Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

REVIEWS

CANCER ORIGINS

An evolutionary perspective on field


cancerization
Kit Curtius, Nicholas A. Wright and Trevor A. Graham
Abstract | Tumorigenesis begins long before the growth of a clinically detectable lesion and,
indeed, even before any of the usual morphological correlates of pre-malignancy are
recognizable. Field cancerization, which is the replacement of the normal cell population by a
cancer-primed cell population that may show no morphological change, is now recognized to
underlie the development of many types of cancer, including the common carcinomas of the
lung, colon, skin, prostate and bladder. Field cancerization is the consequence of the evolution
of somatic cells in the body that results in cells that carry some but not all phenotypes required
for malignancy. Here, we review the evidence of field cancerization across organs and examine
the biological mechanisms that drive the evolutionary process that results in field creation.
We discuss the clinical implications, principally, how measurements of the cancerized field
could improve cancer risk prediction in patients with pre-malignant disease.

Cell lineage Field cancerization is a paradigm for tumorigenesis itself. Pre-malignant diseases — morphologically discern­
A group of cells that share a Before the growth of a malignant lesion, a normal cell ible conditions that increase cancer risk — are examples
recent common ancestor cell; ­lineage can acquire pro-tumorigenic genetic mutations or of field cancerization with associated morphological
also known as a clone.
epimutations (hereafter collectively referred to as ‘muta- change. Pre-malignant disease is recognized in many dif-
Epimutations tions’) that are positively selected for in the microenvi- ferent organs, perhaps most notably in Barrett oesopha-
Gains or losses of DNA ronment of an otherwise healthy organ. Consequently, the gus (BE)3, ductal carcinoma in situ (DCIS) in the breast4
methylation or heritable mutant lineage, also referred to as a ‘mutant clone’, can and prostatic intraepithelial neoplasia (PIN)5. These pre-­
chromatin changes; such grow to produce large patches, or fields, of cells that are malignant lesions all represent the growth of mutant lin-
changes are in contrast to
genetic changes to DNA
predisposed to eventually progress to a neoplasm (FIG. 1). eages that are putatively further on an evolutionary path
nucleotides. A cancerized field is a group of cells that are considered to towards cancer. Because the removal of cancerized fields
be further along an evolutionary path towards cancer 1. is often associated with substantial risk of morbidity, or
Cancerized field The microenvironment surrounding the mutant cells may even mortality (for instance, in BE6 or inflammatory bowel
A collection of cells that have
also be abnormal and/or be altered by the cancerized field; disease (IBD)7), they are typically left in situ, and watchful
gained some but not all the
phenotypic alterations the interplay between the mutant cells and their micro­ waiting (longitudinal surveillance typically with biopsy
required for malignancy; in environment determines which mutations are selected for. sample c­ ollection) is performed8,9 (BOX 1).
general, the altered phenotype The field may or may not exhibit morphological change Discerning the degree of cancerization of a field at
will have been caused by an (for example, cancerized cells may look normal or could the genetic level requires making the important — but
underlying mutation.
exhibit dysplasia). Field cancerization is interchangeably challenging — distinction between mutant lineages and
referred to in the literature as field effects or field defects. cancerized lineages. The former may have many somatic
There is evidence that cancerized field development, mutations that are inconsequential to tumorigenesis,
with or without overt morphological change, occurs whereas the latter will have mutations that, in an appro-
across tissue types and throughout the body (TABLE 1). priate microenvironmental context, increase the rate of,
The size of the cancerized field varies substantially, or drive, cancer development. Mutations accrue steadily
and a cancerized area of tissue will have a microscopic throughout life in ageing tissues10, and the neutral com-
morphology classified as noncancerous, that is, normal, petition (drift) between the adult stem cells that maintains
Centre for Tumour Biology,
Barts Cancer Institute, ­ yperplasia, metaplasia or dysplasia2. In a technical sense,
h the tissue causes the clonal expansion of some of these
EC1M 6BQ London, UK. the smallest cancerized field is a single lineage — even a mutants11; therefore we could broadly consider all mutant
Correspondence to T.A.G. single cell — that has evolved towards cancer but has not cells to be cancerized, and then we would logically have to
t.graham@qmul.ac.uk yet itself become malignant. Together, these data imply believe that the entire body becomes increasingly cancer-
doi:10.1038/nrc.2017.102 that every cancer arises from a potentially detectable ized as it grows older. This definition is clearly not specific
Published online 8 Dec 2017 ­cancerized condition. enough to be useful.

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 1


©
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

a b Mutagenic c
insult

Normal cell Cancerized cell Malignant cell

Figure 1 | Characterization and initiation of a cancerized field. a | A sporadic tumour is a lesionNature
that does not share
Reviews its
| Cancer
tumorigenic mutations with surrounding tissue. The mucosa surrounding the tumour is typically morphologically and
phenotypically normal. b | By contrast, a tumour growing in a cancerized field is one that began from a malignant cell that
initially shared tumorigenic mutations and concomitant altered phenotype with the surrounding tissue; these alterations
led to expansion of its ancestral lineage to form a field of cancer-primed cells. Within the field, the malignancy evolved
because of additional mutations (and phenotypic change) in an evolving and potentially cancer-promoting
microenvironmental context. The cancerized field of cells harbouring tumorigenic mutations may appear morphologically
abnormal (for example, dysplastic) or normal, but the cells in the cancerized field will have an altered phenotype — such
as increased survival — that underpins the expansion of the field. c | A cancerized field may be independently initiated by
many cells following a mutagenic insult. Such an insult may provide cells with an advantageous phenotype as a direct
consequence of DNA damage (for example, mutations caused by ultraviolet radiation) within a key gene and/or by
providing an enabling microenvironment (for example, microenvironmental changes induced by cigarette smoke).
These initiated cells, which occur in varying numbers, will clonally expand to create a (mostly) contiguous patch of
cancer-primed cells.

Rather, we here propose that a cancerized field is particular environmental ­conditions — and furthermore
best described by its phenotypic properties. Our revised that the altered pheno­type of the cancerized lineage will
definition of a cancerized field is a collection of cells be the mechanistic cause of its expansion. Consequently,
that have gained some but not all the phenotypic alter- a cancer arising from a field passes through multiple
Dysplasia ations required for malignancy; in general, this altered pheno­type states, whereas the lineage leading to a s­ poradic
A feature of tissues in which
cells have an abnormal
pheno­type will have been caused by underlying muta- tumour may not have had any p ­ henotypic changes before
morphology or arrangement, tion. These phenotypic changes could include proper- cancer growth.
usually regarded as being an ties such as an increased growth rate, decreased death Inherited cancer risk — for example through the
unequivocal neoplastic rate or increased immune evasion; consequently, the inheritance of a defective copy of a tumour suppressor
alteration.
pheno­typic changes need not be morphological changes. gene — also leads to cells in the body being predisposed
Hyperplasia These phenotypic changes will have been caused by to cancer. We reserve the term ‘field cancerization’ to
A feature of tissues in which an mutations in key cancer-associated genes that act either apply to only a somatic evolutionary process that pro-
increase in cell number occurs autonomously or in tandem with an altered microenvi- duces cells that are ‘closer to’ cancer. Genetically mosaic dis-
without malignant change. ronment. The field need not be monoclonal in origin. ease, resulting from mutation and consequent pheno­typic
Metaplasia
Moreover, this definition avoids the challenge inherent changes arising during development, is an i­ntermediate
A feature of tissues in which in p
­ rescribing importance to individual mutations. example of field cancerization15.
the usual cells of a tissue are We note that cancerized phenotypes may be subtle In this Review, we discuss what combination of
replaced with a cell type that and/or transient. For instance, mutation of TP53 (which geno­types and phenotypes constitutes a cancerized
morphologically resembles
encodes p53) in the skin provides a survival a­ dvantage — field, how such fields are initiated and how they expand,
another tissue type.
a cancerized phenotype — but this phenotype is evident and end by considering the clinical implications of
Inflammatory bowel disease only in response to ultraviolet exposure12. An initial muta- field cancerization.
(IBD). A group of inflammatory tion in the adenomatous polyposis coli (APC) tumour
conditions of the bowel that suppressor gene in the colon is clearly a step towards A brief history of field cancerization
includes ulcerative colitis and
Crohn’s disease.
colorectal cancer (CRC), and cells within crypts with Historical definition of a cancerized field. In 1953,
mono-allelic APC mutation show increased methy­lation Slaughter et al. first proposed the concept of field can-
Sporadic tumour pattern diversity that is indicative of an increased size of cerization in order to explain the statistical enrichment
A tumour that does not share the intestinal stem cell niche and/or increased stem cell of the multifocal (rather than isolated) oral squamous
tumorigenic mutations and/or
lineage survival13,14. Our phenotypic definition excludes carcinomas they found in 783 patients with cancer 16. The
other neoplastic changes in
phenotype with the regions of tissue having DNA damage or mutations that authors hypothesized that the patches of microscopically
surrounding tissue. do not yet have a cancer-related pheno­type, excluding abnormal (but histologically benign) tissue surround-
even oncogene or tumour suppressor gene mutations that ing the tumours suffered from the “preconditioning of
Genetically mosaic do not cause a cancer-­related pheno­type until additional an area of epithelium to cancer growth by a carcino-
A feature of a group of cells
that is composed of two or
mutations emerge. Nevertheless, we suspect that most, genic agent” (REF. 16). In the decades that followed, the
more clonal populations, each if not all, cancer-­associated mutations will have pheno- advent of molecular biology fostered genetic analyses
with a different genotype. typic consequences in normal tissue — at least under that revealed that fields of epithelial cells predisposed

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
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

to malignancy were, in some cases, composed of a clone population. We note that this previous definition implies
derived from a mutated cell2. In one striking example, that the cancerized clone has an altered phenotype that
a patient was reported to have a clonal expansion of a drives its expansion and thus is consistent with our
single TP53 mutation across both lungs17. Thus, the defi- ­current definition.
nition for field cancerization was revised by Braakhuis More recently, fields of epithelial cells carrying
et al. to be “the growth of a mutant clone to produce a somatic epimutations (abnormal DNA methylation)
field of cells predisposed to subsequent tumour growth” have been noted, for example, hypermethylation of
(REF. 1). To frame this previous idea from an overtly evo- various promoters in a Helicobacter pylori-infected
lutionary perspective, we need only note that the mutant stomach18 or similar patterns of unusual DNA methyla-
clone that expands to form the field is fitter (more likely tion in synchronous, spatially separate CRCs19. In such
to produce surviving offspring) than the resident cell cases, the high rate of de novo DNA methylation, the

