandiah Chandrakumaran

Index 249

HEBK001-FM.indd 5 11/02/13 6:40 PM


HEBK001-FM.indd 6 11/02/13 6:40 PM
Contributors

Steven Arnold Mark Harrison


Consultant Surgeon, Department of Surgery, Consultant Clinical Oncologist, Mount Vernon
Basingstoke and North Hampshire Foundation Cancer Centre, Northwood, UK
Trust, Basingstoke, UK R.J. (‘Bill’) Heald
Kirk K.S. Austin Director of Surgery, Pelican Cancer Foundation,
Department of Colorectal Surgery and Surgical Basingstoke, UK
Outcome Research Centre (SOuRCe), Royal Prince Torbjörn Holm
Alfred Hospital, Sydney, Australia Associate Professor of Surgery, Section
Katharine E. Bevan of Coloproctology, Department of Surgical
Basingstoke and North Hampshire Hospital Gastroenterology, Karolinska University Hospital,
Foundation Trust, Basingstoke, UK Stockholm, Sweden
Gina Brown Peter How
Consultant Radiologist and Honorary Senior Colorectal Research Registrar Pelican Centre,
Lecturer, Royal Marsden NHS Foundation Trust, Basingstoke & Croydon University Hospital
Sutton, UK Chris Hunter
Tom D. Cecil Colorectal Surgeon, Royal Marsden NHS
Consultant Surgeon, Department of Surgery, Foundation Trust and Croydon University Hospital,
Basingstoke and North Hampshire Hospital Surrey, UK
Foundation Trust, Basingstoke, UK Gordon G.A. Hutchins
Michael Chadwick Department of Pathology and Tumour Biology, Leeds
Consultant Laparoscopic Colorectal Surgeon, Institute of Molecular Medicine, University of Leeds;
St Helens & Knowsley Teaching Hospitals, and Leeds Teaching Hospitals Trust, Leeds, UK
NHS Trust, Merseyside, UK
Peter J. Lee
Kandiah Chandrakumaran Department of Colorectal Surgery and Surgical
Associate Specialist, Department of Colorectal/ Outcome Research Centre (SOuRCe), Royal Prince
Pseudomyxoma Surgery, Basingstoke and North Alfred Hospital, Sydney, Australia
Hampshire Hospital Foundation Trust, Basingstoke, UK
David Mitchell
Wolfgang B. Gaertner Colorectal Fellow, Department of Surgery,
Division of Colon and Rectal Surgery, Department of Basingstoke and North Hampshire Foundation
Surgery, University of Minnesota, Minneapolis, MN, USA Trust, Basingstoke, UK
Rob Glynne-Jones Brendan Moran
Consultant Clinical Oncologist and Macmillan Lead Consultant Surgeon, Department of Surgery,
Clinician in Gastro-Intestinal Cancer, Mount Vernon Basingstoke and North Hampshire Foundation
Cancer Centre, Northwood, UK Trust, Basingstoke, UK
Mark Gudgeon Tim Palmer
Department of Colorectal Surgery, Department of Pathology and Tumour Biology, Leeds
Frimley Park Hospital, NHS Foundation Trust, Institute of Molecular Medicine, University of Leeds;
Frimley, UK and Leeds Teaching Hospitals Trust, Leeds, UK

HEBK001-FM.indd 7 11/02/13 6:40 PM


Philip Quirke Arcot K. Venkatasubramaniam
Department of Pathology and Tumour Biology, Consultant Colorectal Surgery, Basingstoke and
Leeds Institute of Molecular Medicine, University of North Hampshire Foundation Trust,
Leeds, Leeds, UK Basingstoke, UK
David A. Rothenberger Thilo Wedel
Division of Colon and Rectal Surgery, Department Head of the Center of Clinical Anatomy, Institute of
of Surgery, University of Minnesota, Minneapolis, Anatomy, Christian Albrechts University of Kiel, Kiel,
MN, USA Germany
Oliver Shihab Nick P. West
Basingstoke and North Hampshire Trust, Department of Pathology and Tumour Biology,
Basingstoke, UK Leeds Institute of Molecular Medicine, University of
Leeds; and Leeds Teaching Hospitals Trust,
Michael J. Solomon
Leeds, UK
Department of Colorectal Surgery and Surgical
Outcome Research Centre (SOuRCe), Royal Prince Hideaki Yano
Alfred Hospital and Discipline of Surgery, University Consultant Surgeon, Department of Surgery,
of Sydney, Sydney, Australia National Centre for Global Health and Medicine,
H.S. Tilney Tokyo, Japan
Consultant Colorectal Surgeon, Frimley Park
Hospital, NHS Foundation Trust, Surrey, UK

HEBK001-FM.indd 8 11/02/13 6:40 PM


1
The evolution of a concept: the total
mesorectal excision story (for surgeon
or patient alike)
R.J. (‘Bill’) Heald

Introduction All carcinomas of the lower sigmoid and upper


rectum are tabooed by all practical surgeons on
One German professor, a great friend, suggested that account of their anatomical inaccessibility. All are
I should write the story of how it became possible to abandoned without hope to linger on for a few
conquer the whole world with an idea–a concept– months until death relieves them of their
a guiding principal of surgical technique–to tell in loathsome condition.
fact how the words Total Mesorectal Excision (TME)
moved from humble beginnings in Basingstoke to On this background, all who tackled rectal cancer
become a part of any discussion about rectal cancer surgery had to contend with many failures and ter-
anywhere in the world (Figure 1.1). rible outcomes in the patients who developed local
A personal reflection on the key components of recurrence.
the TME story over a period of 30 years may elu- The TME story starts with its roots in embryol-
cidate some of the issues and encourage others to ogy and its basis in anatomy.
follow an idea born out of clinical observation at
the surgical coalface.
In the late 1970s, when I was a young consult- Embryology
ant, almost half of all patients with rectal cancer
died from local recurrence – that is, regrowth of the Embryology was always a challenge for me as a stu-
cancer within the pelvis. The pelvis became gradu- dent to grasp the mysteries of one cell turning itself
ally permeated by malignant tumour, which infil- into a fetus with a near certainty of becoming a human
trated all pelvic contents, particularly pelvic side-wall being. One special mystery was that ‘tube’ from mouth
nerves and the sacral nerve roots to the legs, eventu- to anus that was somehow programmed to sprout the
ally causing intractable pain, paralysis and inconti- potential liver in one direction and the pancreas in
nence – all adding up to one of the most miserable another, and then, before the fetus is 1 cm long, to take
forms of death ever dreamed up, and all terribly slow. a journey out of the primitive abdominal cavity and
Unsurprisingly, almost 100 years before this, H.W. subsequently back into it, thus setting the components
Maunsell stated in the Lancet in 1892: of the gastrointestinal tract in their appointed order

HEBK001-C01_p01-30.indd 1 11/02/13 3:43 PM


2  The evolution of a concept: the TME story

Figure 1.1. Worldwide TME. Some of the TME venues, comprising more than 450 invited operations in more than
40 countries. At the time of going to press the Queen’s Birthday Honours List presented the CBE to Professor Richard
Heald for services to UK surgery internationally.

and placement and establishing the marvel of the The growing surgeon: ‘on the
human alimentary system in situ. My personal story is shoulders of giants’
of gradual realization that this midline gut tube can be
redefined with its intrinsic ­lymphovascular ­surround These seeds of wonder at embryology lay dormant in
as a midline envelope recognizable for surgeons by me as surgery became practice. Small flashes of wis-
the spider’s web of areolar tissue around it. The dis- dom from along the path of surgical training blinked
tal part of the envelope is around the rectum and as possible explanations for good or bad outcomes:
becomes the mesorectum, and the cobweb around it ‘The best surgeon is the one with a capacity for tak-
where the surgeon dissects is the ‘holy plane’. Each part ing infinite pains’ is one such pearl that continues to
of the envelope has an artery coming from the front flash up with great regularity. Pains to do what exactly?
of the aorta, rather than the side like everything else. Advocating TME eventually became a battle to justify
The key hypothesis was that each part of the envelope, turning a 1 h operation into an increasingly fastidious
if very carefully removed en bloc, might have a very and precise operation taking 3 h or more. Initially the
good chance of enveloping the whole primary field of only obvious justification for such a change was the
spread of a cancer and thus curing all but the most hope of fewer permanent colostomies. The potential
advanced cases (Figure 1.2). for improved outcome, survival and cure would not
The TME story is inextricably linked with embryol- become apparent for 5 years or more and was never
ogy, and it is probable that similar concepts might be dreamed of at the beginning.
applicable to many other cancers, since the constraints It is perhaps a measure of the fundamental ethics
to cancer growth, such as areolar planes that block of the early British National Health Service (NHS)
angiogenesis, are relevant to malignant infiltration of of the 1970s and 1980s that no financial or manage-
many kinds and in many places ­(Figure 1.3). ment pressure was ever exerted on me to desist from

HEBK001-C01_p01-30.indd 2 11/02/13 3:43 PM


Basingstoke, Hampshire  3

Figure 1.2a. The mesorectum surrounded by the holy


plane. Figure 1.3. Pursuable cobweb areolar plane.

such an expensive experiment. Such benign accept-


ance of the time-consuming emerging concept did
not, however, imply that I was not berated and con-
demned by the surgical establishment for the temer-
ity to challenge their supremacy – as outlined later.
One early public condemnation, from Prof Mike
Perry, still rings in my ears today: ‘you are taking on
a grave responsibility by deviating from the radical-
ity of the established orthodoxy of abdominoperi-
neal excision’.

Basingstoke, Hampshire

With an abiding interest in visceral surgery, the early


1970s finds me a consultant surgeon in the little-known
town of Basingstoke in the pleasant countrified county
of Hampshire, about an hour south of London, with
a catchment population of approximately 250 000. I
was the second full-time consultant general surgeon in
the large new hospital under construction. My senior
colleague was the remarkable Mr Frank Tovey, whose
voluntary service in India and China, combined with
his passion for research and teaching, were all truly ser-
endipitous for me. I still remember his standing at my
side in the operating room as I tediously tried to achieve
an early TME. I was embarrassed at my ineptitude, but
he said ‘I think you might have something there, Bill. It
is very different from what everyone else is doing and is
definitely worth persevering with. I will give up doing
rectums and let you have them all.’ This was a key start-
ing point, as becoming the only rectal cancer surgeon in
a District Hospital was then a unique privilege. Speciali-
Figure 1.2b. A perfect TME (PO Nystrom). zation was commencing naturally by mutual consent.

HEBK001-C01_p01-30.indd 3 11/02/13 3:43 PM


4  The evolution of a concept: the TME story

Dividing the mesorectum


1975 (not called that then ....)
No such word in GRAY’s
“ANATOMY”

Figure 1.4. The node contains cancer!

Anterior resection, as I had been taught, involved of the TME story, the seminal moment occurred in
manual extraction of the rectum with much rough tear- the operating room. We were dealing with the all too
ing of the surrounding fat, sometimes ­uncontrollable common random bleeding in the fat behind the rec-
blood loss, and desperate anxiety about leakage from tum when we noticed a small lymph node less than
the anastomosis and later of local recurrence. The latter 1 cm in size. It looked harmless enough, but I lifted it
was assumed to be largely in the hands of the almighty out and asked the scrub nurse to send it for patho-
and pretty much inevitable. It was frequently stated by logical analysis. A week later the node was reported
the great and the good that ‘surgery had gone as far as as containing a focus of cancer (Figure 1.4). It thus
it could go’. More than 75 per cent of rectal cancers, became apparent that I had cut or torn through the
and every cancer that could be felt digitally, was offered field of spread of Anthea’s cancer. What to do? A
only abdominoperineal excision (APE) with perma- week later we performed an APE to be properly ‘radi-
nent colostomy, which was considered much the most cal’, but no further cancer was found in the specimen.
‘radical’ operation and much the safest oncologically. Five years later Anthea died of rectal cancer bony
Anterior resection was believed to add substantially to metastases with a quadriplegia – a memorable and
the risk of local recurrence. terrible end to a historic individual in the story of
Cancer recurrence at the suture line dominated rectal cancer. It still seems to me that cutting through
our ideas of follow-up after anterior resection, and cancer, or its field of spread, almost always leads to a
this dictated a digital examination and sigmoidos- cancer death, unless possibly on occasion when the
copy. Ultrasound, computed tomography (CT) and tissues have been recently irradiated.
magnetic resonance imaging (MRI) did not exist. The concept of total excision of the mesorectum
was developing. The idea seemed strengthened by
the referral of a suture line recurrence later that year
The mesorectum in rectal when the emptying of the pelvis for an ultra-low
cancer: the clue to local anastomosis revealed a large mesorectal remnant
recurrence? with a cancer focus growing from it into the suture
line (Figure 1.5).
Against this background, clinical experiences and The other three cases pointed in the same direc-
histological findings with five particular patients sug- tion. The paper was published in the British Journal
gested that mesorectal residues might be the source of of Surgery entitled ‘The mesorectum in rectal cancer:
those very common pelvic recurrences.1 This ‘tale of the clue to local recurrence?’1 This key publication
five patients’ has, I am told, become the most cited ref- in the rectal cancer literature was simply an idea
erence in the whole surgical literature on bowel cancer. based on clinical observation. One of my much
The most memorable, and perhaps most signifi- loved former chiefs, Mr Peter Philip, wrote me only
cant, of the five thought provoking patients in this one letter; it said ‘That was the most sensible paper I
first paper was a publisher called Anthea. Like most have read for a long time.’

HEBK001-C01_p01-30.indd 4 11/02/13 3:43 PM


MESORECTUM – THE WORD  5

1977

Re-Operation for Suture Line Recurrence 1977


Figure 1.5. Suture line recurrence.

The ‘Russian gun’ Russian gun at about 3 cm was the lowest I had ever
attempted, and the circumferential clearance was a
The ‘Russian gun’ saga preceded TME and landed long slow attempt to circumnavigate the cancer by
me in trouble before the 1970s were out, and it dissecting in clean surgical planes. Anne thrived for
certainly contributed to my preoccupation with 3 years and then sadly developed liver secondaries,
TME. The legendary figure of John Goligher, for which I performed a hemi-hepatectomy. At this
then Professor of Surgery in Leeds, showed me time she had found a new partner. Three years later
the circular stapling gun from Russia, which he she was watching television when she fell uncon-
had bought from a bazaar in Turkey for a few scious from a cerebral secondary and did not wake
stray coins. ‘Take an interest in this, young man: up – merciful but very, very sad. She never had pelvic
it may well have a place in low anterior resection.’ recurrence, so the seeds of hope for lower anterior
So I bought one on my travels, and, to my eternal resections were sewn, and my first TME seemed to
shame, it sat unused in a cupboard for a year until me at least to have been justified with local disease
my encounter with Anne, a young woman with control.
a large ulcerated carcinoma 5 cm from the anal
verge. For a young woman, recently divorced and
in her early twenties, the thought of a permanent Mesorectum – the word
stoma was unbearable.
At this time, the TME story in my own mind was I had heard the word ‘mesorectum’, although it was
at the evolution point where the challenge of meet- not in Gray’s Anatomy or anywhere else that I ever
ing this patient’s hopes was just what was needed found. My mentor and trainer at Guy’s, Mr Rex
to push the TME idea to its logical conclusion. Per- Lawrie, used the word ‘mesorectum’ to describe
haps even such a low cancer, at 5 cm, could be safely the fat behind the rectum that we divided during
removed with all its relevant field of local spread conventional anterior resection. This fat often bled
without sacrificing the pelvic floor and the sphinc- in a tiresome way and so was the object of irritated
ter muscles. The mesorectum as a surgical craft abuse on occasions. We have since seen a report of
entity was being born – a dream of relevant radical- the word ‘mesorectum’ in an ancient radiotherapy
ity without unnecessary mutilation.2 account of the use of radiotherapy in rectal cancer
For Anne, I deemed it possible to achieve sphincter in 1914, but, for practical purposes, the word was
preservation, which, though extremely unorthodox, undiscoverable at the time. It is attributed in my
might just be achievable with that Russian gun. Tor- mind to one of my personal giants, Rex Lawrie, on
tured by the risk of local recurrence, I attempted my whose shoulders much of my surgical thinking had
first TME in 1978. For me, the anastomosis with the certainly been based.

HEBK001-C01_p01-30.indd 5 11/02/13 3:43 PM


6  The evolution of a concept: the TME story

Figure 1.6. Anatomist’s


mesentery.

Words Matter suspending a segment of gut may count as a mesen-


tery and that the term ‘mesentery’ is not applicable
In his book Talking Sense,2 Richard Asher points out to the rectum, since the normal rectum is devoid of
that ‘physicians indulge in conjectural names’ and an encircling, suspensory mesothelial sleeve (Figure
cause a great deal of confusion as a result. He quotes 1.6).3 I replied:
from Socrates: ‘He who first gave names and gave
them according to his conception of the things they To the oncological surgeon the term
signified, if his conception was erroneous, shall we ‘mesentery’ implies an integral visceral,
not be deceived by him?’ lymphovascular and fatty entity that nourishes
I believe that both Socrates and Richard Asher would the organ concerned, and is developed
be happy with the phrase ‘total mesorectal excision’ as embryologically in concert with it. In the case
it describes exactly what the surgeon should try to do. of the mid-rectum, this fatty lymphovascular
It is not easy to do well, but, very fortunately, the sur- structure surrounds the rectum completely, and
geon who believes in the TME concept and attempts to no other word but ‘mesorectum’ seems readily
do TME is hugely rewarded by improved results. available to describe it. Appreciation of the
One enduring problem is that the word ‘mesorec- surgical anatomy of this entity, and exercising
tum’ was, and remains, anathema to many anatomists. particular care when dissecting around it,
Even the current edition of Gray’s Anatomy describes are important components of good surgical
the mesorectum as the mesentery of the sigmoid technique in relation to rectal cancer. Graham
colon. Anatomists describe a ‘meso’ (e.g. mesocolon) Hill’s term ‘extrafascial excision of rectum’ may
as simply two layers of peritoneum with blood vessels be more acceptable to morphologists – but
and lymphatics between them and therefore question is unlikely to displace the term TME from the
the use of the term ‘mesorectum’ (Figure 1.5). surgical nomenclature …’
For me, however, and fortunately for the future
of the concept of TME, this was the point where and ‘his’ fascia is indeed that which surrounds ‘our’
oncology and embryology met, and the choice of mesorectum.
the word became descriptively critical and entirely
acceptable to surgeons who were trying to define
the primary field of spread of a rectal cancer. Thus, The initial impact
the mid-rectum is entirely encompassed circumfer-
entially by its own fat, lymphatics and vascular sup- Early experiences of applying the circular stapling
ply, all enveloped in a fascial covering, with a recog- gun resulted in lower and lower anterior ­resections.4
nizable practical surgical plane outside it. This stimulated interest in the guns by other sur-
Anatomist Professor Morgado proposed that only geons, which led to my first travelling phase, large-
a two-layered mesothelial structure surrounding and ly limited to England. As a young surgeon, I was

HEBK001-C01_p01-30.indd 6 11/02/13 3:43 PM


Data collection and analysis and the pivotal role of Rosemary Sexton  7

Figure 1.7. My first travelling


phase.

instructed to turn up with ‘the gun’ after the cancer Data collection and analysis
had been removed by the host. and the pivotal role of
The outcome of such a visit to St Mark’s Hos- Rosemary Sexton
pital in London to demonstrate the ‘new toy’
is nicely illustrated by my friend Mick Elliot Evidence was appearing of wide variations in the local
(Figures 1.7–1.9). The Russian gun required that recurrence and survival rates between surgeons, and
the staples be individually loaded in addition to Hermanek suggested that TME might be at the heart of
the washer on to which the circular knife cut after this. The maverick practices of a surgeon in Basingstoke
firing of the staples. Undoubtedly the fastidious were unlikely ever to change the practice of surgery in a
Professor Goligher measured each washer’s size fundamental way. Only the most unchallengeable data
and thickness, but I was content for others to do with every possible discrepancy covered were likely to
this, and I assumed the washers were uniform. be of any value if what we were discovering was to turn
What happened at St Mark’s was that the so-called out to be of any importance. Data collection and man-
‘doughnuts’ were not cut through, although the agement were initiated by Rosemary Sexton, formerly
staples had fired. The washer was too thin and my secretary, whose ­husband David worked for Xerox
the gun could not be removed. Perhaps the Bas- and who gave us an enormous computer for this pur-
ingstoke Gazette (Figure 1.10) was right! We all pose. We were ably backed by my cousin Pam who
rejoiced when the US company Autosuture started helped us to create our own dedicated software to cor-
to manufacture disposable, more reliable stapling relate preoperative and operative detail with outcomes
instruments. in a unique way. Thus, my initial data collections were

Figure 1.8. Staples fired, doughnut


not released.

HEBK001-C01_p01-30.indd 7 11/02/13 3:43 PM


8  The evolution of a concept: the TME story

Figure 1.9. Goodbye, Bill!

comprehensive and consecutive so that there were truly of his time, with whom I did a joint clinic. Dr
no exclusions. The quality of the data was enormously Ryall’s view of the role of radiotherapy in rectal
enhanced by the fact that Rosemary made it her busi- cancer was indeed ahead of his contemporaries.
ness to get to know every patient. Of course we made Throughout the USA, and in much of the UK,
some mistakes, and the databases required continuing radiotherapy was being given to many patients
alterations; for example, initially we failed to record postoperatively, which he considered to be infe-
the quadrant of the rectum where the cancer was situ- rior to radical radiation given before surgery. We
ated. Rosemary attended the outpatient clinics and was selected for the latter only patients considered to
truly the forerunner of what is often now called the ‘key be locally inoperable as judged by palpable fixity
worker’ in the UK or ‘case manager’ in Europe as part to adjacent pelvic organs.
of the modern multidisciplinary team management of The other members of our embryo multidis-
cancer. She provided a communication lifeline in the ciplinary team were Rosemary Sexton and stoma
sometimes complex and terrifying journey that follows therapist Sister Anne Leppington-Clarke, who pio-
a diagnosis of rectal cancer. neered irrigation in southern England as a meth-
od of managing a permanent end ­colostomy. She
also worked out ­beautifully how to cope with the
The embryonic tiresome temporary right transverse colostomy
multidisciplinary team that we generally performed to protect a low anas-
and Roger Ryall tomosis for the first 6 weeks. Transverse colostomy
remains my preference to reduce the formation of
The unsung hero of the TME story is Dr Roger small bowel adhesion and abolish the high output
Ryall, a clinical oncologist with an intellect ahead sometimes associated with loop ileostomy, which
enjoys greater popularity at this time.

Early publications

One key early publication emanated from my presi-


dential address to the section of surgery of the ­Royal
Society of Medicine. Two years earlier Mr Geoff
Oates’ presidential address had included the words
‘I don’t know why you don’t believe Bill Heald – he
is a very truthful chap’ – life-saving for me sitting
in the audience at the time. In my own address I
Figure 1.10. From the Basingstoke Gazette, 1979. advanced my preoccupation with the embryology

HEBK001-C01_p01-30.indd 8 11/02/13 3:43 PM


Early publications  9

and ­anatomy to explain the astonishing local recur- one of the medical students who understood log
rence figures that were beginning to earn me so ranking and Kaplan–Meier curves. The local recur-
much derision. Unexpectedly good outcomes led rence of 3.7 per cent was finally calculated based
me to choose as my title ‘The holy plane of rectal on these 115 patients. It is intriguing that, when the
surgery’5 and to suggest that embryologically deter- numbers had quadrupled, the original Kaplan–Mei-
mined planes might be a sounder basis for optimal er prediction was within 0.2 per cent of this initial
oncological specimens and optimal outcomes than calculation, and indeed with gradual narrowing of
random sacrifice of the anus. I suggested that there the confidence intervals over the years very little has
were widespread misunderstandings about the word changed from those original predictions. When a
‘radical’. Abdominoperineal excision was much more local recurrence rate of 3.7 per cent in ‘curative’ 115
radical and therefore much more ‘curative’ than an cases compared so favourably3 with the then accept-
anterior resection, but this was in complete contrast ed figures of 20–40 per cent in various publications,
to the surprising data that we were accumulating. it is perhaps unsurprising that the original data were
Total mesorectal excision, I suggested, was ‘relevant greeted with some scepticism.
precise radicality replacing unnecessary mutilation’. A pointer to the future comes from the following
quote in the discussion section of this Lancet paper:
‘On this evidence it is often safe to limit mural
Publication by a Surgeon and clearance and thus preserve the sphincters provided
Radiotherapist About Patients Not the mesorectum is excised intact with the cancer.’6
Given Radiotherapy
Our little multidisciplinary team published the More than 100 ‘Away Cases’
115 cases in the Lancet with just Heald and Ryall in Sweden and Norway
as the authors with the rather straightforward title
‘Recurrence and survival after total mesorectal exci- By the late 1980s the literature suggested that a Bas-
sion for rectal cancer’.6 ingstoke surgeon could achieve local recurrence rate
This paper, with outcomes at 2–6 years, was based of less than 5 per cent with anterior resection. Many
on only 115 curative cases (i.e. patients without felt that either he was lying or he had redefined a
metastases). I recall quite primitive attempts to work ‘curative’ resection in some way to ­optimize ­outcomes
out confidence intervals myself, eventually aided by reported. The Scandinavians, particularly in ­Norway

Figure 1.11. Using the smaller camera.

HEBK001-C01_p01-30.indd 9 11/02/13 3:43 PM


10  The evolution of a concept: the TME story

Figure 1.12. Professor Pahlman's depiction


of the effects of TME workshops across
Sweden.

and Sweden, had identified the issues of local recur- Professor Bjorn Cedemark and the then young
rence in rectal cancer and, after evaluation, had decid- Torbjörn Holm. They and their colleagues coordinat-
ed that the addition of radiotherapy could result in ed what we often refer to as ‘Stockholm 3’, a succession
slight improvements but at much risk to the patients to the famous Stockholm 1 and 2 radiotherapy trials.
and cost to the health-care system. Professor Soreide Publication of the impact of a series of workshops in
of Oslo invited me as a visiting surgeon to his video- the mid-1990s at the Karolinska Hospital, Stockholm
based ‘super radical cancer surgery’ annual workshops was the first report to demonstrate that workshop
at the Rikshospital. Here in 1 week, assisted by a suc- training in surgical technique can improve cancer
cession of Norwegian surgeons, I performed ten tel- outcomes in a whole population. Ongoing education-
evised TME procedures while trying hard to dodge al activities that were thus started have continued to
around an enormous broadcast-quality camera. This make a real difference throughout Sweden.
news-gathering television camera had been given to Looking back, it is clear that any world conquest by
me by Sony Broadcast, serendipitously a Basingstoke- the TME story occurred very gradually as surgeons
based company (Figure 1.11). grasped the unexpected improvements in outcome
The Swedish approach differed from Norway’s in that could be achieved by the expenditure of time and
that three surgeons from the central Ostergotland painstaking effort in pursuit of the TME principles.
district asked if they might come to Basingstoke The first TME after our first workshop in Perth,
for a week. After three cases, and intensive study of Western Australia makes it very clear that a ‘better’
video clips from my Sony recording kit, Erik Nillsen specimen with intact margins will be much less lia-
and his colleagues returned to Motala and Norrko- ble to spill cells or leave cancer-containing residues. I
ping, clearly convinced of the potential of this tedi- labelled the goal as ‘specimen-oriented surgery’, where-
ous ‘TME surgery’. They grasped that only one sur- by the surgeon’s mind is forever prioritizing the quality
geon in each Swedish district could possibly hope to of the specimen as he or she dissects around it. Naked-
hone the necessary skills and understanding of the eye inspection and a recorded evaluation of the shape,
complexities of the deep pelvic anatomy. contours and perfection of the specimen thus became
The Swedish TME ‘wave’ (Figure 1.12) was carried the key tool in the audit of the quality of the surgery.
forward by Professor Rune Sjodahl, who organized
a series of workshops in Linkoping. This was fol-
lowed by the first of several honorary professorships, Histopathology workshops:
many lasting friendships and ultimately over 80 visits Phil Quirke
across Sweden.
Subsequently, Stockholm became especially impor- As the Scandinavian workshops spread to other
tant, with special friendships established through countries, histopathological evaluation of the

HEBK001-C01_p01-30.indd 10 11/02/13 3:43 PM


A new form of surgical teaching  11

margins of excised TME specimens became part A new form of surgical


of the TME ­crusade in a multidisciplinary fashion. teaching: the live surgery
What had been the ‘Travelling Bill Show’ became, video workshop and the
with Phil Quirke from Leeds, the ‘Travelling Bill birth of nerve recognition
and Phil Show’, and the workshops increasingly and preservation
embraced not only video relay of the live surgery
but also workshops for pathologists with detailed The special problem that has beset the TME story
audit of the margins and the quality of the speci- from the beginning is that the operation is difficult
mens. Naked-eye examination and evaluation to perform, although the concept of ‘embryological
of the perfection of the specimen are potentially envelopes’ is easy enough to grasp in theory. Since
humiliating for many surgeons but a key com- good-quality data were accumulating through the
ponent of introducing objectivity and honesty to late 1980s and the Scandinavian enthusiasm gath-
improve outcomes. ered momentum, the demand for people to see
exactly what was involved grew steadily. Invitations
flooded in from around the world so that the next
Television, BBC2 and 25 years have become for me a steady stream of ‘away
Phil Hammond cases’, armed with a video camera and backed up by
the ever willing Phil Quirke to bring on the histopa-
Without doubt, the most exciting television appear- thology audit, which was the essence of convincing
ance of the TME story occurred on BBC2, when a surgeons of the need to be ‘specimen-oriented’.
series entitled Trust Me I’m a Doctor was conducted Phil’s commitment has become legendary and
by a dynamic doctor and investigatory reporter he has criss-crossed the world with the salutary
called Phil Hammond. His colourful coverage of message that the histopathologist should not only
the ‘Phil and Bill Show’ embraced the experiences report on a surgeon’s work but also audit the qual-
of one young and engaging patient, a natural ity with ruthless tenacity (Figure 1.13), which is apt
television personality who emphasized, straight to strike fear into the surgical heart.
into the camera, ‘When it’s your life, you’ve only got Total mesorectal excision video workshops have
one chance.’ I had often made a similar observation introduced surgeons across the world to the true detail
in relation to rectal cancer that ‘what is omitted at of deep pelvic anatomy. In the late 1980s and 1990s,
the first operation is lost forever’. the demonstration of the seminal vesicles during a
Phil Hammond interviewed Bjorn Cedermark, workshop evoked amazement while video images of
who stated on camera: dissection between them and ­Denonvilliers’ fascia
were hailed a first-time experience. Neither cadav-
[Abdominoperineal resections] and permanent
er dissection nor CT scanning had ever conveyed
colostomies were 50 per cent down to 20
an understanding of Denonvilliers’ fascia, a trapezoi-
per cent, the local recurrence from 22 per cent
dal bib or sheet of collagen extending down from the
down to 5 per cent, and what happened during
apex of the peritoneal reflection between the rectal
1994–1995 was simply the workshops. The
envelope behind and genitourinary organs in front.
important thing about this is that what we didn’t
Denonvilliers described in 1837, in his doctoral the-
really know, we read Bill Heald’s paper for the
sis, ‘l’aponévrose pubio-rectale’. Understanding this
first time and Bill knows as well as everyone else
medially tapering trapezoidal sheet is truly the key to a
that a lot of people did not believe him. They
proper TME in a low anterior cancer in male patients
said this is not right, we have all been doing this
and also the key to preserving potency in all TME
for years, it cannot be true – well it is true! And
dissections in male patients (Figure 1.14). The anteri-
I think we have proven that we can really do
or mesorectal fat, whose very existence was previously
something by changing our surgical habits also
ridiculed by the top surgical establishment, is very real
for a population.
but easy to tear, so we have always advocated dissec-
Stockholm was perhaps the single most signifi- tion anterior to Denonvilliers’ septum (Figure 1.15).
cant episode in the establishment of an extraordi- The emergence and widespread use of axial MRI
nary lifelong series of practical video workshops. finally established in everyone’s mind that the anterior

HEBK001-C01_p01-30.indd 11 11/02/13 3:43 PM


12  The evolution of a concept: the TME story

Before

After

Figure 1.13. Histopathology


audit.

mesorectum did indeed exist and that the septum was cinema operator, found myself with a massive amount
often recognizable on a good-quality image. of broadcast-quality video editing material capable of
handling videotape non-linear editing. In the 1980s
one whole room at my house was taken over by the
Video surgery equipment necessary, and the cost in today’s terms
would have been well over £250 000, provided with-
Many of the initial video workshops were in Basing- out charge by Sony. Thus, we were able to record on
stoke, and it soon became apparent that the quality of professional video format hundreds of operations per-
standard videotapes and video cameras was inadequate formed in a variety of different countries, all in broad-
to convey the detailed images required. Serendipity cast quality and capable of conveying to a watching
intervened in that the advanced products division of surgeon the essence of holy plane dissection and the
Sony Broadcast was situated in Basingstoke, only a few beginning of the identification of the layer of nerves
hundred metres from the operating theatres. Here the that surround the mesorectal envelope.
first commercially aspiring high-definition television Throughout the development of the video tech-
was being developed, and the education department nology necessary to identify the autonomic nerves
in Sony Broadcast became friendly and supportive. of the pelvis, the improvement in picture quality
As a result I, who had started life in my teens as a and the recording and transmission of detail have

HEBK001-C01_p01-30.indd 12 11/02/13 3:43 PM


The identification of the nerves of urogenital function  13

Figure 1.14. Denonvilliers’


septum.

been crucial. Further serendipity provided the TME erection, ejaculation, and control and normality of
story with Jason Flowers, a man of unique talent in urinary and faecal voiding and continence.
the field of computers and computer visualization One notable exception was Sir William Slack,
of images. It is with his help that we have led the a distinguished London surgeon at the Middle-
world in visualizing the fine detail, including the sex Hospital. He had, with his histopathologists,
autonomic nerves and plexuses. looked carefully at the periphery of rectal cancer
specimens and correlated patient impotence with
the nerves inadvertently removed by the surgeon.
I was not innocent of this myself, and I still show
The identification of the an earlier teaching video in which I cut through
nerves of urogenital the superior hypogastric plexus by getting into the
function plane outside it at the aortic bifurcation and then
back into the correct plane by cutting through the
‘Man recognizes only what he knows.’ (Goethe)
nerves on which ejaculation and to some extent the
In the early days some of the most distinguished control of urinary function depend (Figure 1.16).
surgeons totally rejected the idea that we were
seeing the autonomic nerves responsible for male

Figure 1.15. Dissection anterior to Denonvilliers’ septum. Figure 1.16. Right erigent pillar.

HEBK001-C01_p01-30.indd 13 11/02/13 3:44 PM


14  The evolution of a concept: the TME story

Most surgeons had been unaware of these struc- The emergence of magnetic
tures, and I was still failing to grasp fully the onion- resonance imaging in a
like anatomy that must be respected if these nerves Swedish workshop
are to be identified, recognized and preserved. The
idea of the onion came from a Japanese visiting sur- Initial Swedish workshops at the Karolinska Hos-
geon, Professor Yoshi Moriya. The onion is a help- pital involved two adjacent operating theatres and
ful anatomical concept, with the mesorectum as the transmitted images plus continuous review of MRI
core, the autonomic nerve layer as the first layer, the scans with Dr Lennart Blomqvist, their expert
Wolffian ridge ureters and vesicles as the next layer, futuristic radiologist. Groups of up to 20 Swedish
and so on. Holy plane dissection is thus the very surgeons attended, taking it in turns to assist with
careful entry into, and pursuit of, the innermost the operation. The informal relationship between
proper surgical plane around the core outside the small groups of surgeons and personal interaction
mesorectal fascia, while recognizing and preserving has been a unique part of what is undoubtedly a
the nerve layer outside it. completely new form of surgical teaching. The
The detail of the autonomic nerves was the next sense of involvement mixed with informality adds
frontier and the next major controversy. Nerve considerably to the learning experience. Similar
preservation is one of the most difficult challenges arrangements now work extremely well in my regu-
of a good TME. Mesorectal fascial dissection liber- lar Heidelberg workshop series.
ates the inferior hypogastric plexus in an area that
was formerly disastrously and improperly named
the ‘lateral ligaments’. Video teaching dissections Who needs enemies?
made it clear that no actual ligament existed, only
areas of adherence. Simultaneously Japanese anat- Professor Paul Hermanek, a great friend and world-
omists Sato and Sato had also noted that middle renowned pathologist, once told me over breakfast
rectal arteries coming from the internal iliac vessels that the TME project would progress much faster if
were uncommon, being present in only one in five I didn’t have so many people, many from England
patients on one side or the other and bilaterally in and beyond, belittling it. But to every force there is
one in ten patients. The routine of clamping and a counterforce, and such antagonism often breeds
dividing something called a ‘lateral ligament’ that silent supporters from the quiet majority, and
also appeared to contain major arteries and veins opponents often do more benefit than harm.
(traditionally labelled the middle rectal vessels) had One curious, almost dramatic example of this was
to be challenged. the assault by Professor Isbister, at the time a profes-
My own practical experience was, and is, that sub- sor of surgery in Riyadh. His antagonism appeared to
stantial vessels at this level are rare. If the surgeon take the form of an attack on Basingstoke, the inno-
dissects carefully between the mesorectal fat and the cent town from which the TME idea had emanated.
inferior hypogastric plexus, only a variable few nerves Various articles appeared under the titles of ‘Basing-
entering the mesorectum and some minor vessels are stoke visited’, ‘Basingstoke revisited’ and ‘Basingstoke
encountered. These observations suggest that the ‘lat- visited again’. The essence of the articles was that the
eral ligaments’ are no more than areas of adherence data were wrong and meaningless. Since he had never
between inferior hypogastric plexus and mesorectum visited Basingstoke, my replies constantly reiterated a
that tether the specimen inferolaterally. The vessels warm invitation to him to do so, to see what we were
that we used to clamp were the lateral intramesorectal trying to teach, and to give us his comments on our
branches of the superior rectals and a part of the lym- work, favourable or unfavourable. He never came.
phovascular field that should have been encompassed Isbister’s criticisms demanded robust replies
and removed. This stage of the dissection is very chal- and undoubtedly the ‘noise’ helped enormously
lenging and is more achievable with precise monopo- in spreading TME around the globe. Where there
lar diathermy. Diathermy here, however, does bring is no argument there is often little interest. One of
into question the matter of collateral nerve damage, the more colourful condemnations that I earned for
and so low settings and optimal methodology are my impudence was from the then president of the
essential areas for ongoing technical research. South African Surgical Association, who instructed

HEBK001-C01_p01-30.indd 14 11/02/13 3:44 PM


The low colorectal and coloanal anastomosis  15

his members to take no notice of ‘that fellow Heald, the extra benefits of radiotherapy and chemo-
he is a flamboyant charlatan’. I rather cherish this therapy added to standard Mayo Clinic and other
particular accolade. North Central Cancer Treatment Group surgery.
So it has indeed been down to friends and foes
along the way to make the final judgements. The fact
that TME is now a standard established operation The low colorectal and
in virtually every country in the world implies that coloanal anastomosis
the friends have tended to win.
The production and widespread use of disposable
circular staplers coincided with the emergence of
John MacFarlane: a Canadian TME in the early 1980s. Completion of the anasto-
in Basingstoke mosis involved insertion of a manual purse-string
in the anorectal stump. Various operative tricks
Professor John MacFarlane, Professor of Surgery were developed to facilitate this often challeng-
at the University of British Columbia, Vancouver, ing undertaking but have now been superseded by
Canada, felt inclined to reject British and American cross-stapling techniques and hand-sewn coloanal
scepticism concerning the excellent reported out- anastomosis for the very lowest or to salvage rup-
comes from Basingstoke. He offered to join the fray ture of a cross-stapled anorectal stump.
by spending a sabbatical examining the methods During the mid-1980s it became apparent
and validity of the data. In the event he recorded a that it was easier and quicker to perform a linear
slightly greater favourable impact of ‘TME surgery’ staple line across the anorectal stump after the
on outcomes than had been reported, in both local routine washout of the lumen below a Satinsky
recurrence and survival data. His publication in the or Lloyd-Davis right-angled clamp. One of the
Lancet, ‘Mesorectal excision for rectal cancer’,7 was great advances in stapling was the ‘triple stapling
hailed by Professor Hans Troidl of Koln as the first system’ described by my registrar at the time, and
example in the history of surgery of one professor subsequently my first colorectal colleague in Bas-
visiting the unit of another to validate controversial ingstoke, which I have named the ‘Moran triple
data and thus advance the craft of surgery. stapling technique’. The details are outlined in
Figure 1.17 illustrates his comparison between Chapter 7. We have used this to facilitate TME for
Basingstoke patients treated with TME, without more than 20 years.
radiotherapy or chemotherapy, compared with the Throughout and beyond the 1980s, a firm friend-
results of the Krook–Moertel publication extolling ship and cooperation developed with the stapling

100 100
Patients without Recurrence (%)

78%
80 80
58.5%
60 60

40 40
37.3%
20 20

0 0
0 1 2 3 4 5 0 1 2 3 4 5
Years after Randomisation Years after Surgery

Comparison of lifetables for recurrence-free interval between NCCGT series (left)


and this series of TME (right).
Figure 1.17. Our comparable Dukes “B” and “C” stage patients compared with North Central Cancer Treatment Group
patients.

HEBK001-C01_p01-30.indd 15 11/02/13 3:44 PM


16  The evolution of a concept: the TME story

gun manufacturer Autosuture, which later became relation to the build and sex of the patient. Thus, in
Tyco and then Covidien. Mutual cooperation led to a slim woman it often proved to be easy to perform a
sponsorship and support for workshops at home sphincter-preserving stapled anastomosis at or near
and all over the world, and collaboration led to the the dentate line, whereas some cancers in large men
development of the TA45 (45 mm across) linear as high as 7–8 cm could occasionally prove extremely
stapler for the triple stapling technique (the origi- difficult to circumnavigate. The lowest tumours con-
nal technique was described with the TA55 stapler). tinue to be problematic, especially anterior tumours,
We advised that the average human rectum required and the lower the anastomosis the greater the risk
something more than the TA30 stapler while the of poor anal function and continence. This relates
pelvis required something less than the TA55 stapler particularly to the number of visits to the toilet each
for safe ­occlusion in the depths of the pelvis. This day, to the occurrence of episodes of actual inconti-
was a fine example of clinical and industrial coop- nence, and also to the annoying symptom known as
eration in the interests of better surgical technology. ‘clustering’, where multiple visits occur rather close
Similarly, sharp dissection, which was the essence together. Unfortunately, increased use of preopera-
of development of the holy plane, was increasingly tive radiotherapy has greatly increased the incidence
performed by pencil monopolar diathermy dis- of these symptoms over the past two decades.
section, and modern Valleylab Triverse units have Poor function has always been a worry with very
evolved to maximize cut and coagulation and mini- low anastomoses, and the disappointment of a
mize collateral damage. patient cured of rectal cancer but with worse bowel
symptoms after surgery is very problematic. Karanjia,
Schache and I published a comparison of the worst
Why is the plane ‘holy’? function expected from an anastomosis at 3 cm from
the anal verge compared with one at 6 cm.8 This
The phrase ‘holy plane’ came from the practical reality
important message justifies a subtotal mesorectal
that this was the most satisfying interface to develop
excision if it is possible to clear the cancer in higher
and deliver ‘holy space’ – an idea that came to me
tumours with a clear 5 cm of mesorectum. Even after
when visiting Jerusalem. It is a useful concept because
TME, careful preservation of a small rectal reservoir
the line that is clearly visible around the mesorectum
is desirable when oncologically safe.
on MRI, with appropriate traction and counter-trac-
tion and sharp dissection, turns into a potential space.
It has always seemed intrinsically probable to me that
Basil Morson
this potential space, which allows the juxtaposed sur-
faces to move slightly in life, implies an encompassing
During these exciting years of exploration of lower
layer, which is likely to block angiogenesis, the impor-
and lower anastomoses, one of my early friends
tant concept of Judah Folkman, who I once had the
and supporters was Dr Basil Morson, Head of
pleasure of meeting. Angiogenesis is the induction of
Histopathology at St Mark’s Hospital and successor
new blood vessels to nourish a spreading tumour, and
there to the famous Cuthbert Dukes of the Dukes
it does seem intrinsically likely that the areolar tissue
Classification. From the very beginning, Basil helped
between embryologically different organs such as the
the lift-off of the TME idea. ‘I’m a save-the-anus man’
mesorectum and the surrounding nerve layer may
he famously stated to the movie camera when the first
constitute an angiogenic barrier to the spread of can-
TME 16 mm film was produced in the early 1980s.
cer. This is little more than theory based on the unex-
He also made a highly significant scientific obser-
pectedly good results and simple clinical observation.
vation: ‘The palpable lower edge of a rectal carci-
noma is almost always the microscopic lower edge.’
The low and the ultra-low Various papers had demonstrated that intramural
downward spread from the lower margin of a rectal
Total mesorectal excision was a feasible proce- cancer occurred only rarely and only in cases of a
dure for upper and mid-rectal cancers and led to a particularly high malignant potential and a high
smooth muscle tube that is ideal for stapling. There risk of coexistent metastatic disease. This state-
emerged, however, various differences of degree of ment by a person so greatly respected by genera-
difficulty according to the height of the cancer in tions of young surgeons in a TME movie made at

HEBK001-C01_p01-30.indd 16 11/02/13 3:44 PM


Washing out the rectal stump against malignant implantation  17

St Mark’s was a huge boost.9 It reinforced the semi- and patients who have died from the consequences
nal paper from the 1970s by Norman Williams and of anastomotic leaks. In theory, both of these causes
colleagues who examined, and discarded, what used of death could be eliminated, or markedly reduced,
to be called the ‘5 cm rule’ for distal rectal clearance by constant awareness and timely appropriate inter-
below a cancer.10 vention. In practice, especially with modern early
Therefore during the 1990s, we began to focus on discharge, and discontinuity of care aligned with
what we called the ‘close shave’ in rectal cancer and suboptimal recording of patients’ temperature, pulse
published widely on this subject. From the practical and so on, patients still die from these causes.
point of view, if one could get a finger and thumb Reoperation for leakage needs to be timely and
distal to the lower margin of the cancer, and a stapler effective. The words of Rex Lawrie are worth reiter-
could be placed beyond this, there was a high prob- ating: ‘It is really important that the second opera-
ability that even margins of less than 1 cm would tion should go well because the third and fourth
not compromise cure, provided proper TME is per- never work!’ This may not be absolutely true, but
formed. Repeated distal margin frozen sections and it is important that the senior professional is always
histological analysis of the distal ­‘doughnut’ from there at that second operation.
the subsequent circular stapling firing confirmed
cancer clearance, allowing salvage of the sphincters
without sacrificing cure. Why Does Leakage Still Occur, and
One of my highlights in 1996 was presenting the Why Is It So Frequently Mismanaged?
Norman Nigro lecture at the American Society of
Colon and Rectal Surgeons.11 Norman Nigro revo- It is well established that the risks of leakage
lutionized the management of anal carcinoma from increase the nearer to the anal sphincters, with five
major surgery (APE) to chemoradiotherapy alone. to ten times the risk for anastomoses within 6 cm
Years later, with Norman sitting in the hall, I present- of the anal verge compared with higher anastomo-
ed my personal experience of 136 consecutive rectal ses. Contributing factors include haematoma for-
adenocarcinomas within 6 cm of the anal verge – that mation in the presacral space and blood supply at
is, very low cancers that would routinely all have had the bowel ends. Over the years we published three
an APE in that era. The rather provocative title ‘APE: papers on this galling subject.13–15
an endangered operation’ was the same title he had The haunting memory that 3 of the first 100
used years earlier. It incorporated the concepts and TME patients died as a consequence of anastomotic
advantages of TME, as 77 per cent of the patients with leakage probably led to very few in the subsequent
these ultra-low tumours had undergone sphincter- hundreds and largely a consequence of constant
preserving surgery and yet the local recurrence was well anxiety and early intervention combined with a lit-
below 5 per cent (confirmed on extended subsequent tle good fortune.
follow-up). My hypothesis was that the vast majority of Faecal peritonitis arising within the pelvic cav-
abdominoperineal resections, by most surgeons, were ity, away from the pain-sensitive peritoneum of the
being performed for tumours that could safely have anterior abdominal wall, commonly goes unrecog-
been removed without sacrificing the levators or the nized or is treated by repeated doses of analgesia.
anal sphincters. My own very small number of APEs Indeed, anastomotic leaks are commonly confused
had gone very badly, and this was widely quoted, par- with myocardial infarction, pulmonary embolism
ticularly in Japan. The reasons for these poor APE out- or other causes of serious illness with little abdomi-
comes may have been partly technical, although it also nal symptoms or signs. My Dutch friends refer to
represented the worst small subgroup. ‘weekend tragedies’ because the operating team is
not working and the on-call team may have a lower
sensitivity to the possibility of faecal peritonitis and
‘The sword of Damocles’: less of a feeling of ‘ownership’ of the complication.
anastomotic leakage My various phases of defunctioning all or
selected patients finally settled to defunctioning all
For all gastrointestinal surgeons, two major technical patients whose anastomosis is low enough to have
issues haunt us: patients where delay has occurred in followed a TME, somewhere between the dentate
the diagnosis of strangulating intestinal obstruction, line and about 4 cm above it. Loop ileostomy has

HEBK001-C01_p01-30.indd 17 11/02/13 3:44 PM


18  The evolution of a concept: the TME story

recently gained favour over my preference for right mesorectal fascial plane (i.e. TME holy plane) or
­transverse colostomy, which I personally regret. I direct more extended surgery, preoperative neoad-
consider a loop colostomy less likely to give small juvant therapy or combinations of both for locally
bowel adhesions, to defunction more effectively advanced tumours where the potential circumfer-
with little or no faecal residue between the stoma ential margin was involved or threatened.
and the join, and to obviate the risk of a high- Magnetic resonance imaging has revolutionized
output stoma. This is particularly relevant with the the comprehensive planning of treatment strategy,
trend towards omitting mechanical bowel prepara- is part of UK and many other guidelines, and has
tion, although the pendulum appears to be swing- been the ‘mother and father’ of the multidiscipli-
ing back in its favour for these low cancers. nary team concept. With the ability of all to view
the cancer and its surrounding structures, multidis-
ciplinary team decision-making is a major advance,
Washing out the rectal but the experienced surgeon’s finger remains a most
stump against malignant sophisticated probe for tumour height, the type of
implantation
distal margin, and the mobility of the tumour on
the puborectal sling.
My preference had always been to wash the rectal
Looking back, it is interesting to record how I
stump below a clamp. Viable tumour cells have been
‘discovered’ Gina Brown in her formative years as a
shown in animal models to produce tumours, espe-
research registrar. Janet (now Dame Janet) Husband,
cially in crushed devitalized tissue. Thus, a staple line
a leading figure in diagnostic radiology, and a friend
is excellent soil for a viable tumour seed to grow.
from my days at Guy’s Hospital, gave me Gina’s name
A paper from Sweden has reported a reduction in
as the person to talk on MRI at a meeting at the Royal
the local recurrence from 10.2 per cent with no wash-
Society of Medicine. This was when I was President
out compared with 6.0 per cent with a ­washout.12
of the Section of Coloproctology, where a paper was
Although it may be that washout is a surrogate marker
to be read that pointed out how valueless MRI was
for attention to detail, this seems a simple procedure
in the pelvis. Having foresight of this, I exercised
with no complications and major potential benefits.
‘chairman’s prerogative’ and invited Gina to ‘trump’
this paper and tell us what she was achieving. This
The Gina and Lennart story: underpins an important reality, special sequences are
the emergence of magnetic required, fine (2–3 mm) slices, small fields, cuts per-
resonance imaging pendicular to suspect margins, and so on. ‘The devil,
as with TME itself, is in the detail’.
The UK, Sweden and the Netherlands lead the In tracking down Gina to put the record straight
world in the use of MRI in staging and treatment for this meeting, I rang her hospital in Cardiff, where
planning in visceral cancer management. All of this the telephonist revealingly said: ‘Oh, I know where
is a direct consequence of the ‘TME story’. The two she is – even this late at night. She is always work-
radiologists who first grasped the key importance ing in her room in radiology.’ That single-minded
of the precise visualization of the holy plane on work ethic has driven her to the very front of one of
MRI were both intimately involved with TME at a the world’s most exciting imaging advances – rectal
practical level, Gina Brown in the UK and Lennart cancer treatment planning. It is thus through Gina
Blomqvist at the Karolinska Hospital, Sweden. and Lennart that the history of MRI planning is so
Similarly, my visit to Maastricht with the ‘TME intimately bound up with that of TME. So much of
circus’ touched the work of Regina Beets Tan, who the future will be about imaging that this reality is a
has also become one of the world’s great MRI lead- major part of our story.
ers. All three had already recognized the potential
of MRI to delineate cancer and bowel wall mus-
cle, essential for T staging. The ability to delineate The home front: Pelican
the embryologically determined holy plane was of
even greater potential importance. Magnetic reso- My operating career and TME drive would have
nance imaging could predict a clear margin in the been shorter by at least 15 years if it had not been

HEBK001-C01_p01-30.indd 18 11/02/13 3:44 PM


‘Metastatic’ total mesorectal excision to the liver and peritoneum  19

for the organizational skills of Sir Peter Michael, idea to encompass at the same time the concept
creator of the Quantel Electronics Empire, Classic of multidisciplinary cancer board meetings.
FM and countless other enterprises that appealed Serendipity and the wisdom of Sir Peter
to his fertile imagination. He sculpted the Pelican Michael produced at the Pelican a series of first-
Centre, which gave an independent home, an office class chief executive officers – first, Diane Hayter,
and a personal ‘carer’ for me. Of all professionals, who put the show on the road, and then Chris
surgeons seem to need these ‘carers’ most of all. Beagley CBE. Chris brought a determination of a
Jenny, Juliet and Emma have exercised the Pelican naval ship’s captain and the negotiating skills of
hand on the rudder – their names come as worthy a former civil servant at the Ministry of Defence
successors to Jill, Rosemary and Pat in the twentieth to bear at a critical moment. Sir Michael Richards
century. Throughout, the Medical Director Dr John was so determined that his colleagues said ‘He’ll
Fowler has overseen the key decisions and direction get his way in the end’, and Chris delivered for
of the whole foundation with unique wisdom. The Pelican the biggest contract in its history and the
conquest of the world would certainly have faltered most outstanding government-backed training
without Emma and all at Pelican. scheme ever, the ‘multidisciplinary team TME
Pelican also expanded the horizons. The name development project’.
is short for ‘pelvic and liver cancer’. It sought to All but one colorectal cancer multidisciplinary
encompass the two poll positions that our little Bas- teams in England attended between 2003 and 2007
ingstoke Hospital was aspiring to in the world of sur- (148 of 149). Out of all this came complete accept-
gery – rectum and liver. At the same time it sought ance of the principle of multidisciplinary team dis-
to extend at least our intellectual involvement to the cussion for every patient and mandatory MRI for
management of prostate cancer. The disease was each patient. Real planning had started. After 8 years
essentially different – that which is lethal obscured Chris passed the helm to Sarah Crane, who has skil-
by red herrings, and the diagnosis by a fallacious fully negotiated a mass of burgeoning bureaucratic
blood test, prostate-specific antigen (PSA). So it was obstructions to clinical research, and she now keeps
that the Pelican team branched out into focal ther- Pelican securely on course.
apy for prostate cancer, yet another story that spins
off from TME. In particular we have focused on pre-
cision imaging, which has the potential to reduce so ‘Metastatic’ total mesorectal
much suffering, but that is another story! excision to the liver and
peritoneum

Total mesorectal excision Surgical expertise and endeavour propagate and


and British medical politics ‘metastasize’, and thus Basingstoke has wandered
into two other fields where attention to detail and
The greatest tragedy of the NHS is hugely pow- surgical technique are key.
erful managers who do not understand the once The first development was the ‘hepatic element’
powerful, but now impotent, consultants. Com- of TME history with the appointment of Merv Rees
munication blocked is progress deferred. Only in the late 1980s to tackle the problem of color-
those who can build bridges are those who can ectal secondaries in the liver. I had become aware
still achieve real improvement in hospital prac- of the unexpected benefit attached to what preci-
tice. Sir Michael Richards has this in spades. sion hepatic surgery might deliver, especially on
Appointed as the first ‘cancer tsar’, he looked encountering Johannes Scheele from Erlangen in
around him through the fog of UK-critical Euro- Germany. It was a pleasure to be a catalyst in Merv’s
care data for projects to improve outcomes and friendship and cooperation with Johannes and ulti-
increase real cures. He took the trouble to travel mately establishing Basingstoke as a world-class
to Sweden to confirm our own wholly unlikely centre for hepatic resection. In addition Merv has
story of Englishmen and Irishmen training spearheaded the educational centre, which has been
Swedes, and he returned with a burning determi- key to our home efforts in ongoing education and is
nation to repeat and build on the TME workshop properly called ‘Merv’s Ark’.

HEBK001-C01_p01-30.indd 19 11/02/13 3:44 PM


20  The evolution of a concept: the TME story

Another key addition has been the treatment of willingness to believe the unexpected and almost
peritoneal malignancy, in particular pseudomyxoma unbelievable results and their potential benefit also
peritonei of the appendix. Visceral surgeons who started in Norway and ­Sweden, although the impor-
innovate find themselves a part of an international tance of ‘Das Konzept’ was foreseen by Markus Buch-
club of eccentrics. My encounter commenced with a ler in Germany. Much of the Scandinavian story has
telephone call from a general practitioner friend, Ste- already been told, along with some of the film scenar-
ve Tristram, who had been contacted by a childhood ios of operating rooms invaded by coffee machines
friend of his wife whose young husband had been with Swedish and Norwegian surgeons eager to absorb
diagnosed at open-and-close laparotomy ‘with a rare the TME message. This early enthusiasm was greatly
kind of cancer called pseudomyxoma but nothing enhanced in both teachers and taught by the growing
can be done’. The answer to the question ‘Bill, can you involvement of Brendan Moran, whose ready Irish
do anything?’ was ‘I can’t, but I know someone who charm and consummate surgical skill soon established
can – I heard him last week in Hong Kong.’ This cul- him as the future captain of the ‘Good Ship TME’
minated in a visit to Basingstoke by Paul Sugarbaker and ultimately the clinical lead in both the second
from the Washington Cancer Centre; Paul, assisted by Danish national project and the somewhat belated
me and Brendan, performed a 12 h operation, which British Low Rectal Cancer National Development
gave the man 12 years of life. It was the first of what is Programme (LOREC).
now approaching 1000 operative cases, and the name
Basingstoke has emerged once again, spearheaded by
Brendan and now Tom Cecil with three younger col-
Ireland
leagues, Steve Arnold, Faheez Mohamed and Arcot
Many a rejected Englishman, smarting at the indiffer-
Venkat, joining the colorectal team in April 2009.
ence of his fellow countrymen, has found solace and
That first operation will be remembered for
welcome across the Irish Sea. Conversely, Brendan,
many things, including three emergency visits
like many Irish surgeons, found similar solace in leafy
by the fire brigade as a consequence of fire alarm
Hampshire. Many Dublin surgeons, Liam Kirwan
activation by the high-powered diathermy smoke.
in Cork and surgeons from across the Emerald Isle
embraced and practised TME at an early stage. Liam
Worldwide total mesorectal Kirwan published figures identical to those from Bas-
excision ingstoke very soon afterwards, the first firm confir-
mation that our data were real.
National Total Mesorectal Excision To me, Irish TME philosophy runs thus: ‘Viola-
tion of the mesorectum is the clue to surgical failure
Projects – treat it delicately like a virgin’.13
Perhaps the most extraordinary feature of the TME
story is the number of other countries that set up Germany
national training projects in TME technique before
Britain. The Anglo-Saxon world was certainly the last For me, the worldwide TME story begins in and ends
to show any enthusiasm. Professor Norman Williams with Germany. An early great honour was that Markus
voiced dissatisfaction to the press at our failure in the Buchler travelled to Basingstoke to visit one of our first
UK to introduce the training aspects formally at an workshops back in the 1980s. This was followed by an
earlier stage. All the fundamental ideas were crystal- invitation from Bern, Switzerland to present the Koch-
lizing across the world from UK origins, but in other er Memorial Lecture. Researching the life of Theodor
cultures before ours – the ‘Bill, Phil and Gina show’ Kocher is inspirational: he was a brave honest surgeon
has extended to what the Canadians recently called who believed in slow meticulous technique and the
the ‘fab four’, embracing our Pelican revolution- primacy of precision, exactly like the TME dream.
ary oncologist Rob Glynne-Jones. Excluded was our
new leader Brendan Moran, who stayed at home for Markus Buchler
the wedding of Prince William and Kate Middleton. Professor Markus Buchler rapidly recognized
Casting back to earlier days, it must be recorded that the potential importance of TME, and our book

HEBK001-C01_p01-30.indd 20 11/02/13 3:44 PM


Worldwide total mesorectal excision  21

Stunning variability between surgeons in


Germany: Paul Hermanek
Paul Hermanek has been called the ‘father of
­German histopathology’. He is, along with Cuth-
bert Dukes, Basil Morson and more recently Phil
Quirke, one of the all-time greats among ‘sur-
geons’ pathologists’ – those who involve them-
selves in the detail of the specimen, those whose
room is always open to the surgeon bearing fruits
fresh from the operating room of a recent battle
against cancer, eager for consultation and discus-
sion with a fellow warrior. Such pathologists are
one of the greatest weapons we have in the war
Figure 1.18. Das Konzept.
against cancer.
Paul made two seminal contributions in his
paper,15 curiously published in the Italian journal
Das Konzept Der Totalen Mesorektalen Exzision14 Tumori. His observations were crucial: first, there
­(Figure 1.18) published the many implications of was a huge variability between operating surgeons
new thinking. Markus regards cancer surgery as a in their local recurrence rates; second, these varia-
question of compartments and emphasized that tions were reflected in long-term survival. The clear
TME was the careful emptying of the central pel- implication was that poorer surgeons were failing
vic compartment. This amplifies the embryological to remove tissue potentially containing cancer. This
midline mesorectal entity and makes us aware that could then go on to metastasise.
each lateral compartment contains the internal
iliac vessels outside the nerve layer and a septum of Recent times in Germany
significant parietal fascia, which separates paired It is no surprise to me that, despite speaking no Ger-
parietal structures from the midline visceral block. man – except perhaps for ‘zug und gegen zug’ (‘trac-
The visceral envelopes have arteries emerging from tion and counter-traction’) – I now feel most wel-
the front of the aorta, and middle rectal arter- come of all in Germany. Having grown up during the
ies from the internal iliacs are uncommon. At the Second World War and witnessed a Messerschmitt
time the German book was published, Christophe 109 shot down in anger, my life has gone full circle
Maurer translated many chapters for me, and he – what a desperately sad and destructive century the
remains a leading German exponent of TME. twentieth was for our great continent of Europe. At
This first visit by Markus Buchler and his two least we have moved on and the TME story is one of
friends will never be forgotten – a totally unsuit- friendship containing many key German contribu-
able patient, no MRI, a ‘fixed cancer’ and no plan- tions. Professor Paul Hermanek’s observation of the
ning. Videos of the operation survive to illustrate unique, totally unexpected variations in the local
my ‘skeletons in the cupboard’ talk. These show recurrence rates in seven excellent German hospitals
me cutting the left ureter while trying to clear the was seminal to the concept. Equally important,
cancer by excising the seminal vesicles. Lessons and still defying the understanding of some clini-
were learned that day: we needed planning tools cal and medical oncologists, was Paul’s observation
(now MRI), and the ureters in such advanced cases that surgeons who achieved lower local recurrence
should always be stented. rates also had more true cures and fewer metas-
More amusing was the cocktail party in the back tases. This of course is critical to the importance of
garden of our village home when Phil Quirke sat on TME, because the natural history of rectal cancer is
a pillar and cut up the specimen and passed around such that it remains locoregional in one central pel-
the slices; the guests, champagne in their hands, vic compartment for a very long time. During this
were offered these alternating with cocktail fancies period a perfect TME will remove all the satellites of
and sausages on sticks. My wife protested ‘What will cancer and thus cure the patient. Residual mesorec-
people in the village think?’ tum containing cancer or implanted cells on the raw

HEBK001-C01_p01-30.indd 21 11/02/13 3:44 PM


22  The evolution of a concept: the TME story

pelvic side-wall are extremely likely to lead to local that one could stand a cup of coffee on the peri-
regrowth and ultimately death, from either direct neum. This option in a slightly modified form is
extension or metastases from the cancer left behind. now emerging again in the Netherlands under the
Friends in Heidelberg have been the princi- influence of Harm Rutten of Eindhoven. There is
pal basis of my ongoing practical teaching life in no doubt that this difficult dissection in through
­Germany in recent years, together with regular the perineal body with its proximity to the urethra
visits to Erlangen, the home of Paul Hermanek. on the one hand and the front of the specimen on
Two-day craft workshops still seem the premier the other would be very much better effected with-
teaching mode, while the modern video connectiv- out turning the patient if further studies show that
ity of high-definition laparoscopy stacks and the Da equally precise dissection is possible that way. The
Vinci Robot can potentially extend the experience question still remains open, and the most difficult
to larger and larger audiences around the world. In challenge is certainly the perineal body and the
April 2012 I was proud to be a part of the Heidelberg neurovascular bundles subserving erection. The
surgery team transmitting an open TME by satellite problem is that there are no natural planes to guide
to the German Surgical Association in Berlin for the surgeon and there is almost no room for error.
Markus Buchler’s presidential meeting.

Total mesocolic excision: Werner Hohenberger France


The influence of TME extends beyond the rectum.
Professor Werner Hohenberger in Erlangen has put There are probably few stories of human endeavour
down robust TME type principles for mesocolic where the French have not added their own unique
excision in colon cancer. Such principles do seem, to dash of flair. So it is with our story. The word is
rigid oncological surgeons like him, me and Brendan, laparoscopy, and the minimally invasive hall of
sometimes to have been compromised by some sur- fame is littered with the names of Frenchmen –
geons following the rise of minimally invasive sur- Marescaux, Leroy, Rullier, Panis.
gery. Ligation of the ileocolic vessels flush with prop- Even before the laparoscopic revolution I over-
erly exposed superior mesenteric vessels and intact heard a most memorable compliment from Pro-
untorn colonic mesenteries are two examples. fessor Rolland Parc addressing a group of French
It is a personal opinion that we are beginning to surgeons, describing TME as the ‘l’idée la plus
see the rise of German surgery towards its formerly importante du monde’. He, and later Professor
dominant position before the tragedies of the 1930s Emmanuel Tiret, at the Hôpital St Antoine have
and 1940s. It is therefore a source of pride that the established a centre of excellence with TME at the
widespread teaching of TME along practical work- heart of its rectal cancer surgery. It was my privi-
shop lines has been embraced so enthusiastically lege to undertake a three-screen review of their
in Germany and may become a part of this rise in technique as long ago as 1995, and their APE results
other surgical fields. suggest that they are well ahead of us when it comes
to avoidance of the ‘waist’ that bedevils APE.
Joachim Strassburg The rise of laparoscopy has been gradual from
At an early stage in our discussions about how benign disease to malignancies, starting with small-
unsatisfactory our technique for APE indeed was, er, easier and earlier, and progressing with caution
the ‘Berlin position’ entered the argument. This to larger and deeper. Back in 1990, my videos of
was put forward by Professor Joachim Strassburg sharp dissection under direct vision seem to have
from Berlin, who claimed that perfectly satisfactory inspired Professor Joel Leroy at a meeting in Tours:
access to the back of the prostate and the challeng- ‘I determined that day that we would reproduce pre-
ing dissection between it and the front of the speci- cisely with the laparoscope what Heald was doing’.
men could be effected without turning the patient. Over the years this process has progressed like two
Essentially the Berlin position meant a very steep long-distance runners – me for example with three-
Trendelenburg with shoulder support and maxi- directional traction as the key tenet of teaching,
mal flexion and abduction of the hips; the patient and him adding a fourth dimension because of the
was so steeply head-down that Joachim pointed out pneumoperitoneum, and so on.

HEBK001-C01_p01-30.indd 22 11/02/13 3:44 PM


Worldwide total mesorectal excision  23

Joel said: ‘I did probably the first laparoscopic TME invitation of the great laparoscopic surgeon Eric
with coloanal anastomosis in November 1991 in a Rullier tested the idea that certain patients with
young male patient [after chemoradiotherapy]. It was very low cancers could only be performed open. In
a successful procedure and satisfactory oncologically’. 2010 Eric believed that this was true, but he now
A postscript can be added in Joel’s own words that believes that such patients do not really exist for the
provides perhaps a fantasy glimpse of the future. highly experienced laparoscopist, except occasion-
ally for those with certain very large cancers.
I have news for you! I am writing for Archives If we extend this story to liver secondaries,
of Surgery the first NOTES transanal TME France can also boast one of the greatest of all liver
without abdominal assistance and diverting surgeons – Professor Henri Bismuth of the Hôpi-
stoma using a particular procedure to mobilize tal Paul Brosse. He has also exchanged visits with
the sigmoid and descending colon and divide Basingstoke.
the mesenteric vessels. I called the approach
PROGRESS – perirectal oncologic gateway
for retroperitoneal endoscopic single site Sweden
surgery: all the dissection is done by transanal
retroperitoneal approach. Sweden has already featured in the story, but,
around the turn of the century, the Scandinavian
Eligio Floscoli of Covidien has also been persuad- data collection was to prove a new weapon of enor-
ing me of the merits of dissecting upwards in the holy mous power. Throughout my time on the council
plane from the perineum, along with John Marks of the Royal College of Surgeons, the strenuous
from Philadelphia, the delightful son of the illustrious efforts of the maxillofacial surgeon John Williams
Gerald Marks. Who knows? The holy plane may per- to introduce compulsory registration of cancer
haps be accessed entirely by the ‘unholy orifice’! As we and comprehensive clinical outcomes recording
go to press, the charismatic Spaniard Antonio Lacy in the UK had been sidelined by the Department
and I are also focused on this possibility. of Health. The ­manager-dominated NHS was far
more interested in politically sensitive issues such
Video-surgery as waiting lists. Not so the Swedes or Norwegians.
The primacy of surgical ideas being transmitted by National training projects in both countries gath-
video is reflected in the rise of great surgical teach- ered momentum, backed by compulsory pro forma
ing units with ever improving visual relay of ever registration of all the clinically relevant informa-
more surgical detail. Nowhere is this more evident tion – very similar to ‘Rosemary’s database’ back
than in the magnificent Institut de Recherche contre home in Basingstoke. What was being recorded, at
les Cancers de l’Appareil Digestif (IRCAD) Euro- last for a whole country, was the information that
pean Institute of Tele Surgery (EITS) in Strasbourg. cancer doctors were interested in – that is, those
Here I rejoice in the wonderful title of ‘Pope of the things that might influence cure and patients’ suf-
Rectum’, a manifestation of the ready wit of Jacques fering. Such enlightened backing has reaped many
Marescaux, the great French surgical entrepreneur. rewards. Most important was the ‘Martling papers’,
The virtuosity of Leroy and of a rich line-up of which demonstrated without doubt the impact of
invited surgeons provides unique training in the our new workshops.16 More randomly, on a recent
potential of colorectal laparoscopic surgery – always visit to Gothenburg, which had formerly resisted
a wonderful weekend for everyone, faculty and visi- what Lars Pahlman called the ‘Heald wave’, real
tors alike. In the background is the comprehensive evidence emerged of the value of irrigation of the
Websurg providing accessible training on demand anorectal segment before stapled anastomosis.
from cyberspace. Extraordinarily the Marescaux As a believer in the potential of the NHS for
tentacles have replicated the IRCAD miracle in Tai- accumulating clinical data, I can only grieve that we
wan and Brazil, where it is also my privilege to preen have not explored its potential in the way that they
myself as ‘Pope’. The world owes Marescaux a lot. have in Sweden. Fortunately the Association of Col-
Elsewhere in France the saga has continued. An oproctology of Great Britain and Ireland (ACPGBI)
open TME operation by myself in Bordeaux at the is now addressing this matter urgently.

HEBK001-C01_p01-30.indd 23 11/02/13 3:44 PM


24  The evolution of a concept: the TME story

Norway ing component was already under way with the


Japanese colorectal surgeon Yoshihiro Moriya. He
The Norwegian data have a special interest of their visited me at home in Basingstoke and we operated
own. As in other matters, the Norwegians have shown together several times. Memorable to me, and to the
remarkable independence and non-compliance with patient who I later told, was his comment about the
the American National Institutes of Health (NIH) male hypogastric nerves: ‘Englishman has very big
consensus advice that led to neoadjuvant therapy sex nerves’ he announced loudly to the operating
sweeping across the world. Their series reflects a very room. I declined to comment in the hope that we
small usage of radiation and reports results very little might, if this view was to be widely promulgated,
affected thereby, and of course superior functional thereby elevate the street credibility of the Eng-
results because low anastomoses and radiotherapy lish. This may have succeeded as I heard it repeated
are unhappy bedfellows. A substantial book was some years later. Both Yoshi and I criss-crossed the
written early on about the Norwegian project, and traffic-packed roads of the Netherlands in a unique
Norway’s influence endures through the years. enterprise to change surgical habits for a whole
nation of 20 million people. A posse of mentors
followed behind us providing surgical discipline
The Netherlands unique to the Netherlands. For me, a particular and
special Dutchman was a surgeon from the Academic
The formidable Dutch entrepreneurial surgeon Medical Centre (AMC) in Amsterdam, Carlo Taat. It
Cornelis (Cock) van der Velde first approached me was he who took me to every major hospital in the
in 1993. At that time he had no real funding and was Netherlands and attended every one of almost 50
indeed flying by the seat of his pants, with his dream demonstrations that I performed there. Together we
of improving cancer surgery. From our early meet- realized that learning TME is a progressive process,
ings came the Dutch TME trial, which has played the demonstration of the posterior dissection being
in the colorectal concert halls for almost 20 years, much easier than the front. The demonstration of
generating more theses than any other project ever. ‘Bill’s billen’ (Dutch, ‘buttocks’) by a surgeon is per-
The trial was a development of Cock’s earlier D2 haps the first and easiest sign that he has grasped the
Radical Gastrectomy Cancer project, and the train- importance of holy plane dissection (Figure 1.19).

Figure 1.19. Bill’s ‘buttocks’.

HEBK001-C01_p01-30.indd 24 11/02/13 3:44 PM


Worldwide total mesorectal excision  25

His high intellect contributed much unrecognized deaths that clearly followed radiotherapy, apart from
wisdom to the importance and impact of the Dutch some second malignancies in the irradiated pelvis
TME trial in the world of coloproctology. He later and a hint of additional cardiovascular deaths. This
drove me round Baden Wurtenburg in south-west is surely a major challenge to the world of clinical
Germany on a similar mission. Sadly, I later went oncology to demand an answer. If we get such an
to the Netherlands to say goodbye to him when he answer, then TME will make yet another chapter in
decided to exercise the right of Dutch citizens to the cancer story.
end their own lives – his duodenum was obstructed Figure 1.20 shows that the greatest impact of
by an irremovable intractably painful malignancy. the Dutch TME trial was that the surgical train-
As one of the two surgical trainers, I must con- ing improved national outcomes immediately –
fess to disappointment at the high level of reported workshops are the thing.
margin involvement – 23 per cent – in the Dutch
data. So, although we cannot interpret the surgery
in the Dutch trial as being truly optimal TME Belgium
surgery, we salute Cornelis and Carlo’s courage
in tackling the enormous task of attempting to An interesting case report from the Procare Project
convert an entire country’s surgical community states:
to long, slow tedious TME dissection and careful
Patient had had a sigmoid to anorectum
planning.
anastomosis . . . visit by me at 14 days: niggling
The very holding of the workshops had a remark-
fever, normal water-soluble radial opaque
able impact, as analysed by Peeters (Figure 1.19).
enema X-ray shows no leak. However, rectal
Further major information is emerging after 12 years
examination reveals the unique odour of
of fastidious follow-up of this near-perfect prospec-
gangrene on the examining glove. Proctoscopy
tive randomized controlled trial of radiotherapy ver-
shows the colon to be black and necrotic despite
sus no radiotherapy with no chemotherapy. There are
the absence of a leak at that stage. Laparoscopy
more people alive in the group that had not been irra-
was performed to mobilize the splenic flexure,
diated than in those who had received short-course
excise necrotic sigmoid and effect a new stapled
high-dose radiation. This was despite about 6 per
anastomosis. Satisfactory outcome.
cent of extra cancer cures, but these have been more
than cancelled out by additional deaths from other This case emphasizes the importance of the tem-
causes – second malignancies in the irradiated pel- perature chart, the need for constant awareness of the
vis, cardiovascular deaths, et cetera. No explanation risk of anastomotic failure, and the importance of
has emerged for the several largely undocumented examining a suspect anastomosis with a proctoscope

Figure 1.20. The impact workshops,


the Netherlands.
RT, radiotherapy; TME, total mesorectal excision.

HEBK001-C01_p01-30.indd 25 11/02/13 3:44 PM


26  The evolution of a concept: the TME story

to establish viability: viable ends mean that all faeces


must be washed through and the anastomosis can
remain truly defunctioned and not need to be taken
down.

Italy

As in so many aspects of life, Italy TME has


embraced TME with generosity. Many TMEs have
been performed in Rome, Milan, Bologna, Naples,
Palma and Bari.
One Armani-suited elder on Palma station greeted
me as ‘Signor Mesorectum!’

China and Japan

I have been made particularly welcome in these two Figure 1.21. A perfect abdominoperineal excision is a total
so different and important countries. Each deserves mesorectal excision wrapped in levator and sphincters.
a whole chapter in its own right, for rectal cancer is
common in both and extremely individual national Hong Kong
attitudes to the disease are apparent. Controversy
has raged for decades between Japan and the west Hong Kong has played a major part in TME, with
over the oncological importance of the internal iliac more than 20 visits, countless workshops and
and obturator nodes, the prophylactic removal of video-operations, and the huge talent of Profes-
which has been quintessentially Japanese but com- sor Michael Li of the Chinese University. Some of
pletely ignored by myself. My TME story is entirely the earliest laparoscopic versus open TME ‘con-
about what Markus Buchler calls the ‘central tests’ took place here, and Hong Kong has devel-
compartment’. Hideaki Yano and Brendan Moran oped the most modern endoscopic laparoscopic
has addressed this fascinating issue in Chapter 14. operating room, where all combinations of endo-
China is perhaps the deepest mystery in the scopes are available at all times.
world, and rectal cancer surgery is not excluded.
Each time I return from China I am a little more
confused. Several outstanding professors have Russia, Poland and Eastern Europe
befriended me there, and there is no doubt that
TME has made an impact in Beijing, Shanghai, As I near the end of my career as a teacher of surgery,
Chengdu and Wuhan, in all of which I have done the shift towards the eastern part of Europe has been
open TME workshops. One most interesting dif- most noticeable. In 2010–2011 I was invited to Russia,
ference from western practice is that low anas- Poland, the Czech Republic, Hungary, the Ukraine,
tomoses are often simply drained for 2 weeks or Serbia, Slovenia (Figure 1.22) and Romania. The inci-
more against the risk of faecal leakage but with dence of colorectal cancer across the world is highest
no diverting stoma created. Professor Gu at the of all in these countries, the top (or bottom) of the
Beijing Cancer Hospital has undertaken exten- league being Slovakia. The reasons are probably the
sive TME histopathology micro-analysis and usual mix of dietary and genetic factors, but the need
established that small node positivity for cancer for good affordable management plans with good
is far more common than is generally believed – a outcomes is nowhere more urgent.
cogent further reason for the TME ‘oncological The first visit to Moscow in 2009 was memo-
envelope’ concept (Figure 1.21). It is better sim- rable, with many hundreds of surgeons attending
ply to regard mesorectal fat as ‘dangerous stuff ’. from more than 80 cities in 9 countries. The patient

HEBK001-C01_p01-30.indd 26 11/02/13 3:44 PM


Worldwide total mesorectal excision  27

% 100

90
80
70
60
50
40
30
20
10

0
Local 5 year Permanent
recurrence survival stomas
(incl St IV)
%
Before After

Local recurrence 21 4

5 year survival (incl St IV) 34 77

Permanent stomas 67 18

All above p5,0.05; stage distribution and RT rate (54%


and 52%) unchanged. Figure 1.23. The Petrovsky Medal and Professorship.

Figure 1.22. Astonishing results in Slovenia: 195 patients


before and after two TME video workshops in 2009.
a perfect TME (see Figure 1.21) but that the sphinc-
ters and perhaps the levators should be accurately
proudly announced to Russian television that he surrounding the distal part of the TME.
was a wild boar hunter, and he has since sent me Torbjörn Holm had already suggested improv-
more than 50 pictures of his continued activities ing APE by turning the patient into the prone jack-
in this field after his successful operation. This visit knife position, removing the coccyx, and doing the
was the brainchild of Professor Petr Tsarkov, who job from behind. In Poland this was already hap-
has become a firm friend and through whose medi- pening and the words of Kocher from 1897 were
ation Russia bestowed upon us its highest medical key: ‘The surgery of the sacrococcygeal region has
honour, the Petrovsky Professorship and Medal. attained greater interest since it has been recognized
These were formally conferred 2 years later in 2011. that . . . access to the pelvic organs from behind is for
They stand as a pivotal step forward in the interna- many reasons preferable to that obtained from the
tional spread of the TME idea (Figure 1.23). perineum.’
The cost of the disease and the management of Marek Bebenek taught me how right Kocher’s
failure is enormous, with the ever present nightmare words were. I returned to the role of resident and
of massively expensive last-ditch treatments from was taken through what I now like to call the
big pharma being simply unaffordable. Generally ­Kocher–Holm APE. Torbjörn Holm’s own chapter
these buy only weeks of rather unenjoyable life. The in this book is well illustrated with the emerging
real major solutions are earlier diagnosis and better excitement of one very special advantage of the
surgery. Kocher–Holm exposure. Whether Kocher himself
included in his ‘many reasons’ the ability to iden-
Wroclaw, Poland tify the neurovascular bundles as they converge to
Some visits become fact-finding experiences for me. the bulb of the penis is not recorded. This major
In Wroclaw, Professor Marek Bebenek had lined up advance is indeed what we demonstrated and vid-
eight operations to be performed over 2 days on eoed for the very first time at one of our Pelican
two tables side by side in one room. Four were to operative workshops. Tom Cecil performed the
be sphincter-preserving and four were to be APEs. upper end and placed a clip on the hypogastric
It is self-evident that every APE should encompass plexus; later, with me at the audience end, the whole

HEBK001-C01_p01-30.indd 27 11/02/13 3:44 PM


28  The evolution of a concept: the TME story

unique cadaver dissections and animations – all


flexibly coloured and labelled. In my opinion noth-
ing comparable has ever been achieved in the field
of teaching of surgical anatomy. I recommend it.
Please contact Ayhan Kuzu for more information
ayhankuzu@yahoo.com.

The USA and Canada

Figure 1.24. Neurovascular bundles after abdominoperineal For this mighty continent and our impact upon it, a
excision. Note the proximity to the divided right puborectal whole book is required. We will highlight only a few
muscle. themes. First was the excellent surgical technique
I learned as a resident at the Ochsner Clinic in New
Orleans, where John Ray and his colleagues had estab-
neuroanatomy at the back of the prostate was laid lished one of the world’s first colorectal specialist
bare, with visual continuity established by the clip units. At that time, at the Mayo Clinic anything above
(Figure 1.24). The ongoing APE challenge still 5 cm was still under the general surgeons. From Can-
remains and is covered in Chapter 8. ada came the all important intervention from John
MacFarlane from Vancouver, which was critical to the
establishment of our credibility; from Warren Enker
A ‘TME Blockbuster’ from Turkey in New York came the same ideas presented robustly
and backed by immaculate data – plus an enduring
Students of TME now have access to a real treat: friendship for me and the still memorable operation
the perfect companion to this book. Ayhan Kuzu, in Basingstoke where the left-handed Enker did the
Professor of Surgery from Ankara, with help from patient’s left side and, I, right-handed, did the right side
many of our authors (Heald, Quirke, Brown, Rullier, (Figure 1.25). Along the way have been many interac-
Holm), has created a virtual university course on tions with admired American friends – the Norman
the subject in a pack of three DVDs. The con- Nigro lecture, visits and lectures at Memorial, Beth
tents include most of our best videos plus Ayhan’s Israel, Steve Wexner’s presidential invited lecture at the

Figure 1.25. Warren Enker from his


Ernest Miles Centenary Lecture.

HEBK001-C01_p01-30.indd 28 11/02/13 3:44 PM


Conclusion  29

Society of American Gastrointestinal and Endoscopic preparations had been made for anastomosis. The
Surgeons, and so many more. More recently, in 2011 pelvis was packed and Brendan completed the sur-
a major project involved us again in Toronto, where gery 2 days later. The patient is cured 8 years on.
all the concepts – MRI, surgical and pathological – Perhaps the most dramatic incident occurred in
were promoted for the 12 million people of Ontario. front of one of my largest audiences ever, from all
For years these concepts have been a key part of the around the Balkans. The city was Belgrade. Sanc-
standard thinking of so many good American friends, tions were at work, and so the reserve diathermy
including Stan Goldberg, David Rothenburger and machine was of unspecified origin. It later tran-
Phil Paty. Professor Steve Wexner has done much for spired that the settings had been known to no-one
international surgical training, with special empha- and were five times the manufacturer’s recommen-
sis on Central and South America, Russia and Israel. dations. The video shows that the camera recorded
Rob Maddoff and others are promoting these training something amiss as there was electricity in the air
methods in their own constructive ways for the better- and each touch of the diathermy makes the picture
ment of US cancer management. jump. I point towards the right hypogastric nerve,
2012 is a special year for TME as it will provide an arc of electricity leaps to ground on the patient’s
the content for the Harry Bacon lecture at the spe- right external iliac artery and a fountain of blood
cialist meeting of the American Society of Colon hits the light. There is an attempt to control the
and Rectal Surgeons in San Antonio, and later the flow, eventual common sense to ask for audience
centenary cancer lecture of the American College in support, and the appearance of a friendly vascular
Chicago. In summary, TME has established a firm surgeon and subsequently my carefully completed
place in both British and American practice. TME. Many lessons were learned that day, and
the then 21-year-old woman still survives in good
health.
Anecdotal dramas from More quaintly, I have presided over the top end of
the operating room a widely transmitted colovaginal stapled anastomosis.
We did recognize and reverse it, so all was well. The
These are mostly tales that could have had a very sharing of both triumphs and disasters with visi-
unhappy ending. On a visit to Istanbul, the elec- tors and audiences has perhaps been a bumpy road
tricity was cut off as my right hand was poised to towards better surgery around the world, but we claim
cut and we entered 65 minutes of total darkness. A always to have been honest with our visitors.
similar event occurred at a Basingstoke workshop,
when the building works for the Ark cut through
the power line and all electricity, including the Conclusion
emergency reserve, disappeared. The visiting audi-
ence returned to their hotels but were called back This chapter is not finished. Perhaps it never will be.
over an hour later when, mercifully, reconnection There are many gaps in the detail and many individu-
was achieved; the patient lives 10 years on, after als who have contributed so much along the way, and
a brief moment of fame on the front of the local so many patients who have been central to the story.
paper. In this case the blood supply to the rectum It is remarkable to see the same TME principles being
had already been divided so that non-continuation maintained and strived for as new technologies are
was not an option. Transfer to another hospital applied – laparoscopy, robotic, NOTES and all. The
would have involved descent of a stairwell with the cost-effectiveness of new expensive technologies
patient anaesthetized and the abdomen open or such as robots is debatable, but it is gratifying that all
packed shut, as the lift had also failed. are striving to achieve the same aim and are adding
In another Basingstoke workshop I experienced another brick to the TME wall.
uncontrollable bleeding, subsequently discovered Total mesorectal excision has hugely stimulat-
to have been a result of inferior vena cava (IVC) ed the development of MRI cancer planning, has
occlusion from a prophylactic IVC filter as the pre- added sophistication to scientific histopathology,
operative CT had shown common iliac thrombosis. and has provided the most reliable and reproduc-
Total mesorectal excision had been performed and ible reference standard for surgical technique, and

HEBK001-C01_p01-30.indd 29 11/02/13 3:44 PM


30  The evolution of a concept: the TME story

each of these is constantly evolving over time. To   7. MacFarlane JK, Ryall RD, Heald RJ. Mesorectal exci-
list all the major players along the TME pathway is sion for rectal cancer. Lancet 1993; 341: 457–60.
  8. Karanjia ND, Schache DJ, Heald RJ. Function of the
impossible, but together they have made the word
distal rectum after low anterior resection for carci-
‘Basingstoke’ at the heart of any meaningful discus- noma. Br J Surg 1992; 79: 114–6.
sion on optimal surgical technique for cancer and a   9. Karanjia ND, Schache DJ, North WR, Heald RJ. ‘Close
landmark in rectal cancer management. shave’ in anterior resection. Br J Surg 1990; 77: 510–2.
10. Williams NS, Dixon MF, Johnston D. Reappraisal of
the 5 centimetre rule of distal excision for carci-
To Our Registrars noma of the rectum: a study of distal intramural
spread and of patients’ survival. Br J Surg 1983;
Brendan and I share a sense of profound gratitude to 70: 150–4.
11. Heald RJ, Smedh RK, Kald A, Sexton R, Moran
the many surgical registrars who have slaved for hours
BJ. Abdominoperineal excision of the rectum: an
over the human pelvis in the unique quest for greater endangered operation. Norman Nigro Lectureship. Dis
skill and understanding. We started to compile a list Colon Rectum 1997; 40: 747–51.
and settled on avoiding the offence that an omission 12. Kodeda K, Holmberg E, Jörgren F, Nordgren S,
might give. We mention one symbolic name – Marga- Lindmark G. Rectal washout and local recurrence of
ret Farquharson – because she probably slaved longer cancer after anterior resection. Br J Surg 2010; 97:
than any of you. If you have read this far and feel a part 1589–97.
13. Reynolds JV, Joyce WP, Dolan J, Sheahan K, Hyland
of the TME tale, then this ‘thank you’ is to you person-
JM. Pathological evidence in support of TME in the
ally and with great sincerity. You are the very stuff of management of rectal cancer. Br J Surg 1996; 83(8):
which the whole concept was constructed – painstak- 1112–5.
ing precise surgery combined with a passion for doing 14. Buchler MW, Heald RJ, Maurer CA, Ulrich B. Das
things better. Konzept Der Totalen Mesorektalen Exzision. Basel,
Karger, 1998.
15. Hermanek P, Wiebelt H, Staimmer D, et al. Prognostic
References factors of rectal carcinoma: experience of the German
Multicentre Study. Tumori 1995; 81: 60.
  1. Heald RJ, Husband EM, Ryall RD. The mesorectum in 16. Martling AL, Holm T, Rutqvist LE, Moran BJ, Heald RJ,
rectal cancer surgery: the clue to pelvic recurrence? Cedemark B. Effect of a surgical training programme on
Br J Surg 1982; 69: 613–6. outcome of rectal cancer in the county of Stockholm.
  2. Asher R. Richard Asher Talking Sense. London, Lancet 2000; 356: 93–6.
Pitman, 1972.
  3. Morgado P. Total mesorectal excision: a misnomer for
a sound surgical approach. Dis Colon Rectum 1998; Further reading
41: 120–1.
  4. Heald RJ. Towards fewer colostomies: the impact of Heald RJ, Leicester RJ. The low stapled anastomosis. Dis
circular stapling devices on the surgery of rectal can- Colon Rectum 1981; 24: 437–44.
cer in a district hospital. Br J Surg 1980; 67: 198–200. Karanjia ND, Corder AP, Bearn P, Heald RJ. Leakage from
  5. Heald RJ. The ‘holy plane’ of rectal surgery. J R Soc stapled low anastomosis after total mesorectal excision for
Med 1988; 81: 503–8. carcinoma of the rectum. Br J Surg 1994; 81: 1224–6.
  6. Heald RJ, Ryall RD. Recurrence and survival after total Schache D, Stebbing A, Heald RJ. Management of the
mesorectal excision for rectal cancer. Lancet 1986; 1: pelvic space following low anterior resection. Aus N Z
1479–82. J Surg 1989; 59: 339–42.

HEBK001-C01_p01-30.indd 30 11/02/13 3:44 PM


2
Anatomy of the rectum, anal canal
and pelvic floor
Thilo Wedel

Introduction relevant structures located in close topographi-


cal proximity to the rectum. Comparable to the
Undoubtedly, anatomy is the mother discipline nerve-sparing procedures introduced in thyroid
of all surgical interventions. This general notion gland surgery and radical prostatectomy, preser-
holds especially true for rectal cancer surgery, as vation of autonomic nerve plexi passing adjacent
many of the successful advances in surgical treat- to the rectum has become an important issue to
ment of rectal cancer have been derived from ana- improve postoperative outcomes with regard to
tomical considerations. In fact, revisited studies of urogenital functions. Thus, from the anatomical
the anatomy of the rectum, anal canal and pelvic point of view, rectal cancer surgery comprises at
floor have led to substantial procedural modifica- least a twofold challenge: oncologically curative
tions for both anterior rectal resection and ab- removal of the rectum along the mesorectal plane,
dominoperineal excision. and at the same time optimal preservation of the
One of the seminal anatomical contributions surrounding structures, in particular the nerves
with regard to rectal cancer surgery has been the responsible for urinary continence and sexual
clear description of the major routes of lymphatic function. This chapter focuses on the anatomy of
cancer spread. Since the introduction of total me- the organs to be removed and on the anatomy of
sorectal excision (TME), including the removal the structures to be preserved during rectal cancer
of the rectum together with its surrounding lym- surgery.
phatic vessels and lymph nodes, local recurrence With regard to the anatomical nomenclature,
rates have been considerably reduced. This con- one has to be aware that over the past few decades
ceptual change was advocated more than 100 years many different names have been given to the same
ago, exemplified by Berkeley Moynihan (1908), structures in the anatomical and surgical literature.
who stated that ‘we have not yet sufficiently real- These differing nomenclatures have led to ongoing
ised that the surgery of malignant disease is not confusion and, in some instances, controversies re-
the surgery of organs – it is the applied anatomy of garding the exact topography or even the existence
the lymphatic system.’ of a given anatomical structure. Irrespective of these
Another milestone set by anatomical consid- terminological issues, however, a general agreement
erations has been the preservation of functionally has been reached concerning the most relevant

HEBK001-C02_p31-51.indd 31 11/02/13 4:31 PM


32  Anatomy of the rectum, anal canal and pelvic floor

anatomical key landmarks required to meet the ex-


pectations of modern rectal surgery.

Embryology cloacal membrane


allantois

urorectal septum
cloaca
Development of the Rectum hindgut
and Anal Canal
The gastrointestinal tract is an endoderm-derived
structure passing through the body cavity as a primi-
tive gut. The primitive gut can be subdivided into
the foregut, midgut and hindgut, which are all con-
nected to the inner body wall by mesenteries serving
as access routes for blood and lymphatic vessels. The proctodaeum

hindgut includes the distal third of the transverse


colon, the descending and sigmoid colon, and at its
most distal end the rectum and upper anal canal.
The anorectal tube opens into a dilated pouch
termed the endodermal cloaca. Initially the cloaca
corresponds to a common cavity in the develop-
ment of both urogenital and gastrointestinal or-
gans. The Wolffian and Müllerian ducts and the
bladder with the allantois and ureters open into
its ventral portion and the anorectal tube opens urogenital membrane urinary bladder
into its dorsal portion. The cloaca is closed by the perineum urorectal septum
cloacal membrane formed by an inner endoder-
mal layer and an outer ectodermal layer. From anal membrane anorectal canal
outside, the cloacal membrane displays a depres-
sion lined by ectoderm and named proctodaeum
(anal groove). Figure 2.1. Embryonal development of the cloaca. The
cloacal cavity is divided by the primitive urorectal septum
At a later stage, the common endodermal
and separated during the seventh embryonal week into an
cloaca is divided by the primitive urorectal sep-
anterior part (urogenital sinus) and posterior part (anorectal
tum into an anterior part (urogenital sinus) and canal). (Reproduced from Schünke, M. et al., Prometheus
posterior part (anorectal canal) (Figure 2.1). Atlas of Anatomy, Vols. 1 and 2. Stuttgart, Germany,
The urorectal septum is composed of mesenchy- Thieme Publ. 2007/2009, with permission.)
mal tissue growing downwards to fuse with the
cloacal membrane, which is then also separated
into a urogenital and anal membrane. The fu- epithelium along the anorectal canal occurs in a
sion area between the urorectal septum and the craniocaudal direction, starting inside the anal
cloacal membrane corresponds to the primitive canal and proceeding externally.
perineum, which in turn will form the perineal
tendinous centre or perineal body. After degen- Relevance for Rectal Cancer Surgery
eration of the anal part of the cloacal membrane,
the upper anal canal derived from the endoderm The significance of embryological considerations for
is continuous with the lower anal canal derived rectal cancer surgery comes mainly from the observa-
from the ectoderm. Studies have shown that tion that the vascular and lymphatic supply of almost
epithelial differentiation from cylindrical single- the entire rectum is derived from visceral blood and
layered rectal epithelium to keratinized squamous lymphatic vessels travelling within the mesorectum.

HEBK001-C02_p31-51.indd 32 11/02/13 4:31 PM


RECTUM AND ANAL CANAL  33

This notion has been particularly emphasized by The recto-anal segment is 15–20 cm long and ex-
F. Stelzner, who subdivided vertebrates into two tends from the rectosigmoid junction adjacent to
separate organisms based on developmental studies, the promontorium down to the anal orifice. It cor-
namely into an inner ‘visceral individuum’ enveloped responds to the dorsal intrapelvic organ compart-
by an outer ‘somatic individuum’.1 In the visceral in- ment descending in front of the concave sacrum
dividuum, all organs are singular (e.g. stomach, liver, (sacral flexure) towards the anal hiatus of the pel-
spleen, intestines, rectum), with singular blood and vic floor. Due to the contraction of the puborectal
lymphatic vessels; in the somatic individuum, the or- sling, the anal canal is angled in a dorsal direction
gan units are bilaterally symmetric (e.g. two kidneys at the anorectal junction (perineal flexure), form-
and ureters, two gonadal systems, two levator ani ing an angle of 90–1008 with the rectum (anorectal
muscles, two semicircles of external anal sphincter). angulation) (Figure 2.2).
Accordingly, the blood and lymphatic supply of the
rectum including the upper anal canal is provided by
the visceral individuum (superior rectal artery, lym- Rectal Ampulla
phatic drainage towards the inferior mesenteric lymph
nodes), whereas the pelvic floor muscles including the The upper part of the rectum has the same diam-
external anal sphincter are supplied by bilateral sym- eter as the sigmoid colon (approximately 4–6 cm),
metric vessels (internal iliac and pudendal vessels) but the lower part of the rectum widens to form
derived from the somatic individuum. The border of the dilated rectal ampulla, with a diameter of up
these two embryologically different ‘individuals’ re- to 16 cm, depending on how full it is. The rectal
sides along the transition zone between the upper and wall differs morphologically from the colon by the
lower anal canal at the level of the dentate line. confluence of taeniae coli to a continuous longi-
Moreover, embryological studies have highlighted tudinal smooth muscle layer, permanent semilunar
the fact that not only the rectum but also its vascu- transverse folds, and the absence of fatty appendi-
lar and lymphatic supply are commonly ensheathed ces epiploica.
by a fascial system termed the mesorectum, which Most of the rectum is not covered by peritone-
remains clearly discernible in postnatal life. The me- um, and thus the rectum is predominantly an extra-
sorectal fascia represents the embryological delinea- or subperitoneal organ. Only the upper two-thirds
tion of the visceral from the somatic individuum con- of the ventrolateral rectal wall is directly related to
fining the main route of rectal cancer spread. Thus, the peritoneum, which reflects on to the bladder
precise surgical dissection along these embryologi- and seminal vesicles in males (rectovesical pouch)
cally defined planes will enable an oncologically safe and on to the uterus and posterior vaginal fornix in
removal of the specimen with minimal vascular and females (recto-uterine pouch, commonly known as
nerve damage. the pouch of Douglas).

RECTUM AND ANAL CANAL Anal Canal

The rectum and the anal canal are the last segments The rectal ampulla narrows at the level of the anorec-
of the gastrointestinal tract responsible for mediat- tal junction and becomes the anal canal (pars analis
ing stool continence and coordinating the defeca- recti). The anal canal extends down and backwards
tion process. Under normal conditions, the rectum to the anal orifice and is 2.5–4 cm long. The anal ca-
is empty and the anal canal is closed by means of nal is where the gastrointestinal tube of endodermal
the anal sphincter complex and the haemorrhoidal origin (‘visceral individuum’) fuses with the skin
plexus. Filling of the rectum by mass movements of ectodermal origin and the surrounding striated
from the descending and sigmoid colon initiates the pelvic floor and anal sphincter muscles (‘somatic
defecatory reflex, which induces relaxation of the individuum’). Thus, the anal canal represents the
anal sphincters, contraction of the rectal wall and topographical junction where the nerve, blood and
deflation of the haemorrhoidal cushions to allow lymphatic supplies of visceral and somatic structures
the passage of stool. merge and overlap. Accordingly, the anal canal is

HEBK001-C02_p31-51.indd 33 11/02/13 4:31 PM


34  Anatomy of the rectum, anal canal and pelvic floor

Figure 2.2. Rectum and anal


haemorrhoidal
external anal sphincter plexus canal. The frontal section shows
(deep part) internal anal sphincter
dentate line the inner relief of the rectal ampulla
(anal valves & crypts)
and anal canal, and cross-sections
external anal sphincter conjoint longitudinal
(superficial part) muscle of the haemorrhoidal plexus,
the internal and external anal
proctodeal glands
external anal sphincter anoderm sphincters and levator ani muscle.
(subcutaneous part) (Reproduced from Schünke, M.
et al., Prometheus Atlas
perianal skin intersphincteric groove
(anal verge) of Anatomy, Vols. 1 and 2.
Stuttgart, Germany, Thieme Publ.
2007/2009, with permission.)

divided into an upper and lower segment separated endodermal (cloacal) and ectodermal (proctodeal)
by a transitional zone at the dentate line. parts of the anal canal.
The lower segment of the anal canal extends from
Segments of the anal canal the dentate line to the anocutaneous line and cor-
The upper segment of the anal canal extends from responds to the anoderm lined by a non-keratinized
the anorectal junction, defined by the superior bor- stratified squamous epithelium (zona squamosa).
der of the external anal sphincter (anorectal ring) to The anodermal skin is devoid of glands and hairs but
the transitional zone. The inner lining consists of a richly equipped with sensory somatic nerve endings,
pink-coloured mucosa with single-layered cylindri- making it very sensitive to touch, pain and tempera-
cal epithelium (zona colorectalis). ture. At the lower end of the anal canal, an anal inter-
At the transitional zone (zona transitionalis), sphincteric groove (anal verge) is palpable between
the wet mucosa changes into a dry stratified sq- the bulge of the internal and external anal sphincters.
uamous non-keratinized epithelium displaying Whereas the ‘surgical’ anal canal is defined by the en-
a ­histological mosaic of cylindrical, cubic and tire segments from the anal ring to the anal verge,
flat epithelial cells. The transitional zone exhibits the ‘anatomical’ anal canal is confined to the lower
8–12 readily discernible vertical anal columns segment and involves only the anoderm.
(Morgagni) containing terminal branches of the su- At the anal orifice, the anodermal skin changes
perior rectal artery. Between these anal columns ex- into true skin. The hairless perianal skin is of a dull
tend anal sinuses that form pocket-like folds at their brown colour and displays radial folds caused by
lower ends, the anal valves or crypts. The circular the contraction of the corrugator ani muscle. The
line of alternating anal columns and anal valves cor- skin contains sweat, sebaceous and apocrine glands
responds to the dentate line (pectinate line, crypt and is supplied by perianal blood vessels originat-
line) considered to be the junction between the ing from the inferior rectal artery.

