You are on page 1of 11

REVIEWS

CANCER ORIGINS

Metaplasia: tissue injury adaptation


and a precursor to the
dysplasia–cancer sequence
Veronique Giroux and Anil K. Rustgi
Abstract | Metaplasia is the replacement of one differentiated somatic cell type with another
differentiated somatic cell type in the same tissue. Typically, metaplasia is triggered by
environmental stimuli, which may act in concert with the deleterious effects of
microorganisms and inflammation. The cell of origin for intestinal metaplasia in the
oesophagus and stomach and for pancreatic acinar–ductal metaplasia has been posited
through genetic mouse models and lineage tracing but has not been identified in other types
of metaplasia, such as squamous metaplasia. A hallmark of metaplasia is a change in cellular
identity, and this process can be regulated by transcription factors that initiate and/or
maintain cellular identity, perhaps in concert with epigenetic reprogramming. Universally,
metaplasia is a precursor to low-grade dysplasia, which can culminate in high-grade dysplasia
and carcinoma. Improved clinical screening for and surveillance of metaplasia might lead to
better prevention or early detection of dysplasia and cancer.

Dysplasia
Metaplasia is the replacement of one differentiated cell which has been studied more than metaplasia in other
A condition in which cells have type with another mature differentiated cell type that tissues. It is estimated that oesophageal intestinal meta-
abnormal cellular architecture, is not normally present in a specific tissue1. It is impor- plasia, termed Barrett oesophagus, will progress to cancer
with nuclear atypia, nuclear tant to distinguish metaplasia from transdifferentiation. in 1 in 860 (0.12%) individuals with the condition6.
hyperchromasia and loss of
Transdifferentiation is a process in which one differen- The progression of metaplasia to dysplasia may be
cell polarity.
tiated cell type converts into a completely different cell viewed as an ‘oncogenic’ phase, in contrast to the ini-
type present in the tissue2. Although the change from tial development of metaplasia, which is viewed as an
one type of cell to another in metaplasia might be a part ‘adaptive’ phase that occurs in response to environmen-
of the normal adult maturation processes, as will be dis- tal stress. While insights into the mechanisms leading to
cussed later, it is not known to occur during embryonic metaplasia are key to understanding tissue homeostasis
development3. Metaplasia may be induced or accelerated as well as adaptation to stress, studying metaplasia pro-
by some sort of abnormal stimulus (for example, acid or gression to low-grade dysplasia and high-grade dyspla-
base, and hence a change in pH; hormones; cigarette sia can reveal major contributors to malignancy at early
smoke; and alcohol)4. In the context of an abnormal stages. Given the wide prevalence of tissue metaplasia
stimulus, the original cells adapt to the environmental and the limited progression to dysplasia and c­ ancer,
stress by changing identity. If the stimulus that caused there is tremendous intersection of this topic with
metaplasia is removed, it is not clear whether the tissues ­cancer prevention, early detection, risk stratification,
can return to their normal pattern of differentiation. prognosis and therapy. This Review focuses on the types
University of Pennsylvania However, if the condition promoting metaplasia persists, of metaplasia, the potential cellular origins of metaplasia,
Perelman School of metaplasia can progress to dysplasia and occasionally the ways to model certain types of metaplasia and the
Medicine, 951 BRB,
malignancy, as will be discussed later (for example, in the potential opportunities for ­intervention to either reverse
421 Curie Boulevard,
Philadelphia, oesophagus)5. It is not realistically possible to determine or arrest it.
Pennsylvania 19104, USA. the prevalence or incidence of tissue metaplasia of any
Correspondence to A.K.R.  type in the general population, as that information would Tissue metaplasia
anil2@mail.med.upenn.edu require comprehensive and longitudinal monitoring, but Metaplasia tends to occur in tissues constantly exposed
doi:10.1038/nrc.2017.68 one can glean some information about metaplasia and to environmental agents, which are often injurious in
Published online 1 Sep 2017 progression to dysplasia and cancer in the oesophagus, nature. For example, the pulmonary system (lungs and

594 | OCTOBER 2017 | VOLUME 17 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