Table 1 | Examples of field cancerization in human tissues


Cancer type Pre-malignant diagnosis Evidence Clinical importance Comments Refs
(recommended action)
Colon carcinoma in IBD, dysplasia Mutations, aberrant Yes (resection of all Seen in both ulcerative colitis 32,33,
patients with IBD methylation, altered affected area necessary) and Crohn’s disease; can involve 137,138
proteomics whole colon
Colon carcinoma in Adenoma Mutations, aberrant Unknown (removal) Importance unclear; unknown 139–146
non-IBD patients methylation fraction progress
Gastric carcinoma Intestinal metaplasia Mutations, aberrant Yes; occurs in intestinal Not all metaplasias progress 18,66,
methylation metaplasia, a known 147–151
pre-malignant state
(surveillance)
Oesophageal Squamous dysplasia Mutations, aberrant Yes; high possibility of Smoking-associated 152–158
squamous methylation, altered metachronous lesions
carcinoma proteomics (surveillance)
Oesophageal BE, dysplasia Mutations, altered Yes (surveillance) Early evidence implicated the 111,
adenocarcinoma metabolomics development of a cancerized 159–161
field, but later studies showed
carcinoma developed focally
with no selective sweeps
Non-small-cell Dysplasia, carcinoma in situ Mutations Yes (review after Smoking-associated; can be 17,46,
lung squamous resection important) very widespread; amenable to 162–166
carcinoma screening
Non-small-cell lung AAH, AIS Mutations Possible (none) A continuum exists between the 167–171
adenocarcinoma precursor lesions AAH and AIS
and adenocarcinoma
Small-cell lung Diffuse idiopathic Mutations Possible (none) Smoking-associated; can be 167–170,
carcinoma pulmonary neuroendocrine very widespread 172
cell hyperplasia
HNSCC (oral, Leukoplakia, erythroplakia, Mutations Yes; high possibility of These are squamous carcinomas 1,16,
oropharynx, oral submucous fibrosis, metachronous lesions of the oral and pharyngeal 173–179
hypopharynx and lichen planus, dysplasia (surveillance and mucosa
larynx carcinoma) resection)
Breast carcinoma DCIS, ductal hyperplasia Mutations, aberrant Yes; possibility of Explains the presence of 180–184
methylation metachronous lesions multiple areas of DCIS
(resection)
Prostate carcinoma PIN Mutations, altered Possible (surveillance) Multifocal carcinomas of 185–187
proteomics, aberrant prostate are common
methylation
Bladder carcinoma Intestinal metaplasia, Mutations Yes (surveillance, Multifocal tumours are common 188–190
dysplasia, carcinoma in situ resection and therapy)
Skin carcinoma Actinic keratosis, Bowen Mutations Yes (surveillance and Common in face and scalp; 22,
disease treatment) the whole field is treated 191–194
Melanoma Acquired nevi, dysplastic Mutations Yes (surveillance and Suspect lesions are resected 195
nevi (atypical nevi) and treatment)
congenital nevi
Blood cancer None Mutations Yes (surveillance) Clones seen in a substantial 92–94
percentage of newborn babies
AAH, atypical adenomatous hyperplasia; AIS, adenocarcinoma in situ; BE, Barrett oesophagus; DCIS, ductal carcinoma in situ; HNSCC, head and neck squamous
cell carcinoma; IBD, inflammatory bowel disease; PIN, prostatic intraepithelial neoplasia.

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 3


©
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

Box 1 | Pre-malignant disease: a resource for cancer evolution studies simply having a field cancerization driver may not be
sufficient to cause a phenotypic change in the current
Fortuitously for the research community, the clinical practice of watchful waiting for ­microenvironmental condition.
pre-malignant lesions leads to extremely well-characterized longitudinal tissue In skin (squamous epithelium), putative field cancer-
catalogues that facilitate the temporal study of clonal evolution in situ in humans. Such ization driver mutations are found in genes, including
study can reveal the pace and pattern of cancerized clone evolution and/or phenotype
Notch family members and TP53. Mutations in these
generation and spread109,134, and this information can be directly leveraged to optimize
biomarker development and screening regimens135. The exciting multi-institution genes are detected at high frequency in apparently normal
Pre-Cancer Genome Atlas (PCGA) aims to compile a comprehensive characterization sun-exposed skin cells26. Mouse models show that Notch
of the molecular alterations in pre-malignant lesions as well as, importantly, the family mutants are clonally selected for in oesopha­
corresponding changes in the microenvironment and cellular phenotypes136. It is also geal squamous cell epithelium owing to their ability to
crucial that the PCGA contains longitudinal data so that the context-specific nature inhibit differentiation27. TP53 mutation has been long
of cancer evolution and the concomitant intricacies of the genotype–phenotype recognized to be present in nondysplastic skin, and its
mapping, together with the critical rates of cancer evolution, can be fully elucidated. frequency increases with age28 and causes an increase in
the stochastic growth rate of the clone, but only in the
presence of chronic ultraviolet exposure12. Similarly, lin-
potential for demethylation and the likelihood of con- eage tracing in mice has shown that Hedgehog pathway
vergent epi­genetic evolution mean that the clonality of mutations are positively selected for in skin because they
the e­ pigenetic fields is open to question. Instead, the can- inhibit differentiation29. Remarkably, the density of clon-
cerized field is likely to have formed because of exposure ally expanded putative driver alterations in skin was esti-
to a prevalent mutagen and promoter of clonal expan- mated at ~140 driver mutations per square centimetre26
sion (for example, in the case of the H. pylori-infected — for example, more driver mutations can be found in
stomach) and/or subsequent convergent evolution of a small patch of morphologically normal skin than in a
the epigenome (for example, in the case of synchronous skin basal cell carcinoma30.
CRC). Thus, field cancerization can occur because of In the inflamed intestine, mouse models show that
multiple independent clonal expansions. Nevertheless, Trp53 mutation is a field cancerization driver because it
mutational diversity that exists within the cancerized increases stem cell replacement in a chronic inflammatory
field is likely a substrate for natural selection, and so over setting31. Interestingly, in the absence of inflammation, the
time, it is likely that the fittest clone (for example, that Trp53‑mutant clones have no advantage31, indicating
with the most adaptive phenotype) will come to dom- the interplay between mutant lineage and microenviron-
inate the field. Genetic drift will also shape the clonal mental context in determining selective advantage of the
composition of the field20. Related to this, others have phenotype induced by a field cancerization driver muta-
suggested that exposures that have pleiotropic effects on tion. Correspondingly, patients with IBD show clonal
cell phenotypes and microenvironmental inter­actions expansions of TP53‑mutants in nondysplastic bowel32,33,
across many organs — such as smoking — should be and we are unaware of reports of clonal expansion of
thought of as causing a body-wide ‘aetiological field TP53‑mutants in the colon mucosa in patients without
effect’ (REF. 21) that is clearly not clonal in origin. IBD. Patients with IBD also show clonal expansion of
Alterations of stromal cells can also promote field ­KRAS-mutant and/or aneuploid cells in the absence
cancerization. In the skin, epigenetic modification of of dysplasia32,33.
fibroblasts is induced by ultraviolet exposure, leading to To determine precisely which mutations are field can-
the production of diffusible growth factors, inflammatory cerization drivers requires in vivo lineage tracing, pref-
cytokines and matrix-remodelling enzymes that together erably in both model systems and human tissues. Such
predispose to subsequent epithelial tumours22. In this model systems can reveal the mechanism that provides a
particular example, the resulting epithelial tumours clone with a selective advantage (as per the above exam-
show recurrent genetic alterations, implying that the can- ples). Nevertheless, we note that the exact phenotypic
cerized stromal compartment alone is unable to cause consequences of many putative cancer driver mutations
a tumour but provides selective pressure for particular remain elusive, and their common occurrence in cancers
epithelial genotype–phenotype combinations. Because may in fact be because the driver is selected for before
the stroma does not transform into a tumour itself, here, overt cancer growth, that is, during field cancerization.
we do not consider the stroma itself to be cancerized. In cancers, broadly speaking, a mutation will be called a
driver if it occurs more frequently across tumours than
Field cancerization driver mutations. In cancer genom- is expected from the mutation rate alone34 — the excess
ics, a distinction is commonly made between driver frequency is then a consequence of positive selection for
mutations, which confer growth or survival advantages the mutation. However, this analysis does not show when
on tumour cells (within the appropriate microenvi- during the tumorigenic process the driver mutation was
ronment) and passenger (neutral) mutations, which positively selected for or if it is still under positive selec-
passively accumulate in cell lineages23–25. Borrowing tion within the tumour at the time it is observed. In other
that nomenclature, here, we term the mutations that words, the so‑called cancer driver may not actually drive
are functionally important for the phenotypic changes cancer growth per se but rather had previously driven the
Driver mutations
Mutations that are positively
that underpin the cancerized field ‘field cancerization growth of an ancestral lineage. Recognizing that driver
selected for and are implicated drivers’. We note that the phenotypic consequences mutation selection is inherently context-dependent and,
in tumorigenesis. of a driver mutation may be context dependent, so moreover, that the context changes over time provides a

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
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