HEBK001-C02_p31-51.indd 34 11/02/13 4:31 PM


RECTUM AND ANAL CANAL  35

Haemorrhoidal plexus ating the intersphincteric groove (anal verge). Due to


The haemorrhoidal plexus corresponds to a vascular its permanent involuntary contraction, the internal
network extending within the submucosa of the up- anal sphincter is readily palpable as a rigid cylinder, in
per anal canal best described as corpus cavernosum particular when the striated external anal sphincter is
recti (anulus haemorrhoidalis, glomera venosa haem- completely relaxed (e.g. under anaesthesia).
orrhoidalia). The submucosal vascular plexus is char- The longitudinal muscle layer of the rectal wall
acterized by arteriovenous anastomoses and dilated also continues distally into the anal sphincter com-
veins due to sphincter-like constrictions that provide a plex. Smooth muscle bundles descend between the
taut elastic apposition of the anal columns and thus an internal and external anal sphincter, towards the
air- and liquid-tight closure of the anal canal. perianal region, and are joined by striated muscle
The haemorrhoidal plexus is supplied by branch- fibres from the puborectal muscle (conjoint longi-
es from the superior rectal artery, which reach the tudinal muscle). Distally, the muscle fibres diverge,
corpus cavernosum recti from the right (7 and 11 become fibro-elastic and insert into the perianal
o’clock, lithotomy position) and left (3 o’clock) skin producing radial wrinkles (corrugator ani
sides. The blood is drained by veins penetrating muscle). The most peripheral muscular septa radi-
through the internal anal sphincter and collected in ate outwards and pass between the subcutaneous
the external rectal venous plexus. Due to the trans- and superficial parts of the external anal sphinc-
sphincteric blood drainage, the degree of filling of ter into the ischioanal space. These fibres insert in
the haemorrhoidal plexus is determined primarily the superficial perineal fascia and contribute to the
by the contraction of the internal anal sphincter. separation of the ischioanal space from the subcu-
Normally the draining veins are compressed by the taneous perianal space.
resting tone of the internal anal sphincter, result-
External anal sphincter
ing in a physiological cushion-like swelling of the
haemorrhoidal plexus. Relaxation of the internal The external anal sphincter surrounds the internal anal
anal sphincter during defecation allows proper sphincter, but both muscles are separated by connec-
emptying and down-sizing of the haemorrhoidal tive tissue (intersphincteric plane). The external anal
plexus for the passage of stool. sphincter forms an elliptical cylinder approximately
15 mm thick and is divided by septa into three parts.
Although the external anal sphincter is a striated skel-
Anal Sphincter Complex etal muscle, its fibres are composed mainly of slow-
twitch type I fibres mediating prolonged contractions
The anal sphincter complex is composed of two suitable for maintaining an adequate basal tone.
concentric muscles: the internal anal sphincter The deep part of the external anal sphincter is the
composed of smooth musculature derived from thickest and most cranially located segment delineat-
the rectal wall, and the external anal sphincter ing the anorectal ring. Its fibres blend with the pub-
composed of striated musculature, which is in- orectal muscle and are not attached posteriorly to the
timately fused with the surrounding muscles of coccyx, and so this muscular portion is able to follow
the pelvic floor. the contraction of the puborectal muscle anteriorly
to create the anorectal angle. The superficial part is
Internal anal sphincter attached firmly to the perineal tendinous centre an-
The internal anal sphincter consists of circular teriorly and to the anococcygeal ligament posteriorly
smooth muscle bundles and corresponds to a distal and is therefore elliptical in shape. The subcutaneous
thickening of the circular muscle layer of the rectal part circumscribes the anal orifice and extends deep
wall. The ring-shaped muscle is 5–8 mm thick and to the skin below the lower border of the internal anal
2–3 cm long. In relation to the anal canal, the inter- sphincter. Smooth muscle bundles from the conjoint
nal anal sphincter extends from the anorectal junc- longitudinal muscle intermingle with striated mus-
tion down to the anocutaneous line, with the most cle fibres of the external anal sphincter. In males, the
prominent part projecting on to the anoderm. The three parts of the external anal sphincter are of equal
subcutaneous part of the external anal sphincter dimension along the circumference; in females, the
overlies the lower part of the internal sphincter, cre- external anal sphincter muscle is reduced anteriorly

HEBK001-C02_p31-51.indd 35 11/02/13 4:31 PM


36  Anatomy of the rectum, anal canal and pelvic floor

to about one-third of its posterior thickness, in par- Initial attempts to describe the perirectal fas-
ticular due to its less developed deep part. cial anatomy have led to differing nomenclatures
and controversies regarding the exact topography
of those fascial structures enveloping the rectum.
Perirectal Fascia and Spaces Macroscopic dissection studies on fixed and fresh
specimens, histological and immunohistochemi-
Comprehensive understanding of the topographi- cal studies and subtle intraoperative observations
cal anatomy of fascia and spaces that surround the have shown, however, that the system of intrapelvic
rectum is of utmost importance and an essential fascial layers allows clear identification of perirectal
prerequisite for rectal cancer surgery. Since the in- spaces suitable for proper and safe mobilization of
troduction of the TME procedure, it has become ob- the rectum (Figure 2.3).
vious that successful surgery depends first and fore-
most on mobilizing the rectum along the correct Rectal fascia/mesorectum
planes to achieve both oncologically safe margins The inner surface of the pelvic wall, including the pel-
and optimal preservation of urogenital functions. vic muscles (internal obturator, levator ani, coccygeal

middle rectal
peritoneum artery

inferior hypogastric
plexus
rectal
presacral
fascia
fascia

pelvic splanchnic
nerves

superior rectal hypogastric


artery nerve

retrorectal Figure 2.3. Perirectal


space spaces, mediosagittal
section of male pelvis,
presacral space
with presacral vessels
left view. The rectal
fascia and presacral
fascia are highlighted to
illustrate the perirectal
mesorectum with tissue (mesorectum)
lymphatic vessels
and both the retrorectal
and presacral spaces.
Note that the retrorectal
rectoprostatic septum rectosacral fascia
(Denonvillier´s fasica)
,
(Waldeyer s fascia)
space is free of blood
vessels and nerves and
corresponds to the
anococcygeal correct surgical plane
ligament
for mobilization of the
ischioanal space rectum during total
mesorectal excision.
perineal body (Reproduced from
Schünke, M.
et al., Prometheus
Atlas of Anatomy, Vols.
1 and 2. Stuttgart,
Germany, Thieme
Publ. 2007/2009, with
permission.)

HEBK001-C02_p31-51.indd 36 11/02/13 4:31 PM


RECTUM AND ANAL CANAL  37

and piriformis muscles) are covered by the parietal the branches of the superior rectal artery and the
pelvic fascia. The visceral pelvic fascia (endopelvic perirectal lymph nodes and vessels draining in a
fascia) ensheathes the pelvic organs, including the cranial direction towards the inferior mesenteric
rectum. In contrast to urogenital pelvic organs, the lymph nodes.
rectum exhibits an almost completely closed annular The perirectal tissue enclosed by the mesorectum
envelope called the rectal fascia (‘proper’ perirectal is most developed at the dorsal aspect of the rectal
fascia, fascia propria recti). The rectal fascia corre- wall, producing two bulges (‘mesorectal cheeks’).
sponds to a rather thin connective tissue sheath and Anteriorly, the mesorectum border is the rectogeni-
constitutes a morphological barrier, thereby prevent- tal septum (Denonvilliers’ fascia), where the per-
ing early penetration of rectal cancer. For this reason, irectal tissue is much thinner. The dorsal and ven-
it has also been described by Stelzner as a ‘delimiting’ tral parts of the mesorectum resemble a continuous
rectal lamellae (‘rektale Grenzlamellen’).1 plane, but laterally the rectal fascia is not completely
An alternative, clinically widely used term for closed. At these sites the annular pattern of the me-
the rectal fascia is ‘mesorectal’, introduced by sur- sorectum is interrupted bilaterally due to the re-
geons to describe the complete removal of the flection of the rectal fascia towards the pelvic wall
perirectal tissue together with the rectum during (lateral rectal ligaments, rectal pedicles) to give ac-
TME. This surgical concept is based on the ob- cess for blood vessels and autonomic nerves passing
servation that the mesorectum – like the true me- from the inferior hypogastric plexus to the rectal
senteries of the small and large bowel – ensheathes wall. As a consequence, surgical mobilization of the
the tissues harbouring the major routes of blood rectum along the mesorectum requires sharp dissec-
vessel supply and lymphatic drainage of the corre- tion laterally, while posteriorly and anteriorly it can
sponding bowel segment. In fact, the mesorectum be achieved by making use of ‘self-opening’ surgical
surrounds the fatty perirectal tissue that contains planes (Figure 2.4).

ureter
(duplex)

superior hypogastric
plexus

ovarian superior rectal


vessels presacral
fascia artery

presacral space retrorectal


with presacral vessels space

hypogastric rectal
nerve fascia

mesorectum

Figure 2.4. Perirectal spaces, dissected female pelvis, cranial view. The right forceps is inserted into the rectum and pushed
forward to illustrate the rectal fascia that surrounds the perirectal tissue (mesorectum) and contains the superior rectal artery
(red vessel loop). The left forceps grasps the presacral fascia that separates the retrorectal space from the presacral space.
Note that the superior hypogastric plexus (yellow strip) and hypogastric nerves are embedded into and extend along the
presacral fascia. (Reproduced from Schünke, M. et al., Prometheus Atlas of Anatomy, Vols. 1 and 2. Stuttgart, Germany,
Thieme Publ. 2007/2009, with permission.)

HEBK001-C02_p31-51.indd 37 11/02/13 4:31 PM


38  Anatomy of the rectum, anal canal and pelvic floor

superior rectal
artery

pelvic splanchnic presacral space


nerves with presacral vessels
presacral
hypogastric
fascia
nerve
retrorectal
inferior hypogastric space
plexus

rectal
fascia
middle rectal
artery T-junction
mesorectum with
lymphatic vessels

neurovascular rectoprostatic septum


bundles (Denonvillier´s fasica)

seminal vesicles

prostate

urinary bladder

Figure 2.5. Perirectal spaces, transverse section of male pelvis, cranial view. The rectal fascia and presacral fascia are highlighted
to illustrate the perirectal tissue (mesorectum) and both the retrorectal and presacral spaces. Anteriorly the rectal fascia is
bordered by the rectoprostatic septum (Denonvillier’s fascia). Laterally the rectal fascia is pierced on both sides by rectal nerves
diverging from the inferior hypogastric plexus and by small vessels of the middle rectal artery (rectal pedicles, T-junction). Nerves
from the inferior hypogastric plexus continue in an anterior direction as neurovascular bundles to supply the urinary bladder,
prostate and seminal vesicles.

Lateral rectal ligaments/rectal pedicles mentioned above, for complete mobilization of the
On both sides, the rectum is loosely connected to rectum, the T-junction has to be sharply divided on
the pelvic wall by condensed connective tissue, both sides at the level of the rectal pedicles. Care
traditionally described as lateral rectal ligaments. must be taken, however, to stay rather close to the
Originally these were considered to function as rectum to avoid damage to the inferior hypogastric
true ligamentous structures to support the rectal plexus, thereby preserving sexual function and uri-
ampulla above the pelvic floor. From anatomical nary continence (Figure 2.5).
and developmental studies, however, it has become
obvious that these ligaments do not provide sub- Presacral fascia
stantial mechanical fixation but serve primarily as Behind the rectal fascia (posterior mesorectum)
access routes for the vascular and nerve supply of extends another fascial envelope following the con-
the rectum, leading to alternative and more appro- cavity of the sacrum, the presacral fascia. The pre-
priate terms, such as rectal pedicles, rectal stalks sacral fascia is part of the parietal pelvic fascia that
and ‘paraproctial’ compartment. covers the periosteal surface of the sacrum dorsally
The rectal pedicles contain small-sized branches and reflects laterally towards the rectogenital sep-
from the middle rectal arteries (if present) and the tum (Denonvilliers’ fascia). The rectal fascia and
autonomic nerve fibre branches (rectal nerves) that presacral fascia are clearly separated from each oth-
originate from the dorsolaterally located inferior er by the retrorectal space. When descending down
hypogastric plexus and traverse medially to enter the to the pelvic floor, however, approximately at the
rectal wall. This connection between the rectal wall level of the fourth sacral vertebra, the rectal fascia
and the parietal pelvic fascia harbouring the inferior fuses with the presacral fascia. This connective tis-
hypogastric plexus is also called the T-junction. As sue bridge between the rectal and presacral fascia

HEBK001-C02_p31-51.indd 38 11/02/13 4:31 PM


RECTUM AND ANAL CANAL  39

hypogastric
nerve

pelvic splanchnic
nerves

inferior hypogastric
plexus

neurovascular
bundles middle rectal
artery

urinary bladder
T-junction presacral
fascia
seminal vesicles

prostate

rectal
fascia

rectoprostatic septum mesorectum superior rectal


,
(Denonvillier s fascia) artery

Figure 2.6. Perirectal spaces, sagittal section of male pelvis, left view. The rectum and the surrounding mesorectum are
pushed towards the contralateral side to illustrate the course of the autonomic pelvic nerves along the pelvic wall. The
hypogastric nerve and the pelvic splanchnic nerves join the extensive network of the inferior hypogastric plexus. Rectal
nerves leave the inferior hypogastric plexus to pierce the rectal fascia and enter the rectal wall (rectal pedicle, T-junction).
The remaining nerves of the inferior hypogastric plexus extend in an anterior direction as neurovascular bundles to supply
the urinary bladder, prostate and seminal vesicles. (Reproduced from Schünke, M. et al., Prometheus Atlas of Anatomy,
Vols. 1 and 2. Stuttgart, Germany, Thieme Publ. 2007/2009, with permission.)

can be quite thick and is also termed the rectosacral between the iliac vessels on both sides and deline-
fascia or Waldeyer’s fascia. ates the presacral space. Laterally it is pierced by
The presacral fascia is an important surgical pelvic splanchnic nerves that originate from sacral
landmark, as it separates the retrorectal space (sur- spinal nerves and follow along the presacral fascia
gical plane for a TME procedure) from the presac- to join the hypogastric nerves to form the inferior
ral space and it is intimately associated with the hypogastric plexus (Figure 2.6).
hypogastric and pelvic splanchnic nerves that form
the inferior hypogastric plexus. The fascial archi- Rectogenital septum
tecture displays an inner and outer lamella, giving Between the genital organs (vagina in females, pros-
the presacral fascia the aspect of a double-layered tate and seminal vesicles in males) and the rectum
sheath. The inner lamella borders the posterior extends a fascial structure of considerable density
mesorectum, confining the retrorectal space, and originally described by Denonvilliers as ‘aponévrose
contains the autonomic nerve plexus. For this rea- pubio-rectale’,2 which is now recognized as the rec-
son, it has also been called the hypogastric sheath togenital septum. In females the rectovaginal septum
or the pre-hypogastric nerve fascia. Further an- is related to the posterior vaginal wall; in males the
teriorly, the presacral fascia reflects at the level of rectoprostatic septum (Denonvilliers’ fascia) covers
the rectal pedicles and extends medially towards the prostate, seminal vesicles and the most distal
the rectogenital septum to fuse with the prostatic segments of the vasa deferens and ureters, separat-
capsule in the male. The outer lamella extends ing them from the anterior rectal wall.

HEBK001-C02_p31-51.indd 39 11/02/13 4:31 PM


40  Anatomy of the rectum, anal canal and pelvic floor

Although intraoperatively the rectogenital septum narrows and ends at the level of the fourth sacral ver-
seems to comprise a single layer, subtle macroscopic tebra, where the rectal fascia fuses with the presacral
dissections and histological studies have described fascia (rectosacral fascia).
a posterior sheath being the anterolateral lining of
the mesorectum and an anterior sheath adjacent Presacral space
to the prostate. The rectoprostatic septum is clearly The presacral space extends between the presac-
identifiable as a fascial structure at the level of the ral fascia and the concave periosteal surface of the
seminal vesicles and upper half of the prostate, but sacrum. This space is by no means an avascular or
it often joins the prostatic capsule at the lower half nerve-free area, as it contains the medial and lateral
of the prostate and the apex, making an unequivo- sacral arteries, the presacral venous plexus and the
cal identification more difficult. The rectogenital parasympathetic pelvic splanchnic nerves originat-
septum continues laterally to the presacral fascia, in ing from the third and fourth sacral nerves. Thus,
particular to its inner lamella containing the inferior the presacral space corresponds to a ‘forbidden
hypogastric nerve plexus. The rectogenital septum plane’ during the TME procedure, as surgical dis-
is composed of condensed connective tissue and section may lead to nerve injury and, in particular,
smooth muscle cells and is generally well developed. troublesome venous bleeding. When starting dorsal
It is subject, however, to morphological alterations mobilization of the rectum, it is essential to clearly
(e.g. weakening, thinning) with increasing age or dis- identify the proper plane for TME and not to mis-
eases of adjacent organs (e.g. rectocele or enterocele take the presacral space for the retrorectal space.
in females, benign prostatic hyperplasia in males).
Its surgical significance derives from the obser-
vation that multiple nerve branches of the inferior Blood Supply of the Rectum and
hypogastric plexus pass lateral to and in front of the Anal Canal
rectogenital septum. These autonomic nerve fibres
approach the seminal vesicles, vasa deferens, distal As the rectum derives from the hindgut, the rectum,
ureters and prostate dorsolaterally and comprise the including the upper anal canal, is supplied mainly
so-called neurovascular bundles responsible for uri- by the inferior mesenteric artery via the superior
nary continence and sexual functions. Consequently, rectal artery (Figure 2.7). The middle and inferior
this bilateral nerve network is at risk during surgical rectal arteries originate from the internal iliac ves-
mobilization of the anterior rectal wall, in particular sels and contribute to the blood supply of the lower
if the surgical plane is opened not behind but in front anal canal and minor parts of the rectum via intra-
of the rectoprostatic septum with direct exposure of mural anastomoses. Occasionally, the posterior part
the seminal vesicles and prostate. of the anal canal and internal anal sphincter are ad-
ditionally supplied by the median sacral artery.
Retrorectal space
The retrorectal space can be considered as a fascial in- Superior rectal artery
terface extending between the rectal fascia (posterior The superior rectal artery corresponds to the direct
mesorectum) anteriorly and the presacral fascia poste- continuation of the inferior mesenteric artery af-
riorly. This distensible space corresponds to the prop- ter the sigmoid arteries have diverged to supply the
er plane for dorsal mobilization of the rectum which sigmoid mesocolon. The superior rectal artery is of
can be achieved by means of gentle traction and coun- considerable size (diameter 3.0 6 1.1 mm) and con-
ter-traction. Opening of the retrorectal space reveals tributes more than 80 per cent to the entire rectal
loose areolar connective tissue, which has also been blood supply. It approaches the dorsal aspect of the
described quite illustratively as ‘angel’s hair’ (‘cheveux rectal wall by entering the perirectal tissue completely
des anges’) due to its delicate web-like appearance. wrapped by the mesorectal fascia. On its course within
As the retrorectal space corresponds to an avas- the perirectal tissue, the main trunk generally divides
cular and nerve-free area, careful dissection will not into three branches that surround the posterolateral
result in major bleeding or nerve injury. Occasion- wall of the rectum. The branches ramify further, en-
ally blood vessels originating from the presacral ve- ter the rectal muscle layers and the submucosa, and
nous plexus pass through the fascial interface. When descend towards the upper segment of the anal canal.
­approaching the pelvic floor, the retrorectal space Terminal branches supply the haemorrhoidal plexus

HEBK001-C02_p31-51.indd 40 11/02/13 4:31 PM


RECTUM AND ANAL CANAL  41

inferior mesenteric sources of rectal blood supply are essential for effec-
artery median sacral
artery
tive wound healing of deep rectal anastomosis after
removal of the rectum and its major blood supply,
the superior rectal artery.
superior rectal
artery
Lymphatic Drainage of the
Rectum and Anal Canal
internal iliac
artery
Analogous to the blood supply, the lymphatic drain-
internal pudend age from the rectum is mainly in a cranial direction
artery and to visceral lymph nodes. Intramural lymphatic
middle rectal vessels project to perirectal lymph nodes located
artery
within the fatty perirectal tissue, which is completely
inferior rectal enveloped by the mesorectal fascia. Lymphatic col-
artery lection and drainage occur unidirectionally towards
Figure 2.7. Arteries of the rectum and anal canal, dorsal the major lymph node stations along the superior
view. The rectal wall, including the upper anal canal, is rectal and the inferior mesenteric blood vessels.
supplied mainly by the superior rectal artery derived from Depending on the degree of individual develop-
the inferior mesenteric artery. The middle and inferior ment of the lymphatic vessels, lymphatic drainage
rectal arteries originate from the internal iliac vessels and may also take place via the rectal pedicles towards
contribute to the blood supply of the lower anal canal. the internal iliac lymph nodes. In rectal cancer, this
(Reproduced from Schünke, M. et al., Prometheus Atlas of
Anatomy, Vols. 1 and 2. Stuttgart, Germany, Thieme Publ.
route of lymphatic spread is generally confined to
2007/2009, with permission.) advanced tumour stages in which the rectal fas-
cia has been penetrated towards the lateral pelvic
wall. The lower anal canal and the perianal region
(corpus cavernosum recti), creating haemorrhoidal are not hindgut derivates and therefore receive a
cushions at predilected areas (3, 7 and 11 o’clock with somatic rather than a visceral lymphatic drainage.
the patient in the lithotomy position). Lymphatic vessels project mainly to inguinal and
external iliac lymph nodes.
Middle rectal arteries
The middle rectal arteries originate from the in- Nerve Supply of the Rectum and
ternal iliac arteries mostly as direct branches. They Anal Canal
pass above the pelvic floor muscles and approach
the inferior rectum via the rectal pedicles. Thus, the Like the other segments of the gastrointestinal tract,
middle rectal arteries intermingle with rectal nerve the distal end comprising the rectal ampulla and up-
fibres that originate from the inferior hypogastric per anal canal, including the internal anal sphincter, is
plexus and also run within the rectal pedicles (T- controlled by the autonomic nervous system. In con-
junction). In contrast to the superior rectal artery, trast, the somatic nerve supply is confined to the stri-
however, the middle rectal arteries are much smaller ated musculature of the external anal sphincter and to
in size and inconstantly developed and are present the lower anal canal up to the dentate line (Figure 2.8).
bilaterally in only about 10 per cent of individuals. Inside the rectal wall resides the enteric nervous
system, which extends throughout the muscular,
Inferior rectal arteries submucosal and mucosal layers by forming intramu-
The lower anal canal and the anal sphincter complex ral nerve plexi. The myenteric and submucosal plexi
are supplied by anal arteries from the inferior rectal are composed of ganglia and interconnecting nerve
arteries. They diverge from the internal pudendal ar- fibre bundles, which regulate most of the intestinal
teries located within the Alcock canal and approach functions (e.g. motility, perception, secretion, ab-
the anal region via the ischioanal space, dividing into sorption, immune functions). The enteric nervous
ventral and dorsal branches. Functional intramural system is linked to the central nervous system by
anastomoses are established between the inferior, spinal visceral afferents and both sympathetic and
middle and superior rectal arteries. These multiple parasympathetic nerve fibres and ganglia.

HEBK001-C02_p31-51.indd 41 11/02/13 4:31 PM


42  Anatomy of the rectum, anal canal and pelvic floor

superior
hypogastric plexus

hypogastric
S1 nerve

S2
inferior
S3
hypogastric plexus
S4
pelvic splanchnic
nerves
levator nerves

pudendal nerve rectal plexus

rectal wall

external anal
sphincter
internal anal
sphincter

Figure 2.8. Somatic nerve supply of pelvic floor muscles (left side) and autonomic nerve supply of the anorectum (right
side), schematic drawing. Somatic nerves derive from sacral spinal nerves (S2–4), release direct nerve branches (levator
nerves) to the levator ani muscle, and form the pudendal nerve to innervate the external anal sphincter and the perineal
region. Autonomic nerves are composed of lumbar sympathetic nerves (superior hypogastric plexus, hypogastric nerves)
and parasympathetic nerves (pelvic splanchnic nerves), which fuse in the inferior hypogastric plexus responsible for the
nerve supply of anorectum, including the internal anal sphincter. (Reproduced from Schünke, M. et al., Prometheus Atlas
of Anatomy, Vols. 1 and 2. Stuttgart, Germany, Thieme Publ. 2007/2009, with permission.)

It is important to emphasize that sympathetic cavernous bodies (erection in males, swelling of the
and parasympathetic nerves supplying the rectum clitoris in females) and for lubrification of the exter-
originate from an extensive nervous network that nal genital organs in both genders. Sexual dysfunc-
is also responsible for the control of urinary con- tion due to nerve injury is clinically more evident
tinence and mediation of sexual functions. As the in male patients, manifesting as sexual impotence
rectum resides in close proximity to the autonomic characterized by erectile dysfunction and impaired
nerve fibre meshes extending towards the urogeni- ejaculation. The effects and impact of nerve injury
tal organs, these nerves are in danger of damage in female patients, such as inability to experience
during surgical removal of the rectal specimen. orgasm and insufficient lubrication, should not be
Sympathetic nerves support the filling phase of underestimated, however.
the urinary bladder under resting conditions by re- The extent of autonomic nerve injury that results
laxing the detrusor vesicae muscle and closing the in definite postoperative impairment of urinary con-
internal urethral sphincter, thereby contributing to tinence and sexual functions is unclear. In general, the
urinary continence. For controlled micturition, par- autonomic innervation of organs is characterized by a
asympathetic nerves suppress the sympathetic input certain functional redundancy, which is anatomically
and initiate continuous contraction of the detrusor reflected by a bilateral organization of abundantly
vesicae muscle. Thus, injury of these nerves may ramifying intrapelvic nerve plexi and collateral nerve
lead to both urinary incontinence and urinary blad- fibres connecting both sides with each other. Thus,
der dysfunction, including retrograde ejaculation. it has been suggested that unilateral damage may
Both sympathetic and parasympathetic nerves are be compensated by the intact contralateral nervous
also involved in regulating sexual functions. Sym- input, although sexual dysfunction and urinary conti-
pathetic nerves mediate ejaculation, and parasym- nence have also been described after partial damage of
pathetic nerves are responsible for the filling of the autonomic nerves (Figures 2.9 and 2.10).

HEBK001-C02_p31-51.indd 42 11/02/13 4:31 PM


RECTUM AND ANAL CANAL  43

Figure 2.9. Autonomic and somatic


nerves of the pelvis, sagittal section of
male pelvis, left view. The hypogastric
nerves diverge from the superior
hypogastric plexus and extend
laterally to the rectum into the inferior
hypogastric plexus, which is joined by
superior
the pelvic splanchnic nerves. Rectal
hypogastric plexus
nerves leave the inferior hypogastric
plexus to enter the rectal wall (rectal hypogastric
plexus), while the remaining nerve nerve
fibres extend further anteriorly as
neurovascular bundles towards the pelvic splanchnic
urinary bladder, prostate and seminal nerves
vesicles. The neurovascular bundles
include the cavernous nerves, which inferior
enter the cavernous bodies. The hypogastric plexus
pudendal nerve extends below the neurovascular rectal plexus
levator ani muscle and provides the bundles pudendal nerve
somatic nerve supply of the striated cavernous nerves
levator ani muscle
pelvic floor muscles and the skin of
perineal nerves
the perineal region and external genital
dorsal penile nerve inferior rectal nerves
organs. (Reproduced from Schünke,
M. et al., Prometheus Atlas of Anatomy, external anal
Vols. 1 and 2. Stuttgart, Germany, sphincter
Thieme Publ. 2007/2009, with
permission.)

Figure 2.10. Perirectal


spaces, dissected superior hypogastric
pelvis, cranial view. plexus
superior rectal
The rectum is retracted
artery
together with the rectal
fascia to illustrate the
retrorectal space and presacral space
the presacral space with presacral vessels
(forceps inserted). Note
that the presacral fascia
hypogastric
is removed to expose nerve
the autonomic nerves
(superior hypogastric
plexus, hypogastric
inferior
nerves) embedded rectal hypogastric plexus
within this fascia. From fascia
the inferior hypogastric
plexus diverge nerve
fibres via the rectal T-junction
pedicles (T-junction) into
the rectal wall.

HEBK001-C02_p31-51.indd 43 11/02/13 4:32 PM


44  Anatomy of the rectum, anal canal and pelvic floor

Sympathetic nerves nating from the left and right hypogastric nerves
Lumbar sympathetic nerves pass along and in front and parasympathetic nerves originating from pel-
of the descending aorta and form fairly well-defined vic splanchnic nerves (Figure 2.11). The inferior
periarterial nervous networks, the inferior mesenter- hypogastric plexus is embedded in connective tissue
ic and superior hypogastric plexus. These networks of the parietal pelvic fascia. At the level of the rec-
enclose the inferior mesenteric artery up to 5 cm tal pedicles, small branches diverge from the main
from its origin from the aorta. A high-tie arterial li- nerve plexus and enter the rectal wall (T-junction).
gation is virtually impossible without damaging, at The inferior hypogastric plexus extends further in
least in part, adjacent sympathetic nerve fibres. Thus, an anterior direction as a fan-like network radiat-
when disconnection of the rectal blood supply is car- ing cranially as high as the seminal vesicles, distal
ried out, a low-tie ligation of the inferior mesenteric ureters and vasa deferens and caudally towards the
artery is generally preferred to avoid impairment of apex of the prostate and the upper aspect of the
both urinary continence and sexual functions at this perineal body (Figure 2.12).
initial surgical step. The lower part of the inferior hypogastric plexus
When the superior hypogastric plexus has en- has been described by Walsh and colleagues as neu-
tered the pelvic cavity in front of the promonto- rovascular bundles extending dorsolaterally along
rium, it divides into the left and right hypogas- the prostate to approach the apex of the prostate
tric nerve. The hypogastric nerve does not always and penetrate the urogenital diaphragm.3 The neu-
present as a single clearly defined nerve; often it rovascular bundles contain the cavernous nerves re-
consists of different nerve fibre bundles arranged sponsible for erectile functions and terminal nerve
in parallel oriented intermingling strings. Both fibres innervating the internal urethral sphincter.
hypogastric nerves are attached to the inner lamella Thus, special attention should be given to this lower
of the presacral fascia corresponding to the parietal portion of the inferior hypogastric plexus, as these
pelvic fascia extending in front of the sacral con- nerve fibres are closely related to the anterior rectal
cavity. Due to the intimate relationship between wall, before they continue along the transverse peri-
the hypogastric nerves and the presacral fascia, this neal muscles into the penile bulb. The same holds
fascial layer has also been termed the hypogastric true in female patients in whom the lowermost por-
sheath or the pre-hypogastric nerve fascia. Gentle tion of the inferior hypogastric plexus runs along
traction on the hypogastric nerves will lift this fas- the lower lateral wall of the vagina.
cial sheath, helping to visualize the course of the The upper part of the inferior hypogastric plexus
nerves down to the inferior hypogastric plexus. extending along the prostate and seminal vesicles
is separated from the rectum by the rectoprostatic
Parasympathetic nerves septum (Denonvilliers’ fascia) (Figure 2.13). As long
Parasympathetic nerves derive from the sacral por- as the anterior surgical dissection plane is behind
tion of the parasympathetic nervous system located the rectoprostatic septum, preservation of these
within the sacral spinal cord. Together with thick nerves is feasible. Some communicating nerve fibres
ventral branches of the sacral spinal nerves (S2–4), between the left and right inferior hypogastric plexus
they leave throughout the ventral sacral foramina pass and cross in front of the rectoprostatic septum.
as so-called pelvic splanchnic nerves. Due to one This exchange of nerve fibres may explain why uni-
of their main functions, they are also termed eri- lateral lesions of the inferior hypogastric plexus may
gent nerves or erigent pillars. The pelvic splanch- be compensated by the intact contralateral plexus.
nic nerves pierce the presacral fascia at the left and Despite the presence of autonomic nerve fibres at
right sides and follow this fascial plane to join the the midline, this area is still the favoured approach to
inferior hypogastric plexus, where they intermingle initiate anterior mobilization of the rectum, because
with their sympathetic counterparts. nerve fibre density constantly increases towards the
lateral sides of the rectoprostatic septum.
Inferior hypogastric plexus/pelvic plexus
The inferior hypogastric plexus, also termed the Somatic nerves
pelvic plexus, is an extensive network of autonomic The autonomic nerve supply of the rectum and anal
nerve fibres composed of sympathetic nerves origi- canal ends at the level of the dentate line. Below this

HEBK001-C02_p31-51.indd 44 11/02/13 4:32 PM


RECTUM AND ANAL CANAL  45

superior hypogastric
plexus
hypogastric
nerve

pelvic splanchnic
nerves vas deferens
Figure 2.11. Dissected left male
hemipelvis with pelvic organs,
right anterior view. The levator ani
urinary bladder
muscle is pulled back to expose the
course of the autonomic nerves. inferior seminal vesicle
The hypogastric nerve diverges hypogastric plexus
from the superior hypogastric
plexus and extends laterally to the rectal plexus
prostate
rectum into the inferior hypogastric
plexus, which is joined by the pelvic
cavernous nerves
splanchnic nerves. Rectal nerves levator nerves
leave the inferior hypogastric plexus rectum
to enter the rectal wall, while the penile bulb
levator ani
remaining nerve fibres extend
muscle urethra
further anteriorly as neurovascular
bundles towards the urinary
bladder, seminal vesicles, prostate
and cavernous bodies.

embryologically defined border between the gas- tor functions. The lower anal canal comprising the
trointestinal tract (‘visceral individuum’) and the anoderm and perianal region is supplied by perianal
pelvic floor, including the perineal region and the branches of the pudendal nerves. In contrast to the
external anal sphincter (‘somatic individuum’), so- autonomically innervated rectum, the anodermal
matic nerves are in charge for both sensory and mo- segment is highly sensitive to touch, pressure, pain

Figure 2.12. Dissected right male


hemipelvis with pelvic organs partly ureter
reflected, left view. The entire rectum
together with the intact mesorectum urinary bladder
is mobilized and pulled out of the seminal vesicle
pelvic cavity, attached only to hypogastric
the perineal body anteriorly. This prostate nerve
procedure allows optimal exposure
of the right hypogastric nerve, pelvic parietal pelvic fascia
splanchnic nerves and the extensive
nervous network of the inferior inferior
hypogastric plexus
hypogastric plexus (green strips). neurovascular
Nerve fibres of the inferior hypogastric bundle
plexus extend towards the ureter, perineal body pelvic splanchnic
seminal vesicle (yellow strip), urinary nerves
bladder and prostate, and down to
anterior
the cavernous bodies (red vessel
mesorectum
loop) as neurovascular bundles.

HEBK001-C02_p31-51.indd 45 11/02/13 6:38 PM


46  Anatomy of the rectum, anal canal and pelvic floor

urinary bladder
peritoneum

Figure 2.13. Dissected


right male hemipelvis
seminal vesicle with pelvic organs, left
view. The rectoprostatic
neurovascular
septum (Denonvilliers’
bundle
fascia) is grasped by
prostate the right forceps and
retracted dorsally
together with the
anterior rectal wall.
rectoprostatic septum
The left forceps
(Denonvillier´s fasica)
grasps the posterior
anterior wall of the urinary
rectal wall bladder. Between the
rectoprostatic septum
anal and the prostate/
canal
seminal vesicles extend
the neurovascular
bundles.

and temperature, due to densely distributed somato- ani muscle and a urogenital diaphragm comprising
sensory nerve endings. The external anal sphincter is the transverse perineal muscles. In addition to these
innervated by inferior rectal nerve branches provided major striated muscles composed mainly of slow-
by the pudendal nerves. It is notable that a subgroup twitch type I fibres, the pelvic floor is also equipped
of healthy males and females reveals an asymmetry with smooth musculature. The smooth muscle
of external anal sphincter innervation, with a func- components are located predominantly along the
tional preponderance of either the left or right pu- border of the urogenital and anal hiatus created by
dendal nerve. the levator ani muscle. Further smooth muscle fi-
bres have also been described extending from the
rectal wall to the vagina (rectovaginal muscle) to the
PELVIC FLOOR urethra (recto-urethral muscle) and to the coccyx
along the anococcygeal ligament (rectococcygeal
The pelvic floor is composed of both striated and muscle, retractor recti muscle or Treitz muscle).
smooth muscles covered by fasciae and thus corre-
sponds to a rhabdo- and lissomusculofibrous system.
The pelvic floor has a twofold function: closure of the Pelvic Diaphragm/Levator Ani Muscle
pelvic cavity for the support of intrapelvic organs,
and controlled opening for micturition and defeca- The largest of the pelvic floor muscles is the levator
tion in both genders and parturition in females. The ani muscle, and the term ‘levator ani’ is often used to
female pelvic floor is larger than the male, displaying mean the entire pelvic floor. In fact, the levator ani
wider urogenital openings, but its muscle strength muscle covers most of the lower pelvic aperture and
is generally lower and the nerve supply is less devel- leaves only a midline gap for the urethra and vagina
oped. The female pelvic floor is therefore capable of (urogenital hiatus) and the anal canal (anal hiatus)
allowing vaginal delivery on the one hand, but on the (Figure 2.14). This anterior slit-like opening is partly
other hand it is more susceptible to functional insuf- closed by the urogenital diaphragm at the level of
ficiency and pelvic organ prolapse. the urogenital hiatus. The levator ani muscle corre-
The pelvic floor can be subdivided into a pelvic sponds to a broad, flattened, funnel-shaped muscle
diaphragm, which is composed mainly of the levator that originates from the pelvic wall. The muscular

HEBK001-C02_p31-51.indd 46 11/02/13 4:32 PM


PELVIC FLOOR  47

urogenital bone, where it is attached by a tendinous plate. The


hiatus puborectal
internal obturator muscle
pubococcygeal muscle adapts the shape of a ham-
muscle pubococcygeal
mock extending above the ileococcygeal muscles.
tendinous arc muscle Some muscle fibres decussate to the periurethral
ileococcygeal musculature and insert into the walls of the vagina
muscle
anal coccygeal (pubovaginal muscle) and rectum and anal canal
hiatus muscle (puboanal muscle). The puboanal fibres blend with
piriformis
muscle fibres of the longitudinal rectal muscle to form the
conjoint longitudinal muscle.

Puborectal muscle
The puborectal muscle is inseparable from the pub-
Figure 2.14. Pelvic floor, cranial view. The pelvic ococcygeal muscle at its origin at the pubic bone, but
diaphragm is formed by the levator ani muscle composed
more posteriorly the muscle angles at the anorectal
of the puborectal muscle, pubococcygeal muscles and
junction to form a sling behind the rectum. Con-
ileococcygeal muscles. Most of the levator ani muscle
originates from the tendinous arc (‘white line’), which
traction will compress the anal canal by pulling
corresponds to a condensed connective tissue line of the anorectal junction towards its fixed point (the
the obturator fascia. The puborectal sling forms a midline pubic bone), thereby reducing the anorectal angle.
gap for the urethra and vagina (urogenital hiatus) and the The puborectal sling is intimately fused with the
anal canal (anal hiatus). The coccygeal muscles extend deep part of the external anal sphincter. From the
from the ischiadic spine to the lateral margins of the caudal part of the muscle, pre-rectal fibres decus-
coccyx following the course of the sacrospinal ligaments. sate to insert into the perineal tendinous centre.
(Reproduced from Schünke, M. et al., Prometheus Atlas The puborectal muscle delimits the levator ani gap
of Anatomy, Vols. 1 and 2. Stuttgart, Germany, Thieme bordering the urogenital and anal hiatus.
Publ. 2007/2009, with permission.)

Coccygeal muscles
The coccygeal muscles are located dorsocranially to
funnel descends towards the anus and fuses with the the levator ani muscle and extend from the ischial
external anal sphincter. The levator ani muscle is spine to the lateral margins of the coccyx. The mus-
composed of the following parts: cle tissue is generally poorly developed and follows
the course of the sacrospinal ligaments. Lying in the
Ileococcygeal muscles same plane as the levator ani muscle, the coccygeal
The ileococcygeal muscles make up the largest por- muscles complete the pelvic diaphragm at its dor-
tion of the levator ani muscle and originate from socranial end.
the tendinous arc (arcus tendineus fasciae pelvis)
formed by a condensed tissue line (‘white line’) of Urogenital Diaphragm
the obturator fascia. Thus, its muscular insertion
sides are not rigid but correspond to a dynamic sus- The midline gap left by the levator ani muscle for
pension, allowing a degree of compliance against the urogenital hiatus is covered by the urogenital
traction and pressure forces. The flattened muscles diaphragm. This diaphragm extends between both
attach to the coccyx and the last two sacral vertebrae inferior branches of the pubic bone and corre-
and fuse posteriorly in a midline raphe. The funnel- sponds to a musculofibrous plate. In elderly people,
shaped muscle is generally very thin and displays and in particular in females after multiple vaginal
intramuscular slit-like gaps, particularly in females. deliveries, the muscular tissue is almost completely
replaced by connective tissue. In the midline, an
Pubococcygeal muscle anterior opening is left for the urethra surrounded
In contrast to the non-bony origin of the ileococcy- by the omega-shaped external urethral sphincter.
geal muscles, the pubococcygeal muscle runs from Posteriorly, the urogenital diaphragm has an open-
the inner surface of the pubic bone to the coccygeal ing in females for the vaginal wall.

HEBK001-C02_p31-51.indd 47 11/02/13 4:32 PM


48  Anatomy of the rectum, anal canal and pelvic floor

Deep transverse perineal muscle input is provided by direct sacral nerves and the pu-
Most of the urogenital diaphragm is made up of dendal nerves. The pudendal nerves also carry the
the deep transverse perineal muscle. According to somatosensory supply for the perineal region, in-
its name, the muscle fibres extend in a transverse cluding the highly innervated anodermal segment
direction between the left and right inferior pubic of the lower anal canal.
branch. The thin muscular plate is interwoven with
Direct sacral nerves/levator nerves
connective tissue and widens in a posterior direc-
tion, adopting a triangular shape. The muscular The direct sacral nerves for the innervation of the
portion adjacent to the passage of the vaginal tube muscular pelvic diaphragm originate from the
is also termed the constrictor vaginae muscle. third and fourth sacral nerves. The small-sized
nerves, also termed levator nerves, leave the an-
Superficial transverse perineal muscle terior sacral foramina and run from the sacral
The dorsal aspect of the deep transverse perineal concavity directly towards the levator ani muscle.
muscle is bordered by the superficial transverse They approach the levator ani muscle from above
perineal muscle. The slender muscle also extends and penetrate into its muscle fibres. When reach-
between the inferior pubic branches near the ischial ing the pelvic floor from above during a TME pro-
tubercles and is connected to the perineal tendinous cedure, care must be taken to preserve these direct
centre (Figure 2.15). nerve branches.

Nerve Supply of the Pelvic Pudendal nerves


Floor and Perineal Region The pudendal nerves (Figure 2.16) arise from the
second, third and fourth sacral nerves and initial-
All striated muscles of the pelvic floor are innervat- ly leave the pelvic cavity together with the sciatic
ed by nerves diverging from the ventral branches of nerves via the greater sciatic foramen. Once they
the sacral spinal nerves (S2–4). The somatomotor have passed through the infra-piriformis gap,
they curve around the sacrospinal ligament at
the level of the ischial spine to re-enter the pelvis
via the lesser sciatic foramen. The re-entry takes
external urethral
sphincter place at the infralevatory level within the ischio-
deep transverse anal space.
perineal muscle
The main pudendal nerve branch is ensheathed
superficial transverse
perineal muscle by a duplication of the obturator fascia (Alcock’s
internal obturator anal
puborectal canal) together with accompanying internal pu-
muscle muscle
hiatus
ileococcygeal
dendal blood vessels. Along its way towards the
muscle pubic bone, multiple branches leave Alcock’s ca-
piriformis nal, run throughout the fatty tissue of the ischio-
muscle coccygeal
muscle anal space and reach the entire perineal region
with somatomotor and somatosensory terminal
nerve fibres. Inferior rectal nerves innervate the
Figure 2.15. Female pelvic floor, caudal view. The external anal sphincter, the lower portion of the
urogenital hiatus of the pelvic diaphragm is covered levator ani muscles, and the perianal skin and
caudally by the urogenital diaphragm composed of the anoderm. Further ventrally, perineal nerves ap-
deep and superficial transverse perineal muscles. The
proach the urogenital diaphragm for innervation
urogenital diaphragm extends between both inferior
of the transverse perineal muscles and the exter-
branches of the pubic bone and corresponds to a thin
musculofibrous plate. An anterior opening is left for the
nal urethral sphincter and for the sensory nerve
urethra surrounded by the external urethral sphincter. supply of the perineal region, including the scro-
Posteriorly, the urogenital diaphragm gives way for the tal and labial skin. The dorsal nerve of the penis
vaginal wall. (Reproduced from Schünke, M. et al., and clitoris in males and females, respectively,
Prometheus Atlas of Anatomy, Vols. 1 and 2. Stuttgart, travels above the urogenital diaphragm towards
Germany, Thieme Publ. 2007/2009, with permission.) the cavernous bodies.

HEBK001-C02_p31-51.indd 48 11/02/13 4:32 PM


PELVIC FLOOR  49

sacrotuberal levator ani


ligament muscle

pudendal anococcygeal
nerve branches ligament
fascia of internal
Figure 2.16. Female pelvic floor with obturator muscle
dissection of the left perineal region,
dorsocaudal view. The fascia (grasped inferior rectal
by forceps) of the internal obturator ischiadic nerves
muscle is incised and opened to release tubercle external anal
the pudendal nerve fibres from Alcock’s sphincter
canal and to illustrate its terminal
anus
branches (green vessel loop). Inferior
perineal
rectal nerves run towards the external
nerves
anal sphincter and perianal region.
Perineal nerves extend in an anterior vaginal
direction to innervate the urogenital introitus
diaphragm and the vaginal introitus.

The terminal branches of the pudendal nerves The ventral part of the ischioanal fossa is caudally
are located at the outer surface of the puborectal closed by the urogenital diaphragm. The dorsal part
muscle and come into close proximity with the surrounds the lower anal canal and widens towards
neurovascular bundles at the level of the pro- the sacrotuberal ligaments and the gluteus maximus
static apex. At this point, fine branches of the
pudendal nerves intermingle with nerve fibres
from the neurovascular bundles to innervate the
urinary sphincter complex. This region is located
near the anterior dissection plane, and caution
should be taken to preserve these nerves, in par-
ticular if surgical resection is extended down to tendinous
the pelvic floor in abdominoperineal excision of arc
internal obturator
the rectum. muscle ischioanal
space

superficial perineal
Ischioanal Space Alcock´s canal with fascia
pudendal vessels & nerve
superficial perineal/
Below the pelvic diaphragm extends the ischioanal external anal perianal space
sphincter internal anal
space, or fossa, which corresponds to the infradia- sphincter
phragmatic or infralevatory pelvic compartment Figure 2.17. Rectum, anal canal and pelvic floor, frontal
extending below the pelvic floor. In a frontal cross- section, anterior view. The levator ani muscle originates
sectional plane, the ischioanal space appears as a at the tendinous arc on both sides and forms the funnel-
triangle with its base oriented towards the perineal shaped pelvic diaphragm extending down to the external
skin, the lateral side limited by the internal obtu- anal sphincter. The triangular space delimited by the
levator ani muscle, the internal obturator muscle and the
rator muscle, and the medial side bordered by the
superficial perineal fascia corresponds to the ischioanal
funnel-shaped levator ani muscle (Figure 2.17).
space. The main pudendal nerve branch and internal
The apex represents the junction of both muscles pudendal blood vessels are ensheathed by a duplication
along the tendinous arc. At the lateral side-wall ex- of the obturator fascia (Alcock’s canal). (Reproduced from
tends Alcock’s canal, a duplication of the internal Schünke, M. et al., Prometheus Atlas of Anatomy, Vols. 1
obturator fascia harbouring the main branches of and 2. Stuttgart, Germany, Thieme Publ. 2007/2009, with
the internal pudendal vessels and pudendal nerves. permission.)

HEBK001-C02_p31-51.indd 49 11/02/13 6:38 PM


50  Anatomy of the rectum, anal canal and pelvic floor

muscles. The ischioanal fossa is filled with loosely transverse perineal muscle and bulbospongious
arranged areolar fat (corpus adiposum perinei) and muscle in front, the superficial portion of the ex-
contains the terminal branches of the pudendal ternal anal sphincter at the back, and transversely
nerves and blood vessels. Posteriorly, the ischioanal running fibres of the puborectal muscle from the
space is crossed in the midline by the anococcygeal lateral sides.
ligament extending from the external anal sphinc- The perineal body serves as an anterior abutment
ter to the coccygeal bone. for the anorectal junction. It is virtually impossi-
At the perianal region, the ischioanal space is ble to define a clear surgical dissection plane in this
caudally delimited by a thin fascia, the superficial area, because longitudinal smooth muscle bundles
perineal fascia, formed by diverging tendinous end- deriving from the anterior rectal wall (corrugator
ings of the conjoint longitudinal muscle. Below this ani muscle) and the inferior continuation of the
connective tissue plane extends the perianal space. rectogenital septum are intimately connected to the
This space corresponds to the subcutaneous layer perineal body. Moreover, both the urogenital dia-
underlying the perianal skin and contains small fat phragm and the external anal sphincter are firmly
lobules separated by rigid connective tissue septa. attached to the perineal body. In males, fibres of the
The ischioanal space is particularly important in bulbospongious muscle regularly cross the perineal
abdominoperineal excision for low rectal cancer. In body and fuse with fibres of the superficial portion
conventional abdominoperineal excision, a classi- of the external anal sphincter. The condensation of
cal TME is performed and the ischioanal space is smooth muscle fibres that connect the anorectal
dissected only up to the external anal sphincter by junction to the perineal body and thence to the ure-
the perineal approach. This procedure often bears thra is also called the recto-urethralis muscle.
the risk of a ‘tissue waist’ at the tumour site and In abdominoperineal excision, detachment of
thus is associated with increased rates of positive the anterior aspect of the anorectal specimen ap-
circumferential resection margins and inadvertent pears to be one of the most delicate steps, since this
bowel perforation. The modified surgical proce- region lacks self-opening planes. Thus, dissection
dure, also described as ‘cylindrical abdominoperi- has to be carefully carried out behind the superficial
neal excision’, consists of an extended extralevator transverse perineal muscle to avoid perforation of
­abdominoperineal ­excision (ELAPE). The ELAPE the bowel wall and damage of the urethral sphincter
procedure avoids the tissue waist at the anorectal complex and its corresponding nerves.
junction by stopping the TME above the pelvic
floor and by extending the perineal dissection up to
the origin of the levator ani muscle. For this reason, Acknowledgements
the ischioanal space has to be dissected along the
outer surface of the external anal sphincter and of The author wishes to thank Dr Sigmar Stelzner
the funnel-shaped levator ani muscle until reach- (Department of General and Visceral Surgery,
ing its origin at the tendinous arc. Subsequently, Dresden-Friedrichstadt General Hospital, Germany)
the resulting specimen will include the entire anal for his valuable contributions regarding the anatomi-
sphincter complex together with major parts of cal dissections, and Günter-Rudolf Klaws and Stefanie
the levator ani muscle completely enveloping the Gundlach (Institute of Anatomy, Christian Albrechts
tumour-bearing region. University of Kiel, Germany) for their assistance in
the anatomical dissections and photographing.

Perineal Body/Perineal
Tendinous Centre References
1. Stelzner F. Chirurgie an den viszeralen Abschlusssyte-
The perineal body corresponds to the centre of the
men. Stuttgart, Thieme, 1998.
perineal region that serves as a common insertion 2. Valleix MM et al. Bulletin Nr. 10, third série. Juin 1836.
site for several muscles of the pelvic floor and there- 3. Walsh PC, Lepor H, Eggleston JC. Radical prostatecto-
fore is also termed the perineal tendinous ­centre. my with preservation of sexual function: anatomical and
Attached to the perineal body are the superficial pathological considerations. Prostate 1983; 4: 473–85.

HEBK001-C02_p31-51.indd 50 11/02/13 4:32 PM


FURTHER READING  51

FURTHER READING Kinugasa Y, Niikura H, Murakami G, et al. Development of


the human hypogastric nerve sheath with special refer-
ence to the topohistology between the nerve sheath
Aigner F, Zbar AP, Ludwikowski B, Kreczy A, Kovacs P, Fritsch
and other prevertebral fascial structures. Clin Anat
H. The rectogenital septum: morphology, function, and
2008; 21: 558–67.
clinical relevance. Dis Colon Rectum 2004; 47: 131–40.
Kirkham AP, Mundy AR, Heald RJ, Scholefield JH. Cadav-
Baader B, Herrmann M. Topography of the pelvic autonomic
eric dissection for the rectal surgeon. Ann R Coll Surg
nervous system and its potential impact on surgical
Engl 2001; 83: 89–95.
intervention in the pelvis. Clin Anat 2003; 16: 119–30.
Kourambas J, Angus DG, Hosking P, Chou ST. A histologi-
Blaivas JG, Barbalias GA. Characteristics of neural injury after
cal study of Denonvilliers’ fascia and its relationship to
abdomino-perineal resection. J Urol 1983; 129: 84–7.
the neurovascular bundle. Br J Urol 1998; 82: 408–10.
Clausen N, Wolloscheck T, Konerding MA. How to optimize
Lindsey I, Guy RJ, Warren BF, Mortensen NJ. Anatomy
autonomic nerve preservation in total mesorectal
of Denonvilliers’ fascia and pelvic nerves, impotence,
excision: clinical topography and morphology of
and implications for the colorectal surgeon. Br J Surg
pelvic nerves and fasciae. World J Surg 2008; 32:
2000; 87: 1288–99.
1768–75.
Lindsey I, Warren BF, Mortensen NJ. Denonvilliers’ fascia
Fritsch H, Lienemann A, Brenner E, Ludwikowski B. Clinical
lies anterior to the fascia propria and rectal dissection
anatomy of the pelvic floor. Adv Anat Embryol Cell Biol
plane in total mesorectal excision. Dis Colon Rectum
2004; 175: 1–64.
2005; 48: 37–42.
García-Armengol J, García-Botello S, Martinez-Soriano F,
Marr R, Birbeck K, Garvican J, et al. The modern
Roig JV, Lledó S. Review of the anatomic concepts
abdominoperineal excision: the next challenge after
in relation to the retrorectal space and endopelvic
total mesorectal excision. Ann Surg 2005; 242: 74–82.
fascia: Waldeyer’s fascia and the rectosacral fascia.
Maurer CA. Urinary and sexual function after total mesorec-
Colorectal Dis 2008; 10: 298–302.
tal excision. Recent Results Cancer Res 2005; 165:
Havenga K, DeRuiter MC, Enker WE, Welvaart K. Anatomical
196–204.
basis of autonomic nerve-preserving total mesorectal
Nagtegaal ID, van de Velde CJ, Marijnen GC, van Krieken
excision for rectal cancer. Br J Surg 1996; 83: 384–8.
JHJM, Quirke P. Low rectal cancer: a call for a change
Havenga K, Enker WE, McDermott K, Cohen AM, Minsky
of approach in abdominoperineal resection. J Clin
BD, Guillem J. Male and female sexual and urinary
Oncol 2005; 23: 9257–64.
function after total mesorectal excision with autonomic
Schünke M, Schulte E, Schumacher U. Prometheus
nerve preservation for carcinoma of the rectum. J Am
LernAtlas der Anatomie: Allgemeine Anatomie und
Coll Surg 1996; 182: 495–502.
Bewegungssystem. Stuttgart, Thieme, 2007.
Heald BJ, Moran BJ. Embryology and anatomy of the
Schünke M, Schulte E, Schumacher U. Prometheus LernAtlas
rectum. Semin Surg Oncol 1998; 15: 66–71.
der Anatomie: Innere Organe. Stuttgart, Thieme, 2009.
Heald RJ, Smedh RK, Kald A, Sexton R, Moran BJ. Ab-
Standring S. Gray’s Anatomy: The Anatomical Basis of
dominoperineal excision of the rectum: an endangered
Clinical Practice, 39th edn. Edinburgh, Churchill
operation. Dis Colon Rectum 1997; 40: 747–51.
Livingstone, 2004.
Hollabaugh RS Jr, Steiner MS, Sellers KD, Samm BJ,
Stelzner S, Holm T, Moran BJ, et al. Deep pelvic anatomy
Dmochowski RR. Neuroanatomy of the pelvis: im-
revisited for a description of crucial steps in extralevator
plications for colonic and rectal resection. Dis Colon
abdominoperineal excision for rectal cancer. Dis Colon
Rectum 2000; 43: 1390–97.
Rectum 2011; 54: 947–57.
Holm T, Ljung A, Häggmark T, Jurell G, Lagergren J.
Takenaka A, Hara R, Soga H, Murakami G, Fujisawa M. A
Extended abdominoperineal resection with gluteus
novel technique for approaching the endopelvic fascia
maximus flap reconstruction of the pelvic floor for
in retropubic radical prostatectomy, based on an
rectal cancer. Br J Surg 2007; 94: 232–8.
anatomical study of fixed and fresh cadavers. BJU Int
Kinugasa Y, Murakami G, Suzuki D, Sugihara K. Histologi-
2005; 95: 766–71.
cal identification of fascial structures posterolateral to
Uchimoto K, Murakami G, Kinugasa Y, Arakawa T, Matsub-
the rectum. Br J Surg 2007; 94: 620–6.
ara A, Nakajima Y. Rectourethralis muscle and pitfalls
Kinugasa Y, Murakami G, Uchimoto K, Takenaka A, Yajima
of anterior perineal dissection in abdominoperineal re-
T, Sugihara K. Operating behind Denonvilliers’ fascia
section and intersphincteric resection for rectal cancer.
for reliable preservation of urogenital autonomic nerves
Anat Sci Int 2007; 82: 8–15.
in total mesorectal excision: a histologic study using
Uhlenhuth E, Day EC, Smith RD, Middleton EB. The
cadaveric specimens, including a surgical experiment
visceral endopelvic fascia and the hypogastric sheath.
using fresh cadaveric models. Dis Colon Rectum 2006;
Surg Gynecol Obstet 1948; 86: 9–28.
49: 1024–32.

HEBK001-C02_p31-51.indd 51 11/02/13 4:32 PM


3
Clinical ultrasound
Oliver Shihab and Arcot K. Venkatasubramaniam

Introduction but give a poorer depth of penetration.2 In EUS,


frequencies of 7.0 MHz or 10 MHz are most com-
Accurate preoperative staging of rectal cancer influ- monly used and give satisfactory images.
ences the choice of surgery and the use of neoad-
juvant therapies, with the aim of achieving a clear
resection margin. It is also possible that, in selected
STAGING
early tumours, resectional surgery may be avoided
with the use of local excision. The endorectal ultra-
sound (EUS) staging system, its accuracy with Primary Tumour Staging
respect to other imaging modalities, and the clini-
cal role of EUS in the assessment of rectal cancer are Using EUS, it is possible to clearly visualize five
discussed in this chapter. ultrasonic layers of the rectum and surround tis-
sues, which correspond to the five anatomical lay-
ers. The EUS appearances alternate between echo-
genic and echo-poor:3
Imaging technique
l mucosa (echogenic);
Endorectal ultrasound is the oldest imaging modal- l muscularis mucosae (echo-poor);
ity for local staging of rectal cancer1 and can be l submucosa (echogenic);
conveniently performed in the office setting by the l muscularis propria (echo-poor);
colorectal surgeon. Endorectal ultrasound typically l serosa/perirectal fat (echogenic).
uses a rigid probe with a rotating ultrasound trans-
The traditional staging system using EUS fol-
ducer mounted at the tip. This ultrasound trans-
lows the method of Hildebrandt and Feifel:4 They
ducer contains crystals that emit sound waves when
established the use of the TNM (tumour, node,
electrically stimulated, producing images orthogo-
metastasis)-based system for EUS, with the ‘u’ pre-
nal to the probe (Figure 3.1). A balloon filled with
fix to show that the staging had been predicted by
degassed water is at the tip of the probe, passing the
ultrasound, using the relationship of the hypoecho-
emitted sound waves to the rectum via the water,
ic tumour to ultrasonic layers, as described above:
so preventing image distortion by air. The depth of
penetration of these waves is a function of the focal l uT0: tumour confined to the mucosa only.
length of the crystals: those that produce higher- l uT1: tumour confined to the mucosa and sub-
frequency waves produce a higher-resolution image mucosa.

HEBK001-C03_p52-57.indd 52 08/02/13 6:41 PM


STAGING  53

Figure 3.1. Anorectal transducer (courtesy BK medical). Scanning plane for the anorectal transducer.

l uT2: tumour invades muscularis propria but is of the submucosa (SM1) from those with greater
confined to the rectal wall. depth of invasion.20 This is of clinical importance,
l uT3: tumour invades perirectal tissues but does because patients with SM1 or lower disease have
not involve adjacent organs. an extremely low risk of lymph node metastasis.9,21
l uT4: tumour invades adjacent organs. Accurate identification of this subgroup could
therefore allow for safe local excision of the cancer.
Accuracy for depth of invasion is high, with stud-
ies showing this to be in the region of 74–96 per
cent.5–13 In one study, however, after neoadjuvant
radiotherapy the T-stage accuracy of EUS fell from Lymph Node Staging
86 per cent in the non-irradiated patient cohort
to 47 per cent in the irradiated cohort. It was sug- Prediction of lymph node involvement is prob-
gested that irradiation and subsequent rectal wall lematic, because approximately half of all lymph
thickening resulted in poor visualization of the rec- nodes with metastatic spread are less than 5 mm in
tal wall layers.14 Another study demonstrated that diameter22 and up to 18 per cent of lymph nodes
EUS correctly predicts cases of complete response of 4 mm or less may contain tumour metastases.23
following neoadjuvant therapy in 63 per cent of Although there is an association with lymph node
patients and correctly predicts T stage in 48 per cent size and likelihood of tumour metastasis, there is
of patients.15 considerable overlap in size between metastatic
In earlier cancers (SM1-T2), overall accuracy and non-metastatic nodes. As such, lymph node
of EUS is particularly high, in the region of 92 per involvement lacks sufficient discrimination to be
cent.9,16 This compares favourably with magnetic used as a staging criterion. It has been noted, how-
resonance imaging (MRI), which has an accuracy ever, that for grossly enlarged mesorectal nodes
of 67–86 per cent.17–19 The lower accuracy seen with (.9 mm in short-axis diameter), there is almost
MRI is thought to be due to the poor differentiation invariably involvement by tumour.24 Other criteria
of T1/2 cancer from very early T3 cancer, where used in staging include inhomogeneity, long-axis
it can be difficult to distinguish true mesorectal to short-axis diameter ratio (‘roundness index’),
tumour invasion from desmoplastic reactions. Use echogenicity, lymph node hilar reflectivity and
of a 15 MHz probe (resulting in an increased resolu- border contour.23–26
tion but decreased focal length) has a high accuracy A small number of studies have examined the
(86 per cent) in distinguishing the earliest lesions role of fine-needle aspiration (FNA) cytology in

HEBK001-C03_p52-57.indd 53 08/02/13 6:41 PM


54  Clinical ultrasound

improving the accuracy of nodal staging.27,28 Typi- be balanced against the immediate and long-term
cally this uses a longitudinally oriented ultrasound complications, such as perineal wound break-
probe, enabling the biopsy needle to accurately pass down in abdominoperineal excision (APE)29 and
into the target area. Although this has proven tech- well-documented long-term effects, including
nically feasible, the role in rectal cancer is yet to be pelvic fractures, increased risk of second primary
defined. In the study by Harewood and colleagues, tumours and alterations in urinary, bowel and sex-
there was no significant improvement in accuracy ual ­function.30 For these reasons, accurate imaging
of nodal staging using EUS and FNA, compared at this height is crucial in optimal management and
with EUS alone.28 Importantly, with FNA there is poses particular challenges in the lowest rectal can-
a theoretical risk of disseminating cancer cells into cers at the level of the sphincters.
the surrounding mesorectum. Historically, several studies have demonstrated
Nodal disease continues to present a prob- the ability of EUS to define the anal sphincters
lem, regardless of the imaging modality used, and and outline sphincter defects (typically in patients
results are still disappointing when compared with investigated for faecal incontinence) and to define
the staging of direct tumour invasion. Accuracy ­fistulae-in-ano.4,31–34 Furthermore, EUS can pro-
for EUS has been quoted as varying from 61 per vide reliable measurements of the external anal
cent to 83 per cent,6–11 while MRI has an accuracy ­sphincter.35 There have been very few studies, how-
of 57–85 per cent.17,19 Hilar reflectivity and lymph ever, of the ability of EUS to define the extent of
node inhomogeneity appear to have the greatest sphincter infiltration by rectal carcinoma. The
discriminatory value for prediction of lymph node one study found is limited by its small number of
involvement.24 patients (n = 12).36 Endorectal ultrasound does,
however, seem promising when compared with
pathology of the excised specimen, with 100 per
The Low Rectum and Anal Canal cent sensitivity and specificity for pT2 and pT4
tumours and 86 per cent sensitivity and specificity
In the lower third of the rectum (below the origin for pT3 tumours. Furthermore, 5 of the 12 tumours
of the levator muscles and usually within 6 cm of were post-chemoradiotherapy when assessed by
the anal verge), the mesorectum tapers sharply and EUS, and all of these were staged correctly.
no longer forms a protective barrier against tumour There are some reports on the staging accuracy
spread. The anal canal is generally considered to of EUS in anal squamous cell carcinoma, which also
extend from the upper border of puborectalis to has the potential to invade the sphincter complex.
the anal verge and is formed by the circular muscles As with the rectal cancer studies, published reports
of the internal and external anal sphincters, which on anal cancer and sphincter invasion are few in
form a partially overlapping tube of muscle. number and have small study sizes.
At the level of the sphincters the distinction One study involved 12 patients with anal cancer,
between T2 tumours (invading muscularis propria, all of whom were staged by EUS;37 of these, only 5
which, at this level, is the internal anal sphincter), patients underwent surgery, allowing EUS staging
T3 tumours (through the internal sphincter into to be compared with histopathology. Two tumours
the intersphincteric space) and T4 tumours (invad- were classified as uT2a (invading the internal anal
ing the external sphincter) is very small and diffi- sphincter) and three were classified as uT2b (invad-
cult to determine with any degree of certainty. The ing the external sphincter); histology was in exact
depth of tumour invasion at this level has important concordance in these cases.
implications in optimal preoperative and operative
management, particularly the aspects of sphincter
salvage or removal. Three-dimensional
Many centres give neoadjuvant chemoradio- endorectal ultrasound
therapy based on the height of tumours, due to
the greater rate of margin involvement at surgery. Three-dimensional ultrasound reconstructs radial and
Careful anatomical staging allows refinement of longitudinal images to produce a three-­dimensional
this. The benefits of neoadjuvant therapy have to image and may provide a more accurate anatomical

HEBK001-C03_p52-57.indd 54 08/02/13 6:41 PM


References  55

view of the rectum and surrounding structures. Its of the mesorectal fascia is more informative than T
use is said to improve both primary tumour and stage alone in determining the resectability of a rec-
nodal staging.26,38,39 This improved accuracy has also tal tumour,46,47 thereby influencing the decision for
been described in the selection of early tumours for neoadjuvant chemoradiotherapy. The pelvic side-
local excision40 and is also said to produce accurate wall lymph nodes are also typically outside the field
information with regard to the down-staging of of view of the EUS probes, and so other imaging
tumours following neoadjuvant therapies.41 modalities will be required to fully stage this region
to adequately plan treatment.

Limitations of endorectal
ultrasound CONCLUSION
Technical aspects of EUS must also be consid- The management of rectal cancer is complex,
ered. Problems are similar to those faced with the requiring decisions to be made as to the require-
endorectal coils in MRI before the introduction of ment for neoadjuvant therapies and the ability to
pelvic phased-array coils. The size of the probe used perform sphincter-sparing surgery. Endorectal
is significant, as it has been found that larger probes ultrasound is accurate for T staging of tumours
result in stretching of the anal canal. This distorts and is particularly accurate in early disease, where it
the anatomy of the region, so potentially affect- may be possible for selected patients to avoid resec-
ing the image.42 This anatomical distortion is also tional surgery. It also appears to have a promising
seen with the use of a water-filled balloon, used for role in the assessment of tumour relationship to the
acoustic coupling, in the anal canal.26 It is possible components of the anal sphincter complex and may
to overcome this distortion by using a rigid plas- develop a complementary role to MRI in the analy-
tic cone in place of the balloon. A tumour that is sis of cancers of this region. The development of
stenosing or that has a large polypoid intraluminal three-dimensional EUS may enhance the accuracy
component may also provide a physical barrier to of EUS in these roles.
accurate staging, which occurs in around 5 per cent
of rectal cancers.43 Unlike MRI, where planes can be
planned so that they lie orthogonal to the tumour References
and anal canal, it is possible that EUS can produce
images that are oblique to the canal. These oblique   1. Klessen C, Rogalla P, Taupitz M. Local staging of rectal
cancer: the current role of MRI. Eur Radiol 2007; 17:
images have the effect of exaggerating the apparent
379–89.
stage of the tumour.44   2. Saclarides TJ. Endorectal ultrasonography. J Surg
Harewood and colleagues hypothesized that Oncol 1996; 61: 239–41.
small numbers influenced reported accuracy   3. Beynon J, Foy DMA, Roe AM, Temple LN, Mortensen
of EUS.45 They reported an inverse relationship NJM. Endoluminal ultrasound in the assessment of
between study size and accuracy of EUS and specu- local invasion in rectal cancer. Br J Surg 1986; 73:
lated that this could partly result from publication 474–7.
  4. Hildebrandt U, Feifel G. Preoperative staging of rectal
bias. This may indicate that the accuracy of EUS has
cancer by intrarectal ultrasound. Dis Colon Rectum
been historically overestimated. This is emphasized 1985; 28: 42–6.
by the decline in the published accuracy of tumour   5. Skandarajah AR, Tjandra JJ. Preoperative loco-
staging over time.38 regional imaging in rectal cancer. ANZ J Surg 2006;
The biggest limitation for EUS in the staging of 76: 497–504.
rectal cancer is that it permits only limited evalua-   6. Garcia-Aguilar J, Pollack J, Lee SH, et al. Accuracy
tion of the structures surrounding the rectal wall, of endorectal ultrasonography in preoperative staging
of rectal tumors. Dis Colon Rectum 2002; 45:
including the mesorectal fascia. The mesorectal fas-
10–15.
cia represents a crucial landmark in the assessment   7. Mackay SG, Pager CK, Joseph D, Stewart PJ, Solo-
of rectal cancer, as it is the circumferential resection mon MJ. Assessment of the accuracy of transrectal
margin for a total mesorectal excision of the rec- ultrasonography in anorectal neoplasia. Br J Surg
tum. It has been demonstrated that the involvement 2003; 90: 346–50.

HEBK001-C03_p52-57.indd 55 08/02/13 6:41 PM


56  Clinical ultrasound

  8. Nesbakken A, Lovig T, Lunde OC, Nygaard K. Staging presence or absence of metastasis in lymph nodes in
of rectal carcinoma with transrectal ultrasonography. rectal cancer. J Surg Oncol 1993; 54: 252–4.
Scand J Surg 2003; 92: 125–9. 23. Katsura Y, Yamada K, Ishizawa T, Yoshinaka H, Shima-
  9. Akasu T, Kondo H, Moriya Y, et al. Endorectal ultra- zu H. Endorectal ultrasonography for the assessment
sonography and treatment of early stage rectal cancer. of wall invasion and lymph node metastasis in rectal
World J Surg 2000; 24: 1061–8. cancer. Dis Colon Rectum 1992; 35: 362–8.
10. Glaser F, Schlag P, Herfarth C. Endorectal ultrasonog- 24. Hulsmans FH, Bosma A, Mulder PJ, Reeders JW,
raphy for the assessment of invasion of rectal tumours Tytgat GN. Perirectal lymph nodes in rectal cancer: in
and lymph node involvement. Br J Surg 1990; 77: vitro correlation of sonographic parameters and his-
883–7. topathologic findings. Radiology 1992; 184: 553–60.
11. Orrom WJ, Wong WD, Rothenberger DA, Jensen LL, 25. Hildebrandt U, Klein T, Feifel G, Schwarz HP, Koch
Goldberg SM. Endorectal ultrasound in the preopera- B, Schmitt RM. Endosonography of pararectal lymph
tive staging of rectal tumors. A learning experience. nodes: in vitro and in vivo evaluation. Dis Colon
Dis Colon Rectum 1990; 33: 654–9. Rectum 1990; 33: 863–8.
12. Beynon J. An evaluation of the role of rectal 26. Giovannini M, Ardizzone S. Anorectal ultrasound for
endosonography in rectal cancer. Ann R Coll Surg neoplastic and inflammatory lesions. Best Pract Res
Engl 1989; 71: 131–9. Clin Gastroenterol 2006; 20: 113–35.
13. Hildebrandt U, Feifel G, Schwarz HP, Scherr O. 27. Milsom JW, Czyrko C, Hull TL, Strong SA, Fazio VW.
Endorectal ultrasound: instrumentation and clinical Preoperative biopsy of pararectal lymph nodes in
aspects. Int J Colorectal Dis 1986; 1: 203–7. rectal cancer using endoluminal ultrasonography. Dis
14. Napoleon B, Pujol B, Berger F, Valette PJ, Gerard JP, Colon Rectum 1994; 37: 364–8.
Souquet JC. Accuracy of endosonography in the stag- 28. Harewood GC, Wiersema MJ, Nelson H, et al. A
ing of rectal cancer treated by radiotherapy. Br J Surg prospective, blinded assessment of the impact of
1991; 78: 785–8. preoperative staging on the management of rectal
15. Vanagunas A, Lin DE, Stryker SJ. Accuracy of cancer. Gastroenterology 2002; 123: 24–32.
endoscopic ultrasound for restaging rectal cancer 29. Marijnen CAM, Kapiteijn E, van de Velde CJH, et al.
following neoadjuvant chemoradiation therapy. Am J Acute side effects and complications after short-term
Gastroenterol 2004; 99: 109–12. preoperative radiotherapy combined with total mesorec-
16. Glancy DG, Pullyblank AM, Thomas MG. The role of tal excision in primary rectal cancer: report of a multi-
colonoscopic endoanal ultrasound scanning (EUS) in center randomized trial. J Clin Oncol 2002; 20: 817–25.
selecting patients suitable for resection by transanal 30. Holm T, Singnomklao T, Rutqvist L, Cedermark B. Adju-
endoscopic microsurgery (TEM). Colorectal Dis 2005; vant preoperative radiotherapy in patients with rectal
7: 148–50. carcinoma: adverse effects during long term follow-up
17. Blomqvist L, Holm T, Rubio C, Hindmarsh T. Rectal of two randomized trials. Cancer 1996; 78: 968–76.
tumours: MR imaging with endorectal and/or phased- 31. Kumar A, Scholefield JH. Endosonography of the anal
array coils, and histopathological staging on giant canal and rectum. World J Surg 2000; 24: 208–15.
sections – A comparative study. Acta Radiol 1997; 38: 32. Deen KI, Kumar D, Williams JG, Olliff J, Keighley MR.
437–44. Anal sphincter defects: correlation between endoanal
18. Poon FW, McDonald A, Anderson JH, et al. Accuracy ultrasound and surgery. Ann Surg 1993; 218: 201–5.
of thin section magnetic resonance using phased- 33. Tjandra JJ, Milsom JW, Stolfi VM, et al. Endoluminal
array pelvic coil in predicting the T-staging of rectal ultrasound defines anatomy of the anal canal and
cancer. Eur J Radiol 2005; 53: 256–62. pelvic floor. Dis Colon Rectum 1992; 35: 465–70.
19. Brown G, Radcliffe AG, Newcombe RG, Dallimore NS, 34. Schwartz DA, Wiersema MJ, Dudiak KM, et al. A
Bourne MW, Williams GT. Preoperative assessment comparison of endoscopic ultrasound, magnetic
of prognostic factors in rectal cancer using high- resonance imaging, and exam under anesthesia
resolution magnetic resonance imaging. Br J Surg for evaluation of Crohn’s perianal fistulas.
2003; 90: 355–64. Gastroenterology 2001; 121: 1064–72.
20. Harada N, Hamada S, Kubo H, et al. Preoperative 35. Frudinger A, Halligan S, Bartram CI, Price AB, Kamm
evaluation of submucosal invasive colorectal cancer MA, Winter R. Female anal sphincter: age-related
using a 15-MHz ultrasound miniprobe. Endoscopy differences in asymptomatic volunteers with high-
2001; 33: 237–40. frequency endoanal US. Radiology 2002; 224: 417–23.
21. Kikuchi R, Takano M, Takagi K, et al. Management 36. Maier AG, Kreuzer SH, Herbst F, et al. Transrectal
of early invasive colorectal cancer: risk of recurrence sonography of anal sphincter infiltration in lower rectal
and clinical guidelines. Dis Colon Rectum 1995; 38: carcinoma. Am J Roentgenol 2000; 175: 735–9.
1286–95. 37. Tarantino D, Bernstein M. Endoanal ultrasound in the
22. Kotanagi H, Fukuoka T, Shibata Y, et al. The size of staging and management of squamous-cell carcinoma
regional lymph nodes does not correlate with the of the anal canal. Dis Colon Rectum 2002; 45: 16–22.

HEBK001-C03_p52-57.indd 56 08/02/13 6:41 PM


References  57

38. Kim JC, Kim HC, Yu CS, et al. Efficacy of 3-dimen- indications, results and limitations. Eur J Radiol 2007;
sional endorectal ultrasonography compared with con- 61: 480–9.
ventional ultrasonography and computed tomography 43. Manger T, Stroh C. Accuracy of endorectal ultrasonog-
in preoperative rectal cancer staging. Am J Surg 2006; raphy in the preoperative staging of rectal cancer. Tech
192: 89–97. Coloproctol 2004; 8: s14–15.
39. Santoro GA, Fortling B. The advantages of volume 44. Hashimoto BE. Possible invasive rectal wall mass or
rendering in three-dimensional endosonography of the anal sphincter break. In Sanders RC, Winter TC (eds).
anorectum. Dis Colon Rectum 2007; 50: 359–68. Clinical Sonography: A Practical Guide. Philadelphia,
40. Santoro GA, Gizzi G, Pellegrini L, Battistella G, PA, Lippincott Williams & Wilkins, 2006: 203.
Di Falco G. The value of high-resolution three-­ 45. Harewood GC. Assessment of publication bias in the
dimensional endorectal ultrasonography in the man- reporting of EUS performance in staging rectal cancer.
agement of submucosal invasive rectal tumors. Dis Am J Gastroenterol 2005; 100: 808–16.
Colon Rectum 2009; 52: 1837–43. 46. Wolberink SV, Beets-Tan RG, Nagtegaal ID, Wiggers T.
41. Murad-Regadas SM, Regadas FS, Rodrigues LV, et al. Preoperative assessment of the circumferential margin
Role of three-dimensional anorectal ultrasonography in rectal cancer is more informative in treatment plan-
in the assessment of rectal cancer after neoadjuvant ning than the T stage. Techn Coloproctol 2006; 10:
radiochemotherapy: preliminary results. Surg Endosc 171–6.
2009; 23: 1286–91. 47. MERCURY Study Group. Diagnostic accuracy of
42. Saranovic D, Barisic G, Krivokapic Z, Masulovic D, preoperative magnetic resonance imaging in predict-
Djuric-Stefanovic A. Endoanal ultrasound evaluation ing curative resection of rectal cancer: prospective
of anorectal diseases and disorders: technique, observational study. BMJ 2006; 333: 779.

HEBK001-C03_p52-57.indd 57 08/02/13 6:41 PM


4
Magnetic resonance imaging staging
of rectal cancer
Peter How and Gina Brown

Introduction 1982,1 TME involves the meticulous dissection and


complete removal of an intact surgical specimen
Rectal cancer is common, and accurate preopera- containing the tumour-bearing rectum surrounded
tive staging of tumours using high-resolution mag- by the mesorectum, the embryologically derived
netic resonance imaging (MRI) is a crucial part of lymphovascular envelope of the human rectum.1,2
modern multidisciplinary team management. The The principles of TME are outlined in Chapter 7
importance of MRI in staging rectal cancer relates and are based on the following principles:
specifically to the ability to delineate tumour exten-
sion with respect to surgically relevant landmarks, l The rectum is surrounded by the mesorectum
primarily the mesorectal fascia and sphincter com- and an areolar tissue, the mesorectal fascia, that
plex. This has significant therapeutic implications forms a surgically distinct avascular plane.
l Perimesorectal plane dissection should result in
regarding the need or otherwise for neoadjuvant
therapy, sphincter preservation and plane of sur- complete removal of the mesorectum contained
gery that can have a major impact upon the pa- within an intact fascia.
l Careful dissection of the mesorectal fascia
tient’s survival and quality of life. With the develop-
ment of effective coil systems and high-resolution allows identification and preservation of the
surface body coils, MRI looks likely to maintain and autonomic nerves, important for genitourinary
expand its current position as the imaging modality function.
of choice for planning an effective therapeutic strat- In addition, anal sphincter preservation by ante-
egy in patients with advanced rectal cancer. rior resection is generally favoured where feasible, as
it avoids the need for a permanent stoma and is gen-
erally considered oncologically superior to sphincter
MRI and surgical planning removal at abdominoperineal excision.3 TME fa-
for rectal cancer cilitates restorative resection. Since the adoption of
TME, the mesorectal fascia has come to be synony-
Undoubtedly, the most important advance with- mous with the circumferential resection margin, and
in rectal cancer surgery over the past 30 years has tumours located within 1 mm of this margin are con-
been the development of total mesorectal exci- sidered to be margin-positive and at high risk of lo-
sion (TME). Described by Heald and colleagues in cal recurrence. Brown and colleagues demonstrated

HEBK001-C04_p58-70.indd 58 08/02/13 6:40 PM


ANATOMICAL LANDMARKS  59

ANATOMICAL LANDMARKS

The important anatomical structures to consider in


rectal cancer surgery, some or all of which may be
visualized on MRI, are:
l normal rectal wall;
l mesorectum and mesorectal fascia;
l anal sphincter complex;
l Denonvilliers’ fascia/urogenital septum;
l presacral fascia;
l peritoneal reflection;
l pelvic nerve plexuses.
Figure 4.1. Depiction of rectal tumour (red line) relative
to the mesorectal fascia (blue line) on axial magnetic
resonance imaging. Normal Rectal Wall

The rectal wall, in keeping with the rest of the large


the feasibility and reproducibility of identifying such
bowel, is composed of an inner mucosal layer, con-
high-risk tumours on MRI by accurately depicting
centrically surrounded by submucosa and the mus-
the proximity of the tumour to the mesorectal fascia
cularis propria, comprising an inner circular and
(Figure 4.1).4 This is a key determinant for success-
outer longitudinal layer. Interposed between the
ful treatment of rectal cancer, as it is this relationship
two muscle layers lies the myenteric plexus con-
that often dictates the use of neoadjuvant therapy
tained within a thin layer of connective tissue.
and helps determine the optimal surgical plane. In
The mucosal layer on MRI is represented by a del-
cases where tumour extends to or goes through the
icate low-signal intensity line, with the submucosal
mesorectal fascia, the TME or ‘holy plane’ is deemed
layer beneath this depicted as a thicker higher-signal
unsafe; neoadjuvant therapy (preoperative radio- or
intensity structure. With high-quality images, the
chemoradiotherapy) or an extended surgical proce-
muscularis propria can sometimes be visualized as
dure, or a combination of both, may then be required
two distinct layers of circular and longitudinal mus-
to achieve a clear circumferential resection margin.
cle. The outer muscle layer often has an irregular
In a low rectal cancer, an enhanced extralevator ap-
ridged appearance on account of blood vessels enter-
proach may be needed. Such radiological guidance
ing the rectal wall. The perirectal fat appears as high-
is of particular interest with respect to tumours of
signal-intensity tissue, contrasting well with the low
the lower rectum where the mesorectum tapers as it
signal of the muscularis propria (Figure 4.2).
approaches the pelvic floor. It is here where breaches
of the mesorectal fascia and infiltration of the leva-
tor are often observed in advanced tumours. This is Mesorectal Fascia and Mesorectum
outlined in detail later.
In addition to the mesorectal fascia, the proxim- The mesorectal fascia is best seen on axial images
ity of the tumour to other important pelvic viscera and appears as a low-signal intensity linear struc-
plays an important role in surgical planning. These ture surrounding the mesorectum (Figure 4.3). In-
viscera include the prostate, seminal vesicles, bladder, feriorly, this layer fuses with the endopelvic fascia
uterus and vagina. Curative resection may depend on that lies over the surface of the levator muscles; su-
removing parts or all of these adjacent structures in periorly, the fascia fuses with the peritoneal reflec-
locally advanced tumours. Where tumour extends tion anteriorly and parietal fascia posteriorly.
to or invades these structures, conventional surgery The mesorectum on MRI axial images appears as
(anterior resection or abdominoperineal excision) a high-signal intensity (similar to fat) package sur-
will result in cutting through the tumour, and a more rounding the rectum, containing blood vessels and
radical surgical approach such as pelvic exenteration lymphatic tissue. Lymph nodes within the mesorec-
may be required. tum appear as high-signal intensity ovoid structures.

HEBK001-C04_p58-70.indd 59 08/02/13 6:40 PM


60  MRI staging of rectal cancer

ICM, inner circular muscle; M, mucosa; OLM, outer longitudinal muscle; SM, submucosa.
Figure 4.2. Layers of the bowel wall as they appear on magnetic resonance imaging and on histopathology.

Anal Sphincter Complex important for continence (i.e. internal and external
sphincter, puborectalis, levator plate) (Figure 4.4).
The anal canal is composed of inner circular smooth
muscle forming the internal sphincter, surrounded Denonvilliers’ Fascia/Urogenital Septum
by skeletal muscle forming the external sphincter.
The anal canal is seen as a cylindrical structure that Denonvilliers’ fascia or the urogenital septum (see
extends from the insertion of the puborectalis sling Chapter 7) forms a distinctive anterior structure,
on to the rectum to the external anal orifice. The more prominent in the male. The fascia represents
lower portion of the levator ani and external anal
sphincter are anatomically indistinguishable (see
Chapter 2). With high-quality T2 coronal and axial
images, it is possible to accurately depict tumour
proximity to the sphincter complex and delineate
individual components of the sphincter complex

EAS, external anal sphincter; IAS, internal anal sphincter.

Figure 4.3. Mesorectal fascia (blue line) surrounding Figure 4.4. Coronal image showing relationship of tumour
mesorectum and benign lymph node (arrow). (red) to sphincter complex and its components.

HEBK001-C04_p58-70.indd 60 08/02/13 6:41 PM


ANATOMICAL LANDMARKS  61

side-wall. The pelvic side-wall contains a conglomera-


tion of lymph nodes that are related to branches of the
internal and external iliac vessels and are therefore in
a separate compartment from the mesorectum. The
pelvic side-wall nodes are therefore not routinely vis-
ualized in rectal cancer surgery unless this compart-
ment is opened up by dissecting through the presacral
or parietal fascia (see Chapter 7, p. 103).

Peritoneal Reflection

On sagittal MRI, the peritoneal reflection appears as


a low-signal intensity linear structure that extends
over the surface of the bladder and can be traced pos-
Figure 4.5. Axial T2-weighted image showing teriorly to its point of attachment on to the rectum.
Denonvilliers’ fascia as a low-signal layer. On axial section, this point of attachment gives a V-
a barrier to the spread of anterior tumours and shaped configuration, with the anterior covering of
separates the rectum and perirectal structures from the rectum by the peritoneal reflection widening in the
the urogenital organs. It consists of a fibromuscular cranial direction. It is important to note that the peri-
structure composed of a number of layers that are toneal covering of the rectum is not a surgical resec-
fused together and envelope the seminal vesicles. De- tion margin, as there is no adjacent structure – hence,
nonvilliers’ fascia is visible on MRI as a low-signal tumour extending to within 1 mm of the peritoneum
layer that can be traced up to the peritoneum supe- should not be considered as a potentially involved cir-
riorly (Figure 4.5). cumferential resection margin, although of course the
margin of the specimen removed may well be involved
Presacral Fascia with spread through the peritoneum and a high risk
of subsequent peritoneal carcinomatosis (Figure 4.7).
The presacral fascia appears on sagittal MRI as a low-
signal intensity linear structure overlying the presacral Pelvic Nerve Plexuses
vessels (Figure 4.6). The presacral fascia lies posterior
to the mesorectal fascia. It is a thickened parietal fascia Preservation of the autonomic nerve plexuses is cru-
covering the presacral veins and fat. It fuses with and cial if genitourinary dysfunction is to be avoided.
directly covers the muscles and vessels of the pelvic

Figure 4.7. Sagittal magnetic resonance imaging


demonstrating the peritoneal reflection as a low-signal
Figure 4.6. Sagittal magnetic resonance imaging showing the V-shaped linear structure extending over the bladder to the
presacral fascia as a low-signal-intensity linear structure. rectum.

HEBK001-C04_p58-70.indd 61 08/02/13 6:41 PM


62  MRI staging of rectal cancer

The superior hypogastric plexus, situated below the


bifurcation of the aorta, gives rise to the right and
left hypogastric nerves that descend to join the in-
ferior hypogastric plexus. The right and left inferior
hypogastric plexus lie in a parasagittal plane on the
pelvic side-wall. In males, the inferior hypogastric
plexus lies posterolaterally to the seminal vesicles; in
females, its anterior half lies against the upper third
of the vagina. On MRI, the inferior hypogastric plex-
us lies inside and medial to the vessels on the pelvic
side-wall but outside the mesorectal plane. The in-
ferior hypogastric plexus is a distinctive rectangular
fenestrated structure, typically 3–4 cm in anteropos-
terior length, lying in a parasagittal plane that can
often be identified on MRI due to its high-signal
intensity (Figure 4.8). By contrast, on axial and coro-
nal oblique imaging, the plexus appears as a linear
beaded structure.

STAGING RECTAL TUMOURS

Tumour staging is critical for planning of therapy


and determining likely prognosis with regard to lo-
cal recurrence and survival. To develop an effective
therapeutic strategy based on the patient’s indi-
vidual circumstances, discussion of MRI staging in
a multidisciplinary team context is key. It has been
reported that multidisciplinary team discussion of
preoperative MRI with implementation of a selec-
tive preoperative strategy significantly reduces R1
and R2 resections.5 For most cancers, tumour ex-
tension at diagnosis and treatment determines the Figure 4.8. Inferior hypogastric plexus appears as a
high-signal beaded structure on paracoronal (A) and axial
likelihood of cure; this is particularly relevant to
(B) imaging.
rectal cancer, where local and systemic recurrence
are major issues. Cuthbert Dukes highlighted the
importance of extramural spread in predicting lo- Like the Dukes system, the TNM system is a patho-
cal recurrence and survival in patients with rectal logically based system; the prefix ‘p’ indicates ‘patho-
cancer.6 The system he described has stood the test logical’ (e.g. pT1 indicates a T1 tumour at pathology).
of time; the so-called ‘Dukes classification’ focuses The main advantage of the TNM system is its universal
on two key aspects – the local extent of the tumour applicability and the ability to predict the likely stage
in relationship to the bowel wall, and the pres- at preoperative imaging and alter the management
ence or absence of lymph nodal involvement. The appropriately. In rectal cancer, a T1 rectal carcinoma
Dukes system was based on the excised specimen is limited to the submucosa and on MRI is depicted
subjected to pathological analysis and subsequent by intermediate signal intensity within the mucosa
staging. Currently, however, rectal cancer staging and submucosa, with preservation of a thin layer of
is based mostly on the tumour–node–metastasis submucosa lying deep to the tumour. The MRI appear-
(TNM) and Union for International Cancer Con- ances of such a tumour should be recorded as ‘mrT1’.
trol (UICC) staging systems, which have largely su- A T2 tumour invades the muscularis propria and
perseded the Dukes classification. on MRI corresponds to tumour signal extending into

HEBK001-C04_p58-70.indd 62 08/02/13 6:41 PM


STAGING RECTAL TUMOURS  63

the circular muscle, but without extending through The abrupt change in size of the rectal lumen
the full thickness of muscle. Where the full thickness at the anorectal junction limits the usefulness of
of the muscle coat is replaced by intermediate signal oblique axial imaging performed proximally. Be-
intensity, this would equate with a T2 or T3 tumour, yond the anorectal junction, axial imaging cannot
which can be impossible to differentiate on MRI. A T3 demonstrate the rectal wall in its entirety and is un-
rectal cancer extends beyond the muscularis propria able to reliably determine separation of the rectal
and is depicted on MRI by a broad-based infiltrative wall from the levator plate. This appearance could
margin of intermediate signal intensity, extending into lead to potential overstaging of tumours. A high-­
perirectal fat. Clearly, however, T3 cancers are a very resolution coronal imaging sequence, however, can
heterogeneous group, including tumours of variable accurately depict the levator, sphincter complex
prognostic significance related to the depth of extra- and intersphincteric plane, which has important
mural spread.7 More recent versions of the TNM stag- surgical implications.
ing system have sought to rectify this problem with
the introduction of subgroups T3a (, 1 mm beyond
muscularis propria), T3b (1–5 mm beyond muscula- mrT Staging
ris propria), T3c (5–15 mm beyond muscularis pro-
pria) and T3d (. 15 mm beyond muscularis propria), It is widely accepted that MRI is the imaging mo-
which have prognostic significance.8 T4 rectal cancers dality that offers the highest soft-tissue contrast and
are defined as tumours that involve the peritoneal sur- is therefore best suited to T staging rectal cancer.
face or invades adjacent organs. Initial results with MRI were disappointing due
to poor spatial resolution achieved by whole-body
coil systems. The development of endorectal coils
Imaging Technique improved resolution with T staging accuracies com-
parable to endorectal ultrasound, widely regarded
MRI rectal cancer staging is relatively fast and as the gold standard for assessing local invasion
straightforward, because no special patient prepara- of an early rectal cancer. As with endorectal ultra-
tion is required. Some centres advocate the use of an sound, however, endorectal MRI imaging is lim-
antispasmodic agent such as hyoscine butylbromide ited by a small field of view that permits adequate
to prevent artefacts caused by small bowel peristal- evaluation only of early-stage rectal cancer, and not
sis and to help distend the sigmoid and rectum.14 To advanced cancers, as visualization of surrounding
effectively plan the initial localization sequences, it pelvic anatomy is limited. The other major disad-
is important for the radiologist or radiographer to vantage of endoluminal techniques relates to the
know the approximate tumour height on clinical as- invasive nature; technical limitations such as steno-
sessment. The patient is then positioned on his or her sis, stricturing, pain, discomfort, bowel wall move-
back and a phased-array surface coil is placed on the ment and coil migration may render the acquisition
pelvis, such that the edge of the coil is situated a good of good-quality images impossible, particularly in
distance below the pubic bone. The coil is secured advanced tumours.
with belts and the patient is advanced head first into The major breakthrough in MRI rectal can-
the magnetic field. cer staging came with the development of high-­
The first series of localization images is the sagit- resolution phased-array surface-coil systems.
tal T2W-FSE, which identifies the primary tumour. These surface coils can achieve a very high spatial
The second series consists of large field-of-view contrast resolution while maintaining a large field
axial sections of the whole pelvis, extending from of view, enabling both accurate depiction of the in-
the iliac crest to the pubic symphysis. The sagittal testinal wall and important surrounding anatomy,
T2-weighted images are used to plan T2-weighted including the mesorectal fascia. In recent studies,
thin-section axial images through the rectal cancer reported accuracy has varied from 86 per cent to
and along the rectum down to the anus, while en- 100 per cent;9–11 such variability largely reflects the
compassing perirectal tissues including important difficulties in staging borderline tumours as T2 or
pelvic landmarks. The images are achieved using T3. It is well recognized that overstaging T2 tu-
3 mm sections and a 16 cm field of view. mours is often caused by a desmoplastic reaction

HEBK001-C04_p58-70.indd 63 08/02/13 6:41 PM


64  MRI staging of rectal cancer

of peritumoural tissue.12 The most clinically rel- benign, reactive and malignant nodes has been
evant feature of MRI, however, relates to its assess- observed; Brown and colleagues23 have reported
ment of the mesorectal fascia and thus the poten- that a previously purported cut-off size of 5 mm
tial circumferential resection margin in a patient to differentiate involved from uninvolved nodes22
undergoing a TME. The multicentre MERCURY is unfounded. The importance of preoperative N
study revealed MRI to be accurate within 0.5 mm staging is not limited to its prognostic value with
in predicting local tumour spread and specificity regard to systemic recurrence, because lymph
for prediction of a clear margin by MRI to be 92 nodal involvement has also been linked to an in-
per cent.13 creased local recurrence. The presence of involved
lymph nodes close to the mesorectal fascia (cir-
cumferential resection margin in a patient with an
N Staging upper or middle rectal cancer having a TME) has
been reported to increase the risk of local recur-
As with extent of extramural invasion, Dukes was rence,24 and many units consider suspicious nodes
one of the first to identify lymph node involvement near to the mesorectal fascia to be an indication
as having significant prognostic value, with 5-year for neoadjuvant therapy. A publication from 2010
survival being related to the number of affected evaluated the importance of nodes near to the
nodes.19 A number of studies have demonstrated mesorectal fascia detected on MRI and correlated
that the involvement of four or more nodes signifi- whether this resulted in a positive circumferential
cantly worsens survival and highlights the impor- resection margin. In a retrospective review of 396
tance of adequate lymph node dissection during patients with rectal cancer, Shihab and colleagues
surgery and histopathological assessment. Current identified 31 patients with suspicious nodes 1 mm
guidelines from the Royal College of Pathologists or less from the circumferential resection margin.
recommend that an average of more than 12 nodes None of these patients had a positive circumfer-
should be sampled for all surgical specimens con- ential resection margin owing to nodal involve-
taining cancer. ment.25 This would suggest that with regard to
Predicting involvement of the lymph nodes on nodes threatening the circumferential resection
preoperative imaging has been considered by many margin, only those with obvious extracapsular ex-
to be a crucial but challenging task in rectal cancer tension will result in a positive margin and would
staging. Accuracy rates for N staging reported in therefore require neoadjuvant therapy.
the literature show wide variation across all recog-
nized imaging techniques. The reported accuracy
for MRI ranges from 57 per cent to 85 per cent.20–23 Pelvic Side-Wall Nodal Disease
Although it is impossible for any form of preop-
erative imaging to reliably exclude microscopic The use of 16 cm field-of-view thin-section MRI
nodal involvement, the high-contrast resolution enables the pelvic side-wall compartment and the
images offered by MRI have enabled the identifi- mesorectal compartment to be assessed. This is
cation of consistent markers for lymph nodal in- of particular value in identifying patients at risk
volvement. Brown and colleagues demonstrated of residual disease, despite a successful TME op-
that an irregular contour and an inhomogeneous eration, due to pelvic side-wall nodal disease. It is
signal are the most reliable MRI criteria for lymph generally thought that pelvic side-wall disease is a
node metastasis when comparing preoperative feature of aggressive disease and associated with
MRI with histopathological assessment of the re- poor survival.26 Undoubtedly involved nodes are
sected specimen (Figure 4.9).23 Conversely, normal more common in patients with low rectal cancer.
or reactive nodes are characterized by uniform In the UK and Europe, pelvic side-wall nodal dis-
signal intensity and smooth, well-defined borders. section is seldom performed because of the unac-
Although it is natural to suspect that enlarged ceptable morbidity related to this operation. Cur-
nodes are likely to be malignant, using size crite- rently in the West, a high risk of suspicious pelvic
ria alone would result in a high number of false side-wall nodal involvement is generally treated by
positives. Considerable overlap in size between targeted radiotherapy or chemoradiotherapy.

HEBK001-C04_p58-70.indd 64 08/02/13 6:41 PM


STAGING RECTAL TUMOURS  65

Figure 4.9. Irregular contour and inhomogeneous signal are the most reliable magnetic resonance imaging (MRI) criteria
for lymph node metastasis on MRI/histopathological comparison.

Vascular Spread l Location of tumour relative to major vessels:


presence of tumour signal within a vascular
Extramural vascular invasion (EMVI) is defined as structure is highly suggestive of EMVI.
the presence of malignant cells within blood vessels l Calibre of vessel: as tumour invades along the

beyond the muscularis propria in the vicinity of a lumen, the vessel expands. Tumour signal is
colorectal tumour. Histological EMVI has long been intermediate (grey), and hence expansion of a
recognized as an independent predictor of ­local and low-signal vessel by tumour extension is readily
systemic recurrence and poorer overall survival27–29 identifiable.
and has been reported to occur in up to 52 per cent of l Vessel border: tumour may eventually expand

all cases of colorectal cancer.30–34 Extramural vascular through the vessel wall to give variable results;
invasion is therefore considered an adverse prognos- the border may be smooth, irregular or nodular.
tic feature and by some is considered an indication
Based on these criteria, a five-point grading sys-
for neoadjuvant therapy, despite the circumferential
tem (0 to 4) for MRI-predicted EMVI has been pro-
resection margin remaining unthreatened by the
posed, with the lowest score (0) corresponding to
primary tumour. Although MRI can detect EMVI,
an absence of suspicious features and the highest
only recently have the radiological characteristics of
score (4) corresponding to the most overt features
EMVI been documented adequately. This characteri-
(Figure 4.10).
zation is important as it is not always possible to de-
A subsequent study comparing MRI and his-
termine with absolute certainty whether a structure
topathological assessment of EMVI has demon-
is vascular. Larger vessels on T2-weighted images
strated sensitivity and specificity of MRI to be 62
may appear black due to signal void, while smaller
per cent and 88 per cent, respectively (Figure 4.11).36
vessels may be recognized because of tortuosity and
Furthermore, relapse-free survival at 3 years was
branching, but there is a general lack of consistency
reported at 35 per cent for patients with an MRI-
of radiological features. In response to this, Smith
EMVI score of 3–4 compared with 74 per cent for
and colleagues defined four criteria by which MRI-­
patients with an MRI-EMVI score of 0–2. Amalga-
predicted EMVI should be assessed:35
mating five separate scores (0, 1, 2, 3, 4) into these
l Pattern of tumour margin: tumour invasion into two broader groups (0–2, 3–4) achieved a similar
small veins that radiate outward from the bowel prediction of relapse-free survival as histopatho-
wall produces a nodular border that can be logical assessment of EMVI (34 per cent versus 74
distinguished from desmoplasia, which appears per cent). MRI-predicted EMVI can therefore help
as fine stranding of low-signal intensity. predict disease recurrence and is an important part

HEBK001-C04_p58-70.indd 65 08/02/13 6:41 PM


66  MRI staging of rectal cancer

MRI-
EMVI Imaging features Illustration
score

Minimal extramural stranding/nodular


1 extension seen, but not in the vicinity
of any vascular structures.

Stranding demostrated in the vicinity


of extramural vessels, but these
2 vessels are of normal calibre, and
there is no definite tumour signal
seen within the vessel.

Intermediate signal intensity apparent


3 within vessels, although the contour
and calibre of these vessels is only
slightly expanded

Obvious irregular vessel contour or


4 nodular expansion of vessel by definite
tumour signal.

Figure 4.10. Five-point grading system for magnetic resonance imaging-predicted extramural vascular invasion.

of preoperative staging. Although EMVI does pre- higher incidence of PSW disease compared with
dict for an increased risk of local recurrence, the real mid- and upper tumours,37,38 thought to be related
issue is the very high risk of systemic recurrence; in to differing patterns of lymphatic drainage.
future, patients with MRI-detected EMVI may be
treated with neoadjuvant chemotherapy rather than
neoadjuvant chemoradiotherapy. Low Rectal Cancer and an
It has also been demonstrated, however, that a Anatomically Based Staging System
greater proportion of patients with rectal cancer
with MRI-predicted EMVI also have MRI-predicted Low rectal cancer, as defined by a tumour with
pelvic side-wall disease. As alluded to earlier, af- its lower edge at or below the origin of the leva-
fected PSW nodes are generally associated with tors at the pelvic side-wall, usually corresponds to
disseminated systemic disease and in the West are a tumour within 6 cm from the anal verge. At this
an indication for neoadjuvant therapy. Lower rec- level, the mesorectum tapers out and the safety net
tal tumours in particular are noted to have a much of the mesorectum is lost. Thus, tumours at this

HEBK001-C04_p58-70.indd 66 08/02/13 6:41 PM


STAGING RECTAL TUMOURS  67

Figure 4.11. Magnetic resonance imaging (MRI) and histopathological assessments of MRI.

level are more likely to involve the sphincters and The complexity of low rectal cancer has stimu-
neighbouring structures, and surgical treatment lated increasing interest in assessment and manage-
is more problematic. For example, extramural ex- ment of cancers in the distal rectum. At the time
tension located anteriorly is likely to infiltrate the of writing, the English National Cancer Action
prostate and may require clearance of the anterior Team has funded the Low Rectal Cancer National
compartment; similar extramural extension locat- Development Programme aiming to improve can-
ed posteriorly may be amenable to surgical cure by cer outcomes and quality of life in patients with
conventional TME, even for low tumours. Indeed, low rectal cancer. Some of the key objectives are
although preoperative staging techniques are lim- to enhance patient-centred multidisciplinary team
ited in differentiating between T2 and early T3 tu- decision-making to improve patient outcome and
mours, this has minimal therapeutic implications quality of life by optimal preoperative therapy and
in the mid- and upper rectum, given that both have surgical technique. Shihab and colleagues describe
a good prognosis with optimal curative surgery and a two-plane approach to low rectal cancer – the
neoadjuvant therapy is not generally required. The ­mesorectal and extralevator planes.15,16 In response
low rectum is more complex because of anatomical to the apparent oncological inferiority of the tra-
constraints and the lack of mesorectum as a natural ditional abdominoperineal excision compared with
barrier and safety net. low anterior resection in the management of low
Unsurprisingly, interest is growing in anatomically rectal cancer, the authors proposed a modified ap-
based staging classifications that can accurately pre- proach to abdominoperineal excision, as outlined
dict whether a tumour-free circumferential resection in Chapter 8. Histopathological studies have dem-
margin is likely to be achieved or not, regardless of the onstrated a reduction in circumferential resection
T stage. This would enable discrimination between margin positivity and specimen perforation rates
patients with minimal mesorectal infiltration not re- with this approach,17 but possibly more perineal
quiring neoadjuvant therapy and those with threat- wound complications,18 highlighting the impor-
ened or infiltrated mesorectal fascia who would ben- tance of appropriate patient selection.
efit from neoadjuvant therapy. Furthermore, such a Shihab and colleagues advise that for low tumours
system may help identify patients in whom a modi- located above the sphincters and more than 1 mm
fied surgical approach is required such as the extra- from the mesorectal fascia, the mesorectal plane
levator abdominoperineal excision (Figure 4.12) (see may be appropriate and adequate.15,16 Similarly, for
Chapter 8, p. 125). tumours at the anal canal, the mesorectal plane is

HEBK001-C04_p58-70.indd 67 08/02/13 6:41 PM


68  MRI staging of rectal cancer

Resection lines
Level at which resection lines meet
Figure 4.12. The extralevator abdominoperineal excision, whereby perineal dissection is performed outside the levators,
which are divided close to their origin at the pelvic side-wall, with removal en bloc of the rectum and anus.

safe for those that show partial invasion or less of muscle or invade the full thickness of the internal
the internal sphincter – that is, the intersphincteric anal sphincter, or beyond this, at the level of the
plane is not involved or threatened (Figure 4.13). sphincters.
Although the extralevator abdominoperineal exci- Given that MRI can often accurately depict com-
sion remains a topic for debate, the authors seek to ponent parts of the sphincter complex and lower
define the excision planes for this operation and, rectum described earlier, a novel staging system for
in so doing, offer appropriate selection criteria; the low rectal cancer has been proposed.15,16 In keeping
main indication is for low rectal tumours above the with the aforementioned surgical planes, this sys-
sphincters that are predicted to invade the levator tem divides the lower rectum into suprasphincteric

Mesorectal
fascia

Levator
1
insertion

Puborectalis and
Intersphinteric external sphincter
Figure 4.13. A novel magnetic plane
resonance imaging-based staging
2
system for low rectal cancer based Internal
sphincter
on key anatomical landmarks of the
sphincter complex.

