You are on page 1of 18

Seminar

Colorectal cancer
David Cunningham, Wendy Atkin, Heinz-Josef Lenz, Henry T Lynch, Bruce Minsky, Bernard Nordlinger, Naureen Starling
Lancet 2010; 375: 103047
Gastrointestinal Unit, Royal
Marsden Hospital National
Health Service Foundation
Trust, London and Surrey, UK
(Prof D Cunningham MD,
N Starling MRCP); Department
of Surgery and Cancer, Imperial
College, London, UK
(Prof W Atkin PhD); Sharon
Carpenter Laboratory,
University of Southern
California/Norris
Comprehensive Cancer Center,
Keck School of Medicine, Los
Angeles, CA, USA
(Prof H-J Lenz MD); Department
of Preventive Medicine and
Public Health, Creighton
University School of Medicine,
Omaha, NE, USA
(Prof H T Lynch MD); University
of Chicago Medical Centre,
Chicago, IL, USA
(Prof B Minsky MD); and Service
de Chirurgie Gnrale et
Digestive, Hpital Ambroise
Par, Boulogne, France
(Prof B Nordlinger MD)
Correspondence to:
Prof David Cunningham,
Department of Medicine, Royal
Marsden Hospital, Downs Road,
Sutton SM2 5PT, UK
David.cunningham@rmh.
nhs.uk

Substantial progress has been made in colorectal cancer in the past decade. Screening, used to identify individuals
at an early stage, has improved outcome. There is greater understanding of the genetic basis of inherited colorectal
cancer and identication of patients at risk. Optimisation of surgery for patients with localised disease has had a
major eect on survival at 5 years and 10 years. For rectal cancer, identication of patients at greatest risk of local
failure is important in the selection of patients for preoperative chemoradiation, a strategy proven to improve
outcomes in these patients. Stringent postoperative follow-up helps the early identication of potentially radically
treatable oligometastatic disease and improves long-term survival. Treatment with adjuvant uoropyrimidine for
colon and rectal cancers further improves survival, more so in stage III than in stage II disease, and oxaliplatinbased combination chemotherapy is now routinely used for stage III disease, although ecacy must be carefully
balanced against toxicity. In stage II disease, molecular markers such as microsatellite instability might help
select patients for treatment. The integration of targeted treatments with conventional cytotoxic drugs has
expanded the treatment of metastatic disease resulting in incremental survival gains. However, biomarker
development is essential to aid selection of patients likely to respond to therapy, thereby rationalising treatments
and improving outcomes.

Introduction
Worldwide, every year, more than 1 million individuals
will develop colorectal cancer,1 and the disease-specic
mortality rate is nearly 33% in the developed world. Here
we summarise some of the important developments and
advances in molecular carcinogenesis, prognostic and
predictive molecular markers, hereditary predispositions,
screening, diagnosis, and treatment during the past
5 years.

Molecular carcinogenesis
The classic description of colorectal carcinogenesis is the
adenoma-carcinoma sequence and multistep tumourigenesis that is determined by gatekeeper and caretaker
molecular pathways, which takes years to decades.2
Colorectal cancer is increasingly classied into specic
phenotypes on the basis of molecular proles (table 1), two
of which represent genetic instability classes. Most sporadic
cases (about 85%) have chromosomal instability, an allelic
imbalance at several chromosomal loci (including 5q, 8p,
17p, and 18q), and chromosome amplication and
translocation, which together contribute to tumour
aneuploidy.47 By contrast, the remaining cases (about 15%)
have high-frequency microsatellite instability phenotypes

Search strategy and selection criteria


We searched Medline, PubMed, ASCO abstracts, ESMO abstracts, and the Cochrane
database, between 2004 and 2009, for papers published in English. We used the search
terms colorectal cancer, colon cancer, rectal cancer, carcinogenesis,
epidemiology, genetics, screening, diagnosis, staging, surgery, radiation
therapy, chemotherapy (adjuvant, metastatic, and neoadjuvant), and molecular
prognostic and predictive markers. We cross-checked reference lists we found, and asked
other colleagues to recommend references. We selected papers on the basis that they
provided a major contribution to colorectal cancer or drew attention to evolving ideas,
particularly papers in the past 5 years since publication of the previous seminar about
colorectal cancer published in The Lancet in 2005.

1030

ie, frameshift mutations and base-pair substitutions that


commonly arise in short tandemly repeated nucleotide
sequences (microsatellites).8,9 Microsatellite instability is a
measure of the inability of the DNA nucleotide mismatchrepair system to correct errors that often occur during
DNA replication, which is controlled by several genes
(including MLH1, MSH2, and MSH6), and is characterised
by the accumulation of single nucleotide mutations and
length alterations in repetitive microsatellite nucleotide
sequences that are common throughout the genome.10
These tumours are characterised by proximal location,
mucinous histology, poor dierentiation, and lymphocytic
inltration. The genetic mechanism that contributes to
this phenotype is mutation or loss of function through
epigenetic gene silencing of DNA mismatch-repair
genes.11,12 In most sporadic cases, microsatellite instability
occurs when the promoter region of the genes in the
mismatch-repair system (often MLH1) is silenced by hypermethylation of CpG islands.12 The analysis of methylation
of CpG islands as a mechanism of silencing genes in colon
tumours has resulted in the identication of the CpG
island methylator phenotype, which seems to be complex,
and its prognostic signicance in patients with colon
cancer has not been thoroughly investigated.13,14
The serrated pathway, often arising in a serrated
precursor lesion (pathological classication is controversial but includes hyperplastic polyp, sessile serrated
polyp, and serrated adenoma), usually occurs in the right
colon,15 and seems to be governed by progression of
dierent molecular mechanisms to the classic adenomacarcinoma sequence.3 Clinical management of these
precursor lesions is not clear.

Epidemiology
Risk factors
Most cases of colorectal cancer arise sporadically. Risk
factors include increasing age, male sex, previous
colonic polyps, or previous colorectal cancer, and
www.thelancet.com Vol 375 March 20, 2010

Seminar

environmental factors (eg, red meat, high-fat diet,


inadequate intake of bre, obesity, sedentary lifestyle,
diabetes mellitus, smoking, and high consumption of
alcohol).16 Inammatory bowel disease (ulcerative colitis
and Crohns disease) accounts for roughly two-thirds of
the incidence,17,18 and the risk increases with duration of
illness (2% at 10 years, 18% by 30 years18), and severity
and extent of inammation.19 Although colitis-associated
colorectal cancer has many of the same molecular
carcinogenic mechanisms as has sporadic cancer, with
similar frequencies of chromosomal (about 85%) and
microsatellite instability (about 15%), there are important
molecular dierences.20
Of the hereditary syndromes, the frequencies for
inherited syndromes are as follows; the Lynch
syndrome, also known as hereditary non-polyposis
colorectal cancer, occurs in roughly one in 300 people
with colorectal cancer. Familial adenomatous polyposis
is much less frequent, and arises in about one in
7000 people aected by colorectal cancer, whereas
MYH-associated polyposis occurs in about one in
18 000 individuals with colorectal cancer. All familial
cases of colorectal cancer of the syndrome type account
for nearly 6% of casesnamely, 3% of those with Lynch
syndrome, 2% with familial colorectal cancer nonLynch syndrome, and about 1% constitute all of the
others, including the TACSTD1 deletion of MSH2. At
this time, there is no reliable quantitative estimate of
the frequency of TACSTD1 deletion of MSH2 in
Lynch syndrome.

Genetic epidemiology: Lynch syndrome as model


More than a fth of patients with colorectal cancer might
have a familial component, and about 3% of cases will
develop Lynch syndrome,21 which is the most common
hereditary syndrome associated with this cancer (panel 1).
Less than 1% of cases will have familial adenomatous
polyposis or one of the hereditary syndromes of
hamartomatous polyposis.24,25 At least 20% of cases of
colorectal cancer are familial (dened by two or more
rst-degree relatives with this cancer). Although this
familial category remains aetiologically elusive, it is
estimated to have a two-fold to three-fold greater risk
than has the general population. Type X familial colorectal
cancer, a new susceptibility category, meets the
Amsterdam criteria26 for Lynch syndrome but lacks the
molecular genetic features.27,28 The Amsterdam criteria26
and Bethesda guidelines29 are widely used as clinical
screening methods for assessment of risk (panel 2).
The public health implications of hereditary colorectal
cancer are substantial because close relatives of an index
patient could benet from genetic counselling and,
potentially, from mutation testing. Once a diagnosis is
strongly suspected on the basis of family pedigree or
molecular conrmation in the index patient, the next
question is which family members should be tested.30
Many physicians might not be cognisant of the molecular
www.thelancet.com Vol 375 March 20, 2010

Chromosomal
instability pathway

Mismatch
repair pathway

Serrated pathway

Hereditary and sporadic

Hereditary

Hereditary

Sporadic

CIMP status

Negative

Negative

High

MSI status

MSS

MSI-H

MSI-H

MSI-L

Chromosomal instability

+++

---

---

---

KRAS mutation

+++

+/-

---

---

BRAF mutation

---

---

+++

+++

MLH1 status

Normal

Mutation

Methylated

Partial
methylation

Adapted from Nosinger.3 CIMP=CpG island methylator phenotype. MSS=microsatellite stability. MSI=microsatellite
instability. MSI-H=high-level microsatellite instability. MSI-L=low-level microsatellite instability. +++=present. +/-=might
or might not be present. ---=absent.

Table 1: Molecular classication of colorectal carcinoma

Panel 1: Cardinal features of Lynch syndrome (hereditary non-polyposis


colorectal cancer)
Autosomal dominant inheritance pattern of syndrome cancers in the family pedigree.
Onset of colorectal cancer at a younger average age than in the general population:
average age of 45 years in individuals with Lynch syndrome versus 63 years in the
general population.
Proximal (right-sided) colonic cancer predilection: 7085% of colorectal cancers in people
with Lynch syndrome are proximal to the splenic exure.
Accelerated carcinogenesis (small adenomas can develop into carcinomas quickly):
within 23 years in Lynch syndrome versus 810 years in the general population.
High risk of additional colorectal cancers: 2530% of patients having surgery for a cancer
associated with Lynch syndrome will have a second primary colorectal cancer within
10 years of surgical resection if the surgery was less than a subtotal colectomy.
Increased risk of malignancy at specic extracolonic sites:22,23 endometrium
(4060% lifetime risk in carriers of mutation); ovary (1215% lifetime risk in mutation
carriers); stomach (increased risk in Oriental families, reason not known); small bowel;
hepatobiliary tract; pancreas; upper uroepithelial tract (transitional cell carcinoma of
the ureter and renal pelvis); and brain (in Turcots syndrome, a variant of Lynch
syndrome).
Sebaceous adenomas, sebaceous carcinomas, and multiple keratoacanthomas in
Muir-Torres syndrome, a variant of Lynch syndrome.
Pathology of colorectal cancer is often poorly dierentiated, with an excess of mucoid
and signet cell features, a Crohns-like reaction, and an excess of inltrating
lymphocytes within the tumour.
Increased survival when compared by stage in non-Lynch-syndrome-associated
colorectal cancer.
The requirement for diagnosis is the identication of a germline mutation in a
mismatch repair gene (most commonly MLH1, MSH2, or MSH6) that segregates in the
familyie, members who have the mutation have a much higher rate of syndromerelated cancers than do those who do not have the mutation.

genetic and phenotypic features of the syndromes


associated with hereditary colorectal cancer, or their
clinical implications for the patient and family.
Referral should be made to a cancer geneticist so that
sensitive and appropriate counselling, and appropriate
management and surveillance, can be oered.
The genetic basis of Lynch syndrome is germline
mutation in one of the genes in the DNA nucleotide
1031

Seminar

Panel 2: Revised Bethesda guidelines29 and Amsterdam criteria26 for identication of


patients at risk of developing Lynch syndrome
Traditionally, one Bethesda criterion or all Amsterdam criteria should be met to
potentially identify individuals at risk of Lynch syndrome.
Bethesda guidelines:
Colorectal cancer diagnosed in a patient who is younger than 50 years
Presence of synchronous, metachronous colorectal, or other tumours associated with
Lynch syndrome, irrespective of age
Diagnosis of colorectal cancer with histologically high-level microsatellite instability
in a patient younger than 60 years
Colorectal cancer diagnosed in one or more rst-degree relatives with a Lynch
syndrome-associated tumour, with one of the cancers diagnosed before age 50 years
Colorectal cancer diagnosed in two or more rst-degree or second-degree relatives
with Lynch-syndrome-related tumours irrespective of age
Amsterdam I and II criteria:
One individual diagnosed with colorectal cancer (or extracolonic Lynch-syndromeassociated tumours) before age 50 years
Three aected relatives, one a rst-degree relative of the other two
Two successive aected generations
Familial adenomatous polyposis should be excluded
Tumours should be veried by pathological examination

mismatch-repair system, most commonly MSH2 and


MLH1 (accounting for roughly two-thirds of the known
mutations) and less commonly MSH6, PMS1, and PMS2.
Several environmental factors or yet-to-be-identied lowpenetrant gene mutations might aect the ultimate
phenotypic expression of this syndrome. Most cases of
familial adenomatous polyposis are caused by germline
mutations in the APC tumour suppressor gene. Almost all
colorectal tumours associated with Lynch syndrome have
microsatellite instability. Immunohistochemistry can be
used to identify loss of mismatch-repair proteins, such as
MSH2 and MLH1, in tumours positive for microsatellite
instability, and thereby direct mutational testing for a
specic gene, which cannot be done with tests for
microsatellite instability.31 If the tumour is microsatellite
stable, the low probability of an informative immunohistochemistry test needs to be weighed against the cost of
doing the test.32 Microsatellite instability should not be used
as the only basis for selection of patients for mutational
testing for Lynch syndrome, because some patients with
microsatellite-stable tumours can have mutations.32 This
diculty can be resolved more cost eectively with tests for
the BRAF V600E mutation than with those for the genes in
the mismatch-repair system, since the presence of this
mutation excludes Lynch syndrome.33
The clinical phenotypes in Lynch syndrome might
dier according to the mutations. MSH2 is associated
with an increased frequency of extracolonic types of
cancer and includes Muir-Torres syndrome. Expression
of MLH1 in colorectal cancer is higher than the expression
of MSH2, and lower in extracolonic cancer. MSH6 shows
a reduction in expression in colorectal cancer but an
1032

increase in endometrial cancer. PMS2 mutations


contribute importantly to the development of Lynch
syndrome, although penetrance in carriers of monoallelic
PMS2 mutations seems to be lower than that for other
genes in the DNA nucleotide mismatch-repair system.34
These clinical and molecular genetic changes in Lynch
syndrome, and in many other hereditary cancer
syndromes, have led to a new era of genetic counselling.
In 500 consecutive patients with colorectal cancer,21
18 (36%) had Lynch syndrome and were positive for
microsatellite instability, 17 (94%) were correctly predicted
with immunohistochemistry for the presence of mismatch
repair proteins, eight (44%) were diagnosed when they
were younger than 50 years, and 13 (72%) met the revised
Bethesda guidelines29 (table 2). The investigators
concluded that one in 35 patients with colorectal cancer
had Lynch syndrome (a high prevalence for a highly
penetrant, lethal autosomal dominant condition), and
each of these patients had at least three relatives with this
syndrome. These relatives could potentially benet from
increased cancer surveillance and management.
Restriction of molecular screening to patients meeting
the Bethesda guidelines would not identify 28% of cases
of Lynch syndrome. The authors also showed that
immunohistochemistry for mismatch-repair proteins was
almost as sensitive as microsatellite instability for
identication of this syndrome. This nding has
important implications for potential large-scale screening
because immunohistochemistry is readily available in
most pathology laboratories and is useful for directing
gene testing, whereas tests for microsatellite instability
require specialist molecular diagnostics. This nding
supports the argument that all patients with colorectal
cancer, irrespective of family history, should be screened
for microsatellite instability or mismatch-repair protein
deciency by immunohistochemistry. Similar evidence
for molecular genetic screening of all newly diagnosed
cases of colorectal cancer was reported by the Evaluation
of Genomic Applications in Practice and Prevention
Working Group,43 linking such screening with improved
health outcomes in relatives of identied individuals.
However, currently large-scale molecular screening of all
patients is not done.
Surveillance for colorectal cancer in individuals with a
germline mutation in the DNA nucleotide mismatchrepair system is highly eective and cheaper than a lack of
surveillance.44,45 Because of the early age of onset of this
cancer in Lynch syndrome and proximal colon occurrence,
full colonoscopy should be started by age 2025 years in
individuals with mutations in the mismatch-repair system,
and in those thought to be at risk on the basis of pedigree
analysis. Colonoscopy should be done at least every
12 years until 40 years of age, and yearly thereafter,
because of accelerated colorectal carcinogenesis in Lynch
syndrome.46 Similar guidelines apply to familial
adenomatous polyposis. Women with a germline mutation
for Lynch syndrome should have yearly screening for
www.thelancet.com Vol 375 March 20, 2010

