Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

REVIEWS

Clonal evolution of colorectal cancer


in IBD
Chang‑Ho R. Choi1,2*, Ibrahim Al Bakir1,2*, Ailsa L. Hart2 and Trevor A. Graham1
Abstract | Optimizing the management of colorectal cancer (CRC) risk in IBD requires a
fundamental understanding of the evolutionary process underpinning tumorigenesis. In IBD,
clonal evolution begins long before the development of overt neoplasia, and is probably
accelerated by the repeated cycles of epithelial wounding and repair that are characteristic
of the condition. Here, we review the biological drivers of mutant clone selection in IBD with
particular reference to the unique histological architecture of the intestinal epithelium coupled
with the inflammatory microenvironment in IBD, and the unique mutation patterns seen in
IBD-driven neoplasia when compared with sporadic adenomas and CRC. How these data can
be leveraged as evolutionary-based biomarkers to predict cancer risk is discussed, as well as how
the efficacy of CRC surveillance programmes and the management of dysplasia can be improved.
From a research perspective, the longitudinal surveillance of patients with IBD provides
an under-exploited opportunity to investigate the biology of the human gastrointestinal tract
over space and time.

IBD is a chronic relapsing–remitting disorder of the colon in the presence of Crohn’s disease9 or primary
gastrointestinal tract, consisting of two main subtypes: sclerosing cholangitis10, they are more commonly syn­
Crohn’s disease and ulcerative colitis. The aetiology of chronous (15–20% of CA-CRC11,12 compared with 3–5%
IBD is multifactorial, with the disease arising follow­ of sporadic CRC) and have an increased frequency of
ing incompletely defined environmental triggers mucinous or signet ring cell histology13.
in genet­ically predisposed individuals and resulting in Patients with long-standing extensive colonic IBD
an a­ berrant immune-driven inflammatory response are enrolled into surveillance programmes that aim
towards an altered gut microbiota1. to detect precursor colitis-associated dysplasia, a pre-­
The incidence of IBD is increasing worldwide, malignant equivalent of sporadic adenomas. In practice,
with a prevalence now approaching 0.5% in the West2. detecting dysplasia at endoscopy can be challenging.
For patients with long-standing colonic inflammation, These lesions are often flat with subtle borders that
colorectal cancer (CRC) is a recognized and feared are difficult to appreciate in the presence of concomi­
complication. Studies quantifying the increased risk tant inflammation; they might be readily missed in a
of CRC in patients with IBD have generated vari­able background of extensive mucosal scarring and pseudo­
1
Evolution and Cancer
Laboratory, Barts Cancer results reflecting differences in country of origin, polyposis. Missed dysplasia might be a contributing
Institute, Queen Mary study population and disease duration3. The first large ­factor in the stubbornly high rates of interval CA‑CRCs
University of London, meta-­analysis assessing CRC risk in patients with that develop between scheduled endoscopies14.
Charterhouse Square, IBD showed a risk of 2% at 10 years after ulcerative When dysplasia is detected, patient management
London EC1M 6BQ, UK.
2
Inflammatory Bowel Disease
colitis diagnosis, 8% at 20 years and 18% at 30 years remains fraught with challenges. The diagnosis of IBD-
Unit, Level 4 St Mark’s after colitis onset 4. However, other studies suggest associated dysplasia is confirmed through histo­logical
Hospital, Watford Road, that CRC rates in patients with ulcerative colitis might analysis with defining features including nuclear atypia,
London HA1 3UJ, UK. be ­declining5. mucin depletion and an irregular crypt architecture
pacoblue@gmail.com;
Important clinical differences exist between colitis-­ with loss of basal‑to‑luminal epithelial cell matur­
i.albakir@qmul.ac.uk;
ailsa.hart@nhs.net; associated CRC (CA‑CRC) arising in patients with ation15. Immunohistochemical staining for markers
t.graham@qmul.ac.uk IBD, compared with sporadic CRC seen in the general including p53 and β‑catenin are sometimes used to
*These authors contributed population. CA‑CRCs tend to affect younger patients6 complement histological classification16. In practice,
equally to this work. (average age of 50–60 years in IBD7,8 compared with dysplasia ­grading suffers from substantial inter­observer
doi:10.1038/nrgastro.2017.1 65–75 years for sporadic CRCs in the general popula­ variability and can be challenging to differentiate from
Published online 8 Feb 2017 tion), they are more likely to be found in the proximal a regenerating epithelium17. With improved endoscopic

218 | APRIL 2017 | VOLUME 14 www.nature.com/nrgastro


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points In IBD, the clonal evolution of cancer begins long


before the development of a true malignancy. Numerous
• Colorectal cancer development in IBD begins many years before the development studies, using a variety of different molecular techniques,
of neoplasia because of occult evolution within the inflamed bowel have reported extensive genomic and epi­genomic alter­
• The cycles of wounding and repair characteristic of IBD provide a selective pressure ations in morphologically normal intestinal mucosa
for mutant cells that are able to rapidly heal the mucosa and withstand the (TABLE 1). The surprisingly high mutation burden of the
inflammatory insult ostensibly normal IBD epithelium reveals the ‘occult
• Measuring and modulating the occult evolutionary process offers new avenues for evolution’ that is occurring in the inflamed bowel.
effectively predicting and preventing colorectal cancer in IBD These mutant clones clearly experience a pos­itive selec­
• Repositories of IBD surveillance materials offer a surreptitious opportunity to study tive pressure (mutants cells are fitter than non­mutant
in vivo clonal evolution in time and space in humans cells) as they can expand to fill large areas of the intes­
tinal mucosa24 and, in one extreme case, the entire
colon length25.
imaging, areas of low-grade and indefinite dysplasia Mutant clones frequently bear mutations in key
are now detected in up to 10% of surveillance colono­ tumour-suppressor genes including TP53 (encod­
scopies 18. Some patients with IBD-associated low- ing p53) and CDKN2A (encoding p16), and in
grade dysplasia are offered more intensive surveillance, the proto-­oncogene KRAS 24–27. Remarkably, these
whereas others are offered the option of colectomy, cancer-­associated mutations do not alone cause neo­
with all the associated risks of major surgery and con­ plastic growth as the mutations are detected in non-­
sequences of life with a stoma. Data from the largest neoplastic tissue 24–27. These ‘key’ mutations might
UK surveillance registry data has demonstrated that be necessary but insufficient for tumour growth, or
the 10‑year CRC risk from low-grade dysplasia is only the phenotypic effects of these mutations are crit­
30%11; others have shown that low-grade dysplasia ically modu­lated by epigenetic and/or microenviron­
regresses in at least half of patients19 although the mech­ mental constraints present in non-neoplastic mucosa.
anism is unclear. Together, these findings indicate that Nevertheless, the clonal expansion of these cancer-­
the current ­primary aim of dysplasia identification in associated mutants provides a genetic foundation for the
CRC surveillance is inadequate as a cancer-risk stratifi­ phenomenon of ‘field cancer­ization’ (REF. 28), which is
cation tool. A clear clinical need to better understand the preconditioning of a large area of histologically nor­
the molecular aetiology of CA‑CRC remains, which mal epithelium to the future development of neoplastic
would enable the identifi­cation of more efficacious lesions29 (FIG. 1). Field cancerization probably explains
biomarkers of c­ ancer risk. Recognizing the develop­ the high frequency of synchronous and metachronous
ment of CA‑CRC as an evolution­ary process prov­ neoplastic lesions in patients with IBD11,12.
ides a novel and powerful perspective to understand
this disease, in a manner that can be directly lever­ Inflammation accelerates evolution
aged to improve the clinical care of this ­challenging The mutation burden in the morphologically normal
patient group. non-IBD colon is not well characterised; however, the
This Review will summarize the evidence support­ limited evidence available to date suggests that the nor­
ing the presence of extensive inflammation-driven clonal mal mutation rate is less than in patients with IBD30,31
evolution in IBD, long before the development of any and that widespread clonal expansion of mutant cells
clinically apparent neoplasia, and contrast this under­ is rare32. Consequently, the inflamed bowel appears
standing with our current knowledge of sporadic CRC to be a ‘hot bed’ of somatic clonal evolution, but the
carcinogenesis. Finally, we discuss how knowledge of ­reasons why are currently unknown. Our hypothesis is
this occult evolutionary process can aid prognostication that the evolutionary process in IBD is accelerated by
and ultimately improve the efficacy of IBD-associated long-standing repeated cycles of epithelial wounding
cancer surveillance. and repair that are characteristic of IBD. Mechanistically,
IBD-associated inflammation has the potential to
Evidence of clonal evolution in IBD mediate clonal evolution by any, or all, of the following
Occult evolution in colitis three mechanisms: generating a mutagenic pressure;
Carcinogenesis is a process of clonal evolution20–22. providing a selective advantage to those clones able to
Somatic cells acquire mutations that alter their pheno­ survive a (cytotoxic) inflammatory insult; providing a
type, which might confer them and their progeny a selective advantage to those clones able to more rapidly
growth and/or survival advantage within their cur­ ­repopulate the healing mucosa.
rent microenvironment, enabling the cells to persist DSS (dextran sodium sulfate)-treated mice offer a
and clonally expand. A classic example is mutations unique opportunity to assess flat and polypoid dys­plasia
of APC in the context of colorectal adenoma forma­ in a single animal model; both these lesions develop as
tion that result in persistent β‑catenin signalling and a consequence of the induced chemical colitis. Here, the
increased cellular proliferation23. With the occurrence severity of inflammation correlates with the propen­
of further mutations, additional selection pressures sity to generate IBD-like flat dysplastic lesions; inflam­
and clonal expansion could occur. In some cases, this mation scores were an order of magnitude higher in
process eventually results in the development of a colons containing flat dysplasia compared with those
malignant phenotype. with polypoid dysplasia33. Moreover, administration of