HEBK001-C04_p58-70.indd 68 08/02/13 6:41 PM


References  69

and sphincteric compartments, with clear selec- excision of the rectum. AJR Am J Roentgenol 2004;
tion criteria as to which plane of surgery may be 182: 431–9.
  5. Burton S, Brown G, Daniels IR, et al. MRI directed
appropriate, safe and effective. Retrospective evalu-
multidisciplinary team preoperative treatment strategy:
ation of this staging system has yielded promising the way to eliminate positive circumferential margins?
results, and prospective larger-scale studies are un- Br J Cancer 2006; 94: 351–7.
der way. It is hoped that this novel MRI-based stag-   6. Dukes CE. The classification of cancer of the rectum.
ing system will help reduce circumferential resec- J Pathol Bacteriol 1932; 35: 323.
tion margin positivity for low rectal cancers by the   7. Cawthorn SJ, Parums DV, Gibbs NM, et al. Extent of
selective use of neoadjuvant therapy and planned mesorectal spread and involvement of lateral resection
margin as prognostic factors after surgery for rectal
surgery.
cancer. Lancet 1990; 335: 1055–9.
  8. Pollheimer MJ, Kornprat P, Pollheimer VS, et al. Clini-
cal significance of pT sub-classification in surgical
CONCLUSION pathology of colorectal cancer. Int J Colorectal Dis
2010; 25: 187–96.
The development of high-resolution MRI surface-coil   9. Beets-Tan RG, Beets GL, Vliegen RF, et al. Accu-
systems has resulted in accurate preoperative staging, racy of magnetic resonance imaging in prediction of
which has marked a new era in rectal cancer manage- tumour-free resection margin in rectal cancer surgery.
Lancet 2001; 357: 497–504.
ment. Since the widespread adoption of TME, more
10. Gagliardi G, Bayar S, Smith R, Salem RR. Preopera-
attention is being devoted to staging systems that can tive staging of rectal cancer using magnetic resonance
accurately predict circumferential resection margin imaging with external phase-arrayed coils. Arch Surg
status and reliably determine selective use of neoad- 2002; 137: 447–51.
juvant therapy or suggest an alternative surgical ap- 11. Blomqvist L, Holm T, Rubio C, Hindmarsh T. Rectal tu-
proach. Despite its limitations in T staging, MRI is mours: MR imaging with endorectal and/or phased-array
currently the only imaging modality capable of de- coils, and histopathological staging on giant sections – a
comparative study. Acta Radiol 1997; 38: 437–44.
picting the topographic relationship between tumour
12. Laghi A, Ferri M, Catalono C, et al. Local staging of
and the proposed surgical resection margin. Low rectal cancer with MRI using a phased array body coil.
rectal cancer in particular is more complex and with Abdom Imaging 2002; 27: 425–31.
worse outcomes than middle and upper rectal cancer 13. MERCURY Study Group. Extramural depth of tumor
and is currently the focus of ongoing attempts to im- invasion at thin–section MR in patients with rectal can-
prove all aspects of management, including improved cer: results of the MERCURY study. Radiology 2007;
MRI staging based on key anatomical landmarks. Op- 243: 132–9.
14. Klessen C, Rogalla P, Taupitz M. Local staging of rectal
timal staging, enhanced decision-making regarding
cancer: the current role of MRI. Eur Radiol 2007; 17:
preoperative therapy and optimal surgery will lead to 379–89.
improved oncological outcomes. 15. Shihab OC, Heald RJ, Rullier E, et al. Defining the
surgical planes on MRI improves surgery for cancer of
the low rectum. Lancet Oncol 2009; 10: 1207–11.
References 16. Shihab OC, Moran BJ, Heald RJ, Quirke P, Brown G.
MRI staging of low rectal cancer. Eur Radiol 2009; 19:
  1. Heald RJ, Husband EM, Ryall RD. The mesorectum in 643–50.
rectal cancer surgery: the clue to pelvic recurrence? 17. West NP, Finan PJ, Anderin C, et al. Evidence of the
Br J Surg 1982; 69: 613–6. oncologic superiority of cylindrical abdominoperineal
  2. Wibe A, Møller B, Norstein J, et al. A national strategic excision for low rectal cancer. J Clin Oncol 2008; 26:
change in treatment policy for rectal cancer implemen- 3517–22.
tation of total mesorectal excision as routine treatment 18. West NP, Anderin C, Smith KJ, et al. Multicentre
in Norway: a national audit. Dis Colon Rectum 2002; experience with extralevator abdominoperineal
45: 857–66. excision for low rectal cancer. Br J Surg 2010; 97:
  3. Heald RJ, Smedh RK, Kald A, Sexton R, Moran BJ. 588–99.
Abdominoperineal excision of the rectum: an 19. Dukes CE, Bussey HJ. The spread of cancer and its
endangered operation. Norman Nigro Lectureship. Dis effect on prognosis. Cancer 1958; 12: 309–20.
Colon Rectum 1997; 40: 747–51. 20. Kim NK, Kim MJ, Yun SH, Sohn SK, Min JS. Com-
  4. Brown G, Kirkham A, Williams GT, et al. High-resolution parative study of transrectal ultrasonography, pelvic
MRI of the anatomy important in total mesorectal computerized tomography, and magnetic resonance

HEBK001-C04_p58-70.indd 69 08/02/13 6:41 PM


70  MRI staging of rectal cancer

imaging in preoperative staging of rectal cancer. Dis 30. Talbot IC, Ritchie S, Leighton MH, et al. Spread of rec-
Colon Rectum 1999; 42: 770–5. tal cancer within veins: histologic features and clinical
21. Chan TW, Kressel HY, Milestone B, et al. Rectal carci- significance. Am J Surg 1981; 141: 15–17.
noma: staging at MR imaging with endorectal surface 31. Dukes C, Bussey HJ. Venous spread in rectal cancer.
coil – work in progress. Radiology 1991; 181: 461–7. Proc R Soc Med 1941; 34: 571–4.
22. Schnall MD, Furth EE, Rosato EF, Kressel HY. Rectal 32. Knudsen JB, Nilsson T, Sprechler M, Johansen A,
tumor stage: correlation of endorectal MR imaging and Christensen N. Venous and nerve invasion as prog-
pathologic findings. Radiology 1994; 190: 709–14. nostic factors in postoperative survival of patients with
23. Brown G, Richards CJ, Bourne MW, et al. Morphologic resectable cancer of the rectum. Dis Colon Rectum
predictors of lymph node status in rectal cancer with 1983; 26: 613–7.
use of high-spatial-resolution MR imaging with his- 33. Krasna MJ, Flancbaum L, Cody RP, Schneibaum S,
topathologic comparison. Radiology 2003; 227: 371–7. Ben Ari G. Vascular and neural invasion in colorectal
24. Adam IJ, Mohamdee MO, Martin IG, et al. Role of carcinoma: incidence and prognostic significance.
circumferential margin involvement in the local recur- Cancer 1988; 61:1018–23.
rence of rectal cancer. Lancet 1994; 344: 707–11. 34. Sunderland D. The significance of vein invasion by
25. Shihab OC, Quirke P, Heald RJ, Moran BJ, Brown G. cancer of the rectum and sigmoid: a microscopic
Magnetic resonance imaging-detected lymph nodes study of 210 cases. Cancer 1949; 2: 429–37.
close to the mesorectal fascia are rarely a cause of 35. Smith NJ, Shihab O, Arnaout A, Swift RI, Brown G.
margin involvement after total mesorectal excision. Br MRI for detection of extramural vascular invasion in
J Surg 2010; 97: 1431–6. rectal cancer. Am J Roent 2008; 191: 1–6.
26. Yano H, Moran BJ. The incidence of lateral pelvic side- 36. Smith NJ, Barbachano Y, Norman AR, Swift RI,
wall nodal involvement in low rectal cancer may be simi- Abulafi M, Brown G. Prognostic significance of
lar in Japan and the West. Br J Surg 2008; 95: 33–49. magnetic resonance imaging-detected extramural
27. Bokey EL, Chapuis PH, Dent OF, et al. Factors affecting vascular invasion in rectal cancer. Br J Surg 2008;
survival after excision of the rectum for cancer: a multi- 95: 229–36.
variate analysis. Dis Colon Rectum 1997; 40: 3–10. 37. Takahashi T, Ueno M, Azekura K, Ohta H. Lateral node
28. Horn A, Dahl O, Morild I. Venous and neural invasion dissection and total mesorectal excision for rectal can-
as predictors of recurrence in rectal adenocarcinoma. cer. Dis Colon Rectum 2000; 43(10 suppl.): S59–68.
Dis Colon Rectum 1991; 34: 798–804. 38. Ueno M, Oya M, Azekura K, Yamaguchi T, Muto T.
29. Harrison JC, Dean PJ, el-Zeky F, Van der Zwaag R. From Incidence and prognostic significance of lateral lymph
Dukes through Jass: pathological prognostic indicators in node metastasis in patients with advanced low rectal
rectal cancer. Hum Pathol 1994; 25: 498–505. cancer. Br J Surg 2005; 92: 756–63.

HEBK001-C04_p58-70.indd 70 08/02/13 6:41 PM


5
Radiological staging for
systemic disease
Gina Brown and Chris Hunter

Introduction antigen (CEA) alone, and 5-year survival is


improved from 9 per cent to 13 per cent.9
The early accurate identification of metastatic The patient group in which metastasecto-
disease is becoming increasingly important my may be indicated also continues to expand.
in rectal cancer, in both the preoperative and Improvements in surgical technique have result-
follow-up settings. A proportion of patients ed in low operative morbidity and mortality asso-
undergoing resection of metastatic disease now ciated with metastasectomy, and metastasectomy
achieve long-term cure. 1 It is now well estab- is increasingly offered to patients over 70 years
lished that patients undergoing surgery for of age. Even after metastasectomy, continued
resectable liver metastases achieve a 5-year sur- surveillance for further metastatic disease is of
vival in the region or in excess of 40 per cent, benefit, as repeat metastasectomy may improve
compared with almost no survivors at 5 years survival.10,11
for untreated patients.1–4 The proportion of In a proportion of patients, even with optimal
patients in whom long-term cure is achieved surveillance, metastatic disease will be inoper-
following metastasectomy has increased over able at the time of diagnosis. Patients with unre-
time, with more recent studies demonstrating sectable metastatic disease treated with palliative
5-year survival approaching 50 per cent in some chemotherapy have better survival at 6 months and
patients. 5 This is due in part to more accurate 12 months, however, compared with patients
preoperative imaging allowing better patient treated with supportive care alone.12 Patients with
selection.6,7 unresectable metastatic disease may also be selected
The early detection of metastatic disease for palliative rather than aggressive surgical treat-
increases the likelihood of identifying resectable ment. Surgical palliation is an effective treatment
disease. In the follow-up setting, it has been shown that alleviates symptoms, increases survival over
that the early detection of metastatic disease is of non-­surgical supportive care, and may require only
benefit in addition to informing patients of prog- a short hospitalization.13
nosis.8 Intensive follow-up regimes that include In this chapter we discuss the common mecha-
computed tomography (CT) identify local or nisms and sites of metastasis in rectal cancer, and
distant recurrence 8.5 months earlier than those the modalities available to identify metastases to
using clinical follow-up and carcinoembryonic these sites.

HEBK001-C05_p71-86.indd 71 08/02/13 5:59 PM


72  Radiological staging for systemic disease

Mechanisms of spread of cancer, and a further 10.5 per cent of patients devel-
colorectal metastases oped metachronous metastases within 3 years.21 As
well as being the most common site of visceral metas-
Lymphatic Spread tases in colorectal cancer, the liver is the only site of
visceral metastases in 30–40 per cent of patients.
Lymph nodes are the most common site of metas- Other, less common sites of distant metastases
tasis in rectal cancer, with lymph node metastases in colorectal cancer are the lung (11 per cent of
occurring in 51.9 per cent of 1732 patients under- patients have synchronous metastases and 5.8 per
going resection of their primary rectal cancer in cent of patients develop metachronous metastases
the Dukes series.14 The incidence of lymph node within 5 years),22 ovary (2–8 per cent of women
metastases increases with increasing histological within 5 years),23,24 bone (5 per cent) and brain
grade of the primary tumour, and with more local- (2 per cent).25
ly advanced tumours. Lymph node metastases usu- Other sites of colorectal metastases are rare but
ally spread progressively along the lymphatic chain include the spleen, kidneys, pancreas, adrenals,
from lymph node to lymph node. breast, thyroid and skin. It appears, however, that
In rectal cancer, lymphatic spread is initially to unusual sites of metastasis are becoming more
mesorectal lymph nodes adjacent to the primary common with the increased use of systemic agents.
tumour. As metastases move up the lymph node There also appears to be a relationship to the
chain, they may follow the inferior or middle rec- number of systemic therapies received.26
tal arteries to pelvic side-wall lymph nodes, par-
ticularly in low rectal cancer (usually within 6 cm
from the anal verge), and then the internal iliac
Transcoelomic Spread
lymph node chain. The main lymphatic drainage is
The third mechanism of metastasis encountered in
to nodes along the superior rectal artery and thence to
colorectal cancer is spread of tumour cells across
nodes around the inferior mesenteric artery.15
the peritoneal cavity. This usually occurs when
If metastases block the usual lymphatic drain-
tumour has invaded through the peritoneum (T4
age, then retrograde spread of metastases through
disease). This may be apparent on preoperative
the lymphatics may occur. This usually occurs only
imaging, macroscopically at the time of operation,
in advanced tumours with multiple lymph node
or on histopathological assessment postoperatively.
metastases and is one of the mechanisms for spread
Occasionally, this form of spread may result from
to pelvic side-wall and inguinal nodes.16 Spread
peritoneal spillage of tumour cells at the time of
to inguinal lymph nodes in rectal cancer is rare
surgical resection.
(occurring in approximately 2 per cent of patients)
Peritoneal deposits have a predilection for
and usually occurs only in low rectal cancers invad-
certain sites within the abdominal cavity; these
ing the anal canal.
include the superior and inferior paracolic
recesses, the rectovesical pouch (pouch of Doug-
Haematogenous Spread las), the under surface of the diaphragm and the
transverse mesocolon.27–29
Extramural vascular invasion has been known to
be a predisposing factor for the development of
distant metastases for some time.17–20 It is probable SITES OF METASTASES
that haematogenous spread is the main route for IN COLORECTAL CANCER
visceral metastases.
It is therefore unsurprising that the most common Liver Metastases
site for colorectal metastases is the liver, as the venous
drainage of the colon and rectum is primarily via As mentioned previously, the liver is the most com-
the portal vein. In a population-based study of 1325 mon site for visceral colorectal metastases.
patients, 18.8 per cent of patients had liver metastases Figure 5.1 shows a contrast-enhanced CT demon-
occurring within 6 months of diagnosis of colorectal strating liver metastases. Over the period from 1984

HEBK001-C05_p71-86.indd 72 08/02/13 5:59 PM


SITES OF METASTASES IN COLORECTAL CANCER   73

of 24–59 months can be achieved in unresectable


colorectal liver metastases.41 This compares favour-
ably with median overall survival of 16–24 months
with chemotherapy alone. Figure 5.2 shows a hepatic
metastasis following RFA. Portal vein embolization
may be used to induce liver hypertrophy and allow
extended hepatectomy in patients who would other-
wise have insufficient functional reserve.42
A number of different studies have identified
risk factors that predict outcome after resection of
liver metastases. One of the largest studies includ-
ed more than 1000 patients and identified the fol-
lowing adverse prognostic factors: size of metasta-
sis .5 cm, potential involvement of the resection
margin, more than one liver metastasis, poor
prognosis primary, synchronous primary tumour
and liver metastases within 12 months, and extra-
hepatic metastatic disease. Overall 5-year survival
Figure 5.1. Contrast-enhanced computed tomography
was 60 per cent with none of these risk factors,
demonstrating liver metastases (arrowheads).
falling to less than 20 per cent with four or more
risk factors.43
to 2005, 5-year overall survival for patients undergo- It is therefore very important to have accurate
ing resection of hepatic metastases improved from preoperative imaging before treatment for liver
23 per cent to 46 per cent, despite patients undergo- metastases for careful patient selection. The aims of
ing operations with more severe disease.30 imaging for patients undergoing resection of liver
Improved survival following treatment for metastases are:
colorectal liver metastases has been attributed to
l to accurately demonstrate anatomical distribu-
a number of different factors, including improved
tion of liver metastases and segmental sparing;
techniques for anatomical resection,1,2,4 increased
l to exclude widespread micro-metastatic disease
use of intraoperative ultrasound,31,32 reduction in
within the liver;
perioperative morbidity and mortality,3 second
l to exclude extra-hepatic metastases;
resections for recurrent hepatic metastases,33 and
l to discriminate between benign and malignant
the use of chemotherapy.34–37
liver lesions;
Recent developments in interventional radiology
l to stratify patients in terms of prognosis follow-
have also had a significant impact on the treatment of
ing metastasectomy.
colorectal liver metastases. Radiofrequency ablation
(RFA) now has a significant role. Although there is an When assessing patients preoperatively for occult
absence of evidence from randomized controlled tri- intra- or extra-hepatic malignancies, it is worth
als, due in part to the fact that the two attempted ran- bearing in mind that the risk is strongly related to
domized controlled trials of RFA closed early due to the number and distribution of hepatic metastases;
poor recruitment, there is a growing body of evidence both occult intra-hepatic metastases and extra-
from clinical series and non-randomized compara- hepatic metastases are rare with solitary unilobar
tive studies that supports the use of RFA. Although metastases but much more common with multiple
resection of colorectal liver metastases remains the bilobar metastases.44
treatment of choice in suitable patients, RFA is a use-
ful additional treatment modality for metastases of
less than 3–4 cm in size, with local recurrence rates Lung Metastases
of 6.7–17 per cent.38–40 Mean and 5-year survival
with combined chemotherapy and RFA are better The lungs are the second most common site of
than with either modality alone, and median survival metastasis in rectal cancer. If resection can be

HEBK001-C05_p71-86.indd 73 08/02/13 5:59 PM


74  Radiological staging for systemic disease

Figure 5.2. Liver magnetic resonance imaging demonstrating two metastases (in left lobe, black arrow; in right lobe,
white arrow. (A) Before treatment; (B) following chemotherapy; (C) following resection of larger metastasis; (D) following
radiofrequency ablation of smaller metastases.

undertaken safely, then excellent outcomes can this results in the identification of more benign
be achieved, with 5-year survival of 55.4–71 per lesions, a baseline CT and serial examinations allow
cent.45,46 Patients undergoing repeat thoracotomy for indeterminate lesions to be interpreted more confi-
recurrent pulmonary metastases may also achieve dently during follow-up.
good outcomes, but the presence of hilar or medias-
tinal lymph node involvement or extra-pulmonary
metastases is associated with a poor survival rate.11 Ovarian Metastases
As with liver metastases, it is therefore important to
identify lesions early, when curative resection may Ovarian metastases occur in 6–8 per cent of women
still be achieved, and to exclude poor prognosis fea- with colorectal cancer, making the ovaries the third
tures such as extra-pulmonary metastases and hilar most common site of visceral metastases in women.
or mediastinal lymph node involvement. Radiologically, colorectal ovarian metastases may
Lung metastases usually present initially as small be easily mistaken for primary mucinous adenocar-
nodules, which are often less than 5 mm in diam- cinoma of the ovary.48 It is therefore important that
eter. Multidetector CT allows early detection and is the reporting radiologist is aware of any current or
superior to any other modality in the detection of previous diagnosis of colorectal cancer when inter-
these small lesions.47 Figure 5.3 illustrates pulmo- preting radiological imaging. Colorectal ovarian
nary metastases evident on thoracic CT. Although metastases may appear as large oval or lobulated,

HEBK001-C05_p71-86.indd 74 08/02/13 5:59 PM


IMAGING MODALITIES AVAILABLE FOR STAGING SYSTEMIC DISEASE  75

therefore usually occur in the presence of T4b


disease, where the serosa has been breached by
tumour.
The pattern of peritoneal spread is related to
tumour grade, with poorly differentiated tumours
tending to produce diffuse seeding and well-differen-
tiated tumours being more likely to produce solitary
deposits. Peritoneal metastases are the most common
cause of ureteric obstruction due to colorectal cancer
and should be considered when unexplained ureteric
obstruction is identified in patients with colorectal
cancer.
Although peritoneal metastases are usu-
ally managed palliatively, very carefully selected
patients may have a good outcome if treated sur-
gically,51,52 and so patients should be discussed in
a multidisciplinary setting if peritoneal disease
is identified. Figure 5.5 demonstrates peritoneal
Figure 5.3. Contrast-enhanced computed tomography
demonstrating lung metastasis (arrowhead).
metastases on CT.

cystic or solid ovarian masses.49,50 Figure 5.4 shows IMAGING MODALITIES AVAILABLE
a typical ovarian metastasis on CT. FOR STAGING SYSTEMIC DISEASE

The list of imaging modalities available for the


Peritoneal Metastases systemic staging of rectal cancers continues to
increase. No single imaging modality is currently
Peritoneal metastases occur through spread of optimal for identifying all distant metastases, how-
tumour cells across the peritoneal cavity. They ever, so a multimodality approach is necessary.

Figure 5.4. Unenhanced computed tomography Figure 5.5. Contrast-enhanced computed tomography
demonstrating bilateral ovarian metastases (arrowheads). demonstrating peritoneal metastases (arrowheads).

HEBK001-C05_p71-86.indd 75 08/02/13 5:59 PM


76  Radiological staging for systemic disease

Computed Tomography multiple phases of enhancement in the same sitting,


by repeating images through the liver at different
Computed tomography of the thorax, abdomen time delays after the injection of contrast. There is
and pelvis is the most common imaging technique little evidence, however, that arterial phase imaging
used for identifying distant metastases in rectal is of significant benefit in colorectal metastases.57,58
cancer, in both primary staging and follow-up. It is Colorectal liver metastases typically appear as
widely available, rapid and relatively cheap. hypointense lesions with a rim of enhancement.
Despite the fact that colorectal cancer metasta- Occasionally they may contain central necrosis
sizes to the liver via the portal vein, colorectal liver with a low-density ‘cystic’ centre. Liver cysts are the
metastases derive their blood supply from the hepat- most common benign lesions identified. These are
ic artery.53 This fact can be exploited in the iden- well-defined lesions with no rim of enhancement,
tification of liver metastases: liver CT performed no internal structure and very low attenuation.59
during peak hepatic enhancement will identify Although it is often relatively easy to characterize
most colorectal liver metastases due to their rela- larger lesions, small liver lesions (, 10 mm) may be
tive hypovascularity in this phase of enhancement. more challenging. In these circumstances, interval
A contrast-enhanced CT demonstrating hepatic scanning or review of serial imaging is very impor-
metastases is shown in Figure 5.1. This feature can tant. A 3 month interval CT scan improves specifi-
be exploited further using CT portography. In this city from 90 per cent to 99 per cent.60
technique, the superior mesenteric artery is selec- A meta-analysis from 2010 found that the per-
tively catheterized, and CT scanning of the liver is patient sensitivity of CT for detecting liver metas-
performed approximately 60 s after the adminis- tases was 83.6 per cent.61 Computed tomography
tration of contrast material via the catheter. This is therefore a good baseline screening investigation
technique increases the sensitivity and specificity of for distant metastases in patients with rectal cancer,
CT in the identification of colorectal metastases54 in both the preoperative and follow-up settings. It
and has been used in the preoperative assessment may also identify patients with unresectable meta-
of patients being considered for hepatic metastasec- static disease who do not require further imaging.
tomy to delineate the disease extent. The technique In this study, however, the sensitivity of magnetic
is invasive, however, and so has not been used in the resonance imaging (MRI) and 18F-fluorodeoxy-
routine primary staging or follow-up of patients glucose positron-emission tomography (18F-FDG-
with rectal cancer. PET) were both higher than that of CT. Computed
The steady improvements in CT technology tomography also has limited ability to identify
have also increased its accuracy in identifying liv- small-volume peritoneal and liver surface disease.
er metastases. Improved sensitivity and specificity Computed tomography is largely limited to size cri-
in detecting colorectal liver metastases have been teria for the identification of distant nodal metas-
attributed to the use of helical CT with 5-mm tases, which has limited sensitivity. Therefore, when
­collimation.55 Using this technique in the preopera- planning metastasectomy, or if there is a high index
tive assessment of patients undergoing resection of of suspicion of metastatic disease and a normal CT,
liver metastases, 94 per cent of patients selected for other imaging modalities should also be employed.
resection by CT were resectable at operation, and
4-year survival was 58 per cent. Multidetector CT
represents further technological improvement and 18F-Fluorodeoxyglucose
is accurate in detecting lesions over 10 mm in size, Positron-emission Tomography
with 1 mm isotropic voxels giving improved spatial
resolution and the ability to reconstruct images in 18F-Fluorodeoxyglucose positron-emission tom-
any plane.56 This has largely removed the need for ography is a form of functional imaging that uses
CT portography, even in the preoperative assess- an 18F labelled tracer, which is taken up by the glu-
ment for metastasectomy. cose receptor but not metabolized. The tracer accu-
The advent of multidetector CT has also allowed mulates in cells with increased glucose metabolism
imaging of the whole liver in around 10 s. Multi- and upon decay releases high-energy photons,
detector CT allows assessment of the liver during which can be detected by dedicated receptors. As

HEBK001-C05_p71-86.indd 76 08/02/13 5:59 PM


IMAGING MODALITIES AVAILABLE FOR STAGING SYSTEMIC DISEASE  77

increased glucose metabolism is exhibited by many disease in the presence of normal or equivocal ana-
tumour cells, this technique can be used to identify tomical imaging and endoscopy and a high index of
primary and metastatic disease in rectal cancer. suspicion of recurrent rectal cancer, for example in
There has been much interest in the use of 18F-FDG- the presence of rising CEA. In these circumstanc-
PET to identify metastatic disease in colorectal cancer. es, Flamen and colleagues found that 18F-FDG-
In 2005, a meta-analysis of 61 eligible articles sug- PET had a per-lesion sensitivity of 75 per cent,67
gested that on a per-patient basis, 18F-FDG-PET although it should be noted that in this study 38 per
was more sensitive than CT or MRI in detecting liver cent of patients had equivocal findings on conven-
metastases, with a sensitivity of 94.6 per cent, although tional imaging and 70 per cent had not undergone
on a per-lesion basis superparamagnetic iron oxide chest CT.
(SPIO)-enhanced MRI was more sensitive.62 Similar Despite the apparent superiority of systemic stag-
results were found in a further meta-analysis in 2010, ing incorporating 18F-FDG-PET, very few stud-
where the sensitivity of 18F-FDG-PET for detect- ies have looked at the impact of 18F-FDG-PET on
ing colorectal liver metastases on a per-patient basis the primary staging of rectal cancer. Heriot and
was 94.1 per cent and significantly higher than CT or colleagues assessed this issue in 46 patients with
MRI;61 again, on a per-lesion basis, there was no sig- advanced primary rectal cancer and found that man-
nificant difference between MRI and 18F-FDG-PET, agement was changed in 17 per cent of patients,68
and MRI was significantly better than CT in detecting although again it should be noted that patients did
lesions under 1 cm in size. not undergo routine preoperative thoracic CT.
A number of studies have also demonstrated There are a number of potential limitations in
18F-FDG-PET to be accurate in the identification 18F-FDG-PET staging of rectal cancer. Establishing
of extra-hepatic disease. In a meta-analysis, PET/ the precise anatomical location of increased FDG
CT had a sensitivity of 91.2 per cent and a specifi- uptake may be difficult, and because of this it is not
city of 95.4 per cent in identifying extra-hepatic always possible to identify the anatomical location of
metastases.63 liver metastases using 18F-FDG-PET. Physiological or
As a result of this high sensitivity for meta- benign increased FDG uptake may create false posi-
static disease in rectal cancer, 18F-FDG-PET tives, such as granulomatous and inflammatory proc-
has been used in the preoperative assessment of esses in the lung. Following radiotherapy, granulation
patients being considered for resection of hepatic tissue, fibroblast and macrophage activity may give
metastases. A number of studies have shown that false positives. This is a particular problem in the first
18F-FDG-PET significantly alters management in 6 months following radiotherapy. The spatial reso-
patients being considered for metastasectomy, pre- lution of 18F-FDG-PET is limited to around 7 mm,
dominantly by identifying additional extra-hepatic which means that lesions need to be about 1 cm or
metastases in 10–21 per cent of patients,64,65 and so larger in size to be identified. This particularly limits
reduces the rate of negative laparotomies due to the ability of 18F-FDG-PET to identify small-volume
unexpected metastatic disease at the time of surgery peritoneal and liver surface disease.
by approximately 9 per cent.63 The identification of Some authors have suggested that the incidence
lung metastases may also allow successful resection of extra-hepatic metastases in patients with a low
of both lung and liver metastases. clinical risk score being considered for hepatic
18F-Fluorodeoxyglucose positron-emission tom- metastasectomy is so low that 18F-FDG-PET is
ography has also been used to identify pelvic recur- of limited value. Indeed, in this group of patients,
rence in rectal cancer. Following surgery, scarring and the use of 18F-FDG-PET may incorrectly upstage
fibrosis can be difficult to distinguish from recurrence patients and prevent them from undergoing poten-
on anatomical imaging, and this problem can be tially curative resection.69
compounded by post-radiotherapy changes. A meta- To reduce the risk of false positives and false
analysis suggested that the sensitivity and specificity of negatives in 18F-FDG-PET imaging, interpretation
18F-FDG-PET in detecting pelvic recurrence in rectal with correct anatomical information from cross-
cancer are both 94 per cent.66 sectional imaging is very important. For this rea-
18F-Fluorodeoxyglucose positron-emission tomo- son, combined 18F-FDG-PET/CT has superseded
graphy is also used widely to help identify metastatic 18F-FDG-PET in many situations.

HEBK001-C05_p71-86.indd 77 08/02/13 5:59 PM


78  Radiological staging for systemic disease

Combined 18F-Fluorodeoxyglucose
Positron-Emission Tomography and
Computed Tomography
Combining CT and 18F-FDG-PET in the same scan-
ner allows attenuation correction for the PET por-
tion of the imaging, but also allows fusion of the
anatomical component of the CT scanning with the
functional element of the 18F-FDG-PET. The risk of
misregistration of PET and CT images is reduced (but
not eliminated) by combining both types of imaging
in the same machine at the same time. It is hoped that
this combined functional and anatomical imaging will
be more accurate in the identification of rectal cancer
metastases than either imaging modality alone.70
Niekel and colleagues noted in a meta-analysis
that there were not enough studies of 18F-FDG-
PET/CT in the detection of liver metastases to Figure 5.7. Combined 18F-fluorodeoxyglucose positron-
comment on its accuracy.61 In a single series of emission tomography and computed tomography
467 patients, however, Orlacchio and colleagues demonstrating internal iliac lymph node metastasis
have suggested that the combined 18F-FDG-PET/ (arrowhead).
CT modality is more accurate than either modality
alone in identifying colorectal liver metastases, with Davey and colleagues assessed the impact of
reported per-patient sensitivity and specificity of 18F-FDG-PET/CT on the management of 83
98 per cent.71 An 18F-FDG-PET/CT scan demon- patients with primary rectal cancer and found that
strating liver metastases is illustrated in Figure 5.6, management was altered in 12 per cent.72
outlining lymph node metastases in Figure 5.7, and
bilateral ovarian metastases in Figure 5.8.

Figure 5.8. Combined 18F-fluorodeoxyglucose positron-


emission tomography (FDG) and computed tomography
Figure 5.6. Combined 18F-fluorodeoxyglucose positron- demonstrating bilateral ovarian metastases (white
emission tomography and computed tomography arrowheads). Physiological FDG uptake is seen in the right
demonstrating liver metastases (arrowheads). ureter (black arrowhead).

HEBK001-C05_p71-86.indd 78 08/02/13 5:59 PM


IMAGING MODALITIES AVAILABLE FOR STAGING SYSTEMIC DISEASE  79

Pelvic Magnetic Resonance Imaging

As discussed in Chapter 4, pelvic MRI is used pre-


dominantly for the local staging of rectal cancer
and is very accurate for this purpose. In some
instances, however, pelvic MRI may also demon-
strate systemic disease in rectal cancer. Ovarian
metastases may be evident on pelvic MRI; they
most commonly take the form of solid adnexal
masses with intratumoural cysts, although they
may also be predominantly solid or predominant-
ly cystic lesions.50 Figure 5.9 shows a typical ovar-
ian metastasis on MRI.
Lymph node metastases may also be identified
on pelvic MRI. Lymph node metastases occur-
ring within the mesorectal envelope will usually
be removed at the time of optimal total mesorectal
excision surgery and so can be considered with the Figure 5.10. Magnetic resonance imaging demonstrating
local staging. Distant lymph node metastases may internal iliac lymph node metastasis (arrowhead).
also be identified along the internal iliac chain or
less commonly in the inguinal lymph node groups, Peritoneal disease may also be detected and on
however. Although overlap between benign and T2-weighted MRI has an intermediate signal inten-
malignant lymph nodes makes size criteria unre- sity, which appears relatively hyperintense com-
liable, morphological changes such as an irregular pared with the low-signal peritoneum. Figure 5.11
border or inhomogeneous signal intensity have a illustrates peritoneal disease on T2-weighted MRI.
high positive predictive value, and lymph nodes If peritoneal disease is suspected, fat-suppressed
demonstrating these characteristics should be treat- T1-weighted imaging with gadolinium enhance-
ed as metastatic disease.73 Figure 5.10 illustrates a ment may be of benefit in highlighting mesenteric
typical lymph node metastasis identified on MRI. deposits.74

Figure 5.9. Magnetic resonance imaging demonstrating Figure 5.11. Magnetic resonance imaging demonstrating
bilateral ovarian metastases (arrowheads). peritoneal metastases (arrowhead).

HEBK001-C05_p71-86.indd 79 08/02/13 5:59 PM


80  Radiological staging for systemic disease

Hepatic Magnetic Resonance Imaging Additional sequences are useful to help with
lesion characterization and thus help to determine
Dedicated liver MRI is a highly accurate modality for whether lesions represent colorectal liver metastas-
identifying colorectal metastases to the liver. A meta- es. In-phase and out-of-phase T1-weighted imaging
analysis of CT, MRI and PET/CT found that on a per- may help assess areas of focal fatty infiltration, which
lesion basis, for lesions over 1 cm in size, MRI was the may appear as apparent ‘perfusion defects’ on CT
most sensitive modality for identifying colorectal liver and mimic colorectal liver metastases. T2-weighted
metastases.62 This study also found that gadolinium- images, which may be acquired as either respiratory-
enhanced and SPIO-enhanced MRI was significantly triggered fast spin echo (FSE) sequences or breath-
more accurate than unenhanced MRI. A more recent hold single-shot FSE ­sequences, further help with
meta-analysis, which included only prospective stud- the characterization of focal liver lesions.
ies, found that on a per-lesion basis, CT, MRI and A number of different contrast agents have been
18F-FDG-PET had comparable sensitivity of 74.4 per used to increase the sensitivity and specificity of
cent, 80.3 per cent and 81.4 per cent. The sensitivity of liver MRI. Dynamic gadolinium contrast enhance-
MRI had increased since 2004, however, and in studies ment can be obtained with images in arterial, por-
since 2004 the sensitivity of MRI on a per-lesion basis tal venous, equilibrium and delayed phases. This is
is 84.9 per cent; MRI is also the most accurate modali- superior to dual-phase contrast-enhanced spiral CT,
ty in identifying lesions less than 10 mm in diameter.61 especially in lesion characterization.76 Superpara-
A paper comparing liver specific contrast-enhanced magnetic iron oxide may also increase sensitivity and
MRI and 18F-FDG-PET/CT suggested that on a per- specificity of liver MRI in detecting colorectal liver
patient basis PET/CT and MRI had equally high sen- metastases. Superparamagnetic iron oxide is taken
sitivity and specificity at 98 per cent and 100 per cent, up by Kupffer cells, rendering the substance of liver
respectively, but on a per-lesion basis for lesions less dark on T1 gradient echo sequences. Liver metastases
than 1 cm MRI was more sensitive.75 without functioning Kupffer cells therefore appear
The mainstay of liver MRI is the T1-weighted relatively hyperintense. Other lesions including cysts
gradient echo sequence. This allows imaging of and cavernous haemangiomas may also be relatively
the whole liver in a single breath-hold, reduc- hyperintense, and so images must be interpreted with
ing motion artefact. T1-weighted gradient echo T2-weighted images to avoid false positives.77 The
sequences can also be acquired as a volume image American Food and Drug Administration has not
and reconstructed in any plane, allowing accurate approved SPIO contrast agents, however, and in 2011
anatomical localization of lesions. Figure 5.12 illus- all previously available SPIO preparations were with-
trates hepatic metastases on a T1-weighted MRI. drawn from the market.

Figure 5.12. Comparison of gadoxetic acid-enhanced magnetic resonance imaging (MRI) and unenhanced T1-weighted
MRI. Gadoxetic acid MRI (left) shows liver metastases (black arrowhead) not seen on unenhanced T1-weighted MRI.
Second metastasis (white arrowhead) is seen on both gadoxetic acid-enhanced and unenhanced MRI.

HEBK001-C05_p71-86.indd 80 08/02/13 5:59 PM


IMAGING MODALITIES AVAILABLE FOR STAGING SYSTEMIC DISEASE  81

Figure 5.13. Comparison of gadoxetic acid-enhanced magnetic resonance imaging (MRI) and contrast-enhanced
computed tomography. Gadoxetic acid MRI (left) shows liver metastasis (arrowhead) not seen on contrast-enhanced CT.

Liver-specific gadolinium-based contrast agents has been exploited in hepatic MRI to help identify
such as gadoxetic acid and gadobenate dimeglu- and characterize metastatic lesions within the liver.
min are taken up by functioning hepatocytes. This Although evidence is still accumulating for this tech-
causes increased signal in the liver parenchyma nique, a number of studies have suggested that the
on T1-weighted images. Metastatic lesions remain addition of DWI sequences to standard liver MRI
hypointense. These liver-specific contrast agents protocols can increase the sensitivity.79,80 Figure 5.14
increase the sensitivity of liver MRI for colorectal illustrates colorectal liver metastases on DWI.
liver metastases, particularly for small lesions.78
Figure 5.13 illustrates colorectal liver metastases on
gadoxetic acid-enhanced MRI. Transabdominal Ultrasound
Diffusion-weighted imaging (DWI) is an MRI
technique that identifies areas within tissues with The primary use of ultrasound in the systemic stag-
restricted diffusion. As many tumours contain tight- ing of rectal cancer has been to evaluate the liver
ly packed cells with a poorly organized extracellular for the presence of metastases. Liver metastases may
matrix, they frequently have restricted diffusion of lack contrast with the surrounding normal tissue,
water molecules compared with normal tissues. This however, and therefore may be difficult to identify

Figure 5.14. Diffusion-weighted imaging (DWI) and corresponding gadolinium-enhanced magnetic resonance imaging.
Metastasis (white arrowheads) demonstrates restricted diffusion of DWI.

HEBK001-C05_p71-86.indd 81 08/02/13 5:59 PM


82  Radiological staging for systemic disease

on ultrasound. A meta-analysis found that the sen- CT. In a study of 1049 patients who underwent chest
sitivity of ultrasound on a per-patient basis for X-ray for the purposes of preoperative colorectal
identifying liver metastases was 55 per cent, com- cancer staging, only 0.86 per cent of patients had
pared with 72 per cent for CT, 75 per cent for MRI lung metastases correctly identified.87 Chest radi-
and 90 per cent for 18F-FDG-PET.81 ography should therefore be reserved for situations
Micro-bubble contrast agents have been used to where thoracic CT is not available.
improve the ability of ultrasound to identify liver
metastases and detect around 17 per cent more lesions
than conventional B-mode non-contrast-enhanced Assessing response
ultrasound.82 Even when using micro-bubble contrast to chemotherapy
agents, however, ultrasound is still less sensitive than
CT in detecting liver metastases.83 It also does not offer Aside from identifying metastatic disease, imaging
the opportunity to identify metastases at other sites. also has an important role to play in the assessment
Ultrasound is cheap and widely available. There- of response to treatment and in restaging disease
fore, ultrasound may still have a role as a screening following chemotherapy.
or monitoring technique in some institutions and The accuracy of 18F-FDG-PET in assessing met-
occasionally helps to clarify the nature of a hepatic astatic disease appears to be reduced in the period
abnormality. Computed tomography, PET and immediately following chemotherapy. In a study of
MRI are superior imaging modalities where these 138 patients investigating the accuracy of PET in
facilities are available, however. identifying colorectal liver metastases, Glazer and
colleagues found that PET had a negative predic-
tive value of only 13.3 per cent when performed
Intraoperative Ultrasound within 4 weeks of chemotherapy.88 This is probably
due to the metabolic inhibition of chemotherapeu-
Intraoperative ultrasound is used in many institutions tic agents preventing metastatic lesions from taking
for the intraoperative assessment of patients under- up FDG, despite harbouring viable tumour cells.
going resection of hepatic metastases. In this setting, Other studies have supported this finding, report-
ultrasound is helpful; it alters surgery in 20–44 per cent ing a per-lesion sensitivity of PET of only 47–62
of patients and demonstrates anatomy relative to the per cent,89,90 and a per-patient sensitivity of 63 per
metastases to help plan resection.84,85 It is ­particularly cent,91 after chemotherapy.
useful in the intraoperative setting to detect small The accuracy of anatomical imaging per-
lesions for resection or radiofrequency ablation that formed following chemotherapy appears to be
may otherwise have been missed.86 comparable to the accuracy in the absence of
chemotherapy. Superparamagnetic iron oxide-
enhanced MRI has a per-lesion sensitivity of 92
Laparoscopy per cent following chemotherapy,89 and non-con-
trast MRI has a per-lesion sensitivity of 80 per
In certain circumstances, diagnostic laparoscopy cent.92 Contrast-enhanced CT has a per-lesion
with cytology or histology may be useful to assess sensitivity of 65–76 per cent.90–92
the presence of metastatic disease if non-invasive The response of colorectal cancer metastases to
investigations are equivocal. The steady improve- treatment is therefore best assessed using anatomi-
ment in non-invasive cross-sectional and func- cal imaging on either serial CT or MRI scans. An
tional imaging will probably continue to reduce the objective assessment of response can be obtained
situations where laparoscopy is necessary, however. by repeated measurements of identified lesions;
the Response Evaluation Criteria In Solid Tumors
(RECIST) guidelines provide a structure for con-
Chest X-ray sistent assessment.93
Restaging of hepatic metastases following neoadju-
Chest radiography is less sensitive and specific in vant chemotherapy before resection is best performed
detecting colorectal liver metastases than thoracic using MRI with a liver-specific contrast agent.

HEBK001-C05_p71-86.indd 82 08/02/13 5:59 PM


References  83

Future developments provides the most accurate per-lesion preoperative


assessment of liver disease.
Incremental improvements in both MRI and CT 18F-Fluorodeoxyglucose positron-emission tom-
technology have steadily increased the speed and ography (18F-FDG-PET), or 18F-FDG-PET/CT
spatial resolution of these imaging modalities, and where available, should be used as a second-line
it is likely that similar advances will see the spa- investigation for patients with a high index of clinical
tial and perhaps contrast resolution of anatomical suspicion for metastatic disease (such as rising CEA)
imaging continue to increase. and normal first-line investigations. It may also be of
Functional imaging, and in particular fused benefit in assessing pelvic recurrence and in excluding
functional and anatomical imaging, has undergone extra-pulmonary and extra-hepatic disease in patients
rapid development. Alternative radiotracers such as being considered for metastasectomy.
39-deoxy-39-[18F]fluorothymidine (FLT) are being Further studies are required to define the role of
investigated, and it is possible that more specific modalities other than CT in the primary systemic
tracers will improve the specificity of PET in the staging of rectal cancer. It is possible that more
future. It is also likely that the spatial resolution of intensive primary staging may be of benefit in rec-
PET will continue to increase, and this may further tal cancer, particularly in patients who are defined
increase the sensitivity of PET in the future. as high risk on MRI staging of the primary tumour.
Technological hurdles have been overcome to In the future, functional imaging may help in selec-
allow PET and MRI scanning to be incorporated tion of patients most likely to benefit from neoadju-
in the same machine as PET/MRI fusion. Ongoing vant therapy before surgery and in determining the
research is under way to determine whether this likelihood and presence of a complete pathological
offers an advantage over current PET/CT. response and thus selecting patients in whom sur-
gery can be deferred and occasionally avoided.

CONCLUSION References
The ideal modality for systemic staging of rectal   1. Fong Y, Cohen AM, Fortner JG, et al. Liver resection
cancer would be cheap, be widely available, have for colorectal metastases. J Clin Oncol 1997; 15:
a high sensitivity for identifying rectal metastases 938–46.
in all sites, and have a high specificity. Unfortu-   2. Jaeck D, Bachellier P, Guiguet M, et al. Long-term
nately, no modality currently fulfils all these crite- survival following resection of colorectal hepatic
metastases. Br J Surg 1997; 84: 977–80.
ria. It is therefore necessary to use a multimodality
  3. Choti MA, Sitzmann JV, Tiburi MF, et al. Trends
approach and to adapt the staging strategy to the in long-term survival following liver resection for
patient’s situation to try to maximize the benefit hepatic colorectal metastases. Ann Surg 2002; 235:
from available imaging resources. 759–66.
Computed tomography of the chest, abdomen   4. Geoghegan JG, Scheele J. Treatment of colorectal
and pelvis is a good baseline investigation for the liver metastases. Br J Surg 1999; 86: 158–69.
primary systemic staging of rectal cancer. It identi-   5. De Jong MC, Pulitano C, Ribero D, et al. Rates and
patterns of recurrence following curative intent surgery
fies metastatic disease with good sensitivity in the
for colorectal liver metastasis: an international multi-
most commonly affected sites and should be per- institutional analysis of 1669 patients. Ann Surg 2009;
formed in all patients where this facility is available. 250: 440–8.
Review of serial CT images increases the sensitiv-   6. Wei AC, Greig PD, Grant D, Taylor B, Langer B,
ity and specificity of subsequent CT for identifying Gallinger S. Survival after hepatic resection for colorec-
recurrent and metastatic disease in the follow-up tal metastases: a 10-year experience. Ann Surg Oncol
period. 2006; 13: 668–76.
  7. Elias D, Blot F, el Otmany A, et al. Curative treatment
Magnetic resonance imaging should be used
of peritoneal carcinomatosis arising from colorectal
to clarify the nature of indeterminate liver lesions cancer by complete resection and intraperitoneal
identified on baseline or follow-up CT. Liver MRI chemotherapy. Cancer 2001; 92: 71–6.
should also be performed in patients being consid-   8. Bleeker WA, Mulder NH, Hermans J, Otter R, Plukker
ered for resection of liver metastases, as this probably JT. Value and cost of follow–up after adjuvant treatment

HEBK001-C05_p71-86.indd 83 08/02/13 5:59 PM


84  Radiological staging for systemic disease

of patients with Dukes’ C colonic cancer. Br J Surg possible role of chemokines. Clin Colorectal Cancer
2001; 88: 101–6. 2009; 8: 100–5.
  9. Renehan AG, Egger M, Saunders MP, O’Dwyer ST. 26. Sundermeyer ML, Meropol NJ, Rogatko A, Wang
Impact on survival of intensive follow up after curative H, Cohen SJ. Changing patterns of bone and brain
resection for colorectal cancer: systematic review and metastases in patients with colorectal cancer. Clin
meta-analysis of randomised trials. BMJ 2002; 324: 813. Colorectal Cancer 2005; 5: 108–13.
10. Shaw IM, Rees M, Welsh FK, Bygrave S, John TG. 27. Demeo JH, Fulcher AS, Austin RF Jr. Anatomic CT
Repeat hepatic resection for recurrent colorectal liver demonstration of the peritoneal spaces, ligaments,
metastases is associated with favourable long-term and mesenteries: normal and pathologic processes.
survival. Br J Surg 2006; 93: 457–64. Radiographics 1995; 15: 755–70.
11. Ogata Y, Matono K, Hayashi A, et al. Repeat pulmonary 28. Coakley FV, Hricak H. Imaging of peritoneal and
resection for isolated recurrent lung metastases yields mesenteric disease: key concepts for the clinical radi-
results comparable to those after first pulmonary resec- ologist. Clin Radiol 1999; 54: 563–74.
tion in colorectal cancer. World J Surg 2005; 29: 363–8. 29. Meyers MA, Oliphant M, Berne AS, Feldberg MA. The
12. Simmonds PC. Palliative chemotherapy for advanced peritoneal ligaments and mesenteries: pathways of
colorectal cancer: systematic review and meta-analy- intraabdominal spread of disease. Radiology 1987;
sis. BMJ 2000; 321: 531–5. 163: 593–604.
13. Cummins ER, Vick KD, Poole GV. Incurable colorectal 30. Andres A, Majno PE, Morel P, et al. Improved long-
carcinoma: the role of surgical palliation. Am Surg term outcome of surgery for advanced colorectal liver
2004; 70: 433–7. metastases: reasons and implications for management
14. Dukes CE, Bussey HJ. The spread of rectal cancer and on the basis of a severity score. Ann Surg Oncol 2008;
its effect on prognosis. Br J Cancer 1958; 12: 309–20. 15: 134–43.
15. Grinnell RS. Lymphatic metastases of carcinoma of 31. Foroutani A, Garland AM, Berber E, et al. Laparoscop-
the colon and rectum. Ann Surg 1950; 131: 494–506. ic ultrasound vs triphasic computed tomography for
16. Grinnell RS. Lymphatic block with atypical and retro- detecting liver tumors. Arch Surg 2000; 135: 933–8.
grade lymphatic metastasis and spread in carcinoma of 32. Cervone A, Sardi A, Conaway GL. Intraoperative ultra-
the colon and rectum. Ann Surg 1966; 163: 272–80. sound (IOUS) is essential in the management of meta-
17. Dukes C, Bussey HJ. Venous spread in rectal cancer. static colorectal liver lesions. Am Surg 2000; 66: 611–5.
Proc R Soc Med 1941; 34: 571–3. 33. Nordlinger B, Wind P. Repeat resections of primary
18. Sunderland D. The significance of vein invasion by hepatic malignancies. Cancer Treat Res 1994; 69: 53–6.
cancer of the rectum and sigmoid: a microscopic 34. Clavien PA, Selzner N, Morse M, Selzner M, Paulson
study of 210 cases. Cancer 1949; 2: 429–37. E. Downstaging of hepatocellular carcinoma and liver
19. Talbot IC, Ritchie S, Leighton MH, Hughes AO, Bussey metastases from colorectal cancer by selective intra-
HJ, Morson BC. The clinical significance of invasion arterial chemotherapy. Surgery 2002; 131: 433–42.
of veins in cancer of the rectum. Br J Surg 1980; 67: 35. Shankar A, Leonard P, Renaut AJ, et al. Neo-
439–42. adjuvant therapy improves resectability rates for
20. Talbot IC, Ritchie S, Leighton MH, Hughes AO, Bussey colorectal liver metastases. Ann R Coll Surg Engl
HJ, Morson BC. Spread of rectal cancer within veins: 2001; 83: 85–8.
histologic features and clinical significance. Am J Surg 36. Adam R, Avisar E, Ariche A, et al. Five-year survival
1981; 141: 15–7. following hepatic resection after neoadjuvant therapy
21. Leporrier J, Maurel J, Chiche L, Bara S, Segol P, for nonresectable colorectal. Ann Surg Oncol 2001; 8:
Launoy G. A population-based study of the incidence, 347–53.
management and prognosis of hepatic metastases 37. Giacchetti S, Itzhaki M, Gruia G, et al. Long-term
from colorectal cancer. Br J Surg 2006; 93: 465–74. survival of patients with unresectable colorectal cancer
22. Mitry E, Guiu B, Cosconea S, et al. Epidemiology, liver metastases following infusional chemotherapy
management and prognosis of colorectal cancer with with 5-fluorouracil, leucovorin, oxaliplatin and surgery.
lung metastases: a 30-year population-based study. Ann Oncol 1999; 10: 663–9.
Gut 2010; 59: 1383–8. 38. Gillams AR, Lees WR. Radio-frequency ablation of
23. Birnkrant A, Sampson J, Sugarbaker PH. Ovarian colorectal liver metastases in 167 patients. Eur Radiol
metastasis from colorectal cancer. Dis Colon Rectum 2004; 14: 2261–7.
1986; 29: 767–71. 39. Hildebrand P, Kleemann M, Roblick UJ, et al. Radiofre-
24. Segelman J, Floter-Radestad A, Hellborg H, Sjovall quency-ablation of unresectable primary and secondary
A, Martling A. Epidemiology and prognosis of ovarian liver tumors: results in 88 patients. Langenbecks Arch
metastases in colorectal cancer. Br J Surg 2010; 97: Surg 2006; 391: 118–23.
1704–9. 40. Machi J, Oishi AJ, Sumida K, et al. Long-term out-
25. Mongan JP, Fadul CE, Cole BF, et al. Brain metastases come of radiofrequency ablation for unresectable liver
from colorectal cancer: risk factors, incidence, and the metastases from colorectal cancer: evaluation of

HEBK001-C05_p71-86.indd 84 08/02/13 5:59 PM


References  85

prognostic factors and effectiveness in first- and sec- 55. Valls C, Andia E, Sanchez A, et al. Hepatic metastases
ond-line management. Cancer J 2006; 12: 318–26. from colorectal cancer: preoperative detection and
41. Stang A, Fischbach R, Teichmann W, Bokemeyer C, assessment of resectability with helical CT. Radiology
Braumann D. A systematic review on the clinical benefit 2001; 218: 55–60.
and role of radiofrequency ablation as treatment of color- 56. Wong K, Paulson EK, Nelson RC. Breath-hold three-
ectal liver metastases. Eur J Cancer 2009; 45: 1748–56. dimensional CT of the liver with multi-detector row
42. Barbaro B, Caputo F, Tebala C, et al. Preoperative helical CT. Radiology 2001; 219: 75–9.
right portal vein embolisation: indications and results. 57. Scott DJ, Guthrie JA, Arnold P, et al. Dual phase heli-
Radiol Med 2009; 114: 553–70. cal CT versus portal venous phase CT for the detec-
43. Fong Y, Fortner J, Sun RL, Brennan MF, Blumgart LH. tion of colorectal liver metastases: correlation with
Clinical score for predicting recurrence after hepatic intra-operative sonography, surgical and pathological
resection for metastatic colorectal cancer: analysis findings. Clin Radiol 2001; 56: 235–42.
of 1001 consecutive cases. Ann Surg 1999; 230: 58. Ch’en IY, Katz DS, Jeffrey RB Jr, et al. Do arterial
309–18, 318–21. phase helical CT images improve detection or char-
44. Strasberg SM, Dehdashti F, Siegel BA, Drebin JA, Line- acterization of colorectal liver metastases? J Comput
han D. Survival of patients evaluated by FDG-PET before Assist Tomogr 1997; 21: 391–7.
hepatic resection for metastatic colorectal carcinoma: a 59. Jang HJ, Lim HK, Lee WJ, Lee SJ, Yun JY, Choi D.
prospective database study. Ann Surg 2001; 233: 293–9. Small hypoattenuating lesions in the liver on single-
45. Rama N, Monteiro A, Bernardo JE, Eugenio L, phase helical CT in preoperative patients with gastric
Antunes MJ. Lung metastases from colorectal cancer: and colorectal cancer: prevalence, significance, and
surgical resection and prognostic factors. Eur J Cardi- differentiating features. J Comput Assist Tomogr 2002;
othorac Surg 2009; 35: 444–9. 26: 718–24.
46. Borasio P, Gisabella M, Bille A, et al. Role of surgical 60. Glover C, Douse P, Kane P, et al. Accuracy of investi-
resection in colorectal lung metastases: analysis of gations for asymptomatic colorectal liver metastases.
137 patients. Int J Colorectal Dis 2011; 26: 183–90. Dis Colon Rectum 2002; 45: 476–84.
47. Reinhardt MJ, Wiethoelter N, Matthies A, et al. PET 61. Niekel MC, Bipat S, Stoker J. Diagnostic imaging
recognition of pulmonary metastases on PET/CT of colorectal liver metastases with CT, MR imaging,
imaging: impact of attenuation-corrected and non- FDG PET, and/or FDG PET/CT: a meta-analysis of
attenuation-corrected PET images. Eur J Nucl Med prospective studies including patients who have not
Mol Imaging 2006; 33: 134–9. previously undergone treatment. Radiology 2010;
48. Cho KC, Gold BM. Computed tomography of Kruken- 257: 674–84.
berg tumors. AJR Am J Roentgenol 1985; 145: 285–8. 62. Bipat S, van Leeuwen MS, Comans EF, et al. Colorec-
49. Kuhlman JE, Hruban RH, Fishman EK. Krukenberg tal liver metastases: CT, MR imaging, and PET for diag-
tumors: CT features and growth characteristics. South nosis: meta-analysis. Radiology 2005; 237: 123–31.
Med J 1989; 82: 1215–9. 63. Wiering B, Krabbe PF, Dekker HM, Oyen WJ, Ruers
50. Kim SH, Kim WH, Park KJ, Lee JK, Kim JS. CT and TJ. The role of FDG–PET in the selection of patients
MR findings of Krukenberg tumors: comparison with with colorectal liver metastases. Ann Surg Oncol 2007;
primary ovarian tumors. J Comput Assist Tomogr 14: 771–9.
1996; 20: 393–8. 64. Rosa F, Meimarakis G, Stahl A, et al. Colorectal cancer
51. Glehen O, Kwiatkowski F, Sugarbaker PH, et al. Cytore- patients before resection of hepatic metastases:
ductive surgery combined with perioperative intraperi- impact of (18)F-FDG PET on detecting extrahepatic
toneal chemotherapy for the management of peritoneal disease. Nuklearmedizin 2004; 43: 135–40.
carcinomatosis from colorectal cancer: a multi- 65. Truant S, Huglo D, Hebbar M, Ernst O, Steinling M,
institutional study. J Clin Oncol 2004; 22: 3284–92. Pruvot FR. Prospective evaluation of the impact of
52. Sugarbaker PH, Schellinx ME, Chang D, Koslowe [18F]fluoro-2-deoxy-d-glucose positron emission tom-
P, von Meyerfeldt M. Peritoneal carcinomatosis from ography of resectable colorectal liver metastases. Br J
adenocarcinoma of the colon. World J Surg 1996; 20: Surg 2005; 92: 362–9.
585–91, 592. 66. Zhang C, Chen Y, Xue H, et al. Diagnostic value of
53. Ridge JA, Bading JR, Gelbard AS, Benua RS, Daly FDG-PET in recurrent colorectal carcinoma: a meta-
JM. Perfusion of colorectal hepatic metastases. Rela- analysis. Int J Cancer 2009; 124: 167–73.
tive distribution of flow from the hepatic artery and 67. Flamen P, Hoekstra OS, Homans F, et al. 2001.
portal vein. Cancer 1987; 59: 1547–53. Unexplained rising carcinoembryonic antigen (CEA) in
54. Schwartz L, Brody L, Brown K, et al. Prospective, the postoperative surveillance of colorectal cancer: the
blinded comparison of helical CT and CT arterial por- utility of positron emission tomography (PET). Eur J
tography in the assessment of hepatic metastasis from Cancer 2001; 37: 862–9.
colorectal carcinoma. World J Surg 2006; 30: 1892–9, 68. Heriot AG, Hicks RJ, Drummond EG, et al. Does
1900–1. positron emission tomography change management in

HEBK001-C05_p71-86.indd 85 08/02/13 5:59 PM


86  Radiological staging for systemic disease

primary rectal cancer? A prospective assessment. Dis gastrointestinal tract by using noninvasive imaging
Colon Rectum 2004; 47: 451–8. methods (US, CT, MR imaging, PET): a meta-analysis.
69. Schussler-Fiorenza CM, Mahvi DM, Niederhuber J, Radiology 2002; 224: 748–56.
Rikkers LF, Weber SM. Clinical risk score correlates 82. Yarmenitis SD, Karantanas A, Bakantaki A, Papan-
with yield of PET scan in patients with colorectal toniou Y, Gourtsoyiannis N. Detection of colorectal
hepatic metastases. J Gastrointest Surg 2004; 8: cancer hepatic metastases with contrast-enhanced
150–57, 157–8. ultrasound: comparison with conventional B-mode
70. Wahl RL. Why nearly all PET of abdominal and pelvic ultrasound. Dig Dis 2007; 25: 86–93.
cancers will be performed as PET/CT. J Nucl Med 83. Larsen LP, Rosenkilde M, Christensen H, et al. Can
2004; 45(suppl. 1): 82–95S. contrast-enhanced ultrasonography replace multi-
71. Orlacchio A, Schillaci O, Fusco N, et al. Role of PET/ detector-computed tomography in the detection of
CT in the detection of liver metastases from colorectal liver metastases from colorectal cancer? Eur J Radiol
cancer. Radiol Med 2009; 114: 571–85. 2009; 69: 308–13.
72. Davey K, Heriot AG, Mackay J, et al. The impact of 84. Torzilli G, del Fabbro D, Palmisano A, et al. Contrast-
18-fluorodeoxyglucose positron emission tomogra- enhanced intraoperative ultrasonography during
phy–computed tomography on the staging and man- hepatectomies for colorectal cancer liver metastases.
agement of primary rectal cancer. Dis Colon Rectum J Gastrointest Surg 2005; 9: 1148–53, 1153–4.
2008; 51: 997–1003. 85. Mazzoni G, Napoli A, Mandetta S, et al. Intra-operative
73. Taylor FG, Swift RI, Blomqvist L, Brown G. A system- ultrasound for detection of liver metastases from color-
atic approach to the interpretation of preoperative ectal cancer. Liver Int 2008; 28: 88–94.
staging MRI for rectal cancer. AJR Am J Roentgenol 86. Siosteen AK, Elvin A. Intra-operative uses of contrast-
2008; 191: 1827–35. enhanced ultrasound. Eur Radiol 2004; 14(suppl. 8):
74. Cotton F, Pellet O, Gilly FN, Granier A, Sournac L, P87–95.
Glehen O. MRI evaluation of bulky tumor masses in 87. Gielen C, Sanli I, Stroeken L, Botterweck A, Hulsewe
the mesentery and bladder involvement in peritoneal K, Hoofwijk A. Staging chest radiography is not useful
carcinomatosis. Eur J Surg Oncol 2006; 32: 1212–6. in patients with colorectal cancer. Eur J Surg Oncol
75. Kong G, Jackson C, Koh DM, et al. The use of 2009; 35: 1174–8.
18F-FDG PET/CT in colorectal liver metastases: com- 88. Glazer ES, Beaty K, Abdalla EK, Vauthey JN, Curley
parison with CT and liver MRI. Eur J Nucl Med Mol SA. Effectiveness of positron emission tomography for
Imaging 2008; 35: 1323–9. predicting chemotherapy response in colorectal cancer
76. Semelka RC, Martin DR, Balci C, Lance T. Focal liver liver metastases. Arch Surg 2010; 145: 340–5.
lesions: comparison of dual-phase CT and multise- 89. Bacigalupo L, Aufort S, Eberle MC, Assenat E, Ychou
quence multiplanar MR imaging including dynamic M, Gallix B. Assessment of liver metastases from
gadolinium enhancement. J Magn Reson Imaging colorectal adenocarcinoma following chemotherapy:
2001; 13: 397–401. SPIO-MRI versus FDG-PET/CT. Radiol Med 2010;
77. Reimer P, Jahnke N, Fiebich M, et al. Hepatic lesion 115: 1087–100.
detection and characterization: value of nonenhanced 90. Carnaghi C, Tronconi MC, Rimassa L, et al. Utility of
MR imaging, superparamagnetic iron oxide-enhanced 18F-FDG PET and contrast-enhanced CT scan in the
MR imaging, and spiral CT-ROC analysis. Radiology assessment of residual liver metastasis from colorectal
2000; 217: 152–8. cancer following adjuvant chemotherapy. Nucl Med
78. Zech CJ, Herrmann KA, Reiser MF, Schoenberg SO. Rev Cent East Eur 2007; 10: 12–5.
MR imaging in patients with suspected liver metas- 91. Spatz J, Holl G, Sciuk J, Anthuber M, Arnholdt HM,
tases: value of liver-specific contrast agent Gd-EOB- Markl B. Neoadjuvant chemotherapy affects staging
DTPA. Magn Reson Med Sci 2007; 6: 43–52. of colorectal liver metastasis: a comparison of PET,
79. Kenis C, Deckers F, de Foer B, van Mieghem F, van CT and intraoperative ultrasound. Int J Colorectal Dis
Laere S, Pouillon M. Diagnosis of liver metastases: can 2011; 26: 165–71.
diffusion-weighted imaging (DWI) be used as a stand 92. Van Kessel CS, van Leeuwen MS, van den Bosch
alone sequence? Eur J Radiol 2011 [Epub ahead of MA, et al. Accuracy of multislice liver CT and MRI for
print]. preoperative assessment of colorectal liver metastases
80. Koh DM, Brown G, Riddell AM, et al. Detection of after neoadjuvant chemotherapy. Dig Surg 2011; 28:
colorectal hepatic metastases using MnDPDP MR 36–43.
imaging and diffusion-weighted imaging (DWI) alone 93. Eisenhauer EA, Therasse P, Bogaerts J, et al. New
and in combination. Eur Radiol 2008; 18: 903–10. response evaluation criteria in solid tumours: revised
81. Kinkel K, Lu Y, Both M, Warren RS, Thoeni RF. RECIST guideline (version 1.1). Eur J Cancer 2009;
Detection of hepatic metastases from cancers of the 45: 228–47.

HEBK001-C05_p71-86.indd 86 08/02/13 5:59 PM


6
Preoperative radiotherapy and
chemoradiotherapy for rectal cancer
Rob Glynne-Jones and Mark Harrison

Introduction ultrasound and magnetic resonance imaging


(MRI) to stage patients, the success of postoperative
Colorectal cancer is one of the most common solid chemoradiation was extrapolated to the preopera-
tumours, of which approximately 40 per cent of tive setting. The concept of spatial cooperation in
patients have rectal cancer. Historically, surgery preoperative chemoradiation for locally advanced
alone for rectal cancer has been associated with a rectal cancer is attractive.7 Chemotherapy may, as
high incidence of local recurrence. Additionally, a component of chemoradiation, act as a radio-
some 10–40 per cent of patients with rectal cancer sensitizing agent; it may also potentially eradicate
require extirpative procedures leading to a perma- distant micro-metastases. In general, concurrent
nent stoma. A landmark retrospective study high- chemotherapy has proved more effective than the
lighted the poor prognosis for patients with rec- sequential use of these modalities.8
tal cancer, with a 5-year survival of 6 per cent in A series of randomized controlled trials have
patients with pathologically Dukes C cancer.1 clearly shown the superiority of preoperative radia-
In the light of these results, using post-surgical tion or chemoradiation therapy compared with
histology to define risk, several postoperative stud- postoperative treatment.9–13 Current recommenda-
ies investigated chemotherapy and radiotherapy2 tions suggest the use of preoperative chemoradiation
or a combination of both to prevent local recur- followed by total mesorectal excision (TME) surgery
rence and reduce metastases. Overall, these studies as the standard treatment of choice for the majority
showed a significant benefit for the combination of patients diagnosed with advanced rectal cancer.
of chemoradiation.3,4 Interestingly, a retrospective In the past two decades, greater understanding
analysis of pooled data from US trials showed simi- of the natural history of the disease and patterns
lar 5-year overall survival for patients with pT3N0 of recurrence, and more precise histopathological
rectal cancer treated with either surgery or chemo- reporting, have helped to define patients with a high
therapy alone (84 per cent) compared with those risk of local recurrence and metastatic disease fol-
treated with postoperative chemoradiotherapy lowing ‘curative resection’.14 This particular focus
(74–80 per cent).5 Similar data have been reported on the circumferential resection margin (CRM) has
from the National Cancer Data Base.6 driven technical advances in surgical technique with
With the introduction of better preoperative meticulous surgical dissection along ­embryological
imaging such as computed tomography (CT), planes and highlighted the ­importance of

HEBK001-C06_p87-102.indd 87 08/02/13 5:58 PM


88  Preoperative radiotherapy and chemoradiotherapy for rectal cancer

­ reoperative assessment of the tumour with MRI.


p More recently, a number of molecularly targeted
Even technically optimized surgery is unlikely to agents have been integrated into standard palliative
achieve a curative resection when the preoperative chemotherapy regimens for advanced metastatic
MRI shows a threatened or breached CRM. In addi- colorectal cancer. These biologically targeted agents
tion, at the time of diagnosis, some 20–25 per cent appear to improve response rates and extend pro-
of patients with rectal cancer are found to have overt gression-free and overall survival, albeit with varying
metastatic disease, and a further 30–40 per cent sub- success.28–31 Three monoclonal antibodies have now
sequently develop metastases. entered clinical practice in colorectal cancer – cetuxi-
The multimodality treatment of rectal cancer mab, panitumumab and bevacizumab. A strategy
attempts to integrate the three modalities of sur- to incorporate these newer biologically active tar-
gery, radiotherapy and chemotherapy as strategies geted agents into chemoradiation schedules has also
to eradicate cells at the margins or in discontinuous emerged.
areas of tumour within the pelvis, in lymph nodes In this chapter, we examine the rationale for pre-
or in distant metastatic sites to improve both local operative radiation and chemoradiation, the selec-
control and overall survival. tion of patients who are appropriate for long-course
Systematic reviews and several meta-analyses preoperative radiotherapy and short-course preop-
have examined the role of radiotherapy (as opposed erative radiation (SCPRT), the techniques available
to chemoradiation) in resectable rectal cancer.15–17 such as intensity-modulated radiotherapy (IMRT),
These meta-analyses concluded that the evidence optimal doses and field size, the integration of cyto-
from relevant trials favoured preoperative rather toxic chemotherapy and biological agents, potential
than postoperative radiotherapy, and that a bio- biological markers, and future potential strategies
logically equivalent dose greater than 30 Gy is more of treatment.
effective in reducing local relapse. A 2007 Cochrane
review further supports these findings but failed to
confirm a reduction in mortality or any advantage Defining the most
in terms of sphincter sparing from preoperative appropriate treatment
radiotherapy.18 The Cochrane review also high- strategy
lighted the significant risks of increased pelvic and
perineal infections when radiation and surgery are The terms ‘favourable’, ‘early’ and ‘good’, ‘intermedi-
combined, and a detriment to the patient’s anorec- ate’ and ‘bad’, and ‘locally advanced’ and ‘ugly’ have
tal, urinary and sexual function. historically been used for categorizing rectal cancer
Since the early 1980s, the fluoropyrimidine into clinical subgroups. The term ‘locally advanced’
5-fluorouracil (5FU) alone, and more recently in has been commonly used in a very broad and unde-
combinations of cytotoxic chemotherapy using fined way and generally directed the patient to
oxaliplatin or irinotecan, has represented the main- receive preoperative treatment. In the 1980s, preop-
stay of chemotherapy treatment for patients with erative assessment and clinical staging of rectal can-
advanced and metastatic colorectal cancer. These cer was limited to digital rectal examination, pos-
combinations have been integrated into chemora- sibly a rigid sigmoidoscopy and a barium enema. A
diation regimens and tested in a number of prospec- clinical judgement based on these findings assessed
tive randomized trials in rectal cancer, potentially to tumour fixity, but there was no widely accepted and
mirror the success of 5FU and oxaliplatin in deal- validated imaging method of defining either locally
ing with distant micro-metastases in the adjuvant advanced rectal cancer or unresectable disease.
setting in colon cancer.19–21 Investigators have used Currently, we broadly consider patients to have
these cytotoxic agents either as ­radiosensitizers22,23 or easily resectable cancers (which can be cured by
in an attempt to use systemically active chemother- radical surgery alone), borderline resectable dis-
apy preoperatively with an appropriate duration.24–26 ease (i.e. a potentially ‘threatened’ or minimally
Perhaps surprisingly when used as a radiosensitizer, breached circumferential margin as predicted by
these combinations of a cytotoxic agent and radia- MRI), or unresectable cancers with disease outside
tion have had only moderate success in improving the mesorectum (for whom surgery is not possible
outcome in rectal cancer.12,13,22,23,27 without leaving tumour within the pelvis).

HEBK001-C06_p87-102.indd 88 08/02/13 5:58 PM


PREOPERATIVE RADIOTHERAPY  89

Our increased level of knowledge is based on the compromised by surgery and infection; theo-
use of preoperative MRI, which allows division of retically, better oxygenation will improve the
rectal cancers into categories with different risks radiosensitivity of the tumour cells.
and different indications for preoperative radio- l An R0 resection is much more likely if the
therapy. Preoperative treatment may be tailored tumour undergoes a degree of shrinkage.
to the radiological stage of the tumour to facilitate l Killing or sterilizing all the tumour cells with
a reduction of risk of local recurrence, improved radiation before surgery may reduce the risk
likelihood of curative resection, or identification of of tumour cells seeding in the wound during
unresectable disease and prevention of an unneces- surgery, particularly in the posterior pelvis
sary surgical procedure. Use of MRI allows rectal when the patient lies supine, thereby decreasing
cancer to be divided into the following groups: the local recurrence rate.
l With preoperative treatment, the tumour, drain-
1. very early (some cT1 sm1/2)
ing lymph nodes and adjacent mesorectum can
2. low risk (.cT1 sm2 to cT2, some cT3a (to T3c),
be targeted in the gross tumour volume and
CRM 2 /N0)
clinical target volume, rather than an uninvolved
3. locally advanced (cT3, some T4 crm 2 /N1)
length of bowel and mesocolon, which may
4. potentially unresectable advanced (cT3 CRM1
already have a compromised blood supply.
or cT4)
l The clinical target volume will not be required
5. unresectable disease (extension to lateral
to cover the perineum, and there will be no
pelvic side-wall/lateral pelvic lymph node
radiation to the anastomotic site.
involvement/disease outside the pelvis).
l The small bowel is much less likely to be includ-
Other well-recognized factors such as tumour ed in the radiation field, unless the patient has
height, proximity to the CRM, cN stage, lymphovas- had a hysterectomy or previous pelvic surgery.
cular invasion, extramural vascular invasion and l Although still unvalidated, the preoperative
perineural invasion are also relevant to the risk of approach may define good and bad prognos-
local recurrence and the development of metastat- tic groups of patients by their response to
ic disease. Where possible, these factors should be ­treatment.32–36
mentioned in the MRI report with the proviso that
they are categorized with the prefix ‘mr’, e.g. mrN1 For all these reasons, the expectation is that the
or mrEMVI. The relevance of the historical clinical acute and late toxicity of preoperative radiation will
classification into fixed, tethered or mobile tumours be less, and hence compliance will be better and
is unclear if MRI is performed for decision-making, more patients will receive the full dose of radiation.
but it may be complementary, especially in low rectal Three adjuvant radiation approaches have been
tumours. In all patients, a history, clinical examina- in common use: SCPRT, long-course preoperative
tion and rectal examination by the responsible sur- radiation and long-course preoperative chemoradi-
geon is mandatory, and the surgical opinion on cur- ation. Long-course radiation alone has been shown
rent and likely future anorectal function should be to be less effective in terms of local control when
transmitted to the radiation oncologist, ideally at the compared with preoperative 5FU-based chemora-
multidisciplinary team colorectal conference. diation27,37 and hence is used only in patients who
are unfit for chemotherapy; it will be discussed only
briefly here.
There are different rationales for SCPRT and
PREOPERATIVE RADIOTHERAPY chemoradiotherapy, and some differences in
tumour target volume definition that will be dis-
There are a number of theoretical advantages of
cussed later. Short-course preoperative radio-
preoperative radiation therapy compared with
therapy uses a short intensive course of radiation
postoperative radiation therapy. To an extent, these
delivered with a dose of 25 Gy over 5 days. The sin-
advantages have been confirmed in clinical trials:
gle aim of treatment is to reduce the risk of pelvic
l Preoperative radiation can be delivered to an recurrence. This approach has gained widespread
area where the blood supply has not yet been acceptance in Europe following the publication of

HEBK001-C06_p87-102.indd 89 08/02/13 5:58 PM


90  Preoperative radiotherapy and chemoradiotherapy for rectal cancer

the ­Swedish and Dutch rectal cancer studies.38,39 In of combination chemotherapy schedules have been
contrast, the rationale for preoperative chemoradi- completed or are ongoing.
otherapy is more attractive, as it combines early sys- Two randomized trials in unresectable disease
temic chemotherapy with a locoregional treatment. have confirmed an advantage for 5FU-based chem-
Current criteria in our own units for the delivery oradiation over radiation alone.40 One very small
of preoperative chemoradiation rely on MRI scan- phase III trial in unresectable rectal cancer com-
ning in determining the extent of tumour either at pared radiation alone with chemoradiotherapy in
(i.e. within 1 mm) or outside the mesorectal enve- Sweden, demonstrating improved resectability and
lope or below the envelope extending into the leva- local control with the use of chemoradiotherapy.41
tors and sphincter mechanism. Using these MRI
criteria, it is possible to predict patients in whom
there is a high risk that the surgeon will not be able Reducing local recurrence
to perform an R0 resection and will leave gross
macroscopic or undetected microscopic tumour The majority of randomized trials in rectal cancer
behind in the pelvis. have been performed in patients with resectable rec-
tal cancer, where the aim of treatment has been to
lower the risk of local recurrence. Some of these trials
Rendering unresectable were performed before 2000 prior to the acceptance
tumours resectable of the benefits of meticulous surgical dissection in
the form of mesorectal excision. The local recurrence
A minority of patients present with a rectal tumour rates in the surgery alone arm of these studies were in
that cannot be resected without leaving residual the range of 21–36.5 per cent.16
macroscopic or microscopic disease. The multimo- Three retrospective systematic reviews/meta-anal-
dality approach to rectal cancer management allows yses have been published on the role of radiotherapy
these patients to be identified before an (unsuc- in rectal cancer.15–17 The Colorectal Cancer Collabo-
cessful) attempt at surgical resection is made. This rative Group meta-analysis identified 22 randomized
approach facilitates the use of preoperative radio- controlled trials that have compared the use of both
therapy or chemoradiotherapy. preoperative radiotherapy (14 trials, 6350 patients)
Before the development of pelvic MRI, most and postoperative radiotherapy (8 trials, 2157
patients were identified by the presence of tumour patients) in patients intended to undergo a curative
fixity determined either in the clinic or during an resection for rectal cancer, versus surgery alone.16
examination under anaesthetic. Pragmatically this In the entire group of patients, overall survival at
method of assessment is limited to those tumours 5 years was not significantly different, with a 5-year
where the examining finger can assess mobility of overall survival of 45 per cent in the patients receiv-
the tumour. Pelvic MRI can assist in demonstrat- ing radiation versus 42.1 per cent in the patients
ing the relationship of the primary tumour to the treated with surgery alone. Despite the lack of effect
surrounding mesorectal fascia and the surrounding on overall survival, there was an expected reduction
organs, as well as enlarged lymph nodes with MRI in local recurrence. In the preoperative trials, the
appearances of tumour involvement. Thus, patients rate of local recurrence at 5 years was 12.5 per cent
may be selected for preoperative radiation in this in the patients treated with radiation versus 22.2
category if there is evidence of primary tumour per cent in the patients treated with surgery alone.
involving or extending beyond the mesorectal fas- Postoperative radiation also reduced the rate of
cia; primary tumour within 1–2 mm of the mes- local recurrence from 23.8 per cent in the patients
orectal fascia; involved lymph nodes outside the treated with surgery alone to 16.9 per cent in the
mesorectal fascia (usually iliac nodes); and primary patients receiving radiation.
tumour involving the levators. Since response and Trials performed in individual countries have
shrinkage of the tumour is required, these patients influenced their definition of the ‘standard of care.’ A
require preoperative chemoradiotherapy. Current German phase III trial reported improved outcome
approaches use a fluoropyrimidine combined with for preoperative chemoradiotherapy in this setting.
radiation, although many phase II studies of the use A total of 823 patients were randomized between

HEBK001-C06_p87-102.indd 90 08/02/13 5:58 PM


LONG-COURSE PREOPERATIVE 5-FLUOROURACIL-BASED CHEMORADIOTHERAPY  91

preoperative chemoradiotherapy and postopera- with 63.5 per cent in the surgery-only group. Local
tive chemoradiotherapy (patients received postop- recurrence at 5 years was 5.6 per cent and 10.9 per
erative adjuvant chemotherapy in both arms of the cent, respectively (P , 0.001). The low level of local
trial). Local recurrence and acute and late toxicity recurrence rate in this trial has led to the acceptance
were statistically significantly reduced with the pre- of TME as the standard surgical technique. Mature
operative approach, but there was no difference in outcome from the trial has confirmed a reduction
the distant metastases rate or overall survival.12 in local recurrence but no survival benefit.43
In contrast to the German approach, a short In the UK, some colorectal multidisciplinary
accelerated preoperative radiation schedule of teams have adopted the non-selective use of rou-
25 Gy in five fractions (SCPRT) with surgery per- tine SCPRT, while others use pelvic MRI to deter-
formed within a few days of completion of radia- mine patients whose primary tumour is predicted
tion was developed in Sweden to reduce the risk of to be clear of the CRM and in whom initial surgery
local recurrence. The Swedish Rectal Cancer Trial is performed. The Dutch and Medical Research
demonstrated a significant reduction in local recur- Council (MRC) CR07 trials have directly compared
rence and a 10 per cent absolute improvement in these two approaches. The Dutch trial examined
survival; this led to the widespread adoption of the routine use of SCPRT followed by TME, with
SCPRT in the Scandinavian and northern Euro- TME and selective postoperative radiotherapy in
pean countries.38 the event of histopathological evidence of involve-
The Swedish Rectal Cancer Trial is the only major ment of the circumferential resection margin.
modern trial to show a benefit in survival from pre- The Dutch trial appears to show an absolute
operative radiotherapy. This trial randomized 1168 reduction in local recurrence of 6 per cent. Thus,
patients with resectable rectal cancer to SCPRT fol- if 100 patients are irradiated, 6 local recurrences
lowed by surgery, versus surgery alone. The radia- are prevented, i.e. the number of patients needed
tion was delivered over 5 days with 5 Gy per frac- to treat to prevent 1 local recurrence is 16.7. In the
tion. The 5-year overall survival was significantly MRC CR07 trial, if a good mesorectal excision was
better in the radiation–surgery group (58 per cent) performed,44 the number needed to treat appears to
than in the surgery-only group (48 per cent) (P 5 be over 20. As stated above, the present authors do
0.004). The benefit of radiation persisted through a not therefore support the widespread advocacy for
median of 13 years’ follow-up (38 per cent alive v. routine adjuvant radiotherapy as used in the treat-
30 per cent, P 5 0.008). Local recurrence as either ment arms of recent trials.
a first or later event was less common in the group There is increasing evidence that patients with
of patients treated with radiation (9 per cent v. 26 very low tumours that require abdominoperineal
per cent, P , 0.001).42 The survival advantage seen excision are at higher risk of involvement of the
in this trial has been suggested to derive from the CRM, local recurrence and inferior survival. Most
large difference in local recurrence rates between agree that routine preoperative radiotherapy is
the two groups, which may have been a reflection of indicated for this group of patients. It is a source of
the suboptimal surgery before the TME era. considerable debate whether SCPRT may be used
The Dutch Colorectal Cancer Group conducted in some patients, whether chemoradiotherapy is
a large, prospective, randomized trial using an iden- preferred in other patients, and whether a wider
tical radiation schedule to that used in the Swed- cylindrical surgical technique should be adopted to
ish Rectal Cancer Trial. In the Dutch trial a series reduce the rate of an involved CRM.
of surgical training programmes and mentoring
attempted to standardize the surgical procedure,
and all patients underwent surgery by TME prin- LONG-COURSE PREOPERATIVE
ciples. Of 1805 eligible patients, 897 were rand- 5-FLUOROURACIL-BASED
omized to radiation and then immediate surgery CHEMORADIOTHERAPY –
(within 1 week of completion) and 908 were ran- 45–50 GY IN 5 WEEKS
domized to surgery alone. The overall survival
at 5 years was similar in the two groups: 64.2 per Randomized studies in resectable T3/T4 rectal can-
cent in the preoperative radiation group compared cers have compared preoperative chemoradiation

HEBK001-C06_p87-102.indd 91 08/02/13 5:58 PM


92  Preoperative radiotherapy and chemoradiotherapy for rectal cancer

(45 Gy in 25 fractions over 5 weeks) and 5FU-based damage. Mature results of the Swedish Rectal
chemotherapy with radiation alone.27,37 Surgery is Cancer Trial confirm problems such as late bowel
undertaken 4–12 weeks following chemoradio- obstruction and abdominal pain.50 Additionally,
therapy to allow the patient to recover and enable there are unexplained late cardiac effects and an
tumour shrinkage. The addition of 5FU to preoper- increased risk of a second malignancy.51 As follow-
ative radiation increases the pathological complete up in the majority of studies is generally short,
response rate over radiotherapy alone and provides there is likely to be a major underestimate of the
evidence for improvements in locoregional control real risks of late effects.
but has not translated into an improvement in dis- Our caveat would be that in some cases of low
ease-free survival or overall survival. rectal cancer, it may be difficult to discriminate
Trials in unresectable disease have also confirmed between a cT2 and cT3 or even cT4 at the level of the
an advantage for chemoradiation over radiation levators and below. High-quality MRI is not always
alone.40 Local control was significantly better (82 available. Low rectal tumours behave more aggres-
per cent v. 67 per cent at 5 years, P 5 0.03), with sively compared with cancers in the upper rectum.
a trend towards improved overall survival (66 per High-quality surgery cannot always achieve a cura-
cent v. 53 per cent at 5 years, P 5 0.09). tive resection for locally advanced cancers that
extend below the levators. Currently, more aggres-
sive surgical approaches such as the ‘cylindrical’ or
Oral Alternatives to 5-Fluorouracil extralevator approach are being explored, and these
may eventually be shown to be superior to standard
In the neoadjuvant setting in rectal cancer, the safety abdominoperineal excision. Preoperative chemora-
and efficacy of capecitabine45 and a combination of diation still has a major role in many patients with
uracil and tegafur46 have been compared with that low rectal cancer, however.
of a continuous intravenous infusion (CVI) of 5FU. It should be stressed that the choice of not using
For capecitabine, the authors used data from 345 preoperative 5FU-based chemoradiotherapy repre-
patients treated with capecitabine and compared sents a positive decision to forgo effective neoadju-
them with 197 patients treated with CVI 5FU.45 The vant treatment. If the surgical histopathology sub-
pathological complete response was significantly sequently reveals a positive circumferential margin,
higher for capecitabine than CVI 5FU (25 per cent v. the optimal window for treatment may have been
13 per cent). lost.

When is it safe to proceed SHORT-COURSE PREOPERATIVE


to surgery alone? RADIATION VERSUS
CHEMORADIOTHERAPY
The selection of patients in whom initial surgical
resection is safe and who do not require preop- In Europe and Australia, two trials have directly
erative radiotherapy remains a controversial issue. compared SCPRT with chemoradiation for patients
The present authors believe that patients in groups with resectable rectal tumours.52,53 The Polish study
1 and 2, and even some categories of group 3, can found no difference in long-term outcomes of loco-
avoid preoperative radiotherapy in view of the sig- regional control, disease-free survival or overall sur-
nificant associated morbidity in terms of increased vival (Table 6.1). Preliminary results from the Aus-
pelvic and perineal post-surgical infections, and tralian study also failed to show differences in late
significant late anorectal, urinary and sexual dys- outcome. If the premise is accepted that in resectable
function.18 About 5–10 per cent of patients experi- cancers, where the CRM is not threatened, SCPRT
ence grade 3 or 4 late morbidity.47,48 Small bowel and chemoradiotherapy are equivalent in terms of
tolerance is the main dose-limiting factor, and the outcomes such as local recurrence, disease-free sur-
volume of the small bowel in the radiation field is vival and overall survival, then these results raise
crucial,49 although only 1–2 per cent of patients questions about the importance of down-staging
require early surgical intervention for small ­bowel and the effect of pathological ­complete response

HEBK001-C06_p87-102.indd 92 08/02/13 5:58 PM


HEBK001-C06_p87-102.indd 93

Table 6.1. Trial design: preoperative radiotherapy versus preoperative chemoradiation in resectable rectal cancer.@

SHORT-COURSE PREOPERATIVE RADIATION VERSUS CHEMORADIOTHERAPY  93


Pathology
Disease- Pathological circumferential
Patients Patients Primary Local Overall free complete resection
Trial Duration (n) Randomization (n) TME end-point recurrence Metastases survival survival response margin
EORTC 54
1972–1984, 247 34.5 Gy v. 5FU 1 121 v. No Overall 15% v. 30% overall 59% v. 46% 72% v. 2.5% v. 5% Not Quirke
12 years 34.5 Gy 126 survival 15% at at 5 years 68% at
5 years 3 years
EORTC 1992–2004, 1011 45 Gy in 25 fractions 506 v. No Overall 17.1% v. 34.4% 64.8% v. 54.4% v. 5% v. 14% Not Quirke
2292127 12 years v. FUFA 1 45 Gy 505 survival 8.7% at overall 65.8% at 56.1% at
5 years 5 years 5 years
FFCD 1993–2005, 762 45 Gy in 25 fractions 367 v. No Overall 16.5% v. No data 67.9% v. No data 3.6% v. Not Quirke
920337 12 years v. FUFA 1 45 Gy 375 survival 8.1% at 67.4% at 11.4%
5 years 5 years
Polish 1999–2004, 316 25 Gy in 5 fractions/ 155 v. Yes Sphincter- 11% v. No data 67.2% v. 58.4% v. 1% v. 16% 13% v. 4%
trial52 5 years SCPRT v. FUFA 1 157 preserving 16.5% at 66.2% at 55.6% at (CRM)
50 Gy surgery 5 years 4 years 4 years
TROG 01- 2001–2006, 326 25 Gy in 5 fractions/ No data Yes Local 7.5% v. 72% v. 69% 74% v. 70% No data No data Not Quirke v. no
0453 5 years SCPRT v. 50.4 Gy 1 recurrence 4.1% at at 5 years data
PVI 5FU 3 years

EORTC, European Organisation for Research and Treatment of Cancer; FFCD, Fédération Francophone de Cancérologie Digestive; 5FU, 5-fluorouracil; FUFA,
5-fluorouracil plus folinic acid; PVI, protracted venous infusion; TME, total mesorectal excision; TROG, Trans-Tasman Radiation Oncology Group.
08/02/13 5:58 PM
94  Preoperative radiotherapy and chemoradiotherapy for rectal cancer

on survival. Additionally, a positive CRM may not others used synchronous chemoradiation.36,60,61 The
be as significant an adverse prognostic factor after putative hypothesis is supported by evidence from
SCPRT as after chemoradiotherapy. randomized studies. Preoperative chemoradiation
appears to offer a 10 per cent13 or even a 20 per
cent12,62 higher chance overall in achieving sphinc-
Facilitating sphincter- ter-saving surgery. These trials were not specifically
sparing procedures designed to answer the question of the sphincter
preservation rate, however, and these data represent
Many surgeons consider the low position of some subset analysis. The German data are particularly
rectal cancers (below 6 cm from the anal verge) to liable to unforeseen biases, as the Zelen method of
inevitably require an abdominoperineal excision randomization was used, and there are significant
resection, particularly if the sphincter is invaded. differences in the numbers in each group.
In addition, a bulky anterior tumour in an obese
man with a narrow pelvis may prove technically
demanding to achieve sphincter-sparing surgery. RADIOTHERAPY ISSUES
These situations have led to a commonly held
belief that preoperative radiation will allow an Field Size
increase in the rate of sphincter-preserving proce-
dures performed. Possibly, but not consistently, if The optimal clinical target volume in preoperative
a bulky polypoidal tumour shrinks, this may allow radiotherapy and chemoradiotherapy for resectable
the surgeon to safely navigate around the mesorectal rectal cancer before TME remains poorly defined.
fascia down on to the pelvic floor and achieve a low Total mesorectal excision does not remove all nodal
anterior resection. This would not be possible with- stations potentially harbouring subclinical dis-
out prior radiation, in which case abdominoperine- ease. Contouring a clinical target volume is a bal-
al excision may be required. More controversially is ance between encompassing structures at risk of
the choice of the distal resection margin. Should this containing residual cancer cells after TME and the
be chosen as 1 cm below the distal edge of a tumour desire to minimize normal tissue toxicity and surgi-
that has shown major regression, or is it only onco- cal m
­ orbidity.
logically safe to choose the distal margin on the basis Many series in the pre-TME era mapped recur-
of the initial tumour location and size? rence, including the seminal work from Gunderson
One study compared immediate and delayed sur- at the Mayo Clinic, amassed from ‘second-look’ sur-
gery after preoperative radiotherapy.55,56 Sphincter gery.63 Previously, groups have made recommenda-
preservation was achieved in 79 per cent of patients tions for clinical target volume contouring based on
with a long interval (4–6 weeks) following radio- observed sites of local recurrence.64–72 Historically,
therapy, compared with 69 per cent of patients with wide-field radiotherapy using bony landmarks
a shorter interval (2 weeks). A further randomized was delivered to almost all patients. Early studies
study from this group suggests that dose escalation defined the superior border as the junction of L5/
of the radiotherapy with an endoluminal boost may S1, but some studies treated up to the origin of the
offer a higher rate of complete clinical response and inferior mesenteric artery at L4 and were associated
hence increase the chance of sphincter preservation with significant morbidity.
from 44 per cent to 76 per cent.57 Field sizes delivered in the USA are generally
Several non-randomized surgical series have larger than in Europe. The recommended field size
reported results of patients with clinically resect- in one randomized phase II study suggested ‘The
able rectal cancer, in whom clinical assessment by superior border of the treatment volume was at
their surgeon categorized the height of the can- the L5–S1 junction with the inferior border a mini-
cer from the anal verge as requiring an abdomi- mum of 5 cm inferior to the distal-most extent of
noperineal excision. These patients then received the tumour or the anal verge as identified by a mark-
long-course preoperative radiotherapy and were er on simulation’ [our italics].66 In the UK field sizes
subsequently reassessed before definitive surgery. are likely to be at least 4–5 cm shorter in the cranio-
Some studies used radiotherapy alone,55,58,59 and caudal aspect.

HEBK001-C06_p87-102.indd 94 08/02/13 5:58 PM


RADIOTHERAPY ISSUES  95

The studies of local recurrence after TME relate ing to the linear quadratic equation, but in prac-
to SCPRT, and there are very few relating to chemo- tice may be even higher. Early retrospective studies
radiotherapy. Lymph nodes and their precise loca- suggest an increase in local control with a dose of
tions are well described in Far Eastern series but not 50 Gy compared with 40 Gy.74 More recent data from
generally in Western series. Few studies recommend phase II studies suggest that higher doses may be
individualizing clinical target volume according to associated with lower risks of local relapse.75–77
site, stage and risk. Data on quality of life and late A sequential phase II study from Canada has been
morbidity are sparse. We recommend clinical tar- reported. Three sequential schedules combined
get volume-customizing delineations based on site radiation with infusional 5FU, escalated from 40 Gy
and stage, with adaptations for high-risk features in 20 fractions to 46 Gy in 23 fractions and finally
according to clinical and MRI staging. Although to 50 Gy in 25 fractions. A statistically significant
these proposed clinical target volume delineations difference in terms of local control was observed
have not been evaluated in randomized trials, their for doses of 46 Gy and above, but there was no dif-
use appears rational and based on current evidence. ference between doses of 46 Gy and 50 Gy.78 The
same study also appeared to show a trend to higher
pathological complete response rates with increas-
Total Dose ing radiation dose of 13 per cent, 21 per cent and
31 per cent for 40 Gy, 46 Gy and 50 Gy, respectively.
Conventionally, when 1.8 Gy per fraction is used,
total doses in the range 45–50.4 Gy have been deliv-
Hyperfractionation
ered in the preoperative setting, and 50.4 Gy with the
option of a 5.4 Gy boost to the tumour bed in the
Short-course preoperative radiotherapy uses fewer
postoperative setting. It is assumed that the treat-
fractions at a higher dose per fraction (5 3 5 Gy),
ment will be delivered 5 days per week, 1 ­fraction per
which might be expected to cause more late mor-
day, 1.8 Gy per fraction. The most appropriate dose is
bidity64,66 and which cannot allow integration of
not precisely known, but in association with chemo-
chemotherapy. Hence, a number of studies have
therapy the total radiation dose should be at least
used hyperfractionated radiotherapy with a view to
45 Gy in fractions of 1.8–2 Gy. A boost of 4–6 Gy in
reducing late effects.79–81
two to four fractions to the primary tumour is often
Widder and colleagues reported SCPRT with twice-
given, thus reducing the radiation dose to the entire
daily fractions of 2.5 Gy to a total dose of 25 Gy within
volume when chemoradiotherapy is given.
1 week.79 The authors reported a high local control
Ideal preoperative dose rate (98 per cent after 4 years) along with low rates of
toxicity. It should be kept in mind, however, that theo-
Some SCPRT studies have used 20 Gy in five frac-
retical anti-tumour efficiency is significantly reduced
tions,73 but the Swedish Rectal Cancer Study, the
in this fractionation protocol. A more recent study
Dutch study and the CR07 study use 25 Gy in five
modified SCPRT in rectal cancer to deliver twice-
fractions, which most clinicians accept is a dose
daily fractions of 2.9 Gy to a total dose of 29 Gy in 1
fractionation that cannot safely be exceeded.
week immediately before surgery.82 Adjuvant chemo-
With the use of 1.8–2.0 Gy per fraction, how-
therapy was intended to be offered subsequently to
ever, the total dose may range from 45 Gy to 54 Gy
patients with any pathological tumour assessment of
in most reported series. This upper limit is 20 per
stage II or higher in the resected specimen.
cent higher than 45 Gy, and the balance between
improved local control and the increased risk of late
toxicity is continually debated. Brachytherapy
The Colorectal Cancer Collaborative Group
meta-analysis confirmed that when preoperative High-dose-rate intraluminal brachytherapy (HDR-
radiotherapy is used, a biologically equivalent dose ILBT) has the advantage of high conformality, i.e.
greater than 30 Gy is more effective in reducing local a rapid fall-off of radiation dose, which allows the
relapse.16 A dose of 5 Gy daily for 5 days represents delivery of a high dose to the tumour while spar-
a biologically equivalent dose of 37.5 Gy accord- ing normal surrounding structures such as the

HEBK001-C06_p87-102.indd 95 08/02/13 5:58 PM


96  Preoperative radiotherapy and chemoradiotherapy for rectal cancer

contralateral normal rectal mucosa, bladder and Phase II trials provide additional toxicity data
small bowel.83,84 Limited data are available evalu- and potential regimens for future phase III rand-
ating the advantages of HDR-ILBT with external omized trials in patients with rectal cancer. The
beam radiotherapy (EBRT) compared with EBRT Radiation Therapy Oncology Group (RTOG) 0247
alone.85 High-dose-rate intraluminal brachytherapy trial, a randomized phase II trial, compared capecit-
for advanced or inoperable tumours of the rectum abine administered 5 days per week and weekly
has been used both in the palliative setting and to oxaliplatin plus radiation administered before sur-
dose escalate after chemoradiation for curative treat- gery followed by nine cycles of postoperative oxali-
ment.86,87 It has also been used as a single modality in platin with 5FU and folinic acid chemotherapy
the preoperative setting, with 30 per cent pathologi- (FOLFOX) with irinotecan on a weekly schedule
cal complete response;83 again, however, there are substituted for oxaliplatin.
limited data regarding HDR-ILBT as a boost to the Another phase II trial, E3204, combines capecit-
gross tumour volume in combination with preop- abine administered 5 days per week plus weekly
erative chemoradiation for rectal cancers. oxaliplatin and bevacizumab with radiation every
In principle HDR-ILBT as a component or fol- other week; postoperative patients receive 12 cycles
lowing chemoradiation may avoid a colostomy by of FOLFOX and bevacizumab.
increasing down-staging and facilitating sphincter Two randomized phase III studies have examined
preservation57 or by potentiating a non-operative one versus two cytotoxic drugs (Table 6.2).22,23 In
approach.84 the Italian Studio Terapia Adjuvante Retto (STAR)
trial, oxaliplatin added little to early assessable
end-points but increased toxicity when added to a
Intensity-Modulated Radiotherapy standard preoperative chemoradiation regimen for
locally advanced rectal cancer. There were no signif-
Technical advances such as IMRT allow greater icant differences in pathological complete response,
precision and sparing of normal surrounding local tumour response or tumour down-staging
structures such as small bowel compared with con- between patients who received FU-based chemora-
ventional two-dimensional or three-dimensional diation with weekly oxaliplatin infusions.22
planning. Intensity-modulated radiotherapy may The Fédération Francophone de Cancérologie
allow improved compliance or facilitate dose escala- Digestive (FFCD) Fédération Nationale des Centres
tion without increasing late morbidity. In addition, de Lutte Contre le Cancer (FNCLCC) ACCORD
there was no difference with regard to late toxicity 12 trial (NCT00227747) compares neoadjuvant
for any of the four arms in the European Organisa- preoperative capecitabine combined with 45 Gy in
tion for Research and Treatment of Cancer (EORTC) 25 fractions over 5 weeks against capecitabine and
22921 study trial: 522 patients retained their sphinc- oxaliplatin partnered with 50 Gy in 25 fractions over
ters, and of these only 1.4 per cent required surgery 5 weeks. Only data on toxicity and early pathological
for small bowel complications. This low level of late and surgical end-points are currently available.23,37
morbidity calls into question the need to deliver The design of this study is poor, because capecitab-
IMRT with the aim of reducing small bowel toxicity. ine in combination with 45 Gy (arm A) is compared
with capecitabine/oxaliplatin and 50 Gy (arm B).
The pathological complete response appears higher
Other cytotoxic drugs (18.8 per cent) in the capecitabine/oxaliplatin arm
(arm B) than in arm A (13.8 per cent), but it is not
The addition of oxaliplatin and irinotecan to 5FU- clear whether this reflects the addition of oxalipla-
based chemotherapy or oral fluoropyrimidines has tin or the higher radiotherapy dose.23 It remains to
been explored within a chemoradiotherapy sched- be seen whether disease-free survival and overall
ule in numerous phase II studies in an attempt survival are improved by the strategy of adding
to increase tumour shrinkage before surgery and oxaliplatin in this setting, but in the meantime the
­potentially mirror the success of oxaliplatin in deal- addition of irinotecan and ­oxaliplatin to 5FU-based
ing with distant micro-metastases in the adjuvant chemoradiation in patients with resectable tumours
setting in colon cancer.20,21 remains investigational.

