You are on page 1of 31

Rectal Carcinoma: Imaging for

Staging 21
Mit Dattani and Gina Brown

Introduction more, as the absolute TNM stage. Moreover,


additional prognostic determinants such as
In the modern day multimodality management of extramural vascular invasion, which were until
rectal cancer, a growing number of treatment now only evaluable in the post-resection speci-
options are available, the choice of which is largely men, are being diagnosed on pre-operative imag-
determined by tumour related factors. An accurate ing. In this regard, advances in imaging
and reproducible staging system is therefore technology, largely driven by magnetic reso-
invaluable for rectal cancer multidisciplinary nance imaging (MRI) over the last two decades,
teams, which rely increasingly on radiological have enabled the selection of appropriate treat-
prognostication to individualise treatment plans. ment strategies, which optimise the balance
This information is equally important for between oncological and functional outcome.
counselling patients, who will need to make More recently, imaging has found a novel role in
informed decisions by weighing the oncological assessing the response after neo-adjuvant treat-
merits of a radical resection, against the impact on ment, which in selected cases mitigates the need
quality of life ensuing from the multimodality for surgery with the detection of a complete
management of their cancer. response following (chemo)radiotherapy. The
Indeed, pre-operative rectal cancer assess- current imaging modalities available to stage
ment has evolved beyond the determination of rectal cancer include endorectal ultrasound scan
conventional prognostic features such as depth (ERUS), MRI, computed tomography (CT) and
of tumour invasion, nodal involvement and positron emission tomography (PET). These
metastatic spread. Tumour threatened resection techniques have their own strengths and limita-
margins which require neo-adjuvant treatment, tions, and often play a complementary role in
or involved anal sphincters for which extensive what is a challenging aspect of rectal cancer
surgery offers the best chance of cure are just as management. This chapter summarises the evi-
important in guiding treatment decisions, if not dence base and the utility of the different imag-
ing modalities that underpin the current standards
M. Dattani of care in rectal cancer management.
Pelican Cancer Foundation,
Basingstoke, Hampshire, UK
G. Brown (*)
The Royal Marsden NHS Foundation Trust and
Imperial College of London, London, UK
e-mail: gina.brown@rmh.nhs.uk

© Springer International Publishing AG, part of Springer Nature 2019 359


D. E. Beck et al. (eds.), Fundamentals of Anorectal Surgery,
https://doi.org/10.1007/978-3-319-65966-4_21
360 M. Dattani and G. Brown

Imaging Modalities a colorectal surgeon in a clinic setting with the


consent of the patient. Sedation is seldom required,
Endorectal Ultrasound although it is usually necessary to give the patient
an enema to avoid image distortion because faeces
History and Technique in the rectum. The patient is normally positioned in
The clinical application for ultrasound was first the left lateral decubitus position, following which
developed by Wild and Reid in 1952, who used a a digital rectal examination and a proctosigmoid-
linear B-mode transducer in the diagnosis of breast oscopy is performed to clinically assess the size,
lesions, “with a view to future applications at other fixity and position of the lesion being investigated.
sites” [1]. But it was not until 1983 that an advanced Any rectal residue should be suctioned to provide
version of their ‘echoendo probe’ was utilised by an optimal image. A rigid or flexible endorectal
Dragsted and Gammelgaard to evaluate the depth probe is then inserted into the rectal cavity. The tip
of invasion in rectal cancer. They assessed 13 has a rotating transducer covered by a balloon,
patients with rectal cancer using a 4.5 MHz rotat- which is filled with a variable volume of degassed
ing transducer probe, and correctly identified the water after insertion. This ensures coupling
depth of mural infiltration in 11 of the patients between the transducer and the rectal wall without
when compared to subsequent histopathology [2]. any acoustic interference from air within the rec-
In the two remaining patients, the technique was tum. Imaging is normally carried out starting from
limited owing to the presence of a stricturing tumor, the proximal end, gradually withdrawing the probe
which the probe could not access. These initial distally whilst meticulously assessing the preser-
findings were confirmed in 1985 by Hilderbrandt vation or loss of the normal anatomical layers of
and Feifel in a slightly larger series comprising 25 the rectal wall. The adjacent perirectal tissue is
patients [3]. More importantly, they objectively also scanned to evaluate any potentially involved
defined the reporting criteria for depth of rectal mesorectal lymph nodes. Real time images are
wall invasion based on the TNM classification sys- acquired and transmitted to a screen for image cap-
tem, which is still universally used. Denoted by the ture and interpretation in a clockwise manner.
prefix ‘u’ to indicate evaluation by endorectal ultra- A range of probes with varying frequencies are
sound (ERUS), the depth of tumour invasion is available, depending on the intended aim of
staged according to the level of disruption of the ERUS and bearing in mind that the focal length is
five distinct anatomical layers of the rectal wall inversely related to the frequency. As an example,
(Fig. 21.1a, b) by the tumour, which appears as a a high frequency probe will give higher resolution
hypoechoic irregular lesion. Beynon et al. [4] first images, but at a lower depth of penetration. This
proposed the five layer model following their care- may be useful in assessing the depth of invasion,
ful study comparing ultrasonographic appearances whereas a lower frequency may be more relevant
of the rectal wall with that of sequentially dissected in imaging the deeper perirectal tissues, albeit at
out histological layers in the normal rectum. The the expense of image resolution. Frequencies of
differential acoustic impedance across the rectal between 7 and 10 MHz will provide a focal length
wall gives the appearance of a concentric ring of of 1–5 cm and are usually sufficient for the pur-
alternating hyper- and hypoechoic layers as shown pose of staging rectal neoplasms.
in Fig. 21.1c, and described below:
Mucosa: Hyperechoic layer  rimary Rectal Cancer Staging:
P
Muscularis mucosae: Hypoechoic layer The Role of ERUS
Submucosa: Hyperechoic layer
Muscularis propria: Hypoechoic layer Depth of Invasion
Serosa/perirectal fat: Hyperechoic layer The precise assessment of tumour depth into the
Despite vast advances in the ERUS equipment rectal wall is of particular importance, given the
over the years, the basic principles of imaging possibility of a local transanal excision in selected
remain unchanged. The procedure is not labour cases of early rectal cancer, thus avoiding major
intensive, and is simple enough to be performed by resectional surgery and its associated morbidity.
21 Rectal Carcinoma: Imaging for Staging 361

a b

uT0 - tumor confined to the mucosa

1 uT1 - tumor confined to the submucosa

2 uT2 - tumor invades the muscularis


propria but does not penetrate through it
3

4 uT3 - tumor invades the perirectal tissue

5 uT4 - tumor invades an adjacent organ

Fig. 21.1 (a) uT1 lesion. The middle white hyperechoic layer. However, the outermost white layer which is indica-
layer between the white arrows has been disrupted by the tive of the perirectal fat is still preserved, demonstrating that
bulging tumour, indicating invasion into the submucosa. the tumour is still confined to the bowel wall. (c) The five-
However, the outer black hypoechoic line indicating layer rectal wall model of an ERUS and the corresponding
the muscularis propria is preserved. (b) uT1 lesion. depth of invasion (uT stage) as determined by degree of
Submucosal invasion by the tumour (white arrow) is indi- preservation of the different anatomical layers. Images
cated by the loss of the hypoechoic muscularis propria courtesy of Dr. A Corr, St. Mark’s Hospital, London. U.K.

Conversely, a locally advanced and unresectable There is a wide variation in the accuracy of
tumour may benefit from neo-adjuvant treatment, ERUS predicted T-stage, with estimates ranging
rendering it operable and improving the chances between 63% and 96% [5], and a reported mean of
of a curative resection following sufficient down- approximately 85% [6]. A meta-analysis of 42
sizing. These decisions depend on the accuracy studies with over 5000 patients who underwent
of ERUS, or other imaging modalities, at deter- ERUS for rectal cancer staging between 1984 and
mining the depth of rectal wall invasion, or ‘T’ 2006 found a pooled sensitivity of 81–96% and
stage. As discussed previously, ERUS evaluation specificity of 91–98% for evaluating the T stage
of T-stage is reported according to a modified when compared to histopathology [7]. Whilst these
TNM classification system (see Table 19.2) and results are certainly impressive, and comparably
denoted by the prefix ‘u’ to reflect the imaging better than the reported accuracy of CT and MRI in
modality. assessing the depth of mural invasion [8], caution is
362 M. Dattani and G. Brown

warranted in the absolute interpretation of pooled contend with the toxicity of neo-adjuvant treat-
analyses of heterogeneous studies. Several studies ment, particularly in countries where this is stan-
included in the meta-analysis were single centre dard practice for a T3N0 staged rectal cancer with
assessments, often with a small sample size, which no other adverse features [14]. Conversely, an
was less than 30 patients in some cohorts, or a understaged T2 cancer may be offered a local exci-
selection bias tending towards the inclusion of early sion treatment in selective cases, with the prospect
and late stage disease in which the risks of inaccu- of having to undergo a potentially difficult salvage
rate staging are low. Indeed, a common finding of procedure subsequent to histological analysis.
ERUS is the low accuracy in T2 rectal cancer stag- Harewood [6] assessed the accuracy of ERUS
ing, with frequent overstaging cited in several series staging for rectal cancer in his review of 41 pub-
[9]. The main reason postulated is the difficulty in lications in the English literature, and made two
accurately distinguishing between a T2 tumour that important observations worthy of discussion.
invades deep into the muscularis propria, from an Firstly, that the accuracy of ERUS for evaluating
early T3 with microscopic infiltration of the peri- the depth of mural invasion in rectal cancer was
rectal fat [5]. This is a consequence of the inability inversely proportional to the sample size of the
to differentiate peritumoural desmoplastic reaction study. He suggested that publication bias of pre-
from genuine neoplastic invasion in the perirectal viously reported smaller studies may have over
tissues with ERUS [10]. It is argued that the preop- inflated the accuracy of ERUS as the imaging
erative misclassification of a T2 rectal cancer bears modality of choice. In this context, his second
little clinical consequence [11, 12], and whilst this observation was the decline in ERUS accuracy
is true for outcomes of T2 and ‘early’ T3 rectal can- for rectal cancer T-staging in the more recently
cers that undergo primary resectional surgery [13], published data, which is shown in Table 21.1
the risks of overstaging could expose patients to [15–24] for studies with over 100 patients.

Table 21.1 Accuracy of ERUS in determining the depth of rectal wall invasion
T staging
Study Year Patient recruitment Setting No. of patients accuracy (%)
Akasu et al. [15] 2000 1991–1996 Single-centre, Japan 309 80
Garcia-Aguilar 2002 Data not given Single-centre, U.S.A 545 69a
et al. [16]
Marusch et al. [17]b 2002 1999 Multi-centre, Germany 422 63
(n = 49)
Mackay et al. [11] 2003 1991–2001 Multi-centre, Australia 356 77
(n = 2)
Manger et al. [18] 2004 1994–2002 Single-centre, Germany 357 77
Kauer et al. [19] 2004 1990–2000 Single-centre, Germany 458 69
Zammit et al. [20] 2005 1998– Single-centre, U.K 117 76
Ptok et al. [21] 2006 2000–2003 Multi-centre, Germany 3501 66
(n = 331)
Badger et al. [22]c 2007 1999–2004 Single-centre, U.K 131 72
Goertz et al. [23]c 2008 1990–2003 Single-centre, Germany 333 71
Morris et al. [24]c 2011 1999–2007 Single-centre, Australia 233 82
Marusch et al. [25] 2011 2000–2008 Multi-centre, Germany 7096 65
(n = 384)
Ashraf et al. [26]b 2012 1992–2008 Multi-centre, U.K (n = 21) 165 55d
Restivo et al. [27] 2015 1997–2012 Single-centre (Italy) 220 65
a
uT0-T4 tumours; uT1-T4 in all other studies
b
No information on neo-adjuvant treatment provided. In all other studies, patient who had neo-adjuvant treatment
were excluded
c
Some patient had short course neo-adjuvant radiotherapy with immediate surgery
d
uT0-T3 tumours only
21 Rectal Carcinoma: Imaging for Staging 363

A notable pattern in Table 21.1 is the rela- less than 5 mm contained metastases, and whilst
tively lower accuracy of ERUS in multi-centre reducing the size threshold to 3mm improved the
studies, apart from Mackay et al. [11], in which overall accuracy of predicting metastatic nodes,
the staging was carried out by two surgeons this significantly compromised the specificity
across two hospital sites. The inference is that with the inherent risks of over-treatment in this
widespread adoption of ERUS outside expert group [15].
centres can compromise its reliability as an The addition of secondary characteristics,
accurate staging modality. Marusch et al. [25] such as echogenicity, border features and con-
found that the accuracy of T-staging dropped tour, to dimensional assessment of lymph nodes
from 73% at centres performing more than 30 may increase the discriminatory power of
ERUS per year to 63% at centres with less than ERUS in evaluating metastatic nodes. Thus, a
10 cases per year, thus advocating for a centralised hypoechoic round node with a smooth delineated
service provided by experienced operators. border is more likely to be metastatic lymph node
Indeed, in a large study from Minnesota [16], the than an inflammatory one, although even this
pathological T-stage of the tumour and ERUS combination of additive features has been shown
operator were the only two independent factors to give conflicting results [22, 31, 32]. One way
affecting the accuracy of staging. Moreover, of ameliorating these issues, particularly in
ERUS has been plagued with high inter-observer equivocal cases, has been the suggestion of fine
variability, which although in some cases may be needle aspiration (FNA) for cytology sampling of
secondary to procedural aspects, it is more mesorectal lymph nodes [31]. Whilst technically
commonly a consequence of operator experience feasible and safe to perform, ERUS guided FNA
[19]. In the context of a protracted learning curve has not been shown to improve the accuracy of
associated with ERUS [5], the initial operator lymph node staging in rectal cancer [33], and is
variability plateaus with increasing the case load, certainly not routinely practised.
as well as standardising the technique and
interpretation of staging criteria [26–28]. Limitations of ERUS
Technical limitations of ERUS are best consid-
Lymph Node Involvement ered in relation to the rectal anatomy, or the
Beyond the rectal wall is the moderately echo- tumour itself. In the latter case, large polypoid or
genic fatty tissue which contains the lymphovas- stenotic tumours cannot be adequately assessed
cular drainage system of the rectum. Assessment because of the inability of the probe to traverse
of potentially metastatic lymph nodes within the the narrow lumen [5, 23]. Moreover, lesions in
mesorectum continues to present a problem for the upper rectum may be inaccessible for evalua-
all imaging modalities, including ERUS. The tion, and those in the lower rectum have been
reported accuracy for ERUS based nodal staging subject to difficult staging owing to loss of the
is even less than that for evaluating the depth of five layer sonographic appearance in the rectal
invasion. A recent meta-analysis of over 5000 ampulla wall [34]. Further anatomical distortion
patients found a 57% sensitivity and 80% speci- is seen with a wide bore and rigid ERUS probe,
ficity for ERUS predicted involved lymph nodes or the water filled balloon, which both stretch the
[29], with a mean accuracy of 73% [5]. The main rectal wall and impair accurate visualisation.
limitation in assessing lymph nodes is the lack of Finally, the angulation of the probe at the tumour
valid ultrasonographic criteria to discriminate interface, faecal residue in the rectum, prior
between malignant and inflammatory nodal tis- biopsy of the lesion, and the valves of Houston
sue. The commonly applied size criterion of 5mm are all potential confounders that have resulted in
or more to define a malignant lymph node is over-staging of the rectal lesion [9, 23].
arbitrary and of poor predictive value when The major limitation of ERUS, however, is its
compared to gold standard histopathology. Kim inability to accurately define important perirectal
et al. [30] found that up to 18% of lymph nodes anatomical structures, most notably the mesorectal
364 M. Dattani and G. Brown