Glandular stomach trachea) and the gastrointestinal tract are common sites ectocervix, resulting in exposure of the ectocervix to an
The part of stomach that is of metaplasia owing to their contacts with air and food, acidic milieu9. This environment triggers a metaplastic
responsible for normal respectively. As a result, the tissue epithelial structure response, which is initially patchy in the crypts and cul-
physiological functions. adapts through metaplasia, with definitive morpho- minates in total replacement by squamous epithelium10.
Parietal cells
logical changes. The type of metaplasia depends upon Human papillomavirus (HPV) infection of metaplastic
Also known as oxyntic cells, the resident tissue. Metaplasia may be categorized squamous epithelium can result in dysplasia and squa-
these are the acid-producing broadly as squamous metaplasia, intestinal metaplasia mous cell cancer 11. HPV is associated with nearly all
cells in the stomach epithelium. or ­acinar–­ductal metaplasia (ADM) (TABLE 1). cervical dysplastic and cancer tissues12,13.
Other types of squamous metaplasia are rare. Breast
Chief cells
Pepsinogen- and
Squamous metaplasia. Lung squamous metaplasia can squamous metaplasia may be observed in fibroadenomas,
chymosin-producing cells in occur in either the alveolar epithelium or the airway epi- cysts, abscesses and chronic inflammation, and is almost
the stomach epithelium. thelium. Metaplasia of the alveolar epithelium features always benign14,15. It is rare for breast squamous metaplasia
the replacement of alveolar cells (normally a unilayer of to progress to breast squamous cell carcinoma, although
Atrophic gastritis
Loss of segments of the gastric
cuboidal or columnar cells) with squamous epithelium, this cancer accounts for only <1% of all breast malignan-
mucosa in the setting whereas metaplasia of the airway epithelium features cies16,17. Skin squamous metaplasia can be observed in
of inflammation. the replacement of bronchiolar or bronchial epithelium the setting of ulcers or scars and may be a precursor to
with squamous epithelium7 (FIG. 1). Squamous meta­ squamous cell carcinoma18,19. Similarly, squamous meta-
Spasmolytic polypeptide-­
plasia comprises multiple layers of cells and is believed plasia has been noted in sebaceous glands in the skin and
expressing metaplasia
(SPEM). Metaplastic cells that
to be resistant to injury compared with metaplasia of the other issues19.
are marked by spasmolytic alveolar, bronchial or bronchiolar epithelium. As a result
polypeptide expression in the of exposure to injury, squamous metaplasia may be Intestinal metaplasia. The second major tissue meta­
stomach epithelium. accompanied by varying grades of inflammation, fibro- plastic subtype is intestinal metaplasia, which is found in
sis and necrosis. The lung neoplasias that can emerge the distal oesophagus at the gastro-oesophageal junction
from squamous metaplasia include cystic ­keratinizing or in the glandular stomach (TABLE 1). In the case of Barrett
epithelioma and squamous cell carcinoma8. oesophagus, admixture of acid and bile is believed to result
Another illustration of squamous metaplasia is in mucosal injury, inflammation, cellular oxidative stress
observed in the cervix. The endocervix comprises a and the production of reactive oxygen species (ROS),
simple columnar (glandular) epithelium. By contrast, thereby creating a milieu permissive for metaplasia20–24
the ectocervix is composed of stratified squamous (FIG. 2). Acid reflux alone or with bile salts can increase
epithelium, forming a squamocolumnar junction ROS in models of Barrett oesophagus25. In the setting of
between the endocervix and the ectocervix. During chronic inflammation, long-term exposure to ROS can
puberty, the endocervix reconfigures (everts) to the damage proteins, lipids, mitochondria and DNA26. This
damage can lead to dysfunctional mitochondria, altered
gene expression patterns, induction of metaplasia and
Table 1 | Types of tissue metaplasia and environmental stimuli transformation of the epithelium. Barrett oesophagus can
Type of Stimulus Transition in Precancerous progress to low-grade dysplasia and high-grade dysplasia
metaplasia cell lineage and culminate in oesophageal adenocarcinoma (EAC)27,28.
Squamous metaplasia Ablative approaches, such as radiofrequency ablation and
Lung airway Cigarette smoke Columnar Yes
endoscopic mucosal resection, are employed in dysplastic
to squamous states29,30 and result in diminished progression to EAC.
Interestingly, even after ablation, there is a risk of recur-
Cervix Low vaginal pH, Columnar Yes
human papillomavirus to squamous rence of Barrett oesophagus31,32. One might speculate that
all abnormal tissue was not ablated successfully, allowing
Mammary gland* Inflammation, infection ? Yes
expansion of residual Barrett oesophageal tissue, and/or
Sebaceous gland* Inflammation ? Unclear that squamous progenitor cells have been reprogrammed
Skin* Inflammation Squamous Unclear to an intestinal metaplasia fate.
(maintained) In the case of the glandular stomach, chronic
Intestinal metaplasia Helicobacter pylori infection, which is highly prevalent
globally, can lead to the loss of acid-secreting parietal
Oesophagus‡ Acid and/or bile, obesity Squamous Yes
to intestinal cells in some patients33. Pathologists refer to the condi-
tion in which parietal and chief cells are lost as chronic
Stomach High salt intake, low Columnar Yes
vegetable and fruit intake, (stomach) atrophic gastritis. Although the most histologically obvi-
low vitamin C intake, to intestinal ous change that occurs in atrophic gastritis is the loss
Helicobacter pylori infection, of parietal cells, it should be noted that the digestive-­
autoimmune gastritis enzyme-secreting chief cells are also no longer pres-
Acinar–ductal metaplasia ent. As parietal cells die, metaplastic cells that express
Pancreas Inflammation Acinar to ductal Yes abundant spasmolytic polypeptide (SP, also known as
TFF2) emerge; thus, this type of metaplasia is referred
Mammary gland* Inflammation Acinar to ductal Unclear
to as spasmolytic p­ olypeptide-expressing metaplasia
Salivary gland* Inflammation Acinar to ductal Unclear (SPEM)34. Of note, ­parietal cell apoptosis is insufficient
*Much less investigated and inferred from descriptive pathology reports. ‡Barrett oesophagus. to induce metaplasia35.