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 14 | APRIL 2017 | 219


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 | Genetic and epigenetic changes detected in noncancerous IBD mucosa


Abnormality found Methods Spatial extent of change Tissue collection time Ref
DNA aneuploidy in progressors only Cytometry Multiple aneuploid clones co-existing At dysplasia or CRC 123
in the same colon; the largest clones
involve at least half the colon length
DNA aneuploidy Cytometry DNA aneuploidy is persistent and Before CRC, longest 110
spreads over time follow-up period was
8 years
TP53 LOH, DNA aneuploidy Cytometry, SSCP In one case almost the entire colonic At dysplasia or CRC 111
mucosa (95 of 100 biopsies) had
aneuploid DNA
DNA aneuploidy in most progressors Cytometry N/A Up to 2.5 years prior 96
prior to developing overt neoplasia to CRC
TP53 mutation, TP53 LOH Cytometry, SSCP TP53 mutations but not LOH found At dysplasia or CRC 64
in nondysplastic mucosa adjacent to
neoplastic areas
KRAS mutation in non-neoplastic tissue IHC, SSCP N/A At CRC 27
Microsatellite instability seen in up to Microsatellite genotyping Randomly selected non-neoplastic At CRC 124
50% of patients with ulcerative colitis mucosa
but not in patients with acute colitis
(ischaemic or infectious)
CNAs detected in all non-neoplastic FISH and CGH Largest clone extended from the At CRC 125
biopsies of progressors transverse colon to the caecum
KRAS and TP53 mutations, Cytometry, SSCP Up to one-third of the colon length At dysplasia or CRC 26
DNA aneuploidy
CNA at target loci on chromosomes 8, FISH Changes seen at both near At high-grade dysplasia 114
11, 17 and 18 (average 10 cm away) and distal or CRC
(average 50 cm away) tissue
CNAs detected much more commonly CGH and microsatellite The largest clone extended at least At CRC 126
than microsatellite instability instability analysis (10 loci) 36 cm from CRC
Increased ESR1, MYOD1, VCAN Methylation Changes seen in non-adjacent tissue At CRC 63
and CDKN2A methylation
Chromosome arm loss in non-neoplastic FISH N/A At CRC 127
tissue distinguishes progressors from
nonprogressors
In progressors, the degree of genomic PCR-based DNA Weak but significant correlation At dysplasia or CRC 128
instability in non-neoplastic tissue is fingerprinting between the degree of genomic
similar to that of neoplastic tissue in instability and distance from the
the same patient, and is greater than in neoplastic lesion
nonprogressors
Polyclonal TP53 mutations are present Genotyping of individual N/A At dysplasia or CRC 129
in regenerative mucosa and low-grade microdissected crypts
dysplasia; become monoclonal in
high-grade dysplasia and cancer
Increased DNA aneuploidy Cytometry Can involve most of the colon and At CRC; some patients 130
rectum had aneuploidy detected
prior to CRC
Increased mutation burden and clonal Hypermutable microsatellite Clonal patches up to 50 cm long At dysplasia or CRC 107
expansion in progressors locus genotyping (28 loci)
Increased RUNX3, APBA1, and COX2 Methylation Changes seen in non-adjacent tissue At CRC 131
methylation
Mutations in APC, TP53, KRAS, Genotyping of individial The largest clone was at least 14 cm, At CRC 24
chromosome 17p LOH microdissected crypts from which 3 spatially distinct
CRCs arose
Increased aneuploidy and increased Cytometry All patients with extensive aneuploidy Mean 9.2‑year patient 132
S‑phase fraction subsequently developed neoplasia follow‑up
The absolute number of regions with CGH, BAC array Clonal field sizes in progressors are At CRC 92
copy gains distinguishes non-neoplastic large, ranging from 49–161 cm
tissue of progessors from nonprogressors
Mutations in CDKN2A, TP53 and KRAS Genotyping of individual Pancolonic (clonal) 4 years prior to CRC 25
microdissected crypts

220 | APRIL 2017 | VOLUME 14 www.nature.com/nrgastro


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 (cont.) | Genetic and epigenetic changes detected in noncancerous IBD mucosa
Abnormality found Methods Spatial extent of change Tissue collection time Ref
Clonal expansions are more frequent Microsatellite genotyping The largest clone involved most of the At CRC 133
in progressors than nonprogressors. In (17 loci) bowel length
nonprogressors, clonal expansions are
associated with older age
Aneuploidy of indefinite dysplastic Cytometry N/A 28‑month mean follow‑up 134
mucosa predictive of progression of indefinite dysplasia
The table shows a summary of genomic and epigenomic alterations found in nondysplastic colonic mucosa. These changes are evidence of clonal evolution in IBD
prior to cancer development. BAC, bacterial artificial chromosome; CGH, comparative genomic hybridisation; CNA, copy number alteration; CRC, colorectal
cancer; FISH, fluorescence in situ hybridisation; IHC, immunohistochemistry; LOH, loss of heterozygosity; SSCP, single-strand conformation polymorphism.

5‑aminosalicylates, the mainstay of treatment for ulcer­ ­ arrants further investigation. For example, CA‑CRCs
w
ative colits, had a varying effect on the incidence and and dysplastic lesions show a greater infiltration of CD3+
progression of dysplastic lesions on the basis of mor­ and CD8+ lymphocytes, but substantially less granzyme
phology, with fewer flat lesions that were unchanged in B expression than ­sporadic CRCs, suggesting impaired
size, but the same number of polypoid lesions that were cytotoxic function47.
reduced in size34. Emerging studies in the field of microbiome analy­sis
Patient-based studies confirm that inflammation has are revealing the role of the gut microbiota and intestinal
a substantial cumulative role in increasing CRC risk in barrier function in tumorigenesis, and animal studies
IBD, with observational studies consistently demonstrat­ are beginning to shed some light onto the complex and
ing that CA‑CRC risk development is closely linked to dynamic interplay between the altered immune system,
the extent35, duration4 and severity of inflammation7. the aberrant gut microbiome and cancer development
Histological studies suggest that neoplastic lesions arise in IBD37. In practice, it remains difficult to determine
from a field of marked chronic inflammation, telomere whether intestinal dysbiosis and mucosal barrier changes
shortening, DNA damage and senescence, with ‘escape drive mucosal pathology, or represent a secondary con­
from senescence’ a key step in the transition from low- sequence of disease. However, these options are not
grade to high-grade dysplasia36. Thus, a comprehensive mutually exclusive. A prominent example comes from
understanding of carcinogenesis in IBD should not be Arthur et al.48, who used Il10‑knockout mice to demon­
limited to the study of mutation generation and spread strate how chronic inflammation prevents the homeo­
via the epithelial crypt stem cell niche, but should also static elimination of cancer-­associated Escherichia coli,
include a concomitant analysis of the inflammatory stro­ inducing the upregulation of the polyketide synthase
mal microenvironment. Therapeutic interventions that gene responsible for the genotoxic compound coli­
modulate cancer risk in IBD must target these ­aberrant bactin. In turn, E. coli with genotoxic capabilities have
microenvironmental changes. been shown to drive tumorigenesis in this mouse model,
In addition to native cells of the epithelial layer, in a manner independent of inflammation severity49.
immune cells and bacteria migrate up to the crypt as part This effect is probably dependent on a breakdown
of IBD pathogenesis, inducing a myriad of inflammatory in the mucosal barrier50, enabling the direct exposure of
cytokines and intracellular signalling pathways1 that are the epithelium to colibactin. Subsequent human stud­
beyond the scope of this Review37. These pathways act ies confirm that colibactin-equipped E. coli are mark­
directly on non-neoplastic and dysplastic epithelial cells, edly more prevalent in colonic mucosa from patients
which can potentially modulate cancer risk. Animal with sporadic CRC and patients with ulcerative colitis
model studies demonstrate how transcription signal­ compared with controls (patients with sporadic polyps
ling by NF‑κB — a master regulator of inflammation38 or IBS)49,51. Similarly, Fusobacterium have been found
that has a key role in IBD patho­genesis39 — ­promotes to promote tumorigenesis in animal models52 and are
the survival of pre-malignant epithelial clones40. IBD- over-­represented in both sporadic adenomas and CRC53,
mediated inflammation also promotes β‑catenin as well as in non-neoplastic IBD mucosa54. Finally, simi­
stabil­ity through aberrant PI3K–AKT41,42 and NF-κB43 lar changes in colonic mucus affecting IBD, colon polyps
signalling to further enhance canonical Wnt activity. and colon cancer have been described55. A prominent
Epithelial STAT3 signalling is also upregulated in active example is the re‑­expression of the mucin-associated
IBD44. Mouse models show that, although STAT3 path­ oncofoetal carbohydrate, sialyl-Tn‑antigen, at higher
way activation helps amelior­ate the effects of chemical rates in patients with ulcerative colitis who subsequently
colitis by reducing epithelial damage and inflam­mation, progress to dysplasia or cancer than those that remain
it also promotes the survival and progression of pre-­ cancer-free56. This increased expression seems to be
malignant epithelial clones45. Conceivably, non-cell-­ independent of inflammation severity.
autonomous effects similar to those identified in other
systems (for example, in a breast cancer model46) might Somatic mutations in CA‑CRC
also be involved. In this same context, the effect of an The different evolutionary pressures offer an explan­
altered immune system in IBD on immunosurveil­ ation for the altered pattern of somatic mutations
lance necessary to limit CRC initi­ation and progression seen in CA‑CRC compared with sporadic cancers57,58.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 14 | APRIL 2017 | 221