Seminar

endometrial cancer (4060% lifetime risk) beginning at


3035 years. This screening should include endometrial
aspiration and transvaginal ultrasound, although evidencebased data showing survival benet from such screening
is lacking. Prophylactic hysterectomy and salpingooophorectomy in women with germline mutations in the
mismatch repair system substantially reduce the
occurrence of endometrial and ovarian cancer in women
with conrmed Lynch syndrome.47 These surgeries can be
considered when childbearing is complete. Evidencebased data showing survival advantage for urological,
gastric, and small bowel screening are not available.

Screening of colorectal cancer


Survival rates in individuals with colorectal cancer have
increased substantially in the past few years, possibly as a
result of early diagnosis and improved treatment. Although
substantial information about risk factors exists, about
75% of diagnoses are in patients with no apparent risk
factors other than older age.48 5-year survival is still less
than 60% in most European countries.49 Population
screening therefore continues to oer the best prospects
for reduction in mortality rates.
The aim of screening for colorectal cancer is to prevent
the development of advanced cancers through detection
of localised cancers or premalignant adenomas, from
which at least 80% of cancers are thought to arise. Several
technologies exist. Those that are used to target cancers
early reduce mortality rates, but cause a temporary
increase in incidence rates as cancers are typically
diagnosed at screening 23 years earlier than in
symptomatic cases. These tests need to be oered at least
biennially, which has implications for costs and
compliance rates. Tests that are used to detect adenomas
can be oered less frequently and because these tests
should reduce incidence rates of colorectal cancer, they
also reduce the costs of treatment and, thereby, of the
screening programme.50,51 However, since most adenomas
do not develop into symptomatic cancers, screening for
them can result in overtreatment, which can increase the
risk of complications.
The guaiac-based faecal occult blood test is the most
extensively studied, but possibly least sensitive, screening
method. Several randomised trials and a Cochrane
review52 have provided high-quality evidence that this test,
if oered every 2 years, has the potential to reduce
mortality rates associated with colorectal cancer by 16%.
For this home-test kit, two samples are collected from
three consecutive stools and sent for processing in an
accredited laboratory. Collection of one stool sample by a
physician during digital rectal examination is ineective
and is strongly discouraged.53 Investigation by colonoscopy
is recommended if a specic number of the test cards are
positive; the exact number varies according to local
practice.54,55 Several countries have introduced screening
with the faecal occult blood test.55 Costs have been
assessed in the European and US contexts, and were well
www.thelancet.com Vol 375 March 20, 2010

below the commonly used threshold of US$50 000 per


life-year gained.50,56 Immunochemical faecal occult blood
tests have several improved features compared with the
standard test. They are not subject to interference from
animal blood in the diet, only one or two stool samples
are needed, and they are more sensitive for detection of
colorectal cancer and advanced adenomas, though at the
expense of lower specicity.57 Some tests can be automated,
and the cuto for positivity can be adjusted according to
available endoscopy resources.58
Flexible sigmoidoscopy, with a 60 cm endoscope,
allows examination of the sigmoid colon and rectum
where 60% of cancers and adenomas are located. Bowel
preparation for this sigmoidoscopy requires only one
self-administered enema.59 Results from epidemiological
studies suggest that sigmoidoscopy screening reduces
incidence and mortality rates of distal colorectal cancer
by roughly 6080%;60,61 four large trials are in
progress.6265 Screening is recommended every 5 years
from 50 years of age in the USA,66 although the
protection aorded by one exible sigmoidoscopy
might last for many years.60,61,67 The value of a once-in-alifetime screen with this method at about 60 years of
age is being investigated in UK and Italian trials.62,63
Small polyps are removed during the sigmoidoscopy
screening, and colonoscopy follows only if several or
advanced adenomas are found. The results of both trials
will be reported this year. Colonoscopy every 10 years is
the most common method of screening in the USA
despite evidence that it has little ecacy in reducing
rates of proximal colon cancer.68,69 This method is
associated with a higher risk of serious complications
than are other methods,70 and about 48 h are needed for
bowel preparation and recovery from sedation.
Population screening with colonoscopy also presents
manpower diculties that have yet to be resolved,
although high-quality programmes are available in
Germany and Poland.71,72 CT colonography (virtual
colonoscopy) is as sensitive as colonoscopy for the
detection of cancers and large adenomas, but includes
exposure to radiation, requires full bowel preparation,
and colonoscopy is necessary to conrm and remove
detected lesions.57,73 CT colonography also detects
extracolonic lesions, which might lead to further
invasive tests without any clinical benet.74 It is thus an
expensive option for screening.75
Investigations of DNA-based tests of blood or stool are
in progress.76,77 The sensitivity and specicity of markers
need to be optimised, and automated systems need to be
developed to reduce costs and ensure reliability. Many
other colonic imaging technologies are being
investigated78 to improve the acceptability of colonoscopy
or increase the detection of at lesions.
Whatever technology or combination of technologies is
used, population screening is costly with the risk of
exposing healthy individuals to potential harm. To obtain
the maximum benets and cost eectiveness of any
1033

Seminar

Treatment

Number

Phase

Radiation dose
(Gy)/ fraction size

pCR

Local
failure

Grade 3 or more acute toxicity

Gerard et al35 (ACCORD)

Capecitabine vs CAPEOX

379 vs 368

45/18 vs 50/18

14% vs
19%

11% vs 25% (p<0001)

Aschele et al36 (STAR)

CI 5-FU vs FOLFOX

295 vs 291

503/18 vs 504/18

16% vs
15%

15% vs 24% (p<0001)

Crane et al37

Capecitabine+bevacizumab

25

505/18

32%

7% (2 years)

3% wound complication, requiring surgery

Willett et al38

CAPEOX+bevacizumab

32

504/18

16%

22% diarrhoea, 9% hypertension, 6% skin, 3% dehydration,


3% abscess, 3% pain, 3% neuropathy

Rodel et al39

CAPEOX+cetuximab

48

504/18

9%

19% diarrhoea, 4% rash, 4% neuropathy

Mohiuddin et al40 (RTOG


0012)

FOLFIRI vs CI 5-FU

54 vs 52

2R

50454/18 vs
55260/12 BID

28% vs
28%

57% haematological and non-haematological toxicities vs


51% haematological and non-haematological toxicities

Wong et al41 (RTOG 0247)

CAPIRI vs CAPEOX

48 vs 48

2R

504/18 vs 504/18

10% vs
21%

8% haematological, 24% non-haematological toxicities vs


4% haematological, 29% non-haematological toxicities

Fernandez-Martos et al42
(GCR-3)

Induction CAPEOX vs
postoperative CAPEOX

56 vs 52

2R

504/18 vs 504/18

14% vs
13%

17% vs 51% (p=000004)

pCR=pathological complete response rate. CAPEOX= capecitabine and oxaliplatin. CI 5-FU=continuous infusion of uorouracil. FOLFOX=uorouracil and oxaliplatin. FOLFIRI=uorouracil and irinotecan. R=randomised.
BID=twice a day. CAPIRI=capecitabine and irinotecan.

Table 2: Selected novel preoperative chemoradiation regimens

Figure: High-resolution MRI of rectal cancer


Red arrow shows T2 (tumour invading muscularis propria) rectal tumour; white arrow shows lymph node in the
mesorectum; and yellow dotted line shows the rectal fascia.

scheme, delivery of screening within a high-quality


programme is essential.55

Diagnosis and staging


Colorectal cancer is diagnosed on the basis of the results
of colonoscopy or sigmoidoscopy with tumour biopsy.
Treatment strategy is guided by adequate staging. The
pretreatment workup of a newly diagnosed cancer
includes physical examination, a complete colonoscopy
1034

to rule out metachronous tumour, and CT of the chest,


abdomen, and pelvis to identify metastatic disease.79
CT colonography is valuable for precise localisation of
the tumour and can help surgical approaches, especially
in patients who are candidates for laparoscopic resection.
It might also be used to identify other colonic lesions or
polyps that are not detected at colonoscopy, for instance
because of an obstructive lesion.8082 Routine use of PET
with the 18-uoro-2-deoxy-D-glucose (FDG-PET) is not
recommended at the time of initial diagnosis.83 In
patients with rectal cancer, assessment of local tumour
extension is essential for optimum treatment. Highresolution MRI can be used to accurately measure the
spread of tumour in the surrounding mesorectum, and
to assess the circumferential resection margin between
the edge of tumour and the fascia recti (gure).84
Measurement of the depth of invasion in the bowel wall
with endorectal ultrasound is particularly useful for early
rectal cancers.85 Panel 3 shows the advantages and
limitations of pelvic MRI and endorectal ultrasound for
the assessment of local spread of rectal cancer.
In patients with suspected liver metastases from
colorectal cancer, extent of disease is determined by
ultrasound, CT, and MRI. Although these imaging
methods are used to detect metastases, new methods can
be useful to dene the characteristics of liver metastases.
In particular, enhanced hepatic ultrasound86 and enhanced
MRI can help to characterise liver nodules.87 FDG-PET
can be used to rule out occult extrahepatic spread of the
disease that could change the treatment strategy.88,89
Detection of peritoneal carcinomatosis with imaging
remains a challenge, and performance of the dierent
diagnostic methods is inadequate.90 Involvement of the
multidisciplinary team from the start helps to provide the
best treatment. Once diagnosis is made, staging is
described according to the TNM (tumour, node,
metastases) system (panel 4).91
www.thelancet.com Vol 375 March 20, 2010

Seminar

Panel 3: Advantages and limitations of MRI and endorectal


ultrasound for staging of primary tumours (T) and regional
lymph nodes (N) in rectal cancer
MRI
Advantages:
Global staging of rectal tumour
Circumferential resection margin assessment
Assessment of extramural venous invasion
Guide the indication for radiation treatment
Few contraindications
Assessment of pelvic spread of the tumour (iliac nodes)
Limitations:
Node stagingMRI is more accurate than is endorectal
ultrasound, but there are some limitations in correlation
between radiological and pathological ndings
Endorectal ultrasound
Advantages:
Measure of depth of invasion in the bowel wall
Measurement of T staging
Early rectal cancer
Limitations:
Obstructing lesions
User-dependent imaging modality
Measurement of node staging
Assessment of circumferential resection margin assessment
and extramural venous invasion

Surgery
For colon cancer, total resection of the tumour should be
done with adequate margins, and lymphadenectomy.
Distal margins of 5 cm or more are recommended. At least
12 lymph nodes should be taken and analysed to allow
appropriate nodal staging;9395 analysis of fewer than ten
nodes might understage the tumour.12 En-bloc resection of
invaded adjacent organs might be needed for T4 tumours
to obtain R0 resection (no evidence of microscopic cancer
at the margins). Surgical resection of the rectum for
invasive rectal cancer should include total excision of the
mesorectum (TME) with adequate circumferential and
distal margins, and inferior mesenteric lymphadenectomy.
TME is associated with a reduced risk of local recurrence
whether or not combined with preoperative radiotherapy
or chemoradiotherapy.9698 Sphincter-saving surgery is
feasible in most patients with mid and low rectal cancers if
the distal margin is 1 cm or more. Intestinal continuity can
be restored with colorectal or coloanal anastomosis
according to the level of the tumour. For very low tumours,
TME can be combined with resection of the internal
sphincter of the anus without increasing the rate of local
recurrence.99,100 Abdominoperineal resection is a valuable
alternative for very low tumours.101 Functional results after
TME are associated with the level of the anastomosis.
Risk of faecal incontinence is increased in patients
with very low coloanal anastomosis, particularly after
preoperative radiation.102,103
www.thelancet.com Vol 375 March 20, 2010

Panel 4: TMN classication of colon cancer91


T=primary tumour
TX=primary tumour cannot be assessed
T0=no evidence of primary tumour
Tis=carcinoma in situ: intraepithelial or invasion of lamina propria
T1=tumour invades submucosa
T2=tumour invades muscularis propria
T3=tumour invades through the muscularis propria into subserosa or into nonperitonealised pericolic or perirectal tissues
T4a=tumour penetrates the surface of the visceral peritoneum
T4b=tumour directly invades or is histologically adherent to other organs or structures
N=regional lymph nodes
NX=regional lymph nodes cannot be assessed
N0=no regional lymph node metastasis
N1a=metastasis in one regional lymph node
N1b=metastasis in two to three regional lymph nodes
N2a=metastasis in four to six regional lymph nodes
N2b=metastasis in seven or more regional lymph nodes
M=distant metastasis
MX=distant metastasis cannot be assessed
M0=no distant metastasis
M1a=distant metastasis to one site
M1b=distant metastasis to more than one site
Surveillance, Epidemiology and End Results Program data for 5-year stage-specic relative
survival rates in colon cancers:91,92
Stage I (T1, T2, N0): 971%
Stage IIA (T3, N0): 875%
Stage IIB (T4, N0): 715%
Stage IIIA (T1, T2, N1): 877%
Stage IIIB (T1, T2, N2): 750%
Stage IIIB (T3, N1): 687%
Stage IIIC (T3, N2): 473%
Stage IIIC (T4, N1): 505%
Stage IIIC (T4, N2): 271%
5-year stage-specic relative survival rates were similar for rectal cancer as compared with
colon cancer.