NATURE REVIEWS | CANCER VOLUME 17 | OCTOBER 2017 | 595


©
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

Normal columnar epithelium Squamous metaplasia


(NKX2-1+) (SOX2+ p63+)

External
stimuli Stratification

Columnar cell Basement membrane Metaplastic


squamous cell
Figure 1 | Squamous metaplasia. In tissue columnar cells (for example, in the lung or cervix), external
Naturestimuli (for| Cancer
Reviews
example, low vaginal pH in the cervix and cigarette smoke in the lung) promote the conversion to metaplastic squamous
cells, which stratify. In the lung, columnar cells are identified by the expression of homeobox protein Nkx2.1 (NKX2‑1),
and squamous cells are enriched for p63 and SRY-box 2 (SOX2), similar to oesophageal basal cells and their marked
expression of p63 and SOX2.

The factors that contribute to the emergence of gas- adenocarcinoma46,47. Lineage tracing in mice reveals that
tric intestinal metaplasia include but are not limited to ADM occurs in vivo and that these ADM lesions can
ongoing H. pylori infection, bile reflux, cigarette smoking, progress to PanIN48. Epithelial explants from the exocrine
alcohol consumption and a diet low in fruit, vegetable and pancreas lose acinar cells in culture, with subsequent
vitamin C intake and high in salt intake36. Autoimmune expansion of ductal cells49–52. Human acinar cells also
gastritis can also cause atrophic gastritis, SPEM and undergo ADM, as observed by in vitro lineage tracing 53.
intestinal metaplasia34,37. The foci of gastric intesti-
nal metaplasia tend to appear initially at the antrum– Other tissue ADM. ADM has been observed infre-
corpus junction. Over time, the foci enlarge and con- quently in salivary glands and mammary glands
verge, involving the antrum and the corpus38. Complete (TABLE 1); although the meaning of this phenomenon is
gastric intestinal metaplasia resembles the small intesti- unclear, there is much overlap in morphological features
nal epithelium, with evidence of all small intestinal cell among the pancreas, the salivary gland and the mam-
lineages and full expression of brush border enzymes38. mary gland. ADM in the mouse salivary gland is a result
Incomplete gastric intestinal metaplasia resembles of the loss of p120‑catenin (also known as catenin‑δ1),
the colonic epithelium, with incomplete mucin expres- and mice lacking p120‑catenin in the salivary glands die
sion and the absence of a mature brush border (simi- postnatally, likely because the disruptions to salivary
lar to in Barrett oesophagus)38. Dysplastic foci may secretion affect digestion54. E‑cadherin is stabilized by
eventually appear within areas of intestinal metaplasia. p120‑catenin at the cell membrane as part of adher-
Reminiscent of intestinal metaplasia in the oesophagus, ens junctions55,56, and the loss of p120‑catenin creates
intestinal metaplasia in the stomach can also progress an environment for cell–cell disaggregation and local
to low-grade dysplasia and high-grade dysplasia and inflammation and is a likely stimulus for salivary ADM.
culminate in gastric adenocarcinoma36. The true fre-
quency of progression to adenocarcinoma in the stom- Cell of origin
ach is difficult to discern, given the large surface area of Barrett oesophagus. In tissue metaplasia, the epi-
the gastric mucosa (which cannot be easily covered with theli­um often contains a mixture of resident and ectopic
random tissue sampling), the lack of imaging modalities cells. Therefore, the cell of origin for the metaplastic
to guide targeted tissue sampling during endoscopy, and cells becomes ambiguous and sometimes controver-
the fact that the susceptible populations in Asia, Africa, sial. Barrett oesophagus has garnered attention in this
Central America and South America are too large for context 57–60. Studies in transgenic mice have revealed
Brush border uniform and standardized approaches to screening and that the interleukin‑1β (IL‑1β)–IL‑6 signalling cas-
The small intestinal epithelial surveillance39. Nevertheless, clinical progress is being cade and Delta-like protein 1 (DLL1)-dependent Notch
microvilli-covered surface that made with population-based approaches in countries signalling cause Barrett oesophagus61. Lineage trac-
expresses brush border such as Japan, Korea and Colombia40–42. ing indicates that these lesions arise from leucine-rich
enzymes that mediate the
transport of micronutrients
repeat-­containing G‑protein coupled receptor 5 (LGR5)+
from the lumen to within ADM. The third major example of metaplasia is ADM in gastric cardia stem cells61.
the epithelium. the pancreas (FIG. 3; TABLE 1). In mice, ADM is associated Another study in mice observed that Trp63 (which
with acute or chronic inflammation43. In humans, ADM encodes the transcription factor p63)-null embryos
Exocrine pancreas
may be found in chronic pancreatitis44. ADM is revers- rapidly develop intestine-like metaplasia at the squamo­
Compartments of acinar and
ductal cells that secrete and ible in mouse models, where resolution of the acute columnar junction of the oesophagus and stomach62.
transport digestive enzymes. inflammatory stimulus leads to reversion to the original Of note, Trp63‑knockout mice die at birth and also
acinar and ductal cell lineages45. Chronic inflammation have truncation of the limbs and striking absence of
Gastric cardia in the pancreas may also yield some permanent ADM epidermal, prostate, breast and urothelial tissues, find-
The small region that
constitutes the first part of the
lesions. In mice, ADM is a well-defined precursor to ings suggestive of the loss of ectodermal stem cells63.
stomach and is composed of pancreatic intraepithelial neoplasia (PanIN), and even- When Trp63 is deleted, the simple columnar epithe-
columnar cells. tually, PanIN lesions may progress to pancreatic ductal lium expands into the stratified squamous epithelial