fascia. The relationship of a rectal cancer to this only in prognostication of the disease, but also in
crucial oncological landmark is predictive of tailoring treatment that whilst maximising the
circumferential resection margin (CRM) chances of a successful cure, spares the patient
involvement, which is an independent risk factor from the effects of unwarranted multimodality
for local recurrence following TME surgery [35]. treatment. The introduction of phased-array sur-
Current multimodality treatment of rectal cancer face coils has enabled the acquisition of high spa-
therefore mandates the precise evaluation of the tial resolution images with equally good soft
mesorectal fascia, as it determines the need for tissue contrast for interpretation, which makes
neo-adjuvant treatment, or the plane and type of MRI based stratification an ideal staging
surgery where curative resection is achievable, modality.
both of which have significant implications for
the patient. MRI Technique
Whilst institutional protocols will vary some-
 uture Perspectives of ERUS
F what, the main principles of MRI technique for
Advancements in ERUS technology have over- local rectal cancer staging are universal, and
come some of the limitations described above, require thin slice, high spatial resolution
including the advent of endoscopic ultrasound T2-weighted images (1 mm 3 voxel size) with a
miniprobes, which enables access to the more small field of view encompassing the rectal
proximal rectal lesions, as well as assessment of tumour and its surrounding perirectal tissue.
large stricturing tumours [36]. Furthermore, Bowel preparation is unnecessary, and an intra-
three-dimensional (3D) ERUS has recently been muscular injection of 20mg hyoscine butylbro-
show to improve diagnostic performance because mide is helpful to minimise peristaltic artefact
of higher resolution and multi-planar image from the small bowel. Endorectal coil use is not
acquisition of the rectal wall and surrounding recommended because of the several limitations
anatomy, including the mesorectal fascia in few that apply to ERUS as well, and not least because
cases [9, 18, 30]. Strain elastography is another of the superior quality imaging acquired with
novel and complementary technique to ERUS, modern day pelvic surface coils. Moreover, the
which relies on the differential resistance of tis- routine use of endorectal contrast is also not
sues to strain, depending on whether there is any advocated, and may in fact impair accurate inter-
malignant infiltration [37]. Early indications pretation of mural invasion by stretching of a full
from these evolving technologies hold promise rectum. Intravenous gadolinium contrast does not
for the future of accurate preoperative staging, improve diagnostic accuracy, and is also not rec-
which will be dictated by robust evidence of their ommended as it needlessly prolongs the exami-
effectiveness in clinical practice. nation [38]. Similarly diffusion weighted MRI
has not added to the diagnostic accuracy in rectal
cancer staging and results in unacceptable pro-
Magnetic Resonance Imaging longation of the scans which patients may find
distressing.
High-resolution magnetic resonance imaging The patient lies supine on the MRI table and a
(MRI) plays a fundamental role in modern day phased-array surface coil is placed around the
multidisciplinary management of rectal cancer. pelvis to secure the patient in position, thus mini-
The ability of MRI to precisely depict and char- mising movement artefact. MRI scanning is then
acterise a rectal cancer, and its relationship to initiated in a cranio-caudal fashion as the patient
important pelvic anatomical structures such as advances into the magnet core, which should ide-
the sphincter complex and mesorectal fascia, is ally be of 1.5 T field strength, or higher. The first
pivotal in its effectiveness as a preoperative stag- sequences are to locate the rectal tumour being
ing modality. Thus, MRI evaluated local staging investigated, and are acquired as T2-weighted
of rectal cancer has important implications not fast spin-echo (T2W-FSE) images in the sagittal
21 Rectal Carcinoma: Imaging for Staging 365

and coronal plane. Based on the longitudinal axis specimen images, and finally correlating them to
of the tumour from these initial images, thin sec- gold standard histopathology. They argued that
tion orthogonal sequences of the tumour and the poor results in previous studies could be
adjacent perirectal tissues are then obtained in accounted for by the use whole body surface coils
the axial plane. The meticulous planning of this and thick sections that afforded poor quality
angulation, prior to imaging, is critical to enable imaging for interpretation. Since then, high spatial
accurate assessment of depth of invasion by the resolution thin slice MRI has become the imaging
tumour. These images should be acquired using a choice for preoperative evaluation of rectal cancer,
16 cm field of view and a matrix resolution of with a sensitivity of 87% and a specificity of 75%
256 × 256 pixels with a 3 mm thickness to achieve for T-stage assessment as reported in a meta-
a 0.6 × 0.6 mm in plane resolution and 1.1 mm3 analysis of just under 2000 patients [41]. The
voxel resolution. To achieve adequate signal to depth of invasion is reported according to the
noise and interpretability a minimum of 4–6 sig- signal intensity criteria, with the tumour having
nal averages is required (approximately 7 min an intermediate signal intensity that is higher than
scan duration). A further scan should be per- the muscularis propria, but lower than the
formed to cover at least 5 cm above the top of submucosa. The latter is normally seen as a thick
tumour to evaluate the draining nodes and vessels layer just deep to a very fine line of low signal
for tumour spread. The rest of the pelvis is then intensity mucosa. An overall mrT-stage is
imaged from the iliac crests to the pubic symphy- accorded, as derived from TNM staging of rectal
sis with a large field of view. cancers (Table 21.2) [12].
Appropriate consideration must also be given As in the case of ERUS staging, some of the
to MRI planning for distal third rectal cancers, reported variability in the accuracy of mrT-stage
owing to anatomical features of the anorectum in evaluation is because of an apparent difficulty in
the low pelvis. At this level, the mesorectum and differentiating a T2 from an ‘early’ T3 rectal
its fascia taper sharply into non-existence, being cancer. The risk of overstaging at this interface is
replaced by the sphincter complex. The abrupt well recognised, but can be avoided by recognis-
change in calibre and angulation of the rectum
risks a tumour at this level being overstaged,
Table 21.2 MRI staging criteria for depth of invasion
especially if perpendicularity of the imaging (mrT-stage)
planes is not maintained. Optimal angulation to
MRI
achieve high-resolution coronal sequences will MRI signal characteristics T-stage
clearly delineate the sphincter complex and its Low signal in the submucosal layer, T1
relationship to the tumour, the benefits of which replacement of the submucosal layer by
cannot be overstated for the planning of a pri- abnormal signal not extending into circular
muscle layer
mary surgical resection. The awareness of com-
Intermediate signal intensity within T2
mon pitfalls and artefacts related to MRI of the muscularis propria. Outer muscle coat
pelvis, and how to minimise their impact on diag- replaced by tumour of intermediate signal
nostic interpretation have been reviewed by Zand intensity that does not extend beyond the
et al. [39]. outer rectal muscle into the perirectal fat
Broad-based bulge or nodular projection (not T3*
fine spiculation) of intermediate signal
 rimary Rectal Cancer Staging:
P intensity projecting beyond outer muscle coat
The Role of MRI Extension of abnormal signal into adjacent T4
organ, extension of tumour signal through the
Depth of Invasion peritoneal reflection
Brown et al. [40] first reported MRI as an accurate Adapted from [12]
*T3 is further subdivided depending on the depth of extra-
method of staging mural spread in rectal cancer, mural invasion from the muscularis propria to the outer
by prospectively comparing section-for-section edge of the tumour; T3a: <1 mm, T3b: 1–5 mm, T3c:
in vivo MRI images to in vitro rectal cancer 5–15 mm; T3d: >15 mm
366 M. Dattani and G. Brown

a stage. Merkel et al. [13] subdivided a large series


of over 800 pT3 rectal cancers into whether
they had extramural invasion of less than 5 mm
(pT3a), or more than 5 mm (pT3b), and found a
significant difference in the 5 year cancer-specific
survival of 85% and 54%, respectively, irrespec-
tive of the lymph node status. They also showed
that survival for a pT2 cancer was comparable to
a pT3a cancer, and on this basis, it can be argued
that neo-adjuvant treatment for this cohort of
patients is unlikely to be of any significant ben-
efit. The precise evaluation of extramural spread,
and thus the sub-staging of T3 rectal cancers
(Table 21.2), is of major importance in the era of
multimodality treatment. The sub-classification
of this heterogeneous group of rectal cancers has
b been reflected in the pathological TNM staging
of rectal cancers [42]. Based on this classifica-
tion, the MERCURY [43] study found that the
MRI assessment of extramural invasion in rectal
cancer correlated to within 0.5 mm of histological
measurement in 295 cases who had primary sur-
gery. In this context, the group later reported that
for MRI staged ‘good’ prognosis rectal cancers
(mrT1-T3b), the 5-year local recurrence rate was
3% with primary surgery alone, regardless of the
MRI reported lymph node status [44]. The com-
bination of MRI staging followed by good quality
TME surgery avoided the need for neo-adjuvant
treatment in 30% of rectal c­ ancer patients with-
out any major oncological ­compromise [44].
Fig. 21.2 (a) Axial T2-weighted MR sections of a mrT2
rectal cancer confined to the bowel wall. The long, low
intensity spikes of fibrosis in the perirectal fat (arrows in Lymph Node Involvement
a and b) demonstrate extramural desmoplasia, and not T3 The soft tissue contrast afforded by MRI makes
extension (b) Corresponding histopathology slice it the optimal imaging modality to identify local
­(haematoxlin-eosin stain) of the same tumour showing the
tumour margin (arrowheads) which is still intramural mesorectal lymph nodes. Despite this, the lack
(Adapted from Ref. [40]) of a universally accepted set of diagnostic cri-
teria in discriminating between a benign and a
ing the obvious differences between the peritu- malignant lymph node presents the main chal-
moural desmoplastic reaction in the extramural lenge in accurate prediction of nodal involve-
fat and the characteristic nodular broader front of ment. Whilst the presence of any visible
infiltration that is observed in early T3 tumours mesorectal lymph nodes is largely obsolete for
(Fig. 21.2a, b). defining metastatic involvement, size criterion
More recently, the depth of extramural inva- remains an important, if controversial, deter-
sion within the mesorectum has been found to be minant. In a morphometric analysis of nearly
a risk factor for local recurrence, and is therefore 13,000 lymph node retrieved from rectal can-
of prognostic relevance in the preoperative set- cer specimens, Dworak et al. [45] demonstrated
ting, particularly because the largest proportion considerable overlap in the size of reactive and
of rectal cancers seen at presentation are of T3 metastatic lymph nodes. Despite this, a range of
21 Rectal Carcinoma: Imaging for Staging 367

The combination of these features as a diagnos-


tic criteria had a predictive accuracy which was
superior to various size cut-offs applied to the
same cohort [50].
A recent meta-analysis of MRI studies for rec-
tal cancer staging confirmed the poor accuracy
that has plagued preoperative lymph node evalu-
ation with all types of imaging modalities.
Al-Sukhni and colleagues [41] reported a sensi-
tivity of 77% and specificity of only 71% for
MRI predicted lymph node involvement, which
is probably a reflection of the heterogeneity in the
diagnostic criteria employed by the individual
studies. Indeed, most studies continue to use size
criteria of various cut-offs [51, 52], and it is plau-
sible that pooling these results into a meta-analy-
Fig. 21.3 A 8mm mesorectal lymph node (arrow) close sis leads to the reported sub-optimal accuracy for
to the mesorectal fascia on a T2-weighted axial image, MRI nodal evaluation.
showing a homogenous signal and regular border contour. A novel technique to improve the accuracy of
This was subsequently confirmed on histology as a benign
MRI nodal assessment has been the development
node
of lymph node specific contrast agents, most
notably ultra-small superparamagnetic iron oxide
cut-offs have been used over the years to predict particles (USPIO) [53]. These particles are pref-
a metastatic lymph node, with size thresholds of erentially taken up by normal lymphoid tissue
3–10 mm used as a diagnostic criteria [46–48]. and then ingested by macrophages, resulting in a
Unsurprisingly, these arbitrary measurements ‘dark’ signal intensity after delayed imaging on a
have yielded an accuracy of between 43-85% T2*-weighted sequence. Thus, inflammatory
when compared with standard histological nodes with a high concentration of macrophages
assessment, confirming the view that lymph should be discernible from metastatic lymph
node size has a poor predictive value for nodal nodes, which demonstrate a high ‘white’ signal
staging [49], as shown in Fig. 21.3. intensity because of no USPIO uptake. Although
Focus has subsequently shifted to the mor- early results of its application were promising
phological appearances of lymph nodes on MRI, [54, 55], the lack of validating evidence from
which has been shown to be a better discrimina- larger population studies has limited its current
tor of nodal involvement. This is because tumour utilisation in clinical practice.
infiltration into a lymph node disrupts the capsu- The overarching debate in recent times has
lar integrity, as well as the signal intensity owing been the significance of mesorectal lymph nodes
to necrosis within the node. Brown et al. [50] [56, 57], especially with the popularisation of
first demonstrated this phenomenon in their anatomically precise rectal cancer surgery based
meticulous study of comparing lymph nodes in on the principles of TME. Quirke et al. [58] found
the dissected pathology specimen to their corre- that in good quality mesorectal plane TME speci-
sponding MRI images in a section-by-section mens, where the rectum and its surrounding lym-
analysis. They reported that an irregular border phovascular mesorectum were excised as an
and mixed signal intensity in a MRI detected intact package, the risk of local recurrence at
lymph node had a sensitivity of 85%, and a spec- 3 years was 4% without neo-adjuvant radiother-
ificity of 97% in correctly predicting histological apy. Thus, some have argued that as optimal
nodal involvement. Conversely, normal or reac- TME surgery achieves good locoregional con-
tive nodes are likely to have a homogeneous sig- trol, lymph node status should not mandate rou-
nal intensity with a well-defined, smooth border. tine adjunctive treatment [56], suggesting instead
368 M. Dattani and G. Brown

a ‘hierarchical’ set of adverse prognostic features


a
that include extramural venous invasion and the
circumferential resection margin. At present
however, assessment of lymph nodes in the pre-
operative setting has an important role, for exam-
ple where local excision is being considered to
avoid the risks of major resectional surgery.