HEBK001-C06_p87-102.indd 96 08/02/13 5:58 PM


THE FUTURE  97

Table 6.2. Ongoing phase III trials of neoadjuvant chemoradiation in resectable rectal cancer.
Concurrent
chemoradiotherapy Radiotherapy
Study (CRT) randomization dose Patients (n) Preoperative treatment Comments
NSABP R-04, Capecitabine or 5FU/ 50.4 Gy in 28 1606; Recommended to consider Primary end-
NCT00058474 CRT v. capecitabine fractions and closed to ECOG 5204 point: 3-year
or 5FU 1 oxaliplatin 55.8 Gy for recruitment locoregional
(every 2 weeks)/CRT fixed tumours disease control
CAO/ARO/AIO CI 5FU (days 1–15, 50 Gy in 25 1259; 5FU 34 or FOLFOX 34 Primary end-
04 22–35)/CRT v. CI fractions over closed to point: disease-
5FU 1 oxaliplatin as 33 days recruitment free survival at
radiosensitizer (days 3 years
1, 8, 22, 29)
PETACC 6/ Capecitabine/ 45 Gy in 25 Open Capecitabine 36 or XELOX Primary end-
EORTC 40054, radiotherapy v. fractions over 36; adjuvant chemotherapy point: disease-
NCT00766155 capecitabine 1 33 days 6 regimen continues free survival
oxaliplatin (every 5.4 Gy boost preoperative agents
week)/radiotherapy

CI, continuous infusion; ECOG, Eastern Cooperative Oncology Group; FOLFOX, oxaliplatin with 5-fluorouracil
and folinic acid chemotherapy; 5FU, 5-fluorouracil; XELOX, capecitabine with oxaliplatin.

Integration of biologicals Tumors (RECIST) criteria was 19 per cent versus 13


per cent for the addition of cetuximab to cisplatin
Integration of targeted drugs such as cetuximab and 5FU.89 Median progression-free survival was
into preoperative chemoradiation schedules in rec- 9.5 months for cetuximab with cisplatin and 5FU
tal adenocarcinoma is attractive in principle. The and 5.5 months for cisplatin and 5FU, demonstrat-
preliminary results of chemoradiation clinical trials ing that the response may not be the best end-point.
with cetuximab, on the early clinical end-point of The integration of bevacizumab has consider-
pathological complete response, are disappointing able preclinical rationale. Experimental studies in
however. Cetuximab can lead to G1 or G2/M-cell human tumour xenograft models have shown that
cycle arrest; if only a small proportion of cells with- vascular endothelial growth factor (VEGF) block-
in the tumour are affected, this decrease in prolif- ade serves as a potent and non-toxic enhancer of
eration could impact on the chance of achieving a radiation. Anti-VEGF decreases interstitial pres-
complete pathological response. sure, increases oxygenation, and reverses radiation
A large multinational randomized phase II study resistance conferred by hypoxia. Few phase II tri-
EXPERT-C (NCT00383695) has compared neoad- als have been reported, but the combination with
juvant therapy comprising oxaliplatin, capecitabine chemoradiotherapy appears potentially deliverable
and chemoradiotherapy with or without cetuxi- with acceptable toxicity. There are five other ongo-
mab in 164 patients. The study was completed in ing or recently closed phase III trials registered on
July 2008, and results may throw more light on the http://clinicaltrials.gov website.
combinations of cetuximab and chemoradiation
in the clinical setting in locally advanced rectal
cancer. The combination may yet reproduce the THE FUTURE
improvement in long-term results achieved when
cetuximab has been combined with radiation alone The best opportunity to improve survival in patients
in studies of head and neck cancer.88 Also, in a with rectal cancer will require continued focus on
randomized phase II trial of squamous cell carci- adjuvant chemotherapy strategies, since the devel-
noma of the oesophagus, the overall response rate opment of metastases is the predominant cause of
according to Response Evaluation Criteria In Solid recurrence and death. At the same time, we need to

HEBK001-C06_p87-102.indd 97 08/02/13 5:58 PM


98  Preoperative radiotherapy and chemoradiotherapy for rectal cancer

achieve better documentation and understanding of l Current evidence does not support the degree
pursuing detailed assessment of acute and chronic of response to chemoradiation (e.g. pathologi-
toxicity and the risk of second malignancies. cal complete response; down-sizing the primary
Additionally, if treatment strategies are to be suc- tumour; sterilizing the regional nodes; tumour
cessful in patients with rectal cancer, then a degree of regression grades; residual cell density) as a
individualization in the cytotoxic chemotherapy and valid surrogate for long-term local control or
biological agents – and even further individualization survival, although down-staging may predict a
of radiation – is required. To optimize treatment, we role for adjuvant chemotherapy.
need a deeper understanding of tumour biology. l There is no evidence that conventional preop-

erative chemoradiation can facilitate sphincter-


sparing surgery. It should not be a primary
CONCLUSION reason for administering preoperative chemora-
diation, although in the case of excellent down-
A pelvic MRI scan is an essential part of the initial staging this may prove a bonus.
staging of all patients with rectal cancer 0–15 cm l There is no evidence for increased efficacy of

from the anal verge and can help to define the most chemoradiotherapy over SCPRT in moderate-
appropriate preoperative strategy. There is a well- risk resectable cancers.
established role for both SCPRT and down-staging l Preoperative chemoradiation using 45–54 Gy

long-course chemoradiation. For chemoradiation, over 5–6 weeks with an interval to allow
however, there is no consensus regarding the opti- response and shrinkage is recommended where
mum dose, fractionation, field size or chemotherapy the MRI confirms a ‘threatened’ or breached
schedule. Current evidence does not support the circumferential margin, or where there is a
degree of response to chemoradiation (e.g. patho- radiologically unresectable cancer with disease
logical complete response; down-sizing the prima- outside the mesorectum, i.e. surgery is not pos-
ry tumour; sterilizing the regional nodes; tumour sible without leaving tumour within the pelvis.
regression grades; residual cell density) as a valid
surrogate for long-term local control or survival.
Current trials suggest that in resectable cancers References
where the CRM is not threatened, SCPRT and chem-
  1. Thomas WH, Larson RA, Wright HK, Cleveland JC.
oradiotherapy are equivalent in terms of outcomes
Analysis of 830 patients with rectal adenocarcinoma.
such as local recurrence, disease-free survival and Surg Gynecol Obstet 1969; 129: 10–14.
overall survival. In view of the high risk of metastatic   2. Fisher B, Wolmark N, Rockette H, et al. Postoperative
disease, research strategies should be explored to adjuvant chemotherapy or radiation therapy for rectal
address the problem in the preoperative setting. cancer: results from NSABP protocol R-01. J Natl
More rationally designed preclinical and transla- Cancer Inst 1988; 80: 21–9.
tional studies (with recognized negative predictive   3. Gastrointestinal Tumor Study Group. Prolongation of
the disease-free survival in surgically treated rectal
factors such as k-ras mutations, b-raf mutations and
carcinoma. N Engl J Med 1985; 312: 1465–72.
epidermal growth factor receptor (EGFR) gene copy   4. Krook JE, Moertel CG, Gunderson LL, et al. Effective
numbers) may therefore help select out inappropri- surgical adjuvant therapy for high-risk rectal carci-
ate patients and determine the optimal sequence of noma. N Engl J Med 1991; 324: 709–15.
chemotherapy and biological triple combinations.   5. Gunderson LL, Sargent DJ, Tepper JE, et al. Impact of
Only then can we move on to perform large rand- T and N stage and treatment on survival and relapse in
omized phase III trials. adjuvant rectal cancer: a pooled analysis. J Clin Oncol
2004; 22: 1785–96.
  6. Greene FL, Stewart AK, Norton HJ. New tumor–node–
metastasis staging strategy for node positive (stage III)
Recommendations rectal cancer: an analysis. J Clin Oncol 2004; 22:
1778–84.
l A pelvic MRI scan is an essential part of the   7. Bleehen NM. Combined drug–radiation treatments:
initial staging of all patients with rectal cancer indications and clinical problems. Bull Cancer 1981;
0–15 cm from the anal verge. 68: 127–31.

HEBK001-C06_p87-102.indd 98 08/02/13 5:58 PM


References  99

  8. Tannock IF. Treatment of cancer with radiation and 23. Gérard JP, Azria D, Gourgou-Bourgade S, et al.
drugs. J Clin Oncol 1996; 14: 3156–74. Comparison of two neoadjuvant chemoradiotherapy
  9. Stockholm Rectal Cancer Study Group. Preoperative regimens for locally advanced rectal cancer: results of
short-term radiation therapy in operable rectal carci- the phase III trial ACCORD 12/0405-Prodige 2. J Clin
noma: a prospective randomized trial. Cancer 1990; Oncol 2010; 28: 1638–44.
66: 49–55. 24. Chau I, Brown G, Cunningham D, et al. Neoadjuvant
10. Dahl O, Horn A, Morild I, et al. Low-dose preoperative capecitabine and oxaliplatin followed by synchronous
radiation postpones recurrences in operable rectal chemoradiation and total mesorectal excision in
cancer: results of a randomized multicenter trial in magnetic resonance imaging-defined poor-risk rectal
western Norway. Cancer 1990; 66: 2286–94. cancer. J Clin Oncol 2006; 24: 668–74.
11. Pahlman L, Glimelius B. Pre- or postoperative radio- 25. Fernández-Martos C, Pericay C, Aparicio J, et al.
therapy in rectal and rectosigmoid carcinoma: report Phase II, randomized study of concomitant chemo-
from a randomized multicenter trial. Ann Surg 1990; radiotherapy followed by surgery and adjuvant
211: 187–95. capecitabine plus oxaliplatin (CAPOX) compared with
12. Sauer R, Becker H, Hohenberger W, et al. Preopera- induction CAPOX followed by concomitant chemoradi-
tive versus postoperative chemoradiotherapy for rectal otherapy and surgery in magnetic resonance imaging-
cancer. N Engl J Med 2004; 351: 1731–40. defined, locally advanced rectal cancer: Grupo cancer
13. Roh MS, Colangelo LH, O’Connell MJ, et al. Preop- de recto 3 study. J Clin Oncol 2010; 28: 859–65.
erative multimodality therapy improves disease-free 26. Chua YJ, Barbachano Y, Cunningham D, et al.
survival in patients with carcinoma of the rectum: Neoadjuvant capecitabine and oxaliplatin before
NSABP-R03. J Clin Oncol 2009; 27: 5124–30. chemoradiotherapy and total mesorectal excision in
14. Quirke P, Dixon MF, Durdey P, Williams NS. Local MRI-defined poor-risk rectal cancer: a phase 2 trial.
recurrence of rectal cancer due to inadequate surgical Lancet Oncol 2010; 11: 241–8.
resection. Lancet 1986; 2: 996–9. 27. Bosset JF, Collette L, Calais G, et al. Chemotherapy
15. Camma C, Giunta M, Fiorica F, Pagliaro L, Craxi A, with pre-operative radiotherapy in rectal cancer.
­Cottone M. Preoperative radiotherapy for resectable rec- N Engl J Med 2006; 355: 1114–23.
tal cancer: a meta-analysis. JAMA 2000; 284: 1008–15. 28. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevaci-
16. Colorectal Cancer Collaborative Group. Adjuvant zumab plus irinotecan, fluorouracil and leucovorin for
radiotherapy for rectal cancer: a systematic overview metastatic colorectal cancer. N Engl J Med 2004; 350:
of 8507 patients from 22 randomised trials. Lancet 2335–42.
2001; 358: 1291–304. 29. Van Cutsem E, Labianca R, Bodoky G, et al. Rand-
17. Munro AJ, Bentley AHM. Adjuvant radiotherapy in omized phase III trial comparing biweekly infusional
operable rectal cancer: a systematic review. Semin fluorouracil/leucovorin alone or with irinotecan in
Colon Rectal Surg 2002; 13: 31–42. the adjuvant treatment of stage III colon cancer:
18. Wong R, Tandan V, De Silva S, Figueredo A. Preopera- PETACC-3. J Clin Oncol 2009; 27: 3117–25.
tive radiotherapy and curative surgery for the manage- 30. Tol J, Koopman M, Cats A, et al. Chemotherapy,
ment of localized rectal cancer. Cochrane Database bevacizumab, and cetuximab in metastatic colorectal
Syst Rev 2007; (2): CD002102. cancer. N Engl J Med 2009; 360: 563–72.
19. Moertel CG, Fleming TR, Macdonald JS, et al. 31. Folprecht G, Gruenberger T, Bechstein WO, et al.
­Fluorouracil plus levamisole as effective adjuvant Tumour response and secondary resectability of color-
therapy after resection of stage III colon carcinoma: a ectal liver metastases following neoadjuvant chemo-
final report. Ann Intern Med 1995; 122: 321–6. therapy with cetuximab: the CELIM randomised phase
20. Kuebler JP, Wieand HS, O’Connell MJ, et al. Oxalipla- 2 trial. Lancet Oncol 2010; 11: 38–47.
tin combined with weekly bolus fluorouracil and leu- 32. Janjan NA, Crane C, Feig BW, et al. Improved overall
covorin as surgical adjuvant chemotherapy for stage II survival among responders to preoperative chemora-
and III colon cancer: results from NSABP C-07. J Clin diation for locally advanced rectal cancer. Am J Clin
Oncol 2007; 25: 2198–204. Oncol 2001; 24: 107–12.
21. André T, Boni C, Navarro M, et al. Improved overall 33. Read TE, Ogunbiyi OA, Fleshman JW, et al. Neoad-
survival with oxaliplatin, fluorouracil, and leucovorin as juvant external beam radiation and protectomy for
adjuvant treatment in stage II or III colon cancer in the adenocarcinoma of the rectum. Dis Colon Rectum
MOSAIC trial. J Clin Oncol 2009; 27: 3109–16. 2001; 44: 1778–90.
22. Aschele C, Pinto C, Rosati G, et al. Preoperative 34. Hiotis SP, Weber SM, Cohen AM, et al. Assessing
(FU)-based chemoradiation with and without weekly the predictive value of clinical complete response to
oxaliplatin in locally advanced rectal cancer: pathologic neoadjuvant therapy for rectal cancer: an analysis of
response analysis of the Studio Terapia Adjuvante 488 patients. J Am Coll Surg 2002; 194: 131–5.
Retto (STAR)-01 randomized phase III trial. J Clin 35. Ruo L, Tikoo S, Klimstra D, et al. Long term prognos-
Oncol 2009; 27: 804s. tic significance of extended rectal cancer response to

HEBK001-C06_p87-102.indd 99 08/02/13 5:58 PM


100  Preoperative radiotherapy and chemoradiotherapy for rectal cancer

preoperative radiation and chemotherapy. Am Surg 48. Tepper JE, O’Connell, Nieddzwiecki D, et al. Adjuvant
2002; 236: 75–81. therapy in rectal cancer: analysis of stage, sex and
36. Valentini V, Coco C, Picciocchi A, et al. Does down- local control – final report of Intergroup 0114. J Clin
staging predict improved outcome after preoperative Oncol 2002; 20: 1744–50.
chemoradiation for extraperitoneal locally advanced 49. Gallagher MJ, Brereton HD, Rostock RA. A prospec-
rectal cancer? A long term analysis of 165 patients. tive study of treatment techniques to minimise the
Int J Radiat Oncol Biol Phys 2002; 53: 664–74. volume of pelvic small bowel with reduction of acute
37. Gerard JP, Conroy T, Bonnetain F, et al. Preoperative and late effects associated with pelvic irradiation. Int J
radiotherapy with or without concurrent fluorouracil Radiat Oncol Biol Phys 1986; 12: 1565–73.
and leucovorin in T3–T4 rectal cancers: results of 50. Birgisson H, Pahlman L, Glimelius B. Adverse effects
FFCD 9203. J Clin Oncol 2006; 24: 4620–25. of preoperative radiation therapy for rectal cancer:
38. The Swedish Rectal Cancer Trial. Improved survival long-term follow-up of the Swedish Rectal Cancer
with preoperative radiotherapy in resectable cancer. Trial. J Clin Oncol 2006; 23: 8697–705.
N Engl J Med 1997; 336: 980–7. 51. Birgisson H, Pahlman L, Gunnarsson U, Glimelius B.
39. Kapiteijn E, Marijnen CA, Nagtegaal JD, et al. Preop- Occurrence of second cancers in patients treated with
erative radiotherapy combined with total mesorectal radiotherapy for rectal cancer. J Clin Oncol 2005; 23:
excision for resectable rectal cancer. N Engl J Med 6126–31.
2001; 345: 638–46. 52. Bujko K, Nowacki MP, Nasierowska-Guttmejer A, et al.
40. Braendengen M, Tveit KM, Berglund A, et al. Ran- Sphincter preservation following preoperative radio-
domized phase III study comparing preoperative therapy for rectal cancer: report of a randomized trial
radiotherapy with chemoradiotherapy in nonresectable comparing short-term radiotherapy vs. conventionally
rectal cancer. J Clin Oncol 2008; 26: 3687–94. fractionated radiochemotherapy. Radiother Oncol
41. Frykholm GJ, Pahlman L, Glimelius B. Combined 2004; 72: 15–24.
chemo and radiotherapy vs radiotherapy alone in 53. Ngan S. A randomized trial comparing local recur-
the treatment of primary non-resectable cancer of rence rates between short-course and long-course
the rectum. Int J Radiat Oncol Biol Phys 2001; 50: preoperative radiotherapy for clinical T3 rectal cancer:
433–40. an intergroup trial (TROG, AGITG, CSSANZ, RACS).
42. Folkesson J, Birgisson H, Pahlman L, et al. Swedish J Clin Oncol 2010; 28: 15s.
Rectal Cancer Trial: long lasting benefits from radio- 54. Boulis-Wassif S, Gerrard A, Loygue J, et al. Final
therapy on survival and local recurrence rate. J Clin results of a randomised trial on the treatment of rectal
Oncol 2005; 23: 5644–50. cancer with pre-operative radiotherapy alone or in
43. Peeters K, van de Velde C, Leer J, et al. Late side combination with 5-fluoro-uracil followed by radical
effects of short-course preoperative radiotherapy com- surgery. Cancer 1984; 53: 1811–8.
bined with total mesorectal excision for rectal cancer: 55. Francois Y, Nemoz CJ, Baulieux J, et al. Influence of
increased bowel dysfunction in irradiated patients – the interval between preoperative radiation therapy
Dutch Colorectal Cancer Group study. J Clin Oncol and surgery on downstaging and on the rate of
2005; 23: 6199–206. sphincter-sparing surgery for rectal cancer: the Lyon
44. Quirke P, Steele R, Monson J, et al. MRC CR07/NCIC- R90-01 Randomised Trial. J Clin Oncol 1999; 17:
CTG CO16 Trial Investigators, NCRI Colorectal Cancer 2396–402.
Study Group. Effect of the plane of surgery achieved 56. Glehen O, Chapet O, Adham M. Long-term results
on local recurrence in patients with operable rectal of the Lyon R90-01 randomized trial of preoperative
cancer: a prospective study using data from the MRC radiotherapy with delayed surgery and its effect on
CR07 and NCIC-CTG CO16 randomised clinical trial. sphincter-saving surgery in rectal cancer. Br J Surg
Lancet 2009; 373: 821–8. 2003; 90: 996–8.
45. Saif MW, Hashmi S, Zelterman D, et al. Capecitabine 57. Gerard JP, Chapet O, Nemoz C, et al. Improved
vs continuous infusion 5-FU in neoadjuvant treatment sphincter preservation in low rectal cancer with high
of rectal cancer: a retrospective review. Int J Colorectal dose preoperative radiotherapy: the Lyon R96-02
Dis 2008; 23: 139–45. randomized trial. J Clin Oncol 2004; 22: 2404–9.
46. De la Torre A, Garcia-Berical MI, Arrias F, et al. Preop- 58. Wagman R, Minsky BD, Cohen AM, et al. Sphincter
erative chemoradiotherapy for rectal cancer: rand- preservation in rectal cancer with preoperative radia-
omized trial comparing oral uracil and tegafur and oral tion therapy and coloanal anastomosis: long term
leucovorin vs. intravenous 5-fluorouracil and leucov- follow-up. Int J Radiat Oncol Biol Phys 1998; 42:
orin. Int J Radiat Oncol Biol Phys 2008; 17: 102–10. 51–7.
47. Dahlberg M, Glimerlius B, Pahlman L. Preoperative 59. Rouanet P, Fabre JM, Dubois JB, et al. Conservative
radiation effect functional results after surgery for rectal surgery for low rectal carcinoma after high-dose radia-
cancer: results from a randomised study. Dis Colon tion: functional and oncologic results. Ann Surg 1995;
Rectum 1998; 41: 543–9. 221: 67–73.

HEBK001-C06_p87-102.indd 100 08/02/13 5:58 PM


References  101

60. Grann A, Feng B, Wong M, et al. Preoperative com- 74. Fortier GA, Constable WC, Meyers H, et al. Preop-
bined modality therapy for clinically resectable uT3 erative radiation therapy for rectal cancer. Arch Surg
rectal adenocarcinoma. Int J Radiat Oncol Biol Phys 1986; 121: 1380–4.
2001; 49: 987–95. 75. Chan AK, Wong AO, Langevin J, et al. Preoperative
61. Kuvshinoff B, Maghfoor I, Miedema B, et al. Distal chemoradiotherapy and pelvic radiation for tethered or
margin requirements after preoperative chemoradio- fixed rectal cancer: a phase II dose escalation study.
therapy for distal rectal carcinomas: are , or = 1 cm Int J Radiat Oncol Biol Phys 2000; 48: 843–56.
distal margins sufficient? Ann Surg Oncol 2001; 8: 76. Ahmad NR, Marks G, Mohiuddin M. High-dose preop-
163–9. erative radiation for cancer of the rectum: impact of
62. Sauer R, Fietkau R, Wittekind C, et al. Adjuvant vs. radiation dose on patterns of failure and survival. Int J
neoadjuvant radiochemotherapy for locally advanced Radiat Oncol Biol Phys 1993; 27: 773–8.
rectal cancer: the German trial CAO/ARO/AIO-94. 77. Mohiuddin M, Winter K, Mitchell E, et al. Randomized
Colorectal Dis 2003; 5: 406–15. phase II study of neoadjuvant combined-modality
63. Gunderson LL, Sosin H. Areas of failure found at chemoradiation for distal rectal cancer: Radiation
reoperation (second or symptomatic look) following Therapy Oncology Group Trial 0012. J Clin Oncol
‘curative surgery’ for adenocarcinoma of the rectum: 2006; 24: 650–5.
clinicopathologic correlation and implications for 78. Wiltshire KL, Ward IG, Swallow C, et al. Preoperative
­adjuvant therapy. Cancer 1974; 34: 1278–92. radiation with concurrent chemotherapy for resectable
64. Minsky B. Pelvic radiation therapy in rectal cancer: rectal cancer: effect of dose escalation on pathologic
technical considerations. Semin Radiat Oncol 1993; complete response, local recurrence-free survival,
3: 42–7. disease-free survival, and overall survival. Int J Radiat
65. Bagatzounis A, Kolbl O, Muller G, et al. The locore- Oncol Biol Phys 2006; 64: 709–16.
gional recurrence of rectal carcinoma: a computed 79. Widder J, Herbst F, Dobrowsky W, et al. Preoperative
tomographic analysis and a target volume concept for short-term radiation therapy (25 Gy, 2.5 Gy twice daily)
adjuvant radiotherapy. Strahlenther Onkol 1997; 173: for primary resectable rectal cancer (phase II). Br J
68–75. Cancer 2005; 92: 1209–14.
66. Minsky BD. Rectal cancer. In Leibl SA, Phillips TL 80. Brooks S, Glynne-Jones R, Novell R, et al. Short
(eds). Textbook of Radiation Oncology. Philadelphia, course continuous hyperfractionated accelerated
PA, WB Saunders, 1998: 686–71. radiotherapy (CHART) as pre-operative treatment for
67. Lorchel F, Maingon P, Crehange G, et al. Cancer du rectal cancer. Acta Oncol 2006; 45: 1079–85.
rectum: volumes cibles de la radiotherapie preopera- 81. Coucke PA, Notter M, Matter M, et al. Effective timing
toire. Cancer Radiother 2002; 6(suppl. 1): 93–9s. of surgery on survival after preoperative hyperfrac-
68. Arcangeli S, Valentini V, Nori SL, et al. Underlying tionated accelerated radiotherapy (HART) for locally
anatomy for CTV contouring and lymphatic drainage in advanced rectal cancer (LARC): is it a matter of days?
rectal cancer radiation therapy. Rays 2003; 28: 331–6. Acta Oncol 2006; 45: 1086–93.
69. Myerson R, Drzymala R. Technical aspects of image- 82. Guckenberger M, Wulf J, Thalheimer A, et al. Pro-
based treatment planning of rectal carcinoma. Semin spective phase II study of preoperative short-course
Radiat Oncol 2003; 13: 433–40. radiotherapy for rectal cancer with twice daily fractions
70. Gunderson LL, Haddock MG, Gervaz PA. Clinical of 2.9 Gy to a total dose of 29 Gy: long-term results.
target volume in conformal and intensity modulated Radiat Oncol 2009; 4: 67.
radiation therapy: a clinical guide to cancer treatment. 83. Kamikonya N, Hishikawa Y, Kurisu K, Taniguchi M,
In Grégoire V, Scalliet P, Ang KK (eds). Rectal and Anal Miura T. Primary rectal cancer treated with high-dose-
Cancer in Conformal Radiotherapy Planning: Selection rate intraluminal brachytherapy following external
and Delineation of Lymph Node Areas. Heidelberg, radiotherapy. Radiat Med 1991; 9: 85–7.
Springer-Verlag, 2003: 187–97. 84. Vuong T, Belliveau PJ, Michel RP, et al. Conformal
71. Portaluri M, Bambace S, Perez C, et al. Clinical and preoperative endorectal brachytherapy treatment for
anatomical guidelines in pelvic cancer contouring for locally advanced rectal cancer: early results of a phase
radiotherapy treatment planning. Cancer Radiother I/II study. Dis Colon Rectum 2002; 45: 1486–93.
2004; 8: 222–9. 85. Kaufman N, Nori D, Shank B, et al. Remote afterload-
72. Höcht S, Hammad R, Thiel HJ. Recurrent rectal ing intraluminal brachytherapy in the treatment of rec-
cancer within the pelvis: a multicenter analysis of 123 tal, rectosigmoid, and anal cancer: a feasibility study.
patients and recommendations for adjuvant radio- Int J Radiat Oncol Biol Phys 1989; 17: 663–8.
therapy. Strahlenther Onkol 2004; 180: 15–20. 86. Jakobsen A, Mortensen JP, Bisgaard C, Lindebjerg J,
73. Marsh PH, James RD, Schofield PF. Adjuvant Hansen JW, Rafaelsen SR. Preoperative chemoradia-
preoperative radiotherapy for locally advanced rectal tion of locally advanced T3 rectal cancer combined
carcinoma: results of a prospective randomized trial. with an endorectal boost. Int J Radiat Oncol Biol Phys
Dis Colon Rectum 1994; 37: 1205–14. 2006; 64: 461–5.

HEBK001-C06_p87-102.indd 101 08/02/13 5:58 PM


102  Preoperative radiotherapy and chemoradiotherapy for rectal cancer

87. Corner C, Bryant L, Chapman C, Glynne-Jones R, 89. Lorenzen S, Schuster T, Porschen R, et al.
Hoskin PJ. High-dose-rate afterloading intraluminal Cetuximab plus cisplatin–5-fluorouracil versus
brachytherapy for advanced inoperable rectal carci- cisplatin–5-fluorouracil alone in first-line metastatic
noma. Brachytherapy 2010; 9: 66–70. squamous cell carcinoma of the esophagus: a
88. Bonner JA, Harari PM, Giralt J, et al. Radiotherapy randomized phase II study of the Arbeitsgemein-
plus cetuximab for squamous cell carcinoma of the schaft Internistische Onkologie. Ann Oncol 2009;
head and neck. N Engl J Med 2006; 354: 567–78. 20: 1667–73.

HEBK001-C06_p87-102.indd 102 08/02/13 5:58 PM


7
Total mesorectal excision for
rectal cancer
Brendan Moran

Introduction and requires a balance between the desired effect


on the tumour and the immediate and long-term
There have been several key advances in the opti- side effects associated with radiotherapy for rectal
mal management of rectal cancer, but none so cancer.
important as standardization and improvement The benefits of neoadjuvant therapy for locally
in the appropriate surgical procedure. In addition advanced tumours are now generally accepted, with
to surgical advances, neoadjuvant and adjuvant good evidence for both down-staging and down-
radiotherapy and chemoradiotherapy, preoperative sizing after optimal neoadjuvant therapy. Debate
imaging (by computed tomography (CT) of the persists as to what the term ‘locally advanced’
chest and abdomen looking for metastatic disease, means, and until recently the definition lacked
and by magnetic resonance imaging (MRI) to assess the objective fine detail of good-quality MRI. The
local tumour extent), pathological assessment and evidence for neoadjuvant therapy is strongest for
audit have all contributed to better results for a reduction in local recurrence in operable rec-
this complex but eminently curable cancer. These tal cancer and has been reported in a number of
advances, and complex management decisions, are randomized controlled trials such as the Swedish,
undoubtedly best coordinated by a multidiscipli- Stockholm, Dutch and UK CR07 trials. There is lit-
nary team approach, but surgery, and the surgeon, tle evidence of a survival benefit from radiation in
are the key to optimal decision-making and out- these trials, however, with well-documented reports
come. Multidisciplinary team management should of immediate and long-term side effects associated
be individualized to each patient with rectal cancer with radiotherapy. Unquestionably, tissue heal-
and should focus on helping the surgeon and the ing is impaired by radiotherapy, with an increase
patient choose the best course of action. The pre- in perineal wound failure in patients who have an
operative MRI provides optimal local staging and abdominoperineal excision and an increase in anas-
is a key visual component in the multidisciplinary tomotic leakage when reconstruction by anterior
team discussion of the optimal management of a resection is performed. Additionally, in the medi-
patient with rectal cancer. The combination of clin- um to long term, bladder, bowel and sexual func-
ical examination and good-quality MRI facilitates tion are impaired by radiotherapy. There are also
selection for neoadjuvant (preoperative) radiother- increasing reports of long-term complications such
apy or chemoradiotherapy. This decision is crucial as an increase in second primary tumours, excess

HEBK001-C07_p103-123.indd 103 11/02/13 3:51 PM


104  Total mesorectal excision for rectal cancer

cardiovascular deaths, and reoperation for bowel refining and advancing the complex anatomy of
obstruction, all of which emphasize the need for a the human pelvis (see Chapter 2).
selective approach. There is no doubt that patients
with advanced tumours benefit from radiotherapy
Fundamental principles of
and, if used in rectal cancer, radiotherapy should be
total mesorectal excision
given before surgery with randomized trials sup-
surgery
porting this strategy.
There is now general agreement that the opti-
The fundamental components of TME surgery
mal surgical treatment involves the concept of
incorporate a number of principles and include the
total mesorectal excision (TME), and that an R0
following:
excision (all margins of the excised specimen free
of tumour at detailed pathological assessment) l Peri-mesorectal ‘holy plane’ sharp dissection
is crucial. For this reason, the relationship of the by diathermy and scissors under direct ­v ision:
tumour to the mesorectal fascia is key in that a three-directional traction and counter-­
tumour invading or breaching the mesorectal traction is a vital principle for diathermy dis-
fascia will have an involved margin at TME sur- section, as it is essential that the areolar tissue
gery and will not be cured by TME alone. A key is ‘on stretch’ if holy plane dissection is to be
advance of staging MRI is the ability to visualize accurate.
the mesorectal fascia and to accurately determine l Specimen-oriented surgery and histopathol-

whether the mesorectal fascia is clear of tumour, ogy, with the objective of an intact mesorec-
definitely involved or possibly involved (the lat- tum with no tearing of the surface and no
ter often categorized as ‘threatened’) by tumour. circumferential resection margin (CRM)
This crucial information on the relationship of or distal margin involvement (naked eye
the tumour to the mesorectal fascia allows an or microscopic).
informed decision on whether TME alone is l Personal naked-eye assessment as audit for

likely to be curative (clear margins), indicates obvious CRM involvement as the principal
patients who definitely need neoadjuvant thera- immediate outcome measure: this should be
py (involved margin), and facilitates discussion combined with objective assessment of the
on the risks and benefits in patients with a threat- whole specimen by the pathologist, as this
ened margin. These categories are more difficult confirms the optimal planning and completion
to define in the low rectum (at or below the level of the surgery. Surgeons and oncologists may
of the levators), where the mesorectum tapers also base postoperative therapy on this pathol-
out and the margins are more likely to be threat- ogy report, although there must always be a
ened. For this reason, we have an ongoing inter- sense of the ‘horse having bolted’ if cancer has
est, research programme and educational initia- perforated through the resected specimen and
tive in the staging and management of low rectal radiotherapy or chemoradiotherapy (CRT) was
cancers (see www.lorec.nhs.uk). not given before surgery. An involved margin
The broad principles of TME surgery remain generally represents a failure of management
the same, whether the technique is open surgery, planning or surgical technique, or both.
laparoscopic TME or robotic TME. The principles l Recognition during surgery and preservation
of optimal visualization and more precise dis- of the autonomic plexuses and nerves on which
section, improved haemostasis and less collateral sexual and bladder function depend.
damage by diathermy are key. Modern advances l A major increase in anal preservation and reduc-
in technology have helped in these areas. Knowl- tion in the number of permanent colostomies by
edge of the anatomical structures, and their rela- skilful extension into the depths of the pelvis.
tionships in the pelvis, are essential for surgeons, l Stapled low pelvic reconstruction, usually using
regardless of the surgical technique used. It is the Moran triple stapling technique (described
noteworthy that surgical observations and atten- later), plus creation of a short colon pouch or a
tion to the fine detail of the anatomical structures side-to-end anastomosis to the low rectum or
at surgery have been a defining force in developing, anal canal.

HEBK001-C07_p103-123.indd 104 11/02/13 3:51 PM


BASIC PRINCIPLES OF SURGERY FOR RECTAL CANCER  105

BASIC PRINCIPLES OF SURGERY


FOR RECTAL CANCER

Rectal cancer, as defined by an adenocarcino-


ma with its lower edge at or within 15 cm of the
anal verge, is a common cancer and accounts for
approximately 30 per cent of all colorectal cancers.
Surgical excision by anterior resection is curative in
most cases. The term ‘anterior resection’ defines an
operation whereby the inferior mesenteric artery
has been ligated and reconstruction is by anasto-
mosis of the proximal colon to the distal remnant.
Anterior resection is neither required nor appropri-
ate for all patients with rectal cancer. Early tumours
(perhaps in the region of 5–10 per cent of all
patients with rectal cancer, although this percentage
may increase with screening) can be treated by local
excision alone in selected patients (see Chapter 11), Figure 7.1. Inferior mesenteric angiogram outlining
and tumours involving the anal sphincter complex the blood supply to the rectum and demonstrating the
or levator muscle (approximately 10–15 per cent) absence of any significant arterial input from the lateral
require excision of the sphincter complex with a pelvic side-wall.
permanent stoma (see Chapter 8). Thus, restora-
tive anterior resection is required and feasible in
the description and popularization of TME with
approximately 70–80 per cent of all patients with
specimen-oriented surgery. Finally, circumferential
rectal cancer. For each patient, however, the feasi-
pathology reports on the adequacy of excision mar-
bility and benefits of restorative resection will ulti-
gins on the resected specimen and allows continu-
mately depend on the tumour, the patient and, to a
ous quality control of the staging, MDT decisions
lesser extent, the surgeon. Tumour-dependent fac-
and surgical precision.
tors are predominantly the distance of the tumour
The principles of optimal surgery for rectal can-
from the anal verge, the fixity of the tumour, and the
cer revolve around the embryology and anatomy of
presence or absence of diffuse metastatic disease.
the rectum, whereby the lymphatic drainage (which
Patient-dependent factors include the body habitus,
is associated with the arterial blood supply; Figure
the size and depth of the pelvis, and anal sphincter
7.1) is almost exclusively proximal and is generally
integrity and function. Surgeon-dependent factors
confined within the mesorectal fascia.
include the availability of resources (e.g. stapling
The principles of TME focus on specimen-ori-
instruments, adequate retractors) and the availabil-
ented surgery, whereby completeness and intact-
ity of a suitably experienced surgical assistant and
ness of the specimen are crucial factors, such that
theatre team. The principles of anterior resection
ideally it should be one recognizable block of tis-
for rectal cancer currently revolve around what has
sue whose orientation and former relations can
been described as ‘circumferential awareness’ incor-
be identified. In most cases, naked-eye inspection
porating circumferential staging, circumferential
provides the initial necessary quality control. Visual
down-staging, circumferential surgery and circum-
inspection of the front of a well-performed TME
ferential pathology. Circumferential staging incor-
specimen should show three clear landmarks:
porates clinical examination and, more latterly, the
addition of cross-sectional imaging, in particular l the cut edge of the peritoneal reflection;
abdominal and pelvic CT and pelvic MRI. Circum- l the smooth shiny anterior surface of the ante-
ferential down-staging or down-sizing is achieved rior mesorectum of the middle third
by preoperative radiotherapy or chemoradiothera- (the rectogenital septum);
py (neoadjuvant therapy; see Chapter 6). The con- l the almost bare anterior aspect of the anorectal

cept of circumferential surgery has emanated from muscle tube in the lowest anterior resections.

HEBK001-C07_p103-123.indd 105 11/02/13 3:51 PM


106  Total mesorectal excision for rectal cancer

pling instruments,1 the recognition that adenocar-


cinoma rarely spreads distally in the muscle tube,
and the recognition that a 2 cm clearance beyond
the macroscopic tumour provides a safe distal mar-
gin and even less than 1 cm may be adequate for
ultra-low tumours.2
It is important to recognize that, even at its most
scientific, surgery is primarily a craft with very spe-
cial challenges and rewards for excellence. Surgery is
not amenable to study by the same methods as those
applicable to drugs or radiotherapy treatments, and
yet surgical technique has by far the greatest impact
on rectal cancer outcomes. The randomized con-
Figure 7.2. Total mesorectal excision specimen illustrating trolled clinical trial has so far contributed remark-
the shiny mesorectal fascia surrounding the fatty ably little to the development of surgical technique
mesorectum. A linear stapling gun occludes the lumen, in colorectal surgery or most other surgical special-
as used in the Moran triple stapling technique. ties. Even though variations in technique, such as
open, laparoscopic or robotic surgery, lend them-
selves to comparative studies and indeed to rand-
Laterally, the fatty mesorectum expands distally omization, in all such trials the cohort studied can
beyond an anteroposterior groove made by the be only a selected subset of the patient population,
nervi erigentes so that an embryologically per- with the most difficult patients excluded and oper-
fect specimen has a lateral dilatation distally, cor- ated on by open surgical techniques. For this rea-
responding with the part related to the inside of son, the results of such trials are not applicable to
the levator muscles beyond their origins from the all patients and merely represent comparative out-
pelvic side-wall (Figure 7.2). Posteriorly, a perfect comes, generally in the more favourable cases.
specimen exhibits perfectly curved ‘buttocks’ with The technique of TME has never been critically
a central midline groove corresponding to the ano- evaluated by a randomized trial, and nor is likely
coccygeal raphe. to be. In this difficult and complex surgery for rec-
Distal mesorectal spread of the tumour rarely tal cancer, the devil is in the detail, and attention
extends more than 2–3 cm beyond the lower pal- to detail using TME concepts results in optimal
pable luminal edge of the tumour, although for ­outcome.
safety reasons a distal mesorectal clearance of 5 cm,
where feasible and available, is recommended. This
was the fundamental principle underpinning the Preoperative Assessment, Planning
concept of TME, which has now become accepted and Preparation
as the optimal surgical procedure for patients with
resectable mid- and low rectal cancer. There has Preoperative preparation is essential, and to fail
been some confusion that we have advocated TME to plan is to plan to fail. The assessment of a rec-
for all rectal cancers; but TME is not necessary for tal cancer, local staging and management of a rec-
tumours of the upper rectum provided the mes- tal cancer are summarized in Figures 7.3 and 7.4,
orectum and muscle tube can be divided 5 cm distal which, although specific for low rectal cancer, apply
to the lower edge of the tumour and a mesorectal to any rectal adenocarcinoma.
transection, rather than TME, can be performed,
possibly diminishing the postoperative surgical Critical decisions in lower-third cancers
complications such as a reduction in the anasto- One focal area of current interest centres on the
motic leak rate. anatomical and embryological fact that the mes-
Other major advances facilitating safe restora- orectal envelope tapers down in this (infralevator)
tion of continuity, even in low tumours, were the lower third to appear very thin indeed, particu-
development and widespread availability of sta- larly on the crucial coronal oblique MRI cuts on

HEBK001-C07_p103-123.indd 106 11/02/13 3:51 PM


BASIC PRINCIPLES OF SURGERY FOR RECTAL CANCER  107

Suspicion of Rectal Cancer

History, patients perspective and Rectal Examination (PR) by


Experienced Surgeon + CNS ideally present

Too painful PR Rigid Sigmoidoscopy

Mobility
Distance in cms Distance in cms from
from anal verge dentate line
EUA to lower edge to lower edge
(Examination
Under Anaesthetic)
Colonoscopy

Biopsy

Surgeon and CNS

Squamous Other:
Adenocarcinoma Adenoma
Carcinoma Prostate/Carcinoid

Treatment Staging Repeat biopsy Treatment

Systemic Staging CT - Chest


and Abdomen

Disseminated Resectable
No metastases
disease metastases

Local Extent primary


(Figure 7.5)

Pallative treatment ± Chemotherapy ± Radiotherapy ± Local Ablation

CNS, clinical nurse specialist; CT, computed tomography.

Figure 7.3. Suggested algorithm for assessment of rectal cancer.

HEBK001-C07_p103-123.indd 107 11/02/13 3:51 PM


108  Total mesorectal excision for rectal cancer

Low Rectal Cancer

MRI - Low rectal protocol


PR by experienced surgeon EOUS (Endo-anal ultrasound)

MDT

‘Good’ ‘Ugly’
‘Bad’
(early tumour uT1, (Advanced uT3/T4, mr
(mrT3N1-2, EMVI)
mrT1) T3/T4)

SCRT/CRT? CRT

Restage

PR, MRI, EAUS Systemic (CT)

Tumour Clinical and radiological complete


present response
Consider
Deferral of surgery
‘watch and wait’
Recurrence
Surgery Intensive Follow-up

Intensive Follow-up 5–10 years


Local excision Anterior Resection (AR) Abdomino Perineal
perianal, TEM (Including Intersphincteric) Excision (APE)

Defunctioning Stoma Intersphinteric Extralevator ELAPE

Perineal closure
Primary Suture

Primary Suture Biological mesh Omentum Muscle flap

Back to MDT for post op discussion

CRT, chemoradiotherapy; CT, computed tomography; EAUS, endoanal ultrasound; MDT, multidisciplinary team;
MRI, magnetic resonance imaging; PR, per rectal examination; SCRT, short-course radiotherapy; TEM, transanal
endoscopic microsurgery.

Figure 7.4. Suggested algorithm for local assessment of low rectal cancer.

which decisions in modern multidisciplinary teams the tumour in the conscious patient (with muscle
are made. On such an MRI it is tempting to pre- tone). In the author’s opinion, this clinical observa-
dict that this tapering and narrowing area of the tion of mobility on the sphincter complex and adja-
mesorectum will constitute a hazardous margin; cent organs almost invariably means that a TME will
thus, a decision may be made to administer preop- be an achievable surgical objective (see Figure 7.5).
erative down-staging neoadjuvant therapy or even It does not get away from the other issues of the
choose abdominoperineal excision for fear of mar- higher incidence of internal iliac and particularly
gin involvement when in fact a carefully oriented obturator node involvement in tumours less than
axial oblique sequence with the axial cuts precisely 6 cm from the anal margin.3
at right-angles to the tumour segment may dem- A rectal neoplasm should be assessed by an expe-
onstrate a potentially safe clearance. In such cases, rienced clinician performing a rectal examination
where the cancer is below the levator origins on and rigid sigmoidoscopy. The height of the lower
MRI, it is essential that an experienced surgeon edge of the tumour should be measured by rigid
examines the patient to establish free mobility of sigmoidoscopy with the patient awake and recum-

HEBK001-C07_p103-123.indd 108 11/02/13 3:51 PM


BASIC PRINCIPLES OF SURGERY FOR RECTAL CANCER  109

pelvic anatomy, such as ovarian or uterine pathol-


ogy in female patients and prostatic enlargement
in male patients, all of which may limit access. In
some patients, synchronous treatment of ovarian
and rectal pathology may be performed.
The clinical details and radiological investiga-
tions of all patients with rectal cancer should ideally
be discussed at a colorectal multidisciplinary team
meeting to optimize the management strategy,
which in some cases includes neoadjuvant therapy.

Patient consent and immediate


preoperative care
Medical conditions such as diabetes, hyperten-
sion, and cardiac and pulmonary diseases should
be ­optimized. Arrangements should be in place for
postoperative critical care if required. The patient
Figure 7.5. Total mesorectal excision. Dissection follows
should be consented and sites marked in case a stoma
the dotted line. Transection of the muscle tube should be
3–5 cm below the luminal distal edge of the tumour as
is required. It is prudent to consent for a permanent
distal mesorectal spread has been documented so that stoma, in addition to a temporary defunctioning,
mesorectal transection may be adequate for upper rectal should an unexpected event render this necessary.
tumours. In addition to the complications after any intestinal
resection, such as leakage and haemorrhage, the pos-
bent in the left lateral position. The surgeon should sibility of sexual and bladder dysfunction needs to
assess the integrity of the anal canal and if possi- be documented in the consent form for rectal can-
ble assess the mobility, or otherwise, of a palpable cer surgery. These latter complications usually result
tumour. The neoplasm should be biopsied to con- from injury to the pelvic autonomic nerves.
firm the diagnosis. Biopsy may be deferred to a sub- In female patients, the possible need for
sequent urgent colonoscopy, however. oophorectomy should be discussed. Consent for
One useful procedure is to re-examine the lesion, oophorectomy should be documented in case
particularly a low rectal cancer under sedation at the tumour involves the ovaries or the patient
colonoscopy. Occasionally an examination and has a family history of ovarian cancer or requests
biopsy under general anaesthetic is required, and oophorectomy. A personal preference is also to seek
even today this may be a useful addition in the consent for appendicectomy to treat synchronous
work-up of a patient with a rectal cancer. or prevent metachronous appendiceal pathology.
The remainder of the colon should be assessed to Unlike current practice in surgery for colon can-
look for synchronous neoplasia (present in 3–4 per cer, an empty large intestine is desirable for restora-
cent of patients) by colonoscopy, CT colonography tive rectal cancer surgery, particularly if there is a
or barium enema. Colonoscopy has the advantage of need for temporary defunctioning. An ileostomy
allowing biopsy or removal of synchronous lesions proximal to a full colon may not reduce the con-
such as polyps. The benefits of CT colonography sequences of an anastomotic leak. Optimal bowel
are the ability to stage the abdominal cavity and to preparation can be achieved by combining clear
outline the proximal bowel in patients with stenotic fluids by mouth for 48 h before surgery with oral
lesions where colonoscopy may not be ­feasible. laxatives. There is emerging evidence that the
Current best practice also incorporates staging non-prepared colon increases complications after
for systemic disease by chest and abdominal CT scan ­restorative rectal cancer surgery, and many sur-
and local pelvic staging of the rectal tumour, ide- geons who abandoned bowel preparation on the
ally by MRI to assess the relationship of the tumour back of experiences with colonic resection are
to the mesorectal fascia (see Chapter 4 and 5). reverting back to full mechanical bowel preparation
Additionally, MRI can provide details of relevant for restorative rectal cancer surgery.

HEBK001-C07_p103-123.indd 109 11/02/13 3:51 PM


110  Total mesorectal excision for rectal cancer

Systemic antibacterial agents, including anaero-


bic cover such as with metronidazole, are given at
induction of anaesthesia and continued for 24 h
post-surgery.
Deep venous thrombosis (DVT) prophylaxis is
commenced by a combination of heparin or its
analogues (depending on the use of epidural anaes-
thesia). The risk of DVT must always be balanced
against the risk of bleeding, which is catastrophic
if into the dural space and potentially catastrophic
during pelvic dissection. For these reasons, antico-
agulation is best withheld in the immediate preop-
erative period. The use of mechanical calf com-
pression devices, once the patient is positioned
on the operating table, and in the postoperative
period, has been shown to be effective and gener-
ally safe.

Figure 7.6. Midline incision extending from the


Patient Positioning
symphysis pubis, which may need to be extended to the
xiphisternum. The optimal site for a defunctioning stoma is
The lithotomy–Trendelenburg position is opti- on the right side (defunctioning lower right ileostomy) and is
mal as it allows peranal palpation, inspection and marked preoperatively by an enterostomal therapist. A site
washout, together with insertion of the circular is also marked in the left iliac fossa in case a permanent
staple gun to complete the anastomosis. Addition- stoma is deemed necessary.
ally, a second assistant can stand between the legs.
The patient is kept horizontal during the abdomi-
nal phase of the operation and can then be tilted Although some advocate a long transverse inci-
head down by 15–208 or more to facilitate the pel- sion or even a modified extended Pfannensteil inci-
vic dissection. It is important not to maintain steep sion, access to the splenic flexure and pelvis is infe-
Trendelenburg positioning for extended periods, to rior compared with a midline incision.
reduce the risks of calf compartment syndrome. The abdominal cavity is fully palpated, with par-
Good lighting is essential. Optimal lighting can ticular attention directed to the liver and spleen,
be obtained by readjusting the movable focused greater omentum, stomach and small bowel, and
operating lights during different phases, using a the entire colorectum, including the appendix. The
headlight (which many surgeons find irksome for surgical procedure is then planned, including the
a prolonged procedure), and using retractors with sequence. For example, if a low anterior resec-
integrated lights. tion is planned mobilization of the splenic flexure
is almost always needed. Personal preference is to
complete this manoeuvre at the beginning while
On-Table Examination, Skin Incision still fresh and to avoid the temptation to omit this at
and Abdominal Exploration the end of a long procedure and thus compromise
on tension and blood supply to the neorectum.
A rectal examination before painting and draping is
mandatory and must be supplemented by a vaginal
examination in female patients. Commencing the Dissection and
A long vertical midline incision provides optimal Splenic Flexure Mobilization
access to the abdomen and pelvis and may need to be
extended from the pubic symphysis to the xiphister- The operating surgeon stands on the left side of the
num in patients who are overweight (Figure 7.6). patient. The assistant on the right lifts the sigmoid

HEBK001-C07_p103-123.indd 110 11/02/13 3:51 PM


BASIC PRINCIPLES OF SURGERY FOR RECTAL CANCER  111

colon anteriorly and to the patient’s right. The peri- Ligation and division of the inferior
toneal reflection on the left side of the colon (the mesenteric vessels
white line of Toldt) is identified and divided by scis- The left-sided colonic mobilization is continued
sors, or more commonly diathermy, and followed inferiorly by the left-sided operator identifying the
cranially towards the splenic flexure. The plane in ureter (usually positioned medial to the gonadal
the left upper quadrant between the colon and the vessels and crossing the bifurcation of the common
urogenital structures (Gerotas fascia surrounding iliac artery) and the fascial covering of the upper-
the kidney and the gonadal vessels) is developed. most part of the mesorectal package. This manoeu-
At this juncture, if the spleen is mobile on the dia- vre is facilitated by the right-sided assistant apply-
phragm, a large moist swab placed gently between ing traction on the sigmoid, anteriorly and to the
the spleen and diaphragm helps to push the spleen right, taking care not to damage the mesentery of
into view and facilitates splenic flexure mobiliza- the colon. Once the plane has been developed at the
tion (Figure 7.7). The greater omentum is now pelvic brim to just beyond the midline, a small swab
retracted anteriorly and to the patient’s left, and the is pushed behind the colon and rectal mesentery at
bloodless plane between the transverse colon and the level of the pelvic brim. The sigmoid traction is
omentum is developed by sharp scissors dissec- reversed and the surgeon on the patient’s right can
tion or diathermy incision. The apex of the splenic identify the exact place to incise the right-sided per-
flexure attachments are visualized by downwards itoneum by a combination of air in the tissues and
colonic traction from the right side of the patient displacement of the mesentery anteriorly. The swab
with counter-traction by a retractor under the left helps to protect the autonomic nerves at the level of
ribcage. The assistant on the patient’s right, in addi- the pelvic brim by displacing the colonic mesentery
tion to colonic traction, insinuates a finger behind anteriorly. The right-sided peritoneum is incised
the colon on the left. Division of the apical lateral caudally to the pelvic brim and cranially towards
colonic attachments is performed by an operator the root of the inferior mesenteric artery. At this
who stands either on the patient’s left or temporar- point, the surgeon on the patient’s left places the
ily between the patient’s legs. left index finger behind the pedicle, with left thumb
anteriorly, palpating the vessel between index finger
and thumb. The peritoneal attachments are divided
and pre-aortic nerve structures mobilized away
from the right side of the pedicle by sharp dissec-
tion. The index finger is then advanced cranially on
the left side, parallel to the midline, where a window
will be identified above the origin of the inferior
mesenteric artery (IMA) between the aorta and the
inferior mesenteric vein (IMV) and ascending left
colic artery running side by side at this point. This
window is opened, and the autonomic nerves are
freed until the root of the IMA is clearly identified.
It is important to check that the left ureter has not
been elevated in this manoeuvre by visualizing the
structures to the left of the pedicle. Once the IMA
pedicle has been isolated, it is clamped, divided and
ligated approximately 2 cm from the aorta to reduce
injury to the pre-aortic nerves and to achieve a high
but not flush tie of the IMA (Figure 7.8).
For maximum length and mobility of the left
colon, the IMV has to be divided above its last
branch, at the inferior border of the pancreas, where
Figure 7.7. Mobilization of the splenic flexure, incising it disappears upwards to join the splenic vein. In
lateral attachments. 5–10 per cent of patients, a substantial branch of

HEBK001-C07_p103-123.indd 111 11/02/13 3:51 PM


112  Total mesorectal excision for rectal cancer

Figure 7.9. The colon has been divided using a linear


cutting stapler and the posterior plane is being developed
Figure 7.8. Ligation of the inferior mesenteric artery (large anterior to the superior hypogastric plexus and hypogastric
arrow) and high ligation of the inferior mesenteric vein (presacral) nerves.
(small arrow).

the superior mesenteric artery lies near the IMV gentle opening of the perimesorectal planes by trac-
at this point, supplying part of the arterial blood tion and counter-traction in any direction through-
supply to the left colon. Judgement is required to out the pelvic dissection. Having divided the colon,
determine whether this vessel should be divided to the small bowel and right transverse colon can be
provide sufficient length or preserved if it is likely to packed away, upwards and to the right. Personal
be important for colonic viability. preference is to place a large pack under the root
of the caecum wrapping the small bowel and place
a gauze roll beneath the root of the mesentery. An
MOBILIZATION OF THE extendable self-retaining retractor (we generally use
MESORECTUM AND RECTUM a Finochetti chest retractor with the largest retrac-
tor blades) then opens up the space and allows
The pelvic dissection is oncologically one of the excellent views into the top of the pelvis.
most important stages of the operation. The sur-
geon must develop a mental picture of the exact
position and extent of the tumour, based on the Posterior Dissection – Starting ‘Right’
prior clinical and radiological assessment. The cir-
cumferential concepts of TME surgery are applied The pelvic dissection for a rectal cancer is a dynam-
to ensure clear margins on the resected specimen. ic procedure and involves circumferential disection
It is helpful to divide the descending colon well commencing posteriorly, continuing laterally and
above the cancer at this stage, a so-called ‘division anterolaterally and then anteriorly, and constantly
of convenience’ using a linear cutting stapler such moving the site of dissection rather than proceed-
as a GIA 60 (Figure 7.9). This particularly facilitates ing too far at one site. The easiest place is commonly
the posterior pelvic dissection. posteriorly, and commencement in this area is cru-
The ‘division of convenience’ allows optimal cial. For clarity the dissection sites are covered sepa-
mobility of the top of the specimen and facilitates rately, although in reality it is a dynamic procedure

HEBK001-C07_p103-123.indd 112 11/02/13 3:51 PM


MOBILIZATION OF THE MESORECTUM AND RECTUM  113

requiring alteration in the placement and direction operator standing on the patient’s left and the first
of traction of the retractors. The ‘pedicle package’ – assistant on the patient’s right have to position and
the clue to the top of the ‘holy plane’ – involves the control the angulation and retraction force, aided by
key principle that dissection should proceed only the second assistant between the patient’s legs when
in the areolar tissue plane (the ‘holy plane’) within more forceful retraction is needed. It is important
(and thus sparing) the autonomic nerve plexuses, to note that all four hands of the operator and first
the non-visceral presacral fat pad (when present), assistant are needed for retraction and dissection,
the parietal side-wall fascia of the small pelvis, the and a suction device may be a useful retractor, in
hypogastric plexus, the vesicles, and the prostate in addition to its role in removing smoke and fluid. It
male patients and the vagina in female patients. All is really only the operator and first assistant who can
of the dissection should be performed sharp with clearly determine the correct angulation and direc-
diathermy or scissors under direct vision with good tion of the retractors. The second assistant’s role is
light. Throughout, the dedicated assistants should predominantly supportive and to add force rather
provide three-directional traction to open up the than direction to the traction and counter-traction.
planes for the operator: diathermy can be used It is useful to wash out the pelvis on a regular basis;
safely only when the areolar tissue is on stretch. The personal preference is to use water with dilute liq-
overall procedure is lengthy and time-consuming uid proflavine (an antiseptic and cytocidal agent). In
and requires prolonged attention to detail, as a this context, water rather than normal saline is opti-
careful TME plus pouch-to-anus reconstruction mal, as it is hypotonic and therefore cytocidal. Addi-
takes 3–5 h according to the detail of the patient’s tionally, the use of a clear solution helps to visualize
build. the tissue planes, unlike equally effective agents such
Starting correctly involves three-directional trac- as povidone–iodine.
tion on the colon and retroperitoneum to identify Dissection proceeds in the ‘angel hair’ and should
the plane between the back of the pedicle package be predominantly from below upwards, allowing
and the gonadal vessels, ureter and pre-aortic sym- the hypogastric nerves to drop away posterolateral-
pathetic nerves, all of which must be carefully pre- ly. It is important to focus on circumferential mobi-
served. The key to this phase is the recognition of lization rather than proceed too far posteriorly at
the shiny fascial-covered surface of the back of the this stage. Some dissection in the lateral and ante-
pedicle – like a tapering longitudinal sausage with rior plane is recommended at this juncture.
the inferior mesenteric vessels within. This must be
gently lifted forwards.
The avascular areolar tissue plane (mesorectal fas- Lateral Dissection
cia) that surrounds the mesorectum is identified. It
is worth remembering that the mesorectum resem- The lateral attachments are mobilized by extend-
bles a bilobed lipoma. The rectum is lifted gently ing the dissection plane forwards from the midline
forwards from the bifurcation of the hypogastric posteriorly around the side-walls of the pelvis. It is
nerves, and dissection commences in the midline important to remember that the inferior hypogas-
using diathermy, aiming to minimize direct or tric plexuses (formed by the hypogastric nerves and
collateral heat damage to the nerves. Dissection is the pelvic parasympathetic nerves) curve forwards
extended downwards anterior to the curve of the tangentially around the surface of the mesorectum
sacrum on the surface of the mesorectal fascia. Once in close proximity to it. The nervi erigentes (pel-
there is sufficient space, a St Marks rectal retractor vic parasympathetic nerves on which male erection
(one with integral illumination, if available, is opti- depends) lie more posteriorly in the same plane as
mal) is introduced behind the specimen. This helps the hypogastric nerves and should be visualized and
to spread and ‘tent’ the hypogastric nerves and aids preserved, although it is all too easy to ‘tent up’ the
identification. It is important to gently position nerves and cut them at this point.
the retractor and apply firm but gentle pressure to The nervi erigentes curve forwards from the sacral
expose the mesorectal fascia and the layer of are- foramina and converge like a fan to join the hypogas-
olar tissue (sometimes called ‘angel hairs’) where tric nerves and form the neurovascular bundles of
dissection should proceed. In this manoeuvre, the Walsh,4 as elegantly illustrated in Figure 7.10, adapted

HEBK001-C07_p103-123.indd 113 11/02/13 3:51 PM


114  Total mesorectal excision for rectal cancer

Dissection Anteriorly

The traditional approach in low anterior resection in


prostate male patients was to incise the peritoneal reflection
anteriorly, but a more satisfactory approach is to fol-
low the plane forwards, from behind, anterolaterally
neurovascular vesicle on both sides until the vesicles are visualized.
bundle A small swab may be placed on the anterior sur-
face of the specimen, and the plane immediately in
Denonvilliers’
denonvilliers’ front of Denonvilliers’ fascia is developed by sharp
fascia dissection in the midline anteriorly and then care-
fully extended laterally to meet the lateral dissection,
remembering that autonomic nerves converge to
form the neurovascular bundles at the outer edge of
Denonvilliers’ fascia (see Figure 7.11). Denonvilliers’
fascia marks the anterior extent of the ‘tumour pack-
age’ and lies like an apron anterior to the anterior
mesorectum, behind the vesicles, until it fuses pos-
teriorly with the posterior fascia of the prostate. For
this reason, Denonvilliers’ fascia must eventually be
the ‘holy plane’ divided by scissors or diathermy to access the lowest
Figure 7.10. Schematic outline of the mesorectum few centimetres of the anterior rectum. This should
and mesorectal fascia with the neurovascular bundles
anterolaterally in the male pelvis.

from Walsh’s approach to radical prostatic resection


for prostate cancer.
Thus, the nerves lie at the outer edges of Denon-
villiers’ fascia and are in danger at the 10 o’clock
Denonvilliers’
and 2 o’clock anterolateral positions, just behind fascia
the lateral edges of the seminal vesicles in the male.
More distally, they curve forwards out of danger.
As the lateral dissection moves deeper into the
pelvis, one or two middle rectal vessels may be
encountered and occasionally may have to be oc-
cluded by precise diathermy or ligation after appli-
cation of a slender curved artery forceps. There are
almost always some slender nerve branches at this
point, and it is usually these branches that form the
so-called ‘lateral ligament’. When medial traction
is applied, these branches will ‘tent’ the plexus; it
is important to divide them by sharp diathermy or
scissors dissection on the mesorectal surface. The
previously described clamping of the lateral liga-
ments is unnecessary and potentially injurious to
the pelvic nerves.
If bleeding is encountered, it is often wise to
place a gentle pack (personal preference is to use an Figure 7.11. Schematic view with rectum posteriorly. It is
adrenaline-soaked swab) and move the dissection the author’s preference to dissect along the dotted line
to another area, perhaps the other side or anteriorly. anterior to Denonvilliers’ fascia.

HEBK001-C07_p103-123.indd 114 11/02/13 3:51 PM


MOBILIZATION OF THE MESORECTUM AND RECTUM  115

the ‘rectogenital septum’ (the female equivalent of


the male Denonvilliers’ fascia). It is sometimes help-
ful to place a narrow retractor (e.g. narrow Kelly
retractor) or a moist ‘swab-on-a-stick’ in the vagina
to help identify the posterior fornix of the vagina
and find this plane.

Denonvilliers’ Fascia and the Anterior


Part of a Low Anterior Resection
There is ongoing controversy concerning the exact
origin of Denonvilliers’ fascia and some debate as to
whether it exists in female patients. Most now agree
that the term ‘rectogenital septum’ is more compre-
hensive and encompasses Denonvilliers’ fascia, and
Figure 7.12. Total mesorectal excision in female patients. that the most likely origin is two layers of perito-
neum partly or fully fused together and obliterating
what was once a peritoneal cavity extending to the
be well beyond the distal edge of the cancer, except in pelvic floor. In anterior resection, one has to divide
ultra-low resection of a distal rectal cancer. this layer from above to enter the plane between
the rectum and the prostate in male patients, as
Anterior dissection in female patients the fascia fuses with the back of the prostate. The
Anterior dissection in female patients is gener- upper part of the septum is usually adherent to
ally more straightforward, provided the uterus has the anterior mesorectal fat of the middle third. In
been lifted well forward (Figure 7.12). One use- female patients the middle third has a rather thin
ful manoeuvre is to place a strong figure-of-eight and tenuous fatty layer between the rectum and
suture through the apex of the uterus and suture vagina, with the fascia often being scant and dif-
this to the lower end of the wound, which helps ficult to identify. The ‘rectogenital septum’ in male
considerably in uterine retraction. There is a con- patients is commonly a substantial rectangular or
densation of fibrous tissue anteriorly, analogous to trapezoidal layer, like a bib between the hindgut
Denonvilliers’ fascia in the male but almost always a behind and the vesicles and prostate in front. Just
much more tenuous structure. as peritoneum is the integral surface of the ante-
It is often difficult to find a precise avascular rior aspect of the upper, intraperitoneal third of the
plane behind the cervix and posterior fornix with- rectum, so the rectogenital septum ­(Denonvilliers’
out encountering bleeding from the venous plex- fascia) is integral to the fatty anterior mesorectum
us. Often the peritoneal reflection may adhere to in the middle third in male patients.
the posterior fornix. As in male patients, the plane
may be best approached by continuation of the
anterolateral dissection from the side-wall, thus Anterolateral Dissection
finding the relatively avascular plane between the
rectum and vagina. If bleeding occurs from the Connecting the lateral to the anterior plane is a
vagina, attempts to control it may be futile until critical point in the dissection. It is usually best
the vagina is fully mobilized off the anterior rec- to continue the dissection from posterior to ante-
tum, allowing the stretched venous plexus to col- rior, as the autonomic nerves will usually be visible
lapse down. and the correct plane is just medial to the auto-
Additionally, provided the cancer resection per- nomic bundles. There is a tendency to stray too far
mits, the peritoneal incision may be made at the level ­laterally with attendant risks of injury to the auto-
of the posterior fornix to facilitate entering into the nomic nerves or troublesome bleeding from the
plane between the vagina and what is best termed lateral pelvic side-wall vessels. Careful assessment

HEBK001-C07_p103-123.indd 115 11/02/13 3:51 PM


116  Total mesorectal excision for rectal cancer

and reassessment is essential at this point to access Lateral Pelvic Dissection


the correct operative plane. The anterolateral
peritoneal and subperitoneal incisions are curved This involves forward extension of the plane from
medially towards the midline anteriorly to preserve the back around to the sides, gently easing the
the autonomic nerve structures. adherent hypogastric nerves laterally off the mes-
orectal surface under direct vision. The freedom to
lift the divided rectosigmoid forward often means
Deep Posterior Dissection that the tangentially running hypogastric nerves are
first positively identified at this stage, the superior
Posteriorly and posterolaterally, the areolar plane hypogastric plexus itself only becoming obvious
is well defined around the globular expanding proximal to the nerves after they have been dissect-
bilobed mesorectum. A condensation of the fascia ed away from the mesorectal surface on each side.
called the rectosacral ligament (or Waldeyer’s fas- The superior hypogastric plexus may have been
cia) often presents a barrier to the surgeon poste- ensheathed by fatty tissue and not immediately rec-
riorly below the promontory. Just in front of the ognized as a nerve bifurcation.
rectosacral fascia (also referred to as the rectosac-
ral ligament), within the mesorectum, the superior
rectal vessels can often be seen through the back of The ‘Lateral Ligament’ Area
the mesorectal fascia, and around them cancerous
nodes are likely to be present, often only millime- The so-called ‘lateral ligaments’ are now generally
tres away. An intact shiny visceral fascia over these acknowledged not to be true ligaments and appear
must be jealously guarded and tearing avoided at to be attachments between the rectum and pelvic
all costs. This poses one of the greatest dangers of side-wall by small nerves going directly from the pel-
blunt manual extraction or of any haste or rough- vic plexus to the rectum, often accompanied by tiny
ness, since the rectosacral ligament may be stronger arteries and veins. This particular point of adherence
than the surface fascia over the nodes. Thus, tear- in the distal anterolateral sector is one of the most
ing into the lymphatic field by the inserted hand complex and difficult areas in a TME operation.
becomes a real risk and probably occurred often The area is approached by following the ‘holy
in the past. Sharp dissection under direct vision is plane’ down towards the vesicles in male patients,
crucial and good lighting is essential. Beyond this with the expanding plexiform band of inferior
area of attachment, the plane is easy to recognize, hypogastric plexus outside it but increasingly adher-
except that the forward angulation demands strong ent to it. In essence, there is no actual ligament, but
anterodistal retraction to facilitate direct visualiza- there is an area of adherence between mesorectum
tion. Understanding the importance of this forward medially and plexus laterally: small branches of
angulation is critical to mastering the traction and nerves and vessels penetrate through at this point,
counter-traction necessary in open, laparoscopic but none generally reaches more than 1–2 mm in
and robotic TME. diameter. The key nerves entering this flattened
A further reason to positively identify the ‘holy band from above are largely sympathetic hypogas-
plane’ posteriorly, in front of the presacral fat pad tric nerves curving distally from the superior plex-
(when present), is to avoid the risk of tearing thin- uses and more distally the erigent parasympathetic
walled presacral veins, which often have no valves nerves coming forwards to it from behind. These
and can bleed prodigiously when cut or torn. Inju- arise from the front of the roots of the sacral plexus
ry to these veins is much less likely if the correct (especially S3, out of sight behind the parietal side-
plane is followed and tearing is avoided; if they wall fascia). This fascia is quite robust laterally, and
are torn, a small pack and a considerable period of the surgeon will note that they usually cannot even
anterior dissection away from them will provide see the internal iliac vessels, which are outside and
the safest way forward. The key to this dissection positioned laterally. Posteriorly, these erigent pillars
is always to remain on the yellow mesorectal fascia from the nerve roots around S3 curve forwards out-
and to ­display the dissection plane by traction and side the parietal fascia, but medial to the branches
­counter-traction. of the internal iliac vessels.

HEBK001-C07_p103-123.indd 116 11/02/13 3:51 PM


EXTENDED RESECTION IN SPECIAL CASES   117

A little way behind the vesicles in male patients, male patients, as one works distally, there comes a
the erigent pillars pierce the fascia to join the infe- point where the fascia must be divided transversely,
rior hypogastric plexus and often contribute nerve as it becomes adherent to the posterior capsule of
branches to the mesorectum and rectum. These the prostate. Particular care is necessary during this
neural T-junctions are the nearest structures to ‘lat- step to avoid damage to the neurovascular bundles
eral ligaments’ that the most careful surgeon will (of Walsh) that constitute the distal condensation of
find with precise dissection. On occasions, there the inferior hypogastric plexuses, joined by numerous
is a true middle rectal artery of substantial size veins and small arteries, hence the title ‘neurovascular’.
at this point, but this is much less frequent than In a low anterior tumour in male patients, this
reported and may be no more than in the region can be critical: it is essential to avoid exposing
of 20 per cent or so, and then only on one side. We malignant tissue on the front of the specimen at
believe that in the past, what the surgeon thought the very point where the nerves are curving acutely
to be a middle rectal artery was most often a lat- medially. Since they are often impossible to see in
eral intramesorectal artery, and the so-called ‘stalk’ open surgery because of forward angulation behind
being divided represented a ‘coning in’ to the mes- the vesicles, bladder and prostate, they are in partic-
orectum. Alternatively, dissection too laterally will ular danger. In addition, surgeons who ‘take a slice
result in damage to a pelvic side-wall vessel. off the back of the prostate’ are highly likely to cause
Careful dissection between the mesorectum and impotence because of the close relationship of the
the inferior hypogastric plexus may result in bleed- neurovascular bundles to the back of the prostate.
ing but usually nothing more than a tiny vessel that Hand in hand with this dissection anteriorly goes
requires no more than a touch of diathermy. Thus, the development of the lateral side-wall dissection.
the clamp and cut routine generally implied a poor The parasympathetic erigent nerves form poster-
quality of dissection and damaged nerves, and left oanterior lateral pillars on the pelvic side-wall. ­Cadaver
a substantial residue of dangerous tissue – probably dissections have led us all to be taught that the pelvic
all a part of the 30–40 per cent local recurrence rate parasympathetic outflow is tripartite S2–3–4, but
that was once common. to the surgeon there is no doubt that a recognizable
A more distal vessel from the prostatic branch in landmark is often a single or bifid pillar comprising
male patients or from the pelvic floor is often found a nerve root arising from the front of the S3 compo-
later, and lower down, where it can cause troublesome nent of the main sacral plexus, which is out of sight
bleeding. It is likely that the minimal nature of a true posteriorly. The pillar-like appearance is due in part
middle rectal blood supply also implies a minimal to the forcible forward traction on the prostate, vagi-
lymphatic component, accounting for the relatively na and bladder to see the structures during an open
few patients with spread to the pelvic side-wall nodes.3 operation; this tends to bow the nerves medially and
With regard to the so-called ‘lateral ligament’, thus make them stand out. This retraction does not
the surgeon should generally attempt to dissect at occur to the same extent in a laparoscopic operation,
this point of adherence precisely between the out- which may account for some of the reported higher
er aspect of the mesorectum and the triangulated incidence of nerve damage at laparoscopic rectal can-
neural band of nerve plexus, which should be left cer surgery. These pillars and the hypogastric plex-
intact. This technique can be dubbed ‘mesorectal uses curve medially towards the back of the prostate
fat surface dissection’, because no actual loose are- in male patients, where they form the neurovascular
olar tissue exists in those areas where mesorectum bundles, which taper towards the urethra at the apex
and plexus are adherent. The final specimen will of the prostate. Here they become the erectile nerves
often lack the shiny fascial covering over this area. of the corpora cavernosa or cavernous nerves.

Deep Anterior Dissection EXTENDED RESECTION


IN SPECIAL CASES
As outlined above, the key structure anteriorly is
Denonvilliers’ fascia in male patients and the rec- With current optimal preoperative staging, it is
togenital septum equivalent in female patients. In uncommon to unexpectedly find a rectal cancer at

HEBK001-C07_p103-123.indd 117 11/02/13 3:51 PM


118  Total mesorectal excision for rectal cancer

operation that extends into adjacent organs. It is advice and assistance of an experienced urologist
pertinent to be aware that although attachment to should be sought.
or invasion of an adjacent organ may be an inflam-
matory adhesion, in approximately half of patients
cancer invasion is present. Rupture of a malignant Ureters
adhesion will almost certainly result in tumour
spillage and dissemination or local recurrence. The ureters may be involved in rectosigmoid or
Consequently, if feasible, it is prudent to resect an colonic cancers but are seldom involved in tumour
adjacent, adherent structure rather than gamble on of the mid- or low rectum, unless the tumour is very
the adhesion being benign. advanced. In all complex pelvic surgery, however, it is
always prudent to visualize, and in most cases mobi-
lize, the ureters. It is safe to divide the tissues anterior
Uterus and Vagina to the ureteric tunnels in male and female patients as
the ureters are anterior to the pelvic plexuses and are
Involvement of the vagina and uterus is usu- crossed only by the vas deferens in males and the uter-
ally detected at preoperative imaging and vaginal ine vessels in females. A locally advanced tumour may
examination before surgery. A large fixed cancer, invade the distal ureter and require en-bloc resection.
even with neoadjuvant chemoradiotherapy, is best Depending on the extent of the ureteric defect and
removed by en-bloc resection of the uterus and rec- the height of the ureteric resection, it may be possible
tum and as much as is needed of the posterior vagi- to perform a scalloped end-to-end anastomosis over
nal wall to clear the tumour safely. The vagina may a ureteric stent or to reimplant the proximal end into
be closed primarily in most such cases, but if the the bladder or the opposite ureter. Again, urological
defect is large, particularly in a sexually active wom- assistance is strongly recommended.
an, reconstruction using a musculocutaneous flap
may be needed. The need for this should have been
anticipated, and appropriate assistance (perhaps by Bladder
a plastic surgeon) and resources should be available.
Rectal cancer involving the bladder is usually pre-
dicted by preoperative pelvic MRI, and neoadju-
Seminal Vesicles vant chemoradiotherapy is generally indicated.
Cystoscopy is recommended as part of the work-up
Involvement of the vesicles on one or both sides to try to determine the site of involvement relative
may be managed by dissection anterior to the to the ureters. The operative strategy for rectal can-
vesicles, removing them en bloc with the rectum. cer involving the bladder requires consultation with
The ureters are at risk and should be identified a urologist and varies from partial cystectomy, to
and preserved, and it may be prudent to consider excising a disc of the involved bladder en bloc with
preoperative ureteric stenting to facilitate identifi- the rectum (this will almost always be an upper
cation. Additionally, the neurovascular bundles are rectal cancer in this scenario), to total cystectomy
at particular risk, and problems with urinary and by pelvic exenteration, particularly if the trigone is
sexual dysfunction may ensue. involved. Bladder involvement is much more com-
mon in male patients. In female patients, the uterus
and vagina intervene between the rectum and blad-
Prostate der, acting as a barrier to bladder invasion.

It is possible, although technically difficult, to


remove a part of the prostate involved by a rectal Inferior Hypogastric and
cancer. Major prostatic involvement may require Pelvic Plexuses
pelvic exenteration, however, or, in selected patients,
a nerve-preserving prostatectomy. Modern MRI Even though the focus in TME surgery is on iden-
imaging should predict this eventuality, and the tification and preservation of these nerves, a locally

HEBK001-C07_p103-123.indd 118 11/02/13 3:51 PM


DISSECTION OF THE MOST DISTAL MESORECTUM   119

advanced tumour, adherent to the pelvic side-wall, is familiar to proctologists from below – a tube of red
may require nerve resection. The perivascular plane skeletal muscle outside a tube of whiter smooth mus-
outside the nerves along the aorta and major ves- cle within (the internal sphincter).
sels may be developed and followed. Depending on
the site and extent of the tumour, it may be possible
to limit dissection and nerve resection to one side. Partial Mesorectal Excision (High
Bladder and sexual function is likely to be impaired, Anterior Resection and Mesorectal
depending on the particular nerves excised. Transection)
Tumours of the upper rectum (lower edge 11–15 cm
Pelvic Side-Wall Nodal Involvement from the anal verge) may not require TME and may
be optimally managed by mesorectal transection
There is a wide variation in the reported incidence 5 cm below the lower edge of the tumour. The char-
and clinical significance of pelvic side-wall involve- acterization of rectal cancer into high, mid- and low
ment. Increasingly, there is general agreement that has traditionally been measured from the anal verge
lateral nodal involvement is associated mainly with in conscious patients using a rigid sigmoidoscope.
low rectal cancer, is indicative of tumours with a It is worth documenting the height at MRI; indeed,
worse prognosis, and can often be predicted by pel- after much debate, a new definition of low rectal
vic imaging.3 The details are outlined in Chapter 14. cancer is emerging as a tumour with its distal mar-
gin at or below the level of origin of the levators on
the pelvic side-wall (see www.lorec.nhs.uk).
DISSECTION OF THE MOST Rectal cancer heights measured under anaes-
DISTAL MESORECTUM thesia need to be from the dentate line, since the
external sphincter dilates and retracts (add about
The anatomy of the position of the mesorectal 1.5 cm). Care is necessary: any instrument can push
package in relationship to the pelvic floor becomes a mobile tumour upwards, and flexible instru-
difficult for the surgeon to grasp from above ments often give falsely high measurements. With
because of its inaccessibility behind the vesicles and these qualifications, it can be said that, in our view,
prostate in male patients and behind the vagina in most cancers with the lower edge at or below 12 cm
female patients. The situation is complicated fur- should have TME as the key component of a radi-
ther by the fact that the levators are like a funnel in cal cancer operation because the mesorectum is the
continuity with the tube of the external sphincter primary field of lymphovascular spread. The deci-
distally. Conceptually, and because of the distor- sion as to whether the smaller partial mesorectal
tion introduced by upward traction, surgeons tend excision is adequate, however, is confirmed after the
to think of the pelvic floor as being much flatter mobilization has been completed to the point where
than it is in vivo, especially if an assistant applies the mesorectum must be liberated from the adher-
upward pressure on the perineum. If one doubts ent inferior hypogastric plexus – the region usually
this, a careful look at the layers on a coronal MRI referred to as the ‘lateral ligament’. It has long been
scan will make it evident. A clear three-dimension- convention and a very sound rule, borrowed from
al perception of the now globular bilobed mesorec- ­German surgical practice, that a minimum of 5 cm
tum in the depth of the pelvis and the surrounding of mesentery should always be excised both proxi-
neural lamella is the most elusive and challenging mal and distal to any colorectal cancer. Although
conceptual acquisition for the aspiring rectal can- muscle tube margin may safely be reduced to 1 cm
cer surgeon. in the interest of anal conservation, we have always
Careful pursuit of the plane at this level eventually believed that if less than a TME is contemplated, a
liberates the mesorectal package and takes the opera- minimum of 5 cm of mesorectum distal to the low-
tor down to a clean muscle tube. Although crossed er edge of the cancer must be dissected in the per-
by a few small arteries and veins from the puborec- imesorectal (‘holy’) plane. If, therefore, after initial
tal sling and some slips of sphincter muscle, the ‘holy mobilization there is a clear 5 cm of mesorectum,
plane’ here becomes the intersphincteric plane, which then tapering into the mesentery, in the interest of

HEBK001-C07_p103-123.indd 119 11/02/13 3:51 PM


120  Total mesorectal excision for rectal cancer

making a more minor operation and a higher anas-


tomosis, becomes acceptable. An anastomosis in a
patient who has mesorectal transection, provided it
is airtight, is less likely to leak than after a TME. In
this case, selected patients may avoid a temporary
stoma.

Management of the Anorectum Distal


to the Cancer: Stapling Techniques,
Distal Washout and Anastomosis
In more than 90 per cent of rectal cancers, it is tech-
nically feasible, although not necessarily optimal in
terms of future function, to extend the dissection
down to a clean muscle tube where a cross-clamp
may be applied with a finger-and-thumb clearance
beyond the lowest edge of the cancer. This is a dif-
ficult and challenging moment requiring both skill
and experience. We have developed a preference for
the use of the linear stapler in place of the right- Figure 7.13. Schematic drawing of a linear stapler applied
angled clamp (the Moran triple stapling technique). across the anorectal tube below the cancer.
The first TA-45, TA-30 (Covidien) staple line seals
the muscle tube (Figure 7.13) so that the anorectal This is usually, although not invariably, the micro-
lumen beyond can be washed out with water or a scopic edge. Downward spread along the muscle
tumoricidal solution (Figure 7.14). tube is not a significant factor in recurrence: a 2 cm
For very low tumours we have modified the tech- clearance is more than adequate, and 1 cm plus the
nique and now leave the first TA-30 closed and fired ‘doughnut’ is acceptable when the tumour is very
and in place, washout below it and place a second low and survival of the anal sphincters depends on
TA-30 below the first, across the washed muscle such a narrow margin.
tube (Figure 7.15). The bowel is sectioned with a
scalpel between the two TA-30 guns.
A proctoscope is introduced into the anal canal,
and the lumen below the staple line is irrigated with
repeated infusions using a 50 mL bladder syringe
or through a catheter irrigation system. Water (not
saline), povidone–iodine or dilute proflavine are
recommended.
The risk of incorporating viable exfoliated
intraluminal cells in the second staple line is thus
eliminated and a second TA-45 or TA-30 is fired
through the washed bowel while the anatomy is
distorted by upward traction on the first (pathol-
ogist’s) stapler. This process, in our view, justifies
the cost of a second stapler because of the greater
security against spillage of potentially malignant
bowel contents. Only this washed staple line
remains within the patient. Figure 7.14. Schematic drawing of washout below an
The first of these two linear staple lines should be occlusive staple to remove any debris, which may contain
safely clear of the palpable distal edge of the cancer. viable tumour cells.

HEBK001-C07_p103-123.indd 120 11/02/13 3:51 PM


DISSECTION OF THE MOST DISTAL MESORECTUM   121

For troublesome presacral, pelvic side-wall or other


bothersome bleeding, a haemostatic agent such as
Tachosil® may be helpful. Another strategy to keep
in mind is that rather than repeated futile attempts
at diathermy or suturing, packing the pelvis for
10–15 min will usually arrest bleeding.

Neorectal Reservoir

When TME has been performed and a coloanal


anastomosis is required, a neorectal reservoir pro-
vides better functional outcome than a straight
colonic anastomosis. Whatever technique is used,
adequate length of colon from the splenic flex-
ure mobilization is essential for the pouch to lie
without tension in the hollow of the sacrum. For
a reservoir, a side-to-end (side of colon to end of
anorectum) anastomosis is acceptable and easiest
to construct. Alternatives include construction of a
colonic J-pouch or a coloplasty.
For a side-to-end anastomosis, approximately half
of the colonic staple line is excised and the lumen
washed out. This allows assessment of the distal
colonic blood supply and inspection of the mucosa.
An appropriately sized circular stapler is select-
ed (28–31 mm head is optimal), and the head is
Figure 7.15. Modified ‘Triple stapling technique’. The washed detached. The detached head of the gun is inserted
muscle tube is sectioned between the two linear staplers. The into the lumen spike first, and the spike is brought
muscle tube is divided on the proximal surface of the distal out through the antimesenteric border, halfway
stapler (Moran triple stapling).
between the taenia coli, approximately 4 cm from
the distal colonic end. The defect in the staple line
The rectal cancer specimen is now removed. Care- is closed with interrupted sutures. We tend to invert
ful examination of the bowel end in the upper sta- the remaining staple line as well.
pler will confirm clearance: frozen section is general- The side-to-end technique provides a flat surface
ly unnecessary. If there is doubt about clearance, the for the proximal part of the anastomosis and leaves
staples may be removed and the lumen inspected. room for the thicker anorectal end.
Only after checking this should the distal (patient’s) Several variations of pouch construction are
stapler be removed. In the occasional case where the available. Typically a GIA-60 is inserted via a colos-
clearance is marginal, the distal stapler can be pulled tomy 5 cm from the end of the fully mobilized colon
up hard and a further linear stapler applied and fired to create a J-pouch.
beyond the distal one, although clearly this is less per- The CEEA-31 staple gun anvil is inserted into the
fect than first-time clearance of the cancer. same colostomy, which is purse-stringed around
If there are no concerns with clearance after the shaft with 00 Surgipro.
inspection (and removal of the staples if necessary),
the distal linear stapler is removed, leaving a trans-
verse staple line across the anorectal muscle tube. Circular Stapled Anastomosis
The pelvic cavity is washed copiously and
inspected for bleeding. Haemostasis is secured with The anorectal remnant is palpated from between
carefully applied diathermy or suturing if necessary. the legs. The anal canal may have to be dilated

HEBK001-C07_p103-123.indd 121 11/02/13 3:52 PM


122  Total mesorectal excision for rectal cancer

gently to accommodate the lubricated circular sta-


ple gun. Relaxation of the anal sphincter by per-
anal application of glyceryl trinitrate (GTN) cream
applied 30–60 min before or sublingual GTN spray
applied 5 min before may facilitate introduction of
the stapler by relaxing the internal sphincter. The
body of the circular stapler, usually the CEEA-31, is
inserted from below transanally. It is essential with
ultra-low anastomoses to be certain that only the
internal sphincter is purse-stringed into the instru-
ment. To this end, it must be confirmed from above
that only one thickness of muscle can be felt around
the periphery of the cartridge. Care must be taken
not to disrupt the transverse staple line, and the
abdominal surgeon may have to bimanually assist
in this step to ensure safe placement. A St Mark’s
retractor helps to visualize the anorectal stump and
anteriorly retract the vesicles and prostate in male
patients and the vagina in female patients.
Once the circular ring of the gun is visible clearly
through the bowel wall, the gun is opened and the
protruding spike guided through the bowel, ideally
just behind the linear staple line. The anvil (head)
of the gun in the proximal bowel is brought down,
engaged with the shaft. The gun is closed slowly
until the tissues are in apposition, as seen on the
tissue indicator mechanism on the gun shaft (see
Figure 7.16).
At this point, it is mandatory to check the align-
ment of the proximal colon (including the trans-
verse colon) to ensure there is not a 3608 twist of
the colonic mesentery before firing the stapler. Figure 7.16. Side- (of the colon) to-end anastomosis using
The circular stapler is fired according to the a circular stapling gun.
manufacturer’s instructions. A delay of a minute or
so before opening the gun according to the manu- haematoma in the hollow of the sacrum, which may
facturer’s instructions is said to reduce the risk of become infected, form an abscess, and point into the
staple line haemorrhage. bowel at or near the anastomosis, thus creating a late
The doughnuts are inspected for intactness of leak 10–20 days later.
the tissue rings. If there is concern about tumour
clearance in a low rectal cancer, the distal doughnut
should be sent for histology. Defunctioning a Low Anastomosis
The anastomosis is gently palpated for integrity
and can be air-tested by filling the pelvic cavity Even if the anastomosis is airtight, consideration
with water and insufflating air via the anal canal should be given to temporarily defunctioning all
using a bladder syringe. If an air leak is identified, coloanal anastomoses after TME. A randomized tri-
it may be possible to repair it with interrupted al in 2009 reported a 28 per cent leak rate in patients
sutures, if necessary by a transanal approach. after TME without a loop stoma compared with 10
Two low-suction Abdovac drains are used for 48 h, per cent in patients with a stoma.5
unless there is copious drainage, when they may Factors that have been reported by many groups
need to be left for longer. The objective is to avoid to increase the risks of anastomotic leakage are

HEBK001-C07_p103-123.indd 122 11/02/13 3:52 PM


FURTHER READING  123

height of the anastomosis from the anal verge (par- 3. Yano H, Moran B. The incidence of lateral pelvic
ticularly below 5 cm, which includes all patients side-wall nodal involvement in low rectal cancer may
be similar in Japan and the West. Br J Surg 2008; 95:
who have had a TME), male patients, preoperative
33–49.
chemoradiotherapy, and intraoperative techni- 4. Walsh PC, Schiegel PN. Radical pelvic surgery with
cal factors such as major bleeding and absence of preservation of sexual function. Ann Surg 1988; 208:
a loop stoma. There is little to choose between a 391–400.
defunctioning loop transverse colostomy and loop 5. Matthiessen P, Hallböök O, Rutegård J, Simert G,
ileostomy, although an ileostomy has the added Sjödahl R. Defunctioning stoma reduces symptomatic
risks of a high-output stoma and probably more anastomotic leakage after low anterior resection of the
rectum for cancer: a randomized multicenter trial. Ann
adhesional obstruction risks in the longer term.
Surg 2007; 246: 207–14.
Unquestionably, a loop stoma reduces the conse-
quences of a leak and the need for emergency sur-
gery. If the recovery is uneventful, the stoma may be
closed 6–8 weeks later after a water-soluble enema FURTHER READING
and rectal palpation to check there is no leak and to
dilate up any slight narrowing, which is quite com- Birbeck K, Macklin C, Tiffen N et al. Rates of circumferen-
tial resection margin involvement vary between sur-
mon in the defunctioned anastomosis.
geons and predict outcomes in rectal cancer surgery.
If a temporary stoma has not been placed and Ann Surg 2002; 235: 449–57.
there are concerns in the postoperative period, Daniels IR, Fisher SE, Heald RJ, Moran BJ. Accurate
an anastomotic leak should be sought by a rec- staging, selective pre-operative therapy and optimal
tal contrast study (often performed in combina- surgery improves outcome in rectal cancer surgery:
tion with an abdominal and pelvic CT scan). If a a review of the recent evidence. Colorectal Dis 2007;
leak is detected, the patient should have immedi- 9: 290–301.
Dukes CE. The classification of cancer of the rectum.
ate broad-spectrum antibiotics while being pre-
J Pathol Bacteriol 1932; 35: 323.
pared for emergency reoperation surgery, which Edwards DP, Leppington-Clark A, Sexton R, Heald RJ, Moran
will need a stoma. On occasion, it is possible to BJ. Stoma related complications are more frequent after
salvage the anastomosis by a combination of a transverse colostomy than loop ileostomy: a randomized
proximal loop colostomy, distal washout of the controlled trial. Br J Surg 2001; 88: 360–63.
downstream colon and a pelvic drain. In some Heald RJ, Husband EM, Ryall RDH. The mesorectum in
patients, however, anastomotic excision with an rectal cancer surgery: the clue to pelvic recurrence?
Br J Surg 1982; 69: 613–6.
end stoma is required.
Heald RJ. The ‘holy plane’ of rectal cancer. J R Soc Med
1988; 81: 503.
Heald RJ, Moran BJ, Ryall RDH, et al. The Basingstoke
Conclusion experience of total mesorectal excision 1978–1997.
Arch Surg 1998; 133: 894.
Rectal adenocarcinoma is a common cancer and Mercury Study Group. Diagnostic accuracy of pre-­
is curable by surgery alone in most cases. Recent operative magnetic imaging in predicting curative
advances in pelvic MRI, adoption of the surgical resection of rectal cancer; prospective observational
concepts of total mesorectal excision, and ­excellent study. BMJ 2006; 333: 779–84.
MacFarlane JK, Ryall RD, Heald RJ. Mesorectal excision
mechanical stapling instruments have revolution-
for rectal cancer. Lancet 1993; 341: 457.
ized the management and outcomes of this techni- Moran BJ, Docherty A, Finnis D. Novel stapling technique
cally challenging but eminently curable cancer. to facilitate low anterior resection for rectal cancer.
Br J Surg 1994; 81: 1230.
Quirke P, Durdey P, Dixon MF, et al. Local recurrence of
rectal adenocarcinoma due to inadequate surgical
References resection: histopathological study of lateral tumour
spread and surgical excision. Lancet 1986; 2: 996.
1. Moran BJ. Stapling instruments for intestinal anastomo- Quinlan DM, Epstein JL, Careter BS, Walsh PC. Sexual
sis in colorectal surgery. Br J Surg 1996; 83: 902–9. function following radical prostatectomy: influence of
2. Karanjia ND, Schache DJ, North WR, et al. ‘Close preservation of neurovascular bundles. J Urol 1991;
shave’ in anterior resection. Br J Surg 1990; 77: 510. 145: 998–1002.

HEBK001-C07_p103-123.indd 123 11/02/13 3:52 PM


8
Abdominoperineal excision
of the rectum
TorbjÖrn Holm

Introduction This paper had an enormous impact on the sur-


gical community, and ‘Miles’ operation’ became the
The earliest surgical approaches to rectal cancer gold standard procedure for all rectal carcinomas for
were via the perineum, and the techniques used many decades. The concept of removing the entire
were exclusively extraperitoneal. The intraoperative rectum and the anus in all patients with rectal can-
and postoperative mortality was high, the postop- cer gradually changed with time, however, and the
erative functional results were extremely poor, and increasing experience with bowel reconstruction,
the local recurrence rate ranged up to 90 per cent. including the development of stapling instruments,
An important step in the development of the sur- led to the new concept of anterior resection and low
gical treatment for rectal cancer was taken by W. anterior resection, which became the standard proce-
Ernest Miles, a surgeon at St Mark’s Hospital in dures for tumours in the upper and middle rectum.2–6
London, who on 19 December 1908 in the Lancet For tumours in the lower rectum, most surgeons
published a paper entitled ‘A method of perform- continued to perform abdominoperineal excision
ing abdomino-perineal excision for carcinoma (APE), although the extensive perineal approach
of the rectum and of the terminal portion of the described by Miles was more or less forgotten and
pelvic colon’.1 In Miles’ original description of the the synchronous combined APE was introduced as
procedure, the rectum was bluntly mobilized down a feasible procedure, which became popular and
to the sacrococcygeal articulation posteriorly, to the gained widespread use in the treatment of low rectal
prostate anteriorly and to the upper surface of the cancer.7 During the synchronous combined opera-
levator ani laterally. A colostomy was brought out tion, the perineal part is carried out simultaneously
and the abdominal wall was closed. The patient was with the pelvic part of the abdominal procedure,
then turned over and placed in the right lateral and with the patient in the supine lithotomy or Lloyd
semiprone position. The perineal part of the opera- Davies position. The rectum with its mesorectum
tion included a wide excision of skin and fat, and is first mobilized down to the pelvic floor; the peri-
Miles emphasized that the levator muscles should neal surgeon then enters the pelvic cavity just in
be divided ‘as far outwards as their origin from the front of the coccyx, the levator muscles are divided
white line so as to include the lateral zone of spread’. on both sides, and the rectum is dissected off the
The specimen was brought out through the peri- prostate or the vagina and the specimen delivered
neum and the skin was closed over two drains. through the perineum.

HEBK001-C08_p124-139.indd 124 08/02/13 5:54 PM


PROBLEMS ASSOCIATED WITH CONVENTIONAL SYNCHRONOUS COMBINED  125

Despite the gradual improvements in rectal cancer have not improved to the same degree as that seen
treatment during the twentieth century, local control after anterior resection. In one study based on 561
remained a major problem after surgery, with local patients in Leeds in the UK, it was reported that
recurrence rates of up to 50 per cent after potentially compared with patients who had an anterior resec-
curative resections.8 Therefore, preoperative radio- tion during the same time period, patients under-
therapy was evaluated in several randomized trials going APE had a higher local failure rate (22.3 per
during the 1980s; it was shown to reduce the local cent v. 13.5 per cent) and poorer survival (52.3 per
recurrence rate by 50 per cent and also to improve cent v. 65.8 per cent).14
cancer specific survival. It was with the develop- In another paper, based on data from five differ-
ment of total mesorectal excision (TME), however, ent European trials, it was reported that the APE
as described by Bill Heald, that the picture changed procedure was associated with an increased risk of
dramatically.9,10 The TME technique for rectal can- circumferential resection margin (CRM) involve-
cer resection was introduced in many countries ment, an increased local recurrence rate and a
over the past 15–20 years, and subsequently the decreased cancer specific survival.15
results with regard to local control and cancer sur- The difference in oncological results between the
vival have improved significantly. Local recurrence two procedures may be explained by several factors,
rates are now reported to be less than 10 per cent including anatomical difficulties and the surgical
in population-based studies.11–13 The acknowledge- technique associated with standard APE surgery.
ment of TME as the standard surgical technique in In the lower rectum, the surrounding mesorectum
the treatment of rectal cancer has resulted not only in is reduced in size and disappears at the top of the
improved local control but also in increasing rates of sphincters. Below this level, the sphincter muscle
sphincter-saving procedures and improved survival. forms the CRM. As mentioned above, the abdomi-
Therefore, since the mid-1990s, teaching in rectal nal dissection during a conventional synchronous
cancer surgery has focused mainly on the operative combined APE is often carried out along the mes-
technique of TME and anterior resection. Although orectum, all the way down to the pelvic floor and the
the technique used for the abdominal part of APE top of the puborectalis muscle, with the mesorectum
was modified along the lines of TME, little attention being mobilized off the levator muscles. The peri-
was given to the perineal part of this procedure. Thus, neal dissection then follows the external sphincter to
most surgeons adopted the technique of sharp dissec- meet the pelvic dissection at the top of the anal canal
tion under direct vision outside the mesorectal fascia (Figure 8.1a). With this technique, the retrieved
down to the pelvic floor, with the aim to save auto- specimen often has a typical ‘waist’ at 3–5 cm from
nomic nerves and to create a perfect specimen with the distal end, corresponding to the top of the exter-
an intact mesorectal fascia. The perineal part, how- nal sphincter at the level of the puborectalis muscle
ever, has often been completed in the conventional and the lowest part of the mesorectum (Figure 8.1b).
way, with dissection outside the external sphincter This inwards coning at the pelvic floor carries
and with the division of the levator muscles close to the dissection close to the rectal wall, and several
the rectal wall. With the patient in the supine lithoto- studies have reported higher rates of bowel perfo-
my position, it is difficult to achieve an optimal view, ration and tumour involvement of CRM after APE
especially anteriorly (in the front), and therefore parts compared with after anterior resection. Nagtegaal
of the perineal dissection are often done with blunt and colleagues assessed 846 anterior resection
dissection when this approach is used. specimens and 373 APE specimens from the Dutch
TME trial. They found that the plane of resection
was within the sphincter muscle, the submucosa
PROBLEMS ASSOCIATED WITH or lumen in more than a third of the APE cases
CONVENTIONAL SYNCHRONOUS and was on the sphincter muscles in the remain-
COMBINED ABDOMINOPERINEAL ing cases. This resulted in a positive CRM rate of
EXCISION 30.4 per cent after APE versus 10.7 per cent after
anterior resection, and a perforation rate of 13.7
In recent years, several authors have acknowledged per cent after APE versus 2.5 per cent after ­anterior
the fact that local control and survival after APE ­resection.16 Similarly, population-based reports

HEBK001-C08_p124-139.indd 125 08/02/13 5:54 PM


126  Abdominoperineal excision of the rectum

Figure 8.1a. The pelvic dissection


in a conventional synchronous
combined APE is carried along
Levator ani
outside the mesorectal fascia down
Obturator internus to the top of the anal canal (blue
line) and the perineal dissection is
Ischioanal fossa carried along the external sphincter
(red line). The two dissection planes
External sphincter
meet at the level of the puborectal
muscle, which creates a waist on
Internal sphincter the specimen.