596 | OCTOBER 2017 | VOLUME 17 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

Luminal surface Intestinal metaplasia


Goblet cell

Squamous cell Columnar cell


differentiation
• Acid or bile reflux
• Inflammation
Basement • RAS
membrane • Hedgehog signalling
Transcription factors: SOX2 and p63 CDX2 and Notch signalling

Figure 2 | Intestinal metaplasia in the oesophagus. The normal oesophageal squamous epithelial proliferative
Nature Reviewsbasal
| Cancer
cells (p63+ and SRY-box 2 (SOX2)+) undergo early and terminal differentiation as they migrate towards the luminal surface.
In concert with acid or bile reflux and pro-inflammatory stimuli (for example, interleukin 6 (IL‑6)–signal transducer and
activator of transcription 3 (STAT3)), incomplete intestinal metaplasia (presence of columnar cells and goblet cells
and absence of Paneth cells and enteroendocrine cells) appears. Goblet cells produce mucins, which are cytoprotective.
Mesenchymal homeobox protein BarH-like 1 (BARX1) and bone morphogenetic protein 4 (BMP4) (induced by epithelial
sonic hedgehog (SHH)) are critical for the transition from the squamous cell lineage to the intestinal (columnar) cell
lineage. Caudal type homeobox 2 (CDX2) is critical for the induction of columnar cells, and Notch signalling is a key
pathway for the maintenance of the columnar cell lineage. Genomic studies have revealed that TP53 mutation is
important in the earliest dysplastic clones. In this figure, the underlying premise is on the morphological and molecular
changes that occur, but the cell of origin is not implied.