In patients with early rectal cancer, the choice of


treatment is complete local excision or TME, and
depends on the risk of lymph-node involvement, which
is associated with the depth of invasion of the tumour in
the rectal wall. Local excisiontransanal excision or
endoscopic microsurgery for tumours in the upper-third
layer of the submucosa (T1Sm1) and some in the middle
layer (T1Sm2)is valuable if excision is completed with
adequate margins.104106
Laparoscopic colectomy is safe for colon cancer,
particularly left-sided cancer. The long-term oncological
results of this surgery are similar to those of the open
approach.107110 Although laparoscopic colectomy is
technically demanding, advantages are reduced pain,
length of hospital stay, and duration of ileus.111 Comparison
of laparoscopic and open resection (with TME) of rectal
cancer112 showed similar 3-year local recurrence rates and
1035

Seminar

survival despite earlier reports of an increased rate of R1


resection (ie, microscopic tumour present at the resection
margin) with the laparoscopic approach.108 There was a
non-signicantly higher risk of sexual complications
after laparoscopy than after open TME.113
The liver is the most common site of relapse after
surgery for colorectal cancer, and lung recurrence is
common. Resection of the liver and lung metastases is the
standard of care for potentially resectable oligometastatic
disease, and, when feasible, substantially improves 5-year
and 10-year survival rates. Improvement of strategies for
liver surgery, including preoperative embolisation, twostage liver resections, and renements of non-surgical
techniques, such as radiofrequency ablation of small
lesions, have increased the number of patients undergoing
complete local treatment of liver metastases. Important
progress has been made in the treatment of this disease
with use of multimodality and multidisciplinary methods.
Chemotherapy (oxaliplatin-based) given before and after
surgery versus surgery alone reduced the risk of cancer
relapse after surgery (progression-free survival at 3 years
was improved by 73% in all patients [p=0058], and by
92% in patients undergoing resection [p=0025]) in the
EORTC 40983 study.114 In patients with unresectable liver
metastases, initial or conversion chemotherapy can render
metastases resectable if the response to treatment is good.
Preoperative chemotherapy for resectable lung metastases
could improve outcomes, but there are few supporting
data.115 Radiofrequency ablation might also have a role in
non-resectable liver-only metastatic disease, and the
survival outcomes with local radiofrequency ablation
added to systemic palliative chemotherapy are being
assessed in the EORTC CLOCC trial.116

Radiation treatment for rectal cancer


The risk of local failure in treatment of rectal cancer is
aected by involvement of the circumferential resection
margin, lymph-node status, and extramural venous
invasion.84 Since a signicantly lower incidence of local
recurrence and toxicity, and a higher incidence of
sphincter preservation were reported with preoperative
than with postoperative chemoradiation in the German
CAO/ARO/AIO 94 trial,117 the conventional treatment for
clinical stage T3 (cT3) or node-positive rectal cancer is
preoperative treatment, and is the choice in most
countries. Radiation, a local control strategy, is used with
conventional fractionation (504 Gy in 28 fractions) since
short-course radiation (5 Gy5) cannot be safely used
with adequate doses of chemotherapy. Systemic
chemotherapy improves survival. In Scandinavia and
most northern European countries, treatment depends
on preoperative MRI assessment of the circumferential
resection margin.118120 Generally, if this margin is likely to
be negative at the time of surgery, patients undergo
surgery alone or are given ve doses of 5 Gy. If the margin
is likely to be positive, patients are given preoperative
radiation (5 Gy5) or chemoradiation. A positive
1036

circumferential resection margin is not suciently


controlled with postoperative treatment.121,122
Overstaging is a disadvantage of preoperative
treatment (18% in the German trial117), thereby patients
with pathological stage T1-2N0 disease are at risk of
being overtreated. Preoperative treatment is still
preferred to surgery rst since even after preoperative
chemoradiation 22% of patients still have node-positive
disease.123 These patients would then need postoperative
chemoradiation, which has inferior outcome and higher
toxicity than preoperative chemoradiation.
There are no prospective randomised data for local
recurrence based on distance from the anal verge, only
subset analysis from randomised trials that were not
stratied by distance. With univariate analysis, high
tumours (1011 cm) had a lower incidence of local
recurrence than did mid and lower tumours.122,124 By
contrast, there was no signicant dierence between mid
and upper tumours in the German trial.125 On the basis of
the conicting data and the fact that the incidence of
positive nodes after preoperative chemoradiation is the
same from 012 cm from the anal verge,123 treatment
decisions should not be based on distance if it is shorter
than 12 cm.
Chemoradiation and short-course radiotherapy have
been the main preoperative strategies assessed in
randomised studies. Short-course preoperative radiation
in patients with rectal cancer resulted in a survival
advantage for the total treatment group in a Swedish
trial.124 Despite TME, the local recurrence rate with nodepositive disease was 21% in the Dutch CKVO 95-04
trial.126 Therefore patients with node-positive tumours
need adjuvant radiation. The challenge is accurate
identication of positive nodes to allow proper selection
of patients for preoperative treatment. In the randomised
study by Bujko and colleagues,127 patients given
chemoradiation compared with radiation (5 Gy5) had a
signicantly lower incidence of positive circumferential
resection margin (4% vs 13%), but no signicant
dierence in local failure (14% vs 9%) or 4-year survival
(66% vs 67%). Furthermore, although the rate of
pathological complete response was much higher (16% vs
1%), the incidence of sphincter preservation was not
increased. However, because the number of patients
(n=316) was small, surgeons were not encouraged to
modify the operation on the basis of tumour response,
and there was a lack of central review of the quality of
the radiotherapy.
Use of adjuvant chemotherapy after preoperative
chemoradiation has been contentious. Although data
from randomised trials showed a 1015% survival
benet with postoperative uorouracil-based chemotherapy in colon and rectal trials, this result was not
conrmed in the EORTC 22921128 and FFCD 9203129
randomised trials. The negative results might be
partly attributable to the diculty in patients tolerating full doses of chemotherapy after preoperative
www.thelancet.com Vol 375 March 20, 2010

Seminar

chemoradiation. Chemotherapy was better tolerated in


the neoadjuvant than in the adjuvant setting in the
GCR-3 phase II trial (table 2).42 A potential advantage of
neoadjuvant chemotherapy is the early treatment of
micrometastatic disease.
Conventional chemoradiation regimens include
continuous-infusion uorouracil or capecitabine (an
oral uorouracil prodrug). New cytotoxic and targeted
treatments are being investigated (table 2).3542 Most
show higher rates of pathological complete response
than with uorouracil alone. However, acute toxicity
was signicantly higher without a benet in the rate
of pathological complete remission with the addition
of oxaliplatin to continuous-infusion uorouracilbased36
or
capecitabine-based
chemoradiation
(ACCORD 12-040535). Local control and survival data
were not available, but the rate of distant metastases
was lower in the oxaliplatin-combination group than
in the group not given oxaliplatin in the STAR trial.35
Most clinical series indicate improved outcome with
increasing response to preoperative chemoradiation.130 Use
of molecular markers131,132 has had varying success in the
prediction of response rates. Clinical or radiological
responses to preoperative chemoradiation do not
suciently correlate with pathological response.133
Adjuvant treatment should be based on the initial T and N
stages. In one series, the value of radical surgery in patients
(with cT13 disease) with a biopsy-proven complete
response was questioned,134 and although not conrmed by
other investigators remains a pertinent research question.
Novel fractionation and delivery techniques might
improve outcomes in the future. Generally, hyperfractionated radiation improves the rates of pathological
complete response but increases acute toxicity.135 Threedimensional treatment allows planning and localisation of
the target and normal tissues at all levels of the treatment
volume, and to obtain dose-volume histograms.136,137
Intensity-modulated radiotherapy can further reduce the
volume of the small bowel in the eld.138 The clinical
benet of this treatment compared with three-dimensional
or conventional treatment is yet to be determined.
Current and simple guidelines or algorithms for the
multimodality management of rectal cancer, including
surgery and radiation, and all stages of disease have
been reported by the US National Comprehensive
Cancer Network.139

Adjuvant treatment for early stage disease


Surgery is the cornerstone for cure in localised colorectal
cancer. In node-positive (stage III) disease, administration
of adjuvant uorouracil for 6 months reduces the risk of
death by 30%, which is equivalent to an additional 1015%
survival gain.140 The alternative oral uoropyrimidine,
capecitabine, has shown similar ecacy to uorouracil
alone.141 The establishment of 3-year disease-free survival
as a validated surrogate for 5-year overall survival142,143 and
primary endpoint has expedited the timely reporting of
www.thelancet.com Vol 375 March 20, 2010

adjuvant trials and uptake of eective treatments.


However, the magnitude of benet in disease-free survival
is not necessarily matched by that in overall survival
because of various factors.143 Oxaliplatin (a third-generation
platinum) added to infused uorouracil versus infused
uorouracil alone improved 3-year disease-free survival
by 7% in the MOSAIC study,144 leading to a 25% gain in
overall survival at 6 years for stage II and III disease, and
a survival increment of 42% in stage III alone.145 Results
for disease-free survival were replicated in the NSABP
C-07 study of uorouracil and oxaliplatin.146 In the
XELOXA study147 of oral capecitabine plus oxaliplatin
versus uorouracil and folinic acid (stage III disease),
there was an improvement in 3-year disease-free survival
(709% vs 665%, hazard ratio 080, p=00045) but a nonsignicant 5 year benet in overall survival (34%). The
choice between one drug (intravenous uorouracil or
oral capecitabine) or two will depend on the patients
tness or preference, potential for compliance, and
toxicity considerationseg, handfoot syndrome with
capecitabine, and risk of permanent peripheral sensory
neuropathy with oxaliplatin (155% all grades of peripheral
sensory neuropathy at 48 months after treatment145), with
patient-reported improvement in hand peripheral sensory
neuropathy but worsening of foot numbness, tingling, or
discomfort by 18 months.148 Results from the MOSAIC
study144 did not suggest a survival benet with adjuvant
oxaliplatin and uorouracil in patients older than 65 years,
a nding supported for individuals older than 70 years in
an analysis of the ACCENT group database of more than
12 500 patients (17% older than 70 years) given adjuvant
treatment in six randomised studies (including NSABP
C-07 and MOSAIC).149 These data and toxicity
considerations imply uoropyrimidine monotherapy
might be considered in older patients with stage III
colorectal cancer. Irinotecan added to uorouracil did not
signicantly improve disease-free or overall survival,150152
although there was a non-signicant improvement with
combination treatment in the PETAAC-3 trial.152
Adjuvant chemotherapy for node-negative (stage II)
colon cancer (40% of resected cancers) has been
controversial because of the small gains in survival for
this subgroup in studies in which patients with stage II
and III cancer were combined, although high-risk
patients with stage II tumours (T4 tumours, obstructing
presentation, poor dierentiation, extramural venous
invasion, fewer than 1012 harvested lymph nodes,
indeterminate or positive resection margins) are often
oered treatment.153 However, in the QUASAR study154
of 3239 patients (with mostly stage II colorectal cancer)
randomly assigned to uorouracil and folinic acid or to
observation (20% had central pathology review), there
was a signicant improvement in overall survival (risk
reduction of 18% and 36% overall survival gain), with
a similar benet for colon and rectal cancers. This
small benet weighted against treatment toxicities and
logistics, and comorbidities, should be included in
1037

Seminar

Study population

Treatment groups

Patients (n)

Response rate

Median PFS
(months*)

Median OS
(months)

Hazard ratio p value for


primary
for primary
endpoint
endpoint

BOND172

Pretreated
(irinotecanrefractory)

Cetuximab alone vs irinotecan-based


chemotherapy+cetuximab

111 vs 218

108% vs 229%

15 vs 41

69 vs 86

054 (TTP),
091 (OS)

<0001 (TTP),
048 (OS)

NCIC CO.17173

Pretreated
(uorouracil,
irinotecan, and
oxaliplatin)

Best supportive care vs cetuximab

285 vs 287

0 vs 8%

NR

46 vs 61

077

0005 (OS)

EPIC174

Pretreated
(uorouracil/
oxaliplatin)

Irinotecan vs irinotecan+cetuximab

650 vs 648

42% vs 164%

26 vs 40

10 vs 107

098

071 (OS)

Van Cutsem
et al175

Pretreated
(uorouracil,
irinotecan, and
oxaliplatin)

Best supportive care vs panitumumab

232 vs 231

0 vs 10%

73 weeks vs
80 weeks

NR

054

<0001 (PFS)

CRYSTAL176

Untreated

FOLFIRI vs FOLFIRI+cetuximab

599 vs 599

387% vs 469%

80 vs 89

186 vs 199

085

0048 (PFS)

OPUS177

Untreated

FOLFOX vs FOLFOX+cetuximab

168 vs 169

36% vs 46%

72 vs 72

NR

NR

064 (ORR)

CELIM178

Untreated liver-only FOLFOX+cetuximab vs FOLFIRI+


non-resectable
cetuximab vs either regimen for wildmetastases
type KRAS

53 vs 53 vs
67

68% (95% CI 5480) vs


57% (95% CI 4270) vs
70% (95% CI 5881)

COIN179

Untreated wild-type FOLFOX or CAPEOX vs


KRAS
FOLFOX/CAPEOX+cetuximab

367 vs 362

50% vs 59%

86 vs 86

179 vs 170

104

068 (OS)

PRIME180

Untreated wild-type FOLFOX vs FOLFOX+panitumumab


KRAS

331 vs 325

48% vs 55%

80 vs 96

197 vs 239

08

002 (PFS)

181 Study181

Pretreated wild-type FOLFIRI vs FOLFIRI+panitumumab


KRAS

294 vs 303

10% vs 35%

39 vs 59

125 vs 145

073 (PFS),
085 (OS)

0004 (PFS),
012 (OS)

Hurwitz et al182

Untreated

IFL vs IFL+bevacizumab

411 vs 402

348% vs 448%

62 vs 106

156 vs 203

066

<0001 (OS)

E3200183

Treated

FOLFOX vs FOLFOX+bevacizumab vs
bevacizumab

291 vs 286 vs 86% vs 227% vs 33%


243

47 vs 73 vs 27

108 vs 129 vs
102

075 (NR)

00011 (OS)

N016966184

Untreated

XELOX/FOLFOX vs XELOX/
FOLFOX+bevacizumab

701 vs 699

49% vs 47%

80 vs 94

199 vs 213

083

00023 (PFS)

Anti-EGFR trials

Anti-VEGF trials

Combined anti-EGFR and anti-VEGF trials


BOND 2185

Treated (irinotecanrefractory)

Cetuximab+bevacizumab vs irinotecan 40 vs 43
+cetuximab+bevacizumab

20% vs 37%

49 vs 73 (TTP)

114 vs 145

NR

NR

PACCE186

Untreated

Oxaliplatin-based
chemotherapy+bevacizumab vs
oxaliplatin-based chemotherapy+
bevacizumab+ panitumumab

410 vs 413

48% vs 46%

114 vs 100

245 vs 194

127

NR on PFS

CAIRO-2187

Untreated

XELOX+ bevacizumab vs XELOX+


bevacizumab+cetuximab

368 vs 368

50% vs 527%

107 vs 94

203 vs 194

122

p=001 (PFS)

PFS=progression-free survival. OS=overall survival. EGFR=epidermal growth factor receptor. TTP=time to progression. NR=not reported. FOLFIRI=uorouracil and irinotecan. FOLFOX=uorouracil and oxaliplatin.
ORR=overall response rate. CAPEOX=capecitabine and oxaliplatin. VEGF=vascular growth factor receptor. IFL=bolus uorouracil and irinotecan. XELOX=capecitabine and oxaliplatin. *Unless otherwise indicated. For
comparison between control and investigational groups (all-comers and not according to KRAS status). Patients with wild-type or mutated KRAS were randomised in the intent-to-treat population, but data shown
are for the primary and secondary efficacy analyses done in the KRAS wild-type population only. Coprimary endpoints. 230 additional patients were treated with irinotecan-based chemotherapy but the primary
efficacy analysis was restricted to the oxaliplatin cohort (n=823).