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Available data show that CA‑CRCs have increased muta­ mutation. Furthermore, wounding of the intestinal epi­
tion frequencies of various intracellular and intercellu­ thelium induces a host of canonical and noncanonical
lar signalling molecules57,58. The most n­ otable mutations Wnt signalling pathways by mesenchymal cells70.
include IL16, a gene encoding a chemo­attractant Aside from these pertinent genomic differences, the
cytokine that is overexpressed in IBD in an inflamma­ genomes of CA‑CRCs and sporadic CRCs in humans
tion-dependent manner59. IL‑16 has a potential role in interestingly show broad similarities. Both bear a high
directly mediating inflammation60 and so we specu­ frequency of TP53 alteration and mutations in the onco­
late that a gene mutation encoding this protein could genes KRAS, BRAF, PIK3CA, CTNNB1 and the tumour
provide a survival advantage in the inflamed bowel. suppressors SMAD4 and FBXW7 are at relatively high,
Another noteworthy mutation is in RADIL, a gene albeit slightly different, frequencies57,58,71. Microsatellite
encoding a modulator of Rho GTPase signalling in cell instability is also common to a subset of both cancer
migration, which might speculatively provide a selective types72,73. Together, these data imply some convergence
advantage in mucosal healing57. in the outcome of evolutionary processes of sporadic
Mutation signature analysis61 of CA-CRCs57 demon­ CRC and CA‑CRC development. Dysregulation of key
strate novel trinucleotide context changes not seen in pathways such as Wnt signalling and p53 function seem
sporadic CRCs (an over-representation of A>C trans­ to be necessary for tumour formation in the colon,
versions at AA dinucleotides). The predominance of irrespec­tive of the presence or absence of a (prior)
C>T transitions at CG dinucleotides in CA‑CRCs is in inflammatory stimulus. Studies of clonal evolution at
keeping with ‘accelerated ageing’ through rapid cell turn­ the level of the crypt are discussed in the next section,
over62 rather than the accumulation of direct genotoxic advancing our understanding of the survival advan­
hits caused by inflammation-associated carcinogens. tage these genomic changes provide, specifically in the
Indeed, colitis that progresses to high-grade dysplasia ­context of IBD-mediated inflammation.
or CA‑CRC demonstrates field changes in ostensibly
normal epithelium in keeping with accelerated age­ Mechanisms of clonal expansion
ing, including telomere shortening36 and age-related Colonic crypt architecture
CpG methylation63. The basic functional unit of the colon is the crypt,
Although TP53 mutations are found at similar fre­ a finger-­like invagination into the underlying lamina
quencies in both sporadic CRC and CA‑CRC, TP53 propria lined by a single layer of columnar epithelial
alteration is an early event in CA‑CRC that is found cells. Under homeostatic conditions, the majority of
in non-tumour, and even nondysplastic, mucosa64. crypt cells migrate towards the top of the luminal sur­
Indeed, accumulating evidence suggests a role for TP53 face over the course of approximately a week, after which
mutations in promoting cell survival and growth65. they are shed in the lumen74. Migration out of the crypt
Interestingly, the increased frequency and early onset base is associated with differentiation into specialized
of TP53 mutations might provide an additional explan­ cell types, including absorptive enterocytes and sup­
ation for the fairly high proportion of flat dysplastic and porting secretory cell lineages75. These differentiated
cancer­ous lesions seen in IBD. One study66 demonstrated cells are continuously replenished by a small number of
a significant variability in neoplasm morphology that is multi­potent stem cells located in the base of each crypt,
dependent on Tp53 mutation status, with flat lesions known as the stem cell niche76. In the presence of injury
associated with Tp53−/− mice treated with DSS (~85% to this actively cycling stem cell pool, cells from further
flat lesions), and polypoid lesions significantly associated up the crypt axis fall into the crypt base and regain active
with Tp53+/− and Tp53+/+ DSS-treated mice (~17% flat stem cell properties77. Carcinogenic mutations probably
lesions; P <0.0001). first start accumulating in the long-lived stem cell line­
Perhaps most notably, CA‑CRCs in humans show a ages of the crypt, as the lifespan of non-stem cells is
paucity of APC mutations57,58 compared with sporadic too short for them to acquire the necessary mutations
adenomas and CRC, in which APC mutations are a before being shed. Although LGR5+ crypt base stem
common, important and early ‘gatekeeper’ event67. APC cells are clearly implicated in mutant-APC-driven spor­
encodes a key regulator of the canonical Wnt pathway adic CRC and familial adenomatous polyposis78, the
driving β‑catenin signalling. Disruption of normal APC precise stem cell(s) of origin in IBD-driven neoplasia
function is sufficient to constitutively activate this path­ remains undefined.
way and initiate sporadic adenomas in the noninflamed
bowel67,68. The lack of APC mutations in colitis raises the Stem-cell-level evolution
possibility of IBD-generated inflammation-mediated Clonal expansion in the colon begins with the progeny
alterations of underlying Wnt signalling cascades that of a mutant cell repopulating an entire crypt in a process
render APC mutations dispensable for tumorigenesis. referred to as monoclonal conversion of the crypt79. Even
Indeed, immunostaining studies of colonic epithelium in the absence of any mutant cells, the stem cells in a
confirm high levels of Wnt signalling, as demonstrated crypt are randomly lost and replaced by a neighbour­ing
by increased nuclear to cytoplasmic β‑catenin levels in stem cell lineage in a process called drift. This process
human ulcerative colitis tissue69. The β‑catenin levels are eventually results in extinction of all stem cell popula­
midway between normal noninflamed epithelium and tions except for those originating from the one ‘lucky’
established CRC, thereby partially replicating the con­ stem cell clone that repopulates the whole crypt, in a
stitutive canonical Wnt pathway activation of an APC process referred to as niche succession79.

222 | APRIL 2017 | VOLUME 14 www.nature.com/nrgastro


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Some mutations generated in IBD will bias the com­ of Tp53 mutations in an inflamed crypt compared with a
petition between stem cells to make it more likely that a noninflamed crypt reflects the unique selective pressures
particular mutant stem cell will go on to repopulate the that exist in the inflamed bowel.
entire crypt. In vivo lineage tracing studies of intestinal In the healthy bowel, tight morphogen gradients gen­
crypt cells in recombinant mouse models have been used erated by cells in and adjacent to the crypt82 restrict the
to measure the advantage of tumorigenic mutations. size of the stem cell niche, keeping stem cell numbers
Although a wild-type stem cell will replace another wild- constant80. The epithelial wounding that is a hallmark
type stem cell 50% of the time, in keeping with neutral of colitis clearly disrupts these signalling gradients70,82.
competition80,81, a stem cell with a Kras mutation will Additionally, a study in transgenic mice shows how the
replace a wild-type neighbour stem cell in ~75% of com­ inflammation-mediated NF‑κB pathway can induce
petitive divisions81. Interestingly, Tp53 mutations confer dedifferentiation of non-stem cells, thereby increasing
no substantial competitive advantage in a healthy crypt: the size of the pool of long-lived cell lineages that can
these mutant cells will replace wild-type cells only 50% of contribute to carcinogenesis43. Although the precise
the time81. However, in the context of chemically induced mechanisms of morphogen gradient disruption are yet
colitis (via DSS treatment), Tp53 mutation increases the to be characterized in human tissue, the altered cellular
odds of lineage replacement to 58%81. The differing fate composition of the supportive pericryptal fibroblasts in

a Normal colonic mucosa

b Inflamed colonic mucosa

Epithelial cell Stem cell Paneth cell Macrophage Fibroblast Altered microbiota
Mutant epithelial cell Mutant stem cell Mutant Paneth cell Neutrophil Mononuclear cell Ulceration

Figure 1 | Differences in clonal expansion between normal and inflamed Nature


fission (third image), which Reviews
is the | Gastroenterology
main mechanism for clonal&expansion
Hepatology in
mucosa. The accelerated rate of clonal evolution seen in IBD might be the intestine (fourth image). b | In IBD, the altered microenvironment
explained by differences between normal and inflamed mucosa. a | In the generates epithelial cells with different mutant signatures (red, first image)
normal colon, a random mutation generated within a crypt stem cell compared with the normal colon. Active disease provides a selective
(first image, in red) results in an advantageous phenotype that eventually advantage for those mutant cells that can survive an inflammatory insult
results in the extinction of wild-type stem cells (second image). Eventually, (second image). The subsequent healing process selects for those clones
all cells of that crypt are formed by progeny of this mutant cell (monoclonal that can more rapidly repopulate the mucosa (third image). In this manner,
conversion of a crypt). These same mutations often accelerate crypt IBD accelerates clonal evolution and expansion (fourth image).