convenient explanation for the high frequency at which because it is caused by a persistent environmental factor
prominent cancer-related genes are found in cancerized such as tobacco carcinogens, diet or infections21, then
fields35. We also note that in some instances, the same new clones will be continually generated, and the field
phenotype will be positively selected for in both the will continue to appear genetically mosaic.
cancerized field and in a cancer itself: for example, func- For example, mutant cancerized fields, often bear-
tional TP53 mutations appear to drive cancerization of ing mutations in TP53, are seen in the lung and air-
the inflamed intestine31, likely because they provide a sur- ways, and these mutations are associated with smoking
vival and/or growth advantage, and these same pheno­ exposure45,46. In the cervix, high-risk strains of human
types are clearly also important for subsequent cancer papillomavirus impair normal p53 function, a condition
development 36. Hence, a field cancerization driver may that is likely permissive for the development of a field of
also be a cancer driver. preinvasive cervical intraepithelial neoplasia47. H. pylori
Progression to cancer likely involves the accumula- infection is associated with field cancerization via intes-
tion of multiple field cancerization driver mutations37. tinal metaplasia in the stomach48, but recent whole-­
Phenotypic change is caused by either individual genome sequencing on 100 gastric tumours found no
high-impact mutations or epistasis among a synergis- association with mutational spectrum and H. pylori
tically acting group of mutations, each of which would infection49; this suggests that infection induces intestinal
be insufficient to cause a change to a cancerous pheno­ metaplasia fields via a nongenetic route, perhaps by pro-
type without the others. Alternatively, progression may viding a selective pressure for atrophic gastritis-­resistant
be caused by sudden large-scale mutational events that clones that bear epigenetic changes. This intriguing
simultaneously alter large parts of the genome and result highlights the need for further study to elucidate
elicit large phenotypic change: whole-genome doubling the role(s) of H. pylori infection in the formation of
appears to apply a transformative role in BE, for exam- cancerized clones in the stomach. In the oesophagus,
ple38,39. If mutations accrue gradually, then conceivably, gastroesophageal reflux disease (GERD) increases the
the field cancerization driver burden can be used to risk of oesophageal adenocarcinoma50, and a mutational
identify patients at risk of progression, whereas predict- signature thought to be characteristic of acid exposure
ing the likelihood of such ‘catastrophic’ events presents a is observed in the genomes of these cancers51, implying
larger challenge. that the cancer arose from a field mutationally affected
by GERD.
Mapping genotype to phenotype. Nuances in cell mor- Accelerated ageing — the increase in the rate of events
phology and behaviour mean that cells within a cancer- attributable to ageing — is implicated in tumorigenesis
ized field have phenotypes that theoretically occupy an in many tissues. Indeed, the majority of high-frequency
infinite phenotype space, where position in phenotype mutations found in a cancer (of which there are typically
space is determined partly by the underlying genotype very many) is thought to accrue before the initiation of
(FIG. 2). Understanding the genotype–phenotype map is tumour growth52, and it seems likely that these would
critical for understanding the biology and evolution of often be within a cancerized field. In patients with ulcer-
cancerized fields. The example of TP53 and other driver ative colitis, mutational signature analysis53, CpG island
genes given above shows that the genotype–phenotype hypermethylation54 and telomere shortening 55 have indi-
map is many‑to‑many (there is one or more genotype cated accelerated ageing in the colon. Utilizing epigenetic
that underlies each phenotype, but a genotype may incur drift as a molecular clock shows that the ages of BE tissue
different phenotypes on the basis of nongenetic effects); vary dramatically between patients of similar chronologi-
hence, the notion of a ‘genetic blueprint’ that entirely cal age56. Moreover, just as chronological age is recognized
determines the biology of a cancerized field is overly as one of the strongest predictors of cancer risk, biological
simplistic. Pathological diagnosis categorizes the mor- tissue age (that is, the time a tissue has had to accrue addi-
phological features of pre-malignant disease into a small tional stochastic alterations) contributes to field canceriza-
number of groups (for example, grade) — see TABLE 1. tion, and particular attention should be given to exploring
However, we note that the underlying multivariate nature its roles in neoplastic progression57.
of genotypes and phenotypes means that such classifi- It is noteworthy that some early neoplastic lesions,
cations are in fact continuous and fluid. This continuity such as adenomas in the colon58 and nevi in the skin59,
perhaps explains some of the evident challenges in reli­ are derived from multiple independent clones — that is,
ably identifying particular pre-malignant states, such as the neoplastic lesions are polyclonal in origin2. While the
a low-grade dysplasia in BE40,41. mechanism that causes this polyclonality is unknown,
a localized field defect provides a compelling explana-
Epistasis Initiation of the cancerized field tion: an initial mutant clone may produce some kind of
The interaction of multiple
Cancerized fields may be induced by ‘unlucky muta- field (through aberrant signalling, for example) that alters
genes that leads to the
development of a phenotypic tions’42 that occur owing to natural DNA replication the behaviour of surrounding stromal cells, creating an
trait. errors during ageing 43 or by mutagenic insult (FIG. 1c). environment that in some way promotes mutations in
Immediately following the mutagenic insult, the mutated neighbouring cells60. Indeed, in the colon, transformed
Polyclonal field will be composed of many genetically distinct intestinal crypts have profound effects on neighbouring
An attribute of a lesion that is
derived from two or more
clones, but over time, we expect that the clones with the wild-type crypts61,62. Physical compression of intestinal
clones as opposed to a fittest phenotypes will come to dominate the cancer- mucosa induces WNT pathway signalling, and so the
monoclonal origin. ized field44. If the mutagenic insult is ongoing, perhaps signalling induced by an initial expanded mutant clone

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 5


©
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

can result from mechanical cues rather than juxtacrine Glandular epithelium. In order for a clone to grow in
or paracrine signalling 63. Thus, the multitude of inter­ glandular tissue, such as intestine and BE mucosa, it
actions between clones is important in the establishment must first undergo niche succession, whereby a mutant
of cancerized fields. stem cell in the gland replaces all other stem cells (and
so subsequently, all differentiated cells in the gland
Mechanisms of clone growth will also be mutant); second, the repopulated mutant
Human epithelia are organized into two broadly dif- gland must produce a field of mutant glands by gland
ferent types, glandular and squamous, and we discuss fission13,65–67 (FIG. 3). The unit of selection in glandular
how clones grow in each epithelium type below. Tissue tissue therefore switches between individual stem cells
architecture constrains evolution by limiting the ability within a gland to the meta-population of the gland
of mutant clones to expand and in so doing lengthens the as a whole (the differentiated cells in the gland are
waiting time to cancer 64. the somatic lineage derived from the germ stem cells

SMAD4
TP53
Genotype CDKN2A
dimension 2 CDKN2A
TP53
CDKN2A
N

Genotype– Genotype
phenotype dimension 1
map

M
Phenotype C
dimension 2
D

N H

Phenotype dimension 1

b WGD c
TP53 TP53
Genotype
dimension 2
TP53

Genotype dimension 1
Epigenetic and
microenvironmental
influences

End End
Phenotype
dimension 2

Start Start

Phenotype dimension 1
Nature Reviews | Cancer

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
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

in the gland). Measurement of gland division rates and What is selected for? Broadly speaking, phenotypes
the impact of tumorigenic mutations upon the rate are associ­ated with increased growth and/or increased
generally lacking. In the healthy colon, gland fission survival will be positively selected for. In squamous
rates are very low — once every few decades13 — and so lesions, cells that are best able to adhere to the basement
it is clear that a field cancerization driver mutation that membrane will have increased survival because the lin-
clonally expands to form a large patch of glands can do eage will not migrate and differentiate, whereas faster-­
so only by causing a large increase in the fission rate. proliferating basal cells have more opportunities to
replace neighbouring cells. Similarly, in glands, upregu­
Squamous epithelium. Epithelial cells within squa- lation of crypt fission (for example, with clonal expan-
mous tissue, such as epidermis, cervical, squamous oral sion) and increased ability to survive e­ nvironmental
and oesophageal squamous mucosa, expand by basal insult are traits expected to be under selection.
replacement of neighbouring stem cells12,27,68,69. During In the noninflamed colon, KRAS mutation accel-
normal homeostasis, basal cells proliferate, producing erates the rate of crypt fission and is therefore selected
differentiated progeny that go on to form the superfi- for 75. The cycles of wounding and repair that occur in
cial layers of the epithelium and also new basal cells the colonic mucosa of individuals with IBD are expected
that compete neutrally to grow a patch by the lateral to provide a strong selective pressure, first for clones that
replacement of one progenitor cell by another 70 (FIG. 3). can survive the damage and then for fast-proliferating
Tumorigenic mutations, such as in TP53 and Notch clones that can rapidly heal the wound76. This selection
family genes, tilt the competition in favour of the within the cancerized field provides a ready explanation
­expansion of mutant clones12,27 (detailed above). of the genetic differences between sporadic and colitis-­
Localized clonal expansion is the canonical mech­ associated CRC53. BE cells produce mucins that provide
anism of mutant field formation, but there may also be protection against insult from gastric acid reflux; hence,
noncanonical methods. Long-range stem cell replace- BE may be an adaptive response that is selected for in
ment occurs in the Drosophila ovaries71, and similar response to reflux 77. Indeed, mathematical modelling
within-organ metastasis could occur in the human body: of oesophageal adenocarcinoma progression suggests
distant engraftment of genetically labelled organoids that age-dependent GERD symptoms affect not only BE
in the intestine provides a proof‑of‑concept of such a onset but also proliferation rates of high-grade dysplasia
mechanism72. High-resolution lineage tracing studies73,74 due to acid-induced injury 78. Together, these examples
where there is ongoing frequent genetic labelling that show how field cancerization represents an evolution-
provides finer resolution of individual somatic lineages ary trade-off between wound repair and cancer devel-
are required to reveal the frequency of long-range stem opment: faster-healing wounds may come at the cost of
cell migration that is possible in both glandular tissue field cancerization.
and squamous tissue. The mutation burden of apparently healthy tissues
increases with age10 — and consequently, aged epithelia
are a patchwork of different clones. Clonal competition,
or other interactions between these clones, may have an
◀ Figure 2 | Evolutionary trajectories of cancerized clones. a | Common evolutionary
important influence on progression to cancer 79. Clonal
pathways of pretumour progression in Barrett oesophagus (BE)37. The genotype–
phenotype map guides the evolutionary path of cells in the two abstract infinite spaces competition in general slows adaptation, so it may have
of the genotype and phenotype. Progression from normal to a field-cancerized state a cancer-suppressive effect 64,80.
and eventually to cancer is underpinned by changes in genotype; these changes are
due to random somatic mutation (black filled arrows to pink points) that is then Role of the tissue microenvironment
projected by the genotype–phenotype map, which incorporates cells’ genetic Stromal reprogramming is increasingly recognized to
mutations, epimutations, and microenvironmental context to produce a specified play a critical role in tumorigenesis81. In general, the stro-
phenotype (dashed lines to orange points). Natural selection acts in the phenotype mal microenvironment is a key regulator of self-­renewal
space to shift the predominant phenotype of the clone population to one of higher in the epithelium82, and so aberrant changes in the stroma
(contextual) fitness (dashed lines with unfilled arrowheads to purple points). Pink ovals
can enable field cancerization of the epithelium.
represent regions of similar phenotype (N, normal tissue; M, metaplasia; H, hyperplasia;
D, dysplasia; C, cancer). The genotype of fit parents is deterministically preserved back Aberrant stroma (measured by fibroblast phenotypes
in genotype space (dashed lines to blue points). The diagram highlights the overlap in and immune cell population composition) is indeed
defined phenotypic regions — for example, some genotype–phenotype combinations found in pre-malignant disease. For example, patients
could be histologically classified as either metaplastic or dysplastic. b | Punctuated with IBD are at higher risk of CRC, and the severity of
evolution. Here, in contrast to the gradual phenotypic evolution of part a, the inflammation is an independent predictor of cancer
ostensibly normal clone carries a TP53 mutation, which then undergoes risk83. In BE, the stromal compartment shows consist-
whole-genome doubling (WGD) and causes a large change in both genotype and ent changes in gene expression that also have prognostic
phenotype, as has been seen in BE. c | Small changes in genotype space may also lead value, and moreover, these changes are somewhat similar
to large changes in phenotype space, possibly owing to rare variants having large among pre-malignant conditions along the gastrointesti-
phenotypic effects, epistatic effects between accumulated mutations and/or other
nal tract 84. In skin, loss of Notch family signalling in the
epigenetic or microenvironmental effects. For example, a TP53 inactivating mutation
may occur in a morphologically normal clone but not be selected for until subsequent mesenchymal cells of transgenic mouse models induces
small events accumulate to promote large phenotypic changes. In summary, the epithelial tumorigenesis, demonstrating how stromal
degree of phenotypic change need not be proportional to the degree of underlying changes enable field cancerization22. In the breast, trans-
genetic change. CDKN2A, cyclin-dependent kinase inhibitor 2A; SMAD4, mothers genic manipulation and cancer-associated fibroblasts
against decapentaplegic homologue 4. have revealed a critical role for the stromal lineage in