Table 3: Selected randomised trials of targeted drugs for metastatic colorectal cancer

discussions of adjuvant chemotherapy. Tests of


microsatellite instability might contribute to the riskbenet assessment of treatment in stage II disease. In
QUASAR,154 patients with stage II colorectal cancer
older than 70 years did not seem to benet from
chemotherapy, and the question is whether to treat or
not to treat this age group. In the MOSAIC study, the
benet of adjuvant oxaliplatin and uorouracil in stage
II disease seemed small (no gain in 6-year overall
survival, hazard ratio for 5-year disease-free survival
1038

084), with a non-signicant benet in patients with


high-risk stage II cancer (23% gain in 6-year overall
survival, hazard ratio for 5-year disease-free survival
072).145 Central to the decision-making process
for adjuvant treatment (monotherapy or oxaliplatinbased) in all patients (stage II and III disease, older
patients, and those with comorbidities) is discussion
with the patient, and incorporation of the patients
preference after accurate communication of benets
and risks of treatment.
www.thelancet.com Vol 375 March 20, 2010

Seminar

KRAS
assessed
n (%)

Treatment
groups (control
vs EGFR mAB)

KRAS

Wild
type

Mutated

Response rate

All

KRAS wild
type

Progression-free survival (months*)

Overall survival (months)

KRAS
mutated

All

KRAS wild type

KRAS
mutated

All

KRAS wild type

KRAS
mutated

NCIC CO17190

394
(69%)

230
(58%)

164
(42%)

BSC vs cetuximab

0 vs 8% 0 vs
128%

0 vs 12%

NR

19 vs 37, HR
040, p<0001

18 vs 18, HR
099, p=096

46 vs 48 vs 95, HR
61
055, p<0001

EPIC191

300
(23%)

192
(64%)

108
(36%)

Irinotecan vs
cetuximab+
irinotecan

42% vs 74% vs
164% 103%,
p=061

51% vs
122%,
p=029

26 vs 40

28 vs 40,
HR 077,
p=0095

27 vs 26, HR
100, p=098

10 vs
107

116 vs 109,
107 vs 84,
HR 129, p=018 HR 128,
p=029

Amado, et al192

427
(92%)

243
(57%)

184
(43%)

BSC vs
panitumumab

0 vs
10%

0 vs 17%

0 vs 0

73 weeks
vs 80
weeks

73 weeks vs 123 73 weeks vs


weeks, HR 045, 74 weeks,
HR 099
p<0001

NR

76 vs 81,
HR 099

44 vs 49,
HR 102

CRYSTAL189,193
(200910
update)

1063
(89%)

666
(63%)

397
(37%)

FOLFIRI vs
cetuximab+
FOLFIRI

397% vs
593%,
p<00001

361% vs
313%,
p=0348

84 vs 99, HR
77 vs 74, HR
0696, p=00012 1171,
p=02761

20 vs 235, HR
0796,
p=00093

167 vs 162,
HR 1035,
p=07551

OPUS189 (2009
update)

315
(93%)

179
(57%)

136
(43%)

FOLFOX vs
cetuximab+
FOLFOX

34% vs
573%,
p=00027

525% vs
328%,
p=0290

72 vs 83, HR
85 vs 55, HR
0567, p=00064 1720,
p=00153

185 vs 228, HR 175 vs 134,


HR 1290,
0855,
p=02004
p=03854

COIN179

1305
(80%)

724
(56%)

561
(43%)

FOLFOX+CAPEOX
vs cetuximab/OX

50% vs
59%

41% vs
40%

86 vs 86, HR
0959, p=060

69 vs 65, HR
1065,
p=046

179 vs 170, HR
104, p=068

PRIME180

1096
(93%)

656
(60%)

440
(40%)

FOLFOX vs
panitumumab+
FOLFOX

48% vs
55%

40% vs
40%

80 vs 96, HR
08, p=002

88 vs 73, HR
129, p=002

197 vs 239,
193 vs 155,
HR 083, p=007 HR 124,
p=007

181 study181

1083
(91%)

597
(55%)

486
(45%)

FOLFIRI vs
panitumumab+
FOLFIRI

10% vs
35%

14% vs
13%

39 vs 59, HR
073, p=0004

49 vs 50, HR
085, p=014

125 vs 145, HR
085, p=012

46 vs 45, HR
098, p=089

148 vs 136,
HR 098,
p=08

111 vs 118,
HR 094,
p=055

Data are number (%) or %, unless otherwise indicated. mAB=monoclonal antibody. BSC=best supportive care. FOLFIRI=uorouracil and irinotecan. FOLFOX=uorouracil and oxaliplatin. CAPEOX=capecitabine
and oxaliplatin. HR=hazard ratio for comparison between control and EGFR-directed treatment. OX=oxaliplatin-based chemotherapy. NR=not reported. *Unless otherwise indicated. Refers to the unselected
intention-to-treat population. Have been prospectively analysed according to KRAS status.

Table 4: KRAS mutation status as a predictive marker of outcome to epidermal growth factor receptor (EGFR)-directed monoclonal antibodies

Of the targeted treatments assessed, addition of


adjuvant edrecolomab (a monoclonal antibody against
the epithelial cell adhesion molecule ) and bevacizumab
(a humanised monoclonal antibody against vascular
endothelial factor [VEGF]) to chemotherapy did not
improve disease-free survival.155,156 Ecacy results from
another study of adjuvant bevacizumab (AVANT study)
are awaited, and an interim analysis of a randomised
study of cetuximab (a monoclonal antibody targeted at
the epidermal growth factor receptor [EGFR]) added to
adjuvant uorouracil and oxaliplatin has indicated a lack
of benet with the cetuximab combination.157
Disease relapse after surgery, with or without adjuvant
chemotherapy, mostly occurs within 3 years.143 Intensive
follow-up strategies for patients with colorectal cancer can
improve
survival,
with
the
early
detection
of oligometastatic disease (commonly liver or lung)
and potential for further radical treatment in a third of
patients.158160 5-year survival after liver and lung resection
is about 3658% and 2741%, respectively.159,160
Surveillance should be through a combination of clinical
review, monitoring of serum carcinoembryonic antigen
every 36 months for 3 years and then every 612 months
until 5 years, imaging (most commonly CT) once a year
for the rst 3 years, and colonoscopy 1 year after surgery
and then every 35 years.160162
www.thelancet.com Vol 375 March 20, 2010

Treatment of metastatic disease


Palliative chemotherapy for metastatic colorectal cancer
can improve survival, lessen symptoms, improve
quality of life, and downsize liver-only or lung-only
metastases in patients with potentially resectable
disease. Survival has increased from 12 months with
uorouracil monotherapy to roughly 2 years with the
addition of irinotecan, oxaliplatin, and targeted drugs.
The use of infused uorouracil added to oxaliplatin
and to irinotecan has reduced toxicity and is commonly
used.163 Fluorouracil can be safely substituted with
capecitabine when combined with oxaliplatin without
loss of ecacy.164,165 For increments in survival, the
sequence of drugs used at rst presentation of disease
and then on disease progression seems less important
than is exposure to all active drugs during the treatment
pathway.166 Although initial combination treatment
is commonly used, rst-line uoropyrimidine
monotherapy is appropriate in some patients.167,168
When the aim is liver or lung resection, conversion
regimens with high response rates are preferable.169
Treatment might continue until disease progression or
for a xed duration (usually 6 months), depending on
toxicities, preference of the patient, and tumour
response. Intermittent scheduling has also been
investigated.170,171
1039

Seminar

Location

Function

Consequence

Microsatellite
instability

MSI-H

Reduces DNA nucleotide


mismatch repair (deciency)

Improved prognosis

KRAS

Chromosome 12, mutations at Reduces GTPase activity, leading


codons 12 and 13
to constitutive activation of
downstream pathways

Resistance to anti-EGFR
mAB

BRAF

V600E mutation

Constitutive activation of
downstream pathways

Resistance to anti-EGFR
mAB

TS

TSER 28 base pairs (2R)/(3R)


tandem repeat 5UTR

TS expression increased

Response to uorouracil
reduced

TS

TSER 3R G>C single-nucleotide TS expression reduced


polymorphism

Response to uorouracil
increased

ERCC-1

mRNA expression High

DNA excision repair

Resistance to platinumbased chemotherapy

Interleukin 8

Interleukin 8 T-251A (A/A)

VEGF expression increased

Recurrence increased

VEGF

VEGF C+936T (C/C)

VEGF expression increased

Recurrence increased

MSI-H=high-level microsatellite instability. EGFR=epidermal growth factor receptor. mAB=monoclonal antibody.


TS=thymidylate synthase. 2R=two tandom repeats. 3R=three tandem repeats. UTR=untranslated region. TSER=TS
enhancer region. ERCC-1=excision-repair cross-complementing-1. VEGF=vascular-endothelial growth factor.

Table 5: Genetic markers and their clinical consequences in colorectal cancer

The main advance in the management of metastatic


colorectal cancer in the past 5 years has been the
addition of targeted treatments. Cetuximab, bevacizumab,
and panitumumab (a fully human EGFR monoclonal
antibody), the only licensed targeted drugs, have,
however, had a relatively small eect on survival
outcomes. Table 3 shows the main randomised studies
and summarises the ecacy outcomes of these drugs
when used as monotherapy or as part of combination
treatment.
In chemorefractory patients, cetuximab showed activity
when used alone, and reversal of irinotecan-resistance
when used with irinotecan.172 Activity of cetuximab alone
or in combination with irinotecan in chemorefractory
patients has been conrmed in further studies173,174,188 and
activity has now been demonstrated for cetuximab in
combination with uorouracil and irinotecan, and
uorouracil and oxaliplatin regimens in untreated
patients (table 3).176178 Crossover to a cetuximab-based
regimen might have masked survival dierences between
treatment groups in pretreated patients, and subsequent
treatments might have aected the survival results in
untreated patients. First-line treatment with cetuximab
added to uorouracil and irinotecan marginally improved
progression-free survival in the CRYSTAL study.176
However, retrospective subgroup analysis showed
signicantly improved response rates and progressionfree survival in patients with liver-only metastases and
KRAS wild-type tumours, and increased rates of liver
resection with combination treatment. With longer
follow-up, the dierence in progression-free and overall
survival between the two groups in patients with wildtype KRAS has increased and the dierence in overall
survival is now signicant (table 3).189 By contrast,
although comparison of oxaliplatin-based chemotherapy
(investigators choice) with or without cetuximab in the
1040

COIN trial179 showed a higher response rate in the


cetuximab combination group, it did not show a
signicant improvement in progression-free or overall
survival even in patients with wild-type KRAS and BRAF.
Panitumumab has been approved as monotherapy for
pretreated patients.175 It signicantly improved
progression-free and overall survivals when given with
uorouracil and oxaliplatin to untreated patients with
wild-type KRAS tumours (PRIME study).180 Panitumumab
in combination with irinotecan and uorouracil also
resulted in a signicant improvement in progressionfree but not overall survival in pretreated patients with
wild-type KRAS.181 KRAS mutation status has been
consistently identied as a predictor of patient response
to EGFR-directed monoclonal antibodies (table 4), and
cetuximab and panitumumab are now only oered to
patients with wild-type KRAS colorectal cancer. Rash,
diarrhoea, and hypomagnesaemia are characteristic
toxicities of anti-EGFR drugs.
Bevacizumab is an antiangiogenic monoclonal
antibody and can also cause normalisation of tumour
vasculature, thereby improving drug delivery to the
target.
Its
side-eects
include
hypertension,
gastrointestinal perforation, delayed wound healing,
bleeding, and thromboembolism. Bevacizumab, when
administered with chemotherapy that included bolus
irinotecan and uorouracil, improved overall survival
by 47 months and response rates by 10% in untreated
patients with metastatic colorectal cancer.182 However,
these results were not replicated when bevacizumab
was given with oxaliplatin-based chemotherapy to
untreated patients,184 whereas the combination
signicantly prolonged survival in previously treated
patients (table 3).183 Failure to continue combination
treatment until disease progression and possible
occurrence of VEGF rebound might have contributed
to these contradictory ndings.184 Whether bevacizumab
should be continued after disease progression with a
change of the cytotoxic drugs remains an important
question, and an observational series suggested
prolongation in survival with this strategy.194
Maintenance bevacizumab after initial response to
combination treatment is being investigated, but this
drug has little activity when used alone.183 The risk of
postoperative complications, including delayed wound
healing, is increased with bevacizumab. However, the
risk seems to be low when an interval of 6 weeks from
the last bevacizumab dose to surgery is allowed.195
Addition of cetuximab to capecitabine, oxaliplatin, and
bevacizumab (CAIRO trial),187 and of panitumumab to
bevacizumab in combination with irinotecan or
oxaliplatin-based chemotherapy (PACCE trial)186 in
untreated patients with metastatic disease worsened
progression-free survival and increased toxicity,
indicating a possible negative interaction of combining
these targeted drugs despite encouraging results in a
previous phase II trial (table 3).185
www.thelancet.com Vol 375 March 20, 2010