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 14 | APRIL 2017 | 223


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

colitis is a notable example of a niche structure alter­ highly unlikely and so the very large mutant populations
ation83 that has been associated with neoplastic progres­ observed are likely to be clonal in origin. Additionally,
sion in IBD84. Understanding the altered morphogen how mucosal repopulation by fission of adjacent crypts
gradients in IBD is key to understanding how and why alone could explain the same mutation spread across
particular mutant cells have an evolutionary advantage the entire bowel in a matter of a few years is difficult
in IBD. to comprehend. These findings raise the possibility
of unidenti­fied healing mechanisms in colitis. Mouse
Crypt-level evolution model studies show that free-floating organoids intro­
Once a mutant clone has repopulated an entire crypt, duced intra­luminally can successfully engraft on to an
further clone expansion is possible by crypt fission, the inflamed bowel to generate long-lasting functional crypt
mechanism by which human colonic crypts divide79. populations, but not in the context of a non­inflamed
Thus, the initial cell-level evolution within the crypt colon93. In light of the phenotypic elasticity of multiple
switches to crypt-level evolution and, therefore, natural crypt cell populations and the rapid rate of epithelial
selection of the crypt population drives field canceriza­ shedding in active IBD, this finding raises the intriguing
tion. Crypt fission is the main driver of clonal expan­ possibility of long-range stem cell migration following
sion in the human colon85; indeed, the fission of crypts shedding and distant stem cell re‑engraftment. However,
surrounding a wound is the primary mechanism of epi­ to date, no evidence exists for this speculative healing
thelial restitution in the intestine70. Our understanding mechanism in human studies or in IBD a­ nimal models.
of the mechanistic drivers of fission is surprisingly lack­ Conversely, precedence for this process in other model
ing74. 3D imaging studies suggest a critical role for the organisms does exist; long-distance stem cell migra­
relative arrangement of stem cells and their supportive tion has been noted to occur between widely separated
niche-providing Paneth cells86, and mathematical model­ ­ovarian ­follicles in Drosophila94.
ling indicates that a plausible mechanism for triggering
fission is exceeding a threshold number of stem cells87. Evolutionary management of cancer risk
Crypt fission is a mechanism for mucosal healing70, Predicting cancer risk in IBD
and the increase in frequency of crypt-fission events in Great interest exists for the development of cancer
the IBD bowel is a reflection of the wounding or repair biomarkers in IBD95, with DNA aneuploidy96 and/or
that is characteristic of the disease. Cheng and col­ TP53 mutations64,97,98 as the most prominent examples
leagues88 found that, although <1 in 200 crypts in nor­ of  genetic biomarkers. However, extensive within-­
mal human colon are undergoing fission, this rate was lesion genetic and phenotypic heterogeneity are now
increased at least 60‑fold in IBD. Mutations that increase considered hallmarks of both carcinogenesis99 and pre-­
the rate of crypt fission are probably positively selected malignant pathology22. Indeed, in the pre-­malignant
for in the IBD bowel, as these mutants are better able to disease, Barrett oesophagus, which confers a ~30‑fold
heal the damaged mucosa. In line with this hypothesis, increased risk of developing oesophageal adeno­
mutant Kras crypts divide 30‑fold faster than their wild- carcinoma100, sequencing studies of biopsies from non-­
type counterparts in the mouse intestine89, and Tp53 neoplastic Barrett segments demonstrate cancer-related
mutation is also reported to increase the crypt ­fission mutations in a plethora of different genes. With the
rate in mice90. Thus, it is reasonable to expect that some exception of TP53, these mutations occur at similar
mutants which undergo substantial clonal expansion and frequencies across nondysplastic through to high-grade
populate multiple crypts in the non-neoplastic epithe­ dysplasia and even adeno­carcinoma tissues101. In spor­
lium achieve this expansion by the acquisition of muta­ adic CRC, large-scale sequencing projects reveal only
tion(s) that positively regulate crypt fission (FIG. 1). This seven genes that are recurrently mutated in >10%
selection for mucosal healing rather than neoplasia per se of patients, meaning that the overwhelming majority of
might be the underlying reason for the common occur­ mutations were unique to small subsets of patients71.
rence of flat dysplastic lesions in IBD. Consequently, in Collectively, these data indicate the improbability that a
our opinion, we believe that assessing the inflamed epi­ single gene or small set of genetic changes can effectively
thelium for clonally expanded mutations offers a ‘natural assay the vast array of different molecular pathways
experiment’ that can be exploited to identify new genes that lead to cancer102, so it is unlikely that traditional
that modulate crypt fission. This novel approach recog­ candidate-­gene biomarker approaches will yield clin­
nizes one of the old adages of cancer biology: the overlap ically applicable prognostic biomarkers. Nevertheless, as
between wound healing and carcinogenesis91. the cost of sequencing continues to decrease, the option
to cost-effectively sequence a relatively large panel of
Other potential mechanisms genes that encompass all the key pathways involved in
One intriguing finding from IBD mapping studies is CA‑CRC development (once these have all been identi­
the detection of mutant populations containing the fied) could well be a p ­ ossibility — such an approach
exact same point mutation (single nucleotide variant) or already shows promise in acute myeloid leukaemia103.
copy number alteration (CNA), extending across most A different approach involves looking for robust prog­
of the colon length25,92. Although pancolonic selection nostic biomarkers from measurements of the under­lying
pressures could plausibly select for the same altered cel­ evolutionary process itself. The rationale here is that,
lular or crypt functions repeatedly through convergent although the evolutionary paths might differ between
evolution, inducing the exact same point mutation is patients, limiting the efficacy of candidate-gene-based

224 | APRIL 2017 | VOLUME 14 www.nature.com/nrgastro


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

studies, somatic evolution is always subject to the same and how it is subverted during cancer development.
rules and the selective pressures of an inflamed bowel The spatial distribution of genetic alterations across
are similar across patients. Thus, biomarkers based upon surveillance biopsies indicates the order of mutations
measurements of these ­evolutionary rules could have that led to the current clone composition: a ‘trunk’ or
universal applicability. ‘founder’ mutation present in all biopsies arose before a
One example of a candidate ‘evolutionary biomarker’ ‘branch’ mutation that is limited to a subset of biopsies.
is clonal diversity. In ecological studies, the level of A phylogenetic analysis of multiple biopsies at a single
diversity in a population is a major determinant of its time-point (from an individual endoscopy) provides a
evolvability104. If there is no diversity, natural selection mechanism for inferring the p ­ attern of genetic changes
cannot operate, whereas diverse populations are likely to over space and time.
adapt and evolve quickly. Translated to IBD, the clonal Several studies have used mapping biopsies to
diversity within the bowel is a potential evolutionary ­analyse the spatiotemporal changes in clonal popula­
biomarker of cancer risk and, notably, measuring clonal tions, including the relationship between CNAs and key
diversity requires no substantial prior knowledge of the gene mutations. In early studies, analysis of ulcerative
precise genetic and epigenetic pathways driving tumori­ colitis colonoscopic biopsy specimens taken over an
genesis. To date, no studies have used genetic diversity 8‑year period using flow cytometry showed that aneu­
measures for CA‑CRC risk, but the principle has pre­ ploidy can be detected before the onset of dysplasia in
viously been applied to Barrett oesophagus. Genetic at least 10% of patients, with CNAs becoming more
diversity measures were predictors of cancer risk even widespread over time in a subset of patients110. TP53
after controlling for age, Barrett segment length, TP53 gene mutations can also be found in morphologically
mutation and aneuploidy status105, and were robust to normal mucosa and correlate strongly with the presence
the molecular assay used to detect diversity106. A second of aneuploidy64. TP53 loss of heterozygosity was found
approach that also reflects evolvability would be detec­ more commonly in dysplasia, suggesting that it might
tion of large clonal expansions as a measure of ongoing be a later event111. Longitudinal analysis of three patients
evolution. Preliminary studies in patients with ulcer­ with Crohn’s disease using targeted sequencing of TP53,
ative colitis found progressors (high-grade dysplasia or KRAS and CDKN2A revealed that some cancer cases
cancer) have substantially larger clonal expansions than had arisen from a field of mutant clones that expanded
nonprogressors, by using 4–5 biopsies (spaced ~20 cm well beyond the cancer resection margin and formed
apart) for the detection of mutant clones with shared many years before cancer growth25. Similarly, large-scale
point mutation(s)107. This work highlights a promising regional clonal expansions were associated with cancer
avenue to explore. Additionally, changes over time in occurrence in a study that used hypermutable non­
the clonal composition of the epithelium might also be coding polyguanine tracts as neutral lineage markers to
prognostic, irrespective of what those changes might trace clonal expansions across colectomy specimens107.
be, because they are also an indicator of ongoing evolu­ A genome-wide comparative genomic hybridisation
tion. In Barrett oesophagus, neoplastic initiation seems array study found that CNAs of chromosomal segments
to be abrupt, but single nucleotide polymorphism array are present in most colons with pancolitis, and are at
studies suggest that considerable discriminating changes increased frequency in those with neoplasia, without left
in clonal composition occur 24 months before oesoph­ or right colon bias92. Interestingly, this study reported that
ageal adenocarcinoma detection108. Finally, analys­ing most CNAs (~85%) were unique to a single biopsy (with
changes in immune, mesenchyme and/or microbiota substantial differences detected in CNA profiles from
microenvironmental composition (which form the biopsies as little as 2 cm apart) with no shared CNAs
basis for future evolution), and/or their modu­lation between high-grade dysplasia and cancer, and the non­
by the epithelial cells themselves109, offers yet another dysplastic surrounding mucosa. This finding suggests
approach. Concomitant genomic–­microenvironmental that genetic field cancerization does not universally
evolutionary studies could also help answer a ‘chicken- precede cancer development, and instead some patients
and-egg’ question in CA‑CRC: which came first, the show focal genetic instability. Consequently, the detection
altered microenvironment that predisposes to cancer of these different occult evolutionary processes, includ­
risk, or the predisposed clones themselves? ing selective mutant sweeps (in which a clone grows to
cover very large segments of the colon) and pancolonic
Use of surveillance biopsies genomic instability (in which multiple different mutant
In our opinion, IBD-associated cancer surveillance clones arise independently at different location), might
is an ideal ‘model system’ in which to study in vivo provide an indicator for CA‑CRC development risk.
inflammation-associated carcinogenesis in humans.
Multiple biopsies are routinely taken at each surveil­ Optimizing screening intervals
lance colonoscopy and subjected to histological assess­ Endoscopic screening intervals (the time between suc­
ment for inflammation severity and endo­s copically cessive endoscopies) could potentially be optimized by
invisible dysplasia. This process creates an extensive an underlying knowledge of the spatiotemporal dynam­
tissue library of archival specimens reflecting the ics of clonal evolution occurring in IBD. The reason is
spatio­temporal clonal evolutionary changes that occur straightforward: if the minimum time it will take for a
during carcinogenesis. Thus, this rich resource can be cancer to evolve from the current composition of the
utilized to better understand the biology of the intestine bowel is known, surveillance can be planned accordingly.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 14 | APRIL 2017 | 225