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 7


©
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

Stem cell
Somatic alteration Wild-type cell Mutated cell

b c d

Progeny cells Basal cell Mutated cell

Figure 3 | Morphological mechanisms and examples of clonal expansion. a | Stem cell niche succession and fission in
glandular tissue. A single stem cell in the niche acquires a somatic alteration (blue) and may take Nature
over theReviews
entire crypt
| Cancer
population (‘niche succession’) either via neutral genetic drift or via selection, by which the cell with an advantageous
mutation outcompetes the other cell types (brown) to be the source for all future somatic cells in the crypt. The
monoclonally converted crypt then bifurcates, beginning at the niche compartment and producing two mutated crypts.
b | Expansion of a clone in the human colon. A histological en face slide depicts an entirely cytochrome c oxidase
(COX)-deficient crypt (blue) undergoing fission in the colon of a person with familial adenomatous polyposis syndrome.
The two resulting daughter crypts both are COX-deficient and are surrounded by COX-wild-type crypts (brown).
COX-deficiency is a somatically acquired event that usually results from an underlying somatic mutation in the
mitochondrial DNA; consequently, COX-deficiency is a natural heritable mark that facilitates lineage tracing in human
tissues. c | Transverse section from a biopsy sample of a patient with inflammatory bowel disease stained for COX activity
showing two crypts that are part of a single COX-deficient clone. In glandular tissues such as those found in the stomach,
Barrett oesophagus and the colon, clones such as this may continue to grow into a large patch of mutant glands.
d | COX-deficient human epithelial clone expanding in the human airway. Immunofluorescence staining for COX (green)
is shown with counterstains for the mitochondrial marker porin (red) and the nuclear marker 4ʹ,6‑diamidino‑2‑phenylin-
dole (DAPI; blue). COX-deficient cells stain red; COX-proficient cells stain green. A clone of COX-deficient cells has
expanded to form a small mutated patch of cells. The few COX-deficient cells that are separate from the main patch are
likely part of the same clone as the main patch but became separated as the clone grew. e | Expansion of a clone in
squamous epithelium. A single basal cell acquires a somatic alteration (dark red) that confers an advantageous
phenotype, leading the mutant clone to laterally replace neighbouring basal cells (dark green). These basal cells then
produce more superficial progeny cells (lighter red). This process, which occurs in tissues such as the lung, oesophagus
and skin, leads to a patch of clonally derived (and phenotypically altered) cells. Parts b and c are courtesy of Stuart
McDonald, Barts Cancer Institute, Queen Mary University of London, UK. Part d is courtesy of Samuel Janes, University
College London, UK.

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
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

modulating epithelial cell behaviour 85,86. Moreover, for biomarkers that can distinguish cancerized fields that
in the colon, for example, genome-wide association will remain indolent for a long time from those that will
studies for CRC have identified bone morphogenetic rapidly become malignant.
protein (BMP) family members that are regulated and
expressed by stromal cells as key modulators of cancer The pitfalls of pathology. Conventional pathologic grad-
development risk87. ing remains the most common biomarker of cancer risk
Microenvironmental changes enable field can- in conditions associated with field cancerization (for
cerization in the stroma by altering the fitness effects example, IBD and BE) and in morphologically discern-
of m
­ utations in epithelial cells (reviewed in REF.  88) ible pre-malignant disease (for example, DCIS or PIN),
and so promote expansion of cancerized lineages. but in general, the predictive value of pathological sta­
Microenvironmental factors provide selective pressures ging of pre-malignant disease is low. For instance, non­
for phenotype adaptation: within the cancerized field, dysplastic BE has been estimated to incur an ~0.25%
cells explore the adaptive landscape (via genetic muta- annual risk of progression to adenocarcinoma95, whereas
tion or phenotypic plasticity), and the genotypes of high-grade dysplasia carries a near 25‑fold increase in
future generations reflect the phenotype that successfully risk at ~6% annual risk of progression96; however, this still
surmounted any microenvironmental challenges (FIG. 2). means that more than nine out of ten patients with BE
From this theoretical ‘fitness landscapes’ perspective, who are diagnosed with a dangerous high-grade lesion
Gatenby and Gillies proposed a model that identified six will not progress to cancer in the next year. The annual
such micro­environmental barriers for the development of risk of squamous carcinoma of the skin in patients with
a malignant phenotype: apoptosis with loss of basement actinic keratosis ranges between 0.15 and 80%97, and pub-
membrane contact, ­inadequate growth promotion, senes- lished estimates of annual cancer incidence in patients
cence, hypoxia, acid­osis and ischaemia 89. Growth, with BE who have low-grade dysplasia range between
senescence and cell death are clearly also influenced by 0.15 and 36%98. This tremendous variability is due to a
cell-intrinsic factors (for example, oncogenic mutations) multitude of factors, including interobserver variability
but, nevertheless, can also be regulated by intercellular in grading 99, sampling bias (higher-grade lesions may be
signalling (that is, by the microenvironment)90. It is not missed at biopsy 100) and patient cohort differences97,98.
simply serendipitous that a cancerized clone is found in a Moreover, given that field cancerization can occur with-
particular ecosystem — clonal adaptation can occur only out associated morphological change, evidence of evolu-
owing to the current selective microenvironment. tion along a trajectory towards cancer cannot reliably be
assessed by pathology alone.
Clinical implications
The key clinical issue presented by field cancerization is Molecular biomarkers. Molecular profiling for sensitive
that of accurately determining the risk of cancer devel- detection of cancerized fields at high risk of developing
opment from a cancerized lesion, as putatively cancer- malignancy is highly desirable (BOX 2). As for molecular
ized fields are very common but few actually progress prognostication of established cancer, the ideal is to find
to cancer. Thus, simply assuming that the detection of a particular (small and easily assayable) set of molecular
a cancerized field is evidence of likely cancer develop- features (mutation status, RNA expression and/or pro-
ment carries the hazard of overdiagnosis of cancer risk, tein level) that predict those patients at risk of progres-
and moreover, subsequent interventions to ameliorate the sion with high sensitivity and specificity. For example,
risk in most cases constitute overtreatment (with associ- a number of different molecular panels show predictive
ated risks of unnecessary morbidities)91. Take the blood, value for oesophageal adenocarcinoma in patients with
for example: 1% of all newborn babies have cells with an BE101–103 and for lung cancer in current or former smok-
acute lymphoblastic leukaemia-associated mutation and ers104,105. In the lung, gene expression within the prox-
histopathologically identifiable precursor lesions that are imal airway indicates distal cancer, perhaps as a direct
present at ~100 times the corresponding rates of clinical consequence of a cancerized airway 105. Unfortunately, the
cancers92, and moreover, such early genetic events can be studies using such panels typically find that the nega­tive
shared between monozygotic twins, indicating that the predictive value is low; the panels are only moderately
pre-malignant clone formed in utero93. However, bone successful at predicting which patients will not get can-
marrow transplant to remove the cancerized field would cer in the future. While increasing the size of the bio-
seem obscene, given that the intervention is unneces­ marker panels may increase their predictive accuracy,
sary in 99% of patients. In adults, 10% of unselected panel size alone will not address the complicating factors
individuals over the age of 65 show clonal expansion of of inter­patient heterogeneity (not all patients will follow
cancer-associated mutations in their blood94. This appar- the same molecular path to cancer, so a rare dangerous
ent field cancerization in the blood increases cancer risk path may not be assayed by the biomarker panel) and
13‑fold94, in this case indicating that detection of a can- intra­patient heterogeneity (whereby a biopsy may fail to
cerized field is a biomarker of cancer risk. Nevertheless, sample a dangerous lesion).
the absolute risk of haematological cancer development Instead, we emphasize that biomarkers based on
in those patients with clonal haematopoiesis was still quantification of the underlying evolutionary process
found to be very small (~1% annual incidence rate)94, so occurring within the cancerized field (rather than on
detection of the field itself will overdiagnose cancer risk measurement of a molecular feature that may or may
in the majority of people. Clearly, there is an acute need not be an outcome of that process) may have pan-tissue

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 9


©
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

Box 2 | Deriving biomarkers for cancer risk prediction


Traditionally, tissue morphology has been used as the sole indicator that a patient has a certain risk of developing a
malignancy. More recently, the presence or absence of particular molecular features has been used as a biomarker.
However, measures of the evolution of the cancerized field itself, such as genetic diversity107, may also act as biomarkers
for the risk of cancer development. Measuring the field as a whole removes the risk of sampling bias. Serial sampling and
spatially extensive sampling in longitudinal analyses38,109 will continue to provide better insight into the pace of evolution
and thus aid prognostication.
Below, we provide a table of cancer biomarker types alongside a few of their benefits, pitfalls and examples.

Biomarker Benefits Pitfalls Examples


Tissue Standardized histopathology Grey areas of pathological staging; Metaplasia, dysplasia,
morphology methods; large studies completed interobserver variability; missed carcinoma in situ
for validation lesions on exam
Molecular Some genetic defects have higher Single time-point data may not DNA mutations, gene
marker predictive value than morphology reveal event ordering; marker expression patterns,
(single or a (for example, TP53 biallelic sometimes missed in biopsies; protein levels, viral
panel) inactivation in Barrett oesophagus); panels do not typically acknowledge load
ever-decreasing costs marker–marker interactions
Evolutionary Markers of heterogeneity more Some markers require serial Genetic diversity,
measure ubiquitous than single genetic sampling to accurately validate rates clonal expansions,
changes; measures evolutionary of change; limited studies available change over time,
trajectory itself at this time biological tissue age

prognostic value that is robust to the problem of inter- cancer risk — is also invariant over time109. Refinement
patient and intrapatient molecular heterogeneity 106. of screening schedules and associated prognostic bio-
One such ‘evolutionary biomarker’ is the degree of markers requires longitudinal studies of the evolution
genetic diversity within the cancerized field. Genetic within cancerized fields. Mathematical modelling of the
diversity is a proxy measure of field evolvability, as a underlying disease aetiology may further help to opti-
more diverse field is more likely to contain a lineage mize the precise timing of clinical screens to maximize
that is preadapted to a new selective pressure107. Indeed, the probability that an individual is screened within a
genetic diversity has been shown to be prognostic in window of opportunity for timely intervention; here,
two different cohorts of patients with BE, even when evolutionary-minded approaches that explicitly model
using different molecular tools to quantify the d ­ iversity clonal expansions show particular promise114–119.
present 38,108,109. The potential utility of clonal diversity as
a prognostic measure across disease types was under- Chemoprevention. Part of the mechanism of action
lined by a recent pan-cancer study 110. Similarly, genetic of chemopreventive therapies is likely to be through
instability, as another measure of evolvability, has prog- their modulation of pretumour field dynamics.
nostic value in BE111 and potentially also in colitis112. Though direct evidence of this is generally lacking
A second evolutionary measure is the size of clonal — owing to the difficulty inherent in studying pre­
expansions, as this provides both a proxy assay for the tumour clones — studies in BE provide a rare but ele-
size of the cancerized field and evidence for positive gant example. Therein, the cancer-reducing effects of
selection of clones within the field. Detection of clonal non­steroidal anti-inflammatory drugs (NSAIDs) 120
expansions has prognostic value in ulcerative colitis113 may occur because they can reduce the mutation rate
and BE111 and in the blood94. Similarly, changes in lesion and change the microenvironment such that certain
composition over time could have prognostic value as clones that are already established in the tissue will no
a marker of active evolution of the field; quantification longer be selected for 121. Similarly, we hypothesize that
of the mutation rate (a proxy measure of the rate at ­5‑aminosalicylate anti-inflammatory drugs reduce the
which new adaptive lineages are produced) and rates risk of CRC in IBD122 through similar mechanisms,
of clonal expansion (how quickly a new high-­cancer- and more generally, the chemopreventive effects of
risk clone grows to a large size) may hold particular aspirin123 are likely to function through the modulation
prognostic value. of pre­tumour field evolution. Proton-pump inhibitors
used by patients with BE may also influence the for-
Optimizing screening schedules. The intervals between mation of mutant clones, although the relationship, if
cancer screens may be optimized using knowledge of the any, to ­cancer progression remains unclear 124. Further,
biology of cancerized fields. If a field is rapidly evolv- the cancer-­preventive effects of Bacillus Calmette–
ing, then frequent screening may be required, whereas Guérin therapy for patients with superficial bladder
evolutionarily quiescent fields could be surveyed less cancer 125–127 may occur because it modulates clone–­
often. For instance, in BE, longitudinal measurement of microenvironment inter­a ctions 128. Understanding
the clonal composition has revealed that little evolution the mechanisms by which field cancerization driv-
occurs over time, and consequently, the level of genetic ers are selected for is a promising route towards the
diversity within the BE segment — as an indicator of ­development of new chemopreventive drugs.