Seminar

Bevacizumab received regulatory approval in 2004


for rst-line treatment of metastatic colorectal cancer
in combination with uoropyrimidine-based chemotherapy, and was widely considered a standard rst-line
treatment. However, the licence for rst-line cetuximab
plus chemotherapy on the basis of the results of the
CRYSTAL study176 and increased rates of liver resection
in patients with wild-type KRAS tumours and initially
unresectable liver-only metastases provides another
treatment option. Nevertheless, the benets of
cetuximab compared with bevacizumab have not been
proven in this setting. Regulatory approval was granted
for cetuximab to be used as monotherapy or as
combination therapy in 2004 in pretreated
chemotherapy-resistant patients for whom there are
few treatment options, and for panitumumab
monotherapy in 200607 in patients with wild-type
KRAS pretreated tumours. However, outside the USA,
the cost has to some extent restricted the use of
cetuximab, panitumumab, and bevacizumab; thus,
identication of subgroups most likely to benet from
treatment could improve cost eectiveness. For
instance, in the UK, cetuximab was only approved by
the National Institute for Health and Clinical Excellence
(NICE) for wild-type KRAS non-resectable liver-only
metastatic colorectal cancer.196

Prognostic and predictive molecular markers


Prognostic biomarkers are associated with survival that
is independent of the treatment eect. Predictive
biomarkers indicate likely benet of treatment. Markers
can be prognostic and predictive. The most important
development in molecular markers for metastatic
colorectal cancer has been the validation of KRAS
mutation status as predictive of non-response to EGFRtargeted drugs. In the adjuvant setting, prognostic and
predictive molecular markers (microsatellite instability
and 18q imbalance) could potentially be used to
discriminate between molecular phenotypes in stage II
disease (clinically heterogeneous), thereby contributing
to the risk-benet assessment of adjuvant treatment
(table 5). However, complex pathways contribute to
disease progression and, in general, single markers
might not be entirely useful for prediction of ecacy
and outcome. With high-throughput genome-wide
screening, predictive and prognostic molecular
signatures are increasingly being sought but, as yet,
none have been validated for clinical use.
Roughly 40% of patients with metastatic colorectal
cancer have somatic activating KRAS mutations. These
mutations are highly predictive of non-response to
EGFR inhibitors (table 4). The updated results of
CRYSTAL189 showed a signicant improvement in
response rate, progression-free survival, and overall
survival (235 months vs 200 months, p=00094) for
patients with wild-type KRAS tumours given
uorouracil, irinotecan, and cetuximab compared with
www.thelancet.com Vol 375 March 20, 2010

patients with mutant KRAS tumours who did not seem


to benet from treatment. The updated results of
OPUS189 also showed a signicant increase in response
rate and progression-free survival (hazard ratio 0567,
95% CI 03750856, p=0006), but not in overall
survival in patients with wild-type KRAS given
cetuximab plus uorouracil and oxaliplatin (table 4).
The predictive value of KRAS mutation status was
conrmed in a meta-analysis of CRYSTAL and OPUS.189
Similarly, KRAS mutation status is useful for prediction
of the response to panitumumab.180,181,192 Importantly,
patients with KRAS-mutated tumours given rst-line
oxaliplatin-based chemotherapy had worse progressionfree survival and overall survival (table 4) in the PRIME
study,180 worse progression-free survival in OPUS,177
and a non-signicantly inferior overall survival in
COIN.179 In randomised studies of irinotecan-based
chemotherapy plus EGFR-directed monoclonal
antibodies, a detrimental eect on survival in patients
with KRAS mutant tumours has not been observed.181,189
KRAS mutation status should be established before
starting treatment.
However, not all 60% of patients with wild-type KRAS
will respond to treatment. Additional factors, such as
amphiregulin and epiregulin, might contribute to
treatment response; and mutation of BRAF or NRAS,
or loss of PTEN or PIK3CA activation might contribute
to resistance to EGFR-targeted monoclonal antibodies.197
BRAF is the main downstream eector of KRAS.
Mutations that activate BRAF arise in 810% of
metastatic colorectal cancers and are mutually exclusive
of KRAS mutations. BRAF mutation was associated
with poor prognosis (reduced progression-free and
overall survival) in patients (n=113) with metastatic
colorectal cancer given anti-EGFR antibodies.198 The
predictive value of BRAF mutations is yet to be
prospectively assessed, and although the results of the
CRYSTAL study199 suggested no association with
resistance to rst-line cetuximab in combination with
chemotherapy, statistical power was low.
High-frequency microsatellite instability is present
in almost 22% of stage II colorectal cancers compared
with in 12% of stage III tumours.200 In a pooled analysis
of 7642 patients treated with adjuvant uorouracil,
tumours with high-frequency microsatellite instability
were associated with signicantly improved prognosis
compared with tumours that were microsatellite
stable.201 Data for the prognostic eect of this
microsatellite instability were contradictory in patients
given
irinotecan-based
chemotherapy,200,202
and
subgroup analysis suggested a stronger eect in stage
II than in stage III colorectal cancer, possibly
attributable to stage-specic biological eects of
microsatellite instability.200 Patients who have tumours
with high-frequency microsatellite instability do not
seem to benet from adjuvant uorouracil.203 These
ndings were corroborated in a pooled analysis, which
1041

Seminar

also showed that decient mismatch repair (a surrogate


for high-frequency microsatellite instability) was
associated with a signicant reduction in overall
survival (hazard ratio 315, p=003) in patients with
stage II disease given adjuvant uorouracil.204 Hence
microsatellite-instability status might be used to decide
which of these patients (about a fth of those with
stage II colorectal cancer) should not be treated with
adjuvant uorouracil.200
Results from studies have suggested an association
between loss of heterozygosity at chromosome 18q
with poor prognosis in patients with colorectal
cancer.205208 Although the microsatellite-stability status
was not assessed in all the studies, it is an important
covariable since loss of heterozygosity of 18q is rarely
noted in tumours with high-frequency microsatellite
instability. Allelic imbalance at 18q had a negative eect
on overall survival in patients with stage III
microsatellite-stable colorectal cancers,208 but this result
was not replicated in other studies.209212 The 18q region
contains several important candidate genes, including
SMAD7, SMAD4, SMAD2, and DCC, and, in stage III
colon cancer, reduced SMAD4 expression was
associated with poor prognosis with uorouracil-based
chemotherapy.213,214 18q imbalance might also be a
surrogate marker for the complex chromosomal
instability phenotype arising in most colon tumours.215,216
An investigation of the use of microsatellite instability
and 18q loss of heterozygosity to decide which adjuvant
treatment should be assigned in patients with stage II
colorectal cancer is in progress (no treatment if
tumour has high-frequency microsatellite instability or
retention of 18q).
Other extensively studied potential prognostic and
predictive markers include thymidylate synthase (the
primary target of the active metabolite of uorouracil),
excision-repair cross-complementing-1 (implicated in
resistance or sensitivity to platinum drugs), VEGF and its
receptors, and the interleukins (table 5).

Conclusions
There have been considerable advances in understanding
the molecular pathogenesis, in diagnosis (hereditary
and sporadic), and in treatment of colorectal cancer.
Despite the use of active targeted drugs for treatment of
metastatic colorectal cancer in the past decade, and
improvement of overall survival to nearly 2 years for
non-resectable disease, cure rates remain low. Parallel
development of predictive molecular and clinical
markers is paramount to achieve the best outcomes
from targeted treatments, and KRAS is the only
validated
predictive
molecular
marker
in
colorectal cancer for EGFR-directed monoclonal
antibodies. Clinical and translational research that is in
progress will hopefully help to provide the much
promised hope of personalised medicine in the
management of this cancer.
1042

Contributors
All authors provided nal review and approval for the Seminar, and input
into responding to reviewers comments. DC co-wrote sections about
adjuvant treatment for early disease and treatment of metastatic disease,
and also provided overall structure and editing for the Seminar. WA wrote
the section about screening. HJL wrote sections about molecular
carcinogenesis, and prognostic and predictive molecular markers. HTL
wrote the section about genetic epidemiology. BM wrote the section about
radiation for rectal cancer. BN wrote the section about diagnosis and
staging, and surgery. NS co-wrote sections about adjuvant treatment for
early disease and treatment of metastatic disease, collated individual
sections for the whole Seminar, and provided the search strategy.
Conicts of interests
BM has been on speakers bureau for Sano-Aventis, Genentech, and
Roche; and has been a consultant for Sano-Aventis. HJL is a consultant
for Response Genetics. BMS is a consultant for ImClone and Merck,
and holds stock options for Response Genetics. DC has received
research funding from Sano-Aventis, Roche, Amgen, Pzer, Merck,
Novartis. WA, BN, and NS declare that they have no conicts of interest.
Acknowledgments
DC is part funded by the National Institute for Health Research
Biomedical Research Centre.
References
1
Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002.
CA Cancer J Clin 2005; 55:74108.
2
Vogelstein B, Fearon ER, Hamilton SR, et al. Genetic alterations
during colorectal-tumor development. N Engl J Med 1988;
319: 52532.
3
Nosinger AE. Serrated polyps and colorectal cancer: new pathway
to malignancy. Annu Rev Pathol 2009; 4: 34364.
4
Lengauer C, Kinzler KW, Vogelstein B. Genetic instabilities in
human cancers. Nature 1998; 396: 64349.
5
Lothe RA, Peltomaki P, Meling GI, et al. Genomic instability in
colorectal cancer: relationship to clinicopathological variables and
family history. Cancer Res 1993; 53: 584952.
6
Vogelstein B, Fearon ER, Hamilton SR, et al. Genetic alterations
during colorectal-tumor development. N Engl J Med 1988; 319: 52532.
7
Vogelstein B, Fearon ER, Kern SE, et al. Allelotype of colorectal
carcinomas. Science 1989; 244: 20711.
8
Aaltonen LA, Peltomaki P, Leach FS, et al. Clues to the
pathogenesis of familial colorectal cancer. Science 1993; 260: 81216.
9
Ionov Y, Peinado MA, Malkhosyan S, Shibata D, Perucho M.
Ubiquitous somatic mutations in simple repeated sequences reveal
a new mechanism for colonic carcinogenesis. Nature 1993;
363: 55861.
10 Peltomaki P. Role of DNA mismatch repair defects in the
pathogenesis of human cancer. J Clin Oncol 2003; 21: 117479.
11 Kane MF, Loda M, Gaida GM, et al. Methylation of the hMLH1
promoter correlates with lack of expression of hMLH1 in sporadic
colon tumors and mismatch repair-defective human tumor cell
lines. Cancer Res 1997; 57: 80811.
12 Cunningham JM, Christensen ER, Tester DJ, et al.
Hypermethylation of the hMLH1 promoter in colon cancer with
microsatellite instability. Cancer Res 1998; 58: 345560.
13 Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP.
CpG island methylator phenotype in colorectal cancer.
Proc Natl Acad Sci USA 1999; 96: 868186.
14 Shen L, Toyota M, Kondo Y, et al. Integrated genetic and epigenetic
analysis identies three dierent subclasses of colon cancer.
Proc Natl Acad Sci USA 2007; 104: 1865459.
15 Young J, Jenkins M, Parry S, et al. Serrated pathway colorectal
cancer in the population: genetic consideration. Gut 2007;
56: 145359.
16 Colorectal cancer risk factors. 2009. http://www.cdc.
gov/cancer/colorectal/basic_info/risk_factors.htm (accessed Feb 2,
2010).
17 von Roon AC, Reese G, Teare J, Constantinides V, Darzi AW,
Tekkis PP. The risk of cancer in patients with Crohns disease.
Dis Colon Rectum 2007; 50: 83955.
18 Eaden JA, Abrams KR, Mayberry JF. The risk of colorectal cancer in
ulcerative colitis: a meta-analysis. Gut 2001; 48: 52635.

www.thelancet.com Vol 375 March 20, 2010

Seminar

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

Itzkowitz SH, Harpaz N. Diagnosis and management of dysplasia


in patients with inammatory bowel diseases. Gastroenterology 2004;
126: 163448.
Triantallidis JK, Nasioulas G, Kosmidis PA. Colorectal cancer and
inammatory bowel disease: epidemiology, risk factors, mechanisms
of carcinogenesis and prevention strategies. Anticancer Res 2009;
29: 272737.
Hampel H, Frankel WL, Martin E, et al. Feasibility of screening for
Lynch syndrome among patients with colorectal cancer.
J Clin Oncol 2008; 26: 578388.
Watson P, Vasen HFA, Mecklin J-P, et al. The risk of extra-colonic,
extra-endometrial cancer in the Lynch syndrome. Int J Cancer 2008;
123: 44449.
Barrow E, Robinson L, Alduaij W, et al. Cumulative lifetime incidence
of extracolonic cancers in Lynch syndrome: a report of 121 families
with proven mutations. Clin Genet 2009; 75: 14149.
Bussey H. Familial polyposis coli: Family studies, histopathology,
dierential diagnosis, and results of treatment. Baltimore:
Johns Hopkins University Press, 1975.
Bulow S, Faurschou NT, Bulow C, Bisgaard ML, Karlsen L,
Moesgaard F. The incidence rate of familial adenomatous polyposis.
Results from the Danish Polyposis Register. Int J Colorectal Dis 1996;
11: 8891.
Vasen HF, Watson P, Mecklin JP, Lynch HT. New clinical criteria for
hereditary nonpolyposis colorectal cancer (HNPCC, Lynch syndrome)
proposed by the International Collaborative group on HNPCC.
Gastroenterology 1999; 116: 145356.
Lindor NM, Rabe K, Petersen GM, et al. Lower cancer incidence in
Amsterdam-I criteria families without mismatch repair deciency:
familial colorectal cancer type X. JAMA 2005; 293: 197985.
Llor X, Pons E, Xicola RM, et al. Dierential features of colorectal
cancers fullling Amsterdam criteria without involvement of the
mutator pathway. Clin Cancer Res 2005; 11: 730410.
Umar A, Boland CR, Terdiman JP, et al. Revised Bethesda Guidelines
for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and
microsatellite instability. J Natl Cancer Inst 2004; 96: 26168.
Lynch HT, Boland CR, Rodriguez-Bigas MA, Amos C, Lynch JF, Lynch
PM. Who should be sent for genetic testing in hereditary colorectal
cancer syndromes? J Clin Oncol 2007; 25: 353442.
Engel C, Forberg J, Holinski-Feder E, et al. Novel strategy for optimal
sequential application of clinical criteria, immunohistochemistry and
microsatellite analysis in the diagnosis of hereditary nonpolyposis
colorectal cancer. Int J Cancer 2006; 118: 11522.
Lynch HT, Lynch JF, Lynch PM. Toward a consensus in molecular
diagnosis of hereditary nonpolyposis colorectal cancer (Lynch
syndrome). J Natl Cancer Inst 2007; 99: 26163.
Bessa X, Balleste B, Andreu M, et al. A prospective, multicenter,
population-based study of BRAF mutational analysis for Lynch
syndrome screening. Clin Gastroenterol Hepatol 2008; 6: 20614.
Senter L, Clendenning M, Sotamaa K, et al. The clinical phenotype of
Lynch syndrome due to germ-line PMS2 mutations. Gastroenterology
2008; 135: 41928.
Gerard J, Azria D, Gourgou-Bourgade S, et al. Randomized
multicenter phase III trial comparing two neoadjuvant
chemoradiotherapy (CT-RT) regimens (RT45-Cap versus RT50-Capox)
in patients (pts) with locally advanced rectal cancer (LARC): Results of
the ACCORD 12/0405 PRODIGE 2. Proc ASCO Meeting Abstracts
2009; 27 (18 suppl): LBA4007.
Aschele C, Pinto C, Cordio S, et al. Preoperative uorouracil (FU)based chemoradiation with and without weekly oxaliplatin in locally
advanced rectal cancer: Pathologic response analysis of the Studio
Terapia Adiuvante Retto (STAR)-01 randomized phase III trial. Proc
ASCO Meeting Abstracts 2009; 27 (18 suppl): CRA4008.
Crane CH, Eng C, Feig BW, et al. Phase II trial of neoadjuvant
bevacizumab (BEV), capecitabine (CAP), and radiotherapy (XRT) for
locally advanced rectal cancer. Proc ASCO Meeting Abstracts 2008;
26 (15 suppl): 4091.
Willett CG, Duda DG, di TE, et al. Ecacy, safety, and biomarkers of
neoadjuvant bevacizumab, radiation therapy, and uorouracil in rectal
cancer: a multidisciplinary phase II study. J Clin Oncol 2009;
27: 302026.
Rodel C, Arnold D, Hipp M, et al. Phase I-II trial of cetuximab,
capecitabine, oxaliplatin, and radiotherapy as preoperative treatment
in rectal cancer. Int J Radiat Oncol Biol Phys 2008; 70: 108186.