Prediction of Circumferential Resection


Margin Involvement
The relevance of the circumferential resection
margin (CRM) was first postulated in a seminal
paper by Quirke et al. [59] in 1986, in which they b
reported the association of pelvic recurrence with
tumour presence at what they termed the ‘lateral’
margin of rectal cancer specimens. An involved
CRM has since become one of the most important
and well-established prognostic factor in rectal
cancer, and is associated with an increased risk of
local recurrence, distant metastases and reduced
survival [60–63]. The preoperative prediction of
an involved or threatened CRM, therefore, has
significant implications on the need for neo-adju-
vant treatment, planning the type and plane of sur-
gery in order to maximise the chances of a curative
resection, and most crucially, counselling the
Fig. 21.4 (a) Axial T2-weighted MRI section showing the
patient about the potentially adverse outcomes. circumferential mesorectal fascia (white arrows) encasing
The anatomical equivalent of the surgical the mesorectum. In this case, the mesorectal fascia has
CRM is the mesorectal fascia [64]; a thin embry- not been invaded by the tumour (not shown). (b) Axial-
ological fibroareolar sheath that envelopes the Oblique T2-weighted image of a mid rectal tumour (white
arrow) that has perforated through the mesorectal fascia on
rectum and its surrounding fatty lymphovascular the right, with surrounding inflammatory changes
mesorectum, thus forming a natural oncological
barrier. It is beyond this fascia that they ‘holy
plane’ of TME is pursued to achieve the surgical between tumour and mesorectal fascia to predict a
excellence of resecting an oncologically intact threatened margin, a <1 mm threshold is now
specimen. On axial MRI sections, the mesorectal ­universally accepted as a standard, and has the
fascia appears as a low signal intensity circum- benefit of conforming to the histopathological def-
ferential structure that encases the mesorectum inition of an involved CRM [63]. Distance criteria
(Fig. 21.4a, b), which itself is of high signal of <2 mm [65], or even <5 mm [66] have been
intensity similar to fat. used and advocated, and although this unsurpris-
Brown and colleagues [40] first demonstrated ingly increases the sensitivity of predicting CRM
the feasibility of MRI in visualising the mesorectal involvement, the overall accuracy of a <1 mm cut-
fascia in a small, single centre study comprising 28 off is far superior as reported in a recent meta-
patients. Subsequently, they reported a 92% accu- analysis [67]. Moreover, Taylor et al. [68] showed
racy in predicting involvement of the pathological in a large multi-centre study that with a cut-off
CRM if tumour invasion was within 1mm of the criteria of tumour <5 mm from the mesorectal fas-
mesorectal fascia on MRI [12]. Whilst there was cia to predict pathological CRM involvement, up
initial uncertainty about the optimal distance to 13 additional patients would need neo-adjuvant
21 Rectal Carcinoma: Imaging for Staging 369

treatment in order to prevent one local recurrence, meta-analysis of 20 studies, which were mostly
representing overtreatment in this group. retrospective non-randomised cohorts, comparing
The MERCURY study [69], a European curative TME resection and PSWLN dissection
multi-centre prospective observational study, versus TME alone found no difference in the local
reported a MRI accuracy of 88% in predicting recurrence rates between the two strategies [75].
CRM involvement, and an even higher negative In this context, clinical practice in North America
predictive value of 94% when validated against and Europe has been to either ignore suspicious
the gold standard pathological assessment of PSWLN seen on pre-operative imaging, or
CRM involvement. On this basis, when the MRI more commonly, to offer chemoradiotherapy.
predicted mesorectal fascia is clear of tumour, Currently, there is no evidence based consensus
primary surgery based on the principles of TME on which approach offers the best oncological
can be recommended to achieve a R0 resection outcome, largely limited by the lack of an accu-
with a high degree of certainty. The MERCURY rate predictor of PSWLN involvement that can
group subsequently found that MRI assessment guide treatment choice.
of the CRM status was an independent prognostic Debate continues about what constitutes a
marker, predictive of the 5-year local recurrence ‘suspicious’ PSWLN on MRI, much like in the
and survival rate [35]. This series forms the larg- optimal diagnostic criteria for predicting a meta-
est contributor to a recent meta-analysis of 1600 static mesorectal lymph node, as discussed previ-
patients, which found a 74% sensitivity and a ously. Although size is considered a poor
93% specificity in the use of MRI to assess discriminant, it is still employed with various
involvement of the mesorectal fascia in rectal cut-offs to base decisions about neo-adjuvant
cancer staging, prior to any treatment [67]. treatment, or the need to pursue PSWLN dissec-
tion [76, 77]. The MERCURY group have instead
Pelvic Side Wall Lymph Nodes adopted morphological MRI criteria of mixed
As alluded to previously, MRI staging enables signal intensity and irregular contour to define an
evaluation of the whole pelvis, including assess- involved PSWLN (Fig. 21.5a, b) [78]. Judged by
ment of the lateral wall compartment in which these criteria, they reported a 12% incidence of
prominent lymph nodes are occasionally seen. MRI suspicious PSWLN in 325 patients with
The clinical significance and management of rectal cancer, limited of course by the absence of
these pelvic side wall lymph nodes (PSWLN) in histological validation. These patients had a sig-
rectal cancer remains contentious, but it is pur- nificantly poorer disease free survival compared
ported to be one of the reasons for locoregional to those without MRI suspicious PSWLN, how-
failure despite optimal TME surgery, which does ever, when neo-adjuvant radiotherapy was fac-
not address extra-mesorectal disease [70]. This is tored into the analysis, the difference was no
particularly pertinent in the distal third rectal can- longer apparent between the two groups [78].
cers, owing to the preferential lateral lymphatic
spread of low lying tumours along the internal Extramural Vascular Invasion
iliac artery, and then to various lymph node sta- Extramural vascular invasion (EMVI) is defined
tions on the pelvic side wall [71, 72]. Sugihara as the presence of tumour cells in the microvascu-
et al. [73] reported a histologically confirmed lature beyond the muscularis propria, and is a
15% incidence of metastatic PSWLN in cancers well-established adverse prognostic factor
below the peritoneal reflection, compared to a sig- ­[79–81]. Whilst EMVI is more prevalent in locally
nificantly lower 8% incidence in upper rectal can- advanced T3/4 rectal cancers, it can present as
cers from a large multi-centre cohort. In the East, discontinuous foci with early stage intramural
and particularly Japan, PSWLN dissection is tumours [82], and must be sought for to improve
therefore widely pursued and advocated, accept- the accuracy of prognostication. Post-operatively,
ing the risks associated with what is an otherwise histologically confirmed EMVI has been shown
major and high morbidity procedure [72, 74]. A to be associated with a higher risk of local and
370 M. Dattani and G. Brown

a a

b b

Fig. 21.6 (a) Axial T2-weighted MRI section showing


EMVI with the corresponding H&E stained histological
whole mount section. (b) A tongue of tumour is seen
extending into the perirectal fat (arrow) associated with
a vessel, which appears as a signal void on MRI
(arrowheads)

several factors, including the lack of standardised


pathological reporting criteria [90], the process-
ing and number of tissue blocks examined [86,
91], expertise of the reporting pathologist [92],
and the use, or not, of ancillary specialised stains
Fig. 21.5 Axial (a) and Saggital (b) T2-weighted MRI
section showing a 14mm metastatic left pelvic side wall to improve detection [91, 93].
lymph node (white arrow). Note the heterogeneous signal Recent interest has been generated by the
intensity and irregular border which is suggestive of ability of MRI to detect EMVI pre-operatively,
tumour infiltration. (Images courtesy of Mr. B Moran,
as part of standard rectal cancer staging.
Basingstoke and North Hampshire Hospital, U.K.)
Brown and colleagues [12] characterise EMVI
as intermediate signal intensity serpiginous
distant failure [80, 81, 83, 84], and poorer overall structures in the mesorectal fat, similar to
survival [85, 86], regardless of the nodal status or tumour signal, that are seen extending into a sig-
depth of mural invasion [87, 88]. The reported nal void tubular structure which represents a
incidence of histopathological EMVI in rectal blood vessel (Fig. 21.6a, b).
cancer varies from 9 to 61% [89], and to a large In this seminal study, MRI correctly predicted
extent reflects the variability in reporting amongst histological EMVI in 15 out of 18 cases where
pathologist. The diagnostic yield is influenced by large veins were involved, with small calibre
21 Rectal Carcinoma: Imaging for Staging 371

Fig. 21.7 EMVI MRI-EMVI Predicted Typical imaging features Schematic illustration
Grading system. With score EMVI
permission John Wiley
and Sons [95]
Pattern of tumour extension
through the muscle coat is not
0 nodular, and there are no vessels
adjacent ot areas of tumour
penetration

Negative
Minimal extramural standing/
nodular extension, but not in
1 the vicinity of any vascular
structure

Stranding demonstrated in the


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

Intermediate signal intensity


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

Obvious irregular vessel contour


4 or nodular expansion of vessel by
definite tumour signal

vascular invasion being missed even after ret- accuracy of MRI predicted EMVI [94], such that
rosepctive review [12]. grade 0–2 are effectively EMVI negative, whereas
The heterogeneous relationship of perirectal grade 3–4 are strongly suggestive of the presence
vasculature and tumour presence has led to the of EMVI.
formulation of a MRI-detected EMVI grading Based on this grading system, MRI predicted
system which consists of 4 defining criteria pathological EMVI with a high specificity ranging
(Fig. 21.7): pattern of tumour margin; location of between 88% and 96%, but a variable sensitivity
tumour relative to major vessels; calibre of of 29–62% [95–97]. The low sensitivity of MRI
vessels; vessel border [94]. A five-point grading predicted EMVI may be due to the inability to
score based on combination of the above criteria accurately visualise small calibre vessels
has been proposed to improve the diagnostic (<3 mm), even in a high resolution setting.
372 M. Dattani and G. Brown

Alternatively, it is perhaps more likely that EMVI of the rectum to other pelvic viscera, and in
was underreported in histopathology evaluation particular the safety provided by the mesorectum
either because of vessel obliteration during in the mid rectum, or a lack thereof, in the low
specimen handling [95], or as some authors have rectum whereby a locally advanced tumour is
alluded, sub-optimal pathological interpretation, likely to compromise preservation of the anal
leading to false negative reports [97]. Indeed, it sphincters. Given that these distances will vary
has been suggested that MRI evaluation of EMVI somewhat between individual patients, a more
may be more accurate than histopathological objective MRI based definition of low rectal
assessment [91], with an incidence of 22–39% cancer has been proposed as “an adenocarcinoma
reported in the literature using the above MRI with its lower edge at, or below, the origin of the
grading score [95–98]. levators on the pelvic sidewall” [101].
The presence of MRI detected EMVI has been Approximately one third of all rectal cancers
shown to predict disease relapse [95], and in a will be ‘low’ according to this definition [102].
large series of 450 rectal cancer patients, it was The mainstay of a curative surgical resection for
found to be an independent risk factor for syn- low rectal cancer (LRC) is either a low anterior
chronous metastases [97]. Bugg et al. [98] subse- resection (LAR), or in up to 45% of cases, an
quently showed in a cohort of 200 patients that abdominoperineal excision (APE) that results in
MRI detected EMVI was associated with an a permanent stoma [103].
almost fourfold risk of developing metachronous The merit of pre-operatively evaluating LRC
metastases within 1 year of rectal cancer diagno- as a discrete entity, and thus individualising a
sis. In this context, the presence of EMVI on MRI treatment plan, is related to the historically
is of significant relevance, as it may indicate poor outcomes associated with an APE, com-
tumour embolisation into the systemic circulation pared to anterior resection for upper- and mid
with possible micrometastases at the time of pre- rectal tumours which have seen vast prognostic
sentation, which remain undiagnosed because of improvements following the widespread adop-
the limitations of current imaging modalities. tion of TME surgery. Multiple studies have
More crucially, present day multimodality shown that patients undergoing an APE have a
­treatment in the form of neo-adjuvant chemora- higher rate of pathological CRM involvement—
diotherapy and TME surgery does not address this over 30% in some large series—resulting in
issue, and may in part account for the good local higher rates of local recurrence and poorer
control that has been achieved with this approach, survival [104–106]. This marked difference in
but the failure to prevent distant relapse and outcomes can be explained by the surgical chal-
improve survival. Although not routine practice, it lenges of operating within the narrow confines
may be that MRI detected EMVI patients are the of the low pelvis on a segment of the rectal
ideal candidates, amongst other high risk patients, tube which is devoid of the protective mesorec-
for up-front systematic chemotherapy in order to tum, which tapers sharply into non-existence at
secure distant control and improve survival, as is the proximal insertion point of the sphincters.
being trialled in randomised studies [99]. These ­difficulties culminate in the resection of
a sub-optimal surgical specimen, often with a
 RI Evaluation of Low Rectal Cancer
M ‘waist’ around the lowest part of the mesorec-
The rectum is arbitrarily divided into 3 parts tum, or in some cases an iatrogenic perforation
depending on the height from the anal verge, as into the tumour or bowel [106, 107]. A greater
measured by a rigid sigmoidoscope when the awareness of these pitfalls, coupled with the
patient is in the left lateral position; low rectum is refinement of surgical techiniques to ensure a
up to 6 cm from the anal verge, mid rectum is more cylindrical specimen, has led to the use
from 7 to 11 cm, and the upper rectum is from 12 of an extended or ­extralevator APE (ELAPE)
to 15 cm [100]. The importance of this [108], particularly when the levators or sphinc-
classification relates to the anatomical relationship ter complex have been invaded by tumour. But
21 Rectal Carcinoma: Imaging for Staging 373