Figure 8.1b. Photograph showing a


fresh specimen after a conventional
APE, with the typical waist at the level
of the puborectal muscle.

from Sweden, Norway and the Netherlands have anterior resection, even for tumours of the lower
shown a threefold increase in perforation rates after rectum. It has also been shown that these proce-
APE compared with anterior resection (14–15 per dures are feasible and oncologically safe, provided
cent v. 3–4 per cent).17 the tumour can be removed with a clear distal and
Thus, the differences in oncological outcome circumferential margin. In dedicated and highly
between the conventional type of APE and anterior specialized centres adopting intersphincteric ante-
resection may be explained to a substantial part by rior resection for appropriate cases, the overall APE
the increased risk of tumour-involved margins and rate may be below ten per cent.
inadvertent bowel perforations, as both of these On the other hand, the functional results after an
factors are significantly related to local control and ultra-low anterior resection may be poor, especially
survival. if the patient has received preoperative radioche-
With the development of TME, leading to sub- motherapy.18 In patients with a preoperative history
stantially improved results after anterior resection, of gas or faecal incontinence, careful counselling is
many surgeons have advocated low or ultra-low therefore mandatory and information should be

HEBK001-C08_p124-139.indd 126 08/02/13 5:54 PM


PREOPERATIVE PREPARATION  127

100
90
80
70
60
Figure 8.2. Graph from 50 LAR APE Hartmann
the Swedish Rectal Cancer 40
Registry showing the
30
proportion of patients, with
rectal cancer below 6 cm, 20
operated on with APE, AR 10
or Hartmann’s procedure, 0
annually since 1995. 1995 1997 1999 2001 2003 2005 2007 2009 2011

given about the risk of a poor functional outcome the operation follows the standard TME principles,
after an anterior resection. In such patients, a per- there has been no obvious agreement on the surgi-
manent stoma may be preferable. cal details of the perineal part of the operation. This
If the tumour in the lower rectum is more probably explains the significant variability in the
advanced, growing close to or into the distal mes- observed rates of tumour-involved margins, bowel
orectal fascia, the levator muscle or the external perforations, local recurrence and survival.20 Due
sphincter and thereby threatens the potential cir- to this variability, and the suboptimal results after
cumferential resection margin, it may not be pos- APE, there has been a call for a different concept and
sible to perform a safe anterior resection; in these a more standardized approach to APE.21 In recent
cases, an APE is necessary. The decision on when years, a new concept of APE has therefore evolved,
to recommend an APE is therefore related to the which takes into account the specific anatomical
patient and the tumour characteristics. Since such structures of the perineum and the pelvic floor and
variables are interpreted differently between differ- which aims to adopt and standardize the procedure
ent surgeons, the rate of APE varies greatly between according to the characteristics of the patient and the
individual surgeons and between different institu- tumour. Three basic types of APE can be described
tions. Morris and colleagues reported that the rate in relation to the perineal approach and the extent of
of APE varied from 8.5 per cent to 52.6 per cent dissection: the intersphincteric APE, the extralevator
between different English hospitals.19 In Sweden, APE and the ischioanal APE. The mobilization of the
the rate of APE for low rectal cancer, as defined by rectum and the mesorectum during the pelvic dis-
tumours within 6 cm from the anal verge, has varied section in the abdominal part of the operation differs
between 80 per cent and 92 per cent over the past between the intersphincteric APE and the two other
15 years (Figure 8.2). Thus, APE is still a common types. In addition, the indications are different for
operation for low rectal cancer. Since the results the three procedures, as shown in Table 8.1.
have been suboptimal, it is important to change the
concept of APE to reduce the rate of inadvertent
bowel perforations and tumour-involved margins PREOPERATIVE PREPARATION
and thereby obtain improved oncological outcomes.
All patients planned for an APE should be well
informed about the extent of the procedure, the
The new concept of potential complications that may occur postoper-
abdominoperineal excision atively, and the possible late sequel that they may
have to live with. These adverse outcomes after APE
One obvious problem associated with the conven- surgery are discussed below.
tional type of synchronous combined APE is the lack A crucial part of the preoperative preparation
of standardization. Although the abdominal part of and information is to have the patient meet a stoma

HEBK001-C08_p124-139.indd 127 08/02/13 5:54 PM


128  Abdominoperineal excision of the rectum

Table 8.1. Indications for APE in rectal cancer The patient is placed in a modified lithotomy posi-
Inter-sphincteric APE tion, with the buttocks brought down to the edge of
•  Patient unsuitable for bowel reconstruction the table and the legs placed in soft stirrups, but the
•  Preoperative history of incontinence legs are not elevated during the abdominal phase. A
•  High risk of anastomotic leak preoperative briefing is important to allow the sur-
•  Co-morbidity: crucial to prevent leakage + fatal outcome geon to share the plan with the entire operative team
•  Patient’s preference and to confirm the presence of appropriate instru-
Extra-levator APE ments such as self-retaining retractors, St Mark’s pel-
•  Tumour extending less than 1 cm from dentate line vic retractors, a variety of staplers and other devices,
(T2 – T4 cancer) including long instruments. Headlights or lighted
•  Tumour threatening CRM retractors may facilitate dissection deep in the pelvis.
Ischio-anal APE The assistance of an experienced second surgeon is
•  Locally advanced cancer infiltrating levator muscles, invaluable and strongly recommended.
ischio-anal fat or peri-anal skin The surgeon should reassess the rectal cancer by
•  Perforated cancer with abscess or fistula in ischio-anal digital rectal examination and confirm the degree
compartment of involvement of the anal sphincter or other organs
and the level of the distal edge of the tumour. In
female patients, the vagina must be examined to
nurse, well ahead of the operation. The stoma nurse assess the relation of the tumour to the posterior
has an important role in informing the patient vaginal wall. The abdomen and perineum, includ-
about the practicalities around stoma care and how ing the vagina in female patients, should be pre-
to use bags and other aids. It is also very important pared and properly draped.
that the placement of the stoma is assessed carefully
to avoid a suboptimal placement, close to a skin-
fold or a scar. The patient needs to be able to see ABDOMINAL PROCEDURE IN
the stoma, which may be a problem in patients who ABDOMINOPERINEAL EXCISION
are obese if the stoma is placed too low. Thus, the
stoma site should always be marked in advance by With a few exceptions, the approach and the opera-
the stoma nurse. tive technique for the abdominal part of this pro-
Prophylaxis against deep venous thromboem- cedure are identical to those used with TME and
bolism should be administered the evening before anterior resection, as described in Chapter 6.
surgery, and antibiotic prophylaxis against post- The abdomen is opened through a midline inci-
operative infection should be given, either orally sion and the entire abdominal cavity is explored to
in the morning or intravenously within 30 min of detect any metastatic disease or other unexpected
the abdominal incision. Per oral mechanical bowel pathology. The small bowel is packed into the upper
preparation is not necessary for APE, but it is rec- abdomen, the patient is placed in a slight Trendelen-
ommended to give an enema to clear the rectum in burg position, and a self-retaining retractor is insert-
the morning before surgery. After administration of ed. The sigmoid colon may be mobilized from medi-
general anaesthesia, a bladder catheter is inserted. al to lateral or vice versa, although it is our preference
The main reason for this is to identify the urethra to start the mobilization laterally. The sigmoid colon
during the perineal phase of an extralevator APE or is retracted to the right and the peritoneal attach-
ischioanal APE. Our preference is to keep the cath- ments laterally are incised along the avascular plane
eter closed and to insert a suprapubic catheter once (white line of Toldt), distally to the level of the prom-
the abdomen is opened. The urethral catheter is ontory and as far proximally as needed. It is usually
removed directly after surgery and the suprapubic necessary to mobilize a portion of the descending
catheter kept postoperatively. In patients scheduled colon to allow the later construction of a tension-
for an intersphincteric APE, there is no need for a free end colostomy, elevated 1 cm above the skin
urethral catheter, as the perineal dissection is car- level. A complete takedown of the splenic flexure, as
ried out between the internal and external sphinc- is routinely done if a low anastomosis is planned, is
ter, at a safe distance from the urethra. usually unnecessary for an APE. The left ureter and

HEBK001-C08_p124-139.indd 128 08/02/13 5:54 PM


PELVIC DISSECTION IN INTERSPHINCTERIC ABDOMINOPERINEAL EXCISION  129

gonadal vessels are identified and preserved by using in male patients and the vagina in female patients.
sharp and gentle blunt dissection to separate the ret- The lower anterolateral dissection is the most dif-
roperitoneal tissues from the left colonic mesentery. ficult part of the abdominal phase of the operation
The sympathetic nerve plexus in front of the aorta because the correct plane is often difficult to find
is identified, and the dissection continues in front here, and the inferior hypogastric plexus must be
of these nerves and from left to right, just posterior carefully preserved to maintain postoperative sexual
to the inferior mesenteric artery (IMA). The perito- and urinary function. Reducing the angle of the
neum on the right side of the colonic mesentery is Trendelenburg position or even shifting the patient
then opened and the IMA is followed proximally to to a reverse Trendelenburg position may facilitate
its origin at the aorta. the exposure for the anterior dissection.
There is no consensus on where to divide the Mobilization of the rectum with an intact mes-
IMA. Some surgeons prefer a high ligation at the ori- orectum is continued down to the pelvic floor and
gin from the aorta and suggest that this maximizes the puborectalis muscle. Because the intersphinc-
the lymph node yield and may improve oncological teric APE is generally not performed when the
outcome. Other surgeons have a preference for a low tumour is close to the anus, a transverse stapler can
ligation just distal to the left ascending colic artery be put across the rectum below the tumour to seal
and argue that this ensures a better blood supply the bowel and to prevent leakage of mucus or faeces
to the remaining left colon and may prevent nerve from the anus.
damage at the base of the IMA, resulting in less func-
tional impairment. There is not enough evidence to
state that one approach is better than the other. After Perineal Part of Intersphincteric
ligation of the IMA and the inferior mesenteric vein Abdominoperineal Excision
at the same level, the sigmoid mesentery is divided
with diathermy coagulation of small vessels, division The patient’s legs are elevated and the perineum is
and ligation of the marginal artery, and finally divi- exposed. The surgeon and assistant now move from
sion of the colon at the level of the proximal sigmoid the abdomen to perform the perineal phase of the
colon, preferably with a linear stapler to prevent any intersphincteric APE. The anal canal is washed out
faecal contamination. and an incision is made around the anus, just dis-
tal to the intersphincteric groove. A self-retaining
retractor with hooks is recommended to optimize
PELVIC DISSECTION the view and to facilitate the intersphincteric dis-
IN INTERSPHINCTERIC section. Once the skin incision is made, the anus is
ABDOMINOPERINEAL EXCISION closed with a running suture. The dissection then
follows the intersphincteric plane between the inter-
Pelvic dissection in intersphincteric APE is identi- nal and external sphincter, around the circumfer-
cal to that performed for anterior resection, which ence of the anal canal, and all the way up to the pub-
is described in detail in Chapter 6. In summary, the orectal sling and into the pelvic cavity (Figure 8.3A).
loose connective tissue plane (‘holy plane’) separat- The specimen is gently removed through the
ing the mesorectal fascia from the parietal pelvic perineal incision or, if the mesorectum is large
structures is identified and followed first posteriorly, and bulky, lifted up from the pelvis and removed
then to the left and right, and finally anteriorly while from the abdomen via the abdominal incision
the peritoneum is gradually divided. Gentle traction (Figure 8.3B).
on the specimen and counter-traction with appro- The perineal incision is closed subcutaneously
priate retractors is crucial to achieve a good view of with a running or interrupted suture in the pub-
this plane. The superior hypogastric plexus is iden- orectalis and external sphincter. It is our preference
tified at the sacral promontory and the hypogastric to use a running suture in three layers, where the
nerves should be identified, protected and preserved most superficial suture line is placed subdermal to
while the dissection gradually proceeds downwards leave the skin unsealed in order to allow for dis-
in the pelvic cavity. Anteriorly the dissection is con- charge of fluid from the wound.
ducted just posterior to the vesicles and prostate

HEBK001-C08_p124-139.indd 129 08/02/13 5:54 PM


130  Abdominoperineal excision of the rectum

Figure 8.3a. The pelvic dissection


in an inter-sphincteric APE is carried
Levator ani
along outside the mesorectal
Obturator internus fascia down to the top of the anal
canal (blue line) and the perineal
Ischicanal fossa dissection is carried out between
the internal and external anal
External sphincter sphincter (red line). The two
dissection planes meet at the level
Internal sphincter of the puborectal muscle.

Figure 8.3b. Photograph showing


a fresh specimen after an inter-
sphincteric APE.

Finalizing the Abdominal Procedure be mobilized from the transverse colon and from the
greater curvature of the stomach to prepare an omen-
When the perineum is closed, the patient’s legs are toplasty, which can help to fill out the empty pelvic
put down and the abdominal part of the operation cavity. We prefer to place a drain in the pelvic cavity,
continues with a washout of the pelvic cavity, pref- but there is no substantial evidence that this is of any
erably with sterile water or some other cytotoxic value. Finally, a thorough control of haemostasis in
agent, and haemostatic control. the abdomen and pelvic cavity is mandatory before
stoma formation and closure of the abdominal wall.

Omentoplasty Stoma Formation


Bowel obstruction due to entrapment of the small A circular incision is made in the skin at the marked
bowel in the pelvic cavity is not infrequent after APE. stoma site, and the dissection is carried through the
Therefore, if the patient has a large omentum, this may subcutaneous fat down to the aponeurosis covering

HEBK001-C08_p124-139.indd 130 08/02/13 5:54 PM


EXTRALEVATOR ABDOMINOPERINEAL EXCISION  131

the rectus abdominis muscle. There is no consensus pelvic dissection during the abdominal part of an
on how to incise this aponeurosis; some surgeons extralevator APE differs notably from an anterior
make a longitudinal incision, others prefer a cross- resection or an intersphincteric APE (Figure 8.4A).
incision, while some make a round hole and remove
a small disk of the aponeurosis. The fibres of the Pelvic Dissection in Extralevator
rectus abdominis muscle are gently separated and Abdominoperineal Excision
the posterior sheet and peritoneum is opened.
The divided sigmoid or descending colon is now The initial abdominal and pelvic dissection is iden-
picked up and brought out through the stoma site. tical to that described above, but with one very
At this point it is vital to ensure that the bowel is of important difference. In both anterior resection
sufficient length and has a good blood supply. The and intersphincteric APE, the dissection continues
bowel should not be opened at this point, and not all the way down to the pelvic floor and the pub-
until the abdominal wall is closed and the wound orectalis muscle and subsequently the mesorectum
properly dressed. is lifted off the levator muscles. In extralevator APE
In recent years some authors have advocated plac- it is crucial not to take the mobilization of the rec-
ing a lightweight mesh in a sublay position, deep to tum and mesorectum as far down as the pelvic floor.
the rectus abdominis muscle, to prevent parastomal Instead, the dissection should proceed only down to
hernia formation, a complication that is frequent in the sacrococcygeal junction dorsally, just beyond the
patients with a colostomy. The use of a preventive inferior hypogastric plexus anterolaterally, and ante-
mesh in colostomy formation is described in more riorly dissection should stop just below the seminal
detail below. vesicles in male patients or the cervix uteri in female
Once the bowel has been pulled through the sto- patients. By terminating the mobilization of the rec-
ma site, the abdominal wall is closed. This may be tum and mesorectum at this level, the mesorectum
done in different ways, but we strongly advocate a is still attached to the levator muscles of the pelvic
monofilament absorbable or non-resorbable suture, floor, which is a crucial feature of extralevator APE.
a short distance between stitches, and a suture length After completion of the dissection down to this lev-
to wound length ratio of at least 4 to reduce the risk el, the pelvic cavity is rinsed as described above. The
of wound dehiscence and the later development of final steps of the abdominal procedure are carried
incisional hernia. The skin and wound are cleaned out just as for intersphincteric APE, the abdominal
with saline and the wound is dressed with an appro- wall is closed and the stoma fashioned.
priate bandage.
The exteriorized bowel is now opened, everted
and fixed to the skin with interrupted monofila- Perineal Part of Extralevator
ment sutures. Finally, a stoma bag is placed to cover Abdominoperineal Excision
the stoma.
The perineal part of extralevator APE differs consid-
erably from the perineal part of intersphincteric APE.
EXTRALEVATOR This part of the operation can be performed with
ABDOMINOPERINEAL EXCISION the patient in the supine Lloyd Davies position, or in
the prone jack-knife position. Our preference is the
The main objective of an extralevator APE is to prone jack-knife position, due to the excellent expo-
diminish the risk of inadvertent bowel perforation sure of the operative field. Other surgeons prefer the
and tumour involvement of the circumferential supine position, mainly to avoid the time-consuming
resection margins. This is a consequence of excision process of turning the patient and subsequent prepa-
of the levator muscles en bloc with the mesorectum ration and dressing of the perineal area. The pros and
to protect the most distal part of the bowel, thereby cons of the two different positions during the peri-
avoiding the ‘waist’ of the specimen, which has been neal phase of the operation are discussed below.
so common after the conventional type of synchro- With the patient in the prone jack-knife posi-
nous combined APE. Since the levator muscles tion, the exposed, washed and sterile operative field
should not be separated from the mesorectum, the should be well wide of the anus, laterally halfway

HEBK001-C08_p124-139.indd 131 08/02/13 5:54 PM


132  Abdominoperineal excision of the rectum

Figure 8.4a. The pelvic dissection


in an extra-levator APE is carried
Levator ani
along outside the mesorectal fascia
but stops at the top of the levator
Obturator internus
muscle (blue line). The perineal
Ischieanal fossa dissection proceeds just outside
the external sphincter and along
External sphincter the levator muscle fascia, up to
its origin at the obturator internus
Internal sphincter muscle (red line).

Figure 8.4b. Photograph showing


a fresh specimen after an extra-
levator APE. The specimen is more
cylindrical, without a waist, because
the levator muscle is attached to the
mesorectum.

along each buttock and dorsally up to the level of Closing the anus can be done with a double purse-
the mid-sacrum. In male patients the scrotum and string suture or with an inverting running suture
penis should be covered and kept out of vision to after the skin incision has been made around the
expose the whole perineum. In female patients anus. The latter technique is ­especially valuable in
the vagina should be washed out and kept in the very low advanced tumours, which may protrude
operative field. In male patients a urethral catheter through the anus. After closure of the anus, it is rec-
must be in place even if a supra pubic catheter has ommended to wipe the operative field once more
been inserted during the abdominal phase of the with an alcohol solution or another appropriate
operation. This is to ensure that any damage to the antibacterial solution.
urethra during the dissection in the anterior plane, In extralevator APE, less skin and ischioanal fat
between the rectum and the prostate, can be detect- are excised compared with Miles’ original descrip-
ed and taken care of immediately if it occurs. tion of the perineal part of the APE procedure.
The perineal phase starts with closure of the anus Instead, the skin is incised around the anus, with a
to avoid any spillage of faeces or mucus, which could margin of only about 3 cm anteriorly and laterally;
contain tumour cells. The aim of anal closure is thus posteriorly the incision is carried up to the level of
to reduce infection and reduce the risk of tumour the lower sacrum, i.e. 2–3 cm cranial to the sacro-
contamination, which may result in local recurrence. coccygeal junction.

HEBK001-C08_p124-139.indd 132 08/02/13 5:54 PM


EXTRALEVATOR ABDOMINOPERINEAL EXCISION  133

With gentle traction and counter-traction on palpated and visualized, or preferably 1–2 cm poste-
the skin edges, the dissection is continued in the rior to this point. The specimen is now still attached
­subcutaneous fat. As the dissection proceeds deeper, to the anterior aspect of the levator muscles and to
it is important to identify the subcutaneous exten- the prostate or posterior wall of the vagina.
sion of the external sphincter. These fibres of striat- The dissection in the anterior plane during the
ed muscle should be kept medially, and the dissec- perineal phase of extralevator APE is the most dif-
tion follows a plane between the external sphincter ficult, and potentially most dangerous, part of the
and the thin fascia covering the ischioanal fat in the procedure because of the close relationship between
ischioanal compartment (also called the ischiorec- the anterior rectal wall and the prostate or posterior
tal fossa) on both sides. At the top of the external vaginal wall. In addition, the neurovascular bun-
sphincter and puborectal muscle, the levator ani dles derived from the inferior hypogastric plexus
muscles are in direct continuity and the dissection run anterolaterally on each side of the prostate or
is carried along the surface of the levator muscles all vagina and close to the rectum and are easily dam-
the way up to their insertion at the pelvic side-wall, aged if they are not recognized at this stage of the
i.e. the obturator internus muscle. operation. The dissection along the anterior and
The pudendal nerve, derived from S2–4, runs lateral aspects of the lower rectum must therefore
together with the pudendal vessels through Alcock’s be performed meticulously and with great care. If
canal and converges towards the midline on the the dissection is performed close to the rectal wall,
outer anterior surface of the levator muscles. Small there is a risk of inadvertent perforation or tumour-
branches of blood vessels and nerves deriving from involved margin; if the dissection is carried out too
the pudendal vessels and nerve cross the space laterally or too anteriorly, there is a risk of damage
between the ischioanal compartment and the leva- to the neurovascular bundles or to the prostate or
tor muscles, and these are divided by diathermy. vagina. In anteriorly located tumours, it may be
Once the surface of the levator muscles is exposed necessary to include the posterior vaginal wall or
all around the circumference, haemostasis must be a slice of the posterior prostate with the specimen,
controlled before entering the pelvic cavity. and sometimes even to sacrifice the neurovascular
In the midline, the levator muscles are attached bundle on one side, to be able to achieve a nega-
to the anterior surface of the coccyx and continue tive CRM. This extension of the procedure should
as the presacral fascia on the anterior lower aspect ideally be planned in advance, however, so that the
of the sacrum. The dissection follows the proxi- surgeon is prepared for it and the patient is well
mal portion of the levator muscles on both sides informed about the consequences, which may be
of the coccyx so that the coccyx is clearly exposed. impairment of bladder or sexual function.
An incision is made at the sacrococcygeal junction, To facilitate the anterolateral dissection of the
which is easily identified by gentle moving of the lower part of the rectum, it is recommended that the
coccyx. Once the cartilaginous connection between specimen is gently brought out of the pelvic cavity
the sacrum and coccyx has been opened, the coc- so that the anterior aspect of the bowel can be seen.
cyx is pressed anteriorly to stretch the presacral fas- It is now easy to look into the pelvic cavity and to
cia, which is then divided and an entrance into the recognize the seminal vesicles and upper part of the
pelvic cavity created. At this stage it is important to prostate in male patients and the posterior vaginal
identify the mesorectum in order to not injure the wall in female patients. The plane between Denonvil-
mesorectal fascia. lers’ fascia and the prostate or posterior vaginal wall
The pelvic floor, i.e. the levator muscle, is now is now followed carefully while the surgeon attempts
divided from posterior to anterior, first on one side to identify the neurovascular bundles on each side.
and then on the other side. As the division of the pel- Gradually, these planes of dissection are developed
vic floor continues anteriorly, it is important to avoid anteriorly and alternately on the right and left sides,
division of the levator muscles too far laterally and and the remaining part of the levator muscles that
too close to the ischial tuberosity as this may injure are attached to the lowest part of the rectum are
the main pudendal nerve and vessels in Alcock’s divided. Finally, the puborectal muscle on each side
canal. The division of the pelvic floor continues until and the perineal body just posterior to the transverse
the dorsolateral part of the prostate or vagina can be perineal muscle is divided and the specimen can be

HEBK001-C08_p124-139.indd 133 08/02/13 5:54 PM


134  Abdominoperineal excision of the rectum

delivered. The excised specimen is cylindrical, usu-


ally without a waist, due to the fact that the levator
muscle is still attached to the mesorectum, forming
a cuff around the rectal muscle tube (Figure 8.4B).
As soon as the specimen is removed, it is crucial
to control haemostasis at the back of the prostate or
vagina, along the neurovascular bundle on each side
and on the pelvic side-walls. When bleeding is com-
pletely controlled, the pelvic cavity is rinsed with ster-
ile water or another appropriate cytotoxic solution.
If an omentoplasty has been prepared, the omentum
is now gently brought down into the pelvis by gentle
traction. This manoeuvre may be difficult, especially
if a large omentum is to be pulled down into a narrow Figure 8.5. In some very low and advanced tumours the
pelvis. Therefore, it is recommended that the omen- cancer growth may infiltrate the perianal skin. In these patients
tum is attached to the proximal end of the specimen a wide excision of the perianal skin and an ischio-anal APE is
with a couple of sutures during the abdominal phase necessary to achieve a potentially curative resection.
of the operation. This significantly facilitates the
placement of the omentum in the pelvic cavity, since contain tumour cells. Therefore, ischioanal APE is a
the omentum slides down into the pelvis when the valid procedure in these special situations.
rectum is gently pulled out during the perineal phase The abdominal part of ischioanal APE is exactly
of the operation. Care should be taken not to rotate equivalent to the abdominal part of extralevator
the omentum by 360 as this will impair its blood APE. Thus, the dissection stops just above the levator
supply and may result in omental necrosis. At this muscle and leaves the mesorectum attached to the
stage the pelvic drain, placed through the abdomen, pelvic floor (Figure 8.6A). When the abdominal part
is also brought down into the pelvic cavity. of the procedure is completed, with closure of the
The next step of the operation is to reconstruct abdominal wall and formation of a ­colostomy, the
and close the pelvic floor. As discussed below, there patient is turned into the prone jack-knife ­position.
are several alternative methods available to com-
plete this reconstruction.
Perineal Part of Ischioanal
Abdominoperineal Excision
ISCHIOANAL ABDOMINOPERINEAL
EXCISION After proper preparation of the skin of the perineum,
lower sacrum, the medial parts of the buttocks and
In some patients, the rectal tumour is locally the vagina in female patients, a double purse-string
advanced and may infiltrate or even perforate suture is placed to close the anus. The area of the skin
the pelvic floor, i.e. the levator muscle. In other incision in ischioanal APE depends on the extent of
patients, a perianal abscess may be the present- tumour involvement of the skin. Any tumour infiltra-
ing feature of a perforated low rectal cancer, and tion or fistula opening must be included in the excised
after drainage a fistula may persist between the skin area with a margin of at least 2–3 cm. As soon
low rectum and the perianal skin. In a few very low as the incision deepens into the subcutaneous space,
tumours, the growth may extend into the perianal the dissection should be directed laterally towards the
skin (Figure 8.5). In these instances, extralevator ischial tuberosity and progress on to the fascia of the
APE may not be sufficient to achieve a safe tumour- internal obturator muscle. Thus, ­contrary to extrale-
free CRM, and ischioanal APE is usually required to vator APE, the dissection does not follow the external
obtain an oncologically secure margin. In this situ- sphincter and levator muscle but instead is carried
ation, the levator muscle must be removed covered along the fascia of the internal obturator muscle. The
with ischioanal fat, and the ischioanal fat must be dissection is performed along this plane up to where
removed to include the perianal fistula, which may the levator muscle is inserted into the internal obtu-

HEBK001-C08_p124-139.indd 134 08/02/13 5:54 PM


ISCHIOANAL ABDOMINOPERINEAL EXCISION  135

Figure 8.6a. The pelvic dissection A


in an ischio-anal APE is carried
along outside the mesorectal fascia
but stops at the top of the levator
muscle (blue line). The perineal
dissection is directed towards tuber
os ischii and follows the obturator
Levator ani
internus muscle fascia, in order to
remove the fat in the ischio-anal Obturator internus
compartment en bloc. The size of
the skin incision depends on the Ischio-anal fossa
extent of tumour involvement in the
skin and may be extensive (left side) External sphincter
or similar to the skin incision in an
extra-levator APE (right side). Internal sphincter

Figure 8.6b. Photograph showing


a fresh specimen after an ischio-anal
APE. In very advanced tumours,
infiltrating the perineal skin, a wide
skin excision and a complete
clearance of both ischio-anal
compartments may be necessary
for a potentially curative operation.

rator muscle and hence includes the entire fat com- prostate in male patients or the vagina in female
partment of the ischioanal space. This dissection can patients. Once the specimen has been brought out of
be performed unilaterally or bilaterally, depending the pelvic cavity, the anterior and lateral dissection
on the extent of tumour growth. When the dissec- along the prostate or vagina is also carried out as in
tion up to this level is completed, the sacrococcygeal extralevator APE. As mentioned above, the difference
junction is incised and the pelvic cavity is entered between extralevator APE and ischioanal APE is that
in the same fashion as with extralevator APE. The the fat in the ischioanal space is resected en bloc and
subsequent dissection is also similar to that of extra- attached to the levator muscle (Figure 8.6B). This
levator APE, as the levator muscles are divided along procedure is very similar to what Miles described in
the fascia of the internal obturator muscle on to the 1908 in his original paper in the Lancet.

HEBK001-C08_p124-139.indd 135 08/02/13 5:54 PM


136  Abdominoperineal excision of the rectum

LAPAROSCOPIC APPROACH TO cal planes can be followed more easily and the risk
ABDOMINOPERINEAL EXCISION of perforation or involved margins of the specimen
by tumour can be reduced.
The abdominal procedure in APE may be performed In one study, it was shown that the risk of perfo-
with minimally invasive techniques, either laparo- ration in extralevator APE was significantly lower
scopic or robot-assisted. This approach is gaining when perineal dissection was carried out in the
increasing popularity, and it may well be that mini- prone jack-knife position rather than the lithotomy
mally invasive methods for the abdominal part of or Lloyd Davies position (6.4 per cent v. 20.6 per
the operation will become predominant in the near cent).23 As perforation of the specimen is a well-
future. Although some details differ between the known risk factor for adverse prognosis, it may be
open and minimally invasive approach, the main that the prone jack-knife position improves onco-
principles described above for the abdominal part logical outcomes.
of APE should be adopted, irrespective of the sur- Thus, performing the perineal part of APE with
gical method used. The principles and details of the patient in the prone jack-knife position may
minimally invasive procedures in rectal cancer are potentially improve prognosis. In addition, this posi-
described in Chapter 10. tion gives an excellent view, which facilitates teaching
of this part of the operation. With the patient in this
position and with the legs spread apart, the surgeon
Prone or supine position stands between the legs and has one assistant on each
during perineal dissection side. The operative field is well exposed to all three
surgeons, and trainees can perform a step-by-step
In Miles’ original description of the APE procedure, approach to learn how to do the operation, with full
the perineal part of the operation was performed control by the responsible surgeon.
in the right lateral and semiprone position after Many surgeons who have adopted extralevator
completion of the abdominal phase and the stoma APE for low rectal cancer also favour the jack-knife
creation. Subsequently, synchronous combined APE position for the perineal part of the procedure.
gained popularity and became standard for the vast Some surgeons still use the lithotomy or Lloyd Dav-
majority of surgeons. With synchronous combined ies position, however, for the perineal part, even
APE, the perineal part of the operation is performed though they perform extralevator APE.24 There has
with the patient in the supine Lloyd Davies position, been some debate about the pros and cons of the
and the final part of the pelvic dissection from the two approaches, and there have also been reports
abdominal side is often performed simultaneously on excellent oncological outcomes after APE with
with the perineal dissection. In addition, historically the perineal dissection performed with patients in
this perineal dissection was often done blunt and the supine position.25 It is likely that the position
often by the most junior surgeon in the operating of the patient is not crucial, provided that skilled
team. As mentioned above, the consequence of this and properly trained surgeons perform a meticu-
approach was repeatedly that the excised specimen lous dissection to create a perfect specimen in the
had a waist just proximal to the puborectal sling and extralevator plane.26
often also an inadvertent perforation or an involved
circumferential resection margin.
Due to the increasing awareness of the problems REMOVAL OF THE COCCYX
associated with synchronous combined APE, extra-
levator APE was introduced as a different and more With the conventional synchronous combined APE,
radical procedure, performed via the posterior the pelvic dissection during the abdominal part of
perineal approach in the prone jack-knife position, the operation is carried along the mesorectal fascia
which closely mirrors the original Miles opera- all the way down to the pelvic floor and beyond the
tion.22 The main purpose of this prone jack-knife coccyx. With this approach, there is no rationale for
position is to optimize the visibility of the operative removing the coccyx while performing the perineal
field and to have full control of the perineal and pel- part of the operation, because the mesorectum has
vic floor anatomy. As a result, the correct anatomi- already been detached from the levator muscle of

HEBK001-C08_p124-139.indd 136 08/02/13 5:54 PM


PERINEAL RECONSTRUCTION  137

the pelvic floor. Therefore, the division of the leva- a common clinical problem, reported to occur in
tor muscle during conventional APE is often insti- 30–80 per cent of patients with a colostomy. The
gated between the tip of the coccyx and puborectal wide variation in incidence is probably related to
sling and continued forwards bilaterally on to the the definition of a parastomal hernia, the method
prostate or the vagina. With this approach, a lim- used to diagnose the condition and the length of
ited amount of the pelvic floor muscle is removed follow-up. Although many parastomal hernias are
en bloc with the rectum and, as mentioned earlier, only cosmetically disturbing or associated with
the specimen frequently has a waist just above the mild symptoms, about 30–40 per cent of patients
puborectal sling. with parastomal hernias may require surgical repair
During extralevator APE and ischioanal APE, of the hernia due to pain, obstruction, bleeding,
the coccyx is often removed en bloc with the rec- increasing protrusion, poorly fitting stoma bag, fae-
tum. There are two main reasons for doing this: cal leakage or incarceration. Several different surgi-
first, the levator muscle is attached to the anterior cal methods are used to treat parastomal hernias,
surface of the coccyx and continues cranially as including relocation, primary closure of the fascia
the presacral fascia. If the levator muscle is to be and mesh repair, but high complication and recur-
completely removed and fixed to the mesorectum, rence rates have been reported. The use of a pros-
as in extralevator APE, then the coccyx needs to be thetic mesh to repair a parastomal hernia and to
removed. Cutting the levator muscle below the tip reconstruct the abdominal wall has gained increas-
of the coccyx will inevitably leave a part of the leva- ing popularity and improved the rate of successful
tor muscle behind. Second, it is sometimes difficult revisions, but these methods are not without prob-
to bring out the specimen from the pelvic cavity lems. Erosion of the bowel (with subsequent infec-
through a narrow opening in the pelvic floor. Espe- tion and fistula formation) and recurrent hernia
cially in male patients with a narrow pelvis and a development have been reported. The high risk of
large mesorectum, it can be hard to bring out the parastomal hernia and the suboptimal results after
specimen before starting the challenging dissection repair have led to attempts to prevent its occur-
of the anterior part of the rectum and mesorectum rence. Several studies have shown that the use of a
off the prostate or the vagina. By dividing between polypropylene mesh at the time of stoma formation
the sacrum and the coccyx and excising the coccyx is safe and may prevent the occurrence of a paras-
en bloc, the surgeon creates a wider opening of the tomal hernia. A review of three randomized con-
pelvic floor, which facilitates delivery of the speci- trolled trials and three prospective observational
men and the anterior dissection. Some authors have series reported parastomal hernia rates of 55 per
advocated an even more extensive resection includ- cent in patients who did not have a mesh and 7 per
ing the last sacral vertebra to achieve better access cent in patients in whom a mesh was used. Post-
and control of the perineal part of the operation.27 operative morbidity rates were similar, irrespective
With the patient in the supine, lithotomy or of whether a mesh was used.28 Although the place-
Lloyd Davies position, removal of the coccyx is dif- ment of a mesh, preferably by a sublay technique,
ficult due to limited access to the natal cleft. There- seems promising, there are no data on long-term
fore, surgeons who are proponents of removal of results in terms of hernia recurrence or possible late
the coccyx usually also perform the perineal part of complications. Despite this, we recommend the use
extralevator APE in the prone jack-knife position. of a 10 3 10 cm lightweight synthetic mesh around
the bowel behind the rectus muscle and anterior to
the rectus sheet.
Using a mesh to prevent
parastomal hernia
PERINEAL RECONSTRUCTION
One major problem after APE with formation of a
permanent stoma is the subsequent development When the first major paper on APE was published
of a parastomal hernia in many patients, which by Miles in 1908, he described the closure of the
by definition is an incisional hernia related to the perineal wound: ‘Finally, the skin margins are
presence of a stoma in the abdominal wall. This is brought together with sutures and a large drainage-

HEBK001-C08_p124-139.indd 137 08/02/13 5:54 PM


138  Abdominoperineal excision of the rectum

tube is inserted in the anterior and the posterior Some experience with biological mesh reconstruc-
extremities of the median incision.’ Miles later aban- tion of the pelvic floor has also been reported. This
doned this technique due to the high risk of perineal option seems feasible with a reasonable complication
wound infections and the ensuing mortality, and rate.34 The number of reports is limited, however, and
instead the defect was left open and allowed to heal long-term results from biological mesh reconstruc-
by secondary intention. The practice of leaving the tion of the pelvic floor are still lacking. In fact, there
perineal wound open was applied for many decades is no standard solution for pelvic floor reconstruc-
but is now used by very few surgeons. Although this tion after APE, and the method used must be tailored
method avoids some of the infectious complica- according to the patient and the extent of excision.
tions associated with primary closure, the morbid- Thus, primary closure is generally appropriate after
ity associated with a large, slowly healing wound is intersphincteric APE, while some kind of mesh or
significant. Therefore, primary closure of the peri- flap reconstruction is often used after extralevator
neal wound has been the most common method APE. After ischioanal APE, a flap reconstruction is
of ­perineal reconstruction after synchronous com- almost always necessary, especially if the excision of
bined APE. Although the clinical course after prima- skin has been extensive. It is recommended to assess
ry closure is often uneventful, complications due to each patient carefully before surgery to determine
the perineal wound are still one of the major prob- the suitable type of pelvic floor reconstruction and
lems associated with the conventional type of APE, to establish collaboration with a plastic surgeon for
especially in patients who have received preoperative reconstruction after wider excisions.
radiotherapy. Wound problems have been reported
in up to 50 per cent of patients receiving preopera-
References
tive radiotherapy after APE with primary wound
closure.29 Wound infection and delayed healing are   1. Miles WE. A method of performing abdomino-perineal
the most common complications. These problems excision for carcinoma of the rectum and of the
may become even more frequent in patients who terminal portion of the pelvic colon. Lancet 1908; 2:
have received a combination of preoperative radio- 1812–13.
therapy and extralevator APE, with a more extensive   2. Collins DC. End-results of the Miles’ combined
excision of the pelvic floor. abdominoperineal resection versus the segmental
anterior resection: a 25-year postoperative follow-up in
A variety of surgical alternatives to primary clo-
301 patients. Am J Proctol 1963; 14: 258–61.
sure have been used to reconstruct the pelvic floor   3. Fick TE, Baeten CG, von Meyenfeldt MF, Obertop H.
and to reduce the wound healing problems after Recurrence and survival after abdominoperineal and
APE. These procedures include omental pedicle low anterior resection for rectal cancer without adjunc-
flaps (omentoplasty) and different local rotational tive therapy. Eur J Surg Oncol 1960; 16: 105–8.
musculocutaneous flaps. Data from controlled   4. Groves RA, Harrison RC. Carcinoma of the rectum
studies support the use of musculocutaneous flaps and lower sigmoid colon: abdominoperineal or anterior
resection? Can J Surg 1962; 5: 393–403.
for single-stage reconstruction after APE, especial-
  5. Slanetz CA, Herter FP, Grinnell RS. Anterior resection
ly in the presence of chemoradiotherapy. Several versus abdominoperineal resection for cancer of the
reports using the rectus abdominus, gluteus max- rectum and rectosigmoid: an analysis of 524 cases.
imus or gracilis musculocutaneous flaps have been Am J Surg 1972; 123: 110–17.
published.30,31   6. Vandertoll DJ, Beahrs OH. Carcinoma of the rectum
Rectus abdominus flaps have been most com- and low sigmoid: evaluation of anterior resection in
monly used. The reported overall complication 1766 favourable lesions. Arch Surg 1965; 90: 793–8.
  7. Schmitz RL, Nelson PA, Martin GB, Boghossian HM.
rates vary from 10 per cent to 50 per cent, while
Synchronous (two-team) abdominoperineal resection
healing rates during follow-up vary from 95 per of the rectum. AMA Arch Surg 1958; 77: 492–7.
cent to 100 per cent.32,33 Most studies are small,   8. Påhlman L, Glimelius B. Local recurrences after surgi-
with fewer than 50 reported patients; the small cal treatment for rectal carcinoma. Acta Chir Scand
number of patients, the varying definition of 1984; 150: 331–5.
wound complications and the varying follow-up   9. Heald RJ, Husband EM, Ryall RD. The mesorectum in
times probably explain the difference in complica- rectal cancer surgery – the clue to pelvic recurrence.
Br J Surg 1982; 69: 613–16.
tion rates.

HEBK001-C08_p124-139.indd 138 08/02/13 5:54 PM


References   139

10. Macfarlane JK, Ryall RD, Heald RJ. Mesorectal exci- 22. Holm T, Ljung A, Haggmark T, Jurell G, Lagergren J.
sion for rectal cancer. Lancet 1993; 341: 457–60. Extended abdominoperineal resection with gluteus
11. Kapiteijn E, Marijnen CA, Nagtegaal ID, et al. Preop- maximus flap reconstruction of the pelvic floor for
erative radiotherapy combined with total mesorectal rectal cancer. Br J Surg 2007; 94: 232–8.
excision for resectable rectal cancer. N Engl J Med 23. West NP, Anderin C, Smith KJ, Holm T, Quirke P. Mul-
2001; 345: 638–46. ticentre experience with extralevator abdominoperineal
12. Martling AL, Holm T, Rutqvist LE, Moran BJ, Heald RJ, excision for low rectal cancer. Br J Surg 2010; 97:
Cedemark B. Effect of a surgical training programme 588–99.
on outcome of rectal cancer in the County of Stock- 24. Martijnse IS, Dudink RL, West NP, et al. Focus on
holm: Stockholm Colorectal Cancer Study Group, extralevator perineal dissection in supine position for
Basingstoke Bowel Cancer Research Project. Lancet low rectal cancer has led to better quality of surgery
2000; 356: 93–6. and oncologic outcome. Ann Surg Oncol 2012; 19,
13. Wibe A, Syse A, Andersen E, Tretli S, Myrvold HE, 786–93.
Soreide O. Oncological outcomes after total mesorec- 25. De Campos-Lobato LF, Stocchi L, Dietz DW, Lavery
tal excision for cure for cancer of the lower rectum: IC, Fazio VW, Kalady MF. Prone or lithotomy position-
anterior vs. abdominoperineal resection. Dis Colon ing during an abdominoperineal resection for rectal
Rectum 2004; 47: 48–58. cancer results in comparable oncologic outcomes. Dis
14. Marr R, Birbeck K, Garvican J, et al. The modern Colon Rectum 2011; 54: 939–46.
abdominoperineal excision: the next challenge after 26. Bebenek M. Abdominosacral resection is not related
total mesorectal excision. Ann Surg 2005; 242: 74–82. to the risk of neurological complications in patients
15. Den Dulk M, Putter H, Collette L, et al. The abdomi- with low-rectal cancer. Colorectal Dis 2009; 11: 373–6.
noperineal resection itself is associated with an adverse 27. Janes A, Cengiz Y, Israelsson LA. Preventing paras-
outcome: the European experience based on a pooled tomal hernia with a prosthetic mesh: a 5-year follow-
analysis of five European randomised clinical trials on up of a randomized study. World J Surg 2009; 33:
rectal cancer. Eur J Cancer 2009; 45: 1175–83. 118–21, 122–3.
16. Nagtegaal ID, van de Velde CJ, Marijnen CA, van 28. Bullard KM, Trudel JL, Baxter NN, Rothenberger DA.
Krieken JH, Quirke P. Low rectal cancer: a call for a Primary perineal wound closure after preoperative
change of approach in abdominoperineal resection. radiotherapy and abdominoperineal resection has a
J Clin Oncol 2005; 23: 9257–64. high incidence of wound failure. Dis Colon Rectum
17. Eriksen MT, Wibe A, Syse A, Haffner J, Wiig JN. 2005; 48: 438–43.
Inadvertent perforation during rectal cancer resection 29. Khoo AK, Skibber JM, Nabawi AS, et al. Indications for
in Norway. Br J Surg 2004; 91: 210–6. immediate tissue transfer for soft tissue reconstruction
18. Pollack J, Holm T, Cedermark B, Holmstrom B, in visceral pelvic surgery. Surgery 2001; 130: 463–9.
Mellgren A. Long-term effect of preoperative radia- 30. Nisar PJ, Scott HJ. Myocutaneous flap reconstruction
tion therapy on anorectal function. Dis Colon Rectum of the pelvis after abdominoperineal excision. Colorec-
2006; 49: 345–52. tal Dis 2009; 11: 806–16.
19. Morris E, Quirke P, Thomas JD, Fairley L, Cot- 31. Mcallister E, Wells K, Chaet M, Norman J, Cruse W.
tier B, Forman D. Unacceptable variation in Perineal reconstruction after surgical extirpation of
­abdominoperineal excision rates for rectal cancer: time pelvic malignancies using the transpelvic transverse
to intervene? Gut 2008; 57: 1690–7. rectus abdominal myocutaneous flap. Ann Surg Oncol
20. Birbeck KF, Macklin CP, Tiffin NJ, et al. Rates of 1994; 1: 164–8.
circumferential resection margin involvement vary 32. Tobin GR, Day TG. Vaginal and pelvic reconstruction
between surgeons and predict outcomes in rectal with distally based rectus abdominis myocutaneous
cancer surgery. Ann Surg 2002; 235: 449–57. flaps. Plast Reconstr Surg 1988; 81: 62–73.
21. Radcliffe A. Can the results of anorectal (abdomi- 33. Christensen HK, Nerstrom P, Tei T, Laurberg S.
noperineal) resection be improved: are circumferential Perineal repair after extralevator abdominoperineal
resection margins too often positive? Colorectal Dis excision for low rectal cancer. Dis Colon Rectum 2011;
2006; 8: 160–7. 54: 711–7.

HEBK001-C08_p124-139.indd 139 08/02/13 5:54 PM


9
Laparoscopic surgery
Katharine E. Bevan and Tom D. Cecil

Introduction trials and concerns regarding long-term oncologi-


cal outcomes culminated in a logical reluctance
Since the 1940s surgeons have used a laparoscope to embrace laparoscopic colorectal cancer surgery.
to inspect the peritoneal cavity. Advances in equip- There are now a number of randomized control-
ment technology coupled with pioneering ap- led trials comparing laparoscopic and open colon
proaches have moved the role of the laparoscope cancer surgery. The short-term benefits for laparo-
from a diagnostic to an increasingly therapeutic scopic colonic resection have been summarized in a
tool in managing abdominal pathology. The first Cochrane review, with the main disadvantage being
appendicectomy was performed laparoscopically in that the surgery is more time-consuming.2 A subse-
the 1980s, closely followed by laparoscopic chole- quent Cochrane review reported that laparoscopic
cystectomy in 1985. Laparoscopic colonic resec- surgery was feasible for upper rectal cancers, but
tion, particularly for cancer, has been slower in the overall conclusion was that more randomized
development and uptake, however. This is unsur- controlled trials were required to assess long-term
prising, considering the technical challenges in- outcomes.3 A further Cochrane review principally
volved, including the necessity to operate in all four looking at the safety and efficacy of laparoscopic
quadrants of the abdomen and the complexity of versus open total mesorectal excision (TME) for
the arterial and venous anatomy of the colon. The rectal cancer concluded that ‘the limited evidence
need to resect and remove a sometimes substantial suggests that laparoscopic TME has clinically rel-
specimen, and subsequently perform a difficult in- evant short-term advantages in selected patients
testinal anastomosis, are further challenges. When with rectal cancer’.4 Controversy continues regard-
considering mid- to lower rectal cancers, there are ing the role of the laparoscope in rectal cancer as
additional anatomical and technical difficulties. initial reports were from enthusiasts with small
These include bulky tumours in a narrow pelvis, re- series, with little emphasis on the selection of ap-
traction and visualization (especially with the distal propriate patients or the challenges to be overcome
dissection), preservation of autonomic nerves, ac- before widespread adoption of safe laparoscopic
curate circumferential resection (without jeopard- rectal cancer surgery.5
izing the mesorectal envelope) and technological The Conventional versus Laparoscopic-Assisted
challenges (due to current limited angulation of Surgery in Colorectal Cancer (CLASICC) trial was
laparoscopic staplers). the first randomized controlled trial to include pa-
Early anxiety regarding port site metastasis1 tients with rectal cancers. Outcomes were evalu-
combined with a lack of randomized controlled ated separately from colon cancer outcomes. The

HEBK001-C09_p140-153.indd 140 08/02/13 5:50 PM


MODERN MANAGEMENT OF RECTAL CANCER AND THE ROLE OF THE LAPAROSCOPE  141

CLASICC trial highlighted concerns with regard of visceral metastases9 and useful for lymph node
to an increased circumferential resection margin assessment.10 Peritoneal carcinomatosis, especially
(CRM) positivity and suggested that the routine if localized and low volume,11 and to a lesser extent
use of the laparoscope for rectal cancer was not yet local tumour extension, continue to be a challenge to
justified.6 diagnose, however, despite major advances in imag-
In August 2006, the National Institute for Health ing. Thus, there continues to be a role for a more sen-
and Clinical Excellence (NICE) issued revised sitive modality such as laparoscopy, and in colorectal
guidelines recommending that laparoscopic sur- surgery this is particularly so for detection of early,
gery (including laparoscopically assisted surgery) potentially treatable localized carcinomatosis.12
should be offered as an alternative to open surgery A pivotal role for staging laparoscopy in colorectal
if both techniques are suitable and the appropriate cancer surgery is undoubtedly a useful initial inves-
surgical skills are available.7 tigation when a laparoscopic resection is being con-
This chapter focuses on some issues unique to templated or planned. The ultimate decision as to
laparoscopic rectal cancer surgery. We review the whether laparoscopic resection is safe and feasible,
evidence from randomized controlled trials and or whether open surgery is required, is best made at
summarize our approach to laparoscopic rectal this point, and experienced surgeons can rapidly de-
resection. The particular problems needed to be cide on the appropriate modality of surgery in most
addressed in rectal cancer are explored to give a cases.13
balanced and safe approach to the use of the laparo-
scope in rectal cancer and in particular TME.
Laparoscopic Stoma Formation

MODERN MANAGEMENT OF Patients with rectal cancer may, in some circumstanc-


RECTAL CANCER AND THE ROLE es, require a defunctioning stoma, either as palliative
OF THE LAPAROSCOPE treatment or before neoadjuvant therapy for very
symptomatic locally advanced tumours. A stoma may
The modern management of rectal cancer involves be necessary for obstruction, uncontrollable diar-
accurate preoperative staging, appropriate neoad- rhoea, tenesmus or bleeding and often before chemo-
juvant therapy in selected cases (either preoperative radiation, as either down-staging or definitive care.
radiotherapy or chemoradiotherapy), optimal sur- There is considerable variability in the frequency of
gical excision and detailed pathological assessment defunctioning stoma formation before neoadjuvant
of the excised specimen.5 All these aspects interact therapy,14 but it undoubtedly has a role and may be
with the role of the laparoscope in the management needed before or during neoadjuvant therapy.15
of rectal cancer. The laparoscope can be used to perform a defunc-
In the absence of longer-term outcome data from tioning ileostomy or colostomy alone, or as part of
larger clinic trials, a surrogate marker is required to a laparoscopic colorectal resection, or to reduce the
assess operative success. Macroscopic and micro- septic complications secondary to anastomotic leak.
scopic assessment of the specimen, being a prog- In addition, the laparoscopic approach should en-
nostic marker for local recurrence as reported in the able correct identification of the proximal and distal
Medical Research Council (MRC) CR07 trial, may limbs and thus avoid ‘bringing out the wrong limb’,
be the best method for comparing early oncological as surgeons of an older generation have experienced
results from both open and laparoscopic surgery.8 when fashioning a defunctioning trephine stoma.
Some reports suggest shorter time to passage of
flatus and faeces, earlier resumption of a liquid and
Staging and Diagnostic Laparoscopy solid diet, decreased morphine requirements, a re-
duction in 30-day morbidity, and earlier discharge
Despite advances in non-invasive imaging, laparos- in patients having a stoma formed ­laparoscopically.16
copy continues to have an important role in some As a consequence, patients in whom defunctioning
aspects of staging rectal cancer. Cross-sectional im- alone is required, without resection or anastomo-
aging is highly sensitive and specific for detection sis, are likely to achieve maximal benefit from a

HEBK001-C09_p140-153.indd 141 08/02/13 5:50 PM


142  Laparoscopic surgery

laparoscopic approach due to expedited recovery Evidence for the


and therefore minimal interruption to treatment, laparoscopic approach
in comparison with open surgery. There have been to rectal cancer
reports, however, of obstructive complications after
any proximal defunctioning stoma. Short-Term Outcomes
A laparoscopic colostomy is also a very useful
training operation as part of the learning curve for Although there are some conflicting findings, many
more complex surgery involving mobilization of of the short-term benefits of laparoscopic colonic
the colon. resection have also been reported for laparoscopic
rectal resection.4 These include the earlier return
of bowel function and subsequently resumption of
Abdominoperineal Excision diet,6,20–23 shorter length of stay,6,20,23 quicker return
of the Rectum to household activities,20 reduced pain and analge-
sia use,20,21 and decreased blood loss,21–23 sometimes
Abdominoperineal excision (APE) was the first re- equating to a reduction in the requirement for
sectional laparoscopic colorectal procedure to be blood transfusion (Table 9.1).23
performed for malignancy. As the oncological part These findings are unsurprising and are consist-
of the operation is carried out by an open surgi- ent with the evidence of a lesser inflammatory re-
cal technique from the perineum, it could be con- sponse and decreased immunological disturbance
fidently attempted without the concern for a major in laparoscopic colorectal surgery; some of these
compromise in the oncological outcome. Patients beneficial effects may be due to less small bowel
with low rectal cancer undergoing APE, particu- manipulation.24 On the negative side, the duration
larly with the recent developments in extralevator of the laparoscopic operation is longer6,20–23 and the
APE, have perhaps most to gain by a laparoscopic cost of surgery greater.2,20,23
approach.17 In extralevator APE, the main onco-
logical part of the procedure is performed via the
perineum, there is no bowel anastomosis, and the Lymph Node Harvest
specimen can be removed through the perineum,
such that no abdominal incision is required.17 Lymph node harvest has often been considered a short-
The upper part of the TME plane is developed term marker for oncological equivalence of open and
and completed laparoscopically, but without the laparoscopic resection specimens. A small randomized
need to continue the dissection very far distally. controlled trial included 73 patients (39 open surgery,
Subsequently, the perineal plane is developed, usu- 34 laparoscopic TME) and reported equivalent lymph
ally after completing the colostomy, closing the node harvest in both groups.25 Subsequently other
abdomen and turning the patient into the prone studies have reported similar findings.6,20–23
jack-knife position. The dissection proceeds until
the perineal operation meets the extent of the ab-
dominal operation. Additionally, the tumour re- RESECTION QUALITY AND
quiring an APE is generally lower in the rectum, CIRCUMFERENTIAL
well below the peritoneal reflection, compared with RESECTION MARGIN
cases where reconstruction by an anterior resection
is being performed, and is therefore less likely to It is now widely accepted that TME, as described by
obscure the laparoscopic view or be subject to tu- Heald in 1988, is the treatment of choice in ­resectable
mour rupture by traction or retraction. rectal cancer.26 Provided a clear CRM can be achieved,
With the success of laparoscopic APE, enthusi- TME results in an improvement in cancer-specific
asts deemed it as a natural progression to extend the survival. The effect of the quality of the resected speci-
abdominal surgery deeper into the pelvis in suitable men is also emerging as a prognostic indicator for lo-
patients to mobilize the rectum and subsequently cal recurrence after rectal cancer.
proceed to reconstruction to achieve laparoscopic One group showed that both the prospec-
anterior resection.18,19 tive histopathological assessment of the plane of

HEBK001-C09_p140-153.indd 142 08/02/13 5:50 PM


RESECTION QUALITY AND CIRCUMFERENTIAL RESECTION MARGIN   143

Table 9.1 Short- and longer-term outcome results reported in the five largest randomized controlled trials comparing
laparoscopic and open surgery for rectal cancer.
Study Kang et al.21 Lujan et al.22 Braga et al.23 Jayne et al.29 Leung et al.20
Open surgery (n) 170 103 85 253 200
Laparoscopic 170 101 83 128 203
surgery (n)
Blood loss Greater in open Greater in open Greater in open NS NS
group, significant group, P 5 0.001 group, P 5 0.0002
Operative time .48 min in .21 min in .53 min in Increased in .45.7 min in
laparoscopy laparoscopy laparoscopy laparoscopy laparoscopy
group, P , 0.0001 group, P 5 0.020 group, P 5 0.0001 group group, P , 0.001
Conversion rate (%) 1.2 7.9 7.2 34 (rectal) 23.2
Analgesic Reduced in NR NR NR Reduced in
requirements laparoscopy laparoscopy
group, P , 0.0001 group, P , 0.001
Return to bowel Reduced time in NR NR NS NS
function laparoscopy
group, P , 0.0001
Return to diet Reduced time in ,1 day in Reduced time in NS Reduced time in
laparoscopy laparoscopy laparoscopy laparoscopy
group, P , 0.0001 group, NS group, P , 0.0001 group, P , 0.001
Length of stay ,1 day in ,1 day in ,3.6 days in ,2 days in Reduced in
laparoscopy laparoscopy laparoscopy laparoscopy rectal laparoscopy
group, NS group, NS group, P 5 0.004 group group, P , 0.001
Lymph node yield NS NS NS NS NS
CRM positivity (%) NS NS NS 12 (laparoscopy) v. NR
6 (open)
30-day morbidity NS NS NS NS NS
Mortality NS NS NS NS NS
Local recurrence at NR 5.3 (open) v. 4.8 NS at 3 years 10.1, NS 6.6 (laparoscopy)
5 years (%) (laparoscopy), NS v. 4.1 (open), NS
Disease-free survival NR 81 (open) v. 84 NS 52.1 (open) v. 53.2 NS
at 5 years (%) (laparoscopy), NS (laparoscopy), NS
Overall survival at NR 75 (open) v. 72 NS 52.9 (open) v. 60.3 NS
5 years (%) (laparoscopy), NS (laparoscopy), NS
Mean follow-up NR 34 (open) v. 32 53.6 56.3 52.7
(months) (laparoscopy)

CRM, circumferential resection margin; NR, not reported; NS, not statistically significant.

surgery and the CRM margin were independent of dissection.27 It would therefore seem appropri-
prognostic markers for local recurrence. Clear dif- ate to use these two standards (plane of surgery
ferences were noted with suboptimal surgery, with achieved and CRM) as a means of quality control
a statistically significant increase in the local re- when comparing laparoscopic and open rectal
currence rate at 3 years. Circumferential resection cancer surgery.
margin positivity is still one of the most important The evidence base for equivalent clinical out-
predictors of increased risk of local recurrence and comes in rectal cancer treated laparoscopically is
decreased 3-year survival, regardless of the plane limited but emerging.

HEBK001-C09_p140-153.indd 143 08/02/13 5:50 PM


144  Laparoscopic surgery

The CLASICC study initially demonstrated con- resections.30,31 A comparison of the macroscopic
cerns with regard to CRM. The study reported more quality of specimens after laparoscopic32 and open33
positive CRMs in the laparoscopic group compared TME showed that there was a more complete TME
with the open anterior resection group (12 per cent v. with intact visceral pelvic fascia in the laparoscopic
6 per cent). Although the results were not significantly group. The investigators concluded that the im-
different, they were concerning enough to recommend proved visualization at laparoscopy may result in
caution when treating rectal cancer ­laparoscopically.6 improved macroscopic specimens.34
These concerns have been somewhat allayed by the 3- Although the rates of local recurrence vary wide-
and 5-year follow-up data, which have shown that the ly in the published literature, no significant differ-
trend towards positive CRM did not translate into an ences between laparoscopic and open surgery were
increase in local recurrence rates.28,29 reported in the 2006 Cochrane review4 or in sev-
Five randomized controlled trials, all with at least eral randomized controlled trials.20,22,23 It remains to
80 patients in each arm, have reported longer-term be seen whether long-term follow-up consistently
follow-up data. Three of these trials reported on gives results equivalent to those reported for open
rectal cancer outcomes alone.21–23 One trial included TME of less than 5 per cent in curative cases.35
both colonic and rectal cancer and reported on the
rectal subset separately.6 The final trial recruited both
high rectal and sigmoid tumours, but the heteroge- Anastomotic leak and
neity makes interpretation difficult, particularly as its influence on local
the tumours were not analysed independently.20 recurrence
None of these trials demonstrated a statistically sig-
nificant difference in CRM positivity (Table 9.2). An anastomotic leak after restorative resection of
Kang’s group looked at macroscopic quality of rectal cancer continues to be a major issue whether
the TME dissection and showed no difference be- laparoscopic or open surgery is performed. There
tween open and laparoscopic surgery. Three of have been reports that anastomotic leak may sig-
the randomized controlled trials have published nificantly increase the local recurrence rates,36 with
long-term survival figures.6,22,23 These studies have poorer overall and cancer-specific survival.37 There-
shown no statistical difference in local recurrence, fore, it is imperative that we have at least equal or
­disease-free survival or overall survival between preferably lower anastomotic leak rates when per-
surgical approaches (see Table 9.1). forming laparoscopic surgery for rectal cancer.
Some groups have compared the quality of the None of the randomized controlled trials showed a
mesorectal dissection specimen and reported no statistically significant difference between the open
statistical difference between open and laparoscopic and laparoscopic groups when comparing leak rates

Table 9.2 Comparison of leak rate within randomized controlled trials comparing laparoscopic and open surgery for
rectal cancer.
Trial Kang et al.21 Lujan et al.22 Braga et al.23 Jayne et al.29 Leung et al.20
Open (n) 170 103 85 132 200
Laparoscopic (n) 170 101 83 160 202
Inclusions ,9 cm from anal Mid- and low rectal ,15 cm from anal .5 cm from anal Rectosigmoid
verge cancers: mean verge verge (67%
6.2 cm (open) v. open v. 79%
5.5 cm (laparoscopy) laparoscopy)
from anal verge
Stoma formation (%) 88.4 (open) v. 91 59 (open) v. 62 24.7 (open) v. 26.5 NR NR
(laparoscopy) (laparoscopy) (laparoscopy)
Leak rate (%) 0 (open) v. 1.2 12 (open) v. 6 7 (open) v. 6 7 (open) v. 8 2 (open) v. 0.5
(laparoscopy) (laparoscopy) (laparoscopy) (laparoscopy) (laparoscopy)

NR, not reported.

HEBK001-C09_p140-153.indd 144 08/02/13 5:50 PM


OUR APPROACH TO LAPAROSCOPIC ANTERIOR RESECTION  145

(see Table 9.2).6,20–23 Liberal use of defunctioning, ei- equivalence.6,20–23 Taken in conjunction with studies on
ther in all patients having TME or at a minimum all colonic resection,2,40 the available data suggest possible
patients at increased risk of anastomotic leak (e.g. benefits in favour of laparoscopic surgery in frailer and
male patients, patients with a high body mass in- older patients due to fewer complications in the lapar-
dex, patients who have had neoadjuvant treatment, oscopic group, especially in people aged over 70 years.40
patients with a low or complex anastomosis), may
significantly reduce the clinical consequences of a
leak. There may be a case for routine defunction- OUR APPROACH TO
ing of all anastomoses after any mid- or low rec- LAPAROSCOPIC ANTERIOR
tal cancer resection, as in the Comparison of Open RESECTION
versus Laparoscopic Surgery for Mid and Low Rec-
tal Cancer after Neoadjuvant Chemoradiotherapy Preoperative Assessment
(COREAN) study.21 It has been noted, however, that
the addition of a defunctioning stoma significantly The preoperative investigations that we carry out for
increases length of stay after laparoscopic resection, laparoscopic anterior resection for rectal cancer are al-
thereby partly negating this potential benefit of the most identical to those for open surgery. Staging com-
laparoscopic approach.21 puted tomography (CT) of the chest and abdomen
and magnetic resonance imaging (MRI) of the pelvis
are essential. Endoscopic assessment of the entire co-
Length of stay lon (unless an obstructing tumour limits progress)
allows biopsy confirmation of carcinoma and allows
Reduction in length of stay is often quoted as being assessment for synchronous pathology. Unlike open
a particular benefit of laparoscopic surgery. Inter- surgery, tattooing of the location of the tumour is es-
estingly, two of the five randomized controlled tri- sential. For colon cancers above the peritoneal reflec-
als did not demonstrate any statistically significant tion, we recommend marking 5 cm distal to the tu-
reduction in length of stay (see Table 9.1).21,22 Braga mour, and usually in three separate locations around
and colleagues did report a reduction in length of the circumference of the bowel at this level. For rectal
stay with laparoscopic surgery, but this study in- cancer, however, this is unhelpful, as the tumour is be-
cluded high rectal cancers and had a subsequently low the peritoneal reflection and the tattoo can stain
low stoma rate.23 Leung and colleagues also demon- the mesorectal and fascial planes, making dissection
strated a reduction in length of stay, but their study more difficult. For rectal cancer, we rely on MRI as-
included mainly rectosigmoid ­tumours.20 sessment and digital examination or rigid sigmoidos-
The studies of Kang and colleagues21 and Lujan copy for localization of the tumour at surgery.
and colleagues22 showed no significant reduction in We recommend full bowel preparation for any total
length of stay. Of note, all patients in the COREAN mesorectal resection to prevent ongoing pelvic sepsis
trial and over 70 per cent of patients in the Spanish in a defunctioned patient from the column of fae-
trial were operated on after neoadjuvant chemora- ces that will be left from an unprepared colon. Care
diotherapy, and consequently the rates of defunc- needs to be taken to ensure that this is carefully docu-
tioning ileostomy were very high (91 per cent and mented and communicated to the ward and nursing
62 per cent, respectively), which has previously staff, as bowel preparation is not routine, especially
been shown to increase length of stay.37 for patients in enhanced recovery programmes. All
our patients having open or laparoscopic TME are
counselled and sited preoperatively for a defunction-
Morbidity and mortality ing right-sided ileostomy and left iliac fossa colostomy.

The currently reported randomized controlled trials


have not shown a significant difference with regard Operation Table Positioning
to perioperative mortality.20–23,28 Some studies have
shown lower morbidity in laparoscopic patients,38,39 The patient is placed supine on an electrically oper-
but this is a minority and many studies have shown ated table, with the legs in a modified Lloyd Davies

HEBK001-C09_p140-153.indd 145 08/02/13 5:50 PM


146  Laparoscopic surgery

Figure 9.1 Modified Lloyd Davies


position of legs.

position. The legs are placed in leg supports, often the umbilicus. This minimizes both gas leak and the
requiring additional foam padding behind the calf. distance of extension of the port into the abdomen.
This may be placed laterally to reduce the risk of A second 10–12-mm port is placed just medial and
common perineal nerve compression. The legs are superior to the anterior superior iliac spine, with
abducted with the knees at shoulder width apart a 5-mm port at least a hand’s breadth cephalad. A
and flexed to approximately 708. To prevent the further 5-mm port is placed laterally to the umbili-
right thigh getting in the way of the right hand of cus on the left side of the patient. This is used as an
the operating surgeon, the hips are placed in neu- assisting port to provide retraction to the sigmoid
tral or slight extension (Figure 9.1). colon, and as an operating port (Figure 9.2).
All patients have bladder catheterization and a Extra ports are introduced as necessary. A 10-mm
nasogastric tube. suprapubic port at the specimen extraction site can
be useful if the small bowel is difficult to position,
as the camera can be used through this port to give
Port Insertion and Positioning a good view of the omentum and small bowel in
the upper abdomen and right upper quadrant. In
The surgeon stands to the patient’s right, with the addition, this port can be used to provide addi-
camera holder on the same side to the left of the tional retraction in the pelvis. A further right upper
operating surgeon. The assistant stands to the pa- quadrant port may be necessary to retract the small
tient’s left. For mobilization of the splenic flexure, bowel or help with splenic flexure mobilization in
it is often useful for the operating surgeon to stand large patients (see Figure 9.2).
between the patient’s legs and the assistant to stand
to the patient’s right.
We commonly use four ports for an anterior re- Laparoscopy
section. The first port is normally placed just below
the umbilicus. Occasionally, if the distance from The first step is to perform a diagnostic laparoscopy
umbilicus to symphysis pubis is short, this port may and assess the abdomen. This involves evaluation
be placed above the umbilicus. This port is placed of:
using an open or Hasson technique to minimize the
risk of injuring intra-abdominal viscera upon in- l the tumour itself;
sertion. A 10-mm balloon port is our preference at l the surface of the liver for liver metastasis;

HEBK001-C09_p140-153.indd 146 08/02/13 5:50 PM


OUR APPROACH TO LAPAROSCOPIC ANTERIOR RESECTION  147

Transverse Colon

IMV
5

5 DJ flexure
12 5

12

Figure 9.3 Medial approach to the splenic flexure.


12

Figure 9.2 Standard ports are shown in blue and optional


(Figure 9.5). This allows the transverse meso-
extra ports in green. colon to be mobilized off the pancreas. The vein is
then lifted and the two areas of dissection can be
connected to allow a medial to lateral dissection
out over Gerota’s fascia and the pancreas up to the
l the peritoneal surfaces and abdominal cavity
splenic flexure.
for small volume peritoneal disease;
The omentum is then detached from the trans-
l the splenic flexure to assess the need for mobi-
verse colon, which can usually be achieved by sharp
lization. Generally we have a low threshold for
dissection with scissors and diathermy. In patients
fully mobilizing the splenic flexure and would do
who are obese, it may be easier to divide the gas-
this initially as in open surgery. We do not feel
trocolic ligament with LigaSure™, starting medially
that mobilization is obligatory for all patients,
and entering the lesser sac from above and moving
e.g. in the case of a long sigmoid loop, it may be
out laterally.
feasible to omit splenic flexure mobilization.
Finally, the lateral attachments of the descending
colon and splenic flexure are divided to complete
Splenic Flexure Mobilization the mobilization.
Splenic flexure mobilization can be challeng-
Positioning ing, and sometimes moving the point of dissection
The patient is positioned flat or in the reverse Trende-
lenburg position to allow the small bowel to fall into
the pelvis. The omentum is retracted cephalad, above Ascending left colic
Pancreas vein
the liver and spleen. Some right tilt can be useful to
encourage the small bowel mesentery to fall to the
right to allow identification of the duodenal-jejunal
flexure and inferior mesenteric vein (Figure 9.3).
IMV
Approach
The inferior mesenteric vein is mobilized and divid-
ed above the ascending left colic vein as it dives be-
low the pancreas. We use either clips or ­LigaSure™
bipolar diathermy to divide the vein (Figure 9.4).
The lesser sac is opened above the pancreas and
lateral to the middle colic vessels. Figure 9.4 High division of the IMV.

HEBK001-C09_p140-153.indd 147 08/02/13 5:50 PM


148  Laparoscopic surgery

then required to allow gravity to assist the small


transverse mesocolon bowel in falling out of the pelvis.
l The small bowel mesentery needs to be ‘layered’,

right and cephalad. We achieve this with the


blunt graspers (Johan’s type) closed, using blunt
manipulation of the leaves of the small bowel
lesser sac mesentery.
pancreas l The terminal ileum and appendix may need to

be mobilized if they are holding the small bowel


in the pelvis.
l If the small bowel persists in dropping into the

field of view, a tonsil swab can be helpful in


providing additional retraction. If this fails, a
Figure 9.5. Medial approach to the lesser sac. right upper quadrant port can allow a grasper
to act as a retractor across the right iliac fossa.
Finally, if small bowel positioning is still elusive,
­ etween a combination of medial, lateral and superior
b
placing the camera through a 10- or 12-mm
approaches is required to continue to make progress.
suprapubic port allows a better view of the right
An alternative approach, when splenic flexure
upper quadrant and more accurate positioning
mobilization is considered imperative, is to start
of the small bowel and omentum.
the operation with the patient in the right lateral
position. If this is the preferred approach, several Approach
pointers that may be helpful are as follows:
The inferior mesenteric artery (IMA) is approached
l Mark the first port position in the left upper from a medial direction. To facilitate visualization
quadrant midclavicular line just below the cos- of the IMA pedicle as it arises from the aorta, the
tal margin while the patient is prone. Particu- sigmoid colon is retracted superiorly and cephalad.
larly in a patient with a pendulous abdomen, Avoid handling the bowel directly by grasping an
the port placement site can be difficult to assess appendix epiploicae for retraction.
when the patient is in the right lateral position.
l Have the patient positioned at about 70–808
Dissection of the pedicle
(not at 908). The pelvic brim is identified and the dissection begun
l Use two back supports at the level of the at the sacral promontory. The peritoneum is incised
scapula and the anterior superior iliac spine. below and parallel to the ileocolic pedicle. Our prefer-
l Position the monitor above and lateral to the ence is to mark the peritoneal dissection plane with
patient’s left shoulder. closed diathermy scissors. The vascular space is then
l The right knee and hip should be in slight flexion. opened using small downward movements with the
l Cushion between the patient’s legs. scissors. The peritoneal incision is extended in a ce-
phalad direction over the origin of the IMA. The aim
is to separate the mesenteric fat from the retroperito-
Approach to the Inferior neum. Continuing in this direction, Toldt’s plane will
Mesenteric Artery Pedicle be reached (the plane between the posterior mesoco-
lon and the anterior retroperitoneal fascia). Once in
Positioning this plane, it is our preference to use a blunt dissec-
tor (e.g. with 5-mm LigaSure™) to extend the dissec-
The patient is placed in a steep Trendelenburg posi-
tion laterally towards the abdominal wall. The correct
tion with the right lateral tilt left in place. The small
plane should be avascular and allows the hypogastric
bowel needs to be moved into the right upper quad-
nerves, gonadal vessels and ureter to be dropped back
rant. Several tricks and tips can facilitate this:
posteriorly (Figure 9.6). If the bare belly of the psoas
l The omentum needs to be as cephalad as pos- muscle is visible, then the dissection plane is deep to
sible; the maximum degree of Trendelenburg is the ureter. It is imperative that the ureter is positively

HEBK001-C09_p140-153.indd 148 08/02/13 5:50 PM


OUR APPROACH TO LAPAROSCOPIC ANTERIOR RESECTION  149

mesorectal fascial plane. In laparoscopic surgery,


traction and counter-traction on the mesorectum, or
IMA near the tumour, have to be performed with instru-
ments and may be more traumatic (less soft and sen-
sitive) than an expert surgeon’s hand. This may result
in transgression of the ‘holy plane’, which has been
shown to be associated with poorer outcomes.33,35
left ureter Using a tonsil swab underneath a Johan’s retrac-
tor can facilitate atraumatic retraction. Once the
plane is opened up, air will enter the areolar plane
branches of hypogastic and allow sharp dissection with either scissors or
plexus
hook diathermy. At all time, retraction needs to be
gentle and the peritoneal edges should be used for
retraction. The uterus, or the peritoneum above the
Figure 9.6 Mobilizing the inferior mesenteric artery pedicle. seminal vesicles, can be stitched up to the anterior
abdominal wall to facilitate retraction if necessary.
identified before division of the pedicle. If this has not In addition, in female patients a ‘swab-on-a-stick’
been possible, then a lateral approach may be helpful. in the vagina retracting it anteriorly may aid visu-
It is our preference to carefully dissect out the IMA alization of the planes. It is important at all times
at its origin from the aorta and then apply two large during mobilization to be aware of the position of
Haemolock™ clips (gold). Dividing between the the tumour. A combination of digital examination
clips can be done with or without an energy device. If before and during the procedure, and a review of
the pedicle is particularly bulky, it may be necessary the preoperative MRI, are essential to compensate
to divide it with a laparoscopic stapler. A tonsil swab for the loss of tactile sensation at laparoscopic sur-
can be left at the most lateral point of dissection, gery compared with open surgery.
above the ureter, to identify the plane when atten-
tion is turned to the lateral attachments. Following Technique
division of the IMA, the dissection can be continued The peritoneum can then be incised towards the
in a cephalad direction (in Toldt’s plane) towards the pelvis, over the pelvic brim. Here it is important to
splenic flexure. A second low division of the inferior identify the ‘holy plane’ or mesorectal fascial plane
mesenteric vein is then performed for complete mo- (Figure 9.7). The rectum is initially retracted anteri-
bilization. Again, if in the correct plane, Gerota’s fas- orly. This should be achieved with minimal trauma,
cia will be left intact and swept posteriorly. Dissection particularly at the level of the tumour.
is continued in this direction until the physical extent
of dissection is limited. Before taking down the lat-
eral attachments, ensure that the plane has also been rectum
developed as far as is possible in an inferior direction.
To release the lateral attachments, the sigmoid colon
will need to be retracted in a medial direction. If a
tonsil swab was left in situ, it will be visible as the at-
tachments are taken down and the plane identified.
If the medial to lateral dissection was adequate, this TME plane

will be a thin layer.

Mobilization of the Rectum


hypogastric plexus
Principles
The concept of TME revolves around retraction Figure 9.7 Entering the total mesorectal excision plane at
and counter-traction26 to facilitate dissection in the the top of the pelvis.

HEBK001-C09_p140-153.indd 149 08/02/13 5:50 PM


150  Laparoscopic surgery

uterus rectum
rectum retracted to left
swab retractor

holy plane

right hypogastric TME plane at back


nerve

Figure 9.8 Dropping back the right hypogastric nerve. Figure 9.10 Use of a swab as retractor.

Care must be taken not to damage the hypogas- lateral to left lateral or anteriorly to facilitate
tric nerves as they descend over the sacral promon- distal dissection.
tory into the pelvis. Dissection is continued in the Anteriorly, in both male and female patients, a cuff
TME plane posteriorly and laterally, focusing ini- of anterior peritoneum should be preserved by con-
tially on the right side (Figure 9.8). tinuing the dissection from the left and right sides and
When adequate mobilization on the right and meeting in the middle. This acts as a safe area to grasp
posteriorly has been achieved, the peritoneum on with a retractor and allows the surgeon to find the
the left can be divided carefully, dropping back the correct plane behind the seminal vesicles in male pa-
left hypogastric nerve (Figure 9.9). tients and the vagina in female patients (Figure 9.11).
The rectum needs to be fully mobilized poste- On the lateral pelvic side-wall the nervi erigentes
riorly with division of Waldeyer’s fascia and mo- and inferior hypogastric plexus are dropped off
bilized off the pelvic floor. The same principles the rectum. The dissection should continue until
apply as in open surgery, namely working down 1–2 cm clear of the distal extent of the tumour, with
the back of the rectum and coming round to the at least 5 cm of mesorectum below the tumour or
sides with traction and counter-traction, often a total mesorectal excision. For TME, the rectum
with a tonsil swab demonstrating the planes needs to be completely mobilized off the pelvic
(Figure 9.10). As in open surgery, it is necessary floor and on to the muscle tube as it enters the in-
to move the focus of the dissection from right tersphincteric groove (Figure 9.12).

uterus

left hypogastric nerve vagina

rectum
cul de sac

Figure 9.9 Dividing peritoneum on the left. Figure 9.11 Dissecting the rectovaginal plane.

HEBK001-C09_p140-153.indd 150 08/02/13 5:50 PM


CONCLUSION  151

may be impossible to angulate the stapler across


the rectum. The inability to accurately palpate and
inferior hypogastric plexus
correctly identify the lower limit of the tumour
makes mesorectal transection for lower tumours
technically difficult. If there is still uncertainty, a
vagina rigid sigmoidoscope, or digital examination in low
tumours, can be used to accurately determine the
point of transection.
If there is still uncertainty despite these manoeu-
vres, then it is our preference to insert the linear sta-
pelvic floor pler via a Pfannenstiel incision; this incision is also
rectum used as the specimen extraction site. If the rectum
has been fully mobilized, the stapler can be applied
easily through an 8-cm incision and also facilitates
Figure 9.12 Approaching the intersphincteric plane on the formation of a defunctioning ileostomy.
left.

Specimen Extraction
Division of the Rectum
and Anastomosis To facilitate specimen location and retrieval, we
leave a Johan’s retractor attached to the divided
It is our practice to strive to perform the same op- bowel end. If not performed already for cross-sta-
eration laparoscopically as at open surgery, and pling, our preferred extraction site is via a Pfan-
therefore a rectal washout after cross-clamping and nenstiel incision. The patient should be left in the
before rectal transection is imperative. Although head-down position while the abdominal inci-
washout is a simple procedure at open surgery, it sion is made. A wound retractor is inserted; we
is more complex laparoscopically due to difficulty currently favour the Alexis (Applied Medical)™
in occluding the lumen distal to the tumour. A wound system. The bowel is removed and the
number of methods have been reported, including proximal resection point identified. The anvil of
the use of a bulldog clip or Roeder knot or passing a the circular stapler can be inserted in the normal
piece of intravenous line tubing (or similar) via the fashion, before dropping back into the abdomen,
left iliac fossa port, around the rectum and using restoring the pneumoperitoneum and performing
this as an occlusive tourniquet.41 These techniques the anastomosis.
are technically challenging in many cases and po-
tentially result in the procedure being abandoned
and washout not performed. CONCLUSION
Following washout, the rectal transection can be
performed. Rectal transection proximal to the pel- In summary, with regard to the ‘abdominal’ part
vic floor can be challenging, as current laparoscopic of a rectal cancer operation, it is likely that a
staplers can angulate to a maximum of only 658. As laparoscopic minimal-access technique is feasible
in open surgery, the transection should be perpen- and beneficial, in the short term, although operat-
dicular to the rectal muscle tube. Ideally, this should ing time may be prolonged. Equipment costs may
be achieved with a single firing of the stapler, but be greater, but the shorter hospital stay and faster
practically it often requires two or three firings. Not recovery may more than compensate for these
only is this expensive and time-consuming, but it additional initial expenses.
can also result in ‘dog ears’ or a zigzag transection Initial randomized controlled trials report
staple line, raising the possibility of ischaemic areas equivalence in longer-term oncological outcomes,
that may subsequently increase the risk of leak. Fur- with some groups reporting comparable local re-
thermore, within the confines of the narrow male currence rates. The advantage of magnification of
pelvis, especially for a mid- or low rectal cancer, it the TME plane may give better visualization and

HEBK001-C09_p140-153.indd 151 08/02/13 5:50 PM


152  Laparoscopic surgery

s­ ubsequently better macroscopic quality of the   3. Kuhry E, Schwenk W, Gaupset R, Romild U, Bonjer
specimen, particularly when surgeons and operative HJ. Long-term results of laparoscopic colorectal can-
cer resection. Cochrane Database Syst Rev 2008; (2):
teams have overcome the learning curve and gained
CD003432.
experience. Ultimately, the individual surgeon’s ap-   4. Breukink S, Pierie J-P, Wiggers T. Laparoscopic versus
proach to rectal cancer surgery will be determined open total mesorectal excision for rectal cancer.
by many factors. Each case has to be assessed on an ­Cochrane Database Syst Rev 2006; (4): CD005200.
individual basis, taking into account patient fac-   5. Marsden MR, Parvaiz A, Moran B. Resection of rectal
tors, anatomical considerations and the experience cancer: laparoscopy or open surgery? Ann R Coll Surg
of the individual surgeon. The surgeon should also Engl 2010; 92: 106–12.
  6. Guilloi PJ, Quirke P, Thorpe H, et al. Short-term end-
be willing to change the approach intraoperatively
points of conventional versus laparoscopically-assisted
if there is any concern about the oncological out- surgery in patients with colorectal cancer (MRC
come being compromised. Each surgeon should CLASICC trial): multicentre, randomised controlled
audit their laparoscopic rectal cancer outcomes and trial. Lancet 2005; 365: 1718–26.
compare these with their outcomes from open sur-   7. National Institute for Health and Clinical Excellence.
gery. Longer-term outcome data from clinical tri- Colorectal Cancer: Laparoscopic Surgery (Review).
als are awaited to answer some ongoing issues, such Technology appraisal TA105. London, National
­Institute for Health and Clinical Excellence, 2006
as long-term cure rates, incisional hernia rates and
(http://www.nice.org.uk/Guidance/TA105).
postoperative adhesions.   8. Quirke P, Steele R, Monson R, et al. Effect of the plane
The data suggest that optimal oncological colorectal of surgery achieved on local recurrence in patients
cancer resection can be performed laparoscopically with operable rectal cancer: a prospective study us-
in selected patients. Nevertheless, there are still some ing data from the MRC CR07 and NCIC0CTG CO16
ongoing concerns regarding laparoscopic low ante- randomised clinical trial. Lancet 2009; 373: 821–8.
rior resection. The group that described the first   9. Robinson PJ. Imaging liver metastases: current limitations
and future prospects. Br J Radiol 2000; 73: 234–41.
rectal cancer resection have reported higher than
10. Klerkx WM, Bax L, Veldhuis WB, et al. Detection of
expected local recurrence rates in Union for Inter- lymph node metastases by gadolinium–enhanced
national Cancer Control (UICC) stage III disease.42 magnetic resonance imaging: systematic review and
Achieving good oncological outcomes for mid- and meta-analysis. J Natl Cancer Inst 2010; 102: 244–53.
low rectal cancer is a continuing challenge, regard- 11. Dromain C, Leboulleux S, Auperin A, et al. Staging of
less of whether the approach is open or laparoscop- peritoneal carcinomatosis: enhanced CT vs. PET/CT.
ic. Good-quality surgery, according to the experi- Abdom Imag 2008; 33: 87–93.
12. Daniels IR, Fisher SE, Heald RJ, Moran BJ. Accurate
ence and technical ability of the surgeon, should be
staging, selective preoperative therapy and optimal
the gold standard. Whether the marginal short-term surgery improves outcome in rectal cancer: a review of
benefits of the laparoscopic approach justify the po- the recent evidence. Colorectal Dis 2007; 9: 290–301.
tential oncological downside remains controversial.5 13. Neudecker J, Klein F, Bittner R, et al. Short-term out-
In rectal cancer surgery, safe retraction and counter- comes from a prospective randomized trial comparing
traction, an optimal view, good-quality adherence to laparoscopic and open surgery for colorectal cancer.
the TME ‘holy plane’ principles, optimal mesorec- Br J Surg 2009; 96: 1458–67.
14. Morton DG, Sebag-Montefiore D. Defunctioning sto-
tal excision or transection, and rectal division and
mas in the treatment of rectal cancer. Br J Surg 2006;
anastomosis remain the aim in this common, often 93: 650–1.
complex cancer. 15. Koea JB, Guillem JG, Conlon KC, Minsky B, Saltz L,
Cohen A. Role of laparoscopy in the initial multimo-
dality management of patients with near-obstructing
References rectal cancer. J Gastrointest Surg 2000; 4: 105–8.
16. Young CJ, Eyers AA, Solomon MJ. Defunctioning of
  1. Berends FJ, Kazemier G, Bonjer HJ, Lange JF. Sub- the anorectum: historical controlled study of laparo-
cutaneous metastases after laparoscopic colectomy. scopic vs. open procedures. Dis Colon Rectum 1998;
Lancet 1994; 344: 58. 41: 190–4.
  2. Schwenk W, Haase O, Neudecker JJ, Müller JM. 17. West NP, Finan PJ, Anderin C, Lindholm J, Holm T,
Short term benefits for laparoscopic colorectal Quirke P. Evidence of the oncological superiority of
resection. Cochrane Database Syst Rev 2005; (2): cylindrical abdominoperineal excision for low rectal
CD003145. cancer. J Clin Oncol 2008; 26: 3517–22.

HEBK001-C09_p140-153.indd 152 08/02/13 5:50 PM


References  153

18. Morino M, Parini U, Giraudo G, Salval M, Brachet 31. Breukink SO, Grond AJ, Pierie JP, Hoff C, Wiggers T,
Contul R, Garrone C. Laparoscopic total mesorectal Meijerink WJ Laparoscopic vs open total mesorectal
excision: a consecutive series of 100 patients. Ann excision for rectal cancer: an evaluation of the mes-
Surg 2003; 237: 335–42. orectum’s macroscopic quality. Surg Endosc 2005;
19. Leroy J, Jamali F, Forbes L, et al. Laparoscopic total 19: 307–10.
mesorectal excision (TME) for rectal cancer surgery: 32. Law WL, Lee YM, Choi HK, Seto CL, Ho JW. Impact
long-term outcomes. Surg Endosc 2004; 18: 281–9. of laparoscopic resection for colorectal cancer on
20. Leung KL, Kwok SP, Lam SC, et al. Laparoscopic operative outcomes and survival. Ann Surg 2007; 245:
resection of rectosigmoid carcinoma: prospective 1–7.
randomised trial. Lancet 2004; 363: 1187–92. 33. Quirke P. Training and quality assurance for rectal can-
21. Kang SB, Park JW, Jeong SY, et al. Open versus cer: 20 years of data is enough. Lancet Oncol 2003;
laparoscopic surgery for mid or low rectal cancer 4: 695–702.
after neoadjuvant chemoradiotherapy (COREAN trial): 34. Gouvas N, Tsiaoussis J, Pechlivanides G, et al. Quality
short-term outcomes of an open-label randomised of surgery for rectal carcinoma: comparison between
controlled trial. Lancet Oncol 2010; 11: 637–45. open and laparoscopic approaches. Am J Surg 2009;
22. Lujan J, Valero G, Hernandez Q, Sanchez A, Frutos 198: 702–8.
MD, Parrilla P. Randomized clinical trial comparing 35. Heald RJ, Moran BJ, Ryall RD, Sexton R, MacFarlane
laparoscopic and open surgery in patients with rectal JK. Rectal cancer: the Basingstoke experience of total
cancer. Br J Surg 2009; 96: 982–9. mesorectal excision, 1978–1997. Arch Surg 1998;
23. Braga M, Frasson M, Vignali A, Zuliani W, Capretti G, 133: 894–9.
Di Carlo V. Laparoscopic resection in rectal cancer pa- 36. Rahbari NN, Weitz J, Hohenberger W, et al. Definition
tients: outcome and cost–benefit analysis. Dis Colon and grading of anastomotic leakage following anterior
Rectum 2007; 50: 464–71. resection of the rectum: a proposal by the International
24. Hiki N, Shimizu N, Yamaguchi H, et al. Manipulation Study Group of Rectal Cancer. Surgery 2010; 147:
of the small intestine as a cause of the increased 339–51.
inflammatory response after open compared with 37. Cartmell MT, Jones OM, Moran BJ, Cecil TD. A de-
laparoscopic surgery. Br J Surg 2006; 93: functioning stoma significantly prolongs the length of
195–204. stay in laparoscopic colorectal resection. Surg Endosc
25. Pechlivanides G, Gouvas N, Tzortzinis A, et al. Lymph 2008; 22: 2643–7.
node clearance after total mesorectal clearance for 38. Zhou ZG, Hu M, Li Y, et al. Laparoscopic versus open
rectal cancer: laparoscopic versus open approach. Dig total mesorectal excision with anal sphincter preser-
Dis 2007; 25: 94–9. vation for low rectal cancer. Surg Endosc 2004; 18:
26. Heald RJ. The ‘holy plane’ of rectal surgery. J R Soc 1211–5.
Med 1988; 81: 503–8. 39. Vignali A, Braga M, Zuliani W, Frasson M, Radaelli G,
27. Birbeck KF, Macklin CP, Tiffin NJ, et al. Rates of Di Carlo V. Laparoscopic colorectal surgery modifies
circumferential resection margin involvement vary be- risk factors for postoperative morbidity. Dis Colon
tween surgeons and predict outcomes in rectal cancer Rectum 2004; 47: 1686–93.
surgery. Ann Surg 2002; 235: 449–57. 40. Allardyce RA, Bagshaw PF, Frampton CM, et al.
28. Jayne DG, Thorpe HC, Copeland J, Quirke P, Brown Australasian Laparoscopic Colon Cancer Study shows
JM, Guillou PJ. Five-year follow-up of the Medical that elderly patients may benefit from lower postopera-
Research Council CLASICC trial of laparoscopically tive complication rates following laparoscopic versus
assisted versus open surgery for colorectal cancer. Br open resection. Br J Surg 2010; 97: 86–91.
J Surg 2010; 97: 1638–45. 41. García-Granero E, Flor-Lorente B, García-Botello S,
29. Jayne DG, Guillou PJ, Thorpe H, et al. Randomized Muñoz E, Blanco F, Lledó S. The occlusive tourniquet:
trial of laparoscopic–assisted resection of colorectal a simple method for rectal stump washout during
carcinoma: 3-year results of the UK MRC CLASICC open and laparoscopic surgery. Dis Colon Rectum
Trial Group. J Clin Oncol 2007; 25: 3061–8. 2008; 51: 1580–2.
30. Biondo S, Ortiz H, Lujan J, et al. Quality of mesorec- 42. Scheidbach H, Schneider C, Hügel O, et al. Onco-
tum after laparoscopic resection for rectal cancer: logical quality and preliminary long-term results in
results of an audited teaching programme in Spain. laparoscopic colorectal surgery. Surg Endosc 2003;
Colorectal Dis 2010; 12: 24–31. 17: 903–10.

HEBK001-C09_p140-153.indd 153 08/02/13 5:50 PM


10
Robotic total mesorectal excision
M. Chadwick, H.S. Tilney and A.M. Gudgeon

Introduction supported the development of a prototype robot by


SRI International because of its potential to allow
The da Vinci robot is a sophisticated tool for per- surgeons to operate remotely on soldiers wounded
forming minimal access surgery. The actions of the on the battlefield. As a result of this, in 1995 Intui-
instruments are entirely under the control of a sur- tive Surgical Devices Inc. came into being; taking
geon seated separately from the patient. The camera this prototype forward, the da Vinci system evolved.
and instruments are controlled by hand and foot It was first tested in 1997 and then marketed in
movements of the surgeon. The surgeon can oper- ­Europe in 1999. Sales in the USA were soon to fol-
ate the instruments from a comfortable position, low in 2000, with FDA approval for general laparo-
with forearms supported and completely intuitive scopic surgery. The ZEUS robotic surgical system
reproduction of hand movements to the operating developed by the rival company Computer Motion
instruments. The surgery is aided by high-definition allowed the first telerobotic surgery in 2001, with
three-dimensional optics. The surgery may be per- the surgeon in New York and the patient in France.
formed entirely using the robotic instruments or as Fierce competition stalled further developments
a hybrid procedure combining standard laparos- until, in 2003, Intuitive Surgical and Computer
copy with robotics. Motion merged and the ZEUS system was phased
out in favour of the da Vinci system, which is now
used across the world, particularly in South Korea,
History and development of the USA and Europe. The latest refinement is the da
robotic colorectal surgery Vinci Si, released in 2009.
The most common procedure performed with
Robots have been used in surgery since 1985, with the da Vinci system is radical prostatectomy, but
the PUMA 560 first assisting a brain biopsy. In many other urological, gynaecological, cardiotho-
1988 the first prostatic surgery was performed by racic, paediatric and general surgical procedures
the PROBOT at Imperial College, London. The first have been performed, including pyeloplasty, cystec-
robot for clinical use in general surgery was the au- tomy, nephrectomy, ureteral reimplantation, hyster-
tomated endoscopic system for optimal positioning ectomy, myomectomy, sacrocolpopexy, mitral valve
(AESOP) (Computer Motion, Santa Barbara, CA, repair, atrial septal defect closure, cardiac bypass,
USA). In 1994 the United States Food and Drug ablation, mediastinal tumour excision, cholecys-
Administration (FDA) approved AESOP for clini- tectomy,1 Nissen fundoplication, Heller myotomy,
cal use as a robotic camera-holder. In 1990 the De- gastric bypass, donor nephrectomy, adrenalectomy,
fence Advanced Research Projects Administration splenectomy and hepatic resection.2

HEBK001-C10_p154-170.indd 154 11/02/13 3:58 PM


ROBOTIC RECTAL SURGERY  155

The uptake of robotic colorectal surgery has been The view of the surgical field is limited within the
relatively recent. Most work has been done in South pelvis during TME, particularly when operating in
Korea, but increasing numbers of procedures are the narrow male pelvis, and this may have an im-
being performed in Italy and the USA, and its use pact on the quality of the resected mesorectum.10
is gradually expanding in a few centres in the UK. The most important factor relating to rectal dissec-
Procedures include right and left hemicolectomy, tion is the grade of the mesorectal specimen, which
total mesorectal excision (TME), proctectomy and in turn is closely related to oncological outcome.11,12
ileal pouch–anal anastomoses, and ventral mesh A good view is mandatory if precise mesorectal
rectopexy. With increasing experience, confidence is dissection is to be successful. The ability of the da
building and increasingly advanced procedures are Vinci surgical system to offer a three-dimensional
being performed. In 2007 Baik and colleagues re- view with magnification, filtering of hand tremor,
ported simultaneous robotic TME, total abdominal fine dexterity and motion scaling suggests the po-
hysterectomy for rectal cancer and uterine myoma.3 tential for a technical surgical advantage over open
In 2009 Patriti and colleagues reported a series of or laparoscopic TME.
robotic colectomies, including one rectal resection A four-arm technique for robotic TME has been
with synchronous hepatic metastatectomies.4 summarized by Baik and colleagues.13 The instru-
In 2002 Weber and colleagues reported three ments used for dissection were a Cadiere grasper,
robotic right and sigmoid colectomies for benign a PreCise™ Bipolar grasper and a permanent cau-
disease using the da Vinci robotic system.5 In the tery spatula. The Cadiere grasper provides the first
following year, Delaney and colleagues compared traction and the PreCise Bipolar grasper provides
robot-assisted laparoscopic colectomy with case- the second proper traction. Moreover, these grasp-
matched results using standard laparoscopic ap- ers can change to an L-shaped small retractor using
proaches, focusing on clinical outcomes.6 Despite the EndoWrist™ function that is the core technol-
only six robotic colectomies being performed be- ogy of the da Vinci system. The robotic instrument
tween December 2001 and April 2002, there were can be used not only to create traction by grasping
conclusions of feasibility and safety of the da Vinci tissue but also to push the tissue in the narrow pel-
system. D’Annibale and colleagues reported 53 ro- vic space, thereby achieving traction and counter-
botic colorectal cases from May 2001 to May 2003, traction to expose the ideal tissue planes. The com-
including 22 patients with malignant disease.7 They bination of traction and counter-traction using the
concluded that robotic techniques could achieve the robotic instruments can provide an excellent surgi-
same operative and postoperative results as conven- cal view during the rectal dissection in a confined
tional laparoscopic surgery. Rawlings and colleagues space.
reported 30 consecutive robotic cases;8 the series
included 17 right hemicolectomies and 13 anterior
resections, with the conclusion that robotic surgery Evolution of Robotic Total Mesorectal
is technically feasible using the da Vinci system. Two Excision
years later the same authors reported the results
comparing robotic with laparoscopic colectomy.9 Until now, most studies relating to robotic low an-
The outcome in each group was similar. terior resection have reported only initial experi-
ences. The sample sizes have been small and they
are generally non-randomized. Larger, randomized
ROBOTIC RECTAL SURGERY studies are necessary to assess the feasibility of the
robotic system and whether there are real benefits
Robotic low anterior resection is performed in only of the robotic system compared with conventional
a few centres worldwide. Surgery for rectal cancer is laparoscopic surgery.
more difficult than colonic surgery because of the Tables 10.1–10.3 summarize the results of ex-
anatomical characteristics of the rectum and the isting studies comparing outcomes between ro-
pelvis. The principle underlying TME is precise dis- botic and laparoscopic rectal resections. In 2004
section of an avascular plane between the presacral D’Annibale and colleagues compared 12 robotic
fascia and the fascia propria of the mesorectum. rectal resections with laparoscopic rectal resection

HEBK001-C10_p154-170.indd 155 11/02/13 3:58 PM


HEBK001-C10_p154-170.indd 156

156  Robotic total mesorectal excision


Table 10.1. Overall characteristics of studies reporting outcomes of robotic rectal cancer surgery.
Period
Study Institution of study Type of study Patients (n) Procedures (n)
Robotic Laparoscopic
surgery surgery Robotic surgery Laparoscopic surgery
Delaney et al. 6
Cleveland Clinic, 2001–2002 Case-matched 6 6 Right hemicolectomy (2), sigmoid Right hemicolectomy (2), sigmoid
OH, USA resection (3), rectopexy (1) resection (3), rectopexy (1)
D’Annibale et al.7 Padova, Italy 2001–2003 Case-matched 53 53 Right hemicolectomy (10), ileocaecal Right hemicolectomy (13), ileocaecal
resection (0), transverse colectomy (0), resection (1), transverse colectomy (1),
left hemicolectomy (17), sigmoid left hemicolectomy (17),
resection (11), sigmoid resection (4), anterior
anterior resection (10), abdominoperineal resection (15), abdominoperineal
excision of rectum (1), excision of rectum (0),
total colectomy (2), Hartmann’s (1), total colectomy (1), Hartmann’s (1),
Hartmann reversal (0), rectopexy (1) Hartmann reversal (2), rectopexy (0)
Pigazzi et al. 14†
City of Hope National 2004–2005 Non-randomized, 6 6 Low anterior resection Low anterior resection
Medical Center, comparative
CA, USA
Hellan et al.15† City of Hope National 2004–2007 Case series 39 – Rectal cancers: low anterior resection (22), –
Medical Center, coloanal (11), abdominoperineal excision
CA, USA of rectum (6)
Baek et al.16† City of Hope National 2004–2008 Case series 64 – Rectal cancers: colorectal anastomosis (34), –
Medical Center, coloanal anastomosis (18), abdominoperineal
CA, USA excision of rectum (12)
Spinoglio et al. 19
Alessandria, Italy 2005–2007 Non-randomized, 50 161 Right hemicolectomy (18), left hemicolectomy Right hemicolectomy (50), left
comparative (10), anterior resection (19), abdominoperineal hemicolectomy (73), anterior resection
excision of rectum (1), transverse colectomy (1), (26), abdominoperineal excision of
total colectomy (1) rectum (7), transverse
colectomy (2),
total colectomy (3)

(Continues)
11/02/13 3:58 PM
HEBK001-C10_p154-170.indd 157

Table 10.1. Overall characteristics of studies reporting outcomes of robotic rectal cancer surgery. (Continued)

Period
Study Institution of study Type of study Patients (n) Procedures (n)
Robotic Laparoscopic
surgery surgery Robotic surgery Laparoscopic surgery
Baik et al.13* Seoul, South Korea 2006–2007 Case series 9 – Total mesorectal excision –

Baik et al.20* Seoul, South Korea 2006–2007 Randomized 18 18 ‘Tumour-specific total mesorectal ‘Tumour-specific total mesorectal
controlled trial excision’ excision’
Baik et al. *
21
Seoul, South Korea 2006–2007 Non-randomized, 56 57 Low anterior resection Low anterior resection
comparative
Choi et al.22 Korea University Anam 2007–2008 Case series 50 – Rectal cancers –
Hospital, Seoul,
South Korea
Luca et al.23 Milan, Italy 2007–2008 Case series 55 – Anterior resection (17), abdominoperineal –

ROBOTIC RECTAL SURGERY  157


excision of rectum (7), coloanal (4),
left hemicolectomy (27)

*Multiple publications by the same departments with likely duplication of reported outcomes.
†,
11/02/13 3:58 PM
HEBK001-C10_p154-170.indd 158

158  Robotic total mesorectal excision


Table 10.2. Short-term outcomes from studies reporting outcomes of robotic rectal cancer surgery.
Duration of surgery (min) Length of stay (days) Anastomotic
Study Conversion (range)a Blood loss (mL) (range)a (range) leak Complications
Robotic Laparoscopic Robotic Laparoscopic Robotic Laparoscopic Robotic Laparoscopic Robotic Laparoscopic Robotic Laparoscopic
surgery surgery surgery surgery surgery surgery surgery surgery surgery surgery surgery surgery
Delaney – – 216.5 150 (116–165) 100 87.5 (50–200) 3 (2–5) 2.5 (2–7) – – 1 atelectasis 1 incisional
et al.6 (170–274) (50–350) hernia

D’Annibale 6 (2 to 3 240 (661) 222 (677) 21 (680) 37 (6102) – – 0 1 2 bowel injuries, 1

et al.7 laparoscopy, CVA, 1 wound


4 to hand- infection
assisted
laparoscopy)
Pigazzi – – 264 (192–318) 258 (198–312) 104 150 (50–300) 4.5 (3–11) 3.6 (3–6) – – 1 prolonged ileus 1 pelvic abscess
et al.14† (50–318)

Hellan – – 285 – 200 – 4 (2–22) – 4 (12.1%) – 1 intraoperative –

et al.15† (180–540) (25–6000) bleed, 2 wound


infections

Baek 6 (9.4%) – 270 (150–540) – 200 – 5 (2–33) – 4 (7.7%) – 4 pelvic


et al.16† (20–6000) abscesses

Spinoglio 2 (1 to 4 383.8b 266.3b – – 7.74 8.31 2 – 1 incisional hernia, –

et al.18 laparoscopy, 1 atelectasis,


1 to 1 wound
laparotomy) infection,
1 phlebitis,
1 CVA
Baik 0 – 213 (153–315) – – – 7 (5–10) – – –
et al.13*

Baik 0 2 202.5 196.0 (114–297) – – 7 (5–10)b 9 (6–12)b – – – –


et al.20* (149–315)

Baik 0b 6b 178 (120–315) 179 (100–360) – – 5 (5–10)b 6 (4–16)b 1 4 ‘Serious ‘Serious


et al.21* complication’ complication’
5.4%b 19.3%b
(Continues)
11/02/13 3:58 PM
ROBOTIC RECTAL SURGERY  159

and demonstrated that robotic and laparoscopic

Laparoscopic
techniques could achieve the same operative and

surgery
postoperative results.7 Pigazzi and colleagues com-

Complications
pared short-term outcomes between robotic TME


and laparoscopic TME and concluded that robotic
low anterior resection with TME and autonomic
nerve preservation was possible.14 The same group
Robotic
surgery

subsequently reported a series of 39 consecutive


unselected patients with primary rectal cancer and
Laparoscopic

concluded that robotic-assisted surgery for rectal

Figures for continuous outcomes represent median values with range in parentheses unless 6 stated, which denotes standard deviation.
cancer could be carried out safely.15
surgery
Anastomotic

Baik and colleagues reported the first Asian expe-


leak

rience of robotic total mesorectal excision for rectal


cancer patients in June 2006.17 The first published


Robotic
surgery
4 (8.3%)

case of robotic abdominoperineal resection in Asia


was performed in Hong Kong in August 2006.18

One study including 20 robotic rectal cancer exci-


Laparoscopic
Length of stay (days)

sions concluded that robotic colonic surgery was fea-


surgery

sible and safe. Spinoglio and colleagues reported a sig-


Table 10.2. Short-term outcomes from studies reporting outcomes of robotic rectal cancer surgery. (Continued)

(range)

nificant increase in operating time (see Table 10.2),19


but in the authors’ experience this length reduces with


(4–17)
Robotic

(5–24)
surgery

experience as ‘docking time’ reduces. The use of a hy-


brid technique to perform vascular division and left
7.5
9.2

*Multiple publications by the same departments with likely duplication of reported outcomes.

colon/splenic flexure mobilization laparoscopically


Laparoscopic
Blood loss (mL) (range)a

can also help to reduce operative times.