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Our current knowledge of the rates of clone growth Routine colonoscopic biopsies contain ~200 crypts112.
in the inflamed bowel is minimal. Basic questions, such Owing to the sheer length of the whole colon, even the
as the duration of time required for a clone to undergo most intensive IBD mapping biopsy protocols sample
expansion substantial enough to be detectable with <1% of the colonic mucosal surface113. Thus, endoscopy-­
biopsy, and the time required to establish a mutant based studies might result in considerable clonal popu­
cancerization field, remain unanswered. Moreover, lations being missed, unless clonal patch sizes truly
the rate and consistency of mutation accumulation are often as large as those detected in some patients
is unknown. In our opinion, measuring these funda­ with IBD25,92. Understanding the clonal mosaicism of
mental ­e volutionary parameters could have direct the bowel could help guide optimal biopsy protocols.
clinical relevance. Moreover, as ulcerative colitis by definition starts in
A practical factor to consider when using IBD sur­ the rectum and extends proximally, we speculate that the
veillance is the fidelity of standard mapping biopsies at rectum is the site where the most evolution has occurred,
reflecting true clonal composition of the entire colon. suggesting that the evolutionary dynamics in the distal

a IBD nonprogression

* * * * * *
* No active
* *
inflammation
* * *
* * *
* * *
* * This single mutant clonal
*
population has not changed
* * significantly in size with time *
b IBD progression through clonal sweeps

* * * * * *
* Substantial * *
inflammation
* * *
* * *
* * *
* Dysplastic lesion is resected
* Dysplasia recurs, it shares
* Cancer arises
in a field of
from a field of successive rapidly the same truncal mutations rapid clonal
* expanding mutant clones * as the newly formed cancer * expansions

c IBD progression through clonal mosaicism

* * * * * *
* Substantial * *
inflammation
* * * Highest
Mutation burden
* * *
* * *
* * Two cancers and a dysplastic *
Multiple unrelated clonal lesion arise simultaneously through Lowest
* populations arise and
evolve independently * clonal mosaicism; they do not share
the same truncal mutations * Wild-type

Figure 2 | Different potential manifestations of mutant clonal evolution Nature


cancerization). With ongoing Reviews | Gastroenterology
inflammation, & Hepatology
a cancer forms more proximally
in IBD. Colours represent distinct clones and asterisks indicate biopsy sites. (right panel) and a proctocolectomy might be considered. Here, large-scale
a | In IBD nonprogression, although an occasional localized mutant clone and/or rapid clonal expansion of cancer-associated mutations could be a
population might be detected, the overall clonal composition does not biomarker of high cancer risk. c | In IBD progression through clonal
change with time. b | In IBD progression with clonal expansions, a large mosaicism, multiple distinct clonal populations develop and progress
clonal population is already present, with a newly formed clone arising from throughout the colon. The end result is two cancers and a dysplastic region,
this field (left panel). Ongoing inflammation generates further successive all arising in the left colon, without sharing the same ‘truncal’ genomic
clonal expansions (centre panel). A dysplastic lesion in the rectum (white changes. In the absence of large clonal fields, a diagnosis of a ‘dangerous’
star) is resected endoscopically. Biopsies of the surrounding mucosa and the clone is challenging. However, high levels of clonal mosaicism and
left colon confirm that the mutant clone is extensive. This finding suggests substantial changes over time indicate ‘active evolution’ that could be
a substantial risk of developing recurrent neoplasia (a consequence of field utilized as a biomarker of cancer risk.

226 | APRIL 2017 | VOLUME 14 www.nature.com/nrgastro


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

colon might have the most relevance for disease aetio­ determine the spatial extent of these high-risk changes
logy. Indeed, preliminary evidence raises the possibility might enable identification of a subset of patients with
that chromosomal instability of the rectum is a proxy limited cancerization field changes, who could be offered
marker of CRC risk for the whole large intestine114. a segmental colectomy that preserves anorectal function.
Nevertheless, this approach might not apply for patients
with Crohn’s disease and so‑called skip-­lesions, or for Optimizing treatment to prevent cancer
patients with primary sclerosing cholan­gitis and with a With inflammation a key driver of CA‑CRC risk in clin­
propensity to develop right-sided colon cancer owing ical and laboratory studies, assessing how therapy modu­
to possible aberrant enterohepatic biliary circulation115. lates occult evolution provides a means to understanding
Assessing the evolutionary dynamics of the colon how and why drug treatments affect cancer develop­
would enable us to define the window of opportunity ment. Aspirin reduces both sporadic CRC incidence117
for cancer risk prognostication, rationalize colonoscopic and precursor adenoma recurrence118, and evidence from
biopsy protocols (random versus segmental versus endo­ Barrett oesophagus shows that NSAID use slows the
scopic areas of concern) and determine the appropriate rate of somatic mutation accumulation119, suggesting an
time intervals between endoscopies (FIG. 2). To ensure evolutionary mechanism of cancer prevention. Together,
clinical utility, such a biomarker should aim for maximal these data imply that attenuation of inflammation can
sensitivity to avoid missing cancer cases. have profound influence on the carcinogenic process.
Nevertheless, although drug treatment in patients with
Managing neoplastic lesions IBD might modulate clonal evolution of the inflamed
Managing dysplasia remains the most challenging clin­ bowel120, similar studies assessing evolution patterns
ical dilemma in IBD surveillance. Differentiating those before and after therapy are needed to define and
patients requiring immediate colectomy from those who rational­ize the most optimal medical intervention strat­
can be safely managed conservatively remains difficult. egy. As a clear majority of patients undergoing c­ ancer
Consensus guidelines based on dysplasia morphology surveillance show some evidence of active inflammation
and endoscopic resectability aim to standardize clinical on biopsy121, these studies might generate greater clinical
approaches116. Although the majority of patients with impetus towards achieving deep remission122 in high-risk
low-grade dysplasia will never develop cancer, 25% of patients, rather than symptomatic relief alone.
patients with low-grade dysplasia who are advised to
undergo colectomy will already have an established Conclusion
­cancer in their surgical specimen11. Even when a ­cancer Occult evolution is a feature of IBD-associated carcino­
is detected at colonoscopy, it is not uncommon for genesis and begins long before the development
patients to request a segmental resection rather than of clinically detectable neoplasia. Repeated episodes of
proctocolectomy, to avoid living with a stoma and/or inflammation generate mutant clones and select for
ileo­anal pouch. Such requests go against standard clin­ those cells best adapted to this microenvironment, such
ical advice; at least one-third of patients undergoing as clones resistant to apoptosis and those with acceler­ated
colectomy for CA‑CRC detected at surveillance will have growth. These selective pressures, unique to IBD, might
a second s­ ynchronous neoplastic region11. explain the unique characteristics of IBD-associated
Dysplastic lesions offer an ideal opportunity to assess neoplasia in comparison to its sporadic counter­part.
the utility of evolutionary biomarkers by localizing the Assaying this evolutionary process is an exciting new
site of a mutant cancerization field (FIG. 2). These lesions opportunity for developing a new class of biomarkers
and the surrounding mucosa can then be assessed for that are robust to the stochastic nature of tumorigenic
markers of somatic evolution, such as the local burden of mutations, which has hampered the effective­ness of
genomic alteration (or mutational burden). The sizes traditional candidate-gene biomarkers. For clinicians,
of clonally expanded patches and the degree of clonal such a marker would not only aid in cancer risk stratifi­
diversity might enable identification of high-risk lesions cation, but might even identify those patients requir­
that are likely to progress to CRC. This application is ing surgery for dysplasia and/or cancer who would be
particularly useful for patients with recurrent, invisible candidates for segmental resection rather than procto­
or nonresectable low-grade dysplasia, whereas evolu­ colectomy, thereby preserving some colo­rectal function.
tionary markers associated with a high risk of cancer For cancer biologists, the IBD surveillance protocol is
development could be used as an indication for colec­ an ­u nderutilised model for ­u nderstanding human
tomy. Furthermore, analysing the remaining mucosa to intestinal biology.