the radicality of such an operation is associated stage 3 or 4 will require an ELAPE, or exentera-
with a significant perineal defect which is not tive surgery if adjacent viscera are involved in
always suitbale for primary closure, occasion- order to achieve a clear resection margin.
ally requiring reconstructive surgery with myo- Conversely, stage 1 or 2 low rectal cancers can
cutaneous flaps, or a biological mesh to achieve safely achieve a clear CRM with an intersphic-
wound closure and prevent herniation [109]. teric resection, with a colo-anal anastomosis
Despite these techniques, there remains a risk where feasible and indicated (Fig. 21.8).
of perineal wound morbidity which is enhanced A composite MRI staging system for LRC has
with the use of neo-adjuvant radiotherapy [109, subsequently been developed which encom-
110]. Moreover, the early results of studies passes the assessment of tumour proximity to
comparing standard APE against ELAPE have mesorectal fascia, as well as the involvement of
been mixed, and because not every LRC war- the intersphicteric space which can be used to
rants an ELAPE, an accurate staging system to define the surgical plane of excision. Thus, the
tailor the optimal plane of surgery, or appropri- low rectal plane can be considered ‘safe’ to
ately select patients for neo-adjuvant therapy is achieve a complete surgical resection when
highly v­ alued [111]. tumour does not invade both of the aforemen-
To address these complexities in the man- tioned structures (Fig. 21.9).
agement of LRC, an MRI based anatomical The MERCURY II [103] study was the first to
staging system has been devised which assesses prospectively validate this LRC staging system in
the relationship of the tumour to both the inter- 279 patients recruited from 14 centres across
sphinteric space and levator muscle [102, 112] Europe, and achieved and overall pathological
(Table 21.3). CRM involvement of 9%, which is significantly
Salerno et al. [113] retrospectively validated lower than the historically reported rates.
this staging system and demonstrated that stage 3 Reassuringly, when the MRI predicted low rectal
and 4 low rectal tumours had an 18-fold increased cancer plane was safe (Stage 1 and 2 tumours and
risk of a pathologically involved CRM, compared MRF clear), the rate of CRM involvement was
with stage 1 and 2 tumours. When the latter group reduced to only 4%, confirming the utility of
had a standard APE without radical en-bloc MRI in aiding the pre-operative decision making
resection of the levators, the CRM was involved process. Furthermore, the combination of a MRI
in over 50% of the cases. Thus, according to the evaluated safe low rectal cancer plane with no
LRC staging system, tumours categorised as other adverse prognostic features (<mrT3c, mrN2
and no mrEMVI) resulted in CRM involvement
in only 1 out of 62 cases (1.6%). None of these 62
Table 21.3 MRI based staging system for low rectal patients were deemed to require neo-adjuvant
cancer therapy based on the MRI staging, and managed
MRI stage to safely avoid the additional co-morbidity of
for LRC Description of stage assessment chemoradiation by proceeding straight to sur-
Stage 1 Tumour confined to the bowel wall and gery, without any compromise of the short term
does not extend through the full
oncological outcomes.
thickness; intact outer muscle coat
Stage 2 Tumour replaces the muscle coat but On multivariate analyses, three additional
does not extend into the intersphincteric MRI risk factors were identified that predicted an
space involved CRM, namely, a tumour located at a
Stage 3 Tumour invades the intersphincteric height of less than 4cm, anterior quadrant inva-
space or lies within 1mm of the levator
sion, and the presence of EMVI [103]. Based on
muscle
Stage 4 Tumour invades the external anal these findings, the group proposed a risk stratifi-
sphincter and is within 1mm and beyond cation model according to which the presence of
1mm of the levator muscle, with or all 4 adverse features predicted a risk probability
without invading adjacent structures of up to 60% for an incomplete resection. These
374 M. Dattani and G. Brown

a b

Fig. 21.8 Pictorial description of low rectal cancer and on the left in (a) has extended beyond the intersphincteric
plane of resection in an oblique coronal view. (a) The space into the levator ani and puborectalis muscle. This
tumour on the left depicts an early stage low rectal cancer would require an extralevator APE (ELAPE) as shown by
that has not breached the muscularis propria/internal the dashed blue lines in (b) to achieve a clear CRM
sphincter, and would therefore be oncologically suitable (Adapted from Battersby et al. Exp Rev Gastroenterol
for a standard APE, or a restorative intersphicnteric resec- Hepatol 2014;8:703–19)
tion as shown by the dashed green line in (b). The tumour

multivisceral resection beyond the conventional


TME plane in order to achieve a R0 resection
[114]. Despite the risks of significant morbidity
and mortality, and the effect on patients’ quality
of life after what is potentially mutilating surgery,
a clear pathological resection margin is the most
important determinant of survival in patient with
non-metastatic disease [115]. Accurate pre-opera-
tive staging can not only help determine the plane
of surgery required to achieve an en-bloc resec-
tion, but also mitigate the need for a default exen-
terative procedure and its associated consequences.
Pelvic high resolution MRI remains the choice of
imaging in most specialist centres, according to
an expert consensus statement by the ‘Beyond
Fig. 21.9 Axial MRI of low rectal cancer invading the TME’ collaborative [114]. More recently, MRI
intersphincteric plane staging according to pelvic surgical compart-
ments, rather than involvement of individual
are precisely the patients who may benefit from organs, has been proposed as an oncologically
the addition of neo-adjuvant chemoradiotherapy superior method of staging because it facilitates
to achieve downstaging from an ‘unsafe’ to a the selection of anatomical planes of dissection.
‘safe’ low rectal cancer plane, or in the case of Georgiou et al. [116] reported a high diagnostic
poor responders, consideration of exenterative accuracy of compartment invasion by tumour
surgery. (ROC >87%) on such a MRI based staging sys-
tem, in which there was also good inter-observer
 taging for ‘Beyond TME’ Surgery
S agreement for both recurrent and primary rectal
Approximately 5–10% of patients will present cancer requiring curative resection beyond the
with a locally advanced rectal cancer that requires TME plane. However, prospective validation of
21 Rectal Carcinoma: Imaging for Staging 375

Fig. 21.10 Compartmental pelvic staging for ‘Beyond reflection (PR); posterior; infralevator. Adapted and
TME’ surgery using high resolution MRI. Six distinct modified from [117], which also includes a description of
anatomical compartments are shown: Lateral (L); central the individual organs in each compartment
above peritoneal reflection (PR); central below peritoneal

this system in a larger study is warranted, and this tion in its worldwide use [14, 125–127], is detri-
is currently underway in a multi-centre trial mental to the patient and of little clinical benefit
whereby the pelvis is divided into six surgical when optimal TME surgery is performed for
compartments as shown in Fig. 21.10 [117]. operable rectal cancer with no high risk features
[58, 128, 129]. A selective, risk stratifying policy
 RI Staging to Guide Neo-Adjuvant
M that spares patients from the long term toxic
Treatment in Rectal Cancer effects of neo-adjuvant treatment, without neces-
Neo-adjuvant radiotherapy, either alone or in sarily compromising local control, is therefore
combination with chemotherapy as a long course highly desirable [130].
regimen has been shown to reduce local recur- In North America, the National Comprehensive
rence rates [118–121], without any significant Cancer Network (NCCN) recommends
improvement in overall survival. But pelvic irra- neo-adjuvant treatment for all stage II [T3/4; N0]
diation is in itself not without consequence, with and stage III [T1-T4; N1/2] [131] rectal cancers,
genitourinary impairment, bowel dysfunction a policy also adopted by several other interna-
following restorative surgery [122], perineal tional guidelines [14]. However, up to 80% of all
wound morbidity in patients undergoing an APE rectal cancers are stage II or III at presentation,
[123], and the risks of a secondary malignancy and the heterogeneity in this broad group means
[124] all reported in the literature. The unstan- that some patients will inevitably be over treated,
derdised routine use of neo-adjuvant treatment reflecting a recent debate about multimodality
for rectal cancer, as evidenced by the wide varia- treatment in the intermediate risk cancers within
376 M. Dattani and G. Brown

Rectal cancer assessed by MRI

‘The ugly’
Predicted LR risk >20%
Threatened (<1mm)
‘The good’ or
‘The bad’ involved MRF
or
EMVI present
Predicted LR risk <10% Predicted LR risk 10-20%
T1 - T3a/b, N0 T3c/d - T4 or N1/2 Low rectal cancer:
No EMVI MRF clear Involved inter-sphincteric plane
MRF clear or
levators

Primary TME surgery Consider neo-adjuvant short course Offer neo-adjuvant long concourse
radiotherapy (5 x 5 Gy), or long course chemoradiotherapy
chemoradiotherapy

Fig. 21.11 MRI-based risk stratification of local recurrence in rectal cancer, with subsequent recommendation of treat-
ment (Adapted and modified from [129–131])

this prognostically diverse group [128, 132, 133]. advantage of eliminating the sampling biases
Consequently, one approach being adopted in the inherent in selecting patients from a progonosti-
U.K and some of the Northern European coun- cally heterogeneous stage II/III population,
tries has been a shift from the TNM based pre- which may mask the true effect of a given
operative evaluation, to MRI based risk treatment.
stratification in order to guide rectal cancer man-
agement, and in particular, the use of neo-adju-
vant treatment (Fig. 21.11). [134, 135].  ssessment of Response to
A
The distinction between ‘good’, ‘bad’ and Neo-Adjuvant Treatment
‘ugly’ tumours depending on MRI based risk
factors of local recurrence has started to be Over the last decade, the indications for
incorporated into clinical guidelines for rectal neo-adjuvant treatment in rectal cancer have
­
cancer management, most notably in the U.K extended beyond the aim of minimising local
[136] and by the European Society of Medical recurrence rates following surgery. Today, long
Oncology (ESMO) [137]. By selecting patients course chemoradiotherapy followed by delayed
for neo-adjuvant treatment based on these highly surgery is recommended for locally advanced
discriminative MRI features, rectal cancer man- rectal cancers that threaten the surgical CRM,
agement can be personalised to achieve maximal with the intention that downstaging of the tumour
benefit in clinical practice, whilst minimising the will allow for a curative resection [138]. This
risks of over- or under treatment. This strategy is may facilitate a surgical procedure less radical
also increasingly being implemented in the than was initially necessary to achieve a complete
design of clinical trials such as the the joint resection, with the added benefit of sphincter, or
Dutch/Nordic RAPIDO trial [99], and has the even organ preservation through a transanal local
21 Rectal Carcinoma: Imaging for Staging 377

excision in the select few. Moreover, in a prog- partial response is arbitrary, and is unvalidated
nostically favourable subset, sufficient tumour against outcome data [145]. Finally, the RECIST
regression takes place such that there is no clini- criteria do not account for treatment response in
cal evidence of a viable cancer, with the advan- non-tumoural malignant deposits such as EMVI,
tage that patients may be able to avoid surgery or the involvement of vital anatomical structures
and its associated morbidity altogether. These such as the CRM, which remains the most impor-
decisions are challenging, contentious, and high tant determinant of local recurrence in rectal
risk for both patients and clinicians, and are only cancer.
possible in the context of restaging the rectal can- The other issue in restaging rectal cancer after
cer to evaluate its response to neo-adjuvant neo-adjuvant treatment is the choice of imaging
treatment. modality, with MRI and ERUS being the
Whilst the importance of restaging is undis- preferred options [140], and Computed
puted, questions remain unanswered about the Tomography (CT) largely reserved for extra-
optimal time interval between completion of pelvic disease assessment. Both MRI and ERUS
neo-adjuvant treatment and surgical resection are subject to the same limitations discussed
[139], the timing of restaging the tumour in this previously, with the added difficulty that the
hiatus, the imaging modality that best assesses post-chemoradiation effects compromise
response to treatment [140], and the safety of accurate re-staging (denoted by the prefix “y”)
recommending a change in the surgical strategy even further. It is therefore unsurprising that a
[141], or in the presence of a clinical complete meta-analysis reported the average accuracy of
response (cCR), advocating organ preservation predicting ypT with MRI was 52%, and for
under a Watch-and-Wait surveillance programme ERUS at 65% [140], with overstaging of early
[142, 143]. tumours a particular issue for both modalities
[147]. The accuracy of predicting ypN status
Evaluating Tumour Response was modest, with both MRI and ERUS having an
Imaging the assessment of response to neo-adju- estimated average of 72% [140, 148].
vant treatment is vital, albeit challenging, despite But whilst the absolute prediction of the vari-
attempts to standardise the reporting criteria. The ous ypTN stages is desirable, what is perhaps
RECIST (response evaluation criteria in solid more valuable from a therapeutic perspective for
tumours) criteria [144] have been widely adopted a tumour that has partially responded to neo-
and are based on changes of the tumour size in its adjuvant treatment is its relationship to the MRF
longest diameter, which is an objectively measur- or the intersphincteric plane, because of the
able and reproducible parameter. Response can ­possibility of a more conservative curative resec-
be categorised as complete if there has been a tion than was initially possible.
total disappearance of the tumour, partial or pro-
gressive depending on percentage change from  ssessment of CRM Following
A
baseline diameter, or stable disease in case of no Neo-Adjuvant Treatment
appreciable change in measurement. However, MRI retains a good accuracy of predicting CRM
pelvic irradiation results in a combination of tis- involvement after neo-adjuvant treatment, with
sue oedema, inflammation, necrosis and fibrosis reported averages of between 70% and 92% [140].
of the tumour bed [145], all of which can be dif- Whilst the specificity and negative p­redictive
ficult to distinguish from residual tumour when values have been shown to be consistently high,
evaluating the response to neo-adjuvant treat- the main compromising factor is of overstaging,
ment. For example, extensive tumour necrosis in such that the risk of over-treatment is increased
solid organ cancers has been shown take place with a higher number false positive predictions.
without any corresponding tumour shrinkage This arises mainly because of the uncertainty in
[146]. Moreover, the definition of more than a differentiating fibrotic tissues from residual
30% reduction in tumour diameter to indicate a tumour, especially if there has been a concurrent
378 M. Dattani and G. Brown