Baik and colleagues launched the first prospec-
surgery

tive randomized trial of robotic versus laparoscopic


low anterior resection in 2006.20 The short-term


outcomes of this pilot study comparing 18 robotic
(0–600)
Robotic
surgery

low anterior resections with 18 laparoscopic cases


68

highlighted the feasibility and safety of robotic


low anterior resection. A trend towards a superior


Laparoscopic
Duration of surgery (min)

mesorectal grade in the robotic low anterior resec-


surgery

tion group did not achieve statistical significance,


(range)a

but macroscopic grading of the robotic group


showed the mesorectum to be complete in 17 cases


(190–485)

(164–487)

and nearly complete in 1 case. Similar results were


Robotic
surgery

shown in their larger, albeit non-randomized,


304.8

290

comparative study from 2008.23 The quality of the


resected specimen can be considered a surrogate
Laparoscopic

marker for both surgical quality and outcome.24 If


surgery

this can be shown to achieve significance in ongo-


CVA, cerebrovascular accident.
Statistically significant results.
Conversion

ing trials, then it would support a potential onco-


logical benefit for robotic rectal cancer surgery, in


oncological terms. In addition, the use of sharp dis-
Robotic
surgery

section and simple diathermy in the avascular TME


plane may aid identification and preservation of the
0

pelvic autonomic nerves and therefore represent a


secondary benefit in terms of long-term quality-of-
et al.22
et al.2
Study

Luca

life outcomes.
Choi

†,
b
a

HEBK001-C10_p154-170.indd 159 11/02/13 3:58 PM


HEBK001-C10_p154-170.indd 160

160  Robotic total mesorectal excision


Table 10.3. Oncological outcomes from studies reporting outcomes of robotic rectal cancer surgery.
Circumferential
Lymph node yield (n) resection margin
Study (range)a Distal resection margin (cm) Total mesorectal excision assessment involvementb
Robotic Laparoscopic Robotic surgery Laparoscopic Robotic surgery Laparoscopic surgery Robotic Laparoscopic
surgery surgery surgery surgery surgery
Delaney et al. 6
– – – – – – – –
D’Annibale et al.7 17 (610) 16 (69) – – – – – –
Pigazzi et al.14† 14 (9–28) 17 (9–39) 3.8 (1.8–9) 3.5 (2.2–5) – – – –
Hellan et al.15† 13 (7–28) – 2.65 (0.4–7.5) – – – 0 –
Baek et al.16† 14.5 (3–28) – 3.4 (0.2–10) – – – 0 –
Spinoglio et al. 18
22.03 22.85 7.3 7.9 – – – –
Baik et al.13* 22 (2–31) – 2.5 (1–6.5) – Complete (8), nearly complete (1) – – –
Baik et al.20* 18 (6–49) 22 (9–42) 4 (1–5.5) 3.5 (1.5–6.0) Complete (17), nearly complete (1) Complete (13), nearly complete (5) – –
Baik et al. *
21
17.5 (4–43) 17 (4–53) 4 (1–7) 3 (1–9) Complete (52), nearly complete (4), Complete (43), nearly complete 4 5
incomplete (0) (12), incomplete (2)
Choi et al.2 20.6 (4–48) – 1.9 (0.5–4.5) – – – 1 –
Luca et al.22 – – – – – – – –
a
Figures for continuous outcomes represent median values with range in parentheses unless 6 stated, which denotes standard deviation.
b
tumour #1 mm from circumferential cut edge.
†,
*Multiple publications by the same departments with likely duplication of reported outcomes.
11/02/13 3:58 PM
ROBOTIC RECTAL SURGERY  161

Overall, few studies have shown substantial ened resection margins(see Chapter 4). Endolumi-
differences in short-term outcomes between ro- nal ultrasound (see Chapter 3) can help to differen-
botic and laparoscopic resections (Tables 10.1 tiate between T1 and T2 tumours.
and 10.2). Two studies from the same unit have In the authors’ unit, all patients undergoing TME
revealed a significant reduction in length of stay with anastomosis have preoperative oral mechani-
and ­‘serious complications’ in patients undergo- cal bowel preparation and a defunctioning loop ile-
ing robotic surgery, but most of these small, and ostomy. Patients are counselled and sited by a stoma
almost exclusively non-randomized, studies have therapist preoperatively.
shown ­robotic surgery to be at least as safe as, and
oncologically equivalent to, laparoscopic surgery
(see Table 10.3).18,24 There is a need for further Position
exploration in large-scale randomized trials to de-
termine whether robot-assisted TME is superior The patient is most commonly placed in a modi-
in oncological and quality-of-life terms to laparo- fied Lloyd Davies position, with the hips slightly ex-
scopic and open surgery. tended and the knees flexed to 70–908 and placed just
greater than shoulder width apart. In our experience,
the use of a body-length gel mat in direct contact
Patient Preparation with the patient’s skin keeps the patient safely on the
operating table without the need for shoulder sup-
The preoperative work-up for a patient with rec- ports. Thromboembolic deterrent stockings (unless
tal cancer begins with a full history and physi- contraindicated) and intermittent calf compression
cal examination. Blood tests should include a full are used routinely. A warm air blanket is placed over
blood count, electrolytes, liver function tests, car- the patient’s chest. The arms are wrapped at the pa-
cinoembryonic antigen (CEA) levels and a 2 unit tient’s sides and protected from the metalwork of the
cross-match. In our experience, blood transfusion table. The patient is catheterized.
is rarely required during laparoscopic or robotic Some surgeons prefer to use a lateral approach
rectal cancer surgery, but it must be remembered to the splenic flexure, in which case the patient is
that if serious bleeding occurs undocking the ro- positioned accordingly – left side uppermost, with
bot can lead to delays in controlling the bleeding the hips straight and knees bent. Lateral supports
point. An electrocardiogram is requested; where are necessary against the patient’s back. A left arm
appropriate, cardiopulmonary exercise testing is in- support gutter is also used. (If the hips are flexed,
creasingly used to help to stratify operative risk and the left thigh will hinder the smooth and efficient
optimize perioperative management. Full colonic use of instruments via the left lateral port.) After
imaging by colonoscopy, barium enema or com- mobilizing the splenic flexure, the patient is turned
puted tomographic (CT) pneumocolon evaluation on to the back and positioned in the Lloyd Davies
is performed to exclude synchronous tumours, and position, as described above.
histological diagnosis is confirmed on biopsy. The
distance from the lower border of the tumour to
the anal verge is measured using rigid sigmoidos- Surgical Approach
copy; accurate measurement is essential for surgical
planning. Clinical evaluation of sphincter tone and The authors favour routine mobilization of the splenic
careful assessment of premorbid anorectal function flexure when performing an anterior resection with
may help to identify patients in whom a low anas- TME. This is done at the start of the procedure as it
tomosis would lead to unacceptable postoperative is often technically difficult. Once completed, there is
outcomes. Computed tomography of the chest, ab- no temptation to compromise if the rectal dissection
domen and pelvis is performed to assess for distant proves difficult and time-consuming. Although mobi-
metastases and local disease infiltration. lization of the splenic flexure and left colon with high
The rectal cancer is staged by pelvic magnetic ligation of the inferior mesenteric artery and vein can
resonance imaging (MRI) to measure the extent of be performed using the da Vinci system, the authors
local infiltration and identify patients with threat- favour a hybrid approach, with standard laparoscopy

HEBK001-C10_p154-170.indd 161 11/02/13 3:58 PM


162  Robotic total mesorectal excision

for the abdominal component of the operation fol- field of view. The camera port is therefore placed in
lowed by robotic rectal dissection. the mid-position of the instrumental port site arc,
ideally just above or to the right of the umbilicus,
Port sites so that the field of vision is centred on the mid-
The optimum port siting is the subject of much line. The primary operative robotic 8-mm port is
conjecture, and many arrays of ‘standardized’ port placed on the right, 8–10 cm laterally along the arc
site are described. The authors’ preferred set-up has from the umbilicus to the right anterior superior
evolved from assessment of the published techniques iliac spine. The secondary operative robotic 8-mm
and as a result of trial and error. A compromise in port is placed 8–10 cm left laterally along the arc
placement is required to facilitate safe and efficient from the umbilicus to the left anterior superior iliac
laparoscopic mobilization, while still allowing effec- spine. The tertiary operative robotic 8-mm port
tive robotic instrument use in the pelvis, if the over- is placed 8–10 cm laterally to this along the same
all number of port sites is to be minimized. A report arc at a site roughly corresponding to the 12-mm
by Luca and colleagues describes a port site set-up laparoscopic assistant port opposite (Figure 10.1).
that allows for complete low anterior resection and For a small patient, the second operating arm can
does not require the robot cart to be moved.25 The be placed a few centimetres cranially to maintain
authors’ favoured sites are as follows: optimum spacing between ports.
A supraumbilical incision is made and a 12-mm
internal diameter port is inserted using an open Has- Right lateral approach to the splenic flexure
san technique to accommodate the large-diameter If a lateral approach is chosen, the first port to be
three-dimensional robotic telescope and camera. placed is in the left lateral port position using a
The pneumoperitoneum is established with an in- 5-mm optical port, inside which a 5-mm laparo-
sufflation pressure set to 12–15 cmH2O. A 10-mm scope is used to enter the abdominal cavity under
laparoscope is used to assess the feasibility of a min-
imal access approach in terms of adhesions, adipos-
ity of the abdominal wall and mesenteries, and ana-
tomical anomalies, which may hinder or prevent
progress. The procedure continues by insertion of
the operating and assisting robotic 8-mm ports.
These are inserted under direct vision via marked
skin incisions positioned optimally to avoid clash-
ing of the robotic arms. A minimum of 8 cm is rec-
ommended between ports. A2
The robotic cannulae are marked with broad
black bands that represent the fulcrums for angu-
lation. These should lie just within the abdominal 12 R2
wall to allow effective movement and minimize
postoperative pain. R1
R3
The authors favour the use of robotic ports in
standardized positions during the laparoscopic part A1
of the procedure. An additional 12-mm port in the
right iliac fossa, 3–4 cm inferomedial to the ante-
rior superior iliac spine, is used for laparoscopic
instrumentation and for assistant access during the
robotic phase of the procedure. This allows access
for clip applicators, suction/irrigation devices, in- Figure 10.1. Robotic instrument ports R1, R2 and R3.
sertion and removal of tonsil and mastoid swabs, R1 is the main operating port, with traction and counter-
and stapling devices. traction provided by R2 and R3. The 12-mm port is for the
Ports are sited to optimize triangulation of in- camera just to the right of the umbilicus. A1 and A2 are
struments, avoid clashing and provide maximum assistant ports. A1 is also used for stapling the rectum.

HEBK001-C10_p154-170.indd 162 11/02/13 3:58 PM


THE DA VINCI SURGICAL SYSTEM  163

vision. This port can be replaced later by the 8-mm placed back in the left upper quadrant. The small
robotic port. The laparoscopy is performed and the bowel is gently swept out of the pelvis up into the right
12-mm umbilical port can then be inserted under upper quadrant. The pelvic brim is identified at the
direct vision. An additional 5-mm port is placed in sacral promontory, and the sigmoid colon is tented
the left upper midclavicular line for the purposes of up to reveal the fossa beneath the inferior mesenteric
laparoscopic splenic flexure mobilization. artery (IMA) over the bifurcation of the aorta.
The peritoneum is incised along a broad front
Mobilization of the splenic flexure to allow entry into, and then medial to lateral dis-
The right lateral position for splenic flexure mo- section within, Toldt’s plane. The retroperitoneal
bilization is particularly useful in patients who are structures are carefully swept posteriorly, preserv-
obese, as the entire enteric mass falls away under ing the left ureter and gonadal vessels. Once these
the influence of gravity; in some cases, the entire are identified and preserved, attention is focused on
lateral mobilization of the left colon down to the isolation of the IMA. Taken high, this allows for a
pelvic brim can be achieved in this position. More single vessel division. It is important to identify and
commonly, however, splenic flexure mobilization is preserve the pre-aortic sympathetic nerves at this
performed with the patient supine, tilted left side point and to avoid taking the vessel flush with the
up towards the operating surgeon. The small bowel aorta, as this may cause nerve damage. There are
should fall away but may require gentle sweeping. many ways to secure the artery, including clips, sta-
Occasionally, peritoneal adhesions prevent this and pling devices and energy instruments. When taken
require division to partially mobilize the duodeno- more distally, the ascending left colic artery and sig-
jejunal flexure. Once the lateral border of the duo- moid branches may be encountered, necessitating
denojejunal flexure is clearly identified, the inferior further dissection and separate vessel division.
mesenteric vein can be identified running inferola- The dissection is continued in Toldt’s plane as far
terally within the left colonic mesentery. out laterally as the abdominal wall and cranially un-
The peritoneum is incised to allow identification til the previously dissected descending colonic me-
and isolation of the inferior mesenteric vein. The sentery is reached. Inferiorly, the dissection reaches
vein is divided with an appropriate haemostatic the plane between the mesorectal fascia and the pel-
energy source, or between clips, at the level of the vic parietal fascia. The lateral attachments are then
lower border of the pancreas. Dissection continues divided, taking particular care to avoid the left ure-
medial to lateral in the plane between the mesen- ter across the pelvic brim, such that the entire left
tery and Gerota’s fascia towards the lateral abdomi- colon is then mobilized. It is then the authors’ pref-
nal wall. The lesser sac is entered just above the erence to tie a nylon tape around the rectosigmoid
body of the pancreas, where the left branch of the junction to facilitate retraction of the rectum out of
middle colic artery marks the most medial aspect the pelvis during mesorectal dissection.
of the dissection. The posterior wall of the stomach
is clearly seen superiorly, with the tail of the pan-
creas inferiorly and the transverse mesocolon an- THE DA VINCI SURGICAL SYSTEM
teriorly after its disconnection from the pancreas.
The splenic flexure and descending colon are ante- The robotic system consists of three main compo-
rolateral. Lateral attachments of the descending co- nents: the operating console, the electronic tower
lon and splenic flexure are divided, and the flexure and the patient cart.
is freed by re-entering the lesser sac from above by The console (Figure 10.2) is where the surgeon
dissecting the greater omentum free from the trans- controls the robotic arms and instruments. It com-
verse colon. prises scissor-handle-type master controls, which
translate the surgeon’s hand movement directly to
Division of inferior mesenteric artery and the instrument tips, including the ‘wristing’ angu-
sigmoid mobilization lation and rotation (Figure 10.3). There are foot
The patient is placed in a steep head-down position, pedals for camera control, disengaging instruments
with the right lateral tilt maintained. The omentum (clutch), focusing and the application of monopo-
is reflected over the liver and the mobilized flexure lar and bipolar diathermy (Figure 10.4).

HEBK001-C10_p154-170.indd 163 11/02/13 3:58 PM


164  Robotic total mesorectal excision

cart with specially designed transparent sterile covers


(Figure 10.6). The stereoscopic scope, camera and
light lead are also prepared in a sterile fashion with a
special disposable outer sleeve.
There is a range of dedicated robotic instruments,
including dissectors, graspers and retractors. Where
appropriate, these instruments can be connected to
monopolar or bipolar diathermy sources. Each in-
strument has a lifespan of ten procedures. To mini-
mize cost, it is important to develop a technique
using as few instruments as possible. The authors
prefer the use of a diathermy hook or monopolar
diathermy scissors (‘hot shears’) in the right-side
operative port, Maryland bipolar forceps in the pri-
mary assistant port, and a large grasping retractor
or Cadiere forceps in the secondary assistant port.
The central robotic arm holds the camera. Arms 1,
2 and 3 hold the operating instruments introduced
through the 8-mm reusable robotic ports.

Set-Up of Robotic Total Mesorectal


Figure 10.2. Operating console.
Excision
The electronic tower holds the video-processing
system, light source and insufflation equipment. All The patient remains in a steep head-down right
the electronics run through the control tower be- lateral tilt to prevent the small bowel from falling
tween the console and the patient cart. The cart also into the pelvis. In female patients with an intact
has a high-definition screen to transmit a two-di- uterus, a straight-needled polypropylene suture is
mensional image of the operating field and allows a driven through the suprapubic skin through the
trainer to draw lines on the screen to help a trainee fundus of the hitched-up uterus and back through
sitting at the console locate the correct planes of the suprapubic skin and tied over a swab to pre-
dissection. vent damage to the skin and to act as a reminder
The patient-side cart consists of a powered trol- to remove the suture at the end of the operation.
ley holding three or four robotic arms (Figure 10.5). The authors find this gives better uterine eleva-
The scrub nurse is responsible for preparing the robot tion than lifting the uterus by the broad ligament

Figure 10.3. Hand controls for robotic


surgical instruments.

HEBK001-C10_p154-170.indd 164 11/02/13 3:58 PM


THE DA VINCI SURGICAL SYSTEM  165

Figure 10.4. Foot pedals.

and fallopian tubes and also requires only a single The role of the assistant is important and far
suture. from redundant. A scrubbed assistant knowledge-
able in the set-up and function of the da Vinci sys-
tem is absolutely essential to the smooth running
Positioning the Robot and efficiency of the procedure. Set-up (docking)
can be time-consuming and present risk to the pa-
The choice of robot docking position is entirely tient if not performed carefully and meticulously.
personal to the surgeon and will be influenced by The robot is carefully docked between the patient’s
both positive and negative experiences in terms of legs, and the laparoscopic instruments, including the
ease of dissection and minimization of arm clashes camera and scope, are stored in sterile fashion for later
and instrument clashes. The unique disadvantage use. The pneumoperitoneum is maintained and the
of colorectal resection, compared with prostatic
surgery for example, is the necessity to operate in
multiple, and comparatively larger, anatomical sec-
tors (e.g. 30 cm for splenic flexure and descend-
ing colon and 20 cm for the rectum versus 4–5 cm
for the retropubis). A low anterior resection with
TME requires at least two fields of view, and double
docking of the robot adds time to the procedure.
There are few advantages to robotic splenic flexure
mobilization compared with standard laparoscopic
techniques, since the space is less confined and the
surgery often requires relatively large retraction
movements crossing the surgical field. As a result,
a hybrid procedure is often preferred. This elimi-
nates the compromised position of the robot posi-
tioned by the left leg and allows the more optimal
between-the-legs docking position.
The set-up involves manoeuvring the patient-
side cart into position between the patient’s legs.
The central hub, trolley and camera arm of the ro-
bot should all be in line with the patient’s midline.
This enables the ‘sweet spot’ of the camera arm to
be found with ease and without the camera arm
getting in the way of the other instrument arms. Figure 10.5. Patient-side cart with operating arms.

HEBK001-C10_p154-170.indd 165 11/02/13 3:58 PM


166  Robotic total mesorectal excision

Figure 10.6. Patient-side cart set up and


draped.

ports remain in place. The robotic arms are positioned thermy plume and provide tonsil and mastoid swabs
carefully under clutch control to the proximity of the as necessary. The assistant makes a valuable contribu-
robotic ports. Each is then carefully manoeuvred and tion throughout the procedure in helping to provide
fixed to its respective ports using their clasp attach- the essential traction and counter-traction to progress
ments. The assistant is then responsible for insertion with the dissection; this can usually be performed
of the camera and instruments under the careful in- with the suction and irrigation device. It is important
struction of the surgeon now sitting at the console. to remember that each instrument attached to a robot
The camera is introduced first to allow for safe intro- arm has a fixed limited number of uses, and therefore
duction of the other robotic instruments. Although careful instrument choice can limit wastage and un-
in most cases the 08 scope is suitable for visualization, necessary expense.
when difficulty is encountered the 308 scope can be
used with good effect. The angled view is fixed either
‘down’ or ‘up’ for the robotic surgeon, however, who Pelvic Dissection
does not have the advantage afforded the laparoscop-
ic surgeon of being able to rotate the lens of the 308 Sharp dissection using a hook or scissors attached
scope to view the surgical field from varying angles to monopolar diathermy is preferred by the au-
while still maintaining the horizon. Each arm is fitted thors. The harmonic scalpel can be used, but there
in turn with an appropriate robotic instrument. The is no hinge mechanism on the wrist of this instru-
primary arm usually holds the scissors or hook dis- ment. The bloodless mesorectal plane between the
sector with monopolar diathermy attachment. The mesorectum and parietal presacral pelvic fascia is
secondary arm usually holds the Cadiere or Maryland demonstrated easily due to the enhanced view and
bipolar diathermy graspers. The third arm holds a fur- by gentle anterior traction using a tonsil swab held
ther grasper of the surgeon’s choice; a longer grasper by a grasping instrument. The swab provides a
that doubles as a retractor is a useful instrument for blunt broad compression of the posterior mesorec-
this arm. The assistant can lift the rectum out of the tum, minimizing the risk of fascial breach. It also
pelvis using another laparoscopic grasper holding the absorbs small amounts of blood and tissue fluid.
nylon tape as required. Thus, three-point traction can The dissection continues as far as the pelvic floor
be achieved, facilitating precise dissection in the cor- behind and down the right side of the mesorec-
rect plane in a confined space. This dissection is under tum. It is essential for the surgeon to remember
the continuous control of the operating surgeon at that the sacral concavity arcs forwards as dissection
the console. The assistant must be continuously alert progresses, but this is well demonstrated with the
to the risk of robotic arm clashing and communicate three-dimensional view offered by the robot. Extra
regularly with the surgeon. The assistant must also care must be taken not to breach the parietal pelvic
be ready to improve vision by aspiration of the dia- fascia; this is particularly important in the midline

HEBK001-C10_p154-170.indd 166 11/02/13 3:58 PM


THE DA VINCI SURGICAL SYSTEM  167

posteriorly, where the presacral veins can be dam- dle rectal vessels, but these are by no means consistent
aged, leading to haemorrhage that can be difficult and are generally small and insignificant. If progress is
to control. Lateral dissection should be performed not being made in one area, it is important to move to
predominantly from the right side; it is here that another. It is usually possible to dissect the left side of
the hinged robotic instruments offer a significant the mesorectum from the right side of the patient. The
advantage over standard laparoscopy. The perito- mesorectal dissection is started on the right and pos-
neum is opened laterally, taking care not to inad- terolaterally to the left pelvic side-wall from the right
vertently damage the inferior hypogastric nerve or side of the patient; when coming over to the other side
ureter. The posterolateral dissection is taken down of the rectum, all that remains is the division of the
to a comfortable level, where progress starts to be- peritoneum and to complete the TME. Finally, any
come a little more difficult; at this stage, the ante- lower parts of the mesorectum are dissected off the
rior dissection is started. The peritoneum is divided pelvic floor to reveal a clean rectal tube. The muscle
anterior to and above the reflection. The anterior tube can then be divided at the pelvic floor.
dissection continues behind the seminal vesicles in
male patients, at which point Denonvilliers’ fascia
is reached. Denonvilliers’ fascia may exist in one or Rectal Transection
two layers, the anterior layer behind the prostate
and the posterior layer in front of the anterior me- A further nylon tape is tied around the rectal tube, tak-
sorectum. When two layers are present, they fuse ing care that it is tightened well below the lower border
at the lower level of the prostate. At this point, the of the tumour. The authors favour a distal rectal wash-
fascia is divided to enter the plane in front of the out with a cytocidal solution of iodine and water. At
mesorectum. At the lateral corners of Denonvilliers’ this point, there are two choices. The rectum can be di-
fascia, the surgeon needs to be aware of the close vided with the robotic cutting instrument, followed by
proximity of the neurovascular bundles, as these are the insertion of a purse-string suture using the robotic
easily damaged at this point. needle driver;26 this may facilitate transanal extraction
In female patients, the dissection continues down of the specimen.27 Alternatively, if there is space for a
the rectovaginal septum to the pelvic floor. The me- linear laparoscopic stapler, this can be inserted to di-
sorectum is often very thin and easily breached at vide in the standard laparoscopic fashion. The instru-
this level. If dealing with an advanced anterior T3 ment cart can then be undocked after careful removal
or T4 tumour in the lower rectum, it is often neces- of the robotic instruments and camera. An articulated
sary to remove some of the posterior vaginal wall en 45-mm linear stapler cutter is then inserted via the 12-
bloc with the specimen. An advanced upper rectal or 15-mm (right iliac fossa) port. The distal nylon tape
tumour may require an en-bloc hysterectomy. is used to position the rectal tube in the jaws of the sta-
The anterior dissection is perhaps the most chal- pler. Once precise positioning has been achieved, the
lenging part of the operation and may require ante- stapler is closed and then fired after 20 s of tissue com-
rior retraction of the prostate to allow the dissection pression. The stapler is withdrawn and the nylon tape
to proceed to the pelvic floor under direct vision. The is grasped with a ratcheted toothed grasper so that the
mesorectal plane guides the surgeon along the pel- specimen can be easily found for extraction.
vic floor to the rectal tube, which is encountered as it There are often difficulties getting a laparoscopic
passes through the pelvic floor; at this point, the dis- stapler into a tight male pelvis, and it is vital not
section can continue into the intersphincteric plane if to compromise the distal resection margin because
required. The surgeon switches from anterior to pos- of inadequate instrumentation. Currently the tech-
terior to lateral as dictated by view and ease of access. nology is not available to provide the huge forces
Laterally the retraction and three-dimensional view necessary to generate compression and closure of
aids the identification and preservation of the inferior the device around an acute angle without a large
hypogastric plexus and nervi erigentes. A branch of joint between the stapler head and the driving
the inferior hypogastric nerve passes medially to the mechanism. In these circumstances, it is better to
rectum (previously known as the lateral ligament); this enlarge the Pfannenstiel incision and apply an al-
nerve is variable in thickness and will need to be divid- ternative narrow stapling device manually, as com-
ed. Occasionally the nerve is accompanied by the mid- monly used in open surgery. These staplers can be

HEBK001-C10_p154-170.indd 167 11/02/13 3:58 PM


168  Robotic total mesorectal excision

manipulated laparoscopically providing a seal is mum of four degrees of freedom. Movements are
established to prevent leakage of the CO2 pneu- compromised somewhat by dependence upon the
moperitoneum. Alternatively, the rectal muscle tube fulcrum effect of the instruments having to pass via
can be divided distal to the nylon tape using the da ports through the abdominal wall.
Vinci scissors; a purse-string suture can then be in- The intuitive and articulated instrument move-
serted using the da Vinci needle drivers, avoiding ments of the robot have seven degrees of freedom,
the need for a linear stapler. Other options include which are particularly helpful in the confined
a sutured anastomosis performed either robotically space of the pelvis. The ports are simply a means
or with a peranal approach. of access rather than an integral part of instrument
movement, as with standard laparoscopy. The ro-
bot eliminates tremor and scales down movements
Specimen Withdrawal made at the surgeon console, enhancing the accu-
racy of dissection.
A 5- to 7-cm suprapubic transverse incision is made The image of the operative field provided by high-
and a self-retracting dual ring-reinforced wound pro- definition camera systems in laparoscopic surgery is
tector is used to facilitate specimen extraction. When now excellent. Depth of field has to be interpreted by
the mesorectum is not too bulky, the specimen may the surgeon and may adversely affect performance.
be removed through the anal canal. The left colon is The high-definition three-dimensional view provided
divided at a point of convenience; there should be by the robotic optics is vastly superior to the high-def-
a good colonic blood supply based on the marginal inition two-dimensional views produced by standard
artery, and sufficient length to reach the pelvic floor laparoscopic cameras and display monitors. The cam-
without tension. Some surgeons may also favour the era is held perfectly still, avoiding the risk of assistant
fashioning of a colo-pouch. A circular stapler anvil fatigue and reducing disorientation of the operating
is inserted in the proximal colon and secured with a surgeon. These advantages allow for more precise,
purse-string suture. The colon is then returned to the meticulous, sharp dissection of clearly identified tissue
abdomen and the wound protector loosened by un- planes, resulting in less blood loss and reduced surgi-
ravelling the ring reinforcer to allow torsional closure. cal trauma. The enhanced view, intuitive instrument
This is retightened using the ring reinforcer over a movements and improved dissection quality are likely
swab wrapped around the closed core. Pneumoperi- to steepen, and therefore shorten, the learning curve
toneum can then be re-established and an end-to-end, for minimally invasive TME and reduce the chance
end-to-side or end-to-pouch anastomosis fashioned of the need to convert to a standard open procedure.
with the circular stapling device. The anastomosis can Robotic TME may demonstrate improvements in on-
be tested for air leaks, but this is usually unnecessary cological circumferential resection margins and pelvic
as it is the authors’ routine practice to defunction all nerve preservation. It is important to emphasize, how-
TME coloanal/low colorectal anastomoses with a loop ever, that this is purely conjecture until the results of
ileostomy. It is our practice to carefully inspect the clinical trials such as the Robotic versus Laparoscopic
doughnuts and examine the anastomosis digitally to Resection for Rectal Cancer (ROLARR) trial, designed
exclude obvious flaws. to specifically investigate robotic TME compared with
laparoscopic TME, become available.

Advantages and
disadvantages of robotic Disadvantages
colorectal surgery
One of the main disadvantages is the complete lack
Technical Advantages of tactile feedback from the instruments. This can
potentially lead to tissue damage both in and out of
Laparoscopic colorectal surgery is technically very the field of view. A second drawback is the time-con-
demanding and the learning curve is often long. suming procedure of the docking and undocking of
There is considerable restriction of movement with the robotic cart from the patient. If significant bleed-
standard laparoscopic instruments, with a maxi- ing occurs, robotic instruments cannot be changed

HEBK001-C10_p154-170.indd 168 11/02/13 3:58 PM


References  169

quickly to deal with the problem, and the undocking sected specimen, positivity of resection margins, local
process may lead to greater blood loss before control recurrence, quality of life, pelvic nerve function and
is achieved either laparoscopically or by open surgery. health economics analysis.
Technological advances are in progress to create An excellent review by Mirnezami and colleagues
hinged harmonic/ultrasonic energy devices and ro- confirms there is no evidence in the medical litera-
botically controlled suction devices, and the latter ture to demonstrate an advantage of using the da
are now available on the Si model. Suction can eas- Vinci robot for rectal cancer surgery over standard
ily be added via the assistant port, however, and also laparoscopic surgery.28 There is no doubt that the
makes an excellent additional retractor to provide view of the pelvic dissection provided by the da Vinci
counter traction. three-dimensional camera system is unrivalled, mak-
Another disadvantage is the duration of surgery. ing it easier to identify important anatomical struc-
This applies to the set-up (docking) and the proce- tures and dissect without damage to the pelvic nerves.
dure itself. Operating times will reduce with pro- The intuitive hinged instruments also provide an ad-
gression up the learning curve, but until mastery is vantage when dissecting in the narrow confines of
achieved they will always be longer than with stand- the pelvis. These are subjective rather than objective
ard laparoscopic surgery or open surgery. This has observations, and translating them into an evidence-
implications for list planning, service provision and based advantage may prove difficult. The literature
training of juniors in the future. does, however, conclude that robotic TME is at least
A current main issue is cost, as the da Vinci sys- as good and as safe as conventional laparoscopic sur-
tem is expensive. This applies not only to the initial gery, and it is therefore important that we engage with
outlay for the robot but also to the cost of consuma- and develop the technology with the hope of improv-
bles and the annual servicing contract. ing the quality of rectal TME surgery in the future.
These disadvantages must be weighed against the
potential benefits of unrivalled views of the anato-
my, reduced blood loss through accurate dissection References
of the tissue planes, and potentially improved nerve
preservation and quality of the TME specimen.   1. Kang CM, Chi HS, Hyeung WJ, et al. The first Korean
experience of telemanipulative robot-assisted laparo-
scopic cholecystectomy using the da Vinci system.
THE FUTURE Yonsei Med J 2007; 48: 540–5.
  2. Choi SB, Park JS, Kim JK, et al. Early experiences of
robotic-assisted laparoscopic liver resection. Yonsei
It is likely that robotic technology will improve with
Med J 2008; 49: 632–8.
time and become more cost-effective, smaller and ef-   3. Baik SH, Kim YT, Ko YT, et al. Simultaneous robotic
ficient. There is enormous potential to improve the total mesorectal excision and total abdominal hys-
quality of mesorectal dissection, and we believe this terectomy for rectal cancer and uterine myoma. Int J
is another step of progress in the surgical treatment Colorectal Dis 2008; 23: 207–8.
of rectal cancer.28 Cross-stapling the low rectum in a   4. Patriti A, Ceccarelli G, Bartoli A, Spaziani A,
narrow pelvis remains a challenge with the minimal Casciola L. Laparoscopic and robot-assisted one-
stage resection of colorectal cancer with synchronous
access staplers currently available, but robotic purse-
liver metastases: a pilot study. J Hepatobiliary Pan-
string suturing may help overcome some of these creat Surg 2009; 16: 450–7.
difficulties in the short term. There are numerous   5. Weber PA, Merola S, Wasielewski A, Ballantyne GH.
case series and several comparative studies demon- Telerobotic-assisted laparoscopic right and sigmoid
strating equivalence or improvements in oncological colectomies for benign disease. Dis Colon Rectum
and functional outcome measures. 2002; 45: 1689–94, 1695–6.
Robotic rectal cancer surgery is currently being   6. Delaney CP, Lynch AG, Senagore AJ, Fazio VW.
Comparison of robotically performed and traditional
evaluated through international collaboration in a
laparoscopic colorectal surgery. Dis Colon Rectum
worldwide multicentre randomized controlled trial of 2003; 46: 1633–9.
robotic versus laparoscopic TME (the ROLARR trial),   7. D’Annibale A, Morpurgo E, Fiscon V, et al. Robotic
whose main outcome measures are rates of conversion and laparoscopic surgery for treatment of colorectal
to open surgery. Secondary measures are quality of re- disease. Dis Colon Rectum 2004; 47: 2162–8.

HEBK001-C10_p154-170.indd 169 11/02/13 3:58 PM


170  Robotic total mesorectal excision

  8. Rawlings AL, Woodland JH, Crawford DL. Telero- and China with an updated literature review. World J
botic surgery for right and sigmoid colectomies: Gastroenterol 2007; 13: 2514–8.
30 consecutive cases. Surg Endosc 2006; 20: 19. Spinoglio G, Summa M, Priora F, Quarati R, Testa S.
1713–8. Robotic colorectal surgery: first 50 cases experience.
  9. Rawlings AL, Woodland JH, Vegunta RK, Crawford Dis Colon Rectum 2008; 51: 1627–32.
DL. Robotic versus laparoscopic colectomy. Surg 20. Baik SH, Ko YT, Kang CM, et al. Robotic tumor-specific
Endosc 2007; 21: 1701–8. mesorectal excision of rectal cancer: short-term outcome
10. Baik SH, Kim NK, Lee KY, et al. Factors influencing of a pilot randomized trial. Surg Endosc 2008; 22: 1601–8.
pathologic results after total mesorectal excision for 21. Baik SH, Kwon HY, Kim JS, et al. Robotic versus
rectal cancer: analysis of consecutive 100 cases. Ann laparoscopic low anterior resection of rectal cancer:
Surg Oncol 2008; 15: 721–8. short-term outcome of a prospective comparative
11. Heald RJ, Husband EM, Ryall RD. The mesorectum in study. Ann Surg Oncol 2009; 16: 1480–7.
rectal cancer surgery: the clue to pelvic recurrence? Br 22. Choi DJ, Kim SH, Lee PJ, Kim J, Woo SU. Single-stage
J Surg 1982; 69: 613–6. totally robotic dissection for rectal cancer surgery:
12. Nagtegaal ID, van de Velde CJ, van der Worp E, technique and short-term outcome in 50 consecutive
et al. Macroscopic evaluation of rectal cancer resec- patients. Dis Colon Rectum 2009; 52: 1824–30.
tion specimen: clinical significance of the pathologist in 23. Baik SH, Kim NK, Lee KY, et al. Factors influencing
quality control. J Clin Oncol 2002; 20: 1729–34. pathologic results after total mesorectal excision for
13. Baik SH, Lee WJ, Rha KH, et al. Robotic total mes- rectal cancer: analysis of consecutive 100 cases. Ann
orectal excision for rectal cancer using four robotic Surg Oncol 2008; 15: 721–8.
arms. Surg Endosc 2008; 22: 792–7. 24. Quirke P, Steel R, Monson J, et al. Effect of the plane
14. Pigazzi A, Ellenhorn JD, Ballantyne GH, Paz IB. of surgery achieved on local recurrence in patients
Robotic-assisted laparoscopic low anterior resection with operable rectal cancer: a prospective study us-
with total mesorectal excision for rectal cancer. Surg ing data from the MRC CR07 and NCIC-CTG CO16
Endosc 2006; 20: 1521–5. randomised clinical trial. Lancet 2009; 373: 821–8.
15. Hellan M, Anderson C, Ellenhorn JD, Paz B, Pigazzi 25. Luca F, Cenciarelli S, Valvo M, et al. Full robotic left
A. Short-term outcomes after robotic-assisted total colon and rectal cancer resection: technique and early
mesorectal excision for rectal cancer. Ann Surg Oncol outcome. Ann Surg Oncol 2009; 16: 1274–8.
2007; 14: 3168–73. 26. Prasad LM, deSouza AL, Marecik SJ, Park JJ, Abcarian
16. Baek JH, McKenzie S, Garcia-Aguilar J, Pigazzi A. On- H. Robotic pursestring technique in low anterior resec-
cologic outcomes of robotic-assisted total mesorectal tion. Dis Colon Rectum 2010; 53: 230–4.
excision for the treatment of rectal cancer. Ann Surg 27. Kang J, Min B, Hur H, Kim N, Lee K. Transanal speci-
2010; 251: 882–6. men extraction in robotic rectal cancer surgery. Br J
17. Baik SH, Kang CM, Lee WJ, et al. Robotic total mes- Surg 2012; 99: 133–6.
orectal excision for the treatment of rectal cancer. 28. Mirnezami A, Mirnezami R, Venkatasubramaniam A,
J Robotic Surg 2007; 1: 99–102. Chandrakumaran K, Cecil T, Moran B. Robotic color-
18. Ng SS, Lee JF, Yiu RY, Li JC, Hon SS. Telerobotic- ectal surgery: hype or new hope? A systemic review of
assisted laparoscopic abdominoperineal resection for robotics in colorectal surgery. Colorectal Dis 2010; 12:
low rectal cancer: report of the first case in Hong Kong 1084–93.

HEBK001-C10_p154-170.indd 170 11/02/13 3:58 PM


11
Local excision and transanal
endoscopic microsurgery
Wolfgang B. Gaertner and David A. Rothenberger

Introduction anastomosis to achieve near-normal continence


and defecation; preserving genitourinary functions;
The role of local excision (LE) in the treatment of and promoting rapid recovery from surgery, with a
rectal cancer is ever-evolving. Over the past 125 prompt return to normal activities. This broadened
years, its use has varied widely, depending on the list of goals has led many surgeons to question the
intent of therapy (curative, palliative or compro- dogma that all normal-risk patients with a potentially
mised); specific treatment goals; the safety, effec- curable invasive cancer are best served by standard
tiveness and local availability of alternative forms of radical resection (anterior resection or APE). Instead,
therapy for rectal cancer; and the decision-making they suggest a policy that uses LE in the curative-
process and treatment philosophy of those involved intent treatment of patients with selected stage I rectal
in choosing an optimal treatment plan, includ- cancer. Although this approach is highly controver-
ing the patient and the surgeon. At one time, LE sial, recent data confirm that US surgeons are increas-
through a posterior approach was the most com- ingly using LE for curative-intent treatment of rectal
mon method used to treat patients with rectal can- cancer, despite a lack of level I or II evidence that LE is
cer, for both curative and palliative intent. At other oncologically equivalent to standard radical resection.
times in the twentieth century, LE was rarely used Proponents of this practice justify their increased use
for curative-intent treatment of rectal cancer in of LE because it better achieves the other goals now
normal-risk patients, who instead were treated by considered important in treating patients with rectal
standard anterior resection or abdominoperineal cancer and because it is unclear that overall survival
excision (APE). Local excision was used primarily is uniformly better after radical resection for stage I
for palliation or in compromise situations, such as cancer compared with LE. In addition to its use as the
when a patient’s operative risks and comorbidities sole treatment modality in curative surgery for select-
made it too dangerous to undergo standard radical ed rectal cancer, LE is now being used with adjuvant
resection or because the patient refused the recom- or neoadjuvant therapies. These controversies and
mended anterior resection or APE. the indications, techniques and outcomes following
Today, the goals for treating rectal cancer have conventional endoanal LE and transanal endoscopic
broadened to include securing local and distant microsurgery (TEM) for selected patients with rec-
oncological control; minimizing treatment-related tal cancer are the subject of this chapter. The role of
mortality and morbidity; performing restorative LE in the treatment of benign rectal neoplasms and

HEBK001-C11_p171-190.indd 171 08/02/13 5:37 PM


172  Local excision and transanal endoscopic microsurgery

the potential negative consequences of LE of what is tomy under regional anaesthesia. Two weeks later,
thought to be a benign rectal neoplasm, but is found through a transperineal incision, Lockhart-Mum-
by histopathology on the excised specimen to har- mery performed a wide local resection of the anus
bour a focus of invasive adenocarcinoma, are beyond with its surrounding skin, the rectum, the contents
the scope of this chapter. of the ischiorectal fossas, part of the levators and as
much of the sigmoid with its mesentery and lymph
nodes as possible. In 1926, he reported a remark-
Historical evolution of ably low (for the time) operative mortality of 8.5
local excision techniques per cent in 200 patients.3 The Lockhart-Mummery
technique offered the potential advantages of a
Surgeons have devised multiple techniques of LE for more aggressive clearance of the cancer compared
rectal cancer that can be grouped broadly by opera- with the limited resection done with the Kraske
tive approach: posterior, perineal and endoanal. trans-sacral approach, but with significantly less
mortality than the 42 per cent reported in 1908 by
Miles after APE.4 It is interesting to note that Lock-
Posterior Approach hart-Mummery had used the Miles technique but
said he ‘had abandoned it since 1913, except for the
Local resection via the posterior approach was the high cancers at the rectosigmoidal junction’. For all
preferred treatment for most rectal cancers in the other cancers of the rectum, Lockhart-Mummery
late 1800s, when mortality following anaesthesia and preferred his transperineal approach because it ‘was
abdominal surgery was common. The posterior trans- attended with a lower mortality and was applicable
sacral approach was popularized in Europe by Kraske, to a larger proportion of cases’.
who described making a posterior longitudinal inci-
sion over the distal sacrum and coccyx to the midline
raphe of the perineum with the patient in the prone Endoanal Approach
jack-knife position.1 The rectal tumour was exposed
and resected with a 1 cm margin. The rectal wall defect One of the earliest descriptions of use of the endo-
was repaired and the wound closed. This became the anal approach to LE of rectal cancer was by von
most common approach for rectal cancer in Europe in Volkmann in 1878, a mentor to Kocher. He noted
the late 1800s and early 1900s. that it was possible to treat ‘a well-circumscribed
Mason subsequently modified this posterior tumour, with the removal of which requires exci-
approach by dividing the anal sphincters to achieve sion of a small portion of the rectum’, providing it
better exposure.2 The sphincter muscles were care- was possible to close the defect in the rectum pri-
fully preserved and tagged to facilitate suture repair marily by suture, preferably in a transverse fashion
in layers after excising the rectal lesion. Although to avoid the complication of a stricture.
still useful for several benign conditions, both the In 1977 Morson reported the St Marks Hospital,
Kraske trans-sacral and the Mason trans-sphinc- London, experience with LE of rectal cancers using
teric posterior approaches to local resection for the endoanal approach of Sir Alan Parks.5 His endo-
rectal cancer have now essentially been abandoned anal technique is the prototype of the conventional
because of associated operative morbidity (poste- technique used for LE worldwide. The morbidity is
rior faecal fistulas, anal incontinence), the increased markedly less than that observed after radical resec-
risk of incurable local recurrences, and the develop- tion. Technical details and outcomes are described
ment of safer, more effective alternative techniques. below. Transanal endoscopic microsurgery (TEM)
was introduced by Gerhard Buess of Tubingen,
Germany, in 1984.6 This is a modification of LE
Perineal Approach that combines the excellent visualization offered by
endoscopy with advanced-instrument technology.
Lockhart-Mummery advocated a two-stage The technique allows for improved endoanal access
approach to the local resection of rectal cancer.3 to the mid- and upper rectum, thus increasing the
The first stage consisted of constructing a colos- utility of LE. Visual imaging is achieved through a

HEBK001-C11_p171-190.indd 172 08/02/13 5:37 PM


Current techniques of local excision  173

binocular stereoscope, which permits an optimum pudendal nerve block assists with sphincter relaxa-
view during the procedure, thus enhancing the sur- tion and postoperative analgesia. A Lone Star (Lone
geon’s ability to accurately perform full-thickness Star Medical Products, Inc., Stafford, TX, USA)
excisions and to repair the rectal wall defect. The retractor is used to efface the anus and facilitate
greater accuracy is reported to reduce the incidence exposure of the distal rectum. Retractors of the sur-
of positive resection margin and local recurrence geon’s choice are used to dilate the anus and expose
rates compared with conventional endoanal LE.7 the cancer. A fibre-optic headlight or a retractor
As with conventional LE, TEM causes less opera- with a light source is needed to adequately visualize
tive morbidity and mortality than radical resection. the lesion within the rectum.
Technical details and outcomes are described below. After exposure is achieved, electrocautery is
used to define a 1-cm circumferential margin
around the tumour. Traction sutures may be
Current techniques placed in the normal tissue around the tumour
of local excision to facilitate better exposure and manipulation of
the lesion. Care should be taken not to trauma-
Surgeons continue to evolve techniques for LE. tize the lesion or place sutures into the cancer.
Today, the posterior approaches have essentially A full-thickness excision is performed with the
been abandoned for treatment of rectal cancer, dissection extending into the surrounding mes-
while the endoanal LE and TEM approaches have orectal fat. One must avoid injury to the vagi-
increased in popularity. Both endoanal LE and TEM na in female patients and the prostate in male
are increasingly used in conjunction with chemo- patients. Once the specimen is fully excised, it
radiation in an attempt to improve the outcomes should be orientated and pinned before sending
achieved by these surgical techniques when used as it to pathology. After ensuring haemostasis, the
sole therapy for select rectal cancer. Zerz and col- defect is closed transversely to minimize risk of
leagues described a new combined approach, the stricture. For large defects, a sleeve anastomosis
endoscopic posterior mesorectal resection (EPMR), with interrupted absorbable sutures can be used
designed to allow sampling of the mesorectal nodes to advance the proximal rectum over the defect.
while maintaining the advantages of low morbid- Although it is not our practice, if the rectal open-
ity and negligible mortality of LE.7 More recently, ing is below the peritoneal reflection, it can be
Atallah and colleagues described an LE technique left open with no added morbidity.10 A proctos-
using a single-incision laparoscopic port.8 These copy examination is performed at the end of the
techniques are briefly described below. procedure to ensure the rectum is widely patent.
Postoperatively, antibiotics should be stopped
within 24 h. Patients may take a normal diet on
Endoanal Local Excision the evening of the operation. Postoperative anal-
gesia requirements are minimal and patients may
Endoanal LE, as described by Parks in 1968,9 is be discharged on the day of the operation.
the most popular technique of LE. The patient is Most complications after endoanal LE are
positioned depending on surgeon’s preference and minor, with rates ranging from zero to 22 per
the location of the tumour. An anterior tumour is cent.11 Bleeding, local sepsis, urinary tract infec-
best approached with the patient prone, whereas a tion or retention, rectovaginal fistula and pul-
posterior tumour, if distally located, may be better monary emboli are the most common complica-
accessed with the patient in the lithotomy position. tions. Lesions located more than 10 cm from the
Mid- and proximal posteriorly based lesions are dentate line can be difficult to expose and excise
best accessed with the patient in prone jack-knife in one piece with an adequate margin. If radical
position. A limited distal colonic bowel prepara- resection is indicated after LE, it is important to
tion and preoperative antibiotics are generally used. wait at least a month for the wound to contract
Preoperative deep venous thrombosis prophylaxis and heal, otherwise the wound could dehisce
is recommended. The procedure may be performed during rectal mobilization, thus potentially spill-
under general or regional anaesthesia. The use of a ing tumour cells or faeces into the operative site.

HEBK001-C11_p171-190.indd 173 08/02/13 5:37 PM


174  Local excision and transanal endoscopic microsurgery

Transanal Endoscopic Microsurgery trocautery, one can use a variety of commercially


available products such as a harmonic scalpel that
Transanal endoscopic microsurgery has gained can simplify and speed up this step of the opera-
increasing popularity and was developed to allow tion. One must also remember that the rectoscope
excision of more proximal lesions than those ame- should be repositioned several times during the dis-
nable to endoanal LE. The operation is performed section to keep the lesion in the centre of the opera-
through a 40-mm operating rectoscope (12 cm and tive field. If the peritoneum is violated, it should
20 cm in length), which is insufflated with carbon be repaired promptly, but this does not mandate
dioxide (CO2) to obtain vision of the proximal rec- immediate conversion to laparotomy. If pneumor-
tum. Flow can be increased to a rate of 6 L/min ectum is adequately maintained, one can proceed
and intra-rectal pressure is continuously moni- with excision. With regard to the suturing tech-
tored and maintained at 12–15 cmH2O. Specially nique, sutures are started and finished with silver
designed instruments are introduced through the shots applied to the thread with a specially designed
rectoscope’s working ports, similar to the tech- applicator because traditional knot-tying is very
nique used for laparoscopic surgery. Transanal difficult in the rectum. Endoscopic suturing devices
endoscopic microsurgery offers endoscopic mag- (e.g. Endo Stitch™) are also being used for closure
nification and illumination that provide excellent of the rectal wall defect left after the lesion has been
visualization as well as multiple instruments that excised; these add significant cost to the procedure,
allow precise resection and secure suture closure. however. Short sutures are preferable, and bisect-
This is a particular advantage in patients with a ing the wound with an interrupted suture may also
large body habitus, for patients with a proximally facilitate closure. Compared with endoanal LE, CO2
based rectal cancer, and when performing excision distention greatly facilitates closure of the wound.
of locally recurrent lesions. A limited distal colonic Most patients are discharged home the same day or
bowel preparation and preoperative antibiotics are the day following the procedure.
generally used. Bowel preparation is of particular Essentially, any lesion, regardless of size, location
importance if penetration into the peritoneal cav- and degree of circumferential involvement, can be
ity should inadvertently occur. Informed consent removed with TEM, as long as the upper and lower
should include possible laparotomy for lesions at margins of the lesion can be reached and completely
high risk for entering the peritoneum, specifically visualized. After excision of lesions that involve the
anterior tumours in female patients where the loca- entire rectal circumference, intestinal continuity
tion of the anterior peritoneal reflection is unpre- can be re-established with a hand-sewn end-to-end
dictable and may be quite distal. anastomosis performed through the rectoscope.
The rectoscope is inserted up to the lesion under Distal rectal lesions are difficult to excise with TEM
direct vision with manual insufflation and then because of the difficulty obtaining and maintain-
secured to the operating table. The bevel of the ing pneumorectum and difficulty using the instru-
scope must face down at the lesion. The binocular ments in the distal rectum. Whereas exposure in
eyepiece and the accessory scope are inserted. Ade- laparoscopy can be facilitated by inserting addition-
quate pneumorectum should be obtained without al ports or to insert the scope from a different angle,
signs of an air leak. The entire lesion should be vis- this is not possible with TEM. The instruments are
ible, although if not, one can still proceed as long as inserted and manipulated only in parallel, and the
the entire lesion can be pulled down into the opera- scope is fixed in one position. Transanal endoscopic
tive field. Excision of the lesion follows the same microsurgery is thus associated with longer opera-
guidelines as standard endoanal LE. Partial-thick- tive times for such distal lesions compared with tra-
ness or full-thickness excisions can be performed ditional endoanal LE.
with this approach, depending on the location and Disadvantages of TEM include costly equipment
nature of the lesion. In general, cancers should be and slightly longer operating times. Although TEM
removed by a full-thickness excision with a 1 cm was thought to have a steep learning curve, more
margin. Most of the bleeding during full-thickness recent reports have suggested a short learning curve
excisions occurs during the mesorectal dissection. for the procedure in surgeons with prior experience
In addition to conventional haemostasis with elec- in minimally invasive laparoscopic surgery. As with

HEBK001-C11_p171-190.indd 174 08/02/13 5:37 PM


Historical role and outcomes of local excision  175

endoanal LE, TEM is also a safe procedure, with a ping of the superior rectal artery (Figure 11.1).7
low rate of complications. Reported complications Its proponents claim this technique provides
include entry into the peritoneal cavity, conversion to complete tumour staging with minimal morbid-
laparotomy, bleeding, pneumoscrotum, perineal pain, ity after LE of T1 rectal cancers.
rectal stenosis, wound dehiscence, urinary dysfunc-
tion and rectovaginal fistula. Kreissler-Haag and col-
leagues assessed complications in 288 patients under- Laparoscopic Local Excision
going TEM. They reported that tumour localization
higher than 8 cm, tumour diameter of more than Atallah and colleagues reported their experience
2 cm, and localization of the tumour on the lateral with using a single-incision laparoscopic surgery
wall of the rectum were risk factors for surgical com- port for access to the rectum, replacing the con-
plications, mainly bleeding.12 Although there has been ventional operative rectoscope and using ordinary
concern about anal sphincter injury from prolonged laparoscopic instruments to perform LE.8 The
stretch, early studies documented no lasting adverse authors found this technique to be a safe and fea-
effect on anorectal function. Most individual TEM sible alternative to TEM, providing its benefits at a
case series report mild transient incontinence, which fraction of the cost (Figure 11.2).
usually resolves within weeks. Dafnis and colleagues
reported impaired continence in 18 of 48 patients
(37 per cent) at a median follow-up of 22 months.13 Historical role and
These patients did not report improvement in con- outcomes of local excision
tinence over time (pre- v. post-1 year assessment),
which conflicts with other studies reporting signifi- In the late eighteenth and early nineteenth century,
cant improvement within weeks.14,15 Radiotherapy LE via a posterior approach to the rectum was the
may be another factor affecting functional results after most common method of treatment of rectal can-
TEM, since radiation has negatively impacted func- cer. Surgeons adopted this practice because operative
tion when used in conjunction with radical surgery.16 mortality following anaesthesia and abdominal sur-
Data regarding functional outcomes after radiothera- gery was common. Their goals were simple: to resect
py plus TEM are scant and not well defined. Patients the tumour and keep the patient from dying from the
should be aware that significant changes in continence operation. During the first half of the twentieth cen-
may occur in the early postoperative period. Potential tury, mortality after elective colorectal cancer surgery
risk factors include pre-existing decreased continence fell steadily from 41 per cent in 1916–1920 to 7 per
or sphincter defects and large lesions requiring a more cent in 1946–1950.17 As anaesthesia and abdominal
extensive resection and prolonged operating time. The surgery became safer, surgeons embraced the Miles
majority of patients experience a return of pre-proce- APE because of its superior oncological control com-
dure continence at 6 weeks to 3 months. The long-term pared with that achieved by LE. Abdominoperineal
effects of an irradiated rectum after TEM remain to excision became the operation of choice for rectal
be defined. and rectosigmoid cancers in most centres around the
world. In the 1950s, anterior resection and colorectal
anastomosis was shown to achieve oncological out-
Endoscopic Posterior comes identical to APE for distal sigmoid and proxi-
Mesorectal Resection mal rectal cancers without the morbidity of a perma-
nent colostomy.
One significant disadvantage of LE or TEM is the By the 1970s, mortality following radical resec-
lack of information regarding the lymph node tion for rectal cancer had dropped to 5 per cent or
status of the mesorectum. Endoscopic posterior less in most series. A reliable circular stapler that
mesorectal resection includes TEM or endoanal facilitated distal colorectal anastomosis was intro-
LE of selected, favourable T1 rectal cancers fol- duced in 1978. Soon, anterior resection became the
lowed by a minimally invasive transperineal procedure of choice for all but the most distal rec-
resection of the posterior part of the mesorectum, tal cancers, as surgeons pushed the distal limits of
including all relevant lymphatic tissue and clip- sphincter-sparing proctectomy. The goals of surgery

HEBK001-C11_p171-190.indd 175 08/02/13 5:37 PM


176  Local excision and transanal endoscopic microsurgery

Figure 11.1. Technique of endoscopic posterior mesorectal resection. (A) Trocar positions. (B) Access to the retrorectal
space using the index finger. (C) Establishment of a sufficiently large operating space using a dissecting balloon trocar.
(D) Dissection of the mesorectum from the posterior wall of the rectum.

for rectal cancer had expanded beyond safety and tal excision (TME) as part of anterior resection
local oncological control to also include sphincter or APE.19 Various technical modifications to ante-
preservation and avoidance of permanent colosto- rior resection and APE are now included under the
my. Local excision of rectal cancer seemed relegated term TME. Proponents argued that the surgeon
to use only in high-risk patients, in patients refus- could achieve better oncological outcomes, increase
ing radical resection and for palliation. A dissent- sphincter preservation with coloanal anastomosis,
ing opinion came in 1977, when Morson and col- improve anorectal function by using a colo-pouch,
leagues reported the St Marks experience with LE and maintain genitourinary function by using a
of rectal cancers using the endoanal approach of Sir meticulous pelvic dissection technique.
Alan Parks.18 They reported equivalent oncological The role of LE in treatment of rectal cancer varied
results after LE compared with radical resection but widely from centre to centre in the 1980s. Some sur-
with significantly less morbidity and mortality. geons rarely used LE for any indication, while oth-
In the 1980s, reports of high rates of local recur- ers, noting the significant morbidity and less than
rence and major operative morbidity, including ideal oncological results following standard anterior
anastomotic leak, anterior resection syndrome resection and APE, increasingly used LE as an alter-
and genitourinary dysfunction, after low anterior native in patients marginally unfit for radical resec-
resection and distal anastomosis caused physicians tion. At the same time, new and improved imaging
to re-examine the preferred approach to rectal techniques, including endorectal ultrasonography
cancer. Radiation therapists and medical oncolo- (ERUS), magnetic resonance imaging (MRI) and
gists argued for postoperative chemoradiation to computed tomography (CT), held the promise of
enhance oncological outcomes. Heald and others being able to reliably and accurately stage rectal
held workshops to teach surgeons how to optimize cancer, thus minimizing the risk that the use of LE
surgical technique by conducting a total mesorec- would undertreat a patient with rectal cancer. Our

HEBK001-C11_p171-190.indd 176 08/02/13 5:37 PM


Current role and outcomes of local excision  177

from zero to 28 per cent for T1 lesions and from 11


per cent to 45 per cent for T2 lesions, whereas 5-year
survival rates ranged from 74 per cent to 90 per cent
for T1 lesions and from 55 per cent to 75 per cent for
T2 lesions. The wide variation in reported outcomes
probably reflects differences in patient selection,
intent of surgery and surgical technique. We also
found that radical salvage surgery in patients with
local recurrence after LE was not always curative.24
It seemed that LE did not equal the oncological out-
comes historically achieved with standard anterior
resection or APE for similarly staged cancers. As a
result, the use of LE for curative-intent treatment of
rectal cancer was curtailed in many centres, includ-
ing our own.

Current role and outcomes


of local excision

The current role of LE for treating patients with


rectal cancer is controversial. Consensus opinion
as noted in practice guidelines and surgical text-
books is that the standard curative-intent operation
Figure 11.2. Transanal minimally invasive surgery (TAMIS).
Single-port access is used to facilitate minimally invasive
of choice for normal-risk individuals with rectal
surgery so that transanal excision of rectal lesions cancer including stage I lesions is radical resection
can be performed. This is a novel approach that is a (anterior resection or APE) performed with the
hybrid between single-port laparoscopy and transanal meticulous dissection and other techniques now
endoscopic microsurgery. Shown here is the SILS™ Port encompassed in TME. This is supported by data
in position within the anal canal (A). Insufflation of the such as those offered by Blumberg and colleagues.25
rectum is established with a dedicated port. Through the They reported that patients with stage I rectal can-
remaining three cannulas, two are used for laparoscopic cer who undergo radical anterior resection with
instrumentation and one is used for a 5-mm camera (B). sphincter-sparing anastomosis or radical abdomi-
noperineal excision of the rectum with colostomy
experience with increased use of curative-intent LE generally benefit from a high cure rate, with 5-year
for marginally unfit patients was consistent with survival rates around 90 per cent. Given this con-
Morson’s favourable report.18 As a result, we slow- sensus, the appropriate role of LE in curative treat-
ly began using LE for curative-intent treatment in ment of stage I rectal cancer would seem to be very
normal-risk patients. We initially selected what we limited. It is thus surprising that You and collabora-
thought were only the most favourable, small T1 tors reported that LE had been used increasingly for
cancers for LE; when results seemed reasonable, we stage I rectal cancer.26
gradually liberalized our selection criteria and began One may speculate as to why LE has increased
including selected T2 cancers. We were dismayed in use in the USA. It is possible that the increased
when our longer-term follow-up showed high rates enrolment in national screening programmes for
of local recurrence following LE of both T1 and T2 colorectal cancer has led to diagnosis of a higher
lesions.20,21 Soon, results from other large single insti- proportion of early-stage rectal cancers, includ-
tutions that had similarly liberalized the use of LE ing rectal cancers arising in sessile polyps, some of
also showed higher than expected local recurrence which may be potentially amenable to LE. Addi-
rates; some studies showed a decrease in overall sur- tionally, new neoadjuvant treatment regimens may
vival following LE.22,23 Local recurrence rates ranged lead to more effective down-staging of stage II and

HEBK001-C11_p171-190.indd 177 08/02/13 5:37 PM


178  Local excision and transanal endoscopic microsurgery

stage III rectal cancers, potentially making some cal resection). The authors concluded that there is
patients whose cancers demonstrate a complete or an increased use of LE for stage I rectal cancer, but
near-complete clinical response amenable to cur- there are associated risks of local failure and the
ative-intent LE.27–31 Local excision in combination benefits of LE must be balanced against such risks.
with adjuvant or neoadjuvant chemoradiotherapy Winde and colleagues randomized 50 patients
for early-stage rectal cancer is being assessed in ran- with T1 rectal cancer to either TEM or anterior
domized trials. Its use in compromise situations and resection.32 At a mean follow-up of 46 months, there
for palliation is more difficult to assess but is likely was no significant difference in the local recurrence
increasing. With an ageing population, there may rate (4.2 per cent v. 0 per cent) or survival rate (96
be an increase in the number of patients for whom per cent v. 96 per cent) between the two groups.
local therapy is selected as a compromise treatment Lezoche and colleagues randomized patients with
rather than selecting a conventional radical treat- T2N0 rectal cancer to TEM or laparoscopic stand-
ment protocol for an elderly patient with significant ard radical resection with TME after neoadjuvant
operative risk factors. The extent to which any of chemoradiation therapy. Local recurrence (5.7 per
these possible explanations underlies the apparent cent v. 2.8 per cent) and survival (94 per cent for
increased use of LE is unclear. Regardless, it is obvi- both groups) were not significantly different after
ous that the current role of LE in the treatment of 84 months of follow-up.33
rectal cancer is still being defined. For proponents of LE, the apparent lack of a del-
eterious effect of choice of therapy on 5-year over-
all survival was reassuring and added credence to
Curative-Intent Local Excision their opinion that LE should play a significant role
in curative-intent treatment of select rectal cancers.
You and colleagues used the National Cancer Data- Additional support for this view derives from the
base to examine choice of surgical therapy over time observation that radical surgery does not cure all
for 35 179 patients diagnosed with stage I rectal patients with stage I disease and is associated with a
cancer between 1989 and 2003.26 They found that 30-day postoperative mortality of 1.6–4.8 per cent,
LE increased from 26.6 per cent to 43.7 per cent for an often prolonged recovery and major ­morbidity.34
T1 cancers and from 5.8 per cent to 16.8 per cent Anastomotic leak, pelvic sepsis, perineal wound
for T2 lesions. This is a remarkable trend given the problems, colostomy complications, and bowel,
lack of level I or II evidence that LE is oncologically sexual and urinary functional disturbances may
appropriate for curative-intent surgery of stage I adversely affect the patient’s long-term quality
rectal cancer. They further analysed 2124 patients of life. By contrast, LE of rectal cancer will confer
with stage I rectal cancer diagnosed between 1994 undisputed benefits of negligible mortality, mini-
and 1996 to assess perioperative outcomes, local mal morbidity with rapid recovery, and reasonable
recurrence and survival. Local excision was used preservation of genitourinary and anal sphincter
in 765 patients (601 with T1 cancer, 164 with T2 function. In a recent update of the Cancer and
cancer) and standard radical resection was done in Leukemia Group B trial on local excision of distal
1359 patients (493 with T1 cancer, 866 with T2 can- rectal cancer (CALGB 8984), Greenberg and col-
cer). Patients treated by LE experienced significant- laborators presented data supporting the concept
ly lower 30-day morbidity than patients undergo- that patients with selected T1 cancers treated by LE
ing standard radical resection (5.6 per cent v. 14.6 have comparable rates of local recurrence, overall
per cent). The 5-year local recurrence rate after LE survival and disease-free survival as historic con-
versus radical resection, however, was significantly trols treated by radical resection.35 As noted in the
higher (12.5 per cent v. 6.9 per cent for T1 cancers; discussion about Greenberg and colleagues’ paper,
22.1 per cent v. 15.1 per cent for T2 cancers). The the use of historic controls and other design issues
5-year overall survival, however, was influenced by make this a less than totally convincing argument
age and comorbidities but not by choice of surgery to routinely use LE for selected stage I rectal can-
in both T1 cancers (77.4 per cent after LE v. 81.7 cers. The critical and still unresolved controversial
per cent after radical resection) and T2 cancers issues are: does LE for selected stage I rectal can-
(67.6 per cent after LE v. 76.5 per cent after radi- cers provide oncological outcomes equivalent to

HEBK001-C11_p171-190.indd 178 08/02/13 5:37 PM


Current role and outcomes of local excision  179

that achieved by radical resection, and if not, do the tumours), no local recurrences occurred, and only
advantages of LE outweigh the oncological disad- 1 patient with T2 disease had distant recurrence.
vantages? All local recurrences were managed surgically. The
5-year disease-specific survival rate was 100 per
cent for Tis tumours, 100 per cent for T1 tumours
Curative-Intent Transanal and 70 per cent for T2 tumours.
Endoscopic Microsurgery
Local recurrence after TEM has been reported Comparisons of Curative-Intent
mainly in single-institution reviews with lim- Endoanal Local Excision and
ited numbers, which makes comparisons difficult. Transanal Endoscopic Microsurgery
Recurrence rates after TEM range from zero to 13
per cent for patients with T1 tumours and from Although You and colleagues did not distinguish
zero to 80 per cent for patients with T2 tumours. traditional endoanal LE from TEM,26 there is
Bach and colleagues reported the outcomes of 424 increasing literature comparing the two techniques.
patients with rectal cancer treated with TEM and Unfortunately, most studies are retrospective single-
entered into a national database.36 A positive resec- institution experiences (Table 11.1). Moore and col-
tion margin occurred in 11 per cent, 22 per cent and leagues compared endoanal LE (n 5 89) and TEM
42 per cent of patients with T1, T2 and T3 tumours, (n 5 82) and showed no differences in complica-
respectively. Patients who were observed after TEM tions.5 Transanal endoscopic microsurgery was more
(with or without postoperative radiation) were 15 likely to yield clear margins and a non-fragmented
times more likely to develop local recurrence than specimen. At a mean follow-up of 37 months, recur-
those who were converted to standard radical resec- rence was less frequent after TEM than after tradi-
tion (anterior resection or APE) with TME based tional LE (5 per cent v. 27 per cent). At our insti-
on unfavourable histological findings in the TEM tution, we retrospectively compared endoanal LE
specimen. (n 5 129) and TEM (n 5 42). We evaluated quality
We analysed our results with TEM at the Univer- of resection, local recurrence and survival rates in
sity of Minnesota in 95 patients with rectal carci- patients with stage I rectal cancer.40 Although surgi-
noma (58 T1, 26 T2, 11 T3).37 Follow-up time was cal margins were less often positive with TEM, the
49.5 months. Local recurrence rates for 83 malig- estimated 5-year disease-free survival rate was simi-
nant tumours were 9.8 per cent for T1 tumours, lar between the groups (TEM 84 per cent v. endo-
23.5 per cent for T2 tumours and 100 per cent for anal LE 76 per cent). We found that tumour distance
T3 tumours. Ganai and colleagues studied the out- from the anal verge, resection margin status, T stage
comes of 28 patients with malignant rectal tumours and use of adjuvant therapy, but not surgical tech-
(8 T in situ (Tis), 12 T1, 4 T2, 4 T3) who under- nique (LE or TEM), were independent predictors of
went TEM.38 Margin positivity occurred in 14 per local recurrence and disease-free survival.
cent of patients (4/28). Mean follow-up time was 46
months. Local recurrence occurred in 15 per cent of
patients. Depth of invasion was related to the likeli- Curative-Intent Endoscopic Posterior
hood of invasive local recurrence, and lesions larger Mesorectal Resection
than 4 cm had a higher rate of recurrence. Median
time to recurrence was 15 months. The authors rec- Two years after the description of EPMR by Zerz
ommended close endoscopic follow-up after TEM. and colleagues, Tarantino and colleagues reported
Stipa and colleagues reviewed the outcomes of their experience with this promising technique.44
69 patients with early rectal cancer who underwent They performed EPMR 6 weeks after LE in 18 con-
TEM.39 The 5-year local recurrence rate was 8 per secutive patients with T1 tumours (13 endoanal LE,
cent for Tis tumours, 8.6 per cent for T1 tumours 5 TEM) and compared their results with historical
and 9.5 per cent for T2 tumours. The median time controls that underwent low anterior resection with
to recurrence was 10.5 months. Of 14 patients who TME. Both groups included patients with low- and
received preoperative chemoradiation (2 T1, 12 T2 high-risk histology with no significant differences.

HEBK001-C11_p171-190.indd 179 08/02/13 5:37 PM


180  Local excision and transanal endoscopic microsurgery

The median number of lymph nodes resected was

survival (%)
7 for EPMR and 11 for low anterior resection. Of

Overall
importance, an anatomical study found a mean of

NS
93
96

94
77
8.4 lymph nodes in the mesorectum.45 At a median
follow-up of 23 months, one patient developed dis-

recurrence
tant metastases but there was no evidence of local
recurrence in either group. They concluded that
Local

(%) although the lymph node harvesting of EPMR was

9.4

2.8
0

0
not comparable to that of TME, EPMR allowed for
TME

adequate lymph node staging and showed equiva-


Mortality

lent outcomes for T1 tumours. This technique may


Table 11.1. Studies comparing transanal endoscopic microsurgery (TEM) versus total mesorectal excision (TME) for early rectal carcinoma.

(%)

3.7
prove useful to accurately identify otherwise occult
0

0
4
metastatic mesorectal nodes and thus facilitate
appropriate selection of curative-intent therapy for
Complications

apparently-early stage rectal adenocarcinoma.


(%)
48

56
17
64

Curative-Intent Local Excision as Part


of Multimodality Therapy
22
83
18
35
75
n
survival
Overall

Dissatisfaction with oncological control offered by


100
(%)

NR

LE as the sole therapy for rectal cancer led some


95

94
75

centres to use postoperative radiation with or with-


out sensitizing chemotherapy. Advocates argued
recurrence

that by doing LE first, the exact depth of tumour


Local

invasion and histological features of the primary


19.5
(%)
4.1

5.7
10

24

tumour could be assessed properly. Some centres


chose to use adjuvant chemoradiation therapy for
Mortality
TEM

T1 cancers with unfavourable features or for T2


(%)

cancers. The hypothesis was that adjuvant therapy


0

0
0
0

following curative-intent LE would treat occult


Complications

local spread or occult lymph node metastases and


thus improve oncological outcomes without sub-
jecting the patient to the complications of radical
(%)
4.1

7.6

surgery. As neoadjuvant chemoradiation therapy


14
5

has been more accepted for use in advanced-stage


52
22
20
35
80

rectal cancers before radical surgery, its use was


n

suggested for treatment of small early-stage rec-


Follow-up

tal cancers in combination with LE. Its role in this


(months)
31–35

21–43

setting is now the subject of an ongoing national


84
64

trial in the USA. Advocates argue that one can use


local tumour response to estimate the likelihood
T stage

of persistent nodal disease. Whether this is true is


debated.
1
2
1
2
1

NR, not reported.

Adjuvant therapy
43
Lezoche et al.33
De Graaf et al.
42

A multi-institutional phase II prospective trial eval-


Langer et al.
41

uating adjuvant chemoradiation therapy after LE


Lee et al.
Study

divided patients into three groups based on the final


histopathological findings.46 Group 1 patients had T1

HEBK001-C11_p171-190.indd 180 08/02/13 5:37 PM


Current role and outcomes of local excision  181

Table 11.2. Studies assessing chemoradiotherapy before local excision of early rectal carcinoma.
Pathological complete Recurrence, local/
Study T stage response (%) distant (%) Survival (%)
T1 T2
Kim et al.27 26 73 4 92
Bonnen et al. 48
– 26 54 11 86
Lezoche et al.28,29 35 – 32 9 94
Callender et al. 49
– 47 49 21 79
Yeo et al.50 – 11 73 18 89
Garcia-Aguilar et al.51 77 – 43 NR NR

NR, not reported.

lesions with favourable histology and negative margins rence rates compared with LE alone (Table 11.2).
and were observed without adjuvant therapy. Group 2 One of the first reports on preoperative radio-
patients had T1 tumours with unfavourable histology therapy was by Mohiuddin and colleagues.47 Thirty
or lesions greater than or equal to T2 and received the patients with T3 tumours were treated with preop-
standard long-course dose of chemoradiation therapy. erative radiotherapy followed by LE. They reported
Group 3 patients were the same as group 2, except the a pathological complete response in 11 patients (36
resection margins were positive for malignancy, and per cent), a 5-year recurrence rate of 10 per cent
therefore they received a higher dose of chemoradia- and a survival rate of 83 per cent.
tion therapy. After a median follow-up of 6.1 years, Bonnen and colleagues compared the outcomes of
local-regional recurrences developed in 16 per cent of LE and radical resection with TME after preoperative
the patients (8/65). The risk of recurrence correlated chemoradiation therapy.48 Of 26 patients, 54 per cent
with T stage (T1, 4 per cent; T2, 8 per cent; T3, 23 per (n 5 11) had a pathological complete response and 35
cent) and the degree of involvement of the resection per cent had microscopic residual disease. The 5-year
margin. For the two groups treated with chemoradia- local recurrence rate in the patients undergoing LE
tion therapy, the 5-year actuarial freedom from pelvic was 6 per cent and overall survival was 86 per cent,
relapse was 86 per cent, comparing favourably with compared with 8 per cent and 81 per cent, respective-
historical controls treated with TME and suggesting a ly, for the TME group. The results of this study were
beneficial effect of chemoradiation therapy. updated with a median follow-up of 63 months.49
The Cancer and Leukemia Group B conduct- Ten-year actuarial local recurrence was not signifi-
ed a similar prospective multi-institutional trial cantly different between the LE and TME groups (10.6
comparing the outcomes of 59 patients with T1 per cent v. 7.6 per cent), and no significant difference
tumours treated with LE alone and 51 patients in survival was found between the groups. Kim and
with T2 lesions treated with LE and postoperative collaborators at the Lee Moffitt Cancer Center retro-
chemoradiation therapy.35 At a median follow-up spectively reviewed the outcomes of 26 patients who
of 7.1 years, the local recurrence rates were 8 per received neoadjuvant therapy followed by LE for T2
cent for T1 lesions and 18 per cent for T2 cancers. and T3 rectal cancers.27 Pretreatment ERUS staging
The 10-year actuarial overall survival and disease- included 5 T2N0, 13 T3N0, 7 T3N1 and 1 not done.
free survival rates were 84 per cent and 75 per cent Pathological complete and partial responses were
for T1 cancers and 66 per cent and 64 per cent for achieved in 9 of 26 (35 per cent) and 17 of 26 (65 per
T2 cancers, respectively. cent) patients, respectively. At a mean follow-up of
24 months, only one tumour with partial response,
Neoadjuvant therapy in a patient who refused APE, had recurred. None
Although most studies evaluating neoadjuvant of the tumours with complete pathological response
therapy have small numbers and are retrospective recurred. Nair and colleagues reported a recurrence
in nature, they do suggest that preoperative chemo- rate of 16 per cent after neoadjuvant chemoradiation
radiation therapy can indeed down-stage tumours therapy and endoanal LE for T2 and T3 rectal can-
subjected to subsequent LE and also lower recur- cers.52 The overall 5-year survival was 84 per cent in

HEBK001-C11_p171-190.indd 181 08/02/13 5:37 PM


182  Local excision and transanal endoscopic microsurgery

node-negative patients. The results of these studies operative risk for radical surgery. For the most part,
contrast with other series that have shown higher rates even patients with major illnesses can tolerate LE,
of lymph node positivity after TME as well as lower providing the rectal cancer does not exceed the
responses to chemoradiation therapy. These differenc- technical limits of the operation. Similarly, when
es probably reflect selection bias, with patients having patients refuse a recommended radical surgery, usu-
more favourable tumours likely undergoing LE. ally because of the refusal to accept a permanent
In a prospective randomized trial, Lezoche and col- colostomy, the surgeon may consider LE as an alter-
leagues compared TEM and TME after neoadjuvant native therapy. This can pose an ethical dilemma for
chemoradiation therapy in patients with T2N0 rectal the surgeon, since patients often cannot compre-
cancer.28,29 The overall pathological complete response hend that use of LE, even for relatively early-stage
rate was approximately 35 per cent in both groups. In rectal cancer as a compromise indication, may result
an update of this trial with a median follow-up of 84 in incurable painful local recurrence that often
months, 2 of 35 patients (5.7 per cent) had local recur- requires further palliative treatment. Local excision
rence in the TEM group and only 1 of 35 patients (2.8 for palliation is used selectively, but the cancer can-
per cent) had recurrence in the TME group. Disease- not exceed the technical limits of the operation.
free survival after 84 months of median follow-up was
similar (94 per cent) in both groups.
Overall, there seems to be a trend towards lower Patient and cancer selection
recurrence rates and higher disease-free survival for local excision
with neoadjuvant or adjuvant therapy compared
with LE alone. This may be more beneficial in high- Criteria for selecting patients with rectal cancer
risk histology tumours and T2 or T3 tumours. For who would benefit from LE vary depending on the
patients with a pathological complete response intent of therapy, the patient’s desires, the treatment
documented on the surgical specimen, the reported philosophy and techniques offered by the treating
rate of positive nodes in the mesorectum ranges physicians, and, most importantly, the stage and
from zero to 12 per cent. Therefore, LE may not characteristics of the primary cancer. That LE can
guarantee a curative resection, even in patients with cure some rectal cancers is not in dispute, but the
ypT0 tumours after chemoradiotherapy and LE. question is whether we have the ability to accurately
What needs to be defined is the risk of nodal metas- distinguish patients whose rectal cancer is almost
tasis in patients with clinically N0 tumours who certain to be cured by LE from patients whose rectal
become ypT0 after chemoradiation. cancer is unlikely to be cured by LE. There are risk
Another question that remains unanswered is factors that can be identified during pretreatment
the efficacy of neoadjuvant therapy in T2N0 rectal evaluation that are contraindications to curative
adenocarcinoma followed by LE. This is currently intent LE, but there is no widely accepted evidence-
being addressed by the American College of Surgeons based list of criteria that surgeons can use to select
Oncology Group trial Z6041, a multicentre phase II rectal cancers for curative intent LE. Clinical, path-
clinical trial using neoadjuvant chemoradiation ther- ological and imaging assessments of the primary
apy followed by LE in patients with ultrasound-staged rectal cancer are essential to determine whether LE
T2N0 rectal cancer. Preliminary results presented as is even to be considered as a viable option for cura-
an abstract reported a 98 per cent resection rate with tive intent treatment.
negative margins.51 Down-staging occurred in 49 of
77 patients (64 per cent), with a pathological complete
response rate of 43 per cent. Assessing the Primary Rectal Cancer
for Local Excision
Local Excision for Compromise Not all rectal cancers can or should be resected by
and Palliation LE. The surgeon must assess the primary rectal
cancer to determine whether LE is technically pos-
Local excision has long been used for patients sible and whether doing so is likely to achieve the
judged to have comorbidities that put them at high therapeutic goals. It is important that the surgeon

HEBK001-C11_p171-190.indd 182 08/02/13 5:37 PM


Patient and cancer selection for local excision  183

recognizes when not to attempt LE even for pal- offers the ability to more securely suture repair the
liation. Most surgeons agree that LE should be con- rectal wall defect for proximal lesions than is possi-
sidered only if the cancer is in an accessible part of ble with endoanal LE. Although tumour morphol-
the rectum and small enough to be totally excised ogy does not have independent prognostic value
in one piece with a 1 cm margin of normal rectum. when stratified by tumour stage,54,55 it is often used
Additionally, most surgeons would agree that ideally as a surrogate for depth of invasion, since most
the cancer is confined to the rectal wall (T1 or T2). ulcerated lesions are T3 or T4. Thus, many surgeons
Sometimes exceptions are made in cases of com- avoid LE for ulcerated cancers.
promised intent therapy or palliative intent therapy.
Clinical, pathological and imaging assessments are Histopathology
key elements to achieving pretreatment staging of Biopsy of the primary lesion may reveal microscopic
the rectal primary tumour. features associated with lymph node metastases,
local recurrences, and a poor prognosis such as lym-
Tumour size, location and morphology phovascular and perineural invasion or mucinous,
Careful digital rectal examination with proctos- signet ring or poorly differentiated histology. If these
igmoidoscopy to palpate and visualize the rectal are present on pretreatment biopsy, LE is generally
cancer can determine the lesion’s size, precise level contraindicated. The rates of local recurrence and
and location in relation to the anal sphincters and lymph node involvement in T1, grade 1, well-differ-
other structures, and the presence of gross features entiated tumours are 0–3 per cent, whereas T1, grade
associated with a poor prognosis. Although tumour 3 poorly differentiated tumours have 12 per cent
size is not a reliable predictor of depth of invasion nodal involvement and a 10–63 per cent chance of
or nodal metastasis, patients with tumours larger local recurrence.12,56
than 3–4 cm in diameter or involving more than Ulcerated mucinous cancers and lesions with evi-
40 per cent of the circumference of the rectum are dence of perineural and lymphovascular invasion
poor candidates for traditional endoanal LE, sim- are associated with local recurrence rates as high
ply because adequate exposure is difficult to obtain as 25 per cent.12,56 Chemoradiotherapy regimens
with conventional retractors.53 Proponents of TEM are less effective in tumours with undifferentiated
report that this approach provides better exposure histology and lymphovascular invasion.57 Tumour
and visualization, such that the size of the lesion is budding, defined as isolated cancer cells or nests
less of an issue than with endoanal LE. Although of cancer cells in normal tissue at the edge of the
tumour distance from the anal verge does not have main tumour, has been associated with lymph node
independent prognostic value when stratified by metastases in up to 25 per cent of patients with T1
tumour stage,54,55 it is an important consideration cancers and has been identified as a predictor of
if LE is being considered. Traditional endoanal LE worse survival independent of disease stage.58,59 The
provides reliable access to distal and most mid-rec- literature is unclear as to how often histopathol-
tal lesions, while TEM provides access to mid- and ogy of the final specimen obtained by LE results in
proximal lesions but is difficult to use for the most identification of one of these unfavourable features
distal cancers. An additional consideration for the not noted on pretreatment biopsy.
surgeon is whether the location of the cancer allows
full-thickness excision of the rectal wall without Depth of invasion, nodal metastases
entering the peritoneal cavity. Distal and posterior- and local spread
ly based mid-rectal cancers are extraperitoneal and Nodal metastases, direct intrapelvic spread and mes-
generally can be excised safely without much risk orectal spread are contraindications for curative intent
of peritonitis if the rectal wall defect fails to heal. LE in average-risk patients and relative contraindica-
Proximal lesions and anteriorly based mid-rectal tions for LE in other settings (palliation or compro-
cancers are likely to be intraperitoneal, and their mise). All of the risk factors discussed above are sur-
full-thickness excision may increase the risk of peri- rogate markers for nodal metastases or local spread,
tonitis if a leak occurs and risk other organ injury but the most important correlate is depth of invasion
to adjacent structures such as the vagina or pros- of the primary cancer (Table 11.3). At a minimum,
tate. Transanal endoscopic microsurgery reputedly tumours that are considered for curative intent LE

HEBK001-C11_p171-190.indd 183 08/02/13 5:37 PM


184  Local excision and transanal endoscopic microsurgery

Table 11.3. Morphologic features of favourable and Imaging studies used to stage rectal cancer
unfavourable T1 rectal cancers. include CT, MRI and ERUS. New-generation CT
Favourable/ Unfavourable/ scanning and surface-coil MRI are able to detect
low risk high risk tumour invasion outside the rectal wall,67 but they
Differentiation Well differentiated Poorly differentiated cannot reliably distinguish between a T1 and a T2
(G1–G2) (G3) lesion or between a benign lesion and a T1 lesion.
Submucosal 1 2–3 Endorectal ultrasonography and endorectal coil
  invasion or 3-tesla MRI can provide this level of detail, and
Lymphovascular No Yes
either of these studies should be performed in
  invasion
the evaluation of any patient considered for LE.
Although MRI is more expensive and less available
Size ,3 cm .3 cm
than ERUS, it does not suffer from the interobserv-
Wall circumference ,40% .40%
er variation that plagues endosonography, and it is
the optimal way to assess mesorectal invasion and
should be localized to the rectal wall (T1 and T2). determine whether the circumferential radial mar-
Nodal involvement occurs in 0–12 per cent of T1 gin is involved, threatened or free of tumour.
tumours and 12–28 per cent of T2 tumours.56,60 Kudo In a meta-analysis including data from 90 publi-
first introduced the concept of dividing T1 cancers cations, Bipat and colleagues found the sensitivity of
into three levels based on the extent of submucosal ERUS and MRI for tumour invasion outside the rec-
invasion, varying from the superficial third to the tal wall was as high as 90% and 82%, respectively.68
middle third to the deepest third (sm1, sm2, sm3).61 The sensitivity for lymph node involvement was sig-
Several studies have demonstrated an association of nificantly lower, however, at 67 per cent and 66 per
deeper submucosal invasion with increasing risk of cent, respectively. In a systematic review of 53 studies
lymph node metastases in 0–3 per cent for sm1 lesions including 2915 patients, the accuracy of ERUS was 87
to 20–23 per cent for sm3 lesions,53,62 with increased per cent for T stage and 74 per cent for lymph node
rates of local recurrence for sm3 lesions.63 Additional- involvement;69 for MRI with endorectal coil, the cor-
ly, gender may be a predictive marker for lymph node responding numbers were 84 per cent and 82 per cent.
metastasis in early rectal cancer. Kobayashi and col- Data have shown that three-dimensional reconstruc-
leagues showed that 1 per cent of male patients with tion increases the accuracy of ERUS in assessing the
well-differentiated T1 tumours of the lower rectum depth of rectal wall and submucosal invasion and may
had lymph node metastasis, compared with 30 per help in selecting patients for local excision.70
cent in female patients with histological types other Because the gold standard for assessing the accu-
than well-differentiated adenocarcinoma, even when racy of any imaging study should be the final non-
the tumour did not invade the muscularis propria.64 irradiated pathology specimen, many studies are
The authors suggested that these patients should not biased by selective inclusion and exclusion criteria.
undergo LE. Patients with advanced disease who undergo neoad-
Serum carcinoembryonic antigen may be useful juvant therapy are generally excluded, and patients
as a baseline study. A CEA level that is elevated and with clearly localized lesions who are offered LE are
drops to normal after resection is reassuring. Per- similarly often excluded. Thus, the patients left for
sistence of an elevated CEA level after treatment or evaluation are those with more equivocal findings
a rising CEA level should alert the surgeon to the on imaging studies. This selection bias has likely led
possible presence of metastatic disease or incom- to an underestimation of the accuracy of ERUS and
plete resection. Perez and colleagues showed that a MRI.
CEA level below 5 ng/mL after chemoradiotherapy
is a favourable prognostic factor for rectal cancer
and is associated with increased rates of earlier Assessing the patient with
disease staging and complete tumour regression.65 rectal cancer
Conversely, an elevated pre-chemoradiotherapy
serum CEA level (. 5 ng/mL) is associated with The goals of the pretreatment evaluation are to
poor tumour response to chemoradiotherapy.66 identify conditions and issues that will affect the

HEBK001-C11_p171-190.indd 184 08/02/13 5:37 PM


Follow-up and salvage therapy  185

choice of therapy. In addition to carefully staging excuse to avoid a rigorous evidence-based decision-­
the primary rectal cancer as noted above, distant making process for each patient presenting with
metastases, especially to the liver and lungs, and rectal cancer. This admonition is especially relevant
synchronous gastrointestinal or colonic abnormali- to the controversies regarding choice of LE versus
ties, diseases or prior surgery that may complicate radical resection for rectal cancer.
the treatment plan must be assessed. Pre-existing Although the issues regarding appropriate choice
comorbidities, baseline bowel function includ- of curative intent LE to avoid over- and undertreat-
ing continence and defecation and operative risks ment are not resolved, pretreatment review by a
can largely be defined by a thorough history and multidisciplinary team of rectal cancer experts from
physical examination, with additional laboratory or different disciplines (colorectal surgery, medical
other testing as needed. The surgeon must under- oncology, radiation oncology, pathology, diagnostic
stand the patient’s desired outcome and assess their radiology) is recommended to minimize the risk of
psychosocial state, decision-making capability, and personal bias or incorrect interpretation of imaging,
availability of family or others for support during pathology and other relevant data (Figure 11.3). In
the stressful periods likely to arise as the rectal can- addition, this approach couples the pretreatment
cer is treated. A complete review of the pretreat- assessment with the skill, experience and judgement
ment assessment is beyond the scope of this chapter of the treating physicians who have knowledge of
but is available elsewhere.71 locally available treatment modalities and outcomes
achieved in the past as well as the patient’s specific
situation. Intent of therapy (curative or palliative)
Optimal therapy selection: and the occasional need to compromise a preferred
role of a multidisciplinary treatment for a patient whose comorbidities create a
team prohibitive operative risk for radical surgery can be
clarified. A final recommendation for the patient’s
Despite new and improved imaging, pretreatment treatment can be made, informed consent can be
staging of rectal cancer has inherent significant obtained and a treatment protocol implemented.
inaccuracies, especially in detecting lymph node
metastases and making subtle distinctions between
T stages. Thus, what is thought to be a stage I rectal Follow-up and salvage
cancer may actually be a stage II or stage III rectal therapy
cancer. This means that the decision to use endo-
anal LE or TEM, techniques characterized by ‘total The first decision to make following curative
mesorectal neglect’, for an alleged stage I rectal can- intent LE of a rectal cancer is whether the intend-
cer rather than performing a standard radical resec- ed goal is likely to have been achieved. Borschitz
tion with TME may unwittingly compromise onco- and colleagues evaluated and compared oncologi-
logical outcomes. Since nodal metastases occur in cal outcomes of patients undergoing immediate
0–12 per cent of T1 cancers and 12–28 per cent of reoperation versus salvage surgery after LE in the
T2 lesions,56,60 there is a possibility of undetected setting of unsuspected T2 disease or unfavourable
nodal involvement, even in T1 tumours. Inaccu- ­histology.30,31 At a median follow-up of 10 years,
racies in staging can result in undertreatment of patients who underwent salvage surgery had sig-
occult cancer spread to the mesorectum, mesenteric nificantly higher recurrence rates (37 per cent v.
lymph nodes or other sites. Such undertreatment 8 per cent) and significantly lower tumour-free
will inevitably result in increased local recurrences survival (54 per cent v. 86 per cent). The median
and decreased survival that likely would not have time to local recurrence for patients undergoing
occurred had standard radical resection been used. salvage surgery was 12 months. Lee and colleagues
Whether the technique of EPMR can overcome this also reviewed the outcomes of 36 patients with
shortcoming in staging lymph nodes accurately unfavourable histology and positive resection mar-
remains to be seen.9,44 gins who underwent TEM.72 Of the 36 patients, 12
These facts simply reflect the state of current underwent salvage surgery and 24 either refused
medical knowledge and cannot be used as an radical resection or had poor functional status and

HEBK001-C11_p171-190.indd 185 08/02/13 5:37 PM


186  Local excision and transanal endoscopic microsurgery

CT, computed tomography; ERUS, endorectal ultrasound; LE, local excision; MRI, magnetic resonance imaging;
SM1, superficial (third) submucosal invasion; TAE, transanal excision; TEM, transanal endoscopic microsurgery; TME,
total mesorectal excision.

Figure 11.3. Multimodality treatment algorithm for early rectal carcinoma.

would not tolerate a major operation. Of the 12 reoperation have been reported, they are infre-
patients who underwent salvage surgery, 1 had sys- quent. We have found that radical reoperation a few
temic recurrence. Of the 24 patients who did not weeks following LE has been safe.
have radical surgery, 5 had recurrence (3 local, 2 There is no agreed best practice protocol to fol-
distant). Thereafter, many have supported the role low patients long term after apparent successful LE
of immediate salvage surgery after TEM in patients of rectal cancer. Since local recurrence is a major
with unfavourable histology, positive excision mar- concern following LE of rectal cancer, it would seem
gins and a threatened resection margin (, 1 mm), reasonable to regularly perform digital rectal, proc-
reducing the recurrence rate to 6 per cent.73 toscopic and ERUS examinations. Unfortunately,
Based on such data, most surgeons agree that if even with careful follow-up, it is difficult to detect
final pathology after LE or TEM does not confirm residual disease as a curable local recurrence. Many
a rectal cancer with the anticipated favourable fea- tumours recur with unresectable local tumours or
tures and with clear margins (i.e. deeper invasion incurable distant metastases. Patients who are able
than anticipated, unfavourable histology features or to undergo curative intent therapy often require
a positive margin), then further treatment is indi- multimodality therapy with chemoradiotherapy
cated, by radical resection or chemoradiotherapy, and extensive resections. Five-year survival rates
or both. A potential concern when LE has to be range from 50 per cent to 58 per cent after salvage
converted to radical resection with TME is that of surgery and are well below what is expected after
a violated mesorectal plane, which may result in a initial radical surgery with TME. At the University
suboptimal TME. Although recurrences after TME of Minnesota, we followed patients after LE with

HEBK001-C11_p171-190.indd 186 08/02/13 5:37 PM


CONCLUSION  187

digital rectal examination, proctoscopy and ERUS Strong advocates for the expanded use of LE
every 4 months. Despite this intensive surveillance, or TEM for more advanced lesions emphasize the
27 of the 29 patients (93 per cent) we identified small but significant mortality, the high morbid-
with recurrence after LE presented with advanced ity, the long-term functional disabilities and the
disease (stage II or higher).24 Curative resection slow recovery associated with traditional radical
(R0) was possible in only 79 per cent of patients. Of resection. By contrast, they point to the negligible
note, 81 per cent of the patients were asymptomatic mortality, low morbidity, generally excellent func-
at the time of their diagnosis. tion and prompt return to normal life activities
after LE or TEM. They note also that many stud-
ies have shown no significant difference in survival
Surgeon’s philosophy and that new techniques, including the minimally
invasive sampling of mesorectal nodes and combi-
Given the controversies and evolving data regarding nation therapy with chemoradiotherapy, may offer
the use of curative intent LE or TEM, it is not sur- expanded opportunities and improved oncological
prising that colorectal surgeons have widely varying outcomes for these approaches.
philosophies about their roles in normal-risk indi- Endoanal LE or TEM may also be offered to
viduals with rectal cancer. Some surgeons, discour- patients who are medically unfit for a major opera-
aged by our current inability to accurately stage the tion or who refuse radical resection or a colostomy,
T and N stage of rectal cancer in the pretreatment with the option of pre- or postoperative chemo-
evaluation and chastened by the data revealing radiotherapy. In the setting of palliative excision,
compromised oncological outcomes, totally reject however, most primary tumours are bulky or sten-
endoanal LE or TEM as a viable option for all but otic and may not be amenable to LE.
compromised or palliative situations because of the
fear of undertreating a potentially curable rectal
cancer. They argue that radical surgery with TME Conclusion
is the treatment of choice for all rectal cancers,
including stage I lesions. The ideal therapy for rectal cancer cures the patient
Of the surgeons who do use curative intent LE or of the primary lesion and any distant metastases
TEM, most attempt to restrict their use at least as sole without treatment-related mortality or major mor-
therapy to highly selected, low-risk, small, T1 N0 M0 bidity, while preserving pretreatment bowel, sexual
rectal cancers. They argue that despite the inaccuracies and urinary function and allowing prompt return
in our best current imaging studies, they can usually to a high quality of life. Over- and undertreatment
avoid the significant risk of lymph node metastases are avoided. Unfortunately, the ideal is often not
in T2 cancers and the significant morbidity of radi- achievable, in part because there are numerous
cal surgery. Other surgeons attempt to be even more other factors still to be elucidated, such as tumour
restrictive by limiting curative intent LE or TEM to T1 biology, tumour–host interactions and genetics,
cancers with sm1 invasion and otherwise favourable which if known may influence our decisions and, in
features. The problem is that this approach assumes, part, because the information on which we base our
incorrectly, that we have reliable ways to identify pre- decisions is less than complete and less than totally
cise levels of T stage and can accurately assess lymph accurate.
node metastases before treatment. These surgeons The appropriate role of endoanal LE or TEM,
often view the initial LE or TEM as a first step in the or newer modifications of these techniques, for
treatment of such lesions. If final pathology after LE or treatment of rectal cancer is highly controversial.
TEM does not confirm a rectal cancer with the antici- Although radical resection with TME continues to
pated favourable features and with clear margins, they be the standard operation for most patients with
then proceed with further treatment by radical resec- rectal cancer, endoanal LE and TEM are acceptable
tion or chemoradiotherapy, or both. Whether this alternatives, with significantly less morbidity. Most
approach compromises survivorship that may have surgeons restrict their curative intent use to selected
been achieved had more aggressive therapy been used patients with T1 disease or to patients unfit for radical
initially is debated. resection. Compared with endoanal LE, TEM offers

HEBK001-C11_p171-190.indd 187 08/02/13 5:37 PM


188  Local excision and transanal endoscopic microsurgery

a higher likelihood of achieving clear resection mar- 13. Dafnis G, Pahlman L, Raab Y, et al. Transanal endo-
gins, lower recurrence rates and the ability to success- scopic microsurgery: clinical and functional results.
Colorectal Dis 2004; 6: 336–42.
fully excise more proximal tumours. Although the
14. Cataldo PA, O’Brien S, Osler T. Transanal endoscopic
number of studies comparing endoanal LE or TEM microsurgery: a prospective evaluation of functional
to radical resection with TME is small, the results are results. Dis Colon Rectum 2005; 48: 1366–71.
similar, in the sense that both show a higher recur- 15. Herman RM, Richter P, Walega P, Popiela T. Anorectal
rence rate, a trend towards decreased survival, and sphincter function and rectal barostat study in patients
lower morbidity with LE techniques compared with following transanal endoscopic microsurgery. Int J
radical resection. Significant disease progression can Colorectal Dis 2001; 16: 370–6.
16. Peeters KC, van de Velde CJ, Leer JW, et al. Late side
occur after LE despite intense surveillance, which
effects of short-course preoperative radiotherapy com-
may preclude curative salvage. The role of chemora- bined with total mesorectal excision for rectal cancer:
diation therapy and LE techniques in the treatment increased bowel dysfunction in irradiated patients:
of rectal cancer is still under study. a Dutch colorectal cancer group study. J Clin Oncol
2005; 23: 6199–206.
17. Grinnell RS. Results in the treatment of carcinoma of
References the colon and rectum: an analysis of 2,341 cases over
a 35 year period with 5 year survival results in 1,667
  1. Kraske P. Zur exstirpation hoch sitzender mastdarm- patients. Surg Gynecol Obstet 1953; 96: 31–42.
krebske. Verh Dtsch Ges Chir 1885; 14: 464–74. 18. Morson BC, Bussey HJ, Samoorian S. Policy of local
  2. Mason AY. Surgical access to the rectum: a trans- excision for early cancer of the colorectum. Gut 1977;
sphincteric exposure. Proc R Soc Med 1970; 18: 1045–50.
63(suppl.): 91–4. 19. Heald RJ, Ryall RD. Recurrence and survival after total
  3. Lockhart-Mummery J. Two hundred cases of cancer mesorectal excision for rectal cancer. Lancet 1986; 1:
of the rectum treated by perineal excision. Br J Surg 1479–82.
1926; 14: 110–24. 20. Garcia-Aguilar J, Mellgren A, Sirivongs P, et al. Local
  4. Miles WE. A method of performing abdomino-perineal excision of rectal cancer without adjuvant therapy: a
excision for carcinoma of the rectum and of the terminal word of caution. Ann Surg 2000; 231: 345–51.
portion of the pelvic colon. Lancet 1908; 2: 1812–3. 21. Mellgren A, Sirivongs P, Rothenberger DA, et al. Is
  5. Moore JS, Cataldo PA, Osler T, Hyman NH. Transanal local excision adequate therapy for early rectal can-
endoscopic microsurgery is more effective than tradi- cer? Dis Colon Rectum 2000; 43: 1064–71.
tional transanal excision for resection of rectal masses. 22. Paty PB, Nash GM, Baron P, et al. Long-term results
Dis Colon Rectum 2008; 51: 1026–30, 1030–1. of local excision for rectal cancer. Ann Surg 2002; 236:
  6. Buess G, Hutterer F, Theiss J, et al. A system for a 522–30.
transanal endoscopic rectum operation. Chirurg 1984; 23. Madbouly KM, Remzi FH, Erkek BA, et al. Recur-
55: 677–80. rence after transanal excision of T1 rectal cancer:
  7. Zerz A, Muller-Stich BP, Beck J, et al. Endoscopic should we be concerned? Dis Colon Rectum 2005;
posterior mesorectal resection after transanal local 48: 711–9.
excision of T1 carcinomas of the lower third of the 24. Friel CM, Cromwell JW, Marra C, et al. Salvage radical
rectum. Dis Colon Rectum 2006; 49: 919–24. surgery after failed local excision for early rectal can-
  8. Atallah S, Albert M, Larach S. Transanal minimally cer. Dis Colon Rectum 2002; 45: 875–9.
invasive surgery: a giant leap forward. Surg Endosc 25. Blumberg D, Paty PB, Picon AI, et al. Stage I rectal
2010; 24: 2200–05. cancer: identification of high risk patients. J Am Coll
  9. Parks AG. A technique for excising extensive villous Surg 1998; 186: 574–9.
papillomatous change in the lower rectum. Proc R Soc 26. You YN, Baxter NN, Stewart A, et al. Is the increas-
Med 1968; 61: 441–2. ing rate of local excision for stage I rectal cancer in the
10. Strong SA, Lavery IC, Oakley JR, Fazio VW, Church United States justified? A nationwide cohort study from
JM. Transanal excision of rectal tumors: is the case the National Cancer Database. Ann Surg 2007; 245:
closed? Dis Colon Rectum 1992; 35: P25. 726–33.
11. Sengupta S, Tjandra JJ. Local excision of rectal can- 27. Kim CJ, Yeatman TJ, Coppola D, et al. Local excision
cer: what is the evidence? Dis Colon Rectum 2001; of T2 and T3 rectal cancers after downstaging chemo-
44: 1345–61. radiation. Ann Surg 2001; 234: 352–8.
12. Kreissler-Haag D, Schuld J, Lindemann W, et al. Com- 28. Lezoche E, Guerrieri M, Paganini AM, et al. Long-term
plications after transanal endoscopic microsurgical results in patients with T2–3 N0 distal rectal cancer
resection correlate with location of rectal neoplasms. undergoing radiotherapy before transanal endoscopic
Surg Endosc 2008; 22: 612–6. microsurgery. Br J Surg 2005; 92: 1546–52.

HEBK001-C11_p171-190.indd 188 08/02/13 5:37 PM


REFERENCES  189

29. Lezoche E, Guerrieri M, Paganini AM, et al. Transanal 43. De Graaf EJ, Doornebosch PG, Tetteroo GW, et al.
endoscopic versus total mesorectal laparoscopic Transanal endoscopic microsurgery is feasible for
resections of T2-N0 low rectal cancers after neoad- adenomas throughout the entire rectum: a prospective
juvant treatment: a prospective randomized trial with study. Dis Colon Rectum 2009; 52: 1107–13.
a 3-years minimum follow-up period. Surg Endosc 44. Tarantino I, Hetzer FH, Warschkow R, et al. Local exci-
2005; 19: 751–6. sion and endoscopic posterior mesorectal resection
30. Borschitz T, Gockel I, Kiesslich R, Junginger T. Onco- versus low anterior resection in T1 rectal cancer. Br J
logical outcome after local excision of rectal carcino- Surg 2008; 95: 375–80.
mas. Ann Surg Oncol 2008; 15: 3101–8. 45. Canessa CE, Badia F, Fierro S, et al. Anatomic study
31. Borschitz T, Wachtlin D, Mohler M, et al. Neoadjuvant of the lymph nodes of the mesorectum. Dis Colon
chemoradiation and local excision for T2–3 rectal Rectum 2001; 44: 1333–6.
cancer. Ann Surg Oncol 2008; 15: 712–20. 46. Russell AH, Harris J, Rosenberg PJ, et al. Anal sphinc-
32. Winde G, Nottberg H, Keller R, et al. Surgical cure ter conservation for patients with adenocarcinoma of
for early rectal carcinomas (T1): transanal endoscopic the distal rectum: long-term results of radiation therapy
microsurgery vs. anterior resection. Dis Colon Rectum oncology group protocol 89-02. Int J Radiat Oncol Biol
1996; 39: 969–76. Phys 2000; 46: 313–22.
33. Lezoche G, Baldarelli M, Guerrieri M, et al. A prospec- 47. Mohiuddin M, Marks G, Bannon J. High-dose preop-
tive randomized study with a 5-year minimum follow- erative radiation and full thickness local excision: a
up evaluation of transanal endoscopic microsurgery new option for selected T3 distal rectal cancers. Int J
versus laparoscopic total mesorectal excision after Radiat Oncol Biol Phys 1994; 30: 845–9.
neoadjuvant therapy. Surg Endosc 2008; 22: 352–8. 48. Bonnen M, Crane C, Vauthey JN, et al. Long-term
34. Hodgson DC, Zhang W, Zaslavsky AM, et al. Relation results using local excision after preoperative
of hospital volume to colostomy rates and survival for chemoradiation among selected T3 rectal cancer
patients with rectal cancer. J Natl Cancer Inst 2003; patients. Int J Radiat Oncol Biol Phys 2004; 60:
95: 708–16. 1098–105.
35. Greenberg JA, Shibata D, Herndon JE 2nd, et al. 49. Callender GG, Das P, Rodriguez-Bigas MA, et al. Local
Local excision of distal rectal cancer: an update of excision after preoperative chemoradiation results in
cancer and leukemia group B 8984. Dis Colon Rectum an equivalent outcome to total mesorectal excision
2008; 51: 1185–91, 1191–4. in selected patients with T3 rectal cancer. Ann Surg
36. Bach SP, Hill J, Monson JR, et al. A predictive model Oncol 2010; 17: 441–7.
for local recurrence after transanal endoscopic micro- 50. Yeo SG, Kim DY, Kim TH, et al. Local excision fol-
surgery for rectal cancer. Br J Surg 2009; 96: 280–90. lowing pre-operative chemoradiotherapy-induced
37. Tsai BM, Finne CO, Nordenstam JF, et al. Transanal downstaging for selected cT3 distal rectal cancer. Jpn
endoscopic microsurgery resection of rectal tumors: J Clin Oncol 2010; 40: 754–60.
outcomes and recommendations. Dis Colon Rectum 51. Garcia-Aguilar J, Qian S, Thomas CR Jr., et al.
2010; 53: 16–23. Pathologic complete response (pCR) to neoadjuvant
38. Ganai S, Kanumuri P, Rao RS, Alexander AI. Local chemoradiation (CRT) of uT2uN0 rectal cancer (RC)
recurrence after transanal endoscopic microsurgery for treated by local excision (LE): results of the ACOSOG
rectal polyps and early cancers. Ann Surg Oncol 2006; Z6041 trial. J Clin Oncol 2010; 28(suppl.): 15s.
13: 547–56. 52. Nair RM, Siegel EM, Chen DT, et al. Long-term results
39. Stipa F, Burza A, Lucandri G, et al. Outcomes for early of transanal excision after neoadjuvant chemoradiation
rectal cancer managed with transanal endoscopic for T2 and T3 adenocarcinomas of the rectum.
microsurgery: a 5-year follow-up study. Surg Endosc J Gastrointest Surg 2008; 12: 1797–806.
2006; 20: 541–5. 53. Kitajima K, Fujimori T, Fujii S, et al. Correlations
40. Christoforidis D, Cho HM, Dixon MR, et al. Transanal between lymph node metastasis and depth of submu-
endoscopic microsurgery versus conventional cosal invasion in submucosal invasive colorectal carci-
transanal excision for patients with early rectal cancer. noma: a Japanese collaborative study. J Gastroenterol
Ann Surg 2009; 249: 776–82. 2004; 39: 534–43.
41. Lee W, Lee D, Choi S, et al. Transanal endoscopic 54. Leong AF, Seow-Choen F, Tang CL. Diminutive can-
microsurgery and radical surgery for T1 and T2 rectal cers of the colon and rectum: comparison between
cancer. Surg Endosc 2003; 17: 1283–7. flat and polypoid cancers. Int J Colorectal Dis 1998;
42. Langer C, Liersch T, Suss M, et al. Surgical cure 13: 151–3.
for early rectal carcinoma and large adenoma: 55. Chambers WM, Khan U, Gagliano A, et al. Tumour
transanal endoscopic microsurgery (using ultra- morphology as a predictor of outcome after local exci-
sound or electrosurgery) compared to conventional sion of rectal cancer. Br J Surg 2004; 91: 457–9.
local and radical resection. Int J Colorectal Dis 56. Hermanek P, Gall FP. Significance of local control of
2003; 18: 222–9. colorectal cancer. Fortschr Med 1985; 103: 1041–6.