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
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

Metachronous tumours Surgical interventions. The prospect of field canceriza- the increased risk of rapid recurrence from a cancerized
Primary tumours arising tion within a patient undergoing cancer resection has field left in situ. The development of an imaging modality
sequentially in time. implications for deciding surgical margins — should an to visualize the cancerized field during surgery has the
entire cancerized field be removed along with the overt potential to be revolutionary.
lesion? If field cancerization is commonly found in a cer-
tain disease, it is logical to think that patients will be at Summary and conclusions
risk of metachronous tumours if the cancerized field is not The balance of evidence suggests that some tissues in our
removed along with the cancer itself 32,114,129–131. In colitis, bodies inevitably, and unfortunately, become cancerized
there has been a recent tendency for endoscopic (limited) as we age: mutations accrue throughout life, and some
resection of dysplastic pretumour lesions, which may will be unlucky mutations that drive the formation and
underpin a slight increase in cancer incidence rates100,132. phenotypic evolution of a field of mutant cells that is
As seen in the treatment of DCIS with breast-conserving ‘one step closer’ to malignancy. Understanding the evo-
surgery rather than mastectomy 133, choosing optimal lutionary dynamics of cells within these cancerized fields
resection margins requires balancing the competing risks is key to identifying and then controlling those tissues
of morbidities associated with more radical surgery with that have aged ungracefully towards cancer.

1. Braakhuis, B. J., Tabor, M. P., Kummer, J. A., epithelium; clinical implications of multicentric origin. 32. Galandiuk, S. et al. Field cancerization in the intestinal
Leemans, C. R. & Brakenhoff, R. H. A genetic Cancer 6, 963–968 (1953). epithelium of patients with Crohn’s ileocolitis.
explanation of Slaughter’s concept of field This is the original paper proposing the concept of Gastroenterology 142, 855–864.e8 (2012).
cancerization: evidence and clinical implications. field cancerization. This study identifies examples of field cancerization
Cancer Res. 63, 1727–1730 (2003). 17. Franklin, W. A. et al. Widely dispersed p53 mutation by mutant clones in human Crohn’s disease.
This study presents the updated concept of in respiratory epithelium. A novel mechanism for field 33. Leedham, S. J. et al. Clonality, founder mutations,
field cancerization, which includes a genetic carcinogenesis. J. Clin. Invest. 100, 2133–2137 and field cancerization in human ulcerative colitis-
perspective. (1997). associated neoplasia. Gastroenterology 136,
2. Garcia, S. B., Park, H. S., Novelli, M. & Wright, N. A. This study shows field cancerization in the human 542–550.e6 (2009).
Field cancerization, clonality, and epithelial stem cells: lung by interlobe clonal expansion of a TP53‑mutant This study identifies examples of field cancerization
the spread of mutated clones in epithelial sheets. clone. at individual crypt resolution in human colitis.
J. Pathol. 187, 61–81 (1999). 18. Ushijima, T. & Hattori, N. Molecular pathways: 34. Lawrence, M. S. et al. Discovery and saturation
3. Kaz, A. M., Grady, W. M., Stachler, M. D. & Bass, A. J. involvement of Helicobacter pylori-triggered analysis of cancer genes across 21 tumour types.
Genetic and epigenetic alterations in Barrett’s inflammation in the formation of an epigenetic field Nature 505, 495–501 (2014).
esophagus and esophageal adenocarcinoma. defect, and its usefulness as cancer risk and exposure 35. Kato, S., Lippman, S. M., Flaherty, K. T. & Kurzrock, R.
Gastroenterol. Clin. North Am. 44, 473–489 markers. Clin. Cancer Res. 18, 923–929 (2012). The conundrum of genetic “drivers” in benign
(2015). This study describes epigenetic fields as a cause conditions. J. Natl Cancer Inst. 108, djw036
4. Virnig, B. A., Tuttle, T. M., Shamliyan, T. & Kane, R. L. of field cancerization in the stomach. (2016).
Ductal carcinoma in situ of the breast: a systematic 19. Nosho, K. et al. A prospective cohort study shows 36. Bieging, K. T., Mello, S. S. & Attardi, L. D. Unravelling
review of incidence, treatment, and outcomes. unique epigenetic, genetic, and prognostic features mechanisms of p53‑mediated tumour suppression.
J. Natl Cancer Inst. 102, 170–178 (2010). of synchronous colorectal cancers. Gastroenterology Nat. Rev. Cancer 14, 359–370 (2014).
5. Bostwick, D. G. & Cheng, L. Precursors of prostate 137, 1609–1620.e3 (2009). 37. Weaver, J. M. et al. Ordering of mutations in
cancer. Histopathology 60, 4–27 (2012). 20. Simons, B. D. & Clevers, H. Strategies for homeostatic preinvasive disease stages of esophageal
6. Prasad, G. A. et al. Long-term survival following stem cell self-renewal in adult tissues. Cell 145, carcinogenesis. Nat. Genet. 46, 837–843 (2014).
endoscopic and surgical treatment of high-grade 851–862 (2011). This study shows the general lack of
dysplasia in Barrett’s esophagus. Gastroenterology 21. Lochhead, P. et al. Etiologic field effect: reappraisal stage-specificity of genetic mutations in BE.
132, 1226–1233 (2007). of the field effect concept in cancer predisposition 38. Li, X. et al. Temporal and spatial evolution of somatic
7. Nordenvall, C., Ekbom, A., Bottai, M., Smedby, K. E. and progression. Mod. Pathol. 28, 14–29 (2015). chromosomal alterations: a case-cohort study of
& Nilsson, P. J. Mortality after total colectomy in 22. Hu, B. et al. Multifocal epithelial tumors and field Barrett’s esophagus. Cancer Prev. Res. 7, 114–127
3084 patients with inflammatory bowel disease: cancerization from loss of mesenchymal CSL signaling. (2014).
a population-based cohort study. Aliment. Pharmacol. Cell 149, 1207–1220 (2012). 39. Stachler, M. D. et al. Paired exome analysis of
Ther. 40, 280–287 (2014). This study demonstrates that field cancerization Barrett’s esophagus and adenocarcinoma. Nat. Genet.
8. Laine, L. et al. SCENIC international consensus can be initiated by stromal cell changes rather than 47, 1047–1055 (2015).
statement on surveillance and management of by epithelial cell changes. This study illustrates the multiple genetic
dysplasia in inflammatory bowel disease. 23. Calabrese, P., Tavare, S. & Shibata, D. Pretumor pathways, including a punctuated genome-doubling
Gastrointest. Endosc. 81, 489–501.e26 (2015). progression: clonal evolution of human stem cell pathway, that lead from BE to oesophageal cancer.
9. Shaheen, N. J., Falk, G. W., Iyer, P. G. & Gerson, L. B. populations. Am. J. Pathol. 164, 1337–1346 (2004). 40. Curvers, W. L. et al. Low-grade dysplasia in Barrett’s
ACG Clinical Guideline: Diagnosis and management 24. Greaves, M. Evolutionary determinants of cancer. esophagus: overdiagnosed and underestimated.
of Barrett’s esophagus. Am. J. Gastroenterol. 111, Cancer Discov. 5, 806–820 (2015). Am. J. Gastroenterol. 105, 1523–1530 (2010).
30–50 (2016). 25. Stratton, M. R., Campbell, P. J. & Futreal, P. A. 41. Kerkhof, M. et al. Grading of dysplasia in Barrett’s
10. Blokzijl, F. et al. Tissue-specific mutation accumulation The cancer genome. Nature 458, 719–724 (2009). oesophagus: substantial interobserver variation
in human adult stem cells during life. Nature 538, 26. Martincorena, I. et al. Tumor evolution. High burden between general and gastrointestinal pathologists.
260–264 (2016). and pervasive positive selection of somatic mutations Histopathology 50, 920–927 (2007).
This study includes a genome-wide measurement in normal human skin. Science 348, 880–886 (2015). 42. Tomasetti, C., Li, L. & Vogelstein, B. Stem cell
of the accumulation of age-associated mutations This study identifies an extensive amount of divisions, somatic mutations, cancer etiology, and
in the intestine and liver. cancer-associated mutations in morphologically cancer prevention. Science 355, 1330–1334
11. Blanpain, C. & Simons, B. D. Unravelling stem cell normal human skin. (2017).
dynamics by lineage tracing. Nat. Rev. Mol. Cell Biol. 27. Alcolea, M. P. et al. Differentiation imbalance in This study suggests a critical role for pretumour
14, 489–502 (2013). single oesophageal progenitor cells causes clonal mutation accumulation in cancer development.
12. Klein, A. M., Brash, D. E., Jones, P. H. & Simons, B. D. immortalization and field change. Nat. Cell Biol. 16, 43. Alexandrov, L. B. et al. Clock-like mutational processes
Stochastic fate of p53‑mutant epidermal progenitor 615–622 (2014). in human somatic cells. Nat. Genet. 47, 1402–1407
cells is tilted toward proliferation by UV B during 28. Jonason, A. S. et al. Frequent clones of p53‑mutated (2015).
preneoplasia. Proc. Natl Acad. Sci. USA 107, keratinocytes in normal human skin. Proc. Natl Acad. This study identifies mutational processes that
270–275 (2010). Sci. USA 93, 14025–14029 (1996). underlie age-associated mutation accumulation.
13. Baker, A. M. et al. Quantification of crypt and stem 29. Sanchez-Danes, A. et al. Defining the clonal dynamics 44. Driessens, G., Beck, B., Caauwe, A., Simons, B. D. &
cell evolution in the normal and neoplastic human leading to mouse skin tumour initiation. Nature 536, Blanpain, C. Defining the mode of tumour growth by
colon. Cell Rep. 8, 940–947 (2014). 298–303 (2016). clonal analysis. Nature 488, 527–530 (2012).
14. Kim, K. M., Calabrese, P., Tavare, S. & Shibata, D. 30. Bonilla, X. et al. Genomic analysis identifies new 45. de Bruin, E. C. et al. Spatial and temporal diversity in
Enhanced stem cell survival in familial adenomatous drivers and progression pathways in skin basal cell genomic instability processes defines lung cancer
polyposis. Am. J. Pathol. 164, 1369–1377(2004). carcinoma. Nat. Genet. 48, 398–406 (2016). evolution. Science 346, 251–256 (2014).
15. Vortmeyer, A. O. & Alomari, A. K. Pathology of the 31. Vermeulen, L. et al. Defining stem cell dynamics in 46. Steiling, K., Ryan, J., Brody, J. S. & Spira, A. The field
nervous system in Von Hippel-Lindau disease. models of intestinal tumor initiation. Science 342, of tissue injury in the lung and airway. Cancer Prev.
J. Kidney Cancer VHL 2, 114–129 (2015). 995–998 (2013). Res. 1, 396–403 (2008).
16. Slaughter, D. P., Southwick, H. W. & Smejkal, W. This study quantifies the selective advantage of 47. Burd, E. M. Human papillomavirus and cervical
Field cancerization in oral stratified squamous pretumour mutations in the intestine. cancer. Clin. Microbiol. Rev. 16, 1–17 (2003).