retraction of the MRF along with the tumour mutilating surgery are immense and obvious,
[149], leading to a cautious overcall at re-staging. and increasing focus has therefore turned to the
In the case of low rectal cancer, the MERCURY concept of a clinical complete response (cCR) as
II study reported on 92 patients who had sphinc- a surrogate for the former entity.
ter involvement and were deemed to require an A cCR is defined when there is no clinical evi-
ELAPE to achieve a clear pCRM according to the dence of a tumour on imaging, endoluminal visu-
baseline MRI staging. Following neo-adjuvant alisation, or digital palpation where this is
treatment and a re-staging MRI, 33 (36%) of the possible [153]. Patients who exhibit a cCR on re-
cases were downstaged with a clear intersphinc- assessment following neo-adjuvant treatment
teric plane, out of which 7 (21%) avoided a stoma may be offered the option of non-operative man-
by undergoing a less radical operation than the agement under a careful ‘Watch-and-Wait’ sur-
initially planned ELAPE; all 33 patients had a veillance programme to monitor their progress.
clear pCRM [103]. This strategy was initially met with scepticism
A change in the operative strategy based on when first introduced by Habr-Gama’s group
the re-staging MRI scan is not without conten- [142, 154], because of concerns about oncologi-
tion, with concerns about the safety of dissecting cal compromise and the success of salvage sur-
through a ‘sterile’ plane which may still harbour gery for tumour regrowths, but has since been
cancer cells [150]. reported to be both feasible and safe by several
groups internationally [155–158].
 ssessment of EMVI Following
A In addition, the value of imaging assessment
Neo-Adjuvant Treatment in determining a cCR is increasingly being rec-
There is a paucity of data regarding assessment ognised. In a prospective cohort study with pCR
of the ymrEMVI status, and its effect on onco- as the reference endpoint, the addition of MRI
logical outcomes. There is only one study of 188 assessment to clinical evaluation alone (digital
rectal cancer patients by Chand et al. [151] which rectal examination and endoscopy) increased the
showed that patients who had persistent mrEMVI post-test probability of detecting a complete
following neo-adjuvant treatment had a worse response from 90% to 98% [159]. Moreover,
disease-free survival at 3 years (43%) compared pCR prediction by MRI assessment of tumour
to those who were mrEMVI negative on re-stag- response has been shown to be tenfold higher
ing (80%). The stratification was based on a than clinical assessment, which has a lower sen-
novel tumour regression grade in which a greater sitivity because of persistent mucosal
than 50% fibrosis in the mrEMVI was taken as a ­abnormalities [160]. In the latter study, the MRI
substantial response and re-staged as ymrEMVI criteria for evaluating response to neo-adjuvant
negative [152]. treatment are based on the 5-grade Mandard
scoring system for assessing pathological tumour
 umour Regression Grade
T regression, which is dependent on the relative
and Complete Response to proportions of fibrosis and residual tumour in the
Neo-Adjuvant Treatment treated cancer [161]. Radiologically, fibrosis
A proportion of rectal cancer patients who manifests on high resolution T2-weighted images
receive neo-adjuvant treatment will go on to as low signal intensity, whilst tumour is indicated
have what is termed as a pathological complete by an intermediate signal (Table 21.4).
response (pCR)—no evidence of a residual The mrTRG score has been retrospectively
cancer at histopathological analysis of the validated in the MERCURY trial patients, which
surgical specimen. For the commonly used found it to be an important prognostic determinant.
regimens of chemoradiotherapy, the average Good response tumours (mrTRG 1–3) had a
proportion of those who achieve a pCR is significantly better disease-free survival and
estimated at 15–30% [142]. The benefits of overall 5-year survival than poor response tumours
being able to predict a pCR and avoid potentially (mrTRG 4–5) [162]. Moreover, mrTRG 1 and 2
21 Rectal Carcinoma: Imaging for Staging 379

patients had a similar survival outcome to those tumour response following neo-adjuvant treatment.
with a pCR, and may therefore be optimal candi- However, the evidence for these modalities is sub-
dates for a Watch-and-Wait strategy. These find- optimal according to a recent systematic review
ings have now been used to apply mrTRG as an [163], given that most studies comprise of small
imaging biomarker to stratify rectal cancer retrospective cohorts that are not validated against
patients following neo-adjuvant treatment in the established markers of response or long-term
multi-centre randomised controlled trial called outcomes data. Additionally, the uptake of FDG by
TRIGGER (Eudract No.: 2015-003009-40). metabolically active benign tissue is well a
Rectal cancer patients receiving neo-adjuvant recognised issue, and in the context of radiotherapy
therapy as part of their treatment will be induced inflammation, may be a significant con-
randomised to standard TME surgery which is the founder in accurately assessing post-treatment
current standard of care, or mrTRG directed tumour response. This is compounded by a lack of
management where ‘good response’ patients consensus on the optimal cut off values for SUV
(mrTRG1) are offered intensive follow-up in a that predict for a pCR [163].
Watch-and-Wait protocol, whilst the ‘poor
response’ group is recommended intensified
additional chemotherapy (Fig. 21.12a, b). Evaluation of Extra-Pelvic Disease
There is also a growing interest in the use of
functional imaging, particularly diffusion weighted The importance of imaging beyond the primary
MRI (DWI-MRI) and 18F-fluorodeoxyglucose rectal tumour is evidenced by the high risk of
positron emission tomography (18F-FDG PET) synchronous metastatic disease, currently
with the combination of CT (PET/CT) to assess estimated in up to 30% of colorectal cancer
patients at presentation [164]. This has obvious
Table 21.4 The MRI based tumour regression grade implications for the multimodality management,
(mrTRG) prognosis, and most importantly, counselling of
mrTRG 1—complete radiological response (linear low the patient. The liver is the commonest site for
signal intensity scar only) synchronous metastasis, with a reported incidence
mrTRG 2—good response (dense fibrosis, no obvious of 15% [165], followed by lung and peritoneal
tumour signal)
involvement in up to 10% of patients [166, 167].
mrTRG 3—moderate response (>50% fibrosis and
visible intermediate signal) The main focus of evaluating extra-pelvic
mrTRG 4—slight response (mostly tumour) compartments is therefore the detection of
mrTRG 5—no response (intermediate signal concurrent disease at these sites.
comparable to baseline MRI scan)

a b

Fig. 21.12 (a) Axial image of a pre-treatment rectal cancer. (b) Axial MRI image of post-treatment rectal cancer
­showing good response (mrTRG1)
380 M. Dattani and G. Brown

Hepatic Metastases all have to be addressed in the decision making


Computed tomography (CT) represents the most process.
preferred choice of primary imaging modality for The addition of 18F-FDG PET to CT staging of
detecting distant metastatic disease, and the thorax has also been shown to be of sub-opti-
supersedes other imaging modality because of its mal diagnostic value, with a sensitivity of 57.1%
widespread availability, reproducibility and [173] in diagnosing pulmonary metastases. This
lower cost. However, in a meta-analysis of pro- is because of the lower resolution of 18F-FDG
spective trials, the sensitivity of contrast enhanced PET in characterising lesions <10 mm in size
CT for diagnosing colorectal liver metastases compared with thin slice CT scanning, which
was reported to be 83.6% in patient who had no remains the first choice of investigation for pul-
previous treatment, compared to 88.2% and monary staging. Thus, current indications for the
94.1% for MRI and 18F-FDG PET, respectively routine use of 18F-FDG PET/CT in primary rectal
[168]; there was little variation in the specificity cancer staging are limited, and it may be advo-
of each modality (93–96%). Furthermore, in cated in a patient being considered for curative
lesions measuring less than 10 mm in size, MRI hepatic or pulmonary metastectomy to rule out
outperforms CT in being able to characterise a an occult metastasis, where conventional CT or
metastatic deposit. The higher accuracy of MRI MRI imaging may not have been informative.
is because of the enhanced soft tissue resolution,
with further gains in interpretation reported with Peritoneal Metastases
the use of diffusion weighted, or contrast Pre-operative diagnosis of peritoneal carcinoma-
enhanced liver specific MRI [169]. Most hepato- tosis remains a major challenge, and is frequently
biliary units that treat colorectal liver metastases identified for the first time at operation for the
now mandate pre-operative imaging with primary tumour. Contrast enhanced multi-slice
multiparametric MRI because of this accuracy. CT is the principal imaging modality despite a
modest accuracy of between 60% and 88% [174,
Pulmonary Metastases 175], with underestimation of the true extent of
In most instances, a CT of the thorax is performed peritoneal involvement. The main limitation of
as part of the extra-pelvic staging of primary rec- all available imaging modalities is the small size
tal cancer, and is recommended in the American of peritoneal nodules, typically less than 1 cm in
College of Radiology guidelines [170]. The ratio- size, which frequently spread along the normal
nale for routine imaging of the thorax is hae- anatomical planes in the abdomen, thus compro-
matogenous spread of cancer directly to the lungs mising the sensitivity even further. Direct visuali-
via the haemorrhoidal plexus which drain into the sation of the peritoneal surfaces remains the most
vena cava, bypassing the portal circulation alto- accurate way of establishing the tumour burden,
gether. Indeed, isolated pulmonary metastases and is increasingly becoming a common staging
arising from a primary rectal cancer are not modality in patients presenting with metachro-
uncommon [171]. Moreover, rectal cancer metas- nous disease and being considered for surgery
tases to the lung are more common than colon [176]. But unlike primary staging in hepatopan-
cancer. Whilst the importance of detecting pul- creaticobiliary and gastroesophageal cancers, its
monary metastases cannot be overstated, CT role in the detection of synchronous colorectal
imaging frequently identifies small indeterminate peritoneal metastases is unknown at present.
lesions because of its low specificity, with a vari-
able incidence of between 4 and 42% reported in
the literature. Of these, only 1% are eventually  ynoptic Reporting in Rectal
S
confirmed as metastases [172], and there are con- Cancer Staging
siderable costs of follow-up imaging, radiation
exposure, additional tests which may include A key component of the pre-operative evaluation
invasive diagnostics, and patient anxiety which of rectal cancer is the accurate reporting and
21 Rectal Carcinoma: Imaging for Staging 381

communication of radiological findings that the completeness of pre-operative MRI staging


determine patient treatment pathways, in reports [180]. The use of a synoptic report is one
particular the selection of patients for neo- of the required standards of the new American
adjuvant therapies. Traditionally, radiology College of Surgeons Commission on Cancer
reports have been issued in ‘free-form’ and National Accreditation Program for Rectal
unstanderdised text, based on the radiologist’s Cancer [181]. The American College of
opinion of pertinent staging features. However, Radiology has been very supportive of this pro-
this form of unstructured reporting has been gram and has made available a very high quality
associated with the omission of crucial prognostic educational module about both rectal cancer MRI
information such as CRM involvement, both in staging and the use of synoptic reports [182].
histopathology and radiology reports [177, 178].
One solution of enhancing the completeness and
consistency of radiology reports is the use of Summary
synoptic templates that provide summarised
information which various members of the Advances in imaging technology have revo-
multidisciplinary team can access and interpret lutionised the way in which rectal cancer is
easily [179]. An example of a MRI based synoptic managed. The remit of accurate pre-operative
reporting proforma with individually itemised staging goes wider than the traditional prog-
prognostic features, and the option of additional nostic stratification of patients. With a complex
free text is shown in Fig. 21.13. The range of treatment options available, multi-
implementation of a such a reporting system at a disciplinary teams integrate vital information
population level reported a 39% improvement in from radiological staging with clinical evalua-