HEBK001-C11_p171-190.indd 189 08/02/13 5:37 PM


190  Local excision and transanal endoscopic microsurgery

57. Chakravarti A, Compton CC, Shellito PC, et al. Long- 66. Park YA, Sohn SK, Seong J, et al. Serum CEA as a
term follow-up of patients with rectal cancer managed predictor for the response to preoperative chemo-
by local excision with and without adjuvant irradiation. radiation in rectal cancer. J Surg Oncol 2006; 93:
Ann Surg 1999; 230: 49–54. 145–50.
58. Hase K, Shatney C, Johnson D, et al. Prognostic value 67. Muthusamy VR, Chang KJ. Optimal methods for stag-
of tumor ‘budding’ in patients with colorectal cancer. ing rectal cancer. Clin Cancer Res 2007; 13: 6877–84s.
Dis Colon Rectum 1993; 36: 627–35. 68. Bipat S, Glas AS, Slors FJ, et al. Rectal cancer: local
59. Wang HS, Liang WY, Lin TC, et al. Curative resection staging and assessment of lymph node involvement with
of T1 colorectal carcinoma: risk of lymph node metas- endoluminal US, CT, and MR imaging – a metaanalysis.
tasis and long-term prognosis. Dis Colon Rectum Radiology 2004; 232: 773–83.
2005; 48: 1182–92. 69. Kwok H, Bissett IP, Hill GL. Preoperative staging of
60. Gall FP, Hermanek P. Cancer of the rectum: local exci- rectal cancer. Int J Colorectal Dis 2000; 15: 9–20.
sion. Surg Clin North Am 1988; 68: 1353–65. 70. Santoro GA, D’Elia A, Battistella G, et al. The use of
61. Kudo S. Endoscopic mucosal resection of flat and a dedicated rectosigmoidoscope for ultrasound stag-
depressed types of early colorectal cancer. Endoscopy ing of tumours of the upper and middle third of the
1993; 25: 455–61. rectum. Colorectal Dis 2007; 9: 61–6.
62. Ishizaki Y, Takeda Y, Miyahara T, et al. Evaluation of 71. Rothenberger D, Ricciardi R, Madoff RM. Procedures
local excision for sessile–type lower rectal tumors. for rectal cancer. In Ashley SW (ed.). ACS Surgery:
Hepatogastroenterology 1999; 46: 2329–32. Principles and Practice. Philadelphia, PA, BC Decker,
63. Kikuchi R, Takano M, Takagi K, et al. Management of 2010: 933–48.
early invasive colorectal cancer: risk of recurrence and 72. Lee WY, Lee WS, Yun SH, et al. Decision for salvage
clinical guidelines. Dis Colon Rectum 1995; 38: 1286–95. treatment after transanal endoscopic microsurgery.
64. Kobayashi H, Mochizuki H, Kato T, et al. Is total Surg Endosc 2007; 21: 975–9.
mesorectal excision always necessary for T1–T2 lower 73. Borschitz T, Heintz A, Junginger T. The influence of
rectal cancer? Ann Surg Oncol 2010; 17: 973–80. histopathologic criteria on the long-term prognosis of
65. Perez RO, São Julião GP, Habr-Gama A, et al. The role locally excised pT1 rectal carcinomas: results of local
of carcinoembryogenic antigen in predicting response excision (transanal endoscopic microsurgery) and
and survival to neoadjuvant chemoradiotherapy for dis- immediate reoperation. Dis Colon Rectum 2006; 49:
tal rectal cancer. Dis Colon Rectum 2009; 52: 1137–43. 1492–506, 1500–05.

HEBK001-C11_p171-190.indd 190 08/02/13 5:37 PM


12
Pathology assessment
Philip Quirke, Tim Palmer, Gordon G.A. Hutchins and Nick P. West

Introduction via electronic proformas, to cancer registries so that


the work of individuals, teams and institutions can
Pathologists play a key role in the multidisciplinary be compared nationally and internationally and so
team management of rectal cancer. The patho- that excellent and potentially suboptimal practice
logical analysis of the excised specimen provides can be identified.
important prognostic information on the stage of
the tumour, the accuracy of radiology and the qual-
ity assurance of the surgery. Quality assurance ema- Applied anatomy
nates from reporting the completeness of tumour
excision and the precise planes of excision, as rep- The rectum is situated with the confines of the
resented by the macroscopic appearance of the bony pelvis and adjacent to important structures.
specimen. The pathologist can assess the response The pelvic autonomic nerves run very close to the
to preoperative therapy and help to determine the perimesorectal resection planes, the ureters are
need for postoperative or adjuvant therapy, either located laterally, and the prostate in male patients
radiotherapy or chemotherapy. The pathologist can and the posterior vaginal wall in female patients
also identify patients at high risk of metachronous are in close proximity to the anterior surface of the
disease and screen for possible hereditary non- rectum. The sigmoid and upper and middle rec-
polyposis colorectal cancer using immunohisto- tum are encased in peritoneum, which decreases
chemistry for mismatch repair gene expression. in extent distally, but anteriorly at the peritoneal
Pathology may also help to educate radiologists and reflection the surgeon must incise the peritoneum
assure the quality of their reporting. More recently, and create the anterior surgical plane that is so
pathologists have undertaken further molecular critical to a successful surgical operation. Posteri-
testing to help predict the type of therapy that may orly the surgeon enters the perimesorectal fascial
be effective; for example, Ki-ras mutations may be plane, following either the visceral or the parietal
used to predict response to anti-epidermal growth plane. This can be distinguished by the thickness
factor receptor (anti-EGFr) antibodies. The cur- of the fascia applied to the mesorectum. The fas-
rent era is moving into whole-genome sequencing, cia blends with the peritoneum laterally above the
which will have far-reaching implications for the peritoneal reflection, enclosing the surgical ‘pack-
diagnosis, prognosis and treatment of colorectal age’, which needs to be removed intact. Posteriorly,
cancer. A further crucial responsibility is submis- deep within the mesorectum, close to the mesorec-
sion of high-quality pathology reports, preferably tal fascial surgical circumferential resection margin

HEBK001-C12_p191-202.indd 191 08/02/13 5:36 PM


192  Pathology assessment

(CRM), is the ­superior rectal artery and its division lysed, all patients were followed up for 2 years and
into its left and right branches. This artery is impor- the outcomes were published in 1986.1 Just before
tant because lymphatic drainage is associated with this, Chan and colleagues from Hong Kong pub-
the arterial blood supply and lymph nodes can be lished 50 cases showing tumour involvement of the
found clustered near to the artery and adjacent to same margin but without clinical follow-up.2 We
the mesorectal fascia. were able to demonstrate the importance of this
The mesorectum narrows superiorly in its cranial margin as a cause of local recurrence (although
portion, bulges at the level of the mid-rectum and ‘progressive persistent disease’ might be a more
then narrows to taper out at the puborectal sling. accurate term). We reported the high frequency of
The levators are applied to the lowest part of the local recurrence and the importance of a margin
mesorectum and can be taken with the mesorectum greater than 1 mm rather than 0 mm, as had previ-
when excising low rectal cancers or large cancers ously been the gold standard. From this emanated
involving the anal canal. The anatomy of the anal the concept of an involved margin where tumour
sphincters varies between individuals, with variable was found within 1 mm of the surgical resection
bulk of the muscle layers and differing anal canal margin. This initial 1986 publication was followed
lengths. It is a highly complex evacuation control by reports on two larger series3,4 and by others
mechanism and deserves further anatomical study. summarized in a review article.5
The intersphincteric plane, an important potential The principles of CRM involvement have been
surgical plane between the internal and external documented in numerous studies, including many
sphincters, can be used to achieve a very low recon- prospective randomized trials (Dutch Total Mes-
struction with partial or even complete removal of orectal Excision (TME) Trial, Medical Research
the internal sphincter, thus avoiding the need for Council (MRC) CR07 Trial, MRC Conventional
abdominoperineal excision (APE). versus Laparoscopic-Assisted Surgery in Colorec-
The anatomical structures of the anal sphincter tal Cancer (CLASICC) Trial) and have confirmed
are not well known by pathologists but are wor- the importance of a positive or negative CRM.
thy of detailed study to analyse and improve low More recently, a meta-analysis was reported,5 and
rectal cancer surgery and to accurately stage these even with a group of mesorectal trained surgeons it
tumours. Radiologists, surgeons and pathologists retains its importance.6,7
are increasingly skilled at understanding the ana- Involvement of the CRM occurs most frequently
tomical relationships of rectal cancer; optimal pre- anteriorly, where the mesorectum is at its thin-
operative images help in providing the anatomical nest. Additionally, the CRM is at high risk in the
road map for the surgeon to plan the appropriate low mesorectum from surgical coning at the pub-
planes for surgical intervention. The pathologist orectalis and sphincter muscles when the surgeon
can then determine how closely the surgeon fol- undertakes standard APE. Anteriorly there is the
lowed the intended plane. least tissue surrounding the muscle tube,8,9 and fol-
lowing the intended plane under excellent direct
vision is essential. For advanced posterior and lateral
The circumferential margin tumours, removing the levators, or if appropriate the
ischiorectal fat, can increase the potential clearance.
In 1982, while discussing the mismatch between Tumour involvement either at the margin or
the infrequently observed involvement of the dis- 1 mm or closer to the margin carries the highest
tal margin of resection and the high rate of local risks of local recurrence,5,10 and this risk decreases
recurrence in rectal cancer, we decided to look as the distance increases between the tumour and
at the ‘lateral’ surgical margins on the resected the resection margin. The mode of margin involve-
specimens, i.e. the margin created by the surgeon ment does not seem to matter and can be by direct,
around the rectum. It rapidly became apparent discontinuous, venous, lymphatic or neural spread.4
that this margin, initially referred to as the ‘lateral The presence of tumour within a lymph node with-
resection margin’ and subsequently termed the in 1 mm of the margin does not appear to confer
‘circumferential resection margin’, was frequently as high a risk in the two small series identified, but
involved in rectal cancer. Fifty-two cases were ana- the database on this is very small. These areas are

HEBK001-C12_p191-202.indd 192 08/02/13 5:36 PM


PLANES OF SURGERY  193

reviewed elsewhere,5 but the hypothesis is that if plane was within the mesorectum; or mesorectal,
the tumour is confined to lymphatic pathways, then where the planes were followed accurately. Coning
spread will be within this system and will thus be of the specimen near to the distal excision mar-
upwards towards higher nodes rather than towards gin was also frequent, bringing the surgical CRM
the CRM. This does, however, depend on the sur- closer or into the area of the cancer. This classifica-
geon removing the TME plane perfectly. tion into mesorectal, intramesorectal and muscu-
The importance of an involved CRM increases laris propria specimen descriptions was developed
after preoperative treatment, with an increase in from these observations.19,20 The classification was
the risks of local failure,5,11–14 and there is a strong assessed prospectively by local pathologists in the
association with an increased local recurrence but MRC CR07 trial21 and retrospectively by central
an equivalently poor survival. Achieving a complete review of photographs in the Dutch TME Trial.22
excision locally is essential in the curative man- Both studies demonstrated that the plane of sur-
agement of rectal cancer. Although preoperative gery was related to local recurrence; additionally,
treatment helps to achieve a clear margin in some the Dutch study showed the plane of surgery was
advanced cases, if the margin is involved after neoad- related to survival. Three further studies have con-
juvant therapy the outcome is poor. It has been pro- firmed the relationship between specimen quality
posed that consideration should be given to the strat- and local recurrence.23–25 Unsurprisingly, the plane
egy of trying alternative chemotherapy approaches achieved was related to CRM positivity rates,
to achieve down-staging before intervening with with the lowest positive CRM rates in mesorectal
definitive radiotherapy and subsequent surgery. The plane surgery. The specimen plane was not relat-
delivery of induction chemotherapy before chemo- ed directly to the stage of the tumour, with many
radiotherapy in the Expert C trial appears to provide suboptimal specimens in early-stage tumours. The
additional down-staging,15 and in patients resist- frequency of CRM involvement and the plane of
ant to the first therapy an alternative combination surgery have been documented in the Magnetic
may be tried before adding radiation. An alternative Resonance Imaging in Rectal Cancer European
approach is being studied by the Swedish trialists, Equivalence (MERCURY) study,7 with the lowest
where 6 weeks of chemotherapy is given following CRM positivity in mesorectal plane excisions and a
short-course radiotherapy. These approaches are still higher rate of mesorectal excisions than seen in the
experimental and being evaluated. MRC CR07 study. Surgeons showed improvement
in the frequency of mesorectal excisions over the
trial. There was an additive effect of radiotherapy
Planes of surgery on all grades of surgery; with this combination,
good surgery led to a 1 per cent local recurrence
Heald and colleagues described TME in 198216 and rate compared with 6 per cent in patients who
excellent outcomes in 1986.17 Subsequent audit received no irradiation.21
of these results confirmed the survival benefit of In many studies, the quality of surgery was much
this operation in highly skilled hands.18 The differ- poorer in APE specimens. There was no improve-
ences in specimen appearances and surgical planes ment in quality over the period of the trial in CR07
of standard anterior resection and Heald’s TME in APE, whereas there was an improvement in qual-
specimens became apparent when Quirke, Heald ity in anterior resection. Thus, achieving optimal
and others participated in a pathological dissection planes improves outcomes in patients with rectal
workshop in 1993 in Oslo as part of the Norwegian cancer undergoing anterior resection; focusing on
rectal cancer education courses. Total mesorectal this during surgery, and its subsequent pathologi-
excision achieves a smooth surgical margin by fol- cal assessment, has major benefits to patients. This
lowing the anatomical plane provided by the outer can be seen in the reduction in local recurrence
surface of the mesorectal fascia. The standard sur- rates and improving survival rates seen in rectal
gical planes achieved by the Norwegian surgeons cancer where such techniques have been adopted in
at that time were either on the muscularis propria, ­Norway,26 Stockholm,27 the Netherlands,28 Vancou-
where the surgical excision extended down on to ver29 and British Columbia;30 many other results are
the muscularis propria; intramesorectal, where the expected over the next few years.

HEBK001-C12_p191-202.indd 193 08/02/13 5:36 PM


194  Pathology assessment

Table 12.1. CRM involvement in anterior resections moted by Holm.37 The concept of this operation
compared with abdominoperineal excisions. involves removing the levators attached to the
Abdominoperineal Anterior lower mesorectum and the entire anal canal with
Study n excision (%) resection (%) internal and external sphincters and a greater or
Marr et al. 10
686 36.5 22.3 lesser degree of ischiorectal and ischioanal fat.
Taylor et al.6,7 282 33 13 This occurs under direct vision, with the patient
Nagtegaal et al. 22
1586 29 13
in the prone position for the perineal part of the
procedure. The supine position has also been
Guillou et al.31 400 21 10
promoted with an emphasis on excellent direct
M. Peters, 21 10
vision.38 This approach can provide the criti-
 personal
cal few extra millimetres of protection around a
communication,
­locally advanced low rectal tumour and prevent
2008
perforation of the anal canal.
Quirke et al.21 1350 17 8 Holm has categorized excision of the sphincters
Wibe et al.32 2136 12 5 and anal canal into three types of operation: the
standard APE, with its perilous waist, high margin
positivity and high perforation rates; the extraleva-
Abdominoperineal excision tor APE, where the levators are removed en bloc;
and the extended extralevator APE, with a unilat-
The outcome after APE for rectal cancer is generally eral or bilateral extension into the ischiorectal fossa,
inferior to that after anterior resection, with an 8–10 dependent on the local extension of the cancer.
per cent worse overall survival in many studies. The latter is more commonly needed for advanced
The main cause appears to be related to the high recurrent squamous cancer, although on occasions
CRM positivity (Table 12.1) and specimen perfora- it may be required for very advanced adenocarci-
tion (Table 12.2) rates in APE. noma. Any of these operations can include removal
This may be explained on the basis of the sur- of other local organs such as the posterior wall of
gical planes used when resecting these tumours. the vagina in female patients or variable amounts
This aspect of APE specimens was first identi- of the prostate in male patients.
fied in 200236 and subsequently verified in a joint A study compared the specimens from Holm’s
study of the APE specimens in the Dutch TME extralevator operation with the standard opera-
Trial.33 In the latter study, a third of cases showed tion APE specimens from our own institution in
the surgical plane to extend into the lumen, sub- Leeds and demonstrated a markedly reduced CRM
mucosa or internal sphincter. In the other two- positivity and perforation rate.8 An increase in the
thirds of cases, the typical ‘waist’ seen in most amount of tissue removed by extralevator surgery
standard APEs was noted. A solution to reduce compared with standard surgery was also docu-
margin involvement and specimen perforation mented. These studies were extended to collect ext-
was identified at the Karolinska Institute, Stock- ralevator specimens from colleagues in the UK and
holm, when the concept of what is now termed Europe and found identical results, with a drop in
an ‘extralevator APE’ was performed and pro- margin involvement and specimen perforation.9
On reviewing the literature, it also became appar-
ent that there were other units in Paris and Wroclaw
Table 12.2. Perforation rates in anterior resections that used this approach. The Hôpital St Antoine in
compared with abdominoperineal excisions. Paris reported excellent outcomes, with 10 per cent
Abdominoperineal Anterior local recurrence in all patients and 5 per cent in cur-
Study n excision (%) resection (%) ative excisions and a 5-year survival of 76 per cent
Nagtegaal et al.33 1586 13.7 2.5 after APE for rectal cancer.39 Discussions with the
Wibe et al. 32
2136 16 4
authors and photographic proof from Emmanuel
Tiret and Roland Parc confirmed that they adopted
Eriksen et al.34 2873 15.4 3.6
an extralevator approach. Similarly, Bebenek has
Stockholm audit35 613 12 4
reported what he terms ‘abdomino-sacral APE’

HEBK001-C12_p191-202.indd 194 08/02/13 5:36 PM


REPORTING RECTAL CANCER  195

with excellent results of 8 per cent CRM positivity, The accuracy of MRI with regard to nodal
4 per cent perforation, 4.4 per cent local recurrence deposits is debatable, as larger nodal size does not
and 5-year survival of 68 per cent in more advanced always equate with tumour involvement and many
stage low rectal cancers.40 nodal deposits are in small lymph nodes. Indeed,
Thus, there are now substantial data on the large nodal size can equate with a better immune
problems of the ‘standard’ APE and accumulat- response and thus a better outcome. The texture
ing evidence of the superiority of the extraleva- of nodes and their irregularity may give a better
tor approach to APE.9 Ongoing issues pertain to indication of involvement, but it is still suboptimal.
patient selection, the optimum approach, whether The MRI images provide a surgical road map for
prone or supine,38,41 whether open, laparoscopic42,43 the operation, but it is important to note that inac-
or robotic,44 and the best methods of closing the curate calling of MRI images can have a profound
pelvic floor to optimize wound healing and reduce impact on patient treatment, with either over- or
perineal hernias.45,46 While awaiting the outcomes under treatment as a consequence.
of these issues, however, the potentially avoidable
local recurrences and impaired survival from stand-
ard APE should not be discounted. The National Preparation of the Specimen
Health Service in England has commissioned a
pilot programme on training and education in this The following preparation and dissection notes
area for English multidisciplinary teams (www. were prepared for the National Cancer Research
lorec.nhs.uk), and there are similar initiatives in Institute (NCRI) Aristotle trial (see www.control-
Denmark and other health-care systems. led-trials.com/ISRCTN09351447) and are very
similar to those used within the Low Rectal Can-
cer and MERCURY studies. They should be used
Reporting rectal cancer in conjunction with the Royal College of Patholo-
gists’ dissection guidelines and the reporting pro-
Accurate reporting of rectal cancer specimens is formas (see www.rcpath.org/Resources/RCPath/
crucial because the details may have a major impact Migrated%20Resources/Documents/G/G049-
on the multidisciplinary management of what is a ColorectalDataset-Sep07.pdf and www.rcpath.org/
complex and common cancer. It is therefore impor- Resources/RCPath/Migrated%20Resources/
tant to ensure that adequate time and resources are Documents/G/G049ColorectalDatasetAppendixC-
available to undertake this task. Pathologists need Sep07.doc). Whether the specimen is received fresh
to feel like an important component of the team, or fixed, the handling should be equivalent. Before
and to have seen good-quality TME and extral- opening the specimen, the anterior and posterior
evator APE specimens so that they understand the surfaces should be photographed to document
anatomy and potential surgical planes that they the quality of removal by assessing the anatomical
need to evaluate and categorize. Ideally, patholo- planes that have been achieved. Close-up images
gists should understand the magnetic resonance of the front and back of the levator/anal sphincter
imaging (MRI) images so that preoperative scans facilitate assessment of extralevator APE specimens.
can guide specimen dissections and provide accu- Each photograph should include a centimetre scale
rate feedback to radiologists, surgeons and others placed beside the specimen. The pathologist should
in the multidisciplinary team. Increasingly, radiolo- grade the quality of the surgery for both the mes-
gists can accurately describe the site of the tumour, orectum and the levator/anal sphincter area. It is
and report the distance between the margin of the advantageous to paint the surgically created mar-
tumour or tumour deposit and the CRM, whether gin as early as possible to allow the dye to adhere to
this is the mesorectal fascia, levators, anal sphinc- the specimen as securely as possible so it does not
ters, ischiorectal fat or other structures. The dis- spread widely on dissection. The adherence of the
tance from the muscularis propria to the edge of dye increases with the length of fixation.
the tumour, and large vessel extramural vascular The specimen can then be opened from the prox-
invasion, may also be recordable; on occasion, peri- imal margin distally to 2–5 cm above the tumour.
toneal involvement may be documented. The distal end should be kept intact. If the specimen

HEBK001-C12_p191-202.indd 195 08/02/13 5:36 PM


196  Pathology assessment

is received unfixed, and fresh material is required The macroscopic description should be complet-
for storage or research, it should be taken at this ed, specifically noting the presence of a perforation
stage. The tumour can be everted through the upper of the tumour or mucosa and the site of the perfora-
anterior specimen incision and luminal tumour tion. It should be stated whether the tumour perfora-
removed, or a jumbo biopsy forceps can be used to tion is present in an area covered by peritoneum or
obtain tumour material. A piece of foam or tissue a surgical margin, and whether it is above or at the
paper soaked in formalin can be inserted through level of the sphincters. The presence or absence and
the tumour if appropriate to improve fixation; if completeness of the attachment of levator ani to the
received fixed, the specimen can be inflated with mesorectum on the specimen should be described.
formalin and sealed at both ends. The specimen can The descriptions of grading are given below.
then be placed in formalin. The specimen should be sliced as thinly as pos-
Before dissection it is acceptable to inflate the sible, starting from the distal margin to 2–5 cm
specimen with formalin, fix and then take photo- above the tumour. These slices should be laid out
graphs. The anterior surface of the tumour should in good light, starting with the most distal slice at
never be opened as it destroys the assessment of the the top left-hand corner, and the most proximal
anterior CRM, the area most frequently involved slice ending up as the last slice. The face presented
by cancer by direct invasion, peritoneal involve- to the camera should be consistent in all the slices.
ment or perforation. The luminal size or area of the The slices should be photographed including a cen-
tumour does not convey prognostic information. timetre scale.
The anterior high rectum is covered by peritoneum; The minimum distance of the tumour to the
this area, especially the paracolic gutters, should CRM should be described, as should the maximum
be inspected carefully for peritoneal involvement. depth of invasion through the muscularis propria
Although peritoneal involvement is an adverse and the structures invaded by the tumour.
prognostic indicator, peritoneal involvement does If the CRM is free of tumour, it should be not-
not constitute an involved CRM, as this is not a sur- ed whether there is normal tissue at the margin
gically created margin. or whether it is fibrotic tissue following tumour
regression potentially indicating a previously
involved margin.
Dissection If the CRM is involved (confirmed on histology),
then the mode of involvement and the distance of
Consideration should be given to enhancing the involvement at that site should be stated (e.g. small
lymph node yield by the use of methylene or pat- area measuring 2 mm or more extensive involve-
ent blue injection47 into the artery at the time the ment measuring 10 mm).
specimen is removed by the surgeon, or by use of It is preferable to sample the main tumour by
glacial acetic acid, ethanol, distilled water and for- embedding each tumour-bearing slice and cutting
maldehyde (GEWF) fixation48 or post-dissection one to four large mount sections, although this is
using xylene clearance.49,50 This can be especially not possible in many laboratories; where it is not
useful for post-treatment resections where nodal possible, it is important to ensure that a minimum
size is reduced.51,52 of five blocks of tumour are taken, including the
Anterior and posterior non-peritonealized sur- CRM where the tumour is close and any other
faces are painted with ink. It should be remembered important macroscopic pathology (see below).
that the CRM applies only to the surgically incised As many lymph nodes as possible should be
mesorectal planes and not the peritonealized sur- dissected from the specimen. Lymph nodes after
faces. The mesorectal surface is larger posteriorly preoperative therapy are generally smaller and fre-
and extends up to a higher level than it does ante- quently around 1 mm in size; this should be borne
riorly. After the resection surfaces have been inked, in mind, as they will be more difficult to locate. The
the specimen is fixed in formalin for a minimum of use of methylene blue injection or GEWF fixation
48 h, but longer if possible. The firmer the mesorec- will increase the number of nodes found.
tum after fixation, the thinner the slices can be and Involvement of the peritoneum is defined as per
the more thoroughly the tumour can be examined. Shepherd and colleagues’ recommendations.53,54

HEBK001-C12_p191-202.indd 196 08/02/13 5:36 PM


REPORTING RECTAL CANCER  197

Extramural vascular invasion (EMVI) is defined the grade for the mesorectum. For APE, there will be a
by involvement of a thick vascular structure with grade for the mesorectum and another grade for the
a smooth muscle wall that will contain elastin on levator and anal canal area below the mesorectum.
elastic staining. This should be looked for closely.
If an isolated tumour deposit is present close to an
arterial structure, without an accompanying vein, Quality of Resection of
consider vascular invasion. Increased reporting fre- the Mesorectum
quencies of EMVI have been found following the
addition of elastic staining to haematoxylin and The quality of a mesorectal resection can be easily
eosin.55,56 assessed macroscopically and graded as shown in
Table 12.3.
This classification has been used in several series
T Staging of Low Rectal Cancers and two trials. The plane of surgery correlates with
frequency of CRM positivity, with the lowest CRM
The T staging of cancers above the sphincters is positivity rates in mesorectal plane excision speci-
straightforward and is the same as that for colon can- mens and the highest rates in muscularis propria
cers. As some mid-rectal and all low rectal adenocar- excision specimens.
cinomas have a proportion of the lesion within the
region of the sphincters, however, T staging is prob-
lematic. Currently, T staging of adenocarcinoma in Quality of Resection in
the area of the sphincters is unsound. TNM 7 states
that such tumours should be staged as anal cancers
Abdominoperineal Surgery
by tumour size. In the absence of a robust staging
The quality of surgery of the levator/anal canal area
system, the only solution is to describe the maximal
below the mesorectum can be assessed as shown in
anatomical extent of spread, both above the sphinc-
Table 12.4.
ter and at the level of the sphincter separately, to
allow subsequent analysis. We propose that the maxi-
mum level of invasion above and at the level of the Table 12.3. Summary of mesorectal grading.
sphincter should be recorded separately by extent of
Fascial plane Description
maximal spread. This should be captured by describ-
Mesorectal The mesorectum is smooth, with no violation
ing the extent of spread (e.g. submucosal, internal
 of the fat and good bulk to the mesorectum
sphincter, intersphincteric space, external sphinc-
anteriorly and posteriorly. The distal margin
ter) and also measuring the extent of spread from
appears adequate, with no coning near the
the muscularis propria and, where applicable, from
tumour. No defect is more than superficial
the edge of the internal sphincter when a tumour
or 5 mm deep.
involves the sphincters. We need to know whether
Intramesorectal There is moderate bulk to mesorectum but
one or both sphincters are involved by tumour. This
 irregularity of the mesorectal surface. There
is relatively easy to do, as the internal sphincter is an
is moderate coning of the specimen towards
easily identified reference point, being composed of
the distal margin. At no site is the muscularis
pearly-white smooth muscle. Involvement of striated
propria visible, with the exception of the
muscle indicates involvement of the levators, pub-
area of insertion of levator muscles. There is
orectalis or external sphincter.
moderate irregularity of the CRM.
Muscularis There are areas of substantial loss of
Assessment of Quality   propria  mesorectal tissue. Deep cuts and tears

of Surgery: Grading down on to the muscularis propria are


present. On cross-section there is a
very irregular CRM, with little bulk to the
The mesorectum and the anal canal/levator parts of
mesorectal fat; the muscularis propria forms
the specimen should be graded separately. For an ante-
the CRM in places.
rior resection specimen there will only be one grade –

HEBK001-C12_p191-202.indd 197 08/02/13 5:36 PM


198  Pathology assessment

Table 12.4. Summary of abdominoperineal excision grading. l entirely below the level of the peritoneal reflec-
Plane Description tion anteriorly.
Levator The surgical plane lies external to the levators,
Relationship to circumferential resection margin
 with the levators being removed en bloc
with the specimen. This creates a cylindrical Anteriorly the upper rectum is covered by perito-
specimen with the levators forming an extra neum. Only the area below the peritoneal reflection
protective layer on the sphincters. is at risk of surgical CRM involvement. Posteriorly
Sphincteric Either there are no levator muscles attached this area and the area above it, a triangular bare area
 to the specimen or there is only a very small running up to the start of the sigmoid mesocolon,
cuff and the resection margin on the surface is at risk not only from direct tumour spread but
of the sphincters. The specimen has a also from metastatic deposits in lymph nodes that
waisted/apple-core appearance. lie against the circumferential margin.
Intrasphincteric/ The surgeon has inadvertently entered the
It is recommended that the whole of this mar-
  submucosal  sphincters or even deeper into the submucosa
gin (i.e. the mesorectum) be painted with a marker
or perforated the specimen at any point.
such as India ink or Alcian blue followed by acetic
acid to fix the dye on the surface before dissecting
the specimen. The tumour is then best sliced seri-
For an anterior resection specimen, there will ally at 3- to 4-mm intervals to select blocks from the
be a single mesorectal grade. For an APE specimen area above and below the tumour to look for meta-
there will be two grades. static deposits. If lymph nodes lie against the cir-
cumferential margin, then these should be included
in the block.
Definitions Used in Pathology Relationship to extent of extramural
invasion of tumour
Position of tumour
When assessing the relationship to the CRM, on the
The position of the tumour should be noted accu-
whole-mount section the corresponding relation-
rately. Initially this involves documentation of the
ship between the outer surface of the muscle coat
surface involvement (i.e. anterior quadrant, pos-
and the maximum depth of extramural invasion
terior quadrant, lateral quadrant, combinations
needs to be measured. This is performed using the
of these). To correlate the position with the MRI
Vernier scale on the microscope or by overlaying a
report, however, the tumour position should be
scale on the microscope slide.
­reported from the distal resection margin with the
mesorectum posteriorly and the peritoneal reflec- Lymph nodes
tion anteriorly. This can be documented as a rela-
All lymph nodes found in the specimen should be
tionship to a clock face on the reporting proforma,
sampled and counted, regardless of their site and
with anterior at the 12 o’clock position (e.g. ‘maxi-
size. The number of positive lymph nodes must be
mal extent of tumour present between 10 o’clock
equal to or less than the number of lymph nodes
and 3 o’clock’).
sampled.
Relationship to peritoneal reflection Extramural tumour deposits measuring 3 mm or
larger are counted as involved lymph nodes, even if
A crucial landmark for recording the site of rectal
no residual lymph node structure can be identified,
cancers is the peritoneal reflection. This is identified
as defined in TNM 5. If lymph nodes are present
from the exterior surface of the anterior aspect of
but the structure cannot be discerned, the pres-
the specimen. Rectal cancers are classified accord-
ence of satellite nodules should be recorded as well.
ing to whether they are:
Smaller satellite deposits are regarded as apparent
l entirely above the level of the peritoneal reflec- discontinuous extensions of the main tumour, and
tion anteriorly; their presence should be recorded.
l astride or at the level of the peritoneal reflection In TNM 5, pN1 (where ‘p’ corresponds to ‘patho-
anteriorly; logical’) denotes involvement of one to three nodes

HEBK001-C12_p191-202.indd 198 08/02/13 5:36 PM


REPORTING RECTAL CANCER  199

and pN2 denotes involvement of four or more that lowering the ypT stage is of some value, but the
nodes. We do not recommend using TNM 6 or relative risk/benefit ratio is still uncertain for many
7 because of the many issues outlined in several patients. Some studies suggest that it improves out-
papers.57–59 come.14 The occurrence of a complete pathological
response (ypT0 ypN0) is very important, as patients
Distance to distal resection margin with this finding at pathology have excellent
This is measured from the nearest cut end of the cancer-related outcomes.58,59 If ypT0 and ypNx are
specimen to the tumour, not the circumferential included as complete pathological response, then
margin. It is necessary to examine the margins his- the frequency of complete response increases by
tologically only if the tumour extends macroscopi- 7 per cent. One major issue with complete response
cally to within 30 mm of the distal or circumfer- is the pathological dissection and sampling protocol
ential margin. For tumours further away from the followed. Lax pathological dissection and sampling
margin, it can be assumed that the cut ends are not lead to a higher frequency of complete response. For
involved. Exceptions to this recommendation are the Sanofi Capecitabine, Oxaliplatin, Radiotherapy
adenocarcinomas that are found on subsequent his- and Excision (CORE) Trial, an international panel
tology to have an exceptionally infiltrative growth recommended taking five blocks of tumour and, if
pattern, show extensive vascular or lymphatic per- no tumour was identified, to cut three further lev-
meation, or are undifferentiated carcinomas. els. If tumour is still absent, then the whole area of
abnormality should be embedded and, if necessary,
Relationship to dentate line a further three levels sectioned. If at this stage no
This can be measured only for low rectal tumours tumour is identified, then this should be considered
in APE specimens. a complete response. It is critical for the continued
use of complete pathological response that the sam-
Tumour perforation pling is standardized.
If the tumour has perforated into the peritoneal The reports of tumour regression grading stud-
cavity or through the mesorectal fascia or sphinc- ies have been disappointing. Rödel and colleagues
ter complex, then these cases should be recorded as tested regression grading in the German trial. They
a perforation. The TNM staging system states that failed to show a significant relationship to surviv-
only perforation of the peritoneum should be called al with five categories and then amalgamated the
pT4, but perforation/surgical rupture of tumour in groups and obtained significance.60 Gosens and
such areas confers a poor prognosis34 and many of colleagues13 and Rullier and colleagues14 did not
them would be reported as pT4 in routine practice find any significant benefits correlated to tumour
in the UK. regression grading. It is clear that a complete
response ypT0 ypN0 has an excellent prognosis
Tumour differentiation and that good responders, usually regressing to pT1
The differentiation of the tumour should be defined or early pT2 N0, also have excellent cancer related
on the dominant area of tumour as well differenti- outcomes.14 In our experience, it has become appar-
ated or moderately differentiated, equivalent to low ent that it is impossible to distinguish between no
grade and poorly differentiated or undifferenti- response and a poor response if looking at rand-
ated in the World Health Organization ‘blue book’,57 omized trial material blinded to trial arm.
which is equivalent to high-grade tumour. Other
types of differentiation (e.g. mucinous adenocarci-
nomas, signet ring, undifferentiated) and the pres- Optimal Reporting
ence of high-grade dysplasia should be documented.
As outlined above, optimal reporting of a rectal
Assessment after preoperative therapy cancer specimen requires extensive knowledge
The prefix ‘y’ indicates either a staging scan or patho- of the anatomy, radiology, pathology and treat-
logical assessment after preoperative (neoadjuvant) ment modalities. Excellent reporting is key for the
therapy. Such tumours should be staged with the best modern management of this disease. Pathol-
prefix ‘y’ before pTNM staging. There is no doubt ogy reporting goes beyond the crucial benefits to

HEBK001-C12_p191-202.indd 199 08/02/13 5:36 PM


200  Pathology assessment

the individual patient. The pathological findings 11. Mawdsley S, Glynne-Jones R, Grainger J, et al. Can
allow us to give feedback and audit to radiolo- histopathologic assessment of circumferential margin
after preoperative pelvic chemoradiotherapy for T3–T4
gists and surgeons and help oncologists to under-
rectal cancer predict for 3-year disease-free survival?
stand the effects of current and new therapies. This Int J Radiat Oncol Biol Phys 2005; 63: 745–52.
mechanism creates a virtuous loop of continuous 12. Luna-Pérez P, Bustos-Cholico E, Alvarado I, et al.
improvement. Optimal practice and continuous Prognostic significance of circumferential margin
feedback offers our best hope of improving the out- involvement in rectal adenocarcinoma treated with
comes in colorectal cancer, one of the most com- preoperative chemoradiotherapy and low anterior
mon curable gastrointestinal tract tumours. The resection. J Surg Oncol 2005; 90: 20–25.
13. Gosens MJ, Klaassen RA, Tan-Go I, et al. Circum-
combination of screening, optimal surgery and
ferential margin involvement is the crucial prognostic
appropriate preoperative and adjuvant therapy factor after multimodality treatment in patients with
should allow us to make inroads into the mortality locally advanced rectal carcinoma. Clin Cancer Res
associated with this condition. 2007; 13: 6617–23.
14. Rullier A, Laurent C, Capdepont M, Vendrely V, Bioulac-
Sage P, Rullier E. Impact of tumor response on survival
References after radiochemotherapy in locally advanced rectal
carcinoma. Am J Surg Pathol 2010; 34: 562–8.
  1. Quirke P, Durdey P, Dixon MF, et al. Local recurrence 15. Chua YJ, Barbachano Y, Cunningham D, et al. Neoad-
of rectal adenocarcinoma due to inadequate surgical juvant capecitabine and oxaliplatin before chemoradio-
resection: histopathological study of lateral tumour therapy and total mesorectal excision in MRI-defined
spread and surgical excision. Lancet 1986; 328: poor-risk rectal cancer: a phase 2 trial. Lancet Oncol
996–9. 2010; 11: 241–8.
  2. Chan KW, Boey J, Wong SK. A method of reporting 16. Heald RJ, Husband EM, Ryall RD. The mesorectum in
radial invasion and surgical clearance of rectal carci- rectal cancer surgery: the clue to pelvic recurrence?
noma. Histopathology 1985; 9: 1319–27. Br J Surg 1982; 69: 613–6.
  3. Adam IJ, Mohamdee MO, Martin IG, et al. Role of 17. Heald RJ, Ryall RD. Recurrence and survival after total
circumferential margin involvement in the local recur- mesorectal excision for rectal cancer. Lancet 1986;
rence of rectal cancer. Lancet 1994; 344: 707–11. 1: 1479–82.
  4. Birbeck KF, Macklin CP, Tiffin NJ, et al. Rates of 18. MacFarlane JK, Ryall RD, Heald RJ. Mesorectal exci-
circumferential resection margin involvement vary sion for rectal cancer. Lancet 1993; 341: 457–60.
between surgeons and predict outcomes in rectal 19. Quirke P, Dixon MF. The prediction of local recurrence
cancer surgery. Ann Surg 2002; 235: 449–57. in rectal adenocarcinoma by histopathological exami-
  5. Nagtegaal ID, Quirke P. What is the role for the cir- nation. Int J Colorectal Dis 1988; 3: 127–31.
cumferential margin in the modern treatment of rectal 20. Quirke P. Limitations of existing systems of staging
cancer? J Clin Oncol 2008; 26: 303–12. for rectal cancer: the forgotten margin. In Soreide O
  6. Taylor FGM, Quirke P, Heald RJ, et al. One millimetre Norstein J (eds). Rectal Cancer Surgery Optimization,
is the safe cut-off for magnetic resonance imaging Standardization, Documentation. Berlin, Springer-
prediction of surgical margin status in rectal cancer. Verlag, 1997: 63–81.
Br J Surg 2011; 98: 872–9. 21. Quirke P, Steele R, Monson J, et al. Effect of the plane
  7. Taylor FGM, Quirke P, Blomqvist L, et al. Preoperative of surgery achieved on local recurrence in patients
assessment of the circumferential resection margin by with operable rectal cancer: a prospective study using
MRI predicts survival outcomes and recurrence in rec- data from the MRC CR07 and NCIC–CTG CO16
tal cancer: 5 year follow up results of the MERCURY randomised clinical trial. Lancet 2009; 373: 821–8.
study [paper submitted]. Lancet Oncol. 22. Nagtegaal ID, van de Velde CJH, van der Worp E,
  8. West NP, Finan PJ, Anderin C, Lindholm J, Holm T, et al. Macroscopic evaluation of rectal cancer resec-
Quirke P. Evidence of the oncologic superiority of tion specimen: clinical significance of the pathologist in
­cylindrical abdominoperineal excision for low rectal quality control. J Clin Oncol 2002; 20: 1729–34.
cancer. J Clin Oncol 2008; 26: 3517–22. 23. Maslekar S, Sharma A, Macdonald A, Gunn J,
  9. West NP, Anderin C, Smith KJE, Holm T, Quirke P. Monson JR, Hartley JE. Mesorectal grades predict
Multicentre experience with extralevator abdominoper- recurrence after curative excision for rectal cancer. Dis
ineal excision for low rectal cancer. Br J Surg 2010; Colon Rectum 2006; 50: 168–75.
97: 588–99. 24. García-Granero E, Faiz O, Muñoz E, et al. Macro-
10. Marr R, Birbeck K, Garvican J, et al. The modern scopic assessment of mesorectal excision in rectal
abdominoperineal excision: the next challenge after cancer: a useful tool for improving quality control in a
total mesorectal excision. Ann Surg 2005; 242: 74–82. multidisciplinary team. Cancer 2009; 115: 3400–11.

HEBK001-C12_p191-202.indd 200 08/02/13 5:36 PM


REFERENCES  201

25. Leite JS, Martins SC, Oliveira J, Cunhna MF, Castro- abdominoperineal resection for adenocarcinoma of the
Sousa F. Clinical significance for macroscopic com- low rectum. Dis Colon Rectum 2003; 46: 867–4.
pleteness of excision in rectal cancer. Colorectal Dis 40. Bebenek M. Abdominosacral amputation of the
2011; 13: 381–6. rectum for low rectal cancers: ten years of experience.
26. Wibe A, Rendedal PR, Svensson E, et al. Prognostic Ann Surg Oncol 2009; 16: 2211–7.
significance of the circumferential resection margin 41. Holm T. Abdominoperineal resection revisited: is posi-
following total mesorectal excision for rectal cancer. tioning an important issue? Dis Colon Rectum 2011;
Br J Surg 2002; 89: 327–34. 54: 921–2.
27. Martling AL, Holm T, Rutqvist L-E, et al. Effect of a sur- 42. Singh B, Lloyd G, Nilsson PJ, Chaudri S. Laparo-
gical training programme on outcome of rectal cancer scopic extralevator abdominoperineal excision of the
in the County of Stockholm. Lancet 2000; 356: 93–6. rectum: the best of both worlds. Tech Coloproctol
28. Kapiteijn E, Putter H, van de Velde CJ, et al. Impact of 2012; 16: 73–5.
the introduction and training of total mesorectal exci- 43. Vaughan-Shaw PG, King AT, Cheung T, et al. Early
sion on recurrence and survival in rectal cancer in the experience with laparoscopic extralevator abdomi-
Netherlands. Br J Surg 2002; 89: 1142–9. noperineal excision within an enhanced recovery
29. Phang PT, McGahan CE, McGregor G, et al. Effects of setting: analysis of short-term outcomes and quality
change in rectal cancer management on outcomes in of life. Ann R Coll Surg Engl 2011; 93: 451–9.
British Columbia. Can J Surg 2010; 53: 225–31. 44. Marecik SJ, Zawadzki M, Desouza AL, Park JJ,
30. Phang PT, Woods R, Brown CJ, Raval M, Cheifetz R, Abcarian H, Prasad LM. Robotic cylindrical abdomi-
Kennecke H. Effect of systematic education courses noperineal resection with transabdominal levator
on rectal cancer treatments in a population. Am J Surg transection. Dis Colon Rectum 2011; 54: 1320–5.
2011; 201: 640–4. 45. Anderin C, Martling A, Lagergren J, Ljung A, Holm T.
31. Guillou PJ, Quirke P, Thorpe H, et al. Short-term end- Short-term outcome after gluteus maximus myocuta-
points of conventional versus laparoscopic-assisted neous flap reconstruction of the pelvic floor follow-
surgery in patients with colorectal cancer (MRC CLA- ing extra-levator abdominoperineal excision of the
SICC trial): multicentre, randomised controlled trial. rectum [published online ahead of print 8 Oct 2011].
Lancet 2005; 365: 1718–26. ­Colorectal Dis 2011.
32. Wibe A, Syse A, Andersen E, et al. Oncological 46. Horch RE, D’Hoore A, Holm T, Kneser U, Hohen-
outcomes after total mesorectal excision for cure for berger W, Arkudas A. Laparoscopic abdominoperineal
cancer of the lower rectum: anterior vs. abdominoperi- resection with open posterior cylindrical excision and
neal resection. Dis Colon Rectum 2004; 47:48–58. primary transpelvic VRAM flap. Ann Surg Oncol 2012;
33. Nagtegaal ID, van de Velde CJ, Marijnen CA, et al. 19: 502–3.
Low rectal cancer: a call for a change of approach in 47. Martijnse IS, Dudink RL, Kusters M, Rutten HJ,
abdominoperineal resection. J Clin Oncol 2005; 23: ­Nieuwenhuijzen GA, Wasowicz-Kemps DK. Patent
9257–64. blue staining as a method to improve lymph node
34. Eriksen MT, Wibe A, Syse A, et al. Inadvertent perfora- detection in rectal cancer following neoadjuvant treat-
tion during rectal cancer resection in Norway. Br J ment. Eur J Surg Oncol 2012; 38: 252–8.
Surg 2004; 91: 210–6. 48. Iversen LH, Laurberg S, Hageman-Madsen R, Dyb-
35. Anderin C, Martling A, Hellborg H, Holm T. A popula- dahl H. Increased lymph node harvest from colorectal
tion-based study on outcome in relation to the type cancer resections using GEWF solution: a randomised
of resection in low rectal cancer. Dis Colon Rectum study. J Clin Pathol 2008; 61: 1203–8.
2010; 53: 753–60. 49. Jass JR, Miller K, Northover JM. Far clearance method
36. Marr R, Birbeck K, Garvican J, et al. The modern versus manual dissection of lymph nodes in specimens
abdominoperineal excision: the next challenge after of rectal cancer. Int J Colorectal Dis 1986; 1: 155–6.
total mesorectal excision. Ann Surg 2005; 242: 50. Cawthorn SJ, Gibbs NM, Marks CG. Clearance tech-
74–82. nique for the detection of lymph nodes in colorectal
37. Holm T, Ljung A, Häggmark T, Jurell G, Lagergren J. cancer. Br J Surg 1986; 73: 58–60.
Extended abdominoperineal resection with gluteus 51. Sanchez W, Luna-Perez P, Alvarado I, Labastida S,
maximus flap reconstruction of the pelvic floor for Herrera L. Modified clearing technique to identify
rectal cancer. Br J Surg 2007; 94: 232–8. lymph node metastases in post-irradiated surgical
38. Martijnse IS, Dudink RL, West NP, et al. Focus on specimens from rectal adenocarcinoma. Arch Med
extralevator perineal dissection in supine position for Res. 1996; 27: 31–6.
low rectal cancer has led to better quality of surgery 52. Wang H, Safar B, Wexner SD, Denoya P, Berho M.
and oncologic outcome. Ann Surg Oncol 2012; 19: The clinical significance of fat clearance lymph node
786–93. harvest for invasive rectal adenocarcinoma following
39. Dehni N, McFadden N, McNamara DA, Guiguet neoadjuvant therapy. Dis Colon Rectum 2009; 52:
M, Tiret E, Parc R. Oncologic results following 1767–73.

HEBK001-C12_p191-202.indd 201 08/02/13 5:36 PM


202  Pathology assessment

53. Shepherd NA, Baxter KJ, Love SB. The ­prognostic Hruban RH (eds). WHO Classification of Tumours of
importance of peritoneal involvement in colonic the Digestive System. Lyon, World Health Organiza-
­cancer: a prospective evaluation. Gastroenterology tion, 2010: 131–93.
1997; 112: 1096–102. 58. Capirci C, Valentini V, Cionini L, et al. Prognostic value
54. Ludeman L, Shepherd NA. Serosal involvement in of pathologic complete response after neoadjuvant
gastrointestinal cancer: its assessment and signifi- therapy in locally advanced rectal cancer: long-term
cance. Histopathology 2005; 47: 123–31. analysis of 566 ypCR patients. Int J Radiat Oncol Biol
55. Vass DG, Ainsworth R, Anderson JH, et al. The Phys 2008; 72: 99–107.
value of an elastic tissue stain in detecting venous 59. Maas M, Nelemans PJ, Valentini V, et al. Long-term
invasion in colorectal cancer. J Clin Pathol 2004; outcome in patients with a pathological complete
57: 769–72. response after chemoradiation for rectal cancer: a
56. Howlett CJ, Tweedie EJ, Driman DK. Use of an elastic pooled analysis of individual patient data. Lancet
stain to show venous invasion in colorectal carcinoma: Oncol 2010; 11: 835–44.
a simple technique for detection of an important prog- 60. Rödel C, Martus P, Papadoupolos T, et al. Prognostic
nostic factor. J Clin Pathol 2009; 62: 1021–5. significance of tumor regression after preoperative
57. Hamilton SR, Bosman FT, Bofetta P, et al. Carcinoma chemoradiotherapy for rectal cancer. J Clin Oncol
of the colon and rectum. In Bosman FT, Carneiro F, 2005; 23: 8688–96.

HEBK001-C12_p191-202.indd 202 08/02/13 5:36 PM


13
Assessment and management
of recurrence
Peter J. Lee, Kirk K.S. Austin and Michael J. Solomon

Introduction quality of life, with a reduction in pain. In addition,


their quality of life is better than in patients who
Historically, surgery for locally recurrent rectal can- receive non-surgical palliative treatment.11–13
cer (LRRC) has not been a widely accepted modality Improved surgical techniques and adjuvant ther-
of treatment. It was believed to incur high morbidity apies for primary rectal cancer have reduced the
and mortality. Cancer-related deaths with LRRC are rates of local recurrence to less than 10 per cent.14
unpleasant, with intractable pain, offensive perineal Surgery for LRRC, or pelvic exenteration surgery,
discharge and rectal bleeding, all culminating in a has become more complex, requiring more radical
poor quality of life.1–3 The prognosis of such patients extended surgery and the involvement of a multi-
with incurable or inoperable recurrent rectal cancer disciplinary team approach.
without treatment is invariably poor, with a median The most important factor in exenteration surgery
survival of 9 months and less than 5 per cent 5-year is to achieve a microscopically clear (R0) resection
survival.4–6 margin.4,9,10,15–18 Exenteration surgery is challenging
Surgical resection, defined as pelvic exenteration, and can be very difficult due to the anatomical con-
was first described over 50 years ago, but it was not fines of the bony pelvis and undefined surgical planes
until the 1990s that substantial case series were pub- from previous surgery or radiotherapy. Therefore,
lished. As a consequence, rather than surgical refer- such surgery is best done in specialized units.
ral and aggressive treatment, non-invasive palliative The major series on surgery for LRRC in the past
treatments such as chemotherapy or radiotherapy 20 years have reported overall 5-year survival rates
were implemented. This would relieve symptoms of 20–35 per cent.9,10,15,16,19 Data have shown that
for only a short term and had a median survival of exenteration surgery for LRRC can be performed
10–17 months.2,7,8 Improvements in surgical tech- successfully with minimal mortality rates of 0–1
niques and postoperative care and the implemen- per cent.9,10,15,17 Furthermore, the extent of resec-
tation of multimodality therapy have contributed tion does not influence long-term survival.9,10,16,18
to a significant reduction in operative mortality to Surgery is the only modality that offers a chance
somewhere in the region of 1 per cent in specialized for long-term survival.9,10,15,19 Surgery does entail a
units, but morbidity remains high, at about 30 per significant morbidity rate, but unrelated to the ex-
cent.9,10 It is pertinent to note that after pelvic ex- tent of resection.10 Morbidity rates from the recent
enteration, patients report an improvement in their larger series are in the range 24–27 per cent.9,10,17

HEBK001-C13_p203-221.indd 203 08/02/13 5:34 PM


204  Assessment and management of recurrence

Bedrosian and colleagues suggest there is a bio- 3 years and more than 3 years after initial surgery,
logical variance between truly locally recurrent rec- respectively.25 In a study by Heriot and colleagues,
tal cancers and those that disseminate.15 This is sup- time to recurrence was within a year in 28 per cent
ported by Wong and colleagues,20 but their analysis of patients, in the second year in 29.5 per cent, in
was of colonic cancer. They concluded there are the third year in 18 per cent and more than 3 years
two types of colonic cancers – locally active cancers after surgery in 24.5 per cent.10 In a large series from
and cancers that have the tendency to metastasize. the Mayo Clinic, 19 per cent, 26 per cent and 55 per
Approximately 50 per cent of patients with local cent of patients presented within 1 year, 2 years and
pelvic recurrence do not have distant disease.21–23 after 2 years, respectively.9
Approximately two-thirds of this group can have a
curative surgical resection.10,14 These patients with
R0 pathology have 5-year survival rates of up to 49 Preoperative assessment and
per cent.10 preparation for surgery
The encouraging results of the more recent larger
series have fuelled the growing interest in surgical All patients should have localized pelvic disease
resection for LRRC. This chapter defines and out- confirmed by CT, magnetic resonance imaging
lines the principles of assessment and surgical man- (MRI) and positron-emission tomography (PET)
agement of LRRC. before consideration of proceeding to surgery. Al-
though CT and PET imaging give vital information
regarding metastatic disease, MRI is most useful
Presentation in determining resectability and meticulous plan-
ning of the surgical approach. The current con-
Patients usually present with symptoms that reflect sensus amongst the institutions performing pelvic
the location of the recurrence.4,5,9 Symptoms in- exenteration is to perform all three scans. With
clude pelvic pain or referred pain, per rectal bleed- the emergence and popularity of PET/CT, routine
ing and discharge, altered bowel habit and tenes- standard CT scans can be avoided.
mus; the most common is pain. Pain is caused by Less than 5 per cent of pelvic exenterations are
local invasion of bone, the sacrum or pelvic side- said to be irresectable at the time of surgery after
wall, or invasion into nerves, sacral nerve roots or preoperative clinical and radiological assessment
the sciatic nerve. Referred pain is in the distribution including CT, MRI and PET.4
of the involved sciatic nerve.
Asymptomatic patients are identified during rou-
tine colorectal cancer follow-up. Pelvic recurrences Clinical Assessment
may be detected during a digital rectal or vaginal
examination, colonoscopy or routine follow-up Clinical assessment with an examination under
computed tomography (CT) scan. A rising trend in general anaesthetic is frequently performed before
the carcinoembryonic antigen (CEA) should raise pelvic exenteration. Examination without anaes-
the suspicion for recurrence in an asymptomatic thesia is invariably painful and unpleasant for the
patient. This will initiate a cascade of investigations. patient. Tumour fixation and involvement of other
In a Cochrane review published in 2007, intensive pelvic organs can be assessed. Additionally, biopsy
follow-up was found to be associated with improved for tissue confirmation of recurrence, cystoscopy
overall survival and earlier detection of recurrence, and transanal ultrasound can all be performed
suggesting that earlier detection may permit earlier without causing pain to the patient.
intervention leading to improved survival.24
Locally recurrent rectal cancer most commonly
recurs within the first 2–3 years after seemingly Carcinogenic Embryonic Antigen
curative resection.5,24 In a study by Wanebo and
colleagues, 28 per cent of patients presented with- A rise in CEA prompts investigation for recurrence,
in 1 year after initial resection, while 36 per cent, although it does not always rise with recurrence and
17 per cent and 15 per cent presented within 2 years, does not help to determine tumour resectability.26

HEBK001-C13_p203-221.indd 204 08/02/13 5:34 PM


Preoperative assessment and preparation for surgery  205

The specificity is up to 84 per cent, but sensitivity Positron-Emission Tomography


is poor, at 59 per cent. The CEA level may provide
prognostic information, as some studies have dem- Positron-emission tomography scans have been
onstrated that an elevated CEA is associated with shown to be highly accurate in the detection of dis-
worse survival in recurrent pelvic disease.27,28 seminated disease and have been shown to alter the
management of 20–40 per cent of patients by demon-
strating distant disease otherwise undetected on con-
Colonoscopy ventional imaging.26,35 One of the major limitations of
CT and PET scans, however, is that peritoneal disease
Colonoscopy should be performed to biopsy intra- may go undetected until the time of surgery. Further-
luminal recurrent tumour and exclude synchronous more, PET scans are less accurate for the mucinous
colonic malignancies. Anastomotic recurrences are variant of adenocarcinoma and can be false positive
generally discovered during postoperative surveil- with any inflammatory condition.
lance colonoscopies.

Chemotherapy and Radiotherapy


Computed Tomography
Before surgery, patients not previously exposed to
A CT scan of the chest, abdomen and pelvis is per- radiotherapy would usually receive long-course ra-
formed routinely and is the first step for staging re- diotherapy with a chemosensitizer. In an era where
currence before pelvic exenteration. In recent years, radiotherapy is used increasingly, most patients with
PET scans have become more precise in locating recurrent rectal cancer are likely to have already been
the recurrence, and the incorporation of CT in the exposed to radiotherapy; moreover, most patients
form of a PET/CT scan is optimal. will have received the maximum permissible dose. To
circumvent the issue of tissue toxicity, intraoperative
radiotherapy (IORT) has been developed with the
Pelvic Magnetic Resonance Imaging ability to deliver a boost of up to 10 Gy directly to the
tumour bed, which has a biological activity of two-
Magnetic resonance imaging is an essential and to threefold that of external-beam radiotherapy.36
optimal mechanism for locoregional staging of the Intraoperative radiotherapy can be performed as
recurrence and for planning pelvic exenteration. either intraoperative electron-beam radiotherapy
An MRI scan is the best imaging modality avail- or intraoperative high-dose-rate brachytherapy.37,38
able for soft tissue, lymph nodes and tumour defi- Combined with tissue shielding, toxicity to the
nition. The images are superior to those of a CT surrounding tissue can be effectively minimized;
scan. The MRI scan provides accurate assessment therefore, IORT is feasible in previously irradiated
of the tumour, and extent of involvement of adja- patients.10,37–40 This has been advocated for R2 resec-
cent pelvic viscera, including invasion into bone, tions at the time of surgery, but unfortunately it is
nerve, vessels and ligaments. A significant disad- not readily available in all units. Although some au-
vantage of MRI is the difficulty in distinguishing thors have demonstrated significant survival benefit
between recurrent disease and post-radiotherapy with IORT, it does not replace inadequate surgery.40,41
or post-surgical fibrosis and inflammation;29 the Intraoperative radiotherapy becomes less significant
addition of a PET scan, or even better a PET/CT with the advancements in pelvic exenteration, in
scan, complements MRI to help distinguish be- particular with the encouraging results of increasing
tween the two.30–32 In an early study by Popovic proportion of complete tumour clearance rates (R0)
and colleagues, MRI was found to have an accu- for laterally recurrent cancers using the technique of
racy of 83 per cent in determining patients’ eli- en-bloc resection of the internal iliac vasculature and
gibility for pelvic exenteration.33 The accuracy of bone.42 Intraoperative radiotherapy will therefore be
MRI will continue to improve with future devel- beneficial for the scenario where resection was pro-
opments in technology and the implementation of posed as potentially complete but intraoperatively
thinner MRI sections.34 inadvertent dissection through the tumour occurs.10

HEBK001-C13_p203-221.indd 205 08/02/13 5:34 PM


206  Assessment and management of recurrence

Relative and absolute able results.44 They include patients with unilateral liver
contraindications for pelvic metastases or solitary pulmonary recurrence. None-
exenteration theless, these represent only a small group of highly
selected patients.
Many authors have reported absolute and relative Lateral pelvic recurrences are no longer an abso-
contraindications for pelvic exenteration (Table 13.1). lute contraindication, as reported by some encour-
Pelvic exenteration surgery has advanced considerably aging results.42 Extended resection laterally is pos-
over the past decade, pushing the boundaries and re- sible, and extended lateral resections with lateral
defining the extent of surgical techniques. As a con- pelvic bone and ligament resections have been per-
sequence, the lists of contraindications are obsolete formed. This provides access to, and the ability to
and continuously being redefined, and the number of resect, some recurrences that extend to and through
absolute contraindications is diminishing. Nonethe- the greater sciatic foramen. Lateral nerve involve-
less, some of the absolute contraindications have re- ment is not an absolute contraindication, because
mained; these include patient performance, multiple as long as the lumbosacral nerve and S1 nerve roots
distant metastases, including para-aortic lymph node are preserved adequate lower limb motor function
disease, and sacral recurrences necessitating resection can be preserved.
of the whole S1 vertebral body. External iliac vessel encasement is not an absolute
Indications for curative pelvic exenteration surgery contraindication if the vessel can be reconstructed
depend on patient factors and the extent of disease. after obtaining an R0 resection. Lower limb oedema
Due to the high morbidity rates and extent of physi- is also not an absolute contraindication.
ological insult induced during and after pelvic ex- Similarly, ureteric involvement is not a con-
enterations, the patient must be relatively fit (with an traindication unless curative resection was deemed
American Society of Anesthesiologists (ASA) score of impossible preoperatively. Preoperative decisions
3 or less) and have tumour recurrence localized to the and preparations for unilateral nephrectomy, ure-
pelvis without distant metastases, unless distant me- terectomy and reimplantation or, if necessary, radi-
tastases are resectable with curative intent. Pelvic ex- cal cystectomy and conduit need to be planned.
enteration with curative intent has been performed in Essentially, if there is inoperable metastatic dis-
patients with limited visceral metastases with accept- ease or an oncological resection is not possible with

Table 13.1. Absolute and relative contraindications for pelvic exenteration.


Study Absolute Contraindications Relative Contraindications
Boyle et al.4
Encasement of external iliac vessels Distant metastases
Extension of tumour through the sciatic notch Primary stage IV disease
Presence of lower limb oedema from lymphatic or venous Extensive pelvic side-wall involvement
obstruction
Poor performance status Inability to achieve R0 resection
Sacral invasion above S2/3
Pawlik et al. 43
Distant metastases Ureteral obstruction
Involvement of common or external iliac vessels Significant medical comorbidity
Para-aortic lymph node metastases Poor performance status/inability to care for stomas
Involvement of sacrum above S1
Tumour extension through sciatic foramen
Pelvic side-wall involvement
Ogunbiyi et al.6 Tumour invading above S2 Distant metastases
Involvement of pelvic side-wall or pelvic nerves Diffuse intra-abdominal nodal metastases
Involvement of ureters or presence of hydronephrosis on
imaging

HEBK001-C13_p203-221.indd 206 08/02/13 5:34 PM


Anatomy of exenteration surgery  207

a clear pelvic margin, then this would be the major sequent larger study by Hahnloser and colleagues
contraindication for pelvic exenteration. from the Mayo Clinic found recurrence with pain
Metastatic disease no longer automatically pre- and increased fixity of the tumour to be associated
cludes patients from pelvic exenteration. There is with greater operative challenges, difficulty with
some evidence to suggest that patients with limited delivering intraoperative radiotherapy and sig-
and resectable metastasis can survive, and some even nificantly reduced survival.9 In 1999, Wanebo and
beyond 5 years. 41,44–49 Nevertheless, these patients are a colleagues proposed a classification system based
highly select group, and pelvic exenteration was per- on the Union for International Cancer Control
formed after considerable deliberation and debate. (UICC) TNM staging system (Table 13.2), where
Similar to the situation in primary colorectal cancers, the tumour recurrence stage increased depending
there is no consensus on the optimal sequence of sur- on the depth of invasion through the bowel wall
gery. Some surgeons advocate staged procedures while and beyond.25
others perform synchronous resections.41,44–50 The Memorial Sloan Kettering group, Leeds
Rarely, pelvic exenteration can be performed for group and Yamada and colleagues described clas-
palliation. These cases include uncontrollable ma- sification systems based on the anatomical location
lignant masses with small and large bowel to vesi- of local recurrence.
cal, vaginal and or cutaneous fistulae, unmanageable In the classification system by the Memorial
malignant cutaneous and vaginal wounds, and in- Sloan Kettering group, recurrence was classified as
tractable sciatic nerve or pelvic soft tissue pain. In central including anastomotic, perirectal and mes-
addition to the indications and contraindications orectal recurrences, anterior, posterior or lateral.52
for surgery, quality-of-life implications and patient The Leeds group classified recurrence as central,
choice are significant factors to be considered before sacral, side-wall or composite, depending on the
informed consent. structures involved (Table 13.2).4 Yamada and col-
leagues classified recurrence into localized (cen-
tral), lateral or sacral recurrences;28 in their study,
Multidisciplinary team lateral recurrences were associated with a signifi-
planning and management cantly lower overall survival.
Classification of LRRC is important for com-
All decisions regarding the management of patients parative and management purposes, but the sites
considered for pelvic exenteration surgery should or number of points of fixation is significantly im-
be made in a multidisciplinary setting with all in- portant for determining clear margins and therefore
volved relevant medical and surgical specialties. The prognosis.
multidisciplinary team meetings should optimally
involve not only the various surgical, perioperative
anaesthesiology and oncological specialties but also Anatomy of exenteration
allied health specialists, including stoma therapy, surgery
rehabilitation, nutrition and psycho-oncology sup-
port and expertise. Patients who present with localized recurrent pelvic
cancer are a heterogeneous group in terms of the
involved pelvic structures; as such, the definition of
Classification extent of resection is debatable. As a result, there is
no standard defined surgical procedure performed,
Several classification systems have been proposed. but instead the type of operation is dependent on
Anatomical classifications are most commonly the site of the tumour, the size of the tumour and
used, although none is universally accepted. Suzuki the number of organs involved. This means that
and colleagues from the Mayo Clinic published a a number of different pelvic organs, vasculature,
classification system based on symptoms and degree muscles, ligaments, nerves and pelvic bone compo-
of fixation (Table 13.2).51 Although this study did nents are excised. To understand the operative ap-
not reveal any association between survival, symp- proaches, the pelvis can be divided into four main
tomatic recurrence and points of fixation, a sub- compartments (Figure 13.1):

HEBK001-C13_p203-221.indd 207 08/02/13 5:34 PM


208  Assessment and management of recurrence

Table 13.2. Comparison between different classification systems.


Study Description
Suzuki et al.51 Symptoms
  S0 Asymptomatic
  S1 Symptomatic without pain
  S2 Symptomatic with pain
Degree of fixation
 F0 Not fixed
 F1 Single-point fixation
 F2 Two-point fixation
 F3 Three or more points of fixation
Wanebo et al. 25
TR1 Local recurrence at local excision site/anastomosis with invasion of submucosa
TR2 Local recurrence at local excision site/anastomosis with full invasion of muscularis propria
TR3 Local recurrence at anastomosis beyond muscularis with involvement of perirectal soft tissue
TR4 Local/focally extensive invasion of rectum or other organs – anteriorly into vagina, uterus, prostate, bladder or
  seminal vesicles, posteriorly into presacral tissue (tethered, not fixed)
TR5 Extensive invasion of pelvis – bony/ligamentous pelvis
Guillem et al.52 Central – including mesorectal, anastomotic, perirectal recurrences or perineal recurrences after
  abdominoperineal resection
Anterior – uterine, vaginal, prostate, bladder, seminal vesicles
Posterior – sacrum and presacral fascia
Lateral – side-wall soft tissue or bony pelvis
Boyle et al.4 Central – tumour confined to pelvic organs without involvement or invasion into bone
Sacral – tumour in presacral space and abuts or invades sacrum
Side-wall – tumour involving structures on pelvic side-wall, including greater sciatic foramen, sciatic nerve,
  through to piriformis and gluteal region
Composite – simultaneous involvement of sacrum and pelvic side-wall
Yamada et al.28 Localized – localized to adjacent pelvic organs and connective tissue
Sacral invasive – recurrent tumour that invades lower sacrum (S3–5), coccyx and periosteum
Laterally invasive – tumour involving sciatic nerve, greater sciatic foramina, lateral pelvic wall or upper sacrum (S1–2)

l Anterior compartment: consists of the bladder, nerves S1–S4, pelvic bone (sacrum and coccyx),
prostate, seminal vesicles, vas deferens, urethra, anterior sacrococcygeal ligament, medial sacrotu-
urogenital diaphragm, dorsal vein complex, ob- berous and sacrospinous ligaments.
turator internus and externus muscle, anterior l Lateral compartment: consists of the pelvic

half of the vagina, anterior pelvic floor muscle side-wall structures, ureter, internal iliac vessels,
(pubococcygeus and puborectalis part of the external iliac vessels, piriformis and obturator
levator ani), and pelvic bone (pubic symphysis, internus muscle around the ischial spine, coccy-
superior and inferior pubic rami). geus muscle, lateral sacrotuberous and sacros-
l Axial compartment: consists of the posterior half pinous ligaments attached to ischium, ischium
of the vagina, uterus, ovaries, fallopian tubes, including tuberosity and spine, lumbosacral
broad ligament, round ligament of uterus, rectum, trunk and sciatic nerve distal to ischial spine
pelvic floor muscle (iliococcygeus part of levator and obturator nerves and vessels.
ani), lower sacrum (S4 down) and coccyx.
l Posterior compartment: consists of the rectum, pel- In general, the four compartments can be best
vic floor (coccygeus muscle), internal iliac vessel understood by their central points as there is some
branches and tributaries, piriformis muscle, sacral degree of overlap of their peripheries. The central

HEBK001-C13_p203-221.indd 208 08/02/13 5:34 PM


Surgical technique  209

(L5) and upper sacrum, as this can be done ab-


dominally. Lateral higher sacrum and full verte-
bral excision of S2 and S3 require the posterior
prone approach.
Depending on the number and type of pelvic
organs involved in the malignant process, the pro-
cedure requires a multidisciplinary team of highly
skilled consultant surgeons from the surgical spe-
cialties of colorectal surgery, vascular surgery, urol-
ogy, orthopaedics, and plastics and reconstructive
surgery. At our institution, colorectal surgeons pre-
dominantly perform the surgery, with other sur-
gical disciplines being involved at the appropriate
Figure 13.1. Anatomical compartments of the pelvis. time. Specialist anaesthetist and experienced thea-
(Reproduced from O’Connell R, Madoff R, Solomon M
tre nursing staff are also required, as the procedure
(eds). Operative Surgery of the Colon, Rectum and Anus,
can take up to 8 to 20 hours.
6th edn. London, Hodder Arnold, 2013, with permission.)

axis of the anterior compartment is the urethra, for Surgical technique


the axial compartment it is the tip of the coccyx,
for the posterior compartment it is the third sacral The aim of surgery is to achieve an R0 resection.
vertebra, and for the lateral compartment it is the This is the principle predictor of long-term sur-
ischial spine. vival. The resection depends on the location of the
In view of the heterogeneity in the types of re- recurrence in the pelvis and whether the surround-
section, these are best defined as either partial or ing organs and structures are involved. The pelvis is
complete pelvic exenterations. A complete pelvic bounded by bone and, if necessary, the bone is re-
exenteration is defined as removal of the primary or sected as part of the en-bloc specimen. This should
recurrent tumour (with or without attached bone) be performed only if stability of the pelvis can be
with all remaining pelvic viscera – that is, all four preserved.
anatomical components of the pelvis. Partial pelvic The planes for exenteration are ill-defined due to
exenteration is defined as removal of the primary or the previous rectal dissection, quite commonly having
recurrent tumour (with or without attached bone) been a total mesorectal excision (TME). There will in-
with en-bloc resection of up to three anatomical evitably be fibrotic tissue from previous surgery and
components of the pelvis. radiotherapy, which can mimic malignancy.
Pelvic exenteration always involves an abdominal The surgeon must be aware of the necessity to
approach, usually with a perineal completion phase obtain clear margins on the resected specimen and
that can be done in the lithotomy or prone position. must dissect laterally to obtain wider circumfer-
The anterior, axial and lateral compartments are ential margins. Involved lateral structures (ureter,
best done through an abdominal combined with a nerve, vessels) are resected, often with bony mar-
perineal lithotomy approach. Posteriorly, resection gins if needed.
of the sacrum from the fourth sacral vertebra (S4) The pathologist should be aware of the anatomi-
down and the sacrospinous ligaments allows radical cal differences between primary resection and pelvic
excision of posterior pelvic floor that is approached exenteration. Circumferential, proximal and distal
from the abdominal side and is often better visual- margins in numerical values become less significant;
ized than in the prone position. instead, whether each margin is free of malignancy is
Involvement of the third sacral vertebrae (S3), of primary importance (i.e. R0, R1, or R2).
and above, because of the nature of the sacroiliac The principles of surgical oncology are essential
joint attachment, requires a prone approach un- for curative intent. To achieve tumour-free resec-
less resection involves only the anterior cortex of tion margins (R0 resection), resection must be en
the midline bones up to the fifth lumbar vertebra bloc. Moore and colleagues observed that central or

HEBK001-C13_p203-221.indd 209 08/02/13 5:34 PM


210  Assessment and management of recurrence

anterior pelvic recurrences were more likely to have Until recently, a lateral pelvic wall recurrence reflected
R0 resections than other locations, especially lateral a negative prognostic factor with consistently poor
recurrences.53 Other institutions have shown that survival rates. Austin and Solomon reported R0 rates
lateral pelvic side-wall recurrence is a significant as high as 53 per cent for lateral recurrences. Pelvic ex-
negative prognostic indicator for survival.10,28 enteration included the excision of the lateral vascu-
Heriot and colleagues classified pelvic exenteration lature (internal iliac vessels) to achieve clear margins.
into radical and extended radical excisions.10 Radical The overall survival rate after local recurrence surgery
resection is confined to the recurrence involving the was 69 per cent and disease-free survival was 72 per
pelvis and neorectum. Radical resections can be ap- cent at a mean follow-up of 19 months.42
propriate for central recurrences. Predominantly, a
radical resection requires an abdominoperineal exci-
sion but, a redo anterior resection or Hartmann’s pro- Classification of Operations
cedure have been performed for selected cases.4,10,17,54
Extended radical resection involves at least one adja- The magnitude and type of exenteration can be
cent pelvic organ (bladder, prostate, seminal vesicles, broadly described in five approaches (the letters in the
uterus, ovaries, vagina, ureter, iliac vessels, small bowel, list below reflect the planes in Figures 13.2–13.5):
sacrum). The subgroups of anterior (bladder, prostate,
A anterior plane for total pelvic exenteration or
seminal vesicles, uterus, vagina, ovaries), posterior
anterior pelvic exenteration;
(sacrum), anteroposterior, and lateral (internal iliac
B anterior plane for axial pelvic exenteration with
vessels, ureter) extended radical resection are defined
subtotal vaginectomy; in male patients, this
by the organs and structures resected.
includes B and the perineal part of A;
Urinary tract involvement is common and can re-
C anterior plane for axial pelvic exenteration with
quire complex urological interventions. Brunschwig
posterior vaginectomy (in female patients only);
described the first pelvic exenteration with ureteral
D posterior plane for total pelvic exenteration or
diversion into the colon for locally advanced pelvic
axial pelvic exenteration;
cancer in 1948.55
E posterior plane for abdominosacral exenteration.
Wanebo, a pioneer and advocate for surgery for
LRRC, has published extensively on abdominosacral
resections.3,25,56,57 He describes a two-stage procedure.
The first stage is performed in the modified Lloyd Dav-
ies position; the second stage, 2 days later, is done in the
prone position. An extensive nodal dissection is per-
formed from the distal aorta to the pelvic side-walls.
The current contentious issues revolve around
the traditional anatomical limits of resecting with a
clear margin, such as involvement of any bone other E

than S2 down, and major vascular or lumbosacral,


sciatic or femoral nerve involvement. This is in con-
trast to previous issues, when consideration of any
surgical resection for recurrence was contentious. An
understanding of outcomes both with and without
exenteration, including long-term survival data, op-
erative morbidity and mortality, length of hospital
stay, length of rehabilitation and quality of life, must
be considered and discussed in detail. Historically, the
mean hospital stay was 5 weeks,16 but now the average D CB A
is closer to 3 weeks, with recovery taking 3–6 months
Figure 13.2. Resection planes in pelvic exenteration (female).
before a stable quality of life is achieved. Reproduced from O’Connell R, Madoff R, Solomon M
Surgery for LRRC has advanced significantly in (eds). Operative Surgery of the Colon, Rectum and Anus,
the past decade. New boundaries have been defined. 6th edn. London, Hodder Arnold, 2013, with permission.

HEBK001-C13_p203-221.indd 210 08/02/13 5:34 PM


Surgical technique  211

C
B A

Figure 13.3. Resection planes in pelvic exenteration (male).


Reproduced from O’Connell R, Madoff R, Solomon M
(eds). Operative Surgery of the Colon, Rectum and Anus, A R
6th edn. London, Hodder Arnold, 2013, with permission.
Figure 13.5. Lateral pelvic resection. Reproduced from
O’Connell R, Madoff R, Solomon M (eds). Operative
Surgery of the Colon, Rectum and Anus, 6th edn.
In broad principles the resection margin for the London, Hodder Arnold, 2013, with permission.
involved compartment of the pelvis is the soft tissue/
bone attachment or en-bloc excision of the involved
bone. Attempting to obtain soft tissue margins of involved compartments will result in unacceptably
high rates of margin positivity (R1 and R2).

Pelvic Exenteration: Abdominal Phase

Perioperative preparations
Stoma sites are marked. If necessary, the elliptical cu-
taneous incision for the vertical rectus myocutane-
ous flap is marked with a surgical skin marker. If the
patient already has a colostomy, this is covered with
a swab and impervious plastic dressing so that it is
not removed during the lengthy procedure. Bowel
preparation is usually advisable, as the long dura-
tion of the exenteration and previous radiotherapy
may contribute to a common requirement to repair
damaged bowel. Bowel preparation can be omitted
in patients with pre-existing stomas, but the colon
should be prepared if a colostomy is to be converted
into a colonic conduit.
Figure 13.4. Resection planes in pelvic exenteration (axial
The insertion of ureteric stents before the
view, female). Reproduced from O’Connell R, Madoff R,
Solomon M (eds). Operative Surgery of the Colon, Rectum
laparotomy is especially useful for central exentera-
and Anus, 6th edn. London, Hodder Arnold, 2013, with tions where the bladder is to be preserved but is
permission. worth considering in all patients.

HEBK001-C13_p203-221.indd 211 08/02/13 5:34 PM


212  Assessment and management of recurrence

Positioning abdominal wall mesh reconstruction, and surgery


The patient is placed directly on a gel mattress to secure often involving myocutaneous flaps. An open ap-
their position and prevent unexpected slippage dur- pendicectomy post exenteration has the difficulty of
ing steep Trendelenburg positioning. The abdominal access as well as the potential risk of damaging the
phase of pelvic exenteration is performed in the modi- inferior epigastric vessels which will compromise.
fied Lloyd Davies position, with both arms secured at The ureters are identified, mobilized with ad-
the patient’s side. The perineal phase can also be per- equate connective tissue to avoid damage to the
formed in the modified Lloyd Davies or prone posi- ureteric blood supply, and protected with vessel
tion. Sacrectomy above S3 is performed in the prone loops. The ureters may have been displaced, usu-
position. Partial anterior sacrectomy, or a distal sac- ally laterally, due to previous pelvic dissection. The
rectomy below S3, can be performed from a transab- gonadal vessels are preserved if possible in male pa-
dominal approach. The anus, if present, is sutured, pre- tients, but they are ligated in female patients during
venting soiling during the operation. The vagina is also radical hysterectomy. Preservation of the abdomi-
included in the skin preparation. When draping the nal portion of the gonadal vessels is important for
patient, exposure should include the groin and thigh in the arterial blood supply of the ureters. The pelvic
preparation for vein harvest if vascular reconstruction portions of the ureters are transected when an ileal
is necessary with an interposition graft or patch. conduit is planned and performed.
Lateral pelvic wall lymph node dissection is rou-
Laparotomy tine with pelvic exenteration. The lymphadenecto-
Despite resectability having been predicted preopera- my includes the nodes of the common iliac bifurca-
tively by CT, MRI and PET scans in a multidiscipli- tion, internal iliac chain and obturator nodes.
nary setting, the initial role of surgery is to exclude In pelvic exenteration with sacrectomy, a metal-
metastatic peritoneal disease that may not have been lic pin is inserted into the sacrum above the level of
detected by preoperative testing. A thorough laparot- sacral involvement. An image intensifier can locate
omy and adhesiolysis is performed meticulously and this pin in the prone position, thus ensuring the pre-
cautiously to prevent enterotomies in radiotherapy- cision of the level of sacral transection.
damaged small bowel. If negative for small-volume
missed peritoneal disease, then the planned exentera-
tion surgery begins. Of note, lateral pelvic side-wall Perineal Phase
involvement and bone fixity are not contraindications
for pelvic exenteration surgery, although these fea- The perineal phase is performed via a wide lithoto-
tures are best assessed by MRI preoperatively rather my exposure, in the prone position or sequentially
than by palpation at laparotomy. after prior lithotomy to widely resect the anterior
Any nodule suspicious for peritoneal metastasis compartment and then in the prone position for
or hard tissue suspicious for carcinoma encoun- the posterior compartment.
tered during the pelvic dissection should be sent for The prone phase is for the sacrectomy, if needed.
frozen section. Confirmation of malignancy may The sacrectomy is the final step for the complete
change the course of the operation. resection of the recurrence. The en-bloc specimen
with the transected sacrum is delivered and the de-
Preparing the pelvis fect is closed, commonly with a myocutaneous ver-
Adhesiolysis is necessary for a re-laparotomy. The tical rectus abdominus muscle (VRAM) flap.
small bowel is mobilized out of the pelvis; the small
bowel is often adherent to the tumour in recurrent
disease, however, and requires en-bloc resection. Different Surgical Approaches to
The involved segments of small bowel are discon- Pelvic Exenteration
nected from the rest of the small bowel with a linear
gastrointestinal stapling and cutting device. The ap- Pelvic exenteration comprises a heterogeneous
pendix, if still in situ, is usually removed; this is to group of surgical operations commonly involving a
avoid potential future difficulty with abdominal ac- combination of the different subcategories. This is
cess after exenteration with combinations of stomas, to achieve an R0 resection, since resection margin is

HEBK001-C13_p203-221.indd 212 08/02/13 5:34 PM


Surgical technique  213

the single most significant prognostic factor deter- reproductive organs – that is, surgery for anterior
mining long-term survival. and central LRRC (see Table 13.2). Additionally,
The different surgical approaches for pelvic ex- lateral pelvic exenteration may also be required as
enteration have been classified into subgroups ac- part of total pelvic exenteration (see Lateral pelvic
cording to the anatomical classification of LRRC. exenteration below).
Overlap between the groups is common, and inevi- Total pelvic exenteration in male patients involves
table in most cases, due to the nature of LRRC. en-bloc resection of the anterior compartment or-
gans (bladder, prostate and seminal vesicles), the
Central (axial) pelvic exenteration rectum and previous anterior resection anastomo-
Central or axial recurrences represent anastomotic, sis with the recurrence (see Figure 13.3, planes D
perirectal and mesorectal recurrences without lateral and A). This incorporates an abdominoperineal ex-
pelvic wall involvement. This is surgery for central cision with radical cystoprostatectomy and forma-
LRRC using the Memorial Sloan Kettering definition tion of a urostomy.
(see Table 13.2). Central recurrences can occur at the In female patients, the more extensive recur-
anastomosis without extraluminal spread. In such a rence that invades beyond the protective barrier
case, redo low anterior resection (including the pre- of the uterus and vagina, with involvement of the
vious anastomosis) or abdominoperineal excision of trigone of the bladder, warrants radical cystectomy
the rectum can be performed without resection of any and formation of a urostomy with total abdominal
other structures (see Figure 13.3, planes D and C plus hysterectomy, bilateral salpingo-oopherectomy and
pelvic part of A). Heriot and colleagues reported that abdominoperineal excision.
63 of 160 patients (39 per cent) in their series had cen- The posterior plane (plane D) is the same for
tral recurrence.10 Unfortunately, redo anterior resec- central pelvic exenteration and total pelvic ex-
tion is uncommon and reported at 5–10 per cent.9,10 enteration. There is no anatomical plane but rather
Abdominoperineal excision is more common and in ill-defined fibrotic tissue as a result of the previ-
the region of 26 per cent.10 The usual scenario is in- ous rectal resection. Hence, the exenteration dis-
volvement of other organs and structures, and thus an section plane may include the presacral fascia to
extended resection is required. achieve clear margins. If there is any doubt that
In female patients, axial pelvic exenteration in- there is no posterior plane and the sacrum may be
volves en-bloc resection of the recurrence and hys- involved, then there should be no hesitation to pro-
terectomy with vaginectomy. The bladder and ante- ceed to abdominosacral exenteration. At this stage,
rior wall of the vagina can usually be preserved (see a frozen section of ‘suspicious presacral tissue for
Figure 13.2, planes D and C) as the uterus and pos- carcinoma’ should be analysed for histological con-
terior wall of the vagina provide protection. If these firmation. This scenario occurs when the tumour
organs are involved, then total pelvic exenteration is has progressed despite no previous evidence of sac-
performed (see Figure 13.2, planes D and A). ral invasion on MRI scan and post-radiotherapy or
In patients in whom the dome of the bladder is the presacral tissue has been interpreted at imaging
involved, wedge resection with primary closure is as ‘postoperative scar tissue’.
performed. If a unilateral distal ureter is involved,
then resection of the ureter with reimplantation Abdominosacral exenteration
with a Boari flap or psoas hitch can be used. Abdominosacral exenteration is a total exentera-
The TME plane no longer exists and therefore tion with the addition of sacrectomy. This is usually
obtaining a clear circumferential margin becomes necessary in patients with extensive posterior LRRC
a great challenge. Additionally, the induration af- (see Table 13.2).
ter radiotherapy usually adds considerably to the Sacrectomy can be performed at the S1/S2 junc-
difficulty. tion without pelvic instability. After completion
of the abdominal phase with reconstruction and
Total pelvic exenteration abdominal wall closure, the patient is transferred
Total pelvic exenteration is an extension of central to the prone jack-knife position for sacrectomy.
pelvic exenteration that includes the anterior com- During the abdominal phase, a metallic pin is in-
partment organs, namely the bladder and adjacent serted into the sacrum at the level of the proposed

HEBK001-C13_p203-221.indd 213 08/02/13 5:34 PM


214  Assessment and management of recurrence

transection. With the aid of an image intensifier, These limitations have been redefined. Over the past
the pin is located while the patient is in the prone decade, the Royal Prince Alfred Group in Sydney,
jack-knife position to confirm the precise level of Australia has implemented a surgical approach for
the sacrectomy. lateral pelvic wall recurrences with encouraging R0
Sacrectomy does not have to be a complete resec- resection rates and survival.42
tion. In selected patients an anterior cortical or partial Lateral pelvic exenteration is performed for lat-
resection of the sacrum can be performed. This is best eral LRRC (see Table 13.2) and may be necessary in
performed in the abdominal phase. Additionally, dis- total pelvic exenteration. Internal iliac vessel liga-
tal sacrectomy, at the S4/S5 junction and below, can be tion and resection is essential for lateral pelvic ex-
performed during the abdominal phase, thus avoiding enteration to ensure an R0 resection margin.
the necessity for the prone position. The first step is to mobilize the pelvic ureters
Before sacrectomy, especially proximal sacrectomy laterally and then ligate the internal iliac artery
(S3/S4 junction and above), internal iliac vessel liga- bilaterally. It is preferable, if possible, to ligate dis-
ture, referred to as ‘pelvic devascularization’, is recom- tal to the first branch of the internal iliac artery to
mended to minimize pelvic haemorrhage. Ligation of maximize healing of the skin and muscles in the
the internal iliac vessels inherently involves the dissec- buttock region. The distal branches of the internal
tion of the lateral pelvic lymph nodes, starting from iliac vessels usually need to be ligated individually.
the common iliac lymph chain and down to the obtu- Next, the venous system is ligated and resected en
rator lymph nodes. Bleeding from inadvertent injury bloc with lateral tumours. This exposes the lum-
of the internal iliac vessels, especially the veins, can be bosacral trunk and sacral nerve roots (S1, S2, S3) as
profuse and has led to uncontrollable and fatal haem- they converge to form the sciatic nerve at the ischial
orrhage.42 Arterial ligation distal to the first branch of spine. The piriformis muscle lies deep to the nerves.
the internal iliac artery is preferred to promote skin Lateral pelvic recurrences may involve sacral
and muscle flap healing.2 The use of vessel loops of nerve roots and the sciatic nerve. If necessary, the
different colours helps to isolate the different struc- sciatic nerve can be sacrificed to obtain an R0 resec-
tures and for proximal and distal vascular control. tion, but at the cost of an altered gait and requiring
During sacrectomy, especially proximal sacral an orthotic aid for the ankle joint.
transection, sacral nerves are sacrificed. The liga- The ischial spine can be resected to gain maxi-
tion of the proximal sacral nerves (S1, S2) leads to mal lateral exposure and reveal the sciatic nerve as
significant morbidity. In contrast, the lower sacral it exits the pelvis via the greater sciatic foramen.
nerves are relatively less important and adequate Additionally, this helps free the lateral aspect of the
limb function is still achievable. sacrum by releasing the coccygeus muscle with the
The lumbosacral and S1 nerve root must be pre- sacrospinous ligament. This is an important step
served to maintain motor function of the lower limb and aids with the sacrectomy. Furthermore, the sac-
on that side. Gait without a foot-drop is preserved if rotuberous ligament is exposed, allowing it to be
the majority of the sciatic nerve is preserved. transected if required. The Gigli saw is a useful tool
Ligation of S2 and lower sacral nerves will result in to transect the spine.
an atonic bladder. The bladder in a patient with recur- Tumour-encased external iliac vessels can be
rent rectal cancer has commonly been damaged pre- resected as part of the en-bloc specimen, with the
viously by radiotherapy and prior surgery. Therefore, assistance of a vascular surgeon. The vascular re-
sacrectomy above S4 may require a radical cystectomy, construction results are most favourable with vein
even if there is no invasion into the bladder, prostate grafts using the contralateral femoral vein and are
or seminal vesicles. The alternatives of self-catheteri- generally preferred over synthetic grafts. Where
zation and permanent suprapubic urinary catheteri- possible, arterial reconstruction is best performed
zation should also be considered. immediately, rather than at completion of the ex-
enteration, to avoid compartment syndrome. The
Lateral pelvic exenteration encased external iliac vein may commonly be
Previously, lateral side-wall recurrence with ureter- thrombosed and laden with clot. This should be
ic, neural or pelvic vascular involvement were con- identified during preoperative imaging and con-
traindications for pelvic exenteration.4,9,10,15–17,22,28,51,57 sideration given to an inferior vena caval filter

HEBK001-C13_p203-221.indd 214 08/02/13 5:34 PM


Surgical technique  215

insertion before surgery to prevent propagation of closure. Similarly, the Mayo Clinic recommended
the thrombus and pulmonary emboli. In chronic flap reconstruction over both primary closure and
venous obstruction, reconstruction with an inter- primary closure with pedicled omentoplasty.62 Peri-
position venous graft is usually not necessary as neal repair with a flap repair reduced wound-relat-
collaterals are already established. ed complications and length of hospital stay.
The pedicle flaps may also be used to reconstruct
the vagina after posterior vaginectomy. Bell and
Reconstruction colleagues reported a 20 per cent complication rate
with vaginal flap reconstruction.63 A small propor-
Urostomy: ileal and colonic urinary conduits tion of patients will continue to have sexual inter-
The ileum and colon have been used to create the course after vaginal reconstruction.63
urostomy.16,58 In one study, there was no difference
in glomerular filtration rates or complications re-
lated to stenosis between the two types.58 Previ- Unresectable Local Pelvic
ously, ‘wet’ colostomies were performed to avoid Recurrences
double stomas (colostomy and urostomy). There
are various methods for ureteric implantation. The Up to 50 per cent of local recurrences present with-
most common techniques used include bilateral out distant metastases and therefore are potentially
(Wallace) and single (Bricker) anastomoses. Uret- amenable to resection.21–45 Patients eligible, and se-
eric stents are useful aids to promote healing and lected for pelvic exenteration may not proceed be-
protect the anastomosis. yond a laparotomy due to the discovery of multifo-
Ureteric anastomotic leakage is a significant cal disease or peritoneal dissemination.
complication and may require temporary bilateral Advancements in imaging modalities, especially
nephrostomies. The irradiated distal ileum and pel- PET/CT and MRI, have decreased the rate of unnec-
vic ureters increases the risk of anastomotic leaks. essary laparotomies where extensive disease is discov-
To minimize this risk, the abdominal rather than ered and curative resection is not possible. Up to 5 per
the pelvic ureter is used for the anastomosis. The cent of patients do not proceed to pelvic exenteration
blood supply of the ureters is protected by not ‘skel- due to previously undetected peritoneal disease or dis-
etalizing’ the ureters during ureterolysis and pre- tant metastases detected intraoperatively.10
serving the abdominal part of the gonadal vessels.
The use of a colonic urostomy may be worth con-
sidering as this avoids the use of radiation-damaged Factors Influencing Curative
distal ileum. Resection
Perineal flap reconstruction Previously, LRRC has been deemed irresectable
The perineal defect may be closed primarily or when there is extension into the sciatic notch, en-
with a pedicle flap. The most common types in- casement of the external iliac vessels, extension to
clude the VRAM flap, gracilis myocutaneous flap the sacral promontory, and the presence of lower
and gluteal myocutaneous rotational flap.25 Clos- limb oedema from lymphatic or venous obstruc-
ing the defect with flaps has decreased the rate of tion.16 When iliac vessel involvement was deter-
perineal wound complications, notably dehiscence. mined intraoperatively, R0 resection was signifi-
Shibata and colleagues showed a significant reduc- cantly less likely than when the vessels were not
tion in major complications of the perineal wound involved.53 Curative pelvic exenterations with
by closing the defect with a gracilis flap in patients transection of the involved sciatic nerve and resec-
undergoing proctectomy following preoperative tion of the external iliac vessels with reconstruction
radiation.59 Chessin and colleagues60 and Butler have been performed successfully, however.
and colleagues61 reported that perineal closure with Radiological evidence of pelvic side-wall recur-
a rectus abdominis muscle flap post-irradiation rence and hydroureter/hydronephrosis are signifi-
significantly reduced major perineal complications, cant factors that limit an R0 resection but are not
including dehiscence, compared with primary contraindications for surgery. Involvement of the

HEBK001-C13_p203-221.indd 215 08/02/13 5:34 PM


216  Assessment and management of recurrence

genitourinary organs, piriformis muscle, iliac ves- Table 13.3. Pelvic exenteration operative and admission
sels and sacrum do not influence margin status.53 If detailsa.
the recurrence is proximal to S1, however, most in- Median operating time 9 h (range 3–16 h)
stitutions would not aim for a curative resection.16,57 Median blood transfusion rate 6.6 units (range 0–17 units)
Sacrectomy above S2 has a high morbidity, includ- Mean number of specialties 3 (range 1–5)
ing neuropathies and pelvic instability.   involved
Median number of days in ICU 3 days (range 0–11 days)
Mean number of days in hospital 25 days (range 5–126 days)
Post-operation Median number of days in 15 days
  hospital
The patient should be transferred to the intensive a
Unpublished data from Royal Prince Alfred Hospital
care unit from the operating theatre for initial post- Pelvic Exenteration database, Sydney, Australia (n 5 240
operative care. In general, patients who have had cases).
exenterations completed within 10 h can be extu- ICU, intensive care unit.
bated and transferred to the high dependency unit;
patients whose surgery is prolonged over 10 h re-
quire continued intubation and ventilation. The published morbidity rates range from 21
Early commencement of parenteral nutrition is per cent to 72 per cent.1,49,51 Larger, more recent
essential. Prolonged ileus is a common complica- series from major institutions report morbidity
tion following exenteration surgery, and there- rates of 23–27 per cent.9,10,17 The most common
fore enteral nutrition is not recommended. Total complications include perineal wound dehis-
parenteral nutrition is commenced on the first cence, pelvic collections and cardiorespiratory
postoperative day. problems. A more comprehensive list is shown in
It is important to note that patients who un- Table 13.4.
dergo sacrectomy or VRAM flap reconstruction
of the perineal or sacral defect should be nursed
in a 308 lateral tilt position to prevent pressure on Prognosis
the surgical site. Upon stabilization, the patient
can then be transferred to a ward with special- Factors that affect survival have been extensively
ized nursing and allied health staff experienced analysed and reported. Reported factors include
in caring for patients with complex postoperative margin status, site of pelvic recurrence, number
needs. of points of fixity, lymph node status at primary
Hospital stay can be prolonged and is usually de- surgery, intraoperative radiotherapy, preopera-
pendent on postoperative complications. The me- tive and postoperative adjuvant therapy, CEA
dian hospital stay for pelvic exenteration involving levels, symptoms related to recurrence (especially
sacrectomy is 15 days (Table 13.3).64 pain), extent of resection, and operation type at
primary resection.9,10,15–17,28,51,53,57 Statistically sig-
nificant negative prognostic indicators are listed
Complications in Table 13.5.
Resection margins have consistently been
The reported mortality rate has been low, rang- shown to be the most significant factor on sur-
ing from 0.3 per cent to 5.4 per cent.4,9,10,15–17,25 In vival.4,9,10,15–18 If microscopic clear margins are
larger, more recent series, the mortality rate has achieved, then survival is significantly improved
been reported at less than 1 per cent.9,10,17 Occa- compared with positive margins. R0 resection
sional perioperative death may result from uncon- margins have been achieved in 38–84 per cent of
trollable pelvic haemorrhage and cardiorespira- patients.4,10,15–17 The 5-year overall survival rates
tory events. The modern results are a significant for R0 resection range from 23 per cent to 49 per
improvement from the historically high mortality cent.9,10,16,17,57 If the margins are positive, however,
rates (10 per cent or more) of the pioneering era of then the 5-year survival decreases significantly to
pelvic exenteration. 0–23 per cent.9,10,15–17

HEBK001-C13_p203-221.indd 216 08/02/13 5:34 PM


Prognosis  217

Table 13.4. Complications. Table 13.5. Negative prognostic indicators for survival.
Perineal Positive margin
  Wound dehiscence Lateral pelvic side-wall recurrence
  Persistent perineal sinus Number of fixed points in the pelvis
  Perineal flap necrosis Positive lymph node at primary surgery
Pelvic collection Elevated carcinoembryonic antigen level
  Wound infection Presentation with pain
  Pelvic abscess/haematoma Sciatic nerve involvement
Gastrointestinal
  Prolonged ileus (R1 or R2) had a lower 5-year survival rate at 16 per
 Enterocutaneous fistula cent (R1, 22 per cent; R2, 15 per cent) when com-
  Anastomotic leak
pared with a curative resection (R0, 37 per cent).9
These finding are consistent with the data from
  Bowel obstruction
other exenteration units, including Leeds General
Cardiorespiratory/vascular
Infirmary, the Memorial Sloan Kettering group and
  Atrial fibrillation
the Japanese reports (Table 13.6).
  Myocardial infarction The extent of resection did not significantly in-
  Pneumonia fluence survival if R0 resection was achieved.9,10,16,18
 Deep venous thrombosis This is consistent with the principles of surgical
 Chronic lower limb oedema oncology. The ‘holy grail’ of exenteration surgery is
Urological
thus an R0 resection.
Patterns of pelvic recurrence have been studied
 Urinary retention
by Yamada and colleagues.28 Five-year survival
 Ileal conduit leak
rates varied, depending on the original site of
 Colovesical fistula recurrence. Localized recurrence (i.e. recurrence
Neurological involving adjacent organs) had a 5-year survival
  Sciatic nerve palsy of 38 per cent, compared with 10 per cent in pa-
 Obturator nerve palsy tients with sacral invasion and 0 per cent in pa-
tients with lateral recurrence. Lateral recurrence
has been observed as a negative prognostic in-
An extensive pooled analysis of 1569 patients dicator by other authors.10,53 Lateral recurrences
who underwent pelvic exenteration surgery was have inferior results because of the anatomical
published in 2006.5 The overall 5-year survival rate limitations determining resectability. Bone and
was 30.7 per cent. Analysis of the specimens with major pelvic side-wall vessels limit the lateral
clear margins (R0) showed an improvement in the extent of resection. In recent years, extended
5-year survival rate up to 38.2 per cent. Heriot and radical resections including the iliac vessels have
colleagues reported similar survival rates from 160 been performed with encouraging results. Over
patients across three different institutions.10 The the past decade, the Royal Prince Alfred Group
overall 5-year survival was 36.6 per cent. The 5-year in Sydney has implemented a surgical approach
survival rate for negative resection margins (R0) for lateral pelvic wall recurrences.42 The R0 resec-
was significantly higher when compared with those tion rate for lateral pelvic recurrences was as high
with involved margins. Overall, 61 per cent were R0 as 53 per cent, and the overall survival rate was
resections with a 5-year survival rate of 49 per cent, 69 per cent at 19 months’ follow-up. R0 resection
and 25 per cent were positive microscopically (R1) was associated with 46 per cent of patients re-
with a 5-year survival rate of 21 per cent. Macro- maining disease free, with an average disease-free
scopic involvement in 9 per cent of patients resulted interval of 30 months.3
in a 5-year survival rate of 17 per cent. Similarly, An increasing number of fixed points of the re-
the Mayo Clinic reported its series of 304 patients current cancer has been shown to have a poorer
and demonstrated that positive resection margins outcome.9,53

HEBK001-C13_p203-221.indd 217 08/02/13 5:34 PM


218  Assessment and management of recurrence

Table 13.6. Resection margins (R0–2) and 5-year survival rates.