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 11


©
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

48. Yakirevich, E. & Resnick, M. B. Pathology of gastric 73. Frieda, K. L. et al. Synthetic recording and in situ 97. Greaves, M. Does everyone develop covert cancer?
cancer and its precursor lesions. Gastroenterol. Clin. readout of lineage information in single cells. Nature Nat. Rev. Cancer 14, 209–210 (2014).
North Am. 42, 261–284 (2013). 541, 107–111 (2017). This commentary elegantly discusses the inevitably
49. Wang, K. et al. Whole-genome sequencing and 74. McKenna, A. et al. Whole-organism lineage tracing by of cancerization.
comprehensive molecular profiling identify new driver combinatorial and cumulative genome editing. Science 98. Runge, T. M., Abrams, J. A. & Shaheen, N. J.
mutations in gastric cancer. Nat. Genet. 46, 573–582 353, aaf7907 (2016). Epidemiology of Barrett’s esophagus and esophageal
(2014). 75. Snippert, H. J., Schepers, A. G., van Es, J. H., adenocarcinoma. Gastroenterol. Clin. North Am. 44,
50. Rubenstein, J. H. & Taylor, J. B. Meta-analysis: the Simons, B. D. & Clevers, H. Biased competition 203–231 (2015).
association of oesophageal adenocarcinoma with between Lgr5 intestinal stem cells driven by oncogenic 99. Odze, R. D. Diagnosis and grading of dysplasia in
symptoms of gastro-oesophageal reflux. mutation induces clonal expansion. EMBO Rep. 15, Barrett’s oesophagus. J. Clin. Pathol. 59, 1029–1038
Aliment. Pharmacol. Ther. 32, 1222–1227 (2010). 62–69 (2014). (2006).
51. Dulak, A. M. et al. Exome and whole-genome 76. Choi, C. R., Bakir, I. A., Hart, A. L. & Graham, T. A. 100. Choi, C. H. et al. Forty-year analysis of colonoscopic
sequencing of esophageal adenocarcinoma identifies Clonal evolution of colorectal cancer in IBD. Nat. Rev. surveillance for neoplasia in ulcerative colitis: an
recurrent driver events and mutational complexity. Gastroenterol. Hepatol. 14, 218–229 (2017). updated overview. Am. J. Gastroenterol. 110,
Nat. Genet. 45, 478–486 (2013). 77. Reid, B. J., Li, X., Galipeau, P. C. & Vaughan, T. L. 1022–1034 (2015).
52. Tomasetti, C., Vogelstein, B. & Parmigiani, G. Half or Barrett’s oesophagus and oesophageal 101. Bird-Lieberman, E. L. et al. Population-based study
more of the somatic mutations in cancers of self- adenocarcinoma: time for a new synthesis. Nat. Rev. reveals new risk-stratification biomarker panel for
renewing tissues originate prior to tumor initiation. Cancer 10, 87–101 (2010). Barrett’s esophagus. Gastroenterology 143,
Proc. Natl Acad. Sci. USA 110, 1999–2004 (2013). 78. Hazelton, W. D. et al. The role of gastroesophageal 927–935.e3 (2012).
This study suggests that most somatic mutations reflux and other factors during progression to 102. Reid, B. J. et al. Predictors of progression in Barrett’s
in cancer accrue prior to the initiation of cancer esophageal adenocarcinoma. Cancer Epidemiol. esophagus II: baseline 17p (p53) loss of heterozygosity
growth. Biomarkers Prev. 24, 1012–1023 (2015). identifies a patient subset at increased risk for
53. Robles, A. I. et al. Whole-exome sequencing analyses 79. Korolev, K. S., Xavier, J. B. & Gore, J. Turning ecology neoplastic progression. Am. J. Gastroenterol. 96,
of inflammatory bowel disease-associated colorectal and evolution against cancer. Nat. Rev. Cancer 14, 2839–2848 (2001).
cancers. Gastroenterology 150, 931–943 (2016). 371–380 (2014). 103. Timmer, M. R. et al. Derivation of genetic biomarkers
54. Issa, J. P., Ahuja, N., Toyota, M., Bronner, M. P. & 80. Martincorena, I., Jones, P. H. & Campbell, P. J. for cancer risk stratification in Barrett’s oesophagus:
Brentnall, T. A. Accelerated age-related CpG island Constrained positive selection on cancer mutations a prospective cohort study. Gut 65, 1602–1610
methylation in ulcerative colitis. Cancer Res. 61, in normal skin. Proc. Natl Acad. Sci. USA 113, (2016).
3573–3577 (2001). E1128–E1129 (2016). 104. Silvestri, G. A. et al. A bronchial genomic classifier
55. Risques, R. A. et al. Ulcerative colitis is a disease of 81. Mantovani, A., Allavena, P., Sica, A. & Balkwill, F. for the diagnostic evaluation of lung cancer.
accelerated colon aging: evidence from telomere Cancer-related inflammation. Nature 454, 436–444 N. Engl. J. Med. 373, 243–251 (2015).
attrition and DNA damage. Gastroenterology 135, (2008). 105. Spira, A. et al. Airway epithelial gene expression in the
410–418 (2008). 82. Davis, H. et al. Aberrant epithelial GREM1 diagnostic evaluation of smokers with suspect lung
56. Curtius, K. et al. A molecular clock infers heterogeneous expression initiates colonic tumorigenesis from cells cancer. Nat. Med. 13, 361–366 (2007).
tissue age among patients with Barrett’s esophagus. outside the stem cell niche. Nat. Med. 21, 62–70 106. Dhawan, A., Graham, T. A. & Fletcher, A. G. A.
PLoS Comput. Biol. 12, e1004919 (2016). (2015). Computational modeling approach for deriving
57. Campisi, J. Aging, cellular senescence, and cancer. 83. Rutter, M. et al. Severity of inflammation is a risk factor biomarkers to predict cancer risk in premalignant
Annu. Rev. Physiol. 75, 685–705 (2013). for colorectal neoplasia in ulcerative colitis. disease. Cancer Prev. Res. 9, 283–295 (2016).
58. Thirlwell, C. et al. Clonality assessment and clonal Gastroenterology 126, 451–459 (2004). 107. Greaves, M. & Maley, C. C. Clonal evolution in cancer.
ordering of individual neoplastic crypts shows 84. Saadi, A. et al. Stromal genes discriminate preinvasive Nature 481, 306–313 (2012).
polyclonality of colorectal adenomas. Gastroenterology from invasive disease, predict outcome, and highlight 108. Maley, C. C. et al. Genetic clonal diversity predicts
138, 1441–1454 (2010). inflammatory pathways in digestive cancers. progression to esophageal adenocarcinoma.
59. Lin, J. et al. Polyclonality of BRAF mutations in acquired Proc. Natl Acad. Sci. USA 107, 2177–2182 (2010). Nat. Genet. 38, 468–473 (2006).
melanocytic nevi. J. Natl Cancer Inst. 101, 1423–1427 This study shows consistent changes in the gene This study shows how measurement of the
(2009). expression profile of cells within the stromal cancerized field — in this case, the evolvability as
60. Halberg, R. B. & Dove, W. F. Polyclonal tumors in compartment across pre-malignant gastrointestinal measured by genetic diversity — predicts future
the mammalian intestine: are interactions among diseases. cancer risk.
multiple initiated clones necessary for tumor initiation, 85. Bronisz, A. et al. Reprogramming of the tumour 109. Martinez, P. et al. Dynamic clonal equilibrium and
growth, and progression? Cell Cycle 6, 44–51 (2007). microenvironment by stromal PTEN-regulated predetermined cancer risk in Barrett’s oesophagus.
61. Bjerknes, M. & Cheng, H. Colossal crypts bordering miR‑320. Nat. Cell Biol. 14, 159–167 (2011). Nat. Commun. 7, 12158 (2016).
colon adenomas in ApcMin mice express full-length Apc. 86. Quail, D. F. & Joyce, J. A. Microenvironmental 110. Andor, N. et al. Pan-cancer analysis of the extent and
Am. J. Pathol. 154, 1831–1834 (1999). regulation of tumor progression and metastasis. consequences of intratumor heterogeneity. Nat. Med.
62. Thliveris, A. T. et al. Polyclonality of familial murine Nat. Med. 19, 1423–1437 (2013). 22, 105–113 (2016).
adenomas: analyses of mouse chimeras with low tumor 87. Tomlinson, I. P. et al. Multiple common susceptibility 111. Maley, C. C. et al. The combination of genetic
multiplicity suggest short-range interactions. Proc. Natl variants near BMP pathway loci GREM1, BMP4, instability and clonal expansion predicts progression
Acad. Sci. USA 102, 6960–6965 (2005). and BMP2 explain part of the missing heritability to esophageal adenocarcinoma. Cancer Res. 64,
63. Fernandez-Sanchez, M. E. et al. Mechanical induction of colorectal cancer. PLoS Genet. 7, e1002105 (2011). 7629–7633 (2004).
of the tumorigenic beta-catenin pathway by tumour 88. Elinav, E. et al. Inflammation-induced cancer: crosstalk 112. Willenbucher, R. F. et al. Genomic instability is an early
growth pressure. Nature 523, 92–95 (2015). between tumours, immune cells and microorganisms. event during the progression pathway of ulcerative-
64. Martens, E. A., Kostadinov, R., Maley, C. C. & Nat. Rev. Cancer 13, 759–771 (2013). colitis-related neoplasia. Am. J. Pathol. 154,
Hallatschek, O. Spatial structure increases the waiting 89. Gatenby, R. A. & Gillies, R. J. A microenvironmental 1825–1830 (1999).
time for cancer. New J. Phys. 13, 115014 (2011). model of carcinogenesis. Nat. Rev. Cancer 8, 56–61 113. Salk, J. J. et al. Clonal expansions in ulcerative colitis
65. Greaves, L. C. et al. Mitochondrial DNA mutations are (2008). identify patients with neoplasia. Proc. Natl Acad. Sci.
established in human colonic stem cells, and mutated This perspective proposes critical USA 106, 20871–20876 (2009).
clones expand by crypt fission. Proc. Natl Acad. Sci. microenvironmental barriers that must be overcome This study demonstrates that evidence of recent
USA 103, 714–719 (2006). during tumorigenesis. clonal expansion in patients with colitis indicates
66. McDonald, S. A. et al. Mechanisms of field 90. Perez-Mancera, P. A., Young, A. R. & Narita, M. increased cancer risk.
cancerization in the human stomach: the expansion and Inside and out: the activities of senescence in cancer. 114. Curtius, K., Hazelton, W. D., Jeon, J. &
spread of mutated gastric stem cells. Gastroenterology Nat. Rev. Cancer 14, 547–558 (2014). Luebeck, E. G. A. Multiscale model evaluates screening
134, 500–510 (2008). 91. Welch, H. G. & Black, W. C. Overdiagnosis in cancer. for neoplasia in Barrett’s esophagus. PLoS Comput.
This study provides a genetic demonstration of field J. Natl Cancer Inst. 102, 605–613 (2010). Biol. 11, e1004272 (2015).
cancerization in the human stomach. 92. Mori, H. et al. Chromosome translocations and covert This study demonstrates how computational
67. Nicholson, A. M. et al. Barrett’s metaplasia glands are leukemic clones are generated during normal fetal modelling of field cancerization can help optimize
clonal, contain multiple stem cells and share a common development. Proc. Natl Acad. Sci. USA 99, screening protocols.
squamous progenitor. Gut 61, 1380–1389 (2012). 8242–8247 (2002). 115. de Koning, H. J. et al. Benefits and harms of computed
68. Clayton, E. et al. A single type of progenitor cell 93. Greaves, M. F., Maia, A. T., Wiemels, J. L. & Ford, A. M. tomography lung cancer screening strategies:
maintains normal epidermis. Nature 446, 185–189 Leukemia in twins: lessons in natural history. Blood a comparative modeling study for the U.S. Preventive
(2007). 102, 2321–2333 (2003). Services Task Force. Ann. Intern. Med. 160, 311–320
69. Doupe, D. P. et al. A single progenitor population 94. Genovese, G. et al. Clonal hematopoiesis and blood- (2014).
switches behavior to maintain and repair esophageal cancer risk inferred from blood DNA sequence. N. Engl. 116. Hanin, L. & Pavlova, L. Optimal screening schedules
epithelium. Science 337, 1091–1093 (2012). J. Med. 371, 2477–2487 (2014). for prevention of metastatic cancer. Stat. Med. 32,
70. Teixeira, V. H. et al. Stochastic homeostasis in This study shows how the mutation burden in 206–219 (2013).
human airway epithelium is achieved by neutral ostensibly normal white blood cells confers 117. Jeon, J., Meza, R., Moolgavkar, S. H. & Luebeck, E. G.
competition of basal cell progenitors. eLife 2, e00966 leukaemia risk. Evaluation of screening strategies for pre-malignant
(2013). 95. Hvid-Jensen, F., Pedersen, L., Drewes, A. M., lesions using a biomathematical approach.
71. Nystul, T. & Spradling, A. An epithelial niche in the Sorensen, H. T. & Funch-Jensen, P. Incidence of Math. Biosci. 213, 56–70 (2008).
Drosophila ovary undergoes long-range stem cell adenocarcinoma among patients with Barrett’s 118. Kong, C. Y. et al. Exploring the recent trend in
replacement. Cell Stem Cell 1, 277–285 (2007). esophagus. N. Engl. J. Med. 365, 1375–1383 (2011). esophageal adenocarcinoma incidence and
72. Yui, S. et al. Functional engraftment of colon epithelium 96. Spechler, S. J. Barrett esophagus and risk of mortality using comparative simulation modeling.
expanded in vitro from a single adult Lgr5+ stem cell. esophageal cancer: a clinical review. JAMA 310, Cancer Epidemiol. Biomarkers Prev. 23, 997–1006
Nat. Med. 18, 618–623 (2012). 627–636 (2013). (2014).