Fig. 21.13 Proforma for MRI database


382 M. Dattani and G. Brown

tion to personalise management plans for rectal cancer with transrectal sonography: caution is neces-
sary. Radiology. 1994;190(3):715–20.
cancer patients. MRI remains at the mainstay 11. Mackay SG, Pager CK, Joseph D, Stewart PJ,
of this process; local staging with high resolu- Solomon MJ. Assessment of the accuracy of tran-
tion images provides critical information about srectal ultrasonography in anorectal neoplasia. Br J
the tumour and its relation to key anatomical Surg. 2003;90(3):346–50.
12. Brown G, Radcliffe AG, Newcombe RG, Dallimore
landmarks, which determine the need or other- NS, Bourne MW, Williams GT. Preoperative assess-
wise for neo-adjuvant treatment, the plane and ment of prognostic factors in rectal cancer using
radicality of curative surgery, and lately, the high-resolution magnetic resonance imaging. Br J
possibility of avoiding surgery by assessing the Surg. 2003;90(3):355–64.
13. Merkel S, Mansmann U, Siassi M, Papadopoulos T,
downstaging response. Hohenberger W, Hermanek P. The prognostic inho-
Along with emerging technologies, the mogeneity in pT3 rectal carcinomas. Int J Color Dis.
improvements in speed and spatial resolution, 2001;16(5):298–304.
development of functional imaging, and the 14. Nielsen LBJ, Wille-Jørgensen P. National and inter-
national guidelines for rectal cancer. Color Dis.
recognition of novel imaging biomarkers are at 2014;16(11):854–65.
the frontiers of pre-operative rectal cancer 15. Akasu T, Kondo H, Moriya Y, Sugihara K, Gotoda
evaluation. T, Fujita S, et al. Endorectal ultrasonography and
treatment of early stage rectal cancer. World J Surg.
2000;24(9):1061–8.
16. Garcia-Aguilar J, Pollack J, Lee SH, Hernandez de
References Anda E, Mellgren A, Wong WD, et al. Accuracy of
endorectal ultrasonography in preoperative staging of
1. Wild JJ, Reid JM. Further pilot echographic rectal tumors. Dis Colon Rectum. 2002;45(1):10–5.
studies on the histologic structure of tumors 17. Marusch F, Koch A, Schmidt U, Zippel R, Kuhn R,
of the living intact human breast. Am J Pathol. Wolff S, et al. Routine use of transrectal ultrasound
1952;28(5):839–61. in rectal carcinoma: results of a prospective multi-
2. Dragsted J, Gammelgaard J. Endoluminal ultra- center study. Endoscopy. 2002;34(5):385–90.
sonic scanning in the evaluation of rectal cancer: a 18. Manger T, Stroh C. Accuracy of endorectal ultraso-
preliminary report of 13 cases. Gastrointest Radiol. nography in the preoperative staging of rectal cancer.
1983;8(1):367–9. Tech Coloproctol. 2004;8(1):s14–s5.
3. Hildebrandt U, Feifel G. Preoperative staging of 19. Kauer WK, Prantl L, Dittler HJ, Siewert JR. The
rectal cancer by intrarectal ultrasound. Dis Colon value of endosonographic rectal carcinoma stag-
Rectum. 1958;28(1):42–6. ing in routine diagnostics: a 10-year analysis. Surg
4. Beynon J, Foy DMA, Temple LN, Channer JL, Endosc. 2004;18(7):1075–8.
Virjee J, Mortensen NJMC. The endosonic appear- 20. Zammit M, Jenkins JT, Urie A, O'Dwyer PJ,
ances of normal colon and rectum. Dis Colon Molloy RG. A technically difficult endorectal ultra-
Rectum. 1986;29(12):810–3. sound is more likely to be inaccurate. Color Dis.
5. Skandarajah AR, Tjandra JJ. Preoperative loco- 2005;7(5):486–91.
regional imaging in rectal cancer. ANZ J Surg. 21. Ptok H, Marusch F, Meyer F, Wendling P, Wenisch
2006;76(6):497–504. HJ, Sendt W, et al. Feasibility and accuracy of
6. Harewood GC. Assessment of publication bias in the TRUS in the pre-treatment staging for rectal car-
reporting of EUS performance in staging rectal can- cinoma in general practice. Eur J Surg Oncol.
cer. Am J Gastroenterol. 2005;100(4):808–16. 2006;32(4):420–5.
7. Puli SR, Bechtold ML, Reddy JBK, Choudhary A, 22. Badger SA, Devlin PB, Neilly PJ, Gilliland
Antillon MR, Brugge WR. How good is endoscopic R. Preoperative staging of rectal carcinoma by
ultrasound in differentiating various T stages of endorectal ultrasound: is there a learning curve? Int
­rectal cancer? Meta-analysis and systematic review. J Color Dis. 2007;22(10):1261–8.
Ann Surg Oncol. 2008;16(2):254–65. 23. Goertz RS, Fein M, Sailer M. Impact of biopsy on
8. Bipat S, Glas AS, Slors FJ, Zwinderman AH, the accuracy of endorectal ultrasound staging of rec-
Bossuyt PM, Stoker J. Rectal cancer: local staging tal tumors. Dis Colon Rectum. 2008;51(7):1125–9.
and assessment of lymph node involvement with 24. Morris OJ, Draganic B, Smith S. Does a learning
endoluminal US, CT, and MR imaging–a meta-anal- curve exist in endorectal two-dimensional ultrasound
ysis. Radiology. 2004;232(3):773–83. accuracy? Tech Coloproctol. 2011;15(3):301–11.
9. Edelman BR, Weiser MR. Endorectal ultrasound: its 25. Marusch F, Ptok H, Sahm M, Schmidt U, Ridwelski
role in the diagnosis and treatment of rectal cancer. K, Gastinger I, et al. Endorectal ultrasound in rec-
Clin Colon Rectal Surg. 2008;21(3):167–77. tal carcinoma--do the literature results really cor-
10. Hulsmans FJ, Tio TL, Fockens P, Bosma A, Tytgat respond to the realities of routine clinical care?
GN. Assessment of tumor infiltration depth in rectal Endoscopy. 2011;43(5):425–31.
21 Rectal Carcinoma: Imaging for Staging 383

26. Ashraf S, Hompes R, Slater A, Lindsey I, Bach S, ium-based contrast material helpful? Radiology.
Mortensen NJ, et al. A critical appraisal of endorec- 2005;234(1):179–88.
tal ultrasound and transanal endoscopic microsur- 39. Zand KR, Reinhold C, Haider MA, Nakai A,
gery and decision-making in early rectal cancer. Rohoman L, Maheshwari S. Artifacts and pitfalls in
Color Dis. 2012;14(7):821–6. MR imaging of the pelvis. J Magn Reson Imaging.
27. Restivo A, Zorcolo L, Marongiu L, Scintu F, Casula 2007;26(3):480–97.
G. Limits of endorectal ultrasound in tailoring 40. Brown G, Richards CJ, Newcombe RG, Dallimore
treatment of patients with rectal cancer. Dig Surg. NS, Radcliffe AG, Carey DP, et al. Rectal carci-
2015;32(2):129–34. noma: thin-section mr imaging for staging in 28
28. Orrom WJ, Wong WD, Rothenberger DA, patients. Radiology. 1999;211(1):215–22.
Jensen LL, Goldberg SM. Endorectal ultrasound 41. Al-Sukhni E, Milot L, Fruitman M, Beyene J,
in the preoperative staging of rectal tumors. Victor JC, Schmocker S, et al. Diagnostic accuracy
A learning experience. Dis Colon Rectum. of MRI for assessment of T category, lymph node
1990;33(8):654–9. metastases, and circumferential resection margin
29. Li XT, Sun YS, Tang L, Cao K, Zhang XY. Evaluating involvement in patients with rectal cancer: a sys-
local lymph node metastasis with magnetic reso- tematic review and meta-analysis. Ann Surg Oncol.
nance imaging, endoluminal ultrasound and com- 2012;19(7):2212–23.
puted tomography in rectal cancer: a meta-analysis. 42. Hermanek P, Henson DE, Hutter RV, Sobin
Color Dis. 2015;17(6):O129–O35. LH. UICC TNM supplement: a commentary on uni-
30. Kim JC, Kim HC, Yu CS, Han KR, Kim JR, Lee form use. Berlin: Springer; 1993.
KH, et al. Efficacy of 3-dimensional endorec- 43. MERCURY Study Group. Extramural depth of
tal ultrasonography compared with conventional tumor invasion at thin-section MR in patients with
ultrasonography and computed tomography in rectal cancer: results of the MERCURY study.
preoperative rectal cancer staging. Am J Surg. Radiology. 2007;243(1):132–9.
2006;192(1):89–97. 44. Taylor FG, Quirke P, Heald RJ, Moran B,
31. Gleeson FC, Clain JE, Papachristou GI, Rajan E, Blomqvist L, Swift I, et al. Preoperative high-
Topazian MD, Wang KK, et al. Prospective assess- resolution magnetic resonance imaging can
ment of EUS criteria for lymphadenopathy asso- identify good prognosis stage I, II, and III rectal
ciated with rectal cancer. Gastrointest Endosc. cancer best managed by surgery alone: a pro-
2009;69(4):896–903. spective, multicenter, European study. Ann Surg.
32. Catalano MF, Sivak MV Jr, Rice T, Gragg LA, 2011;253(4):711–9.
Van Dam J. Endosonographic features predictive 45. Dworak O. Morphology of lymph nodes in the
of lymph node metastasis. Gastrointest Endosc. resected rectum of patients with rectal carcinoma.
1994;40(4):442–6. Pathol Res Pract. 1991;187(8):1020–4.
33. Harewood GC, Wiersema MJ, Nelson H, Maccarty 46. Vogl TJ, Pegios W, Mack MG, Hünerbein M, Hintze
RL, Olson JE, Clain JE, et al. A prospective, blinded R, Adler A, et al. Accuracy of staging rectal tumors
assessment of the impact of preoperative staging on with contrast-enhanced transrectal MR imaging. Am
the management of rectal cancer. Gastroenterology. J Roentgenol. 1997;168(6):1427–34.
2002;123(1):24–32. 47. Blomqvist L, Machado M, Rubio C, Gabrielsson N,
34. Sailer M, Leppert R, Bussen D, Fuchs KH, Thiede Granqvist S, Goldman S, et al. Rectal tumour stag-
A. Influence of tumor position on accuracy of ing: MR imaging using pelvic phased-array and
endorectal ultrasound staging. Dis Colon Rectum. endorectal coils vs endoscopic ultrasonography. Eur
1997;40(10):1180–6. Radiol. 2000;10(4):653–60.
35. Taylor FG, Quirke P, Heald RJ, Moran BJ, Blomqvist 48. Kim NK, Kim MJ, Park JK, Park SI, Min
L, Swift IR, et al. Preoperative magnetic resonance JS. Preoperative staging of rectal cancer with MRI:
imaging assessment of circumferential resection accuracy and clinical usefulness. Ann Surg Oncol.
margin predicts disease-free survival and local recur- 2000;7(10):732–7.
rence: 5-year follow-up results of the MERCURY 49. Kim JH, Beets GL, Kim M-J, Kessels AGH,
study. J Clin Oncol. 2014;32(1):34–43. Beets-Tan RGH. High-resolution MR imag-
36. Hurlstone DP, Brown S, Cross SS, Shorthouse AJ, ing for nodal staging in rectal cancer: are there
Sanders DS. Endoscopic ultrasound miniprobe stag- any criteria in addition to the size? Eur J Radiol.
ing of colorectal cancer: can management be modi- 2004;52(1):78–83.
fied? Endoscopy. 2005;37(08):710–4. 50. Brown G, Richards CJ, Bourne MW, Newcombe
37. Waage JER, Leh S, Røsler C, Pfeffer F, Bach RG, Radcliffe AG, Dallimore NS, et al. Morphologic
SP, Havre RF, et al. Endorectal ultrasonography, predictors of lymph node status in rectal cancer
strain elastography and MRI differentiation of rec- with use of high-spatial-resolution MR imag-
tal adenomas and adenocarcinomas. Color Dis. ing with histopathologic comparison. Radiology.
2015;17(2):124–31. 2003;227(2):371–7.
38. Vliegen RF, Beets GL, von Meyenfeldt MF, Kessels 51. Akasu T, Iinuma G, Takawa M, Yamamoto S,
AG, Lemaire EE, van Engelshoven JM, et al. Rectal Muramatsu Y, Moriyama N. Accuracy of high-
cancer: MR imaging in local staging--is gadolin- resolution magnetic resonance imaging in preop-
384 M. Dattani and G. Brown

erative staging of rectal cancer. Ann Surg Oncol. 64. Glimelius B, Beets-Tan R, Blomqvist L, Brown
2009;16(10):2787–94. G, Nagtegaal I, Pahlman L, et al. Mesorectal fas-
52. Halefoglu AM, Yildirim S, Avlanmis O, Sakiz cia instead of circumferential resection margin in
D, Baykan A. Endorectal ultrasonography ver- preoperative staging of rectal cancer. J Clin Oncol.
sus phased-array magnetic resonance imaging 2011;29(16):2142–3.
for preoperative staging of rectal cancer. World J 65. Park MJ, Kim SH, Lee SJ, Jang KM, Rhim
Gastroenterol. 2008;14(22):3504–10. H. Locally advanced rectal cancer: added value
53. Koh D-M, Brown G, Temple L, Raja A, Toomey P, of diffusion-weighted MR imaging for predict-
Bett N, et al. Rectal cancer: mesorectal lymph nodes ing tumor clearance of the mesorectal fascia after
at MR imaging with USPIO versus histopatho- neoadjuvant chemotherapy and radiation therapy.
logic findings—initial observations. Radiology. Radiology. 2011;260(3):771–80.
2004;231(1):91–9. 66. Beets-Tan RG, Beets GL, Vliegen RF, Kessels AG,
54. Koh DM, George C, Temple L, Collins DJ, Toomey P, Van Boven H, De Bruine A, et al. Accuracy of mag-
Raja A, et al. Diagnostic accuracy of nodal enhance- netic resonance imaging in prediction of tumour-free
ment pattern of rectal cancer at MRI enhanced with resection margin in rectal cancer surgery. Lancet.
ultrasmall superparamagnetic iron oxide: findings 2001;357(9255):497–504.
in pathologically matched mesorectal lymph nodes. 67. Xie H, Zhou X, Zhuo Z, Che S, Xie L, Fu
AJR Am J Roentgenol. 2010;194(6):W505–13. W. Effectiveness of MRI for the assessment of
55. Lahaye MJ, Engelen SME, Kessels AGH, Bruïne mesorectal fascia involvement in patients with rectal
APD, MFv M, Engelshoven JMAV, et al. USPIO- cancer: a systematic review and meta-analysis. Dig
enhanced MR imaging for nodal staging in patients Surg. 2014;31(2):123–34.
with primary rectal cancer: predictive criteria. 68. Taylor FGM, Quirke P, Heald RJ, Moran B,
Radiology. 2008;246(3):804–11. Blomqvist L, Swift I, et al. One millimetre is the
56. Chand M, Moran BJ, Jones RG, Heald RJ, Brown safe cut-off for magnetic resonance imaging predic-
G. Lymph node status does not predict local recur- tion of surgical margin status in rectal cancer. Br J
rence in the total mesorectal excision era. Dis Colon Surg. 2011;98(6):872–9.
Rectum. 2014;57(1):127–9. 69. Group MS. Diagnostic accuracy of preoperative
57. Kosinski LA, Greene FL. Perfect treatment in an magnetic resonance imaging in predicting curative
imperfect world: surgery alone or radiation for resection of rectal cancer: prospective observational
node positive rectal cancer? Dis Colon Rectum. study. Br Med J. 2006;333(7572):779.
2014;57(1):130–2. 70. Moriya Y. Treatment of lateral pelvic nodes metas-
58. Quirke P, Steele R, Monson J, Grieve R, Khanna tases from rectal cancer: the future prospective. G
S, Couture J, et al. Effect of the plane of surgery Chir. 2013;34(9-10):245–8.
achieved on local recurrence in patients with opera- 71. Bell S, Sasaki J, Sinclair G, Chapuis PH, Bokey
ble rectal cancer: a prospective study using data from EL. Understanding the anatomy of lymphatic drain-
the MRC CR07 and NCIC-CTG CO16 randomised age and the use of blue-dye mapping to determine the
clinical trial. Lancet. 2009;373(9666):821–8. extent of lymphadenectomy in rectal cancer surgery:
59. Quirke P, Durdey P, Dixon MF, Williams NS. Local unresolved issues. Color Dis. 2009;11(5):443–9.
recurrence of rectal adenocarcinoma due to inad- 72. Yano H, Moran BJ. The incidence of lateral pelvic
equate surgical resection. Histopathological study of side-wall nodal involvement in low rectal cancer
lateral tumour spread and surgical excision. Lancet. may be similar in Japan and the West. Br J Surg.
1986;2(8514):996–9. 2008;95(1):33–49.
60. Wibe A, Rendedal PR, Svensson E, Norstein J, Eide 73. Sugihara K, Kobayashi H, Kato T, Mori T, Mochizuki
TJ, Myrvold HE, et al. Prognostic significance of H, Kameoka S, et al. Indication and benefit of pel-
the circumferential resection margin following total vic sidewall dissection for rectal cancer. Dis Colon
mesorectal excision for rectal cancer. Br J Surg. Rectum. 2006;49(11):1663–72.
2002;89(3):327–34. 74. Fujita S, Akasu T, Mizusawa J, Saito N, Kinugasa
61. Nagtegaal ID, Marijnen CA, Kranenbarg EK, van de Y, Kanemitsu Y, et al. Postoperative morbid-
Velde CJ, van Krieken JH. Circumferential margin ity and mortality after mesorectal excision with
involvement is still an important predictor of local and without lateral lymph node dissection for
recurrence in rectal carcinoma: not one millimeter clinical stage II or stage III lower rectal cancer
but two millimeters is the limit. Am J Surg Pathol. (JCOG0212): results from a multicentre, ran-
2002;26(3):350–7. domised controlled, non-inferiority trial. Lancet
62. Birbeck KF, Macklin CP, Tiffin NJ, Parsons W, Oncol. 2012;13(6):616–21.
Dixon MF, Mapstone NP, et al. Rates of circumfer- 75. Georgiou P, Tan E, Gouvas N, Antoniou A,
ential resection margin involvement vary between Brown G, Nicholls RJ, et al. Extended lymph-
surgeons and predict outcomes in rectal cancer sur- adenectomy versus conventional surgery for
gery. Ann Surg. 2002;235(4):449–57. rectal cancer: a meta-analysis. Lancet Oncol.
63. Nagtegaal ID, Quirke P. What is the role for the cir- 2009;10(11):1053–62.
cumferential margin in the modern treatment of rectal 76. Kim TH, Jeong S-Y, Choi DH, Kim DY, Jung KH,
cancer? J Clin Oncol. 2008;26(2):303–12. Moon SH, et al. Lateral lymph node metastasis is
21 Rectal Carcinoma: Imaging for Staging 385