territory and demonstrates certain intestinal-like epithelial separate consideration, bone marrow progenitor cells were
characteristics (for example, Alcian blue-positive stain- putatively shown to act as a cell of origin for oesopha-
ing). The authors of this study further hypothesized that geal metaplasia in a rat model, but it is not clear in what
the Trp63‑null embryonic columnar epithelium persists physi­ological context this might occur 68. To date, there
in the adult squamocolumnar junction62. When the is a lack of in vivo evidence in genetic mouse models that
neighbouring squamous epithelium is damaged, proves that oesophageal stem or progenitor cells are the
the residual Trp63‑null columnar cells can expand and source of Barrett oesophagus.
thereby resemble Barrett metaplasia62. Another possible cell of origin for Barrett oesoph-
Insights have also emerged from human studies of agus is from the oesophageal submucosal glands. DNA
Barrett oesophagus through the use of techniques such sequencing has revealed that human Barrett oesopha-
as microdissection, gene mutation analysis (for exam- gus and the neighbouring submucosal gland epithelium
ple, of mutations in mitochondrial DNA (mtDNA)), share the same mutations and loss of heterozygosity in
gene expression analysis of lineage-specific genes, the tumour suppressors cyclin-dependent kinase inhib-
immunohistochemistry and, occasionally, in  situ itor 2A (CDKN2A) and TP53 (which encodes p53)
hybridization. Some of these studies reveal that Barrett (REF. 69). Histological analysis has revealed an association
glands have features in common with gastric glands, of human ductal metaplastic oesophageal submucosal
resulting in the conclusion that Barrett oesophagus glands with Barrett high-grade dysplasia and EAC70.
originates from potential stem cells in the gastric car- In a porcine model, radiofrequency ablation of the
dia64. As an extension of this logic, through the use of oeso­phageal epithelium resulted in the conversion of
markers of proliferation and specific markers of Barrett oesophageal submucosal glands to a ductal phenotype
glands, it has been suggested that Barrett glands may with expression of the SRY-box 9 (SOX9) transcription
have properties overlapping those of pyloric gastric factor, a ductal marker 71. Overall, it is intriguing that
glands, which are located in the distal antrum and not ADM may represent a possible underpinning of Barrett
near the s­ quamocolumnar junction65. oesophagus if a cell of origin resides in the oesophageal
While these studies suggest that the cellular origin submucosal glands, as the same type of metaplasia is
of Barrett oesophagus is in the gastric cardia region of found in the pancreas. Studies in mice are not feasible,
the squamocolumnar junction, studies with 3D organo- as mice lack oesophageal submucosal glands.
typic cultures have revealed a possible cellular origin in How can one reconcile the seemingly divergent stud-
oesophageal basal cells66. Here, Barrett metaplasia devel- ies that implicate different sources for the cell or cells of
opment may depend upon the coordinated actions of origin? Some aspects are related to differences in model
MYC, the transcription factor caudal type homeobox 2 systems (from mouse to pig to human), experi­mental
(CDX2) and the inhibition of Notch signalling 66. Active approaches (non-genetic to genetic) and nomenclature
Notch signalling may be important in the maintenance of what constitutes a stem cell versus a progenitor cell.
of the columnar cell lineage in Barrett oesophagus61. Although there has been a tendency to conclude that the
Mechanistically, gastro-oesophageal reflux may induce cell of origin resides in only one tissue compartment,
Oesophageal submucosal cellular reprogramming of the oesophageal squamous it is tempting to speculate that more than one cellular
glands
Distinct structures below the
epithelium as a basis for metaplasia67. Presumably, origin may exist for the initiation of Barrett oesophagus.
oesophageal epithelium that this reprogramming could occur in oesophageal stem The cell of origin may depend upon ­context-specific
have secretory functions. or progenitor cells or even in differentiated cells. As a cell-autonomous and non-cell-autonomous cues.

NATURE REVIEWS | CANCER VOLUME 17 | OCTOBER 2017 | 597


©
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

Normal pancreas Pancreatitis


Inflammatory and
Acinar cells Acinar–ductal metaplasia immune cells
(MIST1+ and PTF1A+) (PRRX1+, SOX9+,
Nestin+, HES1+)

Mutant KRAS signalling


and inflammation

Duct cells

Fibroblasts
Acinar cells (↓SHH)

Figure 3 | Acinar–ductal metaplasia. Normal pancreatic acinar cells, which compose the bulk of the pancreatic
parenchyma and express pancreas transcription factor 1 subunit α (PTF1A) and MIST1, can convertNature Reviews | Cancer
into a ductal-cell-like
state, which is referred to as acinar–ductal metaplasia (ADM).  Pro-inflammatory stimuli from T cells, macrophages and
myeloid-derived suppressor cells (MDSCs) and mutant KRAS are critical for the appearance of ADM lesions. These lesions
are enriched for paired mesoderm homeobox protein 1 (PRRX1) and SRY-box 9 (SOX9) transcription factors, as well as for
Nestin+ and the transcription factor HES1+ cells, the latter owing to active Notch signalling. Pancreatic fibroblasts are a
source of sonic hedgehog (SHH), which slows ADM formation.