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
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

119. Ryser, M. D., Lee, W. T., Ready, N. E., Leder, K. Z. & to the formation of an epigenetic field defect. Oncogene 164. McCaughan, F. et al. Genomic evidence of pre-invasive
Foo, J. Quantifying the dynamics of field cancerization 32, 2140–2149 (2013). clonal expansion, dispersal and progression in
in tobacco-related head and neck cancer: a multiscale 141. Damania, D. et al. Nanocytology of rectal colonocytes bronchial dysplasia. J. Pathol. 224, 153–159 (2011).
modeling approach. Cancer Res. 76, 7078–7088 to assess risk of colon cancer based on field 165. Pipinikas, C. P. et al. Cell migration leads to spatially
(2016). cancerization. Cancer Res. 72, 2720–2727 (2012). distinct but clonally related airway cancer precursors.
120. Galipeau, P. C. et al. NSAIDs modulate CDKN2A, TP53, 142. Hawthorn, L., Lan, L. & Mojica, W. Evidence for field Thorax 69, 548–557 (2014).
and DNA content risk for progression to esophageal effect cancerization in colorectal cancer. Genomics 103, 166. Sozzi, G. et al. Genetic evidence for an independent
adenocarcinoma. PLoS Med. 4, e67 (2007). 211–221 (2014). origin of multiple preneoplastic and neoplastic lung
121. Kostadinov, R. L. et al. NSAIDs modulate clonal 143. Kamiyama, H. et al. DNA demethylation in normal lesions. Cancer Res. 55, 135–140 (1995).
evolution in Barrett’s esophagus. PLoS Genet. 9, colon tissue predicts predisposition to multiple cancers. 167. Gomperts, B. N., Walser, T. C., Spira, A. &
e1003553 (2013). Oncogene 31, 5029–5037 (2012). Dubinett, S. M. Enriching the molecular definition of
This study suggests that NSAIDs reduce cancer risk 144. Kaz, A. M. et al. Patterns of DNA methylation in the the airway “field of cancerization:” establishing new
by inhibiting clonal evolution within a cancerized normal colon vary by anatomical location, gender, and paradigms for the patient at risk for lung cancer.
field in BE. age. Epigenetics 9, 492–502 (2014). Cancer Prev. Res. 6, 4–7 (2013).
122. Zhao, L. N. et al. 5‑Aminosalicylates reduce the risk of 145. Milicic, A. et al. Ectopic expression of P‑cadherin 168. Kadara, H. et al. Transcriptomic architecture of the
colorectal neoplasia in patients with ulcerative colitis: correlates with promoter hypomethylation early in adjacent airway field cancerization in non-small cell lung
an updated meta-analysis. PLoS ONE 9, e94208 colorectal carcinogenesis and enhanced intestinal crypt cancer. J. Natl Cancer Inst. 106, dju004 (2014).
(2014). fission in vivo. Cancer Res. 68, 7760–7768 (2008). 169. Lin, M. W., Wu, C. T., Kuo, S. W., Chang, Y. L. &
123. Rothwell, P. M. et al. Effect of daily aspirin on long-term 146. Shen, L. et al. MGMT promoter methylation and field Yang, P. C. Clinicopathology and genetic profile of
risk of death due to cancer: analysis of individual defect in sporadic colorectal cancer. J. Natl Cancer Inst. synchronous multiple small adenocarcinomas:
patient data from randomised trials. Lancet 377, 97, 1330–1338 (2005). implication for surgical treatment of an uncommon
31–41 (2011). 147. Gutierrez-Gonzalez, L. et al. The clonal origins of lung malignancy. Ann. Surg. Oncol. 21, 2555–2562
124. Hvid-Jensen, F., Pedersen, L., Funch-Jensen, P. & dysplasia from intestinal metaplasia in the human (2014).
Drewes, A. M. Proton pump inhibitor use may not stomach. Gastroenterology 140, 1251–1260.e6 170. Nakachi, I. et al. Application of SNP microarrays to the
prevent high-grade dysplasia and oesophageal (2011). genome-wide analysis of chromosomal instability in
adenocarcinoma in Barrett’s oesophagus: a nationwide 148. Kang, G. H. et al. Genetic evidence for the multicentric premalignant airway lesions. Cancer Prev. Res. 7,
study of 9883 patients. Aliment. Pharmacol. Ther. 39, origin of synchronous multiple gastric carcinoma. 255–265 (2014).
984–991 (2014). Lab. Invest. 76, 407–417 (1997). 171. Weichert, W. & Warth, A. Early lung cancer with lepidic
125. Gee, J., Sabichi, A. L. & Grossman, H. B. 149. Takeshima, H. et al. Frequent involvement of chromatin pattern: adenocarcinoma in situ, minimally invasive
Chemoprevention of superficial bladder cancer. remodeler alterations in gastric field cancerization. adenocarcinoma, and lepidic predominant
Crit. Rev. Oncol. Hematol. 43, 277–286 (2002). Cancer Lett. 357, 328–338 (2015). adenocarcinoma. Curr. Opin. Pulm. Med. 20,
126. Herr, H. W. et al. Intravesical bacillus Calmette-Guerin 150. Yamanoi, K. et al. Epigenetic clustering of gastric 309–316 (2014).
therapy prevents tumor progression and death from carcinomas based on DNA methylation profiles at the 172. Wirtschafter, E., Walts, A. E., Liu, S. T. &
superficial bladder cancer: ten-year follow‑up of a precancerous stage: its correlation with tumor Marchevsky, A. M. Diffuse idiopathic pulmonary
prospective randomized trial. J. Clin. Oncol. 13, aggressiveness and patient outcome. Carcinogenesis neuroendocrine cell hyperplasia of the lung
1404–1408 (1995). 36, 509–520 (2015). (DIPNECH): current best evidence. Lung 193,
127. Lamm, D. L. et al. Maintenance Bacillus Calmette- 151. Zaky, A. H. et al. Clinicopathologic implications of 659–667 (2015).
Guerin immunotherapy for recurrent TA, T1 and genetic instability in intestinal-type gastric cancer and 173. Angadi, P. V., Savitha, J. K., Rao, S. S. & Sivaranjini, Y.
carcinoma in situ transitional cell carcinoma of the intestinal metaplasia as a precancerous lesion: proof of Oral field cancerization: current evidence and future
bladder: a randomized Southwest Oncology Group field cancerization in the stomach. Am. J. Clin. Pathol. perspectives. Oral Maxillofac. Surg. 16, 171–180
Study. J. Urol. 163, 1124–1129 (2000). 129, 613–621 (2008). (2012).
128. Redelman-Sidi, G., Glickman, M. S. & Bochner, B. H. 152. Cense, H. A., van Lanschot, J. J., Fockens, P., 174. Boscolo-Rizzo, P. et al. Telomere shortening in mucosa
The mechanism of action of BCG therapy for bladder Obertop, H. & Offerhaus, G. J. A patient with seven surrounding the tumor: biosensor of field cancerization
cancer — a current perspective. Nat. Rev. Urol. 11, primary tumors of the upper aerodigestive tract: the and prognostic marker of mucosal failure in head and
153–162 (2014). process of field cancerization versus distant monoclonal neck squamous cell carcinoma. Oral Oncol. 51,
129. Foo, J., Leder, K. & Ryser, M. D. Multifocality and expansion. Dis. Esophagus 10, 139–142 (1997). 500–507 (2015).
recurrence risk: a quantitative model of field 153. Kammori, M. et al. Squamous cell carcinomas of the 175. Califano, J. et al. Genetic progression model for head
cancerization. J. Theor. Biol. 355, 170–184 esophagus arise from a telomere-shortened epithelial and neck cancer: implications for field cancerization.
(2014). field. Int. J. Mol. Med. 20, 793–799 (2007). Cancer Res. 56, 2488–2492 (1996).
130. Gupta, M. et al. Recurrence of esophageal intestinal 154. Lee, Y. C. et al. Revisit of field cancerization in 176. Narayana, A., Vaughan, A. T., Fisher, S. G. &
metaplasia after endoscopic mucosal resection and squamous cell carcinoma of upper aerodigestive tract: Reddy, S. P. Second primary tumors in laryngeal cancer:
radiofrequency ablation of Barrett’s esophagus: results better risk assessment with epigenetic markers. results of long-term follow‑up. Int. J. Radiat. Oncol.
from a US Multicenter Consortium. Gastroenterology Cancer Prev. Res. 4, 1982–1992 (2011). Biol. Phys. 42, 557–562 (1998).
145, 79–86.e1 (2013). 155. Matsuda, Y. et al. Hypomethylation of Alu repetitive 177. Pentenero, M. et al. Field effect in oral precancer as
131. Rutgeerts, P. et al. Predictability of the postoperative elements in esophageal mucosa, and its potential assessed by DNA flow cytometry and array-CGH.