1. Kaser, A., Zeissig, S. & Blumberg, R. S. Inflammatory 5. Castaño-Milla, C., Chaparro, M. & Gisbert, J. P. 8. Baars, J. E. et al. Age at diagnosis of inflammatory
bowel disease. Annu. Rev. Immunol. 28, 573–621 Systematic review with meta-analysis: the declining bowel disease influences early development of
(2010). risk of colorectal cancer in ulcerative colitis. colorectal cancer in inflammatory bowel disease
2. Kaplan, G. G. The global burden of IBD: from 2015 Aliment. Pharmacol. Ther. 39, 645–659 (2014). patients: a nationwide, long-term survey.
to 2025. Nat. Rev. Gastroenterol. Hepatol. 12, 6. Jess, T., Rungoe, C. & Peyrin-Biroulet, L. Risk of J. Gastroenterol. 47, 1308–1322 (2012).
720–727 (2015). colorectal cancer in patients with ulcerative colitis: 9. Bansal, P. & Sonnenberg, A. Risk factors of
3. Adami, H. et al. The continuing uncertainty about a meta-analysis of population-based cohort studies. colorectal cancer in inflammatory bowel disease.
cancer risk in inflammatory bowel disease. Gut 65, Clin. Gastroenterol. Hepatol. 10, 639–645 (2012). Am. J. Gastroenterol. 91, 44–48 (1996).
889–893 (2016). 7. Rutter, M. D. et al. Thirty-year analysis of a 10. Mattar, M. C., Lough, D., Pishvaian, M. J.
4. Eaden, J. A., Abrams, K. R. & Mayberry, J. F. colonoscopic surveillance program for neoplasia in & Charabaty, A. Current management of
The risk of colorectal cancer in ulcerative colitis: ulcerative colitis. Gastroenterology 130, 1030–1038 inflammatory bowel disease and colorectal cancer.
a meta-analysis. Gut 48, 526–535 (2001). (2006). Gastrointest. Cancer Res. 4, 53–61 (2011).