Gastric intestinal metaplasia. Gastric intestinal meta- origin of squamous metaplasia, but this claim has not
plasia in the glandular stomach arises in the setting of yet been substantiated through genetic in vivo lineage-­
atrophic gastritis. There are two competing models tracing experiments. Other types of tissue metaplasia
for the origin of gastric metaplasia, and they will both are described in pathology specimens, and the dearth
be summarized. Intestinal metaplasia may arise from of studies does not permit conclusions about the cell(s)
SPEM72 (FIG. 4) or perhaps directly from chief cells73,74 of origin.
or gastric isthmus stem cells75. SPEM is associated with
the development of dysplasia in H. pylori-infected mouse Expansion of metaplastic clones
models and is also observed in association with human Lineage tracing in humans, by following certain muta-
gastric cancer 73,76. Is there a specific subset of cells within tions in mtDNA, has demonstrated that the expanded
chief cells that trigger SPEM or cancer? It is known that metaplastic epithelia share the same mtDNA point
LGR5+ cells with properties of self-renewal are located mutations as those in human Barrett oesophagus and
in the gastric antrum and yield long-lived gastric units gastric intestinal metaplasia. These findings support the
in vitro77. Furthermore, reserve LGR5+ cells have now premise that Barrett oesophagus and gastric intestinal
been identified within mouse and human chief cells and metaplasia are clonally evolved64,82–84 and raise the pos-
can catalyse epithelial regeneration after injury in vitro78. sibility that metaplasia in these tissues initially occurs in
Although LGR5+ cells do not appear to give rise to either stem cells and that subsequent gland fission promotes
SPEM79 or intestinal metaplasia, they have been found to the spread of intestinal metaplasia85. The true identity of
give rise to early gastric cancer 78. the original cells remains to be established; alternatively,
Other studies advocate that the cell of origin for gas- gene reprogramming might be occurring in progenitor
tric intestinal metaplasia resides in the gastric isthmus80. cells, or possibly in differentiated cells, as rare events that
For instance, MIST1 (also known as BHLHA15)+ stem trigger metaplasia. Notably, long-lived stem cells have
cells in the gastric isthmus display clonal expansion been identified in both human and mouse oesophagus
in the setting of KrasG12D mutation, resulting in gastric and stomach86–89, with compelling evidence obtained in
intestinal metaplasia75,81. Through the use of genetic the oesophagus from lineage tracing in a genetic mouse
mouse models and lineage tracing over time to deter- model in which keratin 15 (KRT15)+ oesophageal basal
mine the source of proliferation, one compelling study cells are long-lived and contribute to tissue regeneration
found that metaplastic cells are long-lived, suggesting after radiation injury.
that reprogramming occurs in long-lived stem cells and
does not involve transdifferentiation75. Furthermore, Regulation of metaplasia
ablating the stem cells blocked metaplasia and cancer. Transcription factors in metaplasia: p63 and SOX2.
Some drivers of tissue metaplasia have been identified
Other types of metaplasia. From the viewpoint of cer- and revolve around transcription factors whose func-
vical squamous metaplasia, endocervical crypts may be tions contribute to cellular identity and plasticity during
seen deep to the surface epithelium. One might spec- metaplasia. For example, the transcription factor SOX2
ulate that such crypts are a potential morphological plays a critical role in the generation of the stratified

598 | OCTOBER 2017 | VOLUME 17 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

Foveolar
hyperplasia

H. pylori

Antralization SPEM

Surface cells

Normal
progenitor
cells
• Parietal cell loss Dysplasia and
• Chronic inflammation adenocarcinoma
?
Parietal cells

Intestinalization

Chief cells

IM
H. pylori

Figure 4 | Spasmolytic polypeptide-expressing metaplasia and gastric intestinal metaplasia. The gastric epithelium
Nature Reviews | Cancer
harbours chief cells at the base, underneath acid-producing parietal cells, progenitor cells (or stem cells) and surface cells.
In the face of Helicobacter pylori infection, there is parietal cell loss and chronic inflammation. One pathway results in
foveolar hyperplasia and spasmolytic polypeptide-expressing metaplasia (SPEM), which might be a precursor to intestinal
metaplasia (IM, indicated by the dashed arrow). Another pathway leads directly to IM. Both SPEM and IM are precursors to
dysplasia and later adenocarcinoma. Please refer to the main text for a discussion of cell of origin.

squamous epithelium lining the oesophagus and the BarH-like 1 (BARX1), a transcription factor enriched in
anterior portion of the stomach in mice (forestomach; the mesenchyme, also results in the presence of secretory
a part of the stomach that is present in mice but not columnar cells in the squamous cell region of the mouse
humans)90. Significant reductions in SOX2 protein levels oesophagus and forestomach91. The metaplastic epithe-
lead to the formation of simple columnar epithelium in lium ectopically expresses mucin 5AC (MUC5AC)91.
place of stratified squamous cells90. Moreover, the meta- These findings suggest that both epithelial (for example,
plastic cells secrete mucins, similar to the secretion of SOX2 and p63) and mesenchymal (BARX1) factors are
mucins by columnar cells in the glandular stomach and required to establish squamous cell identity in the upper
Foveolar hyperplasia
A characteristic of reactive
small intestine. Interestingly, mesenchymal transcrip- gastrointestinal tract.
gastritis observed in the gastric tion factors can also modulate squamous cell specifica- In the adult lung, the presence of squamous meta­
antrum and body. tion. Deletion of the gene encoding homeobox protein plasia is characterized by an expansion of p63+ SOX2+