5-year 5-year 5-year
Study n R0 (n [%]) survival (%) R1 (n [%]) survival (%) R2 (n [%]) survival (%)
Kusters et al.65 170 92 (54) 40
Heriot et al.10 160 98 (61) 49 40 (25) 21 14 (9) 17
Bell et al.
63
51 41 (80) a
10 (20) a

Bedrosian et al.15 134 65 (49) 30 20 (15) 15 13 (10)


Wiig et al. 66
150 66 (44) 27
Boyle et al.4 64 24 (38)b 24 (38)b 9 (14)b
Moore et al.53 119c 61 (51) 38 (31) 7 (7)
Moriya et al. 16
57 48 (84) 42 9 (16)d 0
Hahnloser et al.9 304 138 (48) 37 27 (6) 22 139 (46) 14
Yamada et al. 57
42 30 (71) 23 12 (29) e
0
Salo et al.17 131 71 (54) 35 13 (10) 23 19 (15) 9

The difference after the sum of all the resections in those patients who had a laparotomy with curative intent but did
not have a resection.
a
4-year survival: R0 41%, R1 10%.
b
3-year survival: R0 68%, R1 34%.
c
18 recurrences from colonic cancers.
d
9 positive margins. 3 had palliative intraoperative radiotherapy. 5-year survival was 0 for positive margins. All
resections includes sacrectomies.
e
12 palliative resections. All 42 patients had sacrectomies.

Some authors have concluded that an elevated quality of life survey performed by Austin and col-
CEA has a worse prognosis,15–17 but other reports leagues showed that patients suffered more in physi-
are inconclusive.10,25 cal than mental terms following pelvic exenteration,
but their overall quality of life was comparable to that
in the general population.67 Miller and colleagues as-
Survival sessed quality of life and cost-effectiveness of therapy
for LRRC; they concluded:
Wanebo and colleagues reported their series in
1999.25 The overall 5-year survival rate was 31 per … surgical resection was a cost-effective treat-
cent, with a 5-year disease-free survival rate of 23 ment for recurrent rectal cancer compared with
per cent. More recent series report overall 5-year non-operative and palliative strategies. Moreover,
survival rates in the range 25–37 per cent, with a the procedure to elicit utility values showed that
median overall survival of 31–43 months.9,10,15,16 patients were more willing to gamble the risks of
The median cancer-specific survival has been re- surgery and the possibility of developing pain or
ported at 48 months,10 with a cancer-specific 5-year complications to have an opportunity to extend life
survival rate in the range 36–41.5 per cent.10,15,16 The than were health care providers. An incremental
overall survival rate after a curative histological re- analysis was performed to combine the costs and
section is in the range 23–49 per cent.2,4,9,10,15,17,18,53,57 benefits of each strategy. The incremental cost–
utility ratio was $109 777 per QALY [quality-
adjusted life-year] for the surgical resection
Quality of Life compared with the non-operative therapy from the
health care provider point of view, and $56  698
The long-term quality of life in survivors of pelvic ex- from the patient view. The diagnostic or palliative
enteration for recurrent and locally advanced primary surgery strategy was always dominated by the
rectal cancer is comparable to that in patients who non-operative option (less costly and more effec-
have undergone primary rectal cancer resections. The tive in terms of survival rate).13

HEBK001-C13_p203-221.indd 218 08/02/13 5:34 PM


References  219

Following pelvic exenteration, patients report an patients with fistulating disease and small bowel
improvement in their quality of life and a reduc- obstruction have pelvic exenteration surgery only
tion in pain. In addition, their quality of life is bet- if cure is possible, otherwise a bowel bypass with
ter than in patients who receive medical palliative anastomosis or a stoma should be considered in the
treatment.11–13 palliative scenario.
Patients with limited and resectable metastatic
disease of the liver or lung may be considered for
Re-recurrence pelvic exenteration if there is potential for cure or
at least disease control.
A second local recurrence occurs in approximately
a third of patients after initial surgery for local re-
currence.68 Bedrosian and colleagues15 and Garcia- References
Aguilar and colleagues19 believe that LRRCs are
biological variants with fewer tendencies to metas-   1. Bakx R, van Tinteren. Surgical treatment of locally
tasize. Garcia-Aguilar and colleagues reported that recurrent rectal cancer. Eur J Surg Oncol 2004; 30:
9 of 14 local recurrences did not metastasize.19 857–63.
Bedrosian and colleagues showed that metastatic   2. Gudderson LL, Sosin H. Areas of failure found at
re-operation (second or symptomatic look) following
disease occurred earlier than the development of a
‘curative surgery’ for adenocarcinoma of the rectum:
second recurrence (9 months v. 16.5 months). Pa- clinicopathologic correlation and implications for adju-
tients lived longer after the second local recurrence vant therapy. Cancer 1974; 34: 1278–92.
(41.5 months) compared with patients who devel-   3. Wanebo HJ, Koness RJ, Vezeridis MP, et al. Pelvic
oped metastatic disease (26.1 months).15 There was resection of recurrent rectal cancer. Ann Surg 1994;
no significant difference in the rate of a second re- 220: 586–95.
currence between an R0 or R1 resection from the   4. Boyle K, Sagar P, Chalmers A, et al. Surgery for locally
recurrent rectal cancer. Dis Colon Rectum 2005; 48:
initial local recurrence surgery.4 The authors stated
929–37.
that they were uncertain of the microscopic margin   5. Heriot AG TP, Darzi A, Mackay J. Surgery for local
status of a second recurrence. Additionally, histolog- recurrence of rectal cancer. Colorectal Dis 2006; 8:
ical orientation and evaluation of the ‘true’ margins 733–47.
may be inaccurate, especially when the specimen   6. Ogunbiyi OA, McKenna K, Birnhaum EH, Fleshman
becomes fragmented.4 Median survival is improved JW, Kodner IJ. Aggressive surgical management of
with R0 resection,54 but other series report no dif- recurrent rectal cancer: is it worthwhile? Dis Colon
Rectum 1997; 40: 150–5.
ference between R0 and R1 resections.17,66
  7. Ito K, Ohtsu A, Ishikura S, et al. Efficacy of chemora-
diotherapy on pain relief on patients with intrapelvic
recurrence of rectal cancer. Jpn J Clin Oncol 2003; 33:
Palliative surgery 180–5.
  8. Wong CS, Cummings BJ, Brierley JD, et al. Treat-
Consistent results have shown that a negative margin ment of locally recurrent rectal carcinoma: results and
for malignancy is the key factor in survival – hence, the prognostic factors. Int J Radiat Oncol Biol Phys 1998;
40: 427–35.
ethical debate about palliative surgery arises. Involved
  9. Hahnloser D, Nelson H, Gunderson L, et al. Curative
surgical margins after pelvic exenteration generally re- potential of multimodality therapy for locally recurrent
sults in poor survival rates and poor quality of life,9,67 rectal cancer. Ann Surg 2003; 237: 502–8.
and therefore it is debatable as to whether it is justifi- 10. Heriot AG, Byrne CM, Lee P, et al. Extended radical
able to perform extensive surgery with considerable resection: the choice for locally recurrent rectal cancer.
morbidity to prolong life for a short period. Dis Colon Rectum 2008; 51: 284–91.
Patients with advanced recurrent pelvic disease 11. Esnaola NF, Cantor SB, Johnson ML, et al. Pain and
quality of life after treatment in patients with locally re-
commonly have intractable pelvic pain and malo-
current rectal cancer. J Clin Oncol 2002; 20: 4361–7.
dorous perineal discharge associated with a signifi- 12. Magrini S, Nelson H, Gunderson LL, et al. Sacropelvic
cant decline in their quality of life. In this scenario, resection and intraoperative electron irradiation in the
pelvic exenteration can be recommended only if management of recurrent anorectal cancer. Dis Colon
surgery is for cure. Similarly, we recommend that Rectum 1996; 39: 1–9.

HEBK001-C13_p203-221.indd 219 08/02/13 5:34 PM


220  Assessment and management of recurrence

13. Miller AR, Cantor SB, Peoples GE, et al. Quality of life 29. Messiou C, Chalmers AG, Boyle K, Wilson D, Sagar
and cost effectiveness analysis of therapy for locally P. Pre-operative MR assessment of recurrent rectal
recurrent rectal cancer. Dis Colon Rectum 2000; 43: cancer. Br J Radiol 2008; 81: 468–73.
1695–701. 30. Arulampalam TH, Loizidou M, Visvikis D, et al. Positron
14. Kapiteijin E, Marijnen CA, Nagtegaal ID, et al. Preop- emission tomography and colorectal cancer. Br J Surg
erative radiotherapy combined with total mesorectal 2001; 88: 176–89.
excision for resectable rectal cancer. N Engl J Med 31. Schlag P, Strauss LG, Georgi P, Herfath C. Scar or
2001; 345: 638–46. recurrent rectal cancer. Positron Emission Tomography
15. Bedrosian I, Giacco G, Pederson L, et al. Outcome af- is more helpful for diagnosis than immunoscintigraphy.
ter curative resection for locally recurrent rectal cancer. Arch Surg 124: 1989.
Dis Colon Rectum 2006; 49: 175–82. 32. Takeuchi O, Koda K, Sarashina H, Nakajima N. Clinical
16. Moriya Y, Akasu T, Fujita S, Yamamoto S. Total pelvic assessment of positron emission tomography for the
exenteration with distal sacrectomy for fixed recurrent diagnosis of local recurrence in colorectal cancer. Br J
rectal cancer in the pelvis. Dis Colon Rectum 2004; Surg 1999; 86: 932–7.
47: 2047–54. 33. Popovic MJ, Hricak H, Sugimura K, Stern JL. The role
17. Salo JC, Paty PB, Guillem J, et al. Surgical salvage of of MR imaging in determining surgical eligibility for pelvic
recurrent rectal carcinoma after curative resection: a exenteration. Am J Roentgenol 1993; 160: 525–31.
10 year experience. Ann Surg Oncol 1999; 6: 171–7. 34. Brown G. Thin section MRI in multidisciplinary pre-
18. Well BJ, Stotland P. Results of an aggressive approach operative decision making for patients with rectal
to resection of locally recurrent rectal cancer. Ann Surg cancer. Br J Radiol 2005; 78: S117–27.
2006; 14: 390–5. 35. Delbeke DVJ, Sandler MP. Staging recurrent colorectal
19. Garcia-Aguilar J, Cormwell J, Marra C, et al. Treat- carcinoma with PET. J Nuclear Med 1997; 38:
ment of the locally recurrent rectal cancer. Dis Colon 1196–201.
Rectum 2001; 44: 1743–8. 36. Vermaas M, Ferenschild FT, Nuyttens JJ, et al. Preop-
20. Wong SKC, Jalaludin BB, Henderson CJA, et al. Di- erative radiotherapy improves outcome in recurrent
rect tumour invasion in colon cancer: Correlation with rectal cancer. Dis Colon Rectum 2005; 48: 918–28.
tumour spread and survival. Dis Colon Rectum 2008; 37. Alektiar KM, Zelefsky MJ, Paty PB, et al. High-dose-
51: 1331–8. rate intraoperative brachytherapy for recurrent
21. McDermott FT, Hughes ES, Pihl E, et al. Local recur- colorectal cancer. Int J Radiat Oncol Biol Phys 2000;
rence after potentially curative resection for rectal 48: 219–26.
cancer in a series of a 1008 patients. Br J Surg 1985; 38. Lowy AM, Rich TA, Skibber JM, et al. Preoperative
72: 34–7. infusional chemoradiation, selective intraoperative
22. Pilipshen SJ, Heilweil M, Quan SH, et al. Patterns radiation, and resection for locally advanced pelvic
of pelvic recurrence following definitive resections of recurrence of colorectal adenocarcinoma. Ann Surg
rectal cancer. Cancer 1984; 53: 1354–62. 1996; 223: 177–85.
23. Rao AR, Kagan AR, Chan PM, et al. Patterns of recur- 39. Lindel K, Willett CG, Shellito PC, et al. Intraoperative
rence following curative resection alone for adeno- radiation therapy for locally advanced recurrent rectal
carcinoma of the rectum and sigmoid colon. Cancer or rectosigmoid cancer. Radiother Oncol 2001; 58:
1981; 48: 1492–5. 83–7.
24. Jeffery M, Hickey BE, Hider PN. Follow up strate- 40. Mannaerts GH, Rutten HJ, Martijn H, et al. Compari-
gies for patients treated for non-metastatic colorectal son of intraoperative radiation therapy-containing
cancer. Cochrane Database Syst Rev 2007; (1): multimodality treatment with historical treatment
CD002200. modalities for locally recurrent rectal cancer. Dis Colon
25. Wanebo HJ, Antoniuk P, Koness RJ, et al. Pelvic Rectum 2001; 44: 1749–58.
resection of recurrent rectal cancer: technical consid- 41. Hashiguchi Y, Sekine T, Sakamoto H, et al. Intraopera-
erations and outcomes. Dis Colon Rectum 1999; 42: tive irradiation after surgery for locally recurrent rectal
1438–48. cancer. Dis Colon Rectum 1999; 42: 886–93.
26. Desai DC, Arnold MW, Burak WE, et al. Positron 42. Austin KK, Solomon MJ. Pelvic exenteration with en
emission tomography affects surgical management in bloc iliac vessel resection for lateral pelvic wall involve-
recurrent colorectal cancer patients. Ann Surg Oncol ment. Dis Colon Rectum 2009; 52: 1223–33.
2003; 10: 59–64. 43. Pawlik TMSJ, Rodriguez-Bigas MA. Educational re-
27. Wanebo HJ, Marcove RC. Abdominosacral resection view pelvic exenteration for advanced pelvic malignan-
of locally recurrent rectal cancer. Ann Surg 1981; 194: cies. Ann Surg Oncol 2006; 13: 612–23.
458–71. 44. Hartley JE, Lopez RA, Paty PB, et al. Resection of
28. Yamada K. Patterns of pelvic invasion are prognostic locally recurrent colorectal cancer in the presence of
in the treatment of locally recurrent rectal cancer. Br J distant metastases: can it be justified? Ann Surg Oncol
Surg 2001; 88: 988–93. 2003; 10: 227–33.

HEBK001-C13_p203-221.indd 220 08/02/13 5:34 PM


References  221

45. Gagliardi G, Hawley PR, Hershman MJ, et al. Prog- 58. Kristjánsson A, Wallin L, Månsson W. Renal function
nostic factors in surgery for local recurrence of rectal up to 16 years after conduit (refluxing or anti-reflux
cancer. Br J Surg 1995; 82: 1401–5. anastomosis) or continent urinary diversion. 1: Glomer-
46. Hashiguchi Y, Sekine T, Kato S, et al. Indicators for ular filtration rate and patency of uretero-intestinal
surgical resection and intraoperative radiation therapy anastomosis. Br J Urol 1995; 76: 539–45.
for pelvic recurrence of colorectal cancer. Dis Colon 59. Shibata D, Hyland W, Busse P, et al. Immediate recon-
Rectum 2003; 46: 31–9. struction of the perineal wound with gracilis muscle
47. Huguier M, Houry S. Treatment of local recurrence of flap following abdominoperineal resection and intraop-
rectal cancer. Am J Surg 1998; 175: 288–92. erative radiation therapy for recurrent carcinoma of the
48. Onodera H, Maetani S, Kawamoto K, et al. Pathologic rectum. Ann Surg Oncol 1996; 6: 33–7.
significance of tumor progression in locally recurrent 60. Chessin DB, Hartley J, Cohen AM, et al. Rectus flap
rectal cancer: different nature from primary cancer. Dis reconstruction decreases perineal wound complica-
Colon Rectum 2000; 43: 775–81. tions after pelvic chemoradiation and surgery: a cohort
49. Wiggers T, de Vries MR. Surgery for local recurrence study. Ann Surg Oncol 2005; 12: 104–10.
of rectal cancer. Dis Colon Rectum 1996; 39: 323–8. 61. Butler CE, Gundeslioglu AO, Rodriguez-Bigas MA.
50. Mirnezami AH, Sagar PM, Kavanagh D, et al. Clinical Outcomes of immediate vertical rectus abdominis
algorithms for the surgical management of locally myocutaneous flap reconstruction for irradiated
recurrent rectal cancer. Dis Colon Rectum 2010; 53: abdominoperineal resection defects. J Am Coll Surg
1248–57. 2008; 206: 694–703.
51. Suzuki K, Dozois RR, Devine RM. Curative reopera- 62. Radice E, Nelson H, Mercill S, et al. Primary myo-
tions for locally recurrent rectal cancer. Dis Colon cutaneous flap closure following resection of locally
Rectum 1996; 39: 323–8. advanced pelvic malignancies. Br J Surg 1999; 86:
52. Guillem J RL. Strategies in operative therapy for locally 349–54.
recurrent rectal cancer. Semin Colon Rectal Surg 63. Bell SW, Dehni N, Chaouat M, et al. Primary rectus
1998; 9: 259–68. abdominis myocutaneous flap for repair of perineal
53. Moore H, Shoup M, Riedel E, et al. Colorectal cancer and vaginal defects after extended abdominoperineal
pelvic recurrences: determinants of respectability. Dis resection. Br J Surg 2005; 92: 482–6.
Colon Rectum 2004; 47: 1599–606. 64. Melton GB, Paty PB, Boland PJ, et al. Sacral resection for
54. Lopez-Kostner F, Fazio V, Vignali A, et al. Locally recurrent rectal cancer: analysis of morbidity and treatment
recurrent rectal cancer: predictors and success of results. Dis Colon Rectum 2006; 49: 1099–107.
salvage surgery. Dis Colon Rectum 2001; 44: 173–8. 65. Kusters M, Dresen RC, Martijn H, et al. Radicality of
55. Brunschwig A. Complete excision of pelvic viscera for resection and survival after multimodality treatment
advanced carcinoma: one-stage abdominoperineal is influenced by subsite of locally recurrent rectal
operation with end colostomy and bilateral ureteral im- cancer. Int J Radiat Oncol Phys 2009; Dec; 75(5):
plantation into colon above colostomy. Cancer 1948; 1444–9.
1: 177–83. 66. Wiig JN. Total pelvic exenteration with preoperative
56. Wanebo HJ, Gaker DL, Whitehill E, et al. Pelvic recur- irradiation for advanced primary and recurrent rectal
rence of rectal cancer: options for curative resection. cancer. Eur J Surg 2002; 168: 42–8.
Ann Surg 1987; 205: 482–95. 67. Austin KK, Young JM, Solomon MJ. Quality of life of
57. Yamada K, Ishizawa T, Niwa K, et al. Pelvic exentera- survivors after pelvic exenteration for rectal cancer. Dis
tion and sacral resection for locally advanced primary Colon Rectum 2010; 53: 1121–6.
and recurrent rectal cancer. Dis Colon Rectum 2002; 68. Sagar PM. Surgical management of locally recurrent
45: 1078–84. rectal cancer. Br J Surg 1996; 83: 293–304.

HEBK001-C13_p203-221.indd 221 08/02/13 5:34 PM


14
Lateral pelvic side-wall nodal
involvement in rectal cancer
Hideaki Yano and Brendan Moran

Introduction has been practically abandoned and has not been


performed due to the following issues:
The risk of metastasis to, and clinical significance
l LPLD is associated with high morbidity;
of, lateral pelvic side-wall nodes in rectal cancer re-
l lateral pelvic side-wall nodal involvement was
mains controversial, although there is now general
considered rare in the West compared with
agreement that involved pelvic side-wall nodes are
Japan and the East;
more common with low rectal cancer.1 Removal of
l involved lateral pelvic side-wall nodes have
these nodes, i.e. lateral pelvic lymph node dissec-
generally been considered to indicate systemic
tion (LPLD), in addition to total mesorectal exci-
disease not amenable to surgery.
sion (TME) is considered a major mechanism in re-
ducing local recurrence and improving survival in Advances in pelvic imaging, however, particular-
Japan (Figure 14.1).2,3 The rationale behind this ap- ly computed tomography (CT) and magnetic reso-
proach is that lateral pelvic side-wall nodal involve- nance imaging (MRI), with the ability to visualize
ment is pathologically confirmed in 10–20 per cent and categorize lymph nodes, together with reviews
of patients with low rectal cancer, and a consider- of the published literature, suggest that lateral nod-
able number of patients with lateral pelvic side-wall al involvement rates may be similar in the West and
nodal involvement survived free of disease after the East and are a particular problem in low rectal
LPLD even before the modern chemotherapy era. cancer.1
Over the past two decades, LPLD has been con-
tinuously developed and refined in Japan, with a
more selective approach in high-risk patients and INCIDENCE OF LATERAL PELVIC
widespread use of autonomic nerve-preserving SIDE-WALL NODE INVOLVEMENT
techniques.1
In Europe and North America, however, the ap- In addition to the predominant lymphatic rectal
proach has been either to ignore lateral pelvic side- drainage that runs upwards or proximally within the
wall nodes or to treat obviously involved nodes mesorectum, the lower rectum has lateral lymphatic
with radiotherapy or, more commonly, chemora- drainage, which may result in involvement of lateral
diotherapy. Lateral pelvic lymph node dissection pelvic side-wall nodes (Figure 14.2). This anatomical

HEBK001-C14_p222-227.indd 222 11/02/13 3:59 PM


INCIDENCE OF LATERAL PELVIC SIDE-WALL NODE INVOLVEMENT  223

Figure 14.1. Lateral pelvic lymph node dissection on the left pelvic side-wall.

feature pertaining to the low rectum has been sup- studies are ongoing to define the sensitivity and
ported not only by numerous experimental studies specificity of imaging in determining the status of
using dye injection and lymphoscintigraphy but also pelvic side-wall nodes. This correlation can only
by clinical observations.1,4 A multicentre study in Ja- be truly documented in patients who have imaging
pan that recruited 2916 patients with rectal cancer followed by LPLD without neoadjuvant therapy.
reported lateral pelvic side-wall node involvement in In addition to modern imaging of the nodes, re-
20.1 per cent of patients with T3/4 low rectal cancer ported predictive factors for lateral nodal involvement
lying at or below the peritoneal reflection.2 It is note- include positive mesorectal nodal status, height of the
worthy that the lower the tumour is, the higher the tumour, depth of tumour invasion, histological grade,
risk of lateral pelvic side-wall node involvement.5 lymphovascular invasion, size of the tumour and fe-
A number of risk factors have been reported to male sex.1,2,6 Some factors can be determined or reliably
predict lateral pelvic side-wall nodal metastasis. predicted before surgery, but others can be determined
Modern pelvic imaging by CT or MRI allows visu- only after surgery. A prospective study reported a high
alization of suspicious pelvic side-wall nodes, and diagnostic accuracy of CT scanning in predicting

Upward
spread

Lateral
spread
Pelvic
plexus

Figure 14.2. Lymphatic drainage from the low rectum exists in the space between the pelvic plexus and the lateral pelvic
side-wall.

HEBK001-C14_p222-227.indd 223 11/02/13 3:59 PM


224  Lateral pelvic side-wall nodal involvement in rectal cancer

Figure 14.3. Imaging suggestive of right lateral pelvic side-wall node involvement.

lateral pelvic side-wall node involvement, allowing pr- in Japan. Key refinements have been a more
eoperative identification of patients most likely to ben- patient-selective approach and nerve-preserving
efit from extended dissection (LPLD) (Figure 14.3).7 surgery.1
In future, MRI or positron-emission tomography Lateral pelvic lymph node dissection for upper
(PET) techniques may further enhance the selection rectal cancer has been almost abandoned in Japan
of patients for appropriate treatment. with a highly selective approach to LPLD. Based on
the large multicentre study, the Japanese guidelines
now suggest LPLD for T3/4 rectal cancer with its
Lateral pelvic side-wall lower edge lying at or below the peritoneal reflec-
node involvement: local tion.8 There is a problem with these criteria be-
or systemic disease? cause the precise depth of the tumour cannot be
determined accurately preoperatively; lateral pelvic
Controversy exists as to whether involved pelvic side-wall nodal involvement was confirmed in only
side-wall nodes represent systemic disease or local- 20 per cent of the patients who had LPLD; and lat-
ized disease that is amenable to surgery. The previ- eral pelvic side-wall node involvement was found
ously mentioned large multicentre study reported in 9.2 per cent of T2 low rectal cancers and would
that the 5-year survival rate of patients with lateral not have been removed based on these criteria. A
pelvic side-wall node involvement was 45.8 per further controversial issue is whether ‘prophylac-
cent, which surpasses the outcomes following liver tic’ LPLD for advanced low rectal cancer offers any
resection for hepatic metastasis.2 survival benefit. This depends on the accuracy of
Patients with lateral pelvic side-wall node in- preoperative assessment of tumour depth, nodal in-
volvement constitute a heterogeneous group. In volvement and other preoperative factors.
some patients the disease is undoubtedly systemic Lateral pelvic lymph node dissection is known
and carries a poor prognosis, indicating a need for to be associated with significant morbidity, longer
neoadjuvant and adjuvant treatment or even pal- operating time, greater blood loss and genitouri-
liative care. Other patients have localized disease as nary dysfunction. Nerve-sparing techniques have
demonstrated by favourable 5-year survival rates been developed and refined, with marked improve-
achieved with LPLD. ments in urinary and sexual function in recent
years.9,10 There are concerns, however, as to whether
preservation of autonomic nerves, particularly the
OPERATION inferior hypogastric plexus, may compromise the
oncological outcomes when cancer cells are likely
In contrast to Western countries, LPLD has been to have metastasized through the lateral lymphatic
continuously promoted, performed and refined channels to the lateral pelvic side-wall nodes.

HEBK001-C14_p222-227.indd 224 11/02/13 3:59 PM


OPERATION  225

Figure 14.4. Dissection between the external iliac artery Figure 14.5. Dissection of the medial aspect of the
and the psoas major muscle. external iliac vessels.

Operative Techniques served. The ureter is then secured using a vessel loop
or tape to reduce the risk of inadvertent damage.
Lateral pelvic lymph node dissection is usually per- The external iliac artery is exposed and secured
formed following completion of rectal resection using a vessel loop or tape. By exposing the medial
using the principles of TME or sphincter resection aspect of the psoas major muscle, the fat tissue
in patients who require abdominoperineal excision. between the external iliac artery and the psoas
In restorative procedures, LPLD is performed be- major is dissected and the obturator nerve is
fore the anastomosis. identified (Figure 14.4). Care should be taken not to
If there are concerns about the margins of the damage the fifth lumbar vein that drains into either
suspicious nodes, the internal iliac artery or vein the inferior vena cava or the external iliac vein.
and the autonomic nerves, i.e. inferior hypogastric Before dissection of the obturator region, the an-
plexus (pelvic plexus), should be excised en bloc. terior aspect of the internal iliac artery is exposed
The main aspects of LPLD on the right pelvic and the fat tissue surrounding the external iliac vein
side-wall are as follows: is dissected from the cranial to the caudal direction.
The hypogastric nerve is secured using a vessel There is generally no need to follow the external
loop or tape. Caudally, the inferior hypogastric plexus iliac vessels to their distal ends, unless there is sus-
and the pelvic splanchnic nerves (nervi erigentes) are picious tissue there, as this may increase the risk of
dissected off the parietal endopelvic fascia and pre- lymphoedema of the leg (Figure 14.5).

Figure 14.6. Dissection of the obturator region.

HEBK001-C14_p222-227.indd 225 11/02/13 3:59 PM


226  Lateral pelvic side-wall nodal involvement in rectal cancer

Figure 14.7. Dissection of the dorsal aspect of the Figure 14.9. Identification of the inferior vesical vessels.
obturator region.
situated on the lumbosacral trunk and the sacral
By retracting the ureter and the external iliac nerve plexus (Figure 14.7). Care should be taken,
vessels laterally, the optimal surgical field will be particularly if excising the internal iliac vessels, not
obtained. The dissection of the obturator region is to damage the small branches of the vein, which can
commenced from where the internal and external result in considerable bleeding.
iliac arteries branch off the common iliac artery. The The caudal part of the obturator area is dissected
superior vesical artery and the obturator vessels are by pulling medially the superior vesical artery and the
then identified, both of which can be excised if nec- ureter. The levator ani muscle is exposed just caudal
essary. Retracting the superior vesical artery medi- to the internal obturator muscle (Figure 14.8).
ally, dissection is continued by exposing the obtura- More medially, identification of the inferior
tor nerve and the internal obturator muscle as far as vesical vessels that are situated just lateral to the
the point where the nerve enters the obturator canal inferior hypogastric plexus is important. This is
(Figure 14.6). When using diathermy dissection, lo- followed by division of the distal branches of the
cal injection of lidocaine is very helpful to prevent inferior vesical artery and vein at the point just
stimulation of the adductor muscles. Alternatively, proximal to the bladder (Figure 14.9). Lateral pel-
ultrasonic coagulating shears may prove useful. vic lymph node dissection is completed by divid-
On the craniomedial aspect, the lymphatic tis- ing the inferior vesical vessels from the internal
sue is dissected off the internal iliac vessels that are iliac vessels (Figures 14.10 and 14.11).

Figure 14.8. Dissection of the caudal aspect of the


obturator region. Figure 14.10. Division of the inferior vesical vessels.

HEBK001-C14_p222-227.indd 226 11/02/13 3:59 PM


References  227

Japan and Netherlands, focusing on the patterns of


local recurrence. Ann Surg 2009; 249: 229–35.
  4. Bell S, Sasaki J, Sinclair G, et al. Understanding the
anatomy of lymphatic drainage and the use of blue-
dye mapping to determine the extent of lymphadenec-
tomy in rectal cancer surgery: unresolved issues.
Colorectal Dis 2009; 11: 443–9.
  5. Ueno M, Oya M, Azekura K, et al. Incidence and prog-
nostic significance of lateral lymph node metastasis in
patients with advanced low rectal cancer. Br J Surg
2005; 92: 756–63.
  6. Ueno H, Yamauchi C, Hase K, et al. Clinicopathologi-
cal study of intrapelvic cancer spread to the iliac area
in lower rectal adenocarcinoma by serial sectioning.
Br J Surg 1999; 86: 1532–7.
  7. Yano H, Saito Y, Takeshita E, et al. Prediction of lateral
Figure 14.11. View after completion of lateral pelvic lymph
pelvic node involvement in low rectal cancer by con-
node dissection on right pelvic side-wall.
ventional computed tomography. Br J Surg 2007;
94: 1014–9.
  8. Japanese Society for Cancer of the Colon and Rectum
References (ed.). JSCCR Guidelines 2010 for the Treatment of
Colorectal Cancer, 2nd edn. Tokyo, Kanehara & Co.,
  1. Yano H, Moran BJ. The incidence of lateral pelvic side- 2010: 13–5.
wall nodal involvement in low rectal cancer may be   9. Moriya Y, Sugihara K, Akasu T, Fujita S. Nerve-sparing
similar in Japan and the West. Br J Surg 2008; surgery with lateral node dissection for advanced lower
95: 33–49. rectal cancer. Eur J Cancer 1995; 31A: 1229–32.
  2. Sugihara K, Kobayashi H, Kato T, et al. Indication and 10. Matsuoka H, Masaki T, Sugiyama M, Atomi Y. Impact
benefit of pelvic sidewall dissection for rectal cancer. of lateral pelvic lymph node dissection on evacuatory
Dis Colon Rectum 2006; 49: 1663–72. and urinary functions following low anterior resection
  3. Kusters M, Beets GL, van de Velde CJH, et al. A com- for advanced rectal carcinoma. Langenbecks Arch
parison between the treatment of low rectal cancer in Surg 2005; 390: 517–22.

HEBK001-C14_p222-227.indd 227 11/02/13 3:59 PM


15
Intestinal stoma and the role
of defunctioning a low anastomosis
after anterior resection
David Mitchell, Kandiah Chandrakumaran and Steven Arnold

Introduction accessing the colon in the lumbar region without


opening the peritoneum. A few surgeons attempted
Surgeons have been constructing intestinal stomas this procedure but failed, and the approach was
for over 200 years, but the modern ileostomy and rapidly abandoned. The first panproctocolectomy
colostomy have been developed from our predeces- and ileostomy was performed by Miller in 1949.
sors’ trial and error. When total mesorectal excision (TME) is per-
Littre proposed the ‘inguinal colostomy’ in 1710 formed, with a very low division of the muscle tube
for infantile imperforate anus but never actually to enable re-anastomosis, most surgeons would
performed the operation. It took more than 65 years elect to use a proximal defunctioning stoma to re-
before the first stoma was performed in 1776, when duce the consequences and possibly the incidence
French surgeon Pillore created a defunctioning of anastomotic leakage. It has been well reported
stoma for obstructing rectal cancer on Monsieur that when a low rectal anastomosis is performed
Morel. This was a caecostomy and the patient died (by definition, less than 7 cm from the anal verge
after 28 days from complications not directly relat- on rigid sigmoidoscopy), then the risk of an anas-
ed to the stoma; this was not reported in the medi- tomotic leak rises significantly; the lower the anas-
cal press until 1840. In 1783, Dubois performed the tomosis, the higher the risk.1
first ‘Littre’s operation’ on a 3-day-old infant for The term ‘anterior resection’ by definition involves
imperforate anus, but the baby died on the tenth an operation whereby the superior rectal artery has
day. The first successful colostomy was performed been ligated and the anastomosis has been performed
in 1793 by Duret, on an infant with a maldeveloped to the top of the rectum. The term is increasingly used
perineum and absence of the rectum. for surgical management of pathology of the sigmoid
With the advent of anaesthesia, diversion colos- colon, especially in the laparoscopic era, where the
tomy became popular for the management of large technique enables modern stapling guns to construct
bowel obstruction. In the early days there was a the anastomosis without the need for intracorporeal
high mortality rate. Amussant, a Parisian surgeon, suturing. Even when resection of the upper rectum is
attributed this high mortality rate to peritonitis as added, the resulting anastomosis is relatively high, the
a result of opening the peritoneum. He suggested risks of anastomotic leakage are much reduced, and a

HEBK001-C15_p228-238.indd 228 08/02/13 5:29 PM


LOOP ILEOSTOMY  229

stoma is frequently unnecessary and can be avoided. marking stoma sites with either surgical skin clips
An ‘anterior resection’ involving TME down to the or a silk stitch as soon as the patient is prepared
pelvic floor, however, is a different matter, with much and draped, as the ink marks may completely rub
higher risks of leakage. off during the operation, especially if the adhesive
dressing is removed for skin preparation.
In the emergency situation, it may not be pos-
PREOPERATIVE PLANNING sible to have optimal planning for siting a stoma,
such as surgery for obstruction, bleeding, ischaemia
Almost all TME operations will be scheduled elec- or abdominal trauma. Nevertheless, the surgeon
tively. This gives the patient and team the chance to should take account of all of the above factors when
prepare for the high likelihood of needing a stoma. choosing where to site a necessary stoma, as the
Preoperatively, there may be the opportunity to ­patient may have the stoma long term and often
meet another patient with a stoma, thereby allow- indefinitely.
ing personal questions and expectations to be ad- A major role of the stomatherapist is to provide
dressed. The patient should meet a specialist stoma the patient with preoperative counselling. It has
nurse (stomatherapist), who has a key role to play been reported that the presence of a stoma may de-
in the preoperative preparation and peri- and post- lay the discharge of an otherwise well patient by as
operative management. much as 4 days.2 With the drive towards enhanced
Optimal abdominal wall stoma siting is a crucial recovery and early discharge, preoperative training
step in successful outcome. The aim is to locate a in the care, emptying and changing of a stoma bag
flat smooth skin surface so that stoma appliances may hasten the patient’s discharge. It is essential to
can be fitted with as near perfect seal as possible. provide contact numbers of the stoma care team to
This reduces the likelihood of leaking and skin ex- the patient on discharge so that they can access spe-
coriation caused by irritant gastrointestinal con- cialist help quickly in the event of problems.
tents. The patient and stoma nurse carefully consid- It is important to note that a stoma may have
er factors such as abdominal shape, previous scars, profound impact on a patient’s life. Body dysmor-
and where the patient can physically see when se- phia is common and the impact is dramatically in-
lecting and marking a suitable stoma site. Personal creased by leaking and poorly functioning stomas.
factors such as clothing styles and where belts are Therefore, the surgeon should take the utmost care
worn are also considered. Siting is carried out away and exemplary surgical techniques to avoid any
from bony landmarks, and skinfolds are avoided to complications. This may be especially important to
prevent the stoma from lying in a crease when the remember at the end of a long operation such as
patient is sitting upright. Note that skinfolds may TME for rectal cancer or total colectomy.
not be apparent when the patient is supine dur-
ing the operation. Some surgeons have suggested
that the risk of developing a parastomal hernia is LOOP ILEOSTOMY
reduced when the bowel is brought through the
rectus abdominis muscle, and hence the stoma site A loop ileostomy is most commonly used to de-
is marked medial to the lateral edge of the rectus function a low rectal anastomosis (Figure 15.1). A
abdominis muscle. Once a suitable site is identified, loop ileostomy appears to have become the pro-
the site is marked with an indelible marker pen. cedure of choice in most units for defunctioning
The site can then be covered with a clear adhesive a low rectal anastomosis and has superseded the
plastic dressing, thereby preventing the mark from equally effective loop colostomy for defunctioning.
becoming faint or rubbing off. It is good practice to A suitable point on the terminal ileum is selected.
have both left- and right-sided stoma sites marked This is usually about 25–30 cm proximal to the ile-
as surgery may not progress according to plan, ocaecal valve. If it is nearer to the ileocaecal valve,
thereby allowing both a defunctioning ileostomy or then in theory back pressure on the segment at
an end colostomy formation. Pre-existing marks al- the time of stoma reversal may increase the likeli-
low whichever stoma is necessary to be formed in a hood of a leak from the ileostomy closure site. A
good position. The authors favour the practice of disc of skin and underlying fat is then excised at

HEBK001-C15_p228-238.indd 229 08/02/13 5:29 PM


230  Intestinal stoma and the role of defunctioning a low anastomosis after anterior resection

the marked site and the underlying sheath exposed. serted inferiorly, attaching the small bowel to the
The assistant can attach a Littlewoods or similar in- skin. This holds the loop in place and marks where
strument to the edge of the midline sheath, allow- the flush afferent limb will be attached to the skin.
ing gentle traction to straighten the abdominal wall Following closure and protection of the main
so the stoma can be brought out straight. Tradition- wound, the ileostomy can then be formed. The il-
ally a cruciate incision was made in the sheath, but eum is opened transversely by an incision a couple
a longitudinal incision is now recommended; the of millimetres above the previously placed effer-
underlying rectus muscle fibres are gently separated ent small bowel-to-skin sutures. This incision is
with Langenbeck’s retractors to expose the poste- extended for about two-thirds of the circumfer-
rior rectus sheath. This is then incised and then ence of the bowel, but preserving the mesenteric
gently stretched to permit the formation of the border, which is left in continuity. Three eversion
stoma. Inserting two fingers through the site usu- sutures are then placed at the 12, 3 and 9 o’clock
ally allows enough stretching for easy passage of the positions. Dissolvable sutures are preferred, such as
bowel, without leading to too big a defect with an 3.0 monocryl or vicryl rapide. The superior-most
increased risk of subsequent herniation. Babcock’s stitch is then used to take a seromuscular bite ap-
forceps are ideal for passing through the site, grasp- proximately 6 cm proximal to the cut end of the
ing the small bowel, and gently guiding the bowel bowel, with the two side sutures taking a similar
through the abdominal wall. Orientation of the bite at approximately 5 cm from the cut end of the
intra-abdominal small bowel is checked, preferably small bowel. Once all three are in situ, gentle trac-
with the efferent limb positioned superiorly. At least tion on all three usually results in eversion of the
6 cm of bowel, measured on the antimesenteric bor- bowel to form a spout approximately 2.5 cm long,
der, should be delivered, to allow sufficient length facing slightly downwards. Sometimes gentle assist-
for formation of a good spout. Before closing the ance is required to evert the spout, and the blunt
abdomen, two or three dissolvable sutures are in- end of Langenbeck’s retractors can be used to facili-
tate this. Once these stitches are secure, further su-
tures can be placed between them to secure the new
mucocutaneous junction around the circumference
of the stoma. Haemostasis is then checked, and a
stoma bag can be cut to shape and fitted.

End ileostomy

In the event of a panproctocolectomy (e.g. a patient


with rectal cancer on a background of ulcerative
colitis), an end ileostomy must be fashioned. After
removing the large bowel, a disc of skin is excised
from the abdominal wall at the marked site, along
Proximal with a disc of subcutaneous fat. The terminal ileum
spouted will usually have been cross-stapled and divided
end close to the caecum. After entering the abdominal
Distal flush cavity as for a loop ileostomy, the end of the ileum
end is grasped gently with Babcock’s forceps and with-
drawn through the opening. The mesentery supply-
ing the end of the bowel is also brought through;
care must be taken not to traumatize the mesentery
and risk a haematoma or disruption of the blood
supply. Small bowel orientation is checked to en-
sure no rotation of the mesentery, and the laparot-
Figure 15.1. Loop ileostomy. omy wound is closed.

HEBK001-C15_p228-238.indd 230 08/02/13 5:29 PM


ALTERNATIVES  231

The staple line is cut off and a spout is created to skin. These should be placed in an extramucosal
similar to the technique described above. The au- fashion. It is common to use a ‘bridge’ under the co-
thors use four sutures placed at 6, 9, 12 and 3 o’clock lonic loop, either purpose-designed or an adapted
on the circumference of the open small bowel, go- piece of tubing (as described above to pull out the
ing from inside out and then through the serosa loop), to help support the stoma. This is removed
and muscle of the proximal small bowel 6 cm from at 5–7 days. The disadvantage of a bridge is that it
the cut end for the uppermost two sutures and 5 cm interferes with appliance application and changing.
from the end for the lower two sutures. Traction on A loop colostomy may also be required in some
all four sutures usually inverts the bowel to produce very symptomatic patients with rectal cancer before
a spout; the slightly longer uppermost length pro- neoadjuvant chemoradiotherapy or occasionally
duces a downward tilt at rest to direct liquid con- to relieve obstruction. The traditional trephine co-
tents into a stoma bag. The sutures are tied and held lostomy method should now be almost exclusively
on forceps while intervening sutures are placed. replaced by a laparoscopic technique to obviate
the reported risks of bringing the wrong end of a
stoma or a rotated loop via a trephine. Difficulty
Loop colostomy often arose in correctly identifying the orientation,
especially with redundant sigmoid or transverse co-
A transverse loop colostomy is a good alternative lon. A laparoscopic technique is optimal to assess
defunctioning stoma, and a premarked site will the mesenteric length, allow mobilization and con-
usually be in the right upper quadrant. Once the firm orientation. If a trephine sigmoid colostomy is
rectal anastomosis is completed, the portion of the being performed, a useful technique is to intubate
transverse colon that can be exteriorized at the pr- the colon peranally using a flexible sigmoidoscope.
eoperatively marked site is selected. It is important If using a sigmoidoscope, it is important not to
that there is no undue tension on the mesentery or inflate too much air and to limit insufflation to a
appearances of ischaemia; full mobilization of the ­minimum.
hepatic flexure may be required. An ellipse of skin
is removed and the subcutaneous fat is split and re-
tracted with Langenbach’s retractors. The anterior End colostomy
rectus sheath is exposed and a cruciate or linear
fascial incision made. The rectus abdominis muscle An end colostomy is performed after abdomi-
is split longitudinally. Great care must be taken to noperineal excision of the rectum and in some
avoid injury to the inferior epigastric vessels. The emergencies in the form of a Hartmann’s proce-
peritoneum is opened and the selected portion of dure. In both cases, the distal end of the proximal
the colon is exteriorized with Babcock’s forceps. colon is usually stapled closed during the laparot-
A useful technique is to pass a length of tubing omy, until the specimen is dissected and removed.
around the transverse colon, through an avascular The closed end is then exteriorized through the
area in the mesentery, and use this to pull the colon ­preoperatively marked site, without tension, similar
through the abdominal wall opening. It is impor- to a loop colostomy. Scissors can be used to excise
tant at this stage to ensure that the orientation is the staple line, and the edges of the colon and skin
correct, there is no undue tension and the opening are approximated with circumferential interrupted
is adequate. In patients who are obese, it may be nec- absorbable sutures.
essary to widen the opening to ensure this. A cou-
ple of ‘stay’ sutures are applied and the laparotomy
wound is closed in the usual manner. The wound ALTERNATIVES
is dressed and protected. A transverse enterotomy
is made in the loop, to include approximately two- Although formal proximal defunctioning gives the
thirds of the circumference, and the bowel ends best protection to a distal anastomosis, alternative
are cleaned with povidone–iodine. Haemostasis is measures have been described.
achieved, and interrupted absorbable stitches are Caecostomy (intubation of the caecum, usually
applied to approximate the open edge of the colon with a urinary catheter) has been used, although

HEBK001-C15_p228-238.indd 231 08/02/13 5:29 PM


232  Intestinal stoma and the role of defunctioning a low anastomosis after anterior resection

the procedure is largely historic. The appendix was the bowel. This may be supplemented by luminal
amputated and the stump brought on to a small endoscopy.
opening on the abdominal wall, in the right iliac If the pathology of the resected specimen is re-
fossa. The hypothesis was that a caecostomy al- ported as an advanced tumour, adjuvant therapy
lowed some venting of the accumulated gas, there- may be offered. The timing of adjuvant therapy is
by reducing the luminal pressure on an anastomo- thought to be important, as undue delay from the
sis. It did not defunction the faecal stream, however, original surgery may render adjuvant therapy less
and this practice has now been superseded by loop effective. Thus, reversal of a defunctioning stoma
ileostomy or colostomy if protection is deemed may delay treatment, so many oncologists prefer
necessary. to treat before stoma reversal, and the reversal may
Other methods proposed to decrease the poten- have to be deferred for 6 months or longer.
tial pressure across an anastomosis involve peranal The principles of stoma reversal are similar for
intubation.3 Again, a urinary catheter has been used loop ileostomy or colostomy reversal. An incision
across a low anastomosis, but a custom-designed is made around the stoma at the interface of the
silastic stent that sits across a low anastomosis and skin and mucosa. Some surgeons recommend a
has an aperture that allows flushing by a bladder sy- transverse elliptical incision a few millimetres away
ringe has been evaluated, with encouraging results.3 from the skin mucosa interface, with the ends ta-
The tube or stent is removed at 5–7 days or when pering together at the 3 and 9 o’clock positions to
the patient has opened their bowel. improve the eventual scar. Once the edge of the
bowel is identified, careful dissection proceeds cir-
cumferentially to free up the adhesions between
Closure of stomas the bowel and the abdominal wall. Sharp dissec-
tion is preferred to diathermy, thereby preventing
Timing of stoma-reversal surgery is important and thermal damage to the bowel. This should be a rel-
often is determined by multiple factors. atively bloodless plane; if difficulty is encountered
Before stoma reversal, most surgeons advocate with small bleeding points, it may be that there is
confirmation of anastomotic healing by a fluoro- a further layer of tissue still attached to the bowel
graphic water-soluble contrast enema. It is recom- wall. The dissection is deepened circumferentially,
mended that the surgeon performs a digital rectal separating the bowel from the sheath and muscle
examination before referral for a contrast study; layers, until the abdominal peritoneal cavity is
often a soft stricture is encountered and should be ­entered. Within the peritoneal cavity, the remain-
digitally dilated. Care is needed in inserting the soft ing adhesions can be freed, often by sweeping a fin-
rubber enema tube to avoid undue trauma, espe- ger in the peritoneal cavity deep to the abdominal
cially to a very low anastomosis. Gastrograffin or a wall. This allows delivery of a good length of both
similar agent is then allowed to flow down the tube bowel ends through the abdominal wall.
to fill the neorectum, and X-ray images are taken. Attention should then be focused on preparing
These X-rays can be taken a few weeks after sur- the bowel for anastomosis. There may be a small
gery, thereby allowing the reversal operation to be rim of skin remaining on the edges of the bowel,
planned. If no adjuvant therapy is proposed, then and this should be excised. Bleeding during this
the patient may be booked for reversal at 6–8  weeks manoeuvre is a good sign, as it signifies that there
after their resection, although generally the longer should be a healthy blood supply to the new anasto-
the time interval (even beyond this), the less diffi- mosis. The ends of a spouted ileostomy should then
cult the procedure. The delay allows the adhesions be unfolded. Often, as the stoma has been formed
between the stoma and abdominal wall to mature only a few weeks previously, the adhesions may
to a point where subsequent dissection is safer, with be flimsy without any undue fibrosis. Sometimes
less peristomal and bowel oedema. gentle digital pressure alone is enough to evert the
Some surgeons rely on clinical examination spout, but on other occasions careful sharp dissec-
alone to determine whether the anastomosis is in- tion may be required. This allows the ends of the
tact. A careful digital examination is performed to bowel to be approximated, assisted by the exist-
feel for an intact join around the circumference of ing continuity of the back wall. The bowel is then

HEBK001-C15_p228-238.indd 232 08/02/13 5:29 PM


STOMA COMPLICATIONS  233

sutured. The authors favour using a 3/0 polydiox- Due to the risk of postoperative infection, some
anone suture with interrupted stitches to form an surgeons prefer to leave the skin wound open. Some
end-to-end anastomosis. The first sutures should have advocated a circumferential skin incision at
be placed at each end of the closure; it can be use- the skin/mucosa interface and then subsequently a
ful to hold these two tied sutures apart using artery nylon purse-string to draw the edges together and
forceps, thereby facilitating further stitch place- an alginate dressing as a plug in the centre. This re-
ment. Further extramucosal stitches are placed and quires regular dressing changes postoperatively, but
tied, until the last suture is placed. Careful inspec- the resulting scar is usually surprisingly neat and
tion of the anastomosis ensures no gaping defects, contracts down, with very low infection rates.
and the bowel can then be gently reduced into the
abdominal cavity. If reduction is difficult, it is better
practice to further open the sheath rather than risk STOMA COMPLICATIONS
haematoma formation or damaging a new anasto-
mosis by forcing it through a tight gap. Haemostasis There are a number of stoma-related complications.
should be checked before fascial closure. Complications related to temporary defunctioning
An alternative way of fashioning the anastomo- stomas may be managed conservatively or tolerated
sis is to use mechanical stapling instruments. Once until reversal, but patients who have a permanent
the two limbs of the bowel have been freed, as de- stoma may require corrective surgery.
scribed above, the two limbs of a linear stapler
are inserted into the lumen of the loops of bowel
and the device approximated but not closed. It is Stricture/Retraction
crucial at this point to check that the mesentery
of the bowel is not caught in the stapler, as firing Stricturing of the skin around a stomal orifice may
through this may lead to troublesome bleeding, be associated with a degree of retraction of the bowel.
with the risk of haematoma formation and bowel Minor separation of the mucocutaneous border is
ischaemia. The stapler can then be closed fully common and managed conservatively with careful as-
and a final check made that the mesentery is clear. sessment to ensure no collection or cellulitis develops.
After firing, the linear stapler device is opened Subsequent healing by secondary intention may lead
and removed, and a transverse stapler is applied to a degree of scarring and subsequent stricturing.
perpendicular to the first staple line to seal the Dilation of a minor stricture may be successful, but
opened bowel ends. A linear stapler for this part if this fails surgery may be warranted if the stoma is
of the technique is no longer recommended, as permanent and the stricture problematic. Often, this
anastomotic leakage from the cross-stapling line is straightforward and requires only incision of the
has been reported. The excess bowel can then be scarred area just away from the orifice, and a small
removed using a scalpel. Many surgeons recom- sliver of the scar tissue is excised. The bowel is then
mend re-enforcing the staple line with sutures and re-approximated to the skin using absorbable sutures.
also placing a separate stitch approximately 1 cm A more significant and early retraction may be
beyond the extent of the first staple line to help seen if there is too much tension on the bowel at the
relieve tension at this point, as this is an area re- time of stoma creation. Abscess formation or cellu-
ported as being prone to leakage. litis may occur in this instance, and a re-laparotomy
Once the bowel has been closed and reduced may be required. This is often a difficult operation,
into the abdomen, the abdominal wall is closed. with oedema and infection, resulting in friability of
The sheath is sutured with a strong monofila- the tissues.
ment nylon using either continuous or inter- For an ileostomy that gradually retracts and loses
rupted sutures. Antiseptic can then be poured its spout, a local revision may be possible. This in-
into the cavity, in an effort to reduce the risk of volves a circumstomal incision and gentle dissec-
infection. Interrupted sutures or skin clips can be tion of the layers between the bowel and the ab-
used to close the skin, allowing some of the clips dominal wall. Often, once freed circumferentially,
or sutures to be removed easily in the event of a the bowel can be advanced gently and the spout
superficial infection. resutured satisfactorily.

HEBK001-C15_p228-238.indd 233 08/02/13 5:29 PM


234  Intestinal stoma and the role of defunctioning a low anastomosis after anterior resection

Prolapse Developments in laparoscopic surgery have ex-


tended to laparoscopic parastomal hernia repair.
Prolapse is more common with a colostomy, par- Laparoscopic ports are introduced and positioned
ticularly loop colostomy. Local amputation of the well away from the stoma orifice, and intra-abdom-
redundant prolapse of an end colostomy with re- inal reduction of the parastomal contents allows
suture of the mucocutaneous junction is a straight- the placement of synthetic mesh around the bowel.
forward procedure that can be done under local Procedure-specific meshes have antiadhesion back-
anaesthetic in an unfit patient. Recurrent prolapse ing layers to minimize adhesion formation. The
is common, however. Loop stomas, particularly mesh can be positioned and fixed into place, and
loop colostomies, are more prone to prolapse (al- the ends overlapped to ensure complete encircle-
most always the distal limb) and are solved by sto- ment of the bowel where it passes through the ab-
ma closure, provided reversal is safe and feasible. If dominal wall.
reversal is not imminently possible, one option with The other option to repair a parastomal hernia
a prolapsing loop is to mobilize the bowel circum- is to perform a re-laparotomy, take down the origi-
ferentially and then staple off the distal limb, which nal stoma, and resite in a new premarked position.
is reduced into the peritoneal cavity. The proximal The defect at the original site can be closed with a
limb is then refashioned into an end stoma, after strong non-absorbable monofilament suture. Some
amputating any redundant length. surgeons insert a prophylactic biological mesh to
reduce the risk of longer-term hernia recurrence.

Herniation
EVIDENCE FOR FAECAL DIVERSION
Parastomal herniation is very common, but the AFTER TOTAL MESORECTAL
risk of strangulation is low. Patients with hernia- EXCISION
tion may have leaking bags caused by difficulty in
getting appliances to adhere and seal properly. Vari- Anastomotic leakage following TME is impossible
ous flanges and belts are available that may prevent to prevent completely. Predicting risk and manag-
this problem, but sometimes surgical intervention ing consequences therefore becomes the aim of the
is r­ equired. surgeon. Rates of leakage after anterior resection
Several options have been evaluated to repair vary between institutions, but a consistent predic-
these herniae, with variable success rates report- tor of an increased risk is the height of the anasto-
ed. All of them have a high incidence of hernia mosis from the anal verge.4 Colorectal anastomoses
recurrence, and patients should be consented below the peritoneal reflection, or coloanal anasto-
accordingly. Peristomal local mobilization, sac moses, have clinical leak rates reported in the range
herniotomy and closure of the sheath with non- 11–37.5 per cent.5 A meta-analysis of defunctioning
absorbable sutures is probably easiest but also stoma versus no stoma following rectal resection
prone to the highest failure rate. A better repair concluded that defunctioning resulted in a lower
may be achieved by making a medial incision leak rate.6 The meta-analysis included data from
away from the stoma site and approaching the sac four randomized controlled trials (RCTs)5,7–9 and 21
anterior to the abdominal wall musculature. The non-randomized studies, with 11 429 patients in to-
mucocutaneous junction is kept intact and the tal. Meta-analysis of the RCTs showed a lower clini-
stoma covered during this operation. Once the cal anastomotic leak rate and a lower reoperation
sac is reduced, the defect can be closed with non- rate in the stoma group. In the non-randomized
absorbable sutures and a synthetic mesh used to studies, a lower clinical anastomotic leak rate, lower
reinforce the area. Using synthetic mesh in this reoperation rate and lower mortality rate in the
way still carries a risk of infection, but newer al- stoma group was reported.
ternatives with collagen-based biological mesh The strength of available evidence in favour of fae-
may be helpful in this scenario. Biological meshes cal diversion with proximal ostomy raises the ques-
are expensive, however, and long-term follow-up tions of how best to divert the faecal stream, the likeli-
is not currently available. hood that a defunctioning stoma will be permanent,

HEBK001-C15_p228-238.indd 234 08/02/13 5:29 PM


EVIDENCE FOR FAECAL DIVERSION AFTER TOTAL MESORECTAL EXCISION  235

and the risks specifically associated with a defunction- with rectal cancer (459 in the radiotherapy group, 465
ing stoma and with stoma reversal. in the non-radiotherapy group) who underwent a low
anterior resection had a defunctioning stoma in the
prospective randomized TME trial involving a total of
Loop Colostomy or Ileostomy? 1530 patients. Creation of stomas and time to stoma
reversal were analysed retrospectively by use of mul-
A systematic review and meta-analysis of RCTs and tivariate analysis. In 523 of 924 patients (57 per cent),
observational studies comparing temporary loop a primary stoma (defined as a stoma created at the
ileostomy and loop colostomy for defunctioning time of TME) was constructed after a low anterior
of colorectal or coloanal anastomoses has reported resection. Loop ileostomy was formed in 329 patients
findings in support of loop ileostomy.10 Clinically and colostomy in 194 patients. Secondary stoma, de-
relevant events were grouped into four study out- fined as any stoma created during a second or follow-
comes: ing procedure after TME, was necessary in 93 of 401
patients (23 per cent). The main reasons for second-
l general outcome measures: dehydration, wound
ary stoma formation were anastomotic leakage (n 5
infection;
61), abscess, sepsis or peritonitis (n 5 18) and fistula
l construction of the stoma outcome measures:
(n 5 6). The total number of patients who therefore
parastomal hernia, stenosis, sepsis, prolapse,
received a temporary stoma either at initial surgery or
retraction, necrosis, haemorrhage;
at a secondary stage was 616 of 924 patients (67 per
l closure of the stoma outcome measures: anasto-
cent). For stomas that were closed, 97 per cent were
motic leak or fistula, wound infection, occlu-
done so within the first postoperative year.
sion, hernia;
Nineteen per cent of these defunctioning stomas
l functioning of the stoma outcome measures:
were never reversed during follow-up. Multivariate
occlusion, skin irritation.
analysis demonstrated that preoperative radiotherapy
Five randomized controlled trials and seven ob- was associated significantly with a decreased likeli-
servational studies were included in this analysis. hood of stoma reversal for secondary stomas, but not
Overall, the included studies reported on 1529 pa- for primary stomas. Older age, secondary stoma con-
tients, just over half of whom (58 per cent) under- struction, an end ileostomy or colostomy, any postop-
went defunctioning loop ileostomy. Loop ileostomy erative complication and recurrence were identified as
had a lower risk of prolapse and sepsis but was as- limiting factors for stoma reversal.
sociated with a higher risk of dehydration due to a The authors concluded that postoperative com-
high output and intestinal obstruction after stoma plications are an important limiting factor for
closure. There were no other significant differences. stoma reversal because, unsurprisingly, after occur-
Included in the analysis was a small randomized rence of these complications, patients and surgeons
trial from the authors’ institution, which reported may be reluctant to reverse a defunctioning stoma.
in favour of loop ileostomy versus loop colostomy The most important trial addressing the issue
after TME surgery, due to fewer abdominal wall of outcomes after TME for rectal cancer has been
problems (incisional hernia, wound infection) in a Swedish RCT multicentre trial, which reported
the ileostomy group.11 6-year follow-up data.7,13 In total, 234 of 821 pa-
tients (28 per cent) undergoing TME were random-
ly assigned to have either a defunctioning stoma
Frequency of Non-Reversal of (n 5 116) or no defunctioning stoma (n 5 118). All
Defunctioning Stoma patients randomized had a satisfactory anastomosis
with a negative air-test. Strict definition of leakage,
Although intended to be temporary, a substantial pro- including pelvic abscess, rectovaginal fistula and
portion of defunctioning stomas are never reversed late leaks, was adhered to. In the original analysis,
and in effect the loop stoma becomes permanent. The symptomatic anastomotic leakage was present in
Dutch Colorectal Cancer Group analysed data from 12 of 116 patients (10.3 per cent) with a defunc-
a TME trial in patients with rectal cancer to identify tioning stoma versus 33 of 118 patients (28.2 per
factors that limit stoma reversal.12 In total, 924 patients cent) without a defunctioning stoma (P , 0.001).7

HEBK001-C15_p228-238.indd 235 08/02/13 5:29 PM


236  Intestinal stoma and the role of defunctioning a low anastomosis after anterior resection

This significant reduction in leak rates was ob- non-closure of stoma, and complication rates fol-
served irrespective of gender. The requirement for lowing reversal have been retrospective reviews of a
emergency reoperation was also significantly high- single-institution experience, with variable results.
er in the group with no stoma (30 of 118 patients, Chun and colleagues reported 123 patients who
25.4 per cent) compared with the group with a underwent a planned temporary defunctioning
­defunctioning stoma (10 of 116 patients, 8 per cent) loop ileostomy performed at the time of a low rec-
(P , 0.001). Of great interest was that with meticu- tal anastomosis.14 The primary outcome measures
lous follow-up, 40 per cent of leaks were diagnosed were the ileostomy complication rate for the en-
after discharge from hospital at a median of 24 days tire spectrum of care, readmission and reoperation
(range 13–172 range). At a median follow-up of rates to treat ileostomy complications, and subse-
42 months postoperatively, more patients who quent closure rate. Of these patients, 64.2 per cent
started out with no stoma had a stoma (20 of 118 experienced some form of morbidity during their
patients v. 16 of 116 patients; P 5 not significant). care, the biggest group being patients readmitted
At 6-years’ follow-up, 1 patient had died and for dehydration following ileostomy formation
the total number of patients with stomas had risen (11.4 per cent). The ileostomy was closed in 76.4
from 36 to 45.13 The total group was analysed with per cent of patients, with 8.6 per cent requiring a
regard to the presence of a permanent stoma, the midline laparotomy (a non-closure rate of 23.6 per
type of stoma, the time point at which the stoma cent, which is higher than in the Dutch and Swedish
was constructed or considered as permanent, and groups). The overall ileostomy-related reoperation
the reasons for requiring a permanent stoma. rate was 10.4 per cent (2.4 per cent during index
During the study period, 45 of 233 patients (19 hospitalization, 1.6 per cent at readmission, and
per cent) were considered to have a permanent sto- 6.4 per cent following ileostomy closure). Obesity
ma: 25 had an end sigmoid colostomy and 20 a loop (body mass index $ 30 kg/m2) was associated with
­ileostomy. End colostomies were constructed at a a higher overall ileostomy complication rate and
median of 22 months after TME, predominantly for outpatient complication rate. Age over 65 years and
anastomotic leakage. Loop ileostomies, performed hypertension increased the risks of high ileostomy
either at initial surgery (n 5 12) or secondarily output and dehydration. Obesity and smoking de-
(n 5 8), were considered as permanent at a medi- creased the likelihood of ileostomy closure.
an of 12.5 months after the initial rectal resection. Akiyoshi and colleagues considered complica-
The reasons for a permanent loop ileostomy were tions of stoma closure to evaluate the risks and
metastatic disease, unsatisfactory anorectal func- benefits of a defunctioning stoma.15 Data were pro-
tion, deterioration in general medical condition, spectively recorded in 125 consecutive patients who
new non-colorectal cancer, patient’s refusal of fur- underwent an elective closure of loop ileostomy
ther surgery, and chronic constipation. The risk of after primary rectal cancer resection. Postopera-
having a permanent stoma in patients with symp- tive complications developed in 21 patients (16.8
tomatic anastomotic leakage was significantly high- per cent), the majority being wound infections;
er compared with patients without symptomatic ­ileus and anastomotic bleeding were also reported.
anastomotic leakage (56 per cent v. 11 per cent). There was no postoperative mortality. Risk factors
Both the Dutch TME study and the Swedish RCT for wound infection included male gender and sur-
demonstrate the importance of consent at the time gical site infection after primary surgery. The mean
of original TME with regard to the possible perma- length of postoperative hospital stay was signifi-
nence of a defunctioning stoma. cantly longer in patients with complications than in
patients without complications.
Luglio and colleagues reported the rate of com-
Risks and Complications of a plications after ileostomy reversal in 944 patients
Defunctioning Loop Ileostomy and its using standardized definitions and perioperative
Subsequent Reversal variables and 30-day outcomes.16 Anastomotic
technique for reversal varied (sutured fold-over,
Most reports on the morbidity of a defunctioning 49.4 per cent; stapled, 33.4 per cent; hand-sewn end
loop ileostomy, predictors of complications and to end, 17.3 per cent). Patients who had sutured

HEBK001-C15_p228-238.indd 236 08/02/13 5:29 PM


References  237

closure had longer operative times, increased times borne in mind. Attention to detail and awareness
to bowel movement and initiation of soft diet, and of the risks and benefits favours consideration of
longer time to discharge, but no difference in other routine defunctioning for all coloanal anastomoses
variables. Overall, complications occurred in 203 after TME surgery.
patients (21.5 per cent), including 45 patients (4.8
per cent) who experienced a major complication.
There was no post-reversal mortality. References
D’Haeninck and colleagues analysed 197 consec-
utive patients who underwent closure of a defunc-   1. Moran BJ, Heald RJ. Risk factors for and manage-
tioning loop ileostomy.17 Again, the method of clo- ment of anastomotic leakage in rectal surgery.
sure varied (transverse closure of enterotomy, 75.6 Colorectal Dis 2001; 3: 135–7.
  2. Cartmell MT, Jones OM, Moran BJ, Cecil TD. A
per cent; segmental enterectomy with hand-sewn
defunctioning stoma significantly prolongs the length
end-to-end anastomosis, 13.2 per cent; stapled side- of stay in laparoscopic colorectal resections. Surg
to-side anastomosis, 11.2 per cent). Overall postop- Endosc 2008; 22: 2643–7.
erative morbidity and mortality were 32.0 per cent   3. Amin AI, Ramalingam T, Sexton R, Heald RJ,
and 0.5 per cent, respectively. The surgical complica- Leppington-Clarke A, Moran BJ. Comparison of
tion rate was 30.5 per cent, including prolonged il- transanal stent with defunctioning stoma in low
eus (11.2 per cent), small bowel obstruction (4.1 per anterior resection for rectal cancer. Br J Surg 2003;
90: 581–2.
cent), anastomotic leak (3.0 per cent) and wound
  4. Matthiessen P, Hallböök O, Andersson M, Rutegård J,
infection (4.6 per cent). Surgical complications were Sjödahl R. Risk factors for anastomotic leakage after
more frequent in male patients. Prolonged ileus was anterior resection of the rectum. Colorectal Dis 2004;
more frequent when the interval to stoma reversal 6: 462–9.
exceeded 12 weeks. The incidence of complications   5. Ulrich AB, Seiler C, Rahbari N, Weitz J, Büchler MW.
was not influenced by the closure technique. Nine- Diverting stoma after low anterior resection: more
teen patients (9.6 per cent) required reoperation for ­arguments in favor. Dis Colon Rectum 2009; 52: 412–8.
  6. Tan WS, Tang CL, Shi L, Eu KW. Meta-analysis of de-
anastomotic leak, wound infection, small bowel ob-
functioning stomas in low anterior resection for rectal
struction or repair of an incisional hernia. cancer. Br J Surg 2009; 96: 462–72.
The complications following closure of a loop   7. Matthiessen P, Hallböök O, Rutegård J, Simert G,
ileostomy are more frequent in male patients Sjödahl R. Defunctioning stoma reduces symptomatic
who are obese. There seems to be no significant anastomotic leakage after low anterior resection of the
difference between whether the closure is sutured rectum for cancer: a randomized multicenter trial. Ann
or stapled. Although these risks should be borne Surg 2007; 246: 207–14.
  8. Pakkastie TE, Ovaska JT, Pekkala ES, Luukkonen PE,
in mind at the time of stoma construction, the
Järvinen HJ. A randomised study of colostomies in
risks are offset by the higher risks of anastomotic low colorectal anastomoses. Eur J Surg 1997; 163:
leakage following TME when not defunctioned, 929–33.
as documented in the Swedish study.7,13 Ideally,   9. Graffner H, Fredlund P, Olsson SA, Oscarson J,
informed consent for TME should include dis- Petersson BG. Protective colostomy in low anterior
cussions that specifically pertain to stoma-related resection of the rectum using the EEA stapling instru-
complications. ment: a randomized study. Dis Colon Rectum 1983;
26: 87–90.
10. Rondelli F, Reboldi P, Rulli A, et al. Loop ileostomy versus
loop colostomy for fecal diversion after colorectal or
CONCLUSION coloanal anastomosis: a meta-analysis. Int J Colorectal
Dis 2009; 24: 479–88.
All colorectal anastomoses are at risk of leakage; the 11. Edwards DP, Leppington-Clarke A, Sexton R, Heald
nearer the anastomosis to the anal verge, the higher RJ, Moran BJ. Stoma-related complications are more
the risk. A defunctioning stoma reduces the con- frequent after transverse colostomy than loop ileos-
tomy: a prospective randomized clinical trial. Br J Surg
sequences (and perhaps the incidence) of leakage
2001; 88: 360–3.
and reduces the requirement for emergency reop- 12. Den Dulk, Smit M, Peeters KC, et al. A multivari-
eration. Nevertheless, a stoma has complications at ate analysis of limiting factors for stoma reversal in
formation, in situ and at reversal, which should be patients with rectal cancer entered into the total

HEBK001-C15_p228-238.indd 237 08/02/13 5:29 PM


238  Intestinal stoma and the role of defunctioning a low anastomosis after anterior resection

mesorectal excision (TME) trial: a retrospective study. 15. Akiyoshi T, Fujimoto Y, Konishi T, et al. Complications
Lancet Oncol 2007; 8: 297–303. of loop ileostomy closure in patients with rectal tumor.
13. Lindgren R, Hallböök O, Rutegård J, Sjödahl R, Mat- World J Surg 2010; 34: 1937–42.
thiessen P. What is the risk for a permanent stoma 16. Luglio G, Pendlimari R, Holubar SD, Cima RR, Nelson
after low anterior resection of the rectum for cancer? H. Loop ileostomy reversal after colon and rectal
A six-year follow-up of a multicenter trial. Dis Colon surgery: a single institutional 5-year experience in 944
Rectum 2011; 54: 41–7. patients. Arch Surg 2011; 146: 1191–6.
14. Chun LJ, Haigh PI, Tam MS, Abbas MA. Defunctioning 17. D’Haeninck A, Wolthuis AM, Penninckx F, D’Hondt
loop ileostomy for pelvic anastomoses: predictors of M, D’Hoore A. Morbidity after closure of a defunc-
morbidity and nonclosure. Dis Colon Rectum 2012; 55: tioning loop ileostomy. Acta Chir Belg 2011; 111:
167–74. 136–41.

HEBK001-C15_p228-238.indd 238 08/02/13 5:29 PM


16
Quality of life in patients undergoing
abdominoperineal excision and anterior
resection for rectal cancer
Peter How and Kandiah Chandrakumaran

Introduction as an important outcome measure in its own right,


and there is evidence for a positive relationship be-
Since the first description of the abdominoperineal tween QOL data or some QOL measures and sur-
excision (APE) by W. Ernest Miles in 1908,1 the vival in cancer patients.3
surgical management of rectal cancer has evolved
dramatically. Total mesorectal excision (TME), de-
scribed by Heald and colleagues in 1982,2 has since Quality-of-life assessment:
been recognized as the gold standard treatment for general principles
most patients with rectal cancer. Other advances in
surgical technique, including stapling devices, and The term ‘quality of life’ intuitively evokes a sense
multimodality treatment, have enabled anterior re- of contentment that is influenced by economical,
section with sphincter preservation to become fea- mental and physical status. Since the World Health
sible for all but very distal rectal cancers. This has Organization’s (WHO) definition of health in 1948
significantly reduced the number of patients bur- as ‘a state of complete physical, mental and social
dened with a permanent stoma and the perceived wellbeing and not merely the absence of disease or
limitations it imposes. Much of the impetus for this infirmity’, health-related quality of life (HRQOL)
change was based on the belief that patients have has come to represent a multidimensional con-
a better quality of life (QOL) after sphincter pres- struct pertaining to four distinct areas of wellbeing:
ervation compared with patients with a permanent physical, psychological, emotional and social.
colostomy. This view is not supported universally, Attempts to measure HRQOL have focused
however, and remains a topic for debate. Over the largely upon these four domains with the use of
past decade, interest has grown in patient-reported specially designed questionnaires. These include,
outcomes. The cultural shift within health care to- among others, patient-derived generic and can-
wards a more patient-centred approach has meant cer-specific modules such as the European Or-
that assessment of treatment efficacy is no longer ganization for Research and Treatment of Cancer
confined to classic biomedical indicators such as (EORTC) Quality of Life Questionnaire – Core 30
survival and mortality. Quality of life has emerged (QLQ-C30) (Figure 16.1), EORTC QLQ-CR38,

HEBK001-C16_p239-248.indd 239 08/02/13 5:28 PM


240  QOL in patients undergoing APE and anterior resection

EORTC QLQ-C30 (version 3)

We are interested in some things about you and your health. Please answer all of the
questions yourself by circling the number that best applies to you. There are no "right" or
"wrong" answers. The information that you provide will remain strictly confidential.

Please fill in your initials:


Your birthdate (Day, Month, Year):
Today's date (Day, Month, Year): 31
___________________________________________________________________________
Not at A Quite Very
All Little a Bit Much
  1. Do you have any trouble doing strenuous activities, like 1 2 3 4
carrying a heavy shopping bag or a suitcase?
  2.  Do you have any trouble taking a long walk? 1 2 3 4
  3. Do you have any trouble taking a short walk outside of the 1 2 3 4
house?
  4.  Do you need to stay in bed or a chair during the day? 1 2 3 4
  5. Do you need help with eating, dressing, washing yourself or 1 2 3 4
using the toilet?

During the past week: Not at A Quite Very


All Little a Bit Much
  6. Were you limited in doing either your work or other daily 1 2 3 4
activities?
  7. Were you limited in pursuing your hobbies or other leisure 1 2 3 4
time activities?
  8.  Were you short of breath? 1 2 3 4
  9.  Have you had pain? 1 2 3 4
10.  Did you need to rest? 1 2 3 4
11.  Have you had trouble sleeping? 1 2 3 4
12.  Have you felt weak? 1 2 3 4
13.  Have you lacked appetite? 1 2 3 4
14.  Have you felt nauseated? 1 2 3 4
15.  Have you vomited? 1 2 3 4
Figure 16.1. The European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire –
Core 30 (QLQ-C30) questionnaire applicable to all cancers. Questions are grouped into functional scales, e.g. physical
function (questions 1–5), symptom scales, e.g. pain (questions 9 and 19), and single items, e.g. insomnia (question 11).
(Continues)

HEBK001-C16_p239-248.indd 240 08/02/13 5:28 PM


QUALITY-OF-LIFE ASSESSMENT: GENERAL PRINCIPLES  241

During the past week: Not at A Quite Very


All Little a Bit Much
16.  Have you been constipated? 1 2 3 4
17.  Have you had diarrhoea? 1 2 3 4
18.  Were you tired? 1 2 3 4
19.  Did pain interfere with your daily activities? 1 2 3 4
20. Have you had difficulty in concentrating on things, like 1 2 3 4
reading a newspaper or watching television?
21.  Did you feel tense? 1 2 3 4
22.  Did you worry? 1 2 3 4
23.  Did you feel irritable? 1 2 3 4
24.  Did you feel depressed? 1 2 3 4
25.  Have you had difficulty remembering things? 1 2 3 4
26. Has your physical condition or medical treatment interfered 1 2 3 4
with your family life?
27. Has your physical condition or medical treatment interfered 1 2 3 4
with your social activities?
28. Has your physical condition or medical treatment caused 1 2 3 4
you financial difficulties?

For the following questions please circle the number between 1 and 7 that best applies
to you
29.  How would you rate your overall health during the past week?
   1     2     3     4    5     6     7
Very poor              Excellent
30.  How would you rate your overall quality of life during the past week?
   1      2      3      4     5     6      7
Very poor              Excellent
Figure 16.1. (Continued).

EORTC QLQ-CR29, short form 36 (SF-36), SF- sufficient precision), sensitivity (ability to detect
12, Functional Assessment of Cancer Therapy – changes), reproducibility (cross-cultural), sim-
Colorectal (FACT-C) and Functional Assessment plicity (enabling self-administration), and easily
of Cancer Therapy – General (FACT-G). Despite interpretable (attaching clinical relevance). Fur-
variation in content and format, certain principles thermore, the questionnaires are intended to focus
and concepts remain central to all of these ques- on dimensions that are influenced by health and
tionnaires, such as validity (measuring what it is can be quantified accurately and appropriately, ac-
intended to measure), reliability (measuring with cording to its component indicators.4

HEBK001-C16_p239-248.indd 241 08/02/13 5:28 PM


242  QOL in patients undergoing APE and anterior resection

Figure 16.2. Quality of life after


surgery.
APE, abdominoperineal excision; LAR, low anterior resection.

Quality of life and the counselling. It is common practice, however, to first


preoperative pathway establish the patient’s understanding of what a sto-
ma is, followed by the introduction of a ‘starter pack’.
Although much of the literature regarding QOL in This pack contains basic anatomical and functional
patients with rectal cancer focuses on the effects of information regarding what to expect both before
surgery, most clinicians agree that appropriate sup- and after surgery. It also includes stoma bags and re-
port and counselling are vitally important from the lated accessories to help prepare the patient, and their
time of diagnosis. The preoperative (pre-cancer re- partner and family, for future daily maintenance.
section) patient pathway is increasingly a compli- Education of the patient’s relatives and carers is
cated one, often encompassing multimodality treat- beneficial and worthwhile. Up to half of patients
ment, restaging and, in some cases, surgery – all of (and their relatives) with an ileostomy and 30 per
which impact heavily upon QOL (Figure 16.2). Pre- cent of patients with a colostomy have expressed
operative baseline QOL scores have been highlight- dissatisfaction at the level of information they have
ed as prognostic indicators, and so it is important received.7 Furthermore, up to 60 per cent of pa-
to provide support at an early stage. This should in- tients are poorly informed about stoma irrigation
clude stoma counselling, sex counselling, and coun- techniques,8 despite good evidence for its effective-
selling about potential faecal incontinence. ness in achieving faecal continence in patients with
end colostomies.9 In addition, a greater proportion
Stoma Counselling of patients are unhappy with their degree of par-
ticipation within the decision-making process. It is
Often the most difficult and psychologically de- clear that although the majority of patients speak
manding hurdle to negotiate regarding rectal can- highly of their stoma counselling experiences, there
cer surgery is the concept of a permanent stoma. It is a need for continually assessing and modifying
is common to encounter patients for whom such this service, with the aim of improving general
an idea is untenable. Retrospective studies show standards of quality of care.
that patients who have access to stoma counselling
before surgery enjoy a better quality of life postop- Sex Counselling
eratively.5 In addition, accurate preoperative stoma
siting and counselling by a stomatherapist helps to Sexual dysfunction remains one of the more long-
improve outcome in such patients.6 lasting side effects of pelvic cancer treatment. Sexu-
Currently, as befits a patient-tailored approach, al problems are associated with both poor physical
there are no national guidelines pertaining to stoma health and emotional distress10 and are considered

HEBK001-C16_p239-248.indd 242 08/02/13 5:28 PM


QUALITY-OF-LIFE OUTCOMES FOLLOWING SURGERY FOR RECTAL CANCER  243

to be a major determinant of global QOL. Despite mote a more positive outlook on the future. Unfor-
the high risk of sexual dysfunction following rectal tunately, precise risks of dysfunction have not been
cancer surgery, sex counselling remains very much well documented, although research is ongoing.
on the periphery of mainstream care.
It has been suggested that an effective way to im-
plement appropriate sex counselling is to provide Quality-of-life outcomes
routine screening and counselling for QOL issues following surgery for
related to cancer treatment. It is proposed that one rectal cancer
person within the colorectal multidisciplinary team
should be assigned the role of assessing and triaging The primary objective in managing rectal cancer is
patients for quality of life problems.11 This could be achieving satisfactory oncological clearance while
conducted via a brief 20- to 30-min interview cen- maintaining QOL acceptable to the patient. Although
tred upon validated questionnaires, which may be much interest surrounds comparative QOL data be-
productive for clinical as well as research purposes. tween anterior resection and abdominoperineal ex-
Sex counselling should include educating and cision, it is pertinent that there never has been, and
warning patients about potential sexual problems now unlikely ever to be, a randomized controlled trial
arising from cancer treatment, helping patients of APE compared with low anterior resection. Thus,
overcome specific sexual problems (which may oc- comparative studies are difficult to interpret, and it is
cur before surgery as a result of stress or neoadju- beneficial to look at aspects of QOL in the individual
vant therapy), and preparing patients for physical procedures to determine how each impacts upon
changes that may adversely affect body image.12 QOL. It is also worth noting that patients treated
with APE will have lower, more advanced tumours,
and a higher percentage will receive neoadjuvant
Faecal Incontinence Counselling therapy,15 making direct comparisons with anterior
resection more complex. Anterior resection has long
For patients looking to avoid a permanent stoma, been associated with superior oncological outcomes
many pay the price in terms of poor functional compared with APE,16 and a similar assumption is
outcome, i.e. faecal incontinence. This is particu- often made regarding QOL. In the following section,
larly true for patients with mid- and lower rectal QOL outcomes following anterior resection and
tumours, where the closer the anastomosis to the APE with regards to function and symptomatology
anal canal, the poorer the functional outcome. are outlined. The results are interpreted to identify
Many patients prefer low anterior resection to APE, major determinants for good and poor outcomes in
even if low anterior resection involves a risk of in- order to optimize treatment policy.
continence on a daily basis.13 Faecal incontinence
rates have been reported to be as high as 61.5 per
cent at 5 years following preoperative radiotherapy Functioning Scores
and TME.14 The potential need for sanitary towels
and incontinence pads on a permanent basis com- Physical functioning has been shown to be better
bined with perianal excoriation is something that following anterior resection compared with APE
all patients undergoing restorative surgery should in comparative studies.8,17 These findings are in
be made aware of. The aim is not to discourage keeping with a meta-analysis of several compara-
patients before surgery but to provide realistic ex- tive studies, demonstrating statistically significantly
pectations of their likely postoperative quality of higher physical function within the anterior resec-
life. The possibility of long-term medication to tion group that also extends to patients with low
help control symptoms and the option of further rectal cancer.18 This is perhaps not surprising given
surgery as definitive treatment should also be made the physical limitations imposed by a permanent
clear to give the patient an appropriate perspective stoma on being able to perform day-to-day tasks
on future events. Knowing that there are options if independently.
problems are encountered can help pre-empt fear Good agreement is generally observed between
and negativity in the lead-up to surgery and pro- physical and role functions, and it is tempting to

HEBK001-C16_p239-248.indd 243 08/02/13 5:28 PM


244  QOL in patients undergoing APE and anterior resection

speculate that if patients undergoing APE are phys- ing specific stoma counselling), reflecting increased
ically limited by their stoma, then everyday work specialization within nursing services. The impor-
and leisure activities (role) may be affected adverse- tance of this is highlighted by the fact that stoma-
ly. Support for this theory comes from observations related complications often do not impact upon
that QOL scores improve upon stoma reversal.19 how patients perceive their quality of care com-
This is not always a true reflection of functional pared with the amount of information they receive,
status, however, as significant improvement in role participation in decision-making and opportunity
functioning has been reported despite severe diar- to discuss sexual matters.22 It would be reasonable
rhoea following stoma reversal.8 Indeed, despite to extrapolate this towards how they regard their
significant improvements in body image and leisure QOL.
activities, stoma reversal has not always resulted in Alternatively, equivalent results may be a result
improved QOL, while significant worsening of gas- of adaptation or response shift, whereby patients
trointestinal problems may persist for up to a year who have survived life-threatening disease appear
following surgery.20 to have new internal standards and are more willing
Interestingly, consistent data suggest equiva- to tolerate the adverse effects of either surgery, i.e.
lent8,21 or better emotional and cognitive function having a stoma or incontinence. Such a phenom-
in patients undergoing APE.18 Reasons for this enon may also account for comparable QOL scores
may include the definitive nature of the surgery, observed both pre- and postoperatively in previ-
whereby the patient feels that by removing both the ously reported and emerging studies.23
rectum and anus, there is less chance of recurrence Despite improvements in APE global QOL scores,
and no further invasive surveillance is required per it is notable that body image is consistently report-
rectum. Patients undergoing anterior resection, on ed to be higher in patients undergoing anterior
the other hand, remain troubled by the possibility resection, although this is not always statistically
of recurrence and further surgery. significant. Unlike functional scores, this aspect of
Although there is general agreement that fatigue, rectal cancer surgery appears resistant to any form
pain, nausea and dyspnoea are comparable between of counselling or psychotherapy and is probably a
anterior resection and APE, there is conflicting evi- contributing factor to the inferior sexual function
dence regarding gastrointestinal tract symptoma- observed in patients undergoing APE. Interestingly,
tology. Much of the earlier literature within the body image is significantly impaired in both sexes,
TME era that reported inferior global QOL scores although women demonstrate a relatively lower sex
in patients undergoing APE demonstrated higher drive preoperatively, while in the months following
levels of gastrointestinal dysfunction (constipation, surgery both men and women are affected negative-
diarrhoea) compared with patients undergoing low ly by a stoma.24 Such studies have called for greater
anterior resection, including defecation problems.8 attention to be paid to preoperative patient coun-
More recent studies, however, have demonstrated selling with regard to body image and sexuality.
no differences in either global QOL or gastrointes-
tinal dysfunction between the two groups,21 with
some reporting higher scores for gastrointestinal Sexual Dysfunction Following
dysfunction in the anterior resection group.17 Rectal Cancer Surgery
It is difficult to explain these differences given
that many of these studies employed similar meth- One critical aspect of TME, introduced by Heald
ods using identical questionnaires. As patient de- and colleagues, is the meticulous preservation of
mographics, tumour stage and follow-up time are the pelvic autonomic nerves, which has resulted in
well documented and broadly comparable, other a reduction in sexual dysfunction following rectal
factors should be considered. One reason may be cancer surgery. Sexual dysfunction continues to
due to the improved level and intensity of preop- be a major complication following anterior resec-
erative stoma counselling. Within the UK, it is now tion, and APE however, with rates varying between
common for multidisciplinary teams to have both 10 per cent and 60 per cent.25 This may be com-
colorectal nurse specialists (offering general sup- pounded further by neoadjuvant and adjuvant ra-
port and guidance) and stoma care nurses (offer- diotherapy.26

HEBK001-C16_p239-248.indd 244 08/02/13 5:28 PM


Quality-of-life outcomes following surgery for rectal cancer  245

There is widespread agreement within the lit- Neorectal reservoir


erature that APE carries a higher risk of sexual If restorative surgery is possible for low rectal tu-
dysfunction than anterior resection. One feature mours, many surgeons perform a circular transanal
of this is altered body image associated with double-stapled low colorectal or coloanal anasto-
a permanent colostomy, described previously. mosis, commonly without a pouch. Such patients
Other reasons accounting for this, particularly often end up having poor function, particularly
regarding male sexual dysfunction, are a higher with regard to continence due to impaired neorec-
proportion of T4 tumours, lower rectal tumours, tal function. Development of the colonic J-pouch
radiotherapy, comorbidity, and increased risk of anal anastomosis29,30 has been reported to reduce
nerve damage during surgery given the extent of stool frequency and defecation urgency.31 System-
the dissection and significant alteration in pelvic atic reviews and meta-analyses of randomized con-
anatomy.27 With respect to physiological sexual trolled trials have reported improved function with
dysfunction, it is perhaps best to focus on com- colonic J-pouch anal anastomosis compared with
bined genitourinary function as a reflection of a conventional straight coloanal anastomosis,32–35
autonomic nerve preservation. Enker and col- suggesting better QOL scores for colonic J-pouch
leagues demonstrated that 57 per cent of patients anal anastomosis.36
undergoing APE compared with 85 per cent of Despite good evidence for the J-pouch improv-
patients undergoing restorative surgery were able ing outcomes and QOL, the technique is not always
to maintain both urinary and sexual function.28 possible. For example, a very narrow pelvis, a fatty
Similar emerging data on sexual function from mesentery or restrictions in length may mean this
our own multicentre prospective study echo technique is impossible to perform in certain pa-
these sentiments, despite global QOL scores re- tients. Encouragingly, similar functional outcomes
maining broadly equivalent. have been observed in side-to-end anastomosis,37
which is technically easier to perform and is a useful
alternative when there is inadequate bowel length
Factors Affecting Functional Outcome
for a J-pouch.
Following Anterior Resection As an additional option, the technique of colo-
plasty has been developed with a similar aim of
Level of anastomosis
improving outcome and QOL. Coloplasty follows
Functional outcome is thought to be worse the the principles of pyloroplasty in that a longitudi-
lower the anastomosis because of smaller reservoir nal incision is closed transversely and the recon-
volume. It has been demonstrated that lower anas- struction is completed by a stapled anastomosis or
tomotic level tends towards increased frequency of a hand-sewn end-to-end pouch anal anastomosis.
incontinence for solid stool and gas.21 These find- Quality-of-life studies based on SF-36 have demon-
ings correlate well with the demonstration of more strated better scores for coloplasty compared with
frequent or painful bowel movements in patients straight anastomosis, including fewer bowel move-
undergoing low anterior resection.8 Interestingly, a ments and less antidiarrhoeal medication,38 and
higher composite score for incontinence is not al- equivalent functional outcome to colonic J-pouch
ways reflected in QOL scores between patients with anal anastomosis and side-to-end anastomosis.39,40
high and low anastomosis. This may be a reflection
of the response shift phenomenon described earlier, Radiotherapy
whereby patients undergoing low anterior resection There is no doubt that the systemic effects of ra-
have lower expectations of their outcome compared diotherapy have a significant detrimental effect on
with patients undergoing high anterior resection. It QOL. Prospective studies using EORTC modules
is not always correct, therefore, to assume that gas- assessing the effects of neoadjuvant radiotherapy
trointestinal symptomatology, depending on the have shown significantly increased scores for diar-
level of the anastomosis, will affect QOL, although rhoea, fatigue and appetite loss and worse outcomes
this does remain a possible reason for the observed for physical function, social function and global
variability between studies comparing patients with QOL.41 As QOL scores tend to return to pretreat-
and without a stoma. ment levels 4–6 weeks after radiotherapy, however,

HEBK001-C16_p239-248.indd 245 08/02/13 5:28 PM


246  QOL in patients undergoing APE and anterior resection

it is perhaps more important to focus upon long- function47 and that patients with low rectal cancer
term local effects. have worse QOL than patients with upper or mid-
Radiotherapy, by way of fibrosis, is purported to dle rectal cancers, further information on surgical
reduce compliance of the rectum, resulting in re- options and outcomes in low rectal cancer would
duced reservoir function. It is also thought that ra- be helpful. Ideally, good-quality prospectively col-
diotherapy-induced fibrosis affecting the myenteric lected QOL data using established questionnaires
plexus prevents adequate closure of the anal canal would represent progress on this front, and this is
in its resting state. currently in progress. There is an increasing interest
Preoperative radiotherapy, whether long or short in comparative QOL for APE and anterior resection
course, has been associated with higher levels of fre- for low rectal cancer, with several studies reporting
quency and urgency42 and faecal incontinence14,43,44 interesting conclusions. Among these are better
when assessed postoperatively in randomized con- sexual functioning scores observed in patients un-
trolled trial settings. A systematic review outlined dergoing APE (both genders) and worse scores for
similar late adverse effects related to radiotherapy gastrointestinal symptoms in patients undergoing
in the treatment of rectal cancer.45 anterior resection.17 This has not impacted on the
In addition, daily urinary incontinence has been general trend of equivalent overall QOL outcomes,
reported more frequently after radiotherapy, with a however (albeit with certain specific differences),
similar adverse effect on social function.43 even within larger studies.48 More recent studies re-
The benefits of radiotherapy in reducing local re- porting inferior QOL in patients with permanent
currence have to be balanced against the adverse ef- stomas remain fewer and are generally hampered
fects of radiotherapy on bowel function and QOL.46 by smaller sample sizes.49 Although it is difficult to
The most effective strategy to help improve this as- draw firm conclusions in the face of such contradic-
pect of patient care would appear to lie in careful tory evidence, there are variations between reported
patient selection, so that substantial overtreatment studies including, in some, a lack of preoperative
is avoided. QOL data, single institution involvement and re-
sponse bias. In addition, length of follow-up is im-
portant given that patients undergoing restorative
Low rectal cancer surgery surgery commonly suffer from anterior resection
and quality of life: the heart syndrome (frequency, urgency, stool fragmentation,
of the matter incontinence), which may persist beyond 1 year.50
Furthermore, there is often little or no information
Given that patients with high and mid-rectal tu- regarding the surgical technique (e.g. level and type
mours routinely undergo restorative surgery (ex- of anastomosis, presence of pouch) or the level of
cept for patients with poor preoperative function), preoperative counselling, factors known to influ-
it is patients with distal rectal cancer (commonly ence outcome.
defined as within 6 cm of the anal verge) who
present the greatest challenge. This includes the
technical challenges of low rectal cancer and the Conclusion
difficult decision as to whether to perform ante-
rior resection or APE. Increasingly, it is oncologi- Interest in QOL outcomes has grown significantly
cally feasible to perform either operation; provided over recent years and is likely to continue, in keep-
the patient has been continent before the onset of ing with growing importance placed on patient-
the cancer and has no strong objections to a per- reported outcome measures. The remit of the sur-
manent stoma, the surgeon must decide which geon is no longer only to remove cancer, but also to
operation will give the best outcome. Techniques ensure the best possible outcome in keeping with
such as anal manometry have been suggested as the patient’s wishes and beliefs. This has important
reliable predictors of functional outcome, but evi- implications regarding the selection of patients
dence for this is inconclusive. Given that low rec- for multimodality treatment and technical factors
tal cancers are associated with more postoperative relating to surgical technique. Although every pa-
complications and poorer anorectal and sexual tient should be considered on an individual basis,

HEBK001-C16_p239-248.indd 246 08/02/13 5:28 PM


REFERENCES  247

i­ nformation, advice and counselling must be cen- higher involved margin rates compared with patients
tred on a strong collective evidence base. Long-term treated by anterior resection. Dis Colon Rectum
2010; 53: 53–6.
QOL prospective studies using internationally vali-
16. Heald RJ, Smedh RK, et al. Abdominoperineal
dated questionnaires currently represent the most excision of the rectum: an endangered operation.
assured way of achieving this. Dis Colon Rectum 1997; 40: 747–51.
17. Kasparek MS, Hassan I, Cima RR, Larson DR,
Gullerud RE, Wolff BG. Quality of life after coloanal
References anastomosis and abdominoperineal resection for distal
rectal cancers: sphincter preservation vs quality of life.
  1. Miles WE. A method of performing abdomino-perineal Colorectal Dis 2011; 13: 872–7.
excision for carcinoma of the rectum and the terminal 18. Cornish JA, Tilney HS, Heriot AG, Lavery IC,
portion of the pelvic colon. Lancet 1908; 2: 1812–3. Fazio VW, Tekkis PP. A meta-analysis of quality of
  2. Heald RJ, Husband EM, Ryall RD. The mesorectum in life for abdominoperineal excision of rectum versus
rectal cancer surgery: the clue to pelvic recurrence? Br anterior resection for rectal cancer. Ann Surg Oncol
J Surgery 1982; 69: 613–6. 2007; 14: 2056–68.
  3. Montazeri A. Quality of life data as prognostic indicators 19. Camilleri-Brennan J, Steele RJ. Prospective analysis
of survival in cancer patients: an overview of the litera- of quality of life after reversal of a defunctioning loop
ture from 1982 to 2008. Health Qual Life Outcomes ileostomy. Colorectal Dis 2002; 4: 167–71.
2009; 7: 102. 20. Siassi M, Hohenberger W, Lösel F, Weiss M. Quality
  4. Lam CLK. Subjective quality of life measures: general of life and patient’s expectations after closure of a tem-
principles and concepts. In Preedy VR (ed.). Hand- porary stoma. Int J Colorectal Dis 2008; 23: 1207–12.
book of Disease Burdens and Quality of Life Measures 21. Guren MG, Eriksen MT, Wiig JN, et al. Quality of
1. Berlin, Springer-Verlag, 2009: 381–9. life and functional outcome following anterior or
  5. Bass EM, Del Pino A, Tan A, Pearl RK, Orsay CP, abdominoperineal resection for rectal cancer. Eur J
Abcarian H. Does preoperative stoma marking Surg Oncol 2005; 31: 735–42.
and education by the enterostomal therapist affect 22. Persson E, Gustavsson B, Hellström AL, Lappas G,
outcome? Dis Colon Rectum 1997; 40: 440–2. Hultén L. Ostomy patients’ perceptions of quality of
  6. Crooks S. Foresight that leads to improved outcome: care. J Adv Nurs 2005; 49: 51–8.
stoma care nurses’ role in siting stomas. Prof Nurse 23. How P, Stelzner S, Branagan G, et al. Comparative
1994; 10: 89–92. quality of life in patients following low anterior resection
  7. Persson E, Gustavsson B, Hellström AL, Fridstedt and abdominoperineal excision for low rectal cancer:
G, Lappas G, Hultén L. Information to the relatives of preliminary results from a prospective multicentre
people with ostomies: is it satisfactory and adequate? study. Dis Colon Rectum 2010; 55: 400–6.
J Wound Ostomy Continence Nurs 2005; 32: 238–45. 24. Burghofer K, Jauch KW. [Sexuality and body image in
  8. Engel J, Kerr J, Schlesinger-Raab A, Eckel R, Sauer H, patients with rectal cancer]. MMW Fortschr Med 2006;
Hölzel D. Quality of life in rectal cancer patients: a four- 148: 36–7.
year prospective study. Ann Surg 2003; 238: 203–13. 25. Banerjee AK. Sexual dysfunction after surgery for
  9. Karadag A, Mentes BB, Ayaz S. Colostomy irrigation: rectal cancer. Lancet 1999; 353: 1900–2.
results of 25 cases with particular reference to quality 26. Mannaerts GH, Schijven MP, Hendrikx A, Martijn H,
of life. J Clin Nurs 2005; 14: 479–85. Rutten HJ, Wiggers T. Urologic and sexual morbidity
10. Laumann EO, Paik A, Rosen RC. Sexual dysfunction following multimodality treatment for locally advanced
in the United States: prevalence and predictors. J Am primary and locally recurrent rectal cancer. Eur J Surg
Med Assoc 1999; 281: 537–44. Oncol 2001; 27: 265–72.
11. Schover LR. Sexuality and fertility after cancer. Hema- 27. Hassan I, Cima RR. Quality of life after rectal resection
tology Am Soc Hematol Educ Program 2005; 523–7. and multimodality therapy. J Surg Oncol 2007; 96:
12. Schover LR. Sexual rehabilitation after treatment for 684–92.
prostate cancer. Cancer 1993; 71(3 suppl.): 1024–30. 28. Enker WE, Havenga K, Polyak T, Thaler H, Cranor M.
13. Bossema E, Stiggelbout A, Baas-Thijssen M, van de Abdominoperineal resection via total mesorectal exci-
Velde C, Marijnen C. Patients’ preferences for low rec- sion and autonomic nerve preservation for low rectal
tal cancer surgery. Eur J Surg Oncol 2008; 34: 42–8. cancer. World J Surg 1997; 21: 715–20.
14. Lange MM, den Dulk M, et al. Risk factors for faecal 29. Parc R, Berger A, Tiret E, Frileux P, Nordlinger B, Han-
incontinence after rectal cancer treatment. Br J Surg noun L. [Colo-anal anastomosis with reservoir in the
2007; 94: 1278–84. treatment of rectal cancer]. Ann Gastroenterol Hepatol
15. Shihab OC, Brown G, Daniels IR, Heald RJ, Quirke P, (Paris) 1987; 23: 329–31.
Moran BJ. Patients with low rectal cancer treated by 30. Lazorthes F, Fages P, Chiotasso P, Lemozy J, Bloom
abdominoperineal excision have worse tumors and E. Resection of the rectum with construction of a

HEBK001-C16_p239-248.indd 247 08/02/13 5:28 PM


248  QOL in patients undergoing APE and anterior resection

colonic reservoir and colo-anal anastomosis for carci- after anterior resection for rectal cancer. Colorectal Dis
noma of the rectum. Br J Surg 1986; 73: 136–8. 12: 624–31.
31. Hallböök O, Hass U, Wänström A, Sjödahl R. Quality 41. Guren MG, Dueland S, Skovlund E, Fossa SD,
of life measurement after rectal excision for cancer: Poulsen JP, Tveit KM. Quality of life during radiothera-
comparison between straight and colonic J-pouch py for rectal cancer. Eur J Cancer 2003; 39: 587–94.
anastomosis. Scand J Gastroenterol 1997; 32: 42. Parc Y, Zutshi M, Zalinski S, Ruppert R, Fürst A, Fazio
490–3. VW. Preoperative radiotherapy is associated with
32. Brown CJ, Fenech DS, McLeod RS. Reconstructive worse functional results after coloanal anastomosis for
techniques after rectal resection for rectal cancer. rectal cancer. Dis Colon Rectum 2009; 52: 2004–14.
Cochrane Database Syst Rev 2008; (2): CD006040. 43. Bruheim K, Guren MG, Skovlund E, et al. Late
33. Koh PK, Tang CL, Eu KW, Samuel M, Chan E. A side effects and quality of life after radiotherapy for
systematic review of the function and complications of rectal cancer. Int J Radiat Oncol Biol Phys 2010;
colonic pouches. Int J Colorectal Dis 2007; 22: 543–8. 76: 1005–11.
34. Heriot AG, Tekkis PP, Constantinides V, et al. Meta- 44. Murata A, Brown CJ, Raval M, Phang PT. Impact of
analysis of colonic reservoirs versus straight coloanal short-course radiotherapy and low anterior resection
anastomosis after anterior resection. Br J Surg 2006; on quality of life and bowel function in primary rectal
93: 19–32. cancer. Am J Surg 2008; 195: 611–5.
35. Peng J, Zhan W, Zhao X, et al. [Comparison of colonic 45. Birgisson H, Påhlman L, Gunnarsson U, Glimelius
J-pouch and straight coloanal anastomosis after low B. Late adverse effects of radiation therapy for rectal
anterior resection for rectal carcinoma: a meta-analysis cancer: a systematic overview. Acta Oncol 2007; 46:
of 8 randomized trials]. Zhonghua Wai Ke Za Zhi 2002; 504–16.
40: 905–8. 46. Sebag-Montefiore D, Stephens RJ, Steele R, et al.
36. Gosselink MP, Busschbach JJ, Dijkhuis CM, Stassen Preoperative radiotherapy versus selective postopera-
LP, Hop WC, Schouten WR. Quality of life after total tive chemoradiotherapy in patients with rectal cancer
mesorectal excision for rectal cancer. Colorectal Dis (MRC CR07 and NCIC-CTG C016): a multicentre,
2006; 8: 15–22. randomized trial. Lancet 2009; 373: 811–20.
37. Machado M, Nygren J, Goldman S, Ljungqvist O. 47. Hoerske C, Weber K, Goehl J, Hohenberger W, Merkel
Functional and physiologic assessment of the colonic S. Long-term outcomes and quality of life after rectal
reservoir or side-to-end anastomosis after low anterior carcinoma surgery. Br J Surg 2010; 97: 1295–303.
resection for rectal cancer: a two-year follow-up. Dis 48. Tilney H, Fazio V, Lavery I, et al. Comparison of long-
Colon Rectum 2005; 48: 29–36. term survival and quality of life following restorative
38. Remzi FH, Fazio VW, Gorgun E, et al. Quality of life, versus non-restorative surgery for lower third rectal
functional outcome, and complications of coloplasty cancer. Dis Colon Rectum 2007; 50: 703–4.
pouch after low anterior resection. Dis Colon Rectum 49. Fucini C, Gattai R, Urena C, Bandettini L, Elbetti C.
2005; 48: 735–43. Quality of life among five-year survivors after treat-
39. Matsuoka H, Masaki T, Kobayashi T, Sato K, Sugiya- ment for very low rectal cancer with or without a
ma M, Atomi Y. Comparison of functional and clinical permanent abdominal stoma. Ann Surg Oncol 2008;
outcomes: colonic J-pouch vs. coloplasty in patients 15: 1099–106.
with low rectal cancer. Hepatogastroenterology 2010; 50. Kakodkar R, Gupta S, Nundy S. Low anterior resec-
57: 70–72. tion with total mesorectal excision for rectal cancer:
40. Liao C, Gao F, Cao Y, Tan A, Li X, Wu D. Meta-analysis functional assessment and factors affecting outcome.
of the colon J-pouch vs transverse coloplasty pouch Colorectal Dis 2006; 8: 650–56.

HEBK001-C16_p239-248.indd 248 08/02/13 5:28 PM


HEBK001-Index.indd 268 08/02/13 5:22 PM

You might also like