www.thelancet.com Vol 375 March 20, 2010

40

41

42

43

44

45

46
47

48

49

50

51

52

53

54

55

56

57

58

59

60

Mohiuddin M, Winter K, Mitchell E, et al. Randomized phase II


study of neoadjuvant combined-modality chemoradiation for distal
rectal cancer: Radiation Therapy Oncology Group Trial 0012.
J Clin Oncol 2006; 24: 65055.
Wong SJ, Winter K, Meropol NJ, et al. RTOG 0247: A randomized
phase II study of neoadjuvant capecitabine and irinotecan versus
capecitabine and oxaliplatin with concurrent radiation therapy for
locally advanced rectal cancer. Proc ASCO Meeting Abstracts 2008;
26 (15 suppl): 4021.
Fernndez-Martos C, Aparicio J, Salud A, et al. Multicenter
randomized phase II study of chemoradiation (CRT) followed by
surgery (S) and chemotherapy (CT) versus induction CT followed
by CRT and S in high-risk rectal cancer: GCR-3 nal ecacy and
safety results. Proc ASCO Meeting Abstracts 2009; 27: 4103.
Evaluation of Genomic Applications in Practice and Prevention
(EGAPP) Working Group. Recommendations from the EGAPP
Working Group: genetic testing strategies in newly diagnosed
individuals with colorectal cancer aimed at reducing morbidity and
mortality from Lynch syndrome in relatives. Genet Med 2009; 11: 3541.
Jarvinen HJ, Aarnio M, Mustonen H, et al. Controlled 15-year trial
on screening for colorectal cancer in families with hereditary
nonpolyposis colorectal cancer. Gastroenterology 2000; 118: 82934.
Dove-Edwin I, Sasieni P, Adams J, Thomas HJ. Prevention of
colorectal cancer by colonoscopic surveillance in individuals with a
family history of colorectal cancer: 16 year, prospective, follow-up
study. BMJ 2005; 331: 1047.
Lynch HT, de la CA. Genetic susceptibility to non-polyposis
colorectal cancer. J Med Genet 1999; 36: 80118.
Schmeler KM, Lynch HT, Chen LM, et al. Prophylactic surgery to
reduce the risk of gynecologic cancers in the Lynch syndrome.
N Engl J Med 2006; 354: 26169.
American Cancer Society. Detailed guide: colon and rectum cancer.
What are the risk factors for colorectal cancer? May 18, 2009. http://
www.cancer.org/docroot/CRI/content/CRI_2_4_2X_What_are_the_
risk_factors_for_colon_and_rectum_cancer.asp (accessed Feb 2, 2010).
Verdecchia A, Francisci S, Brenner H, et al. Recent cancer survival
in Europe: a 200002 period analysis of EUROCARE-4 data. Lancet
Oncol 2007; 8: 78496.
Tappenden P, Chilcott J, Eggington S, Sakai H, Karnon J, Patnick J.
Option appraisal of population-based colorectal cancer screening
programmes in England. Gut 2007; 56: 67784.
Loeve F, Brown ML, Boer R, van BM, van Oortmarssen GJ,
Habbema JD. Endoscopic colorectal cancer screening: a cost-saving
analysis. J Natl Cancer Inst 2000; 92: 55763.
Towler B, Irwig L, Glasziou P, Kewenter J, Weller D, Silagy C. A
systematic review of the eects of screening for colorectal cancer
using the faecal occult blood test, hemoccult. BMJ 1998; 317: 55965.
Collins JF, Lieberman DA, Durbin TE, Weiss DG. Accuracy of
screening for fecal occult blood on a single stool sample obtained by
digital rectal examination: a comparison with recommended
sampling practice. Ann Intern Med 2005; 142: 8185.
Steele RJ, Parker R, Patnick J, et al. A demonstration pilot trial for
colorectal cancer screening in the United Kingdom: a new concept in
the introduction of healthcare strategies. J Med Screen 2001; 8: 197202.
Benson VS, Patnick J, Davies AK, Nadel MR, Smith RA, Atkin WS.
Colorectal cancer screening: a comparison of 35 initiatives in
17 countries. Int J Cancer 2008; 122: 135767.
Pignone M, Saha S, Hoerger T, Mandelblatt J. Cost-eectiveness
analyses of colorectal cancer screening: a systematic review for the
US Preventive Services Task Force. Ann Intern Med 2002;
137: 96104.
Whitlock EP, Lin JS, Liles E, Beil TL, Fu R. Screening for
colorectal cancer: a targeted, updated systematic review for the U.S.
Preventive Services Task Force. Ann Intern Med 2008; 149: 63858.
Castiglione G, Grazzini G, Miccinesi G, et al. Basic variables at
dierent positivity thresholds of a quantitative immunochemical
test for faecal occult blood. J Med Screen 2002; 9: 99103.
Atkin WS, Hart A, Edwards R, et al. Single blind, randomised trial
of ecacy and acceptability of oral picolax versus self administered
phosphate enema in bowel preparation for exible sigmoidoscopy
screening. BMJ 2000; 320: 150408.
Selby JV, Friedman GD, Quesenberry CP Jr, Weiss NS. A
case-control study of screening sigmoidoscopy and mortality from
colorectal cancer. N Engl J Med 1992; 326: 65357.

1043

Seminar

61

62

63

64

65

66

67
68

69

70

71

72

73

74

75

76

77

78
79

80

81

82

1044

Newcomb PA, Storer BE, Morimoto LM, Templeton A,


Potter JD. Long-term ecacy of sigmoidoscopy in the reduction
of colorectal cancer incidence. J Natl Cancer Inst 2003;
95: 62225.
Atkin WS, Edwards R, Wardle J, et al. Design of a multicentre
randomised trial to evaluate exible sigmoidoscopy in colorectal
cancer screening. J Med Screen 2001; 8: 13744.
Segnan N, Senore C, Andreoni B, et al. Baseline ndings of the
Italian multicenter randomized controlled trial of once-only
sigmoidoscopySCORE. J Natl Cancer Inst 2002; 94: 176372.
Weissfeld JL, Schoen RE, Pinsky PF, et al. Flexible sigmoidoscopy
in the PLCO cancer screening trial: results from the baseline
screening examination of a randomized trial. J Natl Cancer Inst
2005; 97: 98997.
Bretthauer M, Gondal G, Larsen K, et al. Design, organization and
management of a controlled population screening study for
detection of colorectal neoplasia: attendance rates in the NORCCAP
study (Norwegian Colorectal Cancer Prevention).
Scand J Gastroenterol 2002; 37: 56873.
Pignone M, Rich M, Teutsch SM, Berg AO, Lohr KN. Screening for
colorectal cancer in adults at average risk: a summary of the
evidence for the U.S. Preventive Services Task Force.
Ann Intern Med 2002; 137: 13241.
Atkin WS, Northover JMA, Cuzick J, Whynes DK. Prevention of
colorectal cancer by once-only sigmoidoscopy. Lancet 1993; 341: 73640.
Lako J, Paszat LF, Saskin R, Rabeneck L. Risk of developing
proximal versus distal colorectal cancer after a negative
colonoscopy: a population-based study. Clin Gastroenterol Hepatol
2008; 6: 111721.
Baxter NN, Goldwasser MA, Paszat LF, Saskin R, Urbach DR,
Rabeneck L. Association of colonoscopy and death from colorectal
cancer. Ann Intern Med 2009; 150: 18.
Pignone M, Rich M, Teutsch S, Berg A, Lohr K. Screening for
colorectal cancer in adults. Systematic evidence review number 7.
Rockville: Agency for Healthcare Research and Quality, 2002.
http:// www.ahrq.gov/clinic/serles.htm (accessed Feb 2, 2010).
Pox C, Schmiegel W, Classen M. Current status of screening
colonoscopy in Europe and in the United States. Endoscopy 2007;
39: 16873.
Regula J, Rupinski M, Kraszewska E, et al. Colonoscopy in
colorectal-cancer screening for detection of advanced neoplasia.
N Engl J Med 2006; 355: 186372.
Halligan S, Altman DG, Taylor SA, et al. CT colonography in the
detection of colorectal polyps and cancer: systematic review, metaanalysis, and proposed minimum data set for study level reporting.
Radiology 2005; 237: 893904.
Pickhardt PJ, Hanson ME, Vanness DJ, et al. Unsuspected
extracolonic ndings at screening CT colonography: clinical and
economic impact. Radiology 2008; 249: 15159.
Vijan S, Hwang I, Inadomi J, et al. The cost-eectiveness of CT
colonography in screening for colorectal neoplasia.
Am J Gastroenterol 2007; 102: 38090.
Imperiale TF, Ransoho DF, Itzkowitz SH, Turnbull BA,
Ross ME. Fecal DNA versus fecal occult blood for colorectalcancer screening in an average-risk population.
N Engl J Med 2004; 351: 270414.
Diehl F, Schmidt K, Durkee KH, et al. Analysis of mutations in
DNA isolated from plasma and stool of colorectal cancer patients.
Gastroenterology 2008; 135: 48998.
Fockens P. Advances in colonic imaging. Eur J Gastroenterol Hepatol
2005; 17: 78384.
Mauchley DC, Lynge DC, Langdale LA, Stelzner MG, Mock CN,
Billingsley KG. Clinical utility and cost-eectiveness of routine
preoperative computed tomography scanning in patients with colon
cancer. Am J Surg 2005; 189: 51217.
Mainenti PP, Romano M, Imbriaco M, et al. Added value of CT
colonography after a positive conventional colonoscopy: impact on
treatment strategy. Abdom Imaging 2005; 30: 4247.
Copel L, Sosna J, Kruskal JB, Raptopoulos V, Farrell RJ,
Morrin MM. CT colonography in 546 patients with incomplete
colonoscopy. Radiology 2007; 244: 47178.
Morrin MM, Kruskal JB, Farrell RJ, Goldberg SN, McGee JB,
Raptopoulos V. Endoluminal CT colonography after an incomplete
endoscopic colonoscopy. AJR Am J Roentgenol 1999; 172: 91318.

83

84

85

86

87
88

89

90

91
92

93

94

95

96

97

98

99

100
101

102

103

Furukawa H, Ikuma H, Seki A, et al. Positron emission


tomography scanning is not superior to whole body multidetector
helical computed tomography in the preoperative staging of
colorectal cancer. Gut 2006; 55: 100711.
Brown G, Radclie AG, Newcombe RG, Dallimore NS, Bourne
MW, Williams GT. Preoperative assessment of prognostic factors in
rectal cancer using high-resolution magnetic resonance imaging.
Br J Surg 2003; 90: 35564.
Bipat S, Glas AS, Slors FJ, Zwinderman AH, Bossuyt PM, Stoker J.
Rectal cancer: local staging and assessment of lymph node
involvement with endoluminal US, CT, and MR imaging. A
meta-analysis. Radiology 2004; 232: 77383.
Fioole B, de Haas RJ, Wicherts DA, et al. Additional value of
contrast enhanced intraoperative ultrasound for colorectal liver
metastases. Eur J Radiol 2008; 67: 16976.
Ward J. New MR techniques for the detection of liver metastases.
Cancer Imaging 2006; 6: 3342.
Bipat S, van Leeuwen MS, Comans EF, et al. Colorectal liver
metastases: CT, MR imaging, and PET for diagnosismetaanalysis. Radiology 2005; 237: 12331.
Truant S, Huglo D, Hebbar M, Ernst O, Steinling M, Pruvot FR.
Prospective evaluation of the impact of [18F]uoro-2-deoxy-Dglucose positron emission tomography of resectable colorectal liver
metastases. Br J Surg 2005; 92: 36269.
Gonzalez-Moreno S, Gonzalez-Bayon L, Ortega-Perez G,
Gonzalez-Hernando C. Imaging of peritoneal carcinomatosis.
Cancer J 2009; 15: 18489.
Sobin LH, Wittikind C. TMN classication of malignant tumours.
7th edn. London: Wiley and sons, 2010.
Gunderson LL, Jessup JM, Sargent DJ, Greene FL, Stewart AK.
Revised TN categorization for colon cancer based on national
survival outcomes data. J Clin Oncol 2010; 28: 26471.
Le Voyer TE, Sigurdson ER, Hanlon AL, et al. Colon cancer survival
is associated with increasing number of lymph nodes analyzed:
a secondary survey of intergroup trial INT-0089. J Clin Oncol 2003;
21: 291219.
Chang GJ, Rodriguez-Bigas MA, Skibber JM, Moyer VA.
Lymph node evaluation and survival after curative resection of
colon cancer: systematic review. J Natl Cancer Inst 2007;
99: 43341.
Prandi M, Lionetto R, Bini A, et al. Prognostic evaluation of stage B
colon cancer patients is improved by an adequate lymphadenectomy:
results of a secondary analysis of a large scale adjuvant trial.
Ann Surg 2002; 235: 45863.
Quirke P, Steele R, Monson J, et al, on behalf of the MRC
CR07/NCIC-CTG CO16 trial investigators and the NCRI colorectal
cancer study group. Eect of the plane of surgery achieved on local
recurrence in patients with operable rectal cancer: a prospective
study using data from the MRC CR07 and NCIC-CTG CO16
randomised clinical trial. Lancet 2009; 373: 82128.
Peeters KC, Marijnen CA, Nagtegaal ID, et al. The TME trial after a
median follow-up of 6 years: increased local control but no survival
benet in irradiated patients with resectable rectal carcinoma.
Ann Surg 2007; 246: 693701.
Bosset JF, Collette L, Calais G, et al. Chemotherapy with
preoperative radiotherapy in rectal cancer.
N Engl J Med 2006; 355: 111423.
Rullier E, Laurent C, Bretagnol F, Rullier A, Vendrely V, Zerbib F.
Sphincter-saving resection for all rectal carcinomas: the end of the
2-cm distal rule. Ann Surg 2005; 241: 46569.
Chamlou R, Parc Y, Simon T, et al. Long-term results of intersphincteric resection for low rectal cancer. Ann Surg 2007; 246: 91621.
Cornish JA, Tilney HS, Heriot AG, Lavery IC, Fazio VW, Tekkis PP.
A meta-analysis of quality of life for abdominoperineal excision of
rectum versus anterior resection for rectal cancer.
Ann Surg Oncol 2007; 14: 205668.
Bretagnol F, Rullier E, Laurent C, Zerbib F, Gontier R, Saric J.
Comparison of functional results and quality of life between
intersphincteric resection and conventional coloanal anastomosis for
low rectal cancer. Dis Colon Rectum 2004; 47: 83238.
van DP, Slors JF, Taat CW, et al. Prospective evaluation of anorectal
function after total mesorectal excision for rectal carcinoma with or
without preoperative radiotherapy. Am J Gastroenterol 2002;
97: 228289.