NATURE REVIEWS | CANCER VOLUME 17 | OCTOBER 2017 | 599


©
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

cells in the proximal lung airways92. Although both p63 transcription regulator 1 (Nupr1)118 and regenerating
and SOX2 are expressed exclusively in the basal progeni- islet-derived 1 (Reg1)119, genes that are expressed in
tor cells of the oesophagus and trachea, the latter is lined embryonic pancreatic cells.
by a simple columnar epithelium in adults. Therefore, the Deletion of Mist1 also blocks the differentiation
presence of abundant p63+ SOX2+ basal cells in the lung of chief cells in the base of gastric glandular units120.
metaplastic squamous epithelium suggests the adoption Consequently, uncommitted epithelial cells accumu-
of an oesophageal basal cell lineage93,94. late along the pathway where the derivatives of isthmus
progenitor cells migrate120. More importantly, SPEM
Transcription factors in metaplasia: CDX2. Intestinal occurs in the glandular stomach of Mist1−/− mice upon
metaplasia identity may be fostered by the transcrip- treatment with the protonophore DMP‑777 or its ana-
tion factor CDX2, which is overexpressed in both logue L635, which induces the loss of parietal cells and
Barrett oesophagus and gastric intestinal metaplasia95,96. inflammation121. These findings suggest that MIST1 is
Indeed, transgenic mice with Cdx2 expression targeted critical for the maturation of epithelial progenitor cells in
to the glandular stomach develop intestinal meta­plasia97. the developing pancreas and glandular stomach. In both
Provocatively, conditional knockout of Cdx2 in the organs, blocking the differentiation process seems to
mouse intestine yields squamous metaplasia accom- facilitate the incidence of metaplasia. It will be interest-
panied by the ectopic presence of p63+ SOX2+ basal ing to determine whether conditional deletion of Mist1
cells98. CDX2 controls chromatin access for interactions in the adult also interferes with epithelial differentiation
with other transcription factors to modulate cell cycle and facilitates metaplasia.
­progression, proliferation and differentiation99,100.
Epigenetic and genomic considerations
Transcription factors in ADM: PTF1A and MIST1. It is conceivable that these transcription factors, in spe-
Pancreatic ADM is driven by the loss of acinar-­lineage- cific tissues, function in a coordinated fashion with epi-
specific transcription factors, such as pancreas transcrip- genetic regulators that fuel chromatin remodelling to
tion factor 1 subunit α (PTF1A), and the induction of the permit the binding of genomic loci by the transcription
paired mesoderm homeobox protein 1 (PRRX1)–SOX9 factors to create a cellular framework for metaplasia.
axis101–104. The molecular basis of ADM involves trans- This type of regulation has yet to be definitively identi-
forming growth factor-α (TGFα) and epidermal growth fied in tissue metaplasia but has been suggested to occur
factor receptor (EGFR) signalling 43. Transgenic mice with in Barrett oesophagus122,123.
Tgfa overexpression in the exocrine pancreas show evi- Ideally, genomic analysis of metaplasia across tissue
dence of duct-like cells49. Acinar-specific expression of types might reveal common processes, but anatomic
activated KRAS in mice (LSL-KrasG12D/+;Ptf1aCre/+ mice) location often serves as a barrier to obtaining tissues for
results in ADM105. These metaplastic structures are pro- analysis. In the case of Barrett oesophagus, tissue is acces-
liferative and express Notch target genes106–108. Transgenic sible via upper endoscopy. To that end, whole and paired
expression of pancreatic and duodenal homeobox 1 exomic sequencing of normal and Barrett oeso­phagus
(Pdx1) in the Ptf1a locus109 results in the transition of has unravelled a crucial role for mutations of TP53 and
acinar cells to duct-like cells and is regulated through sig- CDKN2A124–126. Although the exact consequences of
nal transducer and activator of transcription 3 (STAT3) TP53 mutations in the development of Barrett oesoph-
activation110. Pancreatic ADM lesions are associated with agus remain to be elucidated, it is possible that such
inflammation with a concomitant increase in MEK–ERK mutations might serve as a gatekeeper for the mainte-
signalling 111. In human pancreatic sections, the close nance of metaplasia and/or drive the conversion of meta-
juxtaposition of ADM and PanIN suggests but does not plasia to low-grade dysplasia in the oesophagus124 and
prove progression of ADM to PanIN112. perhaps the stomach as well127. Loss of CDKN2A results
MIST1 is a global regulator of secretory cell archi- in enhanced cell cycle progression. Additionally, muta-
tecture in multiple professional secretory cells, such tions of other transcription factors have been detected in
as the acinar cells of the pancreas and salivary gland Barrett oesophagus, for example, GATA6 amplification
and the chief cells of the stomach113. During metaplasia and SMAD4 deletion125. Specifically, SMAD4 deletion
in the stomach, salivary gland and pancreas, Mist1 is could foster a pro-proliferative environment, and GATA6
one of the first genes whose expression is decreased as amplification might also have oncogenic properties. It
cells scale down their secretion114, and forced expres- will be interesting in the future to determine the role of
sion or loss of MIST1 interferes with the induction these transcription factors in the development of Barrett
of and recovery from metaplasia, respectively 115–117. oesophagus and gastric intestinal metaplasia.
Specifically, deletion of Mist1 leads to reduced lev- Although EAC is highly aneuploid, with multiple high-
els of amylase, an enzyme specifically produced by level gains and losses, Barrett oesophagus is typically copy
mature exocrine pancreatic cells. Lineage tracing number neutral (99.7%, range 62.8–100%, of genomes had
with cells expressing lacZ in the Mist1 locus (Mist1− a copy number of 2)126. Regarding specific alterations,
lacZ+ cells) has demonstrated that these cells express recurrent loss of 9p heterozygosity was found in 11 of
ductal genes (for example, keratin 20 (Krt20)), sug- 23 (48%) samples, and no high-level amplifications were
gesting that a cell-fate switch occurs in differentiated observed. The number of focal deletions was found to
exocrine cells102. Of note is that the same Mist1− lacZ+ increase with increasing stage from non-­dysplastic Barrett
cells also maintain moderate levels of nuclear protein oesophagus to dysplastic Barrett oesophagus to EAC125.