a major cause of locoregional recurrence in rec- of local peritoneal involvement and extramural
tal cancer treated with preoperative chemoradio- venous invasion in colonic cancer. Histopathology.
therapy and curative resection. Ann Surg Oncol. 2009;55(4):407–13.
2007;15(3):729–37. 91. Messenger DE, Driman DK, Kirsch R.
77. Otowa Y, Yamashita K, Kanemitsu K, Sumi Y, Developments in the assessment of venous inva-
Yamamoto M, Kanaji S, et al. Treating patients sion in colorectal cancer: implications for future
with advanced rectal cancer and lateral pelvic practice and patient outcome. Hum Pathol.
lymph nodes with preoperative chemoradiotherapy 2012;43(7):965–73.
based on pretreatment imaging. OncoTargets Ther. 92. Messenger DE, Driman DK, McLeod RS, Riddell
2015;8:3169–73. RH, Kirsch R. Current practice patterns among
78. Shihab OC, Taylor F, Bees N, Blake H, Jeyadevan pathologists in the assessment of venous invasion
N, Bleehen R, et al. Relevance of magnetic reso- in colorectal cancer. J Clin Pathol. 2011;64(11):
nance imaging-detected pelvic sidewall lymph 983–9.
node involvement in rectal cancer. Br J Surg. 93. Sternberg A, Amar M, Alfici R, Groisman G.
2011;98(12):1798–804. Conclusions from a study of venous invasion in
79. Freedman LS, Macaskill P, Smith AN. Multivariate stage IV colorectal adenocarcinoma. J Clin Pathol.
analysis of prognostic factors for operable rectal 2002;55(1):17–21.
cancer. Lancet. 1984;2(8405):733–6. 94. Smith NJ, Shihab O, Arnaout A, Swift RI. Brown
80. Horn A, Dahl O, Morild I. Venous and neural inva- G. MRI for detection of extramural vascular inva-
sion as predictors of recurrence in rectal adenocarci- sion in rectal cancer. AJR Am J Roentgenol.
noma. Dis Colon Rectum. 1991;34(9):798–804. 2008;191(5):1517–22.
81. Talbot IC, Ritchie S, Leighton MH, Hughes AO, 95. Smith NJ, Barbachano Y, Norman AR, Swift RI,
Bussey HJ, Morson BC. The clinical significance Abulafi AM, Brown G. Prognostic significance of
of invasion of veins by rectal cancer. Br J Surg. magnetic resonance imaging-detected extramu-
1980;67(6):439–42. ral vascular invasion in rectal cancer. Br J Surg.
82. Thomson E, Scott N, Tolan D. Re: the prog- 2008;95(2):229–36.
nostic significance of MRI-detected extramural 96. Jhaveri KS, Hosseini-Nik H, Thipphavong S,
venous invasion in rectal carcinoma. Clin Radiol. Assarzadegan N, Menezes RJ, Kennedy ED, et al.
2015;70(1):111–2. MRI detection of extramural venous invasion in
83. Bokey EL, Öjerskog B, Chapuis PH, Dent OF, rectal cancer: correlation with histopathology using
Newland RC, Sinclair G. Local recurrence after elastin stain. Am J Roentgenol. 2016;206:1–9.
curative excision of the rectum for cancer without 97. Sohn B, J-s L, Kim H, Myoung S, Choi J, Kim NK,
adjuvant therapy: role of total anatomical dissection. et al. MRI-detected extramural vascular invasion is
Br J Surg. 1999;86(9):1164–70. an independent prognostic factor for synchronous
84. Rich T, Gunderson LL, Lew R, Galdibini JJ, metastasis in patients with rectal cancer. Eur Radiol.
Cohen AM, Donaldson G. Patterns of recurrence 2014;25(5):1347–55.
of rectal cancer after potentially curative surgery. 98. Bugg WG, Andreou AK, Biswas D, Toms AP,
Cancer. 1983;52(7):1317–29. Williams SM. The prognostic significance of
85. Courtney ED, West NJ, Kaur C, Ho J, Kalber B, ­MRI-detected extramural venous invasion in rectal
Hagger R, et al. Extramural vascular invasion is an carcinoma. Clin Radiol. 2014;69(6):619–23.
adverse prognostic indicator of survival in patients 99. Nilsson PJ, van Etten B, Hospers GAP, Påhlman
with colorectal cancer. Color Dis. 2009;11(2):150–6. L, van de Velde CJH, Beets-Tan RGH, et al. Short-
86. Betge J, Pollheimer MJ, Lindtner RA, Kornprat P, course radiotherapy followed by neo-adjuvant che-
Schlemmer A, Rehak P, et al. Intramural and extra- motherapy in locally advanced rectal cancer – the
mural vascular invasion in colorectal cancer. Cancer. RAPIDO trial. BMC Cancer. 2013;13:279.
2012;118(3):628–38. 100. Salerno G, Sinnatamby C, Branagan G, Daniels IR,
87. Petersen VC, Baxter KJ, Love SB, Shepherd Heald RJ, Moran BJ. Defining the rectum: surgi-
NA. Identification of objective pathological prog- cally, radiologically and anatomically. Color Dis.
nostic determinants and models of prognosis in 2006;8:5–9.
Dukes’ B colon cancer. Gut. 2002;51(1):65–9. 101. Moran BJ, Holm T, Brannagan G, Chave H,
88. Chand M, Bhangu A, Wotherspoon A, Stamp GWH, Quirke P, West N, et al. The English National
Swift RI, Chau I, et al. EMVI-positive stage II rec- Low Rectal Cancer Development Programme:
tal cancer has similar clinical outcomes as stage III key messages and future perspectives. Color Dis.
disease following pre-operative chemoradiotherapy. 2014;16(3):173–8.
Ann Oncol. 2014;25(4):858–63. 102. Shihab OC, Moran BJ, Heald RJ, Quirke P, Brown
89. Chand M, Siddiqui MRS, Swift I, Brown G. MRI staging of low rectal cancer. Eur Radiol.
G. Systematic review of prognostic importance of 2009;19(3):643–50.
extramural venous invasion in rectal cancer. World J 103. Battersby NJ, How P, Moran B, Stelzner S, West NP,
Gastroenterol. 2016;22(4):1721–6. Branagan G, et al. Prospective validation of a low
90. Littleford SE, Baird A, Rotimi O, Verbeke CS, rectal cancer magnetic resonance imaging staging
Scott N. Interobserver variation in the reporting system and development of a local recurrence risk
386 M. Dattani and G. Brown

stratification model: The MERCURY II study. Ann 117. Balyasnikova S, Brown G. Imaging advances in
Surg. 2016;263(4):751–60. colorectal cancer. Curr Colorectal Cancer Rep.
104. Marr R, Birbeck K, Garvican J, Macklin CP, Tiffin 2016;12:162–9.
NJ, Parsons WJ, et al. The modern abdominoperi- 118. Sebag-Montefiore D, Stephens RJ, Steele R, Monson
neal excision: the next challenge after total mesorec- J, Grieve R, Khanna S, et al. Preoperative radiother-
tal excision. Ann Surg. 2005;242(1):74–82. apy versus selective postoperative chemoradiother-
105. den Dulk M, Putter H, Collette L, Marijnen CA, apy in patients with rectal cancer (MRC CR07 and
Folkesson J, Bosset JF, et al. The abdominoperi- NCIC-CTG C016): a multicentre, randomised trial.
neal resection itself is associated with an adverse Lancet. 2009;373(9666):811–20.
outcome: the European experience based on a 119. van Gijn W, Marijnen CA, Nagtegaal ID, Kranenbarg
pooled analysis of five European randomised EM, Putter H, Wiggers T, et al. Preoperative radio-
clinical trials on rectal cancer. Eur J Cancer. therapy combined with total mesorectal excision
2009;45(7):1175–83. for resectable rectal cancer: 12-year follow-up of
106. Nagtegaal ID, van de Velde CJ, Marijnen CA, van the multicentre, randomised controlled TME trial.
Krieken JH, Quirke P. Low rectal cancer: a call for a Lancet Oncol. 2011;12(6):575–82.
change of approach in abdominoperineal resection. J 120. Bosset J-F, Collette L, Calais G, Mineur L, Maingon
Clin Oncol. 2005;23(36):9257–64. P, Radosevic-Jelic L, et al. Chemotherapy with pre-
107. Eriksen MT, Wibe A, Syse A, Haffner J, Wiig operative radiotherapy in rectal cancer. N Engl J
JN. Inadvertent perforation during rectal cancer Med. 2006;355(11):1114–23.
resection in Norway. Br J Surg. 2004;91(2):210–6. 121. Gérard J-P, Conroy T, Bonnetain F, Bouché O,
108. Holm T, Ljung A, Häggmark T, Jurell G, Lagergren Chapet O, Closon-Dejardin M-T, et al. Preoperative
J. Extended abdominoperineal resection with glu- radiotherapy with or without concurrent fluo-
teus maximus flap reconstruction of the pelvic floor rouracil and leucovorin in T3-4 rectal cancers:
for rectal cancer. Br J Surg. 2007;94(2):232–8. results of FFCD 9203. J Clin Oncol. 2006;24(28):
109. Foster JD, Pathak S, Smart NJ, Branagan G, 4620–5.
Longman RJ, Thomas MG, et al. Reconstruction of 122. Battersby NJ, Juul T, Christensen P, Janjua AZ,
the perineum following extralevator abdominoperi- Branagan G, Emmertsen KJ, et al. Predicting the
neal excision for carcinoma of the lower rectum: a risk of bowel-related quality-of-life impairment
systematic review. Color Dis. 2012;14(9):1052–9. after restorative resection for rectal cancer: a mul-
110. West NP, Anderin C, Smith KJ, Holm T, Quirke ticenter cross-sectional study. Dis Colon Rectum.
P. Multicentre experience with extralevator abdomi- 2016;59(4):270–80.
noperineal excision for low rectal cancer. Br J Surg. 123. Marijnen CA, Kapiteijn E, van de Velde CJ, Martijn
2010;97(4):588–99. H, Steup WH, Wiggers T, et al. Acute side effects
111. Salerno G, Daniels I, Heald RJ, Brown G, Moran and complications after short-term preoperative
BJ. Management and imaging of low rectal carci- radiotherapy combined with total mesorectal exci-
noma. Surg Oncol. 2004;13(2–3):55–61. sion in primary rectal cancer: report of a multicenter
112. Shihab OC, Heald RJ, Rullier E, Brown G, Holm randomized trial. J Clin. 2002;20(3):817–25.
T, Quirke P, et al. Defining the surgical planes on 124. Birgisson H, Pahlman L, Gunnarsson U, Glimelius
MRI improves surgery for cancer of the low rectum. B. Occurrence of second cancers in patients treated
Lancet Oncol. 2009;10(12):1207–11. with radiotherapy for rectal cancer. J Clin Oncol.
113. Salerno GV, Daniels IR, Moran BJ, Heald RJ, 2005;23(25):6126–31.
Thomas K, Brown G. Magnetic resonance imag- 125. Morris EJ, Finan PJ, Spencer K, Geh I, Crellin A,
ing prediction of an involved surgical resection Quirke P, et al. Wide variation in the use of radio-
margin in low rectal cancer. Dis Colon Rectum. therapy in the management of surgically treated
2009;52(4):632–9. rectal cancer across the English National Health
114. Beyond TME Collaborative. Consensus state- Service. Clin Oncol. 2016;22:522–31.
ment on the multidisciplinary management of 126. Fitzgerald TL, Zervos E, Wong JH. Patterns of pel-
patients with recurrent and primary rectal cancer vic radiotherapy in patients with stage II/III rectal
beyond total mesorectal excision planes. Br J Surg. cancer. J Cancer Epidemiol. 2013;2013:408460.
2013;100(8):1009–14. 127. van Leersum NJ, Snijders HS, Wouters MWJM,
115. Bhangu A, Ali SM, Brown G, Nicholls RJ, Tekkis Henneman D, Marijnen CAM, Rutten HR, et al.
P. Indications and outcome of pelvic exenteration for Evaluating national practice of preoperative radio-
locally advanced primary and recurrent rectal can- therapy for rectal cancer based on clinical auditing.
cer. Ann Surg. 2014;259(2):315–22. Eur J Surg Oncol. 2013;39(9):1000–6.
116. Georgiou PA, Tekkis PP, Constantinides VA, Patel 128. Gunderson LL, Sargent DJ, Tepper JE, Wolmark N,
U, Goldin RD, Darzi AW, et al. Diagnostic accuracy O'Connell MJ, Begovic M, et al. Impact of T and N
and value of magnetic resonance imaging (MRI) in stage and treatment on survival and relapse in adju-
planning exenterative pelvic surgery for advanced vant rectal cancer: a pooled analysis. J Clin Oncol.
colorectal cancer. Eur J Cancer. 2013;49(1):72–81. 2004;22(10):1785–96.
21 Rectal Carcinoma: Imaging for Staging 387