course of Crohn’s disease. Gastroenterology 99, contribution to the epigenetic field for cancerization. J. Oral Pathol. Med. 41, 119–123 (2012).
956–963 (1990). Cancer Causes Control 23, 865–873 (2012). 178. Shaw, R. J. et al. Molecular staging of surgical margins
132. Choi, C. H. et al. Low-grade dysplasia in ulcerative 156. Oka, D. et al. The presence of aberrant DNA in oral squamous cell carcinoma using promoter
colitis: risk factors for developing high-grade dysplasia methylation in noncancerous esophageal mucosae in methylation of p16INK4A, cytoglobin, E‑cadherin, and
or colorectal cancer. Am. J. Gastroenterol. 110, association with smoking history: a target for risk TMEFF2. Ann. Surg. Oncol. 20, 2796–2802 (2013).
1461–1471 (2015). diagnosis and prevention of esophageal cancers. 179. Van der Vorst, S., Dekairelle, A. F., Weynand, B.,
133. Morrow, M. et al. Society of Surgical Oncology– Cancer 115, 3412–3426 (2009). Hamoir, M. & Gala, J. L. Assessment of p53 functional
American Society for Radiation Oncology–American 157. Roesch-Ely, M. et al. Proteomic analysis of field activity in tumor cells and histologically normal mucosa
Society of Clinical Oncology consensus guideline on cancerization in pharynx and oesophagus: a from patients with head and neck squamous cell
margins for breast-conserving surgery with whole- prospective pilot study. J. Pathol. 221, 462–470 carcinoma. Head Neck 34, 1542–1550 (2012).
breast irradiation in ductal carcinoma in situ. Ann. Surg. (2010). 180. Dworkin, A. M., Huang, T. H. & Toland, A. E. Epigenetic
Oncol. 23, 3801–3810 (2016). 158. Yakoub, D., Keun, H. C., Goldin, R. & Hanna, G. B. alterations in the breast: Implications for breast cancer
134. Okosun, J. et al. Integrated genomic analysis identifies Metabolic profiling detects field effects in nondysplastic detection, prognosis and treatment. Semin. Cancer
recurrent mutations and evolution patterns driving the tissue from esophageal cancer patients. Cancer Res. Biol. 19, 165–171 (2009).
initiation and progression of follicular lymphoma. 70, 9129–9136 (2010). 181. Ellsworth, D. L. et al. Outer breast quadrants
Nat. Genet. 46, 176–181 (2014). 159. Galipeau, P. C., Prevo, L. J., Sanchez, C. A., demonstrate increased levels of genomic instability.
135. Esserman, L. J. et al. Addressing overdiagnosis and Longton, G. M. & Reid, B. J. Clonal expansion and Ann. Surg. Oncol. 11, 861–868 (2004).
overtreatment in cancer: a prescription for change. loss of heterozygosity at chromosomes 9p and 17p 182. Foschini, M. P. et al. Genetic clonal mapping of in situ
Lancet Oncol. 15, e234–e242 (2014). in premalignant esophageal (Barrett’s) tissue. and invasive ductal carcinoma indicates the field
This article highlights the current scale of the J. Natl Cancer Inst. 91, 2087–2095 (1999). cancerization phenomenon in the breast. Hum. Pathol.
overdiagnosis and overtreatment problem in 160. Maley, C. C. et al. Selectively advantageous mutations 44, 1310–1319 (2013).
pre-malignant disease. and hitchhikers in neoplasms: p16 lesions are selected 183. Rivenbark, A. G. & Coleman, W. B. Field cancerization
136. Campbell, J. D. et al. The case for a Pre-Cancer Genome in Barrett’s esophagus. Cancer Res. 64, 3414–3427 in mammary carcinogenesis - Implications for
Atlas (PCGA). Cancer Prev. Res. 9, 119–124 (2016). (2004). prevention and treatment of breast cancer.
137. Koizumi, K. et al. Array-based identification of common 161. van Dekken, H., Vissers, C. J., Tilanus, H. W., Exp. Mol. Pathol. 93, 391–398 (2012).
DNA methylation alterations in ulcerative colitis. Tanke, H. J. & Rosenberg, C. Clonal analysis of a case of 184. Trujillo, K. A. et al. Breast field cancerization: isolation
Int. J. Oncol. 40, 983–994 (2012). multifocal oesophageal (Barrett’s) adenocarcinoma by and comparison of telomerase-expressing cells in tumor
138. van Dekken, H. et al. Genomic analysis of a case of comparative genomic hybridization. J. Pathol. 188, and tumor adjacent, histologically normal breast tissue.
multifocal adenocarcinoma in ulcerative colitis. 263–266 (1999). Mol. Cancer Res. 9, 1209–1221 (2011).
Virchows Arch. 449, 716–721 (2006). 162. Chang, Y. L. et al. Clonality and prognostic implications 185. Haaland, C. M. et al. Differential gene expression in
139. Alonso, S. et al. Methylation of MGMT and ADAMTS14 of p53 and epidermal growth factor receptor somatic tumor adjacent histologically normal prostatic tissue
in normal colon mucosa: biomarkers of a field defect for aberrations in multiple primary lung cancers. indicates field cancerization. Int. J. Oncol. 35,
cancerization preferentially targeting elder African- Clin. Cancer Res. 13, 52–58 (2007). 537–546 (2009).
Americans. Oncotarget 6, 3420–3431 (2015). 163. Kadara, H. & Wistuba, I. I. Field cancerization in 186. Nonn, L., Ananthanarayanan, V. & Gann, P. H. Evidence
140. Asada, K. et al. FHL1 on chromosome X is a single-hit non-small cell lung cancer: implications in disease for field cancerization of the prostate. Prostate 69,
gastrointestinal tumor-suppressor gene and contributes pathogenesis. Proc. Am. Thorac Soc. 9, 38–42 (2012). 1470–1479 (2009).

NATURE REVIEWS | CANCER ADVANCE ONLINE PUBLICATION | 13


©
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

187. Trujillo, K. A., Jones, A. C., Griffith, J. K. & Bisoffi, M. field cancerization. Cancer Res. 55, 3604–3609 Acknowledgements
Markers of field cancerization: proposed clinical (1995). The authors thank D. Brash for helpful conversations during
applications in prostate biopsies. Prostate Cancer 192. Stern, R. S., Bolshakov, S., Nataraj, A. J. & the writing of this manuscript. The authors acknowledge fund-
2012, 302894 (2012). Ananthaswamy, H. N. p53 mutation in nonmelanoma ing from Cancer Research UK (grants A19771 to T.A.G. and
188. Hafner, C., Knuechel, R., Stoehr, R. & Hartmann, A. skin cancers occurring in psoralen ultraviolet a‑treated A21870 to N.A.W.), the Wellcome Trust (202778/Z/16/Z to
Clonality of multifocal urothelial carcinomas: 10 years patients: evidence for heterogeneity and field T.A.G.) and the Barts Charity (472–2300 to K.C. and T.A.G.).
of molecular genetic studies. Int. J. Cancer 101, 1–6 cancerization. J. Invest. Dermatol. 119, 522–526
(2002). (2002). Author contributions
189. Vriesema, J. L., Aben, K. K., Witjes, J. A., 193. Szeimies, R. M. et al. Clinical, histopathological and All authors researched data for the article, made substan­
Kiemeney, L. A. & Schalken, J. A. Superficial and immunohistochemical assessment of human skin field tial contributions to discussion of the content, wrote the
metachronous invasive bladder carcinomas are cancerization before and after photodynamic therapy. article and reviewed and edited the manuscript before
clonally related. Int. J. Cancer 93, 699–702 (2001). Br. J. Dermatol. 167, 150–159 (2012). submission.
190. Wang, Y., Lang, M. R., Pin, C. L. & Izawa, J. I. 194. Vatve, M., Ortonne, J. P., Birch-Machin, M. A. &
Comparison of the clonality of urothelial carcinoma Gupta, G. Management of field change in actinic Competing interests statement
developing in the upper urinary tract and those keratosis. Br. J. Dermatol. 157 (Suppl. 2), 21–24 The authors declare no competing interests.
developing in the bladder. SpringerPlus 2, 412 (2007).
(2013). 195. Shain, A. H. et al. The genetic evolution of melanoma Publisher’s note
191. Kanjilal, S. et al. p53 mutations in nonmelanoma skin from precursor lesions. N. Engl. J. Med. 373, Springer Nature remains neutral with regard to jurisdictional
cancer of the head and neck: molecular evidence for 1926–1936 (2015). claims in published maps and institutional affiliations.

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrc


©
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