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 14 | APRIL 2017 | 227


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

11. Choi, C.‑H. R. et al. Forty-year analysis of colonoscopic 34. Clapper, M. L. et al. 5‑aminosalicylic acid inhibits 59. Seegert, D. et al. Increased expression of IL‑16
surveillance program for neoplasia in ulcerative colitis: colitis-associated colorectal dysplasias in the mouse in inflammatory bowel disease. Gut 48, 326–332
an updated overview. Am. J. Gastroenterol. 110, model of azoxymethane/dextran sulfate sodium- (2001).
1022–1034 (2015). induced colitis. Inflamm. Bowel Dis. 14, 1341–1347 60. Wang, P. et al. IL‑16 induces intestinal inflammation
12. Lam, A. K.‑Y., Chan, S. S.‑Y. & Leung, M. Synchronous (2008). via PepT1 upregulation in a pufferfish model:
colorectal cancer: clinical, pathological and molecular 35. Ekbom, A., Helmick, C., Zack, M. & Adami, H. O. new insights into the molecular mechanism of
implications. World J. Gastroenterol. 20, 6815–6820 Ulcerative colitis and colorectal cancer. A population- inflammatory bowel disease. J. Immunol. 191,
(2014). based study. N. Engl. J. Med. 323, 1228–1233 1413–1427 (2013).
13. Itzkowitz, S. H. & Yio, X. Inflammation and cancer IV. (1990). 61. Alexandrov, L. B. et al. Signatures of mutational
Colorectal cancer in inflammatory bowel disease: 36. Risques, R. A. et al. Ulcerative colitis-associated processes in human cancer. Nature 500, 415–421
the role of inflammation. Am. J. Physiol. Gastrointest. colorectal cancer arises in a field of short telomeres, (2013).
Liver Physiol. 287, G7–G17 (2004). senescence, and inflammation. Cancer Res. 71, 62. Alexandrov, L. B. et al. Clock-like mutational processes
14. Mooiweer, E. et al. Incidence of interval colorectal 1669–1679 (2011). in human somatic cells. Nat. Genet. 47, 1402–1407
cancer among inflammatory bowel disease patients 37. Grivennikov, S. I. Inflammation and colorectal cancer: (2015).
undergoing regular colonoscopic surveillance. colitis-associated neoplasia. Semin. Immunopathol. 63. Issa, J. P., Ahuja, N., Toyota, M., Bronner, M. P.
Clin. Gastroenterol. Hepatol. 13, 1656–1661 (2015). 35, 229–244 (2013). & Brentnall, T. A. Accelerated age-related CpG
15. Harpaz, N. & Polydorides, A. Colorectal dysplasia 38. Ghosh, S. & Hayden, M. S. New regulators of island methylation in ulcerative colitis. Cancer Res. 61,
in chronic inflammatory bowel disease: pathology, NF‑kappaB in inflammation. Nat. Rev. Immunol. 8, 3573–3577 (2001).
clinical implications, and pathogenesis. Arch. Pathol. 837–848 (2008). 64. Brentnall, T. et al. Mutations in the p53 gene: an early
Lab. Med. 134, 876–895 (2010). 39. Atreya, I., Atreya, R. & Neurath, M. F. NF‑κB in marker of neoplastic progression in ulcerative colitis.
16. van Schaik, F. D. M. et al. Endoscopic and pathological inflammatory bowel disease. J. Intern. Med. 263, Gastroenterology 107, 369–378 (1994).
aspects of colitis-associated dysplasia. Nat. Rev. 591–596 (2008). 65. Kato, S. et al. Understanding the function-structure
Gastroenterol. Hepatol. 6, 671–678 (2009). 40. Greten, F. R. et al. IKKβ links inflammation and and function-mutation relationships of p53 tumor
17. DeRoche, T. C., Xiao, S.‑Y. & Liu, X. Histological tumorigenesis in a mouse model of colitis-associated suppressor protein by high-resolution missense
evaluation in ulcerative colitis. Gastroenterol. Rep. 2, cancer. Cell 118, 285–296 (2004). mutation analysis. Proc. Natl Acad. Sci. USA 100,
178–192 (2014). 41. Brown, J. B. et al. Mesalamine inhibits epithelial 8424–8429 (2003).
18. Mooiweer, E. et al. Chromoendoscopy for surveillance beta‑catenin activation in chronic ulcerative colitis. 66. Chang, W. C. I. et al. Loss of p53 enhances the
in inflammatory bowel disease does not increase Gastroenterology 138, 595–605.e3 (2010). induction of colitis-associated neoplasia by dextran
neoplasia detection compared with conventional 42. Lee, G. et al. Phosphoinositide 3‑kinase signaling sulfate sodium. Carcinogenesis 28, 2375–2381
colonoscopy with random biopsies: results from mediates B‑catenin activation in intestinal epithelial (2007).
a large retrospective study. Am. J. Gastroenterol. stem and progenitor cells in colitis. 139, 869–881 67. Lamlum, H. et al. APC mutations are sufficient for the
110, 1014–1021 (2015). (2010). growth of early colorectal adenomas. Proc. Natl Acad.
19. Zisman, T. L. et al. Prospective study of the 43. Schwitalla, S. et al. Intestinal tumorigenesis initiated Sci. USA 97, 2225–2228 (2000).
progression of low-grade dysplasia in ulcerative colitis by dedifferentiation and acquisition of stem-cell-like 68. Dow, L. E. et al. Apc restoration promotes
using current cancer surveillance guidelines. properties. Cell 152, 25–38 (2013). cellular differentiation and reestablishes crypt
Inflamm. Bowel Dis. 18, 2240–2246 (2012). 44. Mudter, J. et al. Activation pattern of signal homeostasis in colorectal cancer. Cell 161,
20. Nowell, P. C. The clonal evolution of tumor cell transducers and activators of transcription (STAT) 1539–1552 (2015).
populations. Science 194, 23–28 (1976). factors in inflammatory bowel diseases. 69. Shenoy, A. K. et al. Transition from colitis to cancer:
21. Merlo, L. M. F., Pepper, J. W., Reid, B. J. & Maley, C. C. Am. J. Gastroenterol. 100, 64–72 (2005). high Wnt activity sustains the tumor-initiating
Cancer as an evolutionary and ecological process. 45. Bollrath, J. et al. gp130‑mediated Stat3 activation potential of colon cancer stem cell precursors.
Nat. Rev. Cancer 6, 924–935 (2006). in enterocytes regulates cell survival and cell-cycle Cancer Res. 72, 5091–5100 (2012).
22. Greaves, M. & Maley, C. C. Clonal evolution in cancer. progression during colitis-associated tumorigenesis. 70. Miyoshi, H. et al. Wnt5a potentiates TGF‑β signaling
Nature 481, 306–313 (2012). Cancer Cell 15, 91–102 (2009). to promote colonic crypt regeneration after tissue
23. Sansom, O. J. et al. Loss of Apc in vivo immediately 46. Marusyk, A., Tabassum, D. P., Altrock, P. M., injury. Science 338, 108–113 (2012).
perturbs Wnt signaling, differentiation, and migration. Almendro, V. & Michor, F. Non-cell-autonomous 71. The Cancer Genome Atlas Network. Comprehensive
Genes Dev. 18, 1385–1390 (2004). driving of tumour growth supports sub-clonal molecular characterization of human colon and rectal
24. Leedham, S. J. et al. Clonality, founder mutations, heterogeneity. Nature 514, 54–58 (2014). cancer. Nature 487, 330–337 (2012).
and field cancerization in human ulcerative 47. Michael-Robinson, J. M. et al. Characterization 72. Schulmann, K. et al. Molecular phenotype of
colitis–associated neoplasia. Gastroenterology of tumour-infiltrating lymphocytes and apoptosis inflammatory bowel disease-associated neoplasms
136, 542–550.e6 (2009). in colitis-associated neoplasia: comparison with with microsatellite instability. Gastroenterology 129,
25. Galandiuk, S. et al. Field cancerization in the sporadic colorectal cancer. J. Pathol. 208, 381–387 74–85 (2005).
intestinal epithelium of patients with Crohn’s (2006). 73. van Dieren, J. M. et al. Chromosomal and
ileocolitis. Gastroenterology 142, 855–864.e8 48. Arthur, J. C. et al. Microbial genomic analysis reveals microsatellite instability of adenocarcinomas
(2012). the essential role of inflammation in bacteria-induced and dysplastic lesions (DALM) in ulcerative colitis.
26. Holzmann, K. et al. Comparative analysis of histology, colorectal cancer. Nat. Commun. 5, 4724 (2014). Diagn. Mol. Pathol. 15, 216–222 (2006).
DNA content, 53 and Ki‑ras mutations in colectomy 49. Arthur, J. C. et al. Intestinal inflammation targets 74. Vermeulen, L. & Snippert, H. J. Stem cell dynamics
specimens with long-standing ulcerative colitis. cancer-inducing activity of the microbiota. Science in homeostasis and cancer of the intestine. Nat. Rev.
Int. J. Cancer 76, 1–6 (1998). 338, 120–123 (2012). Cancer 14, 468–480 (2014).
27. Chaubert, P., Benhattar, J., Saraga, E. & Costa, J. 50. Schwerbrock, N. M. J. et al. Interleukin 10‑deficient 75. Barker, N. et al. Identification of stem cells in small
K‑Ras mutations and p53 alterations in neoplastic mice exhibit defective colonic Muc2 synthesis before intestine and colon by marker gene Lgr5. Nature 449,
and nonneoplastic lesions associated with longstanding and after induction of colitis by commensal bacteria. 1003–1007 (2007).
ulcerative colitis. Am. J. Pathol. 144, 767–775 (1994). Inflamm. Bowel Dis. 10, 811–823 (2004). 76. Ritsma, L. et al. Intestinal crypt homeostasis revealed
28. Slaughter, D. P., Southwick, H. W. & Smejkal, W. 51. Prorok-Hamon, M. et al. Colonic mucosa-associated at single-stem-cell level by in vivo live imaging. Nature
‘Field cancerization’ in oral stratified squamous diffusely adherent afaC+ Escherichia coli expressing 507, 362–365 (2014).
epithelium. Clinical implications of multicentric origin. lpfA and pks are increased in inflammatory bowel 77. Tian, H. et al. A reserve stem cell population in
Cancer 6, 963–968 (1953). disease and colon cancer. Gut 63, 761–770 (2014). small intestine renders Lgr5‑positive cells dispensable.
29. Braakhuis, B. J. M., Tabor, M. P., Kummer, J. A., 52. Kostic, A. D., Xavier, R. J. & Gevers, D. The Nature 478, 255–259 (2011).
Leemans, C. R. & Brakenhoff, R. H. A genetic microbiome in inflammatory bowel disease: current 78. Barker, N. et al. Crypt stem cells as the
explanation of Slaughter’s concept of field status and the future ahead. Gastroenterology 146, cells‑of‑origin of intestinal cancer. Nature 457,
cancerization: evidence and clinical implications. 1489–1499 (2014). 608–611 (2009).
Cancer Res. 63, 1727–1730 (2003). 53. McCoy, A. N. et al. Fusobacterium is associated with 79. Humphries, A. & Wright, N. A. Colonic crypt
30. Zhu, D. et al. K‑Ras gene mutations in normal colorectal adenomas. PLoS ONE 8, e53653 (2013). organization and tumorigenesis. Nat. Rev. Cancer
colorectal tissues from K‑ras mutation-positive 54. Strauss, J. et al. Invasive potential of gut mucosa- 8, 415–424 (2008).
colorectal cancer patients. Cancer Res. 57, derived Fusobacterium nucleatum positively 80. Snippert, H. J. et al. Intestinal crypt homeostasis
2485–2492 (1997). correlates with IBD status of the host. Inflamm. Bowel results from neutral competition between
31. Hsieh, J. C. F., Van Den Berg, D., Kang, H., Dis. 17, 1971–1978 (2011). symmetrically dividing Lgr5 stem cells. Cell 143,
Hsieh, C.‑L. L. & Lieber, M. R. Large chromosome 55. Rhodes, J. M., Campbell, B. J. & Yu, L.‑G. 134–144 (2010).
deletions, duplications, and gene conversion events Lectin‑epithelial interactions in the human colon. 81. Vermeulen, L. et al. Defining stem cell dynamics in
accumulate with age in normal human colon crypts. Biochem. Soc. Trans. 36, 1482–1486 (2008). models of intestinal tumor initiation. Science 342,
Aging Cell 12, 269–279 (2013). 56. Itzkowitz, S. H. et al. Sialosyl‑Tn antigen is prevalent 995–998 (2013).
32. Nosho, K. et al. A prospective cohort study shows and precedes dysplasia in ulcerative colitis: 82. Biswas, S. et al. Microenvironmental control of stem
unique epigenetic, genetic, and prognostic features a retrospective case-control study. Gastroenterology cell fate in intestinal homeostasis and disease.
of synchronous colorectal cancers. Gastroenterology 110, 694–704 (1996). J. Pathol. 237, 135–145 (2015).
137, 1609–1620.e3 (2009). 57. Robles, A. I. et al. Whole-exome sequencing analyses 83. Francoeur, C. et al. Degeneration of the pericryptal
33. Cooper, H. S., Murthy, S., Kido, K., Yoshitake, H. of inflammatory bowel disease-associated colorectal myofibroblast sheath by proinflammatory cytokines
& Flanigan, A. Dysplasia and cancer in the dextran cancers. Gastroenterology 150, 931–943 (2016). in inflammatory bowel diseases. Gastroenterology
sulfate sodium mouse colitis model. Relevance to 58. Yaeger, R. et al. Genomic alterations observed in 136, 268–277.e3 (2009).
colitis-associated neoplasia in the human: a study colitis-associated cancers are distinct from those found 84. Yao, T. & Talbot, I. C. The demonstration of pericryptal
of histopathology, B‑catenin and p53 expression in sporadic colorectal cancers and vary by type of fibroblasts in background mucosa and dysplasia
and the role of inflammation. Carcinogenesis 21, inflammatory bowel disease. Gastroenterology 151, complicating ulcerative colitis. Histopathology 28,
757–768 (2000). 278–287.e6 (2016). 325–331 (1996).