www.thelancet.com Vol 375 March 20, 2010

Seminar

104 Nascimbeni R, Burgart LJ, Nivatvongs S, Larson DR. Risk of lymph


node metastasis in T1 carcinoma of the colon and rectum.
Dis Colon Rectum 2002; 45: 20006.
105 Tytherleigh MG, Warren BF, Mortensen NJ. Management of early
rectal cancer. Br J Surg 2008; 95: 40923.
106 Bach SP, Hill J, Monson JR, et al. A predictive model for local
recurrence after transanal endoscopic microsurgery for rectal cancer.
Br J Surg 2009; 96: 28090.
107 Leung KL, Kwok SPY, Lam SCW, et al. Laparoscopic resection of
rectosigmoid carcinoma: prospective randomised trial. Lancet 2004;
363: 118792.
108 Guillou PJ, Quirke P, Thorpe H, et al, for the MRC CLASICC trial
group. Short-term endpoints of conventional versus laparoscopicassisted surgery in patients with colorectal cancer (MRC CLASICC
trial): multicentre, randomised controlled trial. Lancet 2005;
365: 171826.
109 Lacy AM, Delgado S, Castells A, et al. The long-term results of a
randomized clinical trial of laparoscopy-assisted versus open surgery
for colon cancer. Ann Surg 2008; 248: 17.
110 A comparison of laparoscopically assisted and open colectomy for
colon cancer. N Engl J Med 2004; 350: 205059.
111 Hewett PJ, Allardyce RA, Bagshaw PF, et al. Short-term outcomes of
the Australasian randomized clinical study comparing laparoscopic
and conventional open surgical treatments for colon cancer: the
ALCCaS trial. Ann Surg 2008; 248: 72838.
112 Jayne DG, Guillou PJ, Thorpe H, et al. Randomized trial of
laparoscopic-assisted resection of colorectal carcinoma: 3-year
results of the UK MRC CLASICC Trial Group. J Clin Oncol 2007;
25: 306168.
113 Jayne DG, Brown JM, Thorpe H, Walker J, Quirke P, Guillou PJ.
Bladder and sexual function following resection for rectal cancer in
a randomized clinical trial of laparoscopic versus open technique.
Br J Surg 2005; 92: 112432.
114 Nordlinger B, Sorbye H, Glimelius B, et al, for the EORTC
Gastro-Intestinal Tract Cancer Group, Cancer Research UK,
Arbeitsgruppe Lebermetastasen undtumoren in der
Chirurgischen Arbeitsgemeinschaft Onkologie (ALM-CAO),
Australasian Gastro-Intestinal Trials Group (AGITG), and
Fdration Francophone de Cancrologie Digestive (FFCD).
Perioperative chemotherapy with FOLFOX4 and surgery versus
surgery alone for resectable liver metastases from colorectal
cancer (EORTC Intergroup trial 40983): a randomised controlled
trial. Lancet 2008; 371: 100716.
115 Negri F, Musolino A, Cunningham D, Pastorino U, Ladas G,
Norman AR. Retrospective study of resection of pulmonary
metastases in patients with advanced colorectal cancer: the
development of a preoperative chemotherapy strategy.
Clin Colorectal Cancer 2004; 4: 10106.
116 Ruers T, van Coevorden F, Pierie J, et al. Radiofrequency
ablation (RFA) combined with chemotherapy for unresectable
colorectal liver metastases (CRC LM): interim results of a randomised
phase II study of the EORTC-NCRI CCSG-ALM Intergroup 40004
(CLOCC). J Clin Oncol 2008; 26 (suppl): 4012 (abstr).
117 Sauer R, Becker H, Hohenberger W, et al. Preoperative versus
postoperative chemoradiotherapy for rectal cancer. N Engl J Med
2004; 351: 173140.
118 MERCURY Study Group. Diagnostic accuracy of preoperative
magnetic resonance imaging in predicting curative resection of
rectal cancer: prospective observational study. BMJ 2006;
333: 779.
119 Burton S, Brown G, Daniels I, et al. MRI identied prognostic
features of tumors in distal sigmoid, rectosigmoid, and upper
rectum: treatment with radiotherapy and chemotherapy.
Int J Radiat Oncol Biol Phys 2006; 65: 44551.
120 Rutten H, Sebag-Monteore D, Glynne-Jones R, et al. Capecitabine,
oxaliplatin, radiotherapy, and excision (CORE) in patients with
MRI-dened locally advanced rectal ademocarcinoma: results of an
international multicenter phase II study. Proc ASCO 2006; 24: 153s
(abstr).
121 Sebag-Monteore D, Stephens RJ, Steele R, et al, on behalf of all the
trial collaborators. Preoperative radiotherapy versus selective
postoperative chemoradiotherapy in patients with rectal cancer
(MRC CR07 and NCIC-CTG C016): a multicentre, randomised trial.
Lancet 2009; 373: 81120.

www.thelancet.com Vol 375 March 20, 2010

122 Marijnen CAM, Nagtegaal ID, Kapiteijn E, et al, Cooperative


investigators of the Dutch Colorectal Cancer Group. Radiotherapy
does not compensate for positive resection margins in rectal cancer
patients: report of a multicenter randomized trial.
Int J Radiat Oncol Biol Phys 2003; 55: 131120.
123 Guillem JG, Diaz-Gonzalez J, Minsky BD, et al. cT3N0 rectal cancer:
potential overtreatment with preoperative chemoradiotherapy is
warranted. J Clin Oncol 2008; 26: 36873.
124 Birgisson H, Pahlman L, Glimelius B. Adverse eects of
preoperative radiation therapy for rectal cancer: long-term follow-up
of the Swedish Rectal Cancer Trial. J Clin Oncol 2005; 23: 869705.
125 Sauer R, Rodel C. Chemoradiotherapy for rectal cancer.
N Engl J Med 2005; 352: 50911.
126 Peeters KCMJ, Marijnen CAM, Nagtegaal ID, et al, Dutch
Colorectal Cancer group. The TME trial after a median follow-up
of 6 years: increased local control but no survival benet in
irradiated patients with resectable rectal carcinoma. Ann Surg
2007; 246: 693701.
127 Bujko K, Nowacki MP, Nasierowska-Guttmejer A, Michalski W,
Bebenek M, Kryj M. Long term results of a randomized trial
comparing preoperative short-course radiotherapy with preperative
conventionally fractionated chemoradiation for rectal cancer.
BrJ Surg 2006; 93: 121523.
128 Bosset JF, Collette L, Bardet E, Beny A, Ollier JC. Chemotherapy
with preoperative radiotherapy in rectal cancer. N Engl J Med 2006;
355: 111423.
129 Gerard JP, Conroy T, Bonnetain F. Preoperative radiotherapy with
or without concurrent uorouracil and leucovorin in T3-4 rectal
cancers: results of FFCD 9203. J Clin Oncol 2006; 28: 462025.
130 Guillem JG, Chessin DB, Cohen AM, et al. Long term oncologic
outcome following preoperative combined modality therapy and
total mesorectal excision of locally advanced rectal cancer. Ann Surg
2005; 241: 82938.
131 Bertolini F, Bengala C, Losi L, et al. Prognostic and predictive value
of baseline and posttreatment molecular marker expression in
locally advanced rectal cancer treated with neoadjuvant
chemoradiotherapy. Int J Radiat Oncol Biol Phys 2007; 68: 145561.
132 Johnston PG. Prognostic markers of local relapse in rectal cancer:
are we any further forward? J Clin Oncol 2006; 24: 404950.
133 Glynne-Jones R, Wallace M, Livingstone JI, Meyrick Thomas J.
Complete clinical response after preoeprative chemoradiation in
rectal cancer: is a wait and see policy justied? Dis Colon Rectum
2008; 51: 1020.
134 Habr-Gama A, Santinho B, de Souza PM, et al. Low rectal cancer.
Impact of radiation and chemotherapy on surgical treatment.
Dis Colon Rectum 1998; 41: 108796.
135 Ceelen W, Boterberg T, Pattyn P, et al. Neoadjuvant chemoradiation
versus hyperfractionated accelerated radiotherapy in locally
advanced rectal cancer. Ann Surg Oncol 2007; 14: 42431.
136 Meyer J, Czito B, Yin FF, Willett C. Advanced radiation therapy
technologies in the treatment of rectal and anal cancer: intensitymodulated photon therapy and proton therapy.
Clin Colorectal Cancer 2007; 6: 34856.
137 Aristu JJ, Arbea L, Rodriguez J, et al. Phase I-II trial of concurrent
capeciabine and oxaliplatin with preoperative intensity modulated
radiotherapy in patients with locally advanced rectal cancer.
Int J Radiat Oncol Biol Phys 2008; 71: 74855.
138 Callister MD, Ezzell GA, Gunderson LL. IMRT reduces the dose to
small bowel and other pelvic organs in the preoperative treatment
of rectal cancer. Int J Radiat Oncol Biol Phys 2006; 66: s290 (abstr).
139 NCCN Clinical Practice Guidelines in Oncology. 2009. http://www.
nccn.org/professionals/physician_gls/f_guidelines.asp (accessed
Nov 11, 2009).
140 International Multicentre Pooled Analysis of Colon Cancer Trials
(IMPACT) investigators. Ecacy of adjuvant uorouracil and
folinic acid in colon cancer. Lancet 1995; 345: 93944.
141 Twelves C, Wong A, Nowacki MP, et al. Capecitabine as adjuvant
treatment for stage III colon cancer. N Engl J Med 2005;
352: 2696704.
142 Sargent DJ, Wieand HS, Haller DG, et al. Disease-free survival
versus overall survival as a primary end point for adjuvant colon
cancer studies: individual patient data from 20,898 patients on
18 randomized trials. J Clin Oncol 2005; 23: 866470.

1045

Seminar

143 Sargent DJ, Patiyil S, Yothers G, et al. End points for colon cancer
adjuvant trials: observations and recommendations based on individual
patient data from 20,898 patients enrolled onto 18 randomized trials
from the ACCENT Group. J Clin Oncol 2007; 25: 456974.
144 Andre T, Boni C, Mounedji-Boudiaf L, et al. Oxaliplatin,
uorouracil, and leucovorin as adjuvant treatment for colon cancer.
N Engl J Med 2004; 350: 234351.
145 Andre T, Boni C, Navarro M, et al. Improved overall survival with
oxaliplatin, uorouracil, and leucovorin as adjuvant treatment in
stage II or III colon cancer in the MOSAIC trial. J Clin Oncol 2009;
27: 310916.
146 Kuebler JP, Wieand HS, OConnell MJ, et al. Oxaliplatin combined
with weekly bolus uorouracil and leucovorin as surgical adjuvant
chemotherapy for stage II and III colon cancer: results from
NSABP C-07. J Clin Oncol 2007; 25: 2198204.
147 Haller D, Tabernero J, Maroun J, et al. First ecacy ndings from a
randomised phase III trial of capecitabine + oxaliplatin versus bolus
5FU/LV for stage III colon cancer (NO16968/XELOXA study).
Eur J Cancer 2009; 7 (suppl 3): 5LBA (abstr).
148 Land SR, Kopec JA, Cecchini RS, et al. Neurotoxicity from
oxaliplatin combined with weekly bolus uorouracil and leucovorin
as surgical adjuvant chemotherapy for stage II and III colon cancer:
NSABP C-07. J Clin Oncol 2007; 25: 220511.
149 Jackson McCleary NA, Meyerhardt J, Green E, et al. Impact of older
age on the ecacy of newer adjuvant therapies in >12,500 patients
(pts) with stage II/III colon cancer: Findings from the ACCENT
Database. Proc ASCO Meeting Abstracts 2009; 27 (15 suppl): 4010.
150 Saltz LB, Niedzwiecki D, Hollis D, et al. Irinotecan uorouracil plus
leucovorin is not superior to uorouracil plus leucovorin alone as
adjuvant treatment for stage III colon cancer: results of CALGB
89803. J Clin Oncol 2007; 25: 345661.
151 Ychou M, Raoul JL, Douillard JY, et al. A phase III randomised trial
of LV5FU2 + irinotecan versus LV5FU2 alone in adjuvant high-risk
colon cancer (FNCLCC Accord02/FFCD9802). Ann Oncol 2009;
20: 67480.
152 Van CE, Labianca R, Bodoky G, et al. Randomized phase III trial
comparing biweekly infusional uorouracil/leucovorin alone or
with irinotecan in the adjuvant treatment of stage III colon cancer:
PETACC-3. J Clin Oncol 2009; 27: 311725.
153 Cunningham D, Starling N. Adjuvant chemotherapy of colorectal
cancer. Lancet 2007; 370: 198081.
154 Quasar Collaborative Group. Adjuvant chemotherapy versus
observation in patients with colorectal cancer: a randomised study.
Lancet 2007; 370: 202029.
155 Fields AL, Keller A, Schwartzberg L, et al. Adjuvant therapy with the
monoclonal antibody Edrecolomab plus uorouracil-based therapy
does not improve overall survival of patients with stage III colon
cancer. J Clin Oncol 2009; 27: 194147.
156 Wolmark N, Yothers G, OConnell MJ, et al. A phase III trial
comparing mFOLFOX6 to mFOLFOX6 plus bevacizumab in stage
II or III carcinoma of the colon: Results of NSABP Protocol C-08.
Proc ASCO Meeting Abstracts 2009; 27 (18 suppl): LBA4.
157 Addition of Cetuximab to Standard Chemotherapy in Early-Stage
Colon Cancer Shows No Benet in Phase 3 Clinical Trial. 2009.
http://www.cancer.gov/newscenter/pressreleases/
CetuximabClosure (accessed March 3, 2010).
158 Renehan AG, Egger M, Saunders MP, ODwyer ST. Impact on
survival of intensive follow up after curative resection for colorectal
cancer: systematic review and meta-analysis of randomised trials.
BMJ 2002; 324: 813.
159 Jeery GM, Hickey BE, Hider P. Follow-up strategies for patients
treated for non-metastatic colorectal cancer.
Cochrane Database Syst Rev 2002; 1: CD002200.
160 Tsikitis VL, Malireddy K, Green EA, et al. Postoperative surveillance
recommendations for early stage colon cancer based on results
from the clinical outcomes of surgical therapy trial.
J Clin Oncol 2009; 27: 367176.
161 Chau I, Allen MJ, Cunningham D, et al. The value of routine
serum carcino-embryonic antigen measurement and computed
tomography in the surveillance of patients after adjuvant
chemotherapy for colorectal cancer. J Clin Oncol 2004; 22: 142029.
162 Van CE, Oliveira J. Primary colon cancer: ESMO clinical
recommendations for diagnosis, adjuvant treatment and followup. Ann Oncol 2009; 20 (suppl 4): 4950.