129. Lai LL, Fuller CD, Kachnic LA, Thomas CR Jr. 144. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz
Can pelvic radiotherapy be omitted in select patients LH, Sargent D, Ford R, et al. New response
with rectal cancer? Semin Oncol. 2006;33(6 Suppl evaluation criteria in solid tumours: revised
11):S70–4. RECIST guideline (version 1.1). Eur J Cancer.
130. Glynne-Jones R, Tan D, Goh V. Pelvic MRI for guid- 2009;45(2):228–47.
ing treatment decisions in rectal cancer. Oncology. 145. Evans J, Patel U, Brown G. Rectal cancer: primary
2014;28(8):667–77. staging and assessment after chemoradiotherapy.
131. Benson AB, Bekaii-Saab T, Chan E, Chen Y-J, Choti Semin Radiat Oncol. 2011;21(3):169–77.
MA, Cooper HS, et al. Rectal cancer. J Natl Compr 146. Minocha J, Lewandowski RJ. Assessing imag-
Cancer Netw. 2012;10(12):1528–64. ing response to therapy. Radiol Clin N Am.
132. Guillem JG, Díaz-González JA, Minsky BD, 2015;53(5):1077–88.
Valentini V, Jeong S-Y, Rodriguez-Bigas MA, et al. 147. Zhao RS, Wang H, Zhou ZY, Zhou Q, Mulholland
cT3N0 rectal cancer: potential overtreatment with MW. Restaging of locally advanced rectal cancer
preoperative chemoradiotherapy is warranted. J Clin with magnetic resonance imaging and endoluminal
Oncol. 2008;26(3):368–73. ultrasound after preoperative chemoradiotherapy:
133. Sautter-Bihl M-L, Hohenberger W, Fietkau R, a systemic review and meta-analysis. Dis Colon
Roedel C, Schmidberger H, Sauer R. MRI-based Rectum. 2014;57(3):388–95.
treatment of rectal cancer: is prognostication of 148. de Jong EA, ten Berge JCEM, Dwarkasing RS,
the recurrence risk solid enough to render radiation Rijkers AP, van Eijck CHJ. The accuracy of MRI,
redundant? Ann Surg Oncol. 2014;21(1):197–204. endorectal ultrasonography, and computed tomogra-
134. Smith N, Brown G. Preoperative staging of rectal phy in predicting the response of locally advanced
cancer. Acta Oncol. 2008;47(1):20–31. rectal cancer after preoperative therapy: a metaanal-
135. Blomqvist L, Glimelius B. The ‘good’, the ‘bad’, and ysis. Surgery. 2016;159(3):688–99.
the ‘ugly’ rectal cancers. Acta Oncol. 2008;47(1):5–8. 149. Pierredon-Foulongne MA, Nougaret S, Bibeau F,
136. Colorectal cancer: diagnosis and management Rouanet P, Delhom E, Lonjon J, et al. Utility of
[CG131]. National Institute for Health and Clinical reassessment after neoadjuvant therapy and dif-
Excellence. 2011(November). ficulties in interpretation. Diagn Interv Imaging.
137. Glimelius B, Tiret E, Cervantes A, Arnold D, Group 2014;95(5):495–503.
obotEGW. Rectal cancer: ESMO clinical practice 150. Tanaka S, Martling A, Lindholm J, Holm T, Palmer
guidelines for diagnosis, treatment and follow-up. G. Remaining cancer cells within the fibrosis
Ann Oncol. 2013;24(suppl 6):vi81–8. after neo-adjuvant treatment for locally advanced
138. Valentini V, Glimelius B, Haustermans K, rectal cancer. Eur J Surg Oncol. 2015;41(9):
Marijnen CAM, Rödel C, Gambacorta MA, et al. 1204–9.
EURECCA consensus conference highlights about 151. Chand M, Evans J, Swift RI, Tekkis PP, West NP,
rectal cancer clinical management: The radia- Stamp G, et al. The prognostic significance of post-
tion oncologist’s expert review. Radiother Oncol. chemoradiotherapy high-resolution MRI and histo-
2014;110(1):195–8. pathology detected extramural venous invasion in
139. Glimelius B. Optimal time intervals between pre- rectal cancer. Ann Surg. 2015;261(3):473–9.
operative radiotherapy or chemoradiotherapy and 152. Chand M, Swift RI, Tekkis PP, Chau I, Brown
surgery in rectal cancer? Front Oncol. 2014;4:50. G. Extramural venous invasion is a potential imag-
140. Memon S, Lynch AC, Bressel M, Wise AG, Heriot ing predictive biomarker of neoadjuvant treatment in
AG. Systematic review and meta-analysis of the rectal cancer. Br J Cancer. 2014;110(1):19–25.
accuracy of MRI and endorectal ultrasound in the 153. Dattani M, Moran BJ. Watch and wait after neo-
restaging and response assessment of rectal can- adjuvant therapy for rectal cancer. Br J Surg.
cer following neoadjuvant therapy. Color Dis. 2016;103:629–31.
2015;17(9):748–61. 154. Habr-Gama A, Perez RO, Nadalin W, Sabbaga J,
141. Hanly AM, Ryan EM, Rogers AC, McNamara Ribeiro U, Silva e Sousa AH, et al. Operative ver-
DA, Madoff RD, Winter DC. Multicenter evalu- sus nonoperative treatment for stage 0 distal rectal
ation of rectal cancer reimaging post neo- cancer following chemoradiation therapy: long-term
adjuvant (MERRION) therapy. Ann Surg. results. Ann Surg. 2004;240(4):711–8.
2014;259(4):723–7. 155. Smith JD, Ruby JA, Goodman KA, Saltz LB,
142. Glynne-Jones R, Wallace M, Livingstone JIL, Guillem JG, Weiser MR, et al. Nonoperative man-
Meyrick-Thomas J. Complete clinical response after agement of rectal cancer with complete clinical
preoperative chemoradiation in rectal cancer: is a response after neoadjuvant therapy. Ann Surg.
“wait and see” policy justified? Dis Colon Rectum. 2012;256(6):965–72.
2007;51(1):10–20. 156. Renehan AG, Malcomson L, Emsley R, Gollins S,
143. Marijnen CAM. Organ preservation in rectal cancer: Maw A, Myint AS, et al. Watch-and-wait approach
have all questions been answered? Lancet Oncol. versus surgical resection after chemoradiotherapy
2015;16(1):e13–22. for patients with rectal cancer (the OnCoRe project):
388 M. Dattani and G. Brown

a propensity-score matched cohort analysis. Lancet gery in combination with hyperthermic intraperito-
Oncol. 2015;17:174–83. neal chemotherapy improves survival in patients
157. Appelt AL, Pløen J, Harling H, Jensen FS, Jensen with colorectal peritoneal metastases compared
LH, Jørgensen JCR, et al. High-dose chemoradio- with systemic chemotherapy alone. Br J Cancer.
therapy and watchful waiting for distal rectal cancer: 2014;111(8):1500–8.
a prospective observational study. Lancet Oncol. 168. Niekel MC, Bipat S, Stoker J. Diagnostic imaging
2015;16(8):919–27. of colorectal liver metastases with CT, MR imaging,
158. Martens MH, Maas M, Heijnen LA, Lambregts DM, FDG PET, and/or FDG PET/CT: a meta-analysis
Leijtens JW, Stassen LP, et al. Long-term outcome of prospective studies including patients who have
of an organ preservation program after neoadju- not previously undergone treatment. Radiology.
vant treatment for rectal cancer. J Natl Cancer Inst. 2010;257(3):674–84.
2016;108(12):djw171. 169. Knowles B, Welsh FK, Chandrakumaran K, John TG,
159. Maas M, Lambregts DMJ, Nelemans PJ, Heijnen Rees M. Detailed liver-specific imaging prior to pre-
LA, Martens MH, Leijtens JWA, et al. Assessment operative chemotherapy for colorectal liver metasta-
of clinical complete response after chemo- ses reduces intra-hepatic recurrence and the need for
radiation for rectal cancer with digital rectal a repeat hepatectomy. HPB. 2012;14(5):298–309.
examination, endoscopy, and MRI: selection for organ- 170. Dewhurst C, Rosen MP, Blake MA, Baker ME, Cash
saving treatment. Ann Surg Oncol. 2015;22(12): BD, Fidler JL, et al. ACR appropriateness criteria
3873–80. pretreatment staging of colorectal cancer. J Am Coll
160. Bhoday J, Smith F, Siddiqui MR, Balyasnikova S, Radiol. 2012;9(11):775–81.
Swift RI, Perez R, et al. Magnetic resonance tumor 171. Vatandoust S, Price TJ, Karapetis CS. Colorectal
regression grade and residual mucosal abnormality cancer: metastases to a single organ. World J
as predictors for pathological complete response in Gastroenterol. 2015;21(41):11767–76.
rectal cancer postneoadjuvant chemoradiotherapy. 172. Nordholm-Carstensen A, Wille-Jorgensen PA,
Dis Colon Rectum. 2016;59(10):925–33. Jorgensen LN, Harling H. Indeterminate pulmonary
161. Mandard AM, Dalibard F, Mandard JC, Marnay nodules at colorectal cancer staging: a systematic
J, Henry-Amar M, Petiot JF, et al. Pathologic review of predictive parameters for malignancy. Ann
assessment of tumor regression after preop- Surg Oncol. 2013;20(12):4022–30.
erative chemoradiotherapy of esophageal carci- 173. Bamba Y, Itabashi M, Kameoka S. Value of PET/
noma. Clinicopathologic correlations. Cancer. CT imaging for diagnosing pulmonary metasta-
1994;73(11):2680–6. sis of colorectal cancer. Hepato-Gastroenterology.
162. Patel UB, Taylor F, Blomqvist L, George C, Evans 2011;58(112):1972–4.
H, Tekkis P, et al. Magnetic resonance imaging– 174. Koh J-L, Yan TD, Glenn D, Morris DL. Evaluation
detected tumor response for locally advanced rec- of preoperative computed tomography in esti-
tal cancer predicts survival outcomes: MERCURY mating peritoneal cancer index in colorectal
experience. J Clin Oncol. 2011;29(28):3753–60. peritoneal carcinomatosis. Ann Surg Oncol.
163. Joye I, Deroose CM, Vandecaveye V, Haustermans 2009;16(2):327–33.
K. The role of diffusion-weighted MRI and (18) 175. Esquivel J, Chua TC, Stojadinovic A, Melero JT,
F-FDG PET/CT in the prediction of pathologic Levine EA, Gutman M, et al. Accuracy and clinical
complete response after radiochemotherapy for rec- relevance of computed tomography scan interpre-
tal cancer: a systematic review. Radiother Oncol. tation of peritoneal cancer index in colorectal can-
2014;113(2):158–65. cer peritoneal carcinomatosis: a multi-institutional
164. Cunningham D, Atkin W, Lenz HJ, Lynch HT, study. J Surg Oncol. 2010;102(6):565–70.
Minsky B, Nordlinger B, et al. Colorectal cancer. 176. Iversen LH, Rasmussen PC, Laurberg S. Value
Lancet. 2010;375(9719):1030–47. of laparoscopy before cytoreductive surgery
165. Mantke R, Schmidt U, Wolff S, Kube R, Lippert and ­ hyperthermic intraperitoneal chemother-
H. Incidence of synchronous liver metastases in apy for peritoneal carcinomatosis. Br J Surg.
patients with colorectal cancer in relationship to 2013;100(2):285–92.
clinico-pathologic characteristics. Results of a 177. Bull AD, Biffin AH, Mella J, Radcliffe AG,
German prospective multicentre observational study. Stamatakis JD, Steele RJ, et al. Colorectal cancer
Eur J Surg Oncol. 2012;38(3):259–65. pathology reporting: a regional audit. J Clin Pathol.
166. Mitry E, Guiu B, Cosconea S, Jooste V, Faivre 1997;50(2):138–42.
J, Bouvier AM. Epidemiology, management and 178. Taylor F, Mangat N, Swift IR, Brown G. Proforma-
prognosis of colorectal cancer with lung metas- based reporting in rectal cancer. Cancer Imaging.
tases: a 30-year population-based study. Gut. 2010;10:S142–50.
2010;59(10):1383–8. 179. Spiegle G, Leon-Carlyle M, Schmocker S, Fruitman
167. Mirnezami R, Mehta AM, Chandrakumaran K, M, Milot L, Gagliardi AR, et al. Development of
Cecil T, Moran BJ, Carr N, et al. Cytoreductive sur-
21 Rectal Carcinoma: Imaging for Staging 389

a synoptic MRI report for primary rectal cancer. 181. National Accreditation Program for Rectal Cancer,
Implement Sci. 2009;4:79. American College of Surgeons, https://www.facs.
180. Kennedy ED, Milot L, Fruitman M, Al-Sukhni org/quality-programs/cancer/naprc.
E, Heine G, Schmocker S, et al. Development 182. ACR Catalog, Rectal Cancer Staging, https://
and implementation of a synoptic MRI report shop.acr.org/Default.aspx?TabID=55&Product
for preoperative staging of rectal cancer on Id=1753677070.
a population-based level. Dis Colon Rectum.
2014;57(6):700–8.

You might also like