228 | APRIL 2017 | VOLUME 14 www.nature.com/nrgastro


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

85. Greaves, L. C. et al. Mitochondrial DNA mutations are for predicting outcome in cancer. Proc. Natl Acad. 121. Gupta, R. B. et al. Histologic inflammation is a risk
established in human colonic stem cells, and mutated Sci. USA 103, 5923–5928 (2006). factor for progression to colorectal neoplasia in
clones expand by crypt fission. Proc. Natl Acad. 103. Papaemmanuil, E. et al. Genomic classification and ulcerative colitis: a cohort study. Gastroenterology
Sci. USA 103, 714–719 (2006). prognosis in acute myeloid leukemia. N. Engl. J. Med. 133, 1099–1105 (2007).
86. Langlands, A. J. et al. Paneth cell-rich regions 374, 2209–2221 (2016). 122. Zallot, C. & Peyrin-Biroulet, L. Deep remission
separated by a cluster of Lgr5+ cells initiate crypt 104. Masel, J. & Trotter, M. V. Robustness and evolvability. in inflammatory bowel disease: looking beyond
fission in the intestinal stem cell niche. PLoS Biol. 14, Trends Genet. 26, 406–414 (2010). symptoms. Curr. Gastroenterol. Rep. 15, 315
e1002491 (2016). 105. Maley, C. C. et al. Genetic clonal diversity predicts (2013).
87. Loeffler, M., Bratke, T., Paulus, U., Li, Y. Q. progression to esophageal adenocarcinoma. 123. Levine, D. S. et al. Distribution of aneuploid cell
& Potten, C. S. Clonality and life cycles of intestinal Nat. Genet. 38, 468–473 (2006). populations in ulcerative colitis with dysplasia or
crypts explained by a state dependent stochastic 106. Martinez, P. et al. A dynamic clonal equilibrium cancer. Gastroenterology 101, 1198–1210 (1991).
model of epithelial stem cell organization. J. Theor. predicts cancer risk in Barrett’s oesophagus. 124. Brentnall, T. A. et al. Microsatellite instability in
Biol. 186, 41–54 (1997). Nat. Commun. 7, 12158 (2016). nonneoplastic mucosa from patients with chronic
88. Cheng, H., Bjerknes, M., Amar, J. & Gardiner, G. 107. Salk, J. J. et al. Clonal expansions in ulcerative colitis ulcerative colitis. Cancer Res. 56, 1237–1240
Crypt production in normal and diseased human identify patients with neoplasia. Proc. Natl Acad. (1996).
colonic epithelium. Anat. Rec. 216, 44–48 (1986). Sci. USA 106, 20871–20876 (2009). 125. Willenbucher, R., Zelman, S., Ferrell, L., Moore, D.
89. Snippert, H. J., Schepers, A. G., Van Es, J. H., 108. Li, X. et al. Temporal and spatial evolution of somatic & Waldman, F. Chromosomal alterations in ulcerative
Simons, B. D. & Clevers, H. Biased competition chromosomal alterations: a case-cohort study of colitis-related neoplastic progression.
between Lgr5 intestinal stem cells driven by oncogenic Barrett’s esophagus. Cancer Prev. Res. (Phila.) 7, Gastroenterology 113, 791–801 (1997).
mutation induces clonal expansion. EMBO Rep. 15, 114–127 (2014). 126. Willenbucher, R. F. et al. Genomic instability is
62–69 (2014). 109. Itzkowitz, S. H. et al. Sialosyl‑Tn antigen: initial report an early event during the progression pathway
90. Pruitt, S. C., Freeland, A. & Kudla, A. Cell cycle of a new marker of malignant progression in long- of ulcerative-colitis-related neoplasia. Am. J. Pathol.
heterogeneity in the small intestinal crypt and standing ulcerative colitis. Gastroenterology 109, 154, 1825–1830 (1999).
maintenance of genome integrity. Stem Cells 28, 490–497 (1995). 127. O’Sullivan, J. N. et al. Chromosomal instability in
1250–1259 (2010). 110. Lofberg, R., Brostrom, O., Karlen, P., Ost, A. ulcerative colitis is related to telomere shortening.
91. Schäfer, M. & Werner, S. Cancer as an overhealing & Tribukait, B. DNA aneuploidy in ulcerative-colitis — Nat. Genet. 32, 280–284 (2002).
wound: an old hypothesis revisited. Nat. Rev. Mol. Cell reproducibility, topographic distribution, and relation to 128. Chen, R. et al. DNA fingerprinting abnormalities can
Biol. 9, 628–638 (2008). dysplasia. Gastroenterology 102, 1149–1154 (1992). distinguish ulcerative colitis patients with dysplasia
92. Lai, L. A. et al. Pan-colonic field defects are detected 111. Burmer, G. C. et al. Neoplastic progression in and cancer from those who are dysplasia/cancer-free.
by CGH in the colons of UC patients with dysplasia/ ulcerative colitis: histology, DNA content, and loss Am. J. Pathol. 162, 665–672 (2003).
cancer. Cancer Lett. 320, 180–188 (2012). of a p53 allele. Gastroenterology 103, 1602–1610 129. Yoshida, T., Mikami, T., Mitomi, H. & Okayasu, I.
93. Yui, S. et al. Functional engraftment of colon (1992). Diverse p53 alterations in ulcerative colitis-associated
epithelium expanded in vitro from a single adult Lgr5+ 112. Greaves, L. C. et al. Clonal expansion of early to low-grade dysplasia: full-length gene sequencing
stem cell. Nat. Med. 18, 618–623 (2012). mid‑life mitochondrial DNA point mutations drives in microdissected single crypts. J. Pathol. 199,
94. Nystul, T. & Spradling, A. An epithelial niche in the mitochondrial dysfunction during human ageing. 166–175 (2003).
Drosophila ovary undergoes long-range stem cell PLoS Genet. 10, e1004620 (2014). 130. Sjöqvist, U. et al. Colorectal cancer in colonic
replacement. Cell Stem Cell 1, 277–285 (2007). 113. Shergill, A. K. et al. The role of endoscopy in Crohn’s disease — high frequency of DNA-aneuploidy.
95. Thorsteinsdottir, S., Gudjonsson, T., Nielsen, O. H., inflammatory bowel disease. Gastrointest. Endosc. 81, Anticancer Res. 25, 4393–4397 (2005).
Vainer, B. & Seidelin, J. B. Pathogenesis and 1101–1121.e13 (2015). 131. Garrity-Park, M. M., Loftus, E. V. J., Sandborn, W. J.,
biomarkers of carcinogenesis in ulcerative colitis. 114. Rabinovitch, P. S. et al. Pancolonic chromosomal Bryant, S. C. & Smyrk, T. C. Methylation status
Nat. Rev. Gastroenterol. Hepatol. 8, 395–404 instability precedes dysplasia and cancer in ulcerative of genes in non-neoplastic mucosa from patients
(2011). colitis. Cancer Res. 59, 5148–5153 (1999). with ulcerative colitis-associated colorectal cancer.
96. Rubin, C. E. et al. DNA aneuploidy in colonic 115. Torres, J., de Chambrun, G. P., Itzkowitz, S., Am. J. Gastroenterol. 105, 1610–1619 (2010).
biopsies predicts future development of dysplasia Sachar, D. B. & Colombel, J.‑F. Review article: 132. Söderlund, S. et al. Colitis-associated DNA
in ulcerative colitis. Gastroenterology 103, colorectal neoplasia in patients with primary aneuploidy and dysplasia in Crohn’s disease and
1611–1620 (1992). sclerosing cholangitis and inflammatory bowel disease. risk of colorectal cancer. Inflamm. Bowel Dis. 17,
97. Lashner, B. A., Shapiro, B. D., Husain, A. Aliment. Pharmacol. Ther. 34, 497–508 (2011). 1101–1107 (2011).
& Goldblum, J. R. Evaluation of the usefulness of 116. Laine, L. et al. SCENIC international consensus 133. Salk, J. J. et al. Clonal expansions and short telomeres
testing for p53 mutations in colorectal cancer statement on surveillance and management of are associated with neoplasia in early-onset, but not
surveillance for ulcerative colitis. Am. J. Gastroenterol. dysplasia in inflammatory bowel disease. late-onset, ulcerative colitis. Inflamm. Bowel Dis. 19,
94, 456–462 (1999). Gastroenterology 148, 639–651.e28 (2015). 2593–2602 (2013).
98. Gerrits, M. M. et al. Biomarker-based prediction 117. Algra, A. M. et al. Effects of regular aspirin on long- 134. Choi, W.‑T. et al. Outcome of ‘indefinite for dysplasia’
of inflammatory bowel disease-related colorectal term cancer incidence and metastasis: a systematic in inflammatory bowel disease: correlation with
cancer: a case-control study. Cell. Oncol. (Dordr.) 34, comparison of evidence from observational studies DNA flow cytometry and other risk factors of
107–117 (2011). versus randomised trials. Lancet Oncol. 13, 518–527 colorectal cancer. Hum. Pathol. 46, 939–947 (2015).
99. Marusyk, A., Almendro, V. & Polyak, K. Intra-tumour (2012).
heterogeneity: a looking glass for cancer? Nat. Rev. 118. Asano, T. K. & McLeod, R. S. Non steroidal Acknowledgements
Cancer 12, 323–334 (2012). anti‑inflammatory drugs (NSAID) and aspirin for The authors are grateful for funding from Cancer Research
100. Solaymani-Dodaran, M., Logan, R. F. A., West, J., preventing colorectal adenomas and carcinomas. UK, the Medical Research Council, the Derek Willoughby
Card, T. & Coupland, C. Risk of oesophageal cancer Cochrane Database Syst. Rev. 2, CD004079 (2004). Fund for Inflammatory Research, the St Mark’s Hospital
in Barrett’s oesophagus and gastro-oesophageal 119. Kostadinov, R. L. et al. NSAIDs modulate clonal Foundation and Barts Charity.
reflux. Gut 53, 1070–1074 (2004). evolution in Barrett’s esophagus. PLoS Genet. 9,
101. Weaver, J. M. J. et al. Ordering of mutations e1003553 (2013). Author contributions
in preinvasive disease stages of esophageal 120. Zhao, L.‑N. et al. 5‑aminosalicylates reduce the risk The authors contributed equally to the review.
carcinogenesis. Nat. Genet. 46, 837–843 (2014). of colorectal neoplasia in patients with ulcerative
102. Ein-Dor, L., Zuk, O. & Domany, E. Thousands of colitis: an updated meta-analysis. PLoS ONE 9, Competing interests statement
samples are needed to generate a robust gene list e94208 (2014). The authors declare no competing interests.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 14 | APRIL 2017 | 229


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like