1046

163 Tournigand C, Andre T, Achille E, et al. FOLFIRI followed by


FOLFOX6 or the reverse sequence in advanced colorectal cancer:
a randomized GERCOR study. J Clin Oncol 2004; 22: 22937.
164 Cassidy J, Clarke S, az-Rubio E, et al. Randomized phase III study
of capecitabine plus oxaliplatin compared with uorouracil/folinic
acid plus oxaliplatin as rst-line therapy for metastatic colorectal
cancer. J Clin Oncol 2008; 26: 200612.
165 Porschen R, Arkenau HT, Kubicka S, et al. Phase III study of
capecitabine plus oxaliplatin compared with uorouracil and
leucovorin plus oxaliplatin in metastatic colorectal cancer: a nal
report of the AIO Colorectal Study Group. J Clin Oncol 2007;
25: 421723.
166 Grothey A, Sargent D, Goldberg RM, Schmoll HJ. Survival of
patients with advanced colorectal cancer improves with the
availability of uorouracil-leucovorin, irinotecan, and oxaliplatin
in the course of treatment. J Clin Oncol 2004; 22: 120914.
167 Seymour MT, Maughan TS, Ledermann JA, et al, for the FOCUS
Trial Investigators and the National Cancer Research Institute
Colorectal Clinical Studies Group. Dierent strategies of
sequential and combination chemotherapy for patients with poor
prognosis advanced colorectal cancer (MRC FOCUS):
a randomised controlled trial. Lancet 2007; 370: 14352.
168 Cunningham D, Sirohi B, Pluzanska A, et al. Two dierent
rst-line 5-uorouracil regimens with or without oxaliplatin in
patients with metastatic colorectal cancer. Ann Oncol 2009;
20: 24450.
169 Folprecht G, Grothey A, Alberts S, Raab HR, Kohne CH.
Neoadjuvant treatment of unresectable colorectal liver metastases:
correlation between tumour response and resection rates.
Ann Oncol 2005; 16: 131119.
170 Maughan TS, James RD, Kerr DJ, et al, on behalf of the Medical
Research Council Colorectal Cancer Group. Comparison of
intermittent and continuous palliative chemotherapy for advanced
colorectal cancer: a multicentre randomised trial. Lancet
2003; 361: 45764.
171 Tournigand C, Cervantes A, Figer A, et al. OPTIMOX1:
a randomized study of FOLFOX4 or FOLFOX7 with oxaliplatin in
a stop-and-Go fashion in advanced colorectal cancera GERCOR
study. J Clin Oncol 2006; 24: 394400.
172 Cunningham D, Humblet Y, Siena S, et al. Cetuximab monotherapy
and cetuximab plus irinotecan in irinotecan-refractory metastatic
colorectal cancer. N Engl J Med 2004; 351: 33745.
173 Jonker DJ, OCallaghan CJ, Karapetis CS, et al. Cetuximab for
the treatment of colorectal cancer. N Engl J Med 2007;
357: 204048.
174 Sobrero AF, Maurel J, Fehrenbacher L, et al. EPIC: phase III trial
of cetuximab plus irinotecan after uoropyrimidine and
oxaliplatin failure in patients with metastatic colorectal cancer.
J Clin Oncol 2008; 26: 231119.
175 Van Cutsem E, Peeters M, Siena S, et al. Open-label phase III trial
of panitumumab plus best supportive care compared with best
supportive care alone in patients with chemotherapy-refractory
metastatic colorectal cancer. J Clin Oncol 2007; 25: 165864.
176 Van Cutsem E, Kohne CH, Hitre E, et al. Cetuximab and
chemotherapy as initial treatment for metastatic colorectal cancer.
N Engl J Med 2009; 360: 140817.
177 Bokemeyer C, Bondarenko I, Makhson A, et al. Fluorouracil,
leucovorin, and oxaliplatin with and without cetuximab in the
rst-line treatment of metastatic colorectal cancer. J Clin Oncol
2009; 27: 66371.
178 Folprecht G, Gruenberger T, Bechstein WO, et al. Tumour response
and secondary resectability of colorectal liver metastases following
neoadjuvant chemotherapy with cetuximab: the CELIM randomised
phase 2 trial. Lancet Oncol 2010; 11: 3847.
179 Maughan T, Adams RA, Smith CG, et al. Addition of cetuximab to
oxaliplatin-based combination chemotherapy in patients with
KRAS wild-type advanced colorectal cancer: a randomised
superiority trial (MRC COIN). Eur J Cancer 2009; 7 (suppl): 6LBA
(abstr).
180 Siena S, Cassidy J, Tabernero J, et al. Randomized phase III study
of panitumumab (pmab) with FOLFOX4 compared to FOLFOX4
alone as rst-line treatment (tx) for metastatic colorectal cancer
(mCRC): PRIME trial. Gastrointestinal Symposium 2010; 283
(abstr).

www.thelancet.com Vol 375 March 20, 2010

Seminar

181 Peeters M, Price TJ, Hotko YS, et al. Randomized phase III study of
panitumumab (pmab) with FOLFIRI versus FOLFIRI alone as
second-line treatment (tx) in patients (pts) with metastatic colorectal
cancer (mCRC): patient-reported outcomes (PRO). Gastrointestinal
Symposium 2010; 282 (abstr).
182 Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus
irinotecan, uorouracil, and leucovorin for metastatic colorectal
cancer. N Engl J Med 2004; 350: 233542.
183 Giantonio BJ, Catalano PJ, Meropol NJ, et al. Bevacizumab in
combination with oxaliplatin, uorouracil, and leucovorin
(FOLFOX4) for previously treated metastatic colorectal cancer:
results from the Eastern Cooperative Oncology Group Study E3200.
J Clin Oncol 2007; 25: 15391544.
184 Saltz LB, Clarke S, az-Rubio E, et al. Bevacizumab in combination
with oxaliplatin-based chemotherapy as rst-line therapy in
metastatic colorectal cancer: a randomized phase III study.
J Clin Oncol 2008; 26: 201319.
185 Saltz LB, Lenz HJ, Kindler HL, et al. Randomized phase II trial of
cetuximab, bevacizumab, and irinotecan compared with cetuximab
and bevacizumab alone in irinotecan-refractory colorectal cancer:
the BOND-2 study. J Clin Oncol 2007; 25: 455761.
186 Hecht JR, Mitchell E, Chidiac T, et al. A randomized phase IIIB trial
of chemotherapy, bevacizumab, and panitumumab compared with
chemotherapy and bevacizumab alone for metastatic colorectal
cancer. J Clin Oncol 2009; 27: 67280.
187 Tol J, Koopman M, Cats A, et al. Chemotherapy, bevacizumab, and
cetuximab in metastatic colorectal cancer. N Engl J Med 2009;
360: 56372.
188 Wilke H, Glynne-Jones R, Thaler J, et al. Cetuximab plus irinotecan
in heavily pretreated metastatic colorectal cancer progressing on
irinotecan: MABEL Study. J Clin Oncol 2008; 26: 533543.
189 Van Cutsem E, Rougier P, Kohne CH, Stroh C, Schlichting M,
Bokemeyer C. A meta-analysis of the CRYSTAL and OPUS studies
combining cetuximab with chemotherapy as 1st-line treatment for
patients with metastatic colorectal cancer: results according to KRAS
and BRAF mutation status. Eur J Cancer 2009; 7: 6077 (abstr).
190 Karapetis CS, Khambata-Ford S, Jonker DJ, et al. K-ras mutations
and benet from cetuximab in advanced colorectal cancer.
N Engl J Med 2008; 359: 175765.
191 US Food and Drug Administration. http://www.fda.gov/ohrms/
dockets/ac/08/slides/2008-4409s1-04-ImClone.pdf. (accessed
March 3, 2010).
192 Amado RG, Wolf M, Peeters M, et al. Wild-type KRAS is required
for panitumumab ecacy in patients with metastatic colorectal
cancer. J Clin Oncol 2008; 26: 162634.
193 Van Cutsem E, Lang I, Folprecht G, et al. Cetuximab plus FOLFIRI
in the treatment of metastatic colorectal cancer (mCRC): the
inuence of KRAS and BRAF biomarkers on outcome: Updated
data from the CRYSTAL trial. Gastrointestinal Symposium 2010; 281
(abstr).
194 Grothey A, Sugrue MM, Purdie DM, et al. Bevacizumab beyond
rst progression is associated with prolonged overall survival in
metastatic colorectal cancer: results from a large observational
cohort study (BRiTE). J Clin Oncol 2008; 26: 532634.
195 Scappaticci FA, Fehrenbacher L, Cartwright T, et al. Surgical wound
healing complications in metastatic colorectal cancer patients
treated with bevacizumab. J Surg Oncol 2005; 91: 17380.
196 Cetuximab for the rst-line treatment of metastatic colorectal
cancer. 2009. http://guidance.nice.org.uk/TA176/Guidance/pdf/
English (accessed Aug 26, 2009).
197 Bardelli A, Siena S. Molecular mechanisms of resistance to
cetuximab and panitumumab in colorectal cancer. J Clin Oncol
2010; 28: 125461.
198 Di NF, Martini M, Molinari F, et al. Wild-type BRAF is required for
response to panitumumab or cetuximab in metastatic colorectal
cancer. J Clin Oncol 2008; 26: 570512.

www.thelancet.com Vol 375 March 20, 2010

199 Kohne C, Stroiakovski D, Chang-chien C, et al. Predictive


biomarkers to improve treatment of metastatic colorectal cancer
(mCRC): Outcomes with cetuximab plus FOLFIRI in the CRYSTAL
trial. Proc ASCO Meeting Abstracts 2009; 27: 4068.
200 Tejpar S, Bosman F, Delorenzi M, et al. Microsatellite instability
(MSI) in stage II and III colon cancer treated with 5FU-LV or 5FULV and irinotecan (PETACC 3-EORTC 40993-SAKK 60/00 trial).
Proc ASCO Meeting Abstracts 2009; 27: 4001.
201 Popat S, Hubner R, Houlston RS. Systematic review of
microsatellite instability and colorectal cancer prognosis.
J Clin Oncol 2005; 23: 60918.
202 Bertagnolli MM, Niedzwiecki D, Compton CC, et al. Microsatellite
instability predicts improved response to adjuvant therapy with
irinotecan, uorouracil, and leucovorin in stage III colon cancer:
Cancer and Leukemia Group B Protocol 89803. J Clin Oncol 2009;
27: 181421.
203 Ribic CM, Sargent DJ, Moore MJ, et al. Tumor microsatelliteinstability status as a predictor of benet from uorouracil-based
adjuvant chemotherapy for colon cancer. N Engl J Med 2003;
349: 24757.
204 Sargent DJ, Marsoni S, Thibodeau SN, et al. Conrmation of decient
mismatch repair (dMMR) as a predictive marker for lack of benet
from 5-FU based chemotherapy in stage II and III colon cancer (CC):
A pooled molecular reanalysis of randomized chemotherapy trials.
Proc ASCO Meeting Abstracts 2008; 26 (15 suppl): 4008.
205 Jen J, Kim H, Piantadosi S, et al. Allelic loss of chromosome 18q
and prognosis in colorectal cancer. N Engl J Med 1994; 331: 21321.
206 Ogunbiyi OA, Goodfellow PJ, Herfarth K, et al. Conrmation that
chromosome 18q allelic loss in colon cancer is a prognostic
indicator. J Clin Oncol 1998; 16: 42733.
207 Popat S, Houlston RS. A systematic review and meta-analysis of the
relationship between chromosome 18q genotype, DCC status and
colorectal cancer prognosis. Eur J Cancer 2005; 41: 206070.
208 Watanabe T, Wu TT, Catalano PJ, et al. Molecular predictors of
survival after adjuvant chemotherapy for colon cancer.
N Engl J Med 2001; 344: 1196206.
209 Halling KC, French AJ, McDonnell SK, et al. Microsatellite
instability and 8p allelic imbalance in stage B2 and C colorectal
cancers. J Natl Cancer Inst 1999; 91: 1295303.
210 Popat S, Zhao D, Chen Z, et al. Relationship between chromosome
18q status and colorectal cancer prognosis: a prospective, blinded
analysis of 280 patients. Anticancer Res 2007; 27: 62733.
211 Ogino S, Nosho K, Irahara N, et al. Prognostic signicance and
molecular associations of 18q loss of heterozygosity: A cohort study
of microsatellite stable colorectal cancers. J Clin Oncol 2009; 20:
459198.
212 Barratt PL, Seymour MT, Stenning SP, et al, the UKCCCR AXIS
trial collaborators. DNA markers predicting benet from adjuvant
uorouracil in patients with colon cancer: a molecular study. Lancet
2002; 360: 138191.
213 Alazzouzi H, Alhopuro P, Salovaara R, et al. SMAD4 as a prognostic
marker in colorectal cancer. Clin Cancer Res 2005; 11: 260611.
214 Alhopuro P, Alazzouzi H, Sammalkorpi H, et al. SMAD4 levels and
response to 5-uorouracil in colorectal cancer. Clin Cancer Res 2005;
11: 631116.
215 Gaasenbeek M, Howarth K, Rowan AJ, et al. Combined arraycomparative genomic hybridization and single-nucleotide
polymorphism-loss of heterozygosity analysis reveals complex
changes and multiple forms of chromosomal instability in
colorectal cancers. Cancer Res 2006; 66: 347179.
216 Trautmann K, Terdiman JP, French AJ, et al. Chromosomal
instability in microsatellite-unstable and stable colon cancer.
Clin Cancer Res 2006; 12: 637985.

1047

You might also like