600 | OCTOBER 2017 | VOLUME 17 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

These deletions in Barrett oesophagus were often not mediated by CD11b+ GR1+ immature myeloid cells or
shared with the patients’ paired EAC tissues, suggesting myeloid-derived suppressor cells134.
that they may not be important in progression to cancer. Activated fibroblasts interplay with epithelial cells
Focal amplifications seemed to be observed almost exclu- in the development of metaplasia through aberrant
sively in EAC, with rare examples of these changes in sam- sonic hedgehog (SHH) signalling 135–138. In the context
ples of high-grade dysplasia125. Genome doubling events of Barrett oesophagus, it has been proposed that epi-
were also commonly identified in EAC and only rarely thelial cells induce SHH expression, which in turn pro-
identified in high-grade dysplasia. No genome doubling motes stromal expression of SHH target genes such as
was seen in low-grade dysplasia or non-dysplastic Barrett patched 1 (PTCH1) and bone morphogenetic protein 4
oesophagus125. Taken together, these studies suggest a pat- (BMP4), and may result in the expression of epithelial
tern where there is acquisition of a small number of copy SOX9 and cytokeratins137. Similarly, it has been shown
number changes in non-dysplastic Barrett oesophagus that in the pancreas, genetic ablation of smoothened
and low-grade dysplasia125,126. Many of these changes seem (Smo), which mediates SHH signalling, in fibroblasts
to be present in both patients who progress to higher-stage results in the activation of AKT and the transcrip-
disease and those who have stable non-progressive dis- tion factor GLI2 and increased ADM135,139. Another
ease. However, around the time of development of high- factor in the stroma is the transcription factor ETS2.
grade dysplasia and then EAC, there is a dramatic rise in Stromal ETS2 in the mouse pancreas driven by KrasG12D-
the number of copy number changes found throughout induced ADM has been shown to be important for the
the genome125,126. recruitment of chemokines and immune cells, such as
T regu­latory cells, myeloid-derived suppressor cells and
Pathogens, inflammation and immune cells are key mature macrophages140.
factors in metaplasia. The non-epithelial actions Cell intrinsic events also appear to be of functional
(effects on immune cells, inflammatory cells and fibro- relevance in tissue metaplasia. To that end, mutant
blasts) of acid and/or bile, pathogens, cigarette smoke, KRAS signalling is critical for pancreatic ADM141,142.
alcohol and nutritional deficiencies conspire to foster Similarly, mutant KRAS in gastric chief cells results in
a stressful environment in which epithelial cells are gastric SPEM143. MYC, which is downstream of mutant
forced to change identity and lineage specification. KRAS, is associated with intestinal metaplasia in the
Interestingly, microorganisms may find these shifts oesophagus and stomach and with squamous ­metaplasia
in environmental cues very attractive. For example, in the lung 144–146.
chronic infection with H. pylori, especially virulent
strains, results in loss of parietal cells and in acid pro- Future translational directions
duction, which are key drivers of gastric intestinal meta- Each type of metaplasia is identified through character-
plasia33. These environmental changes may be mediated istic morphology, histopathology and lineage-specific
in part through the local recruitment of lymphocytes markers (including transcription factors). However, the
and the secretion of cytokines. Invariably, HPV onco- roles of aetiological factors (such as acid, bile and cig-
genic strains are associated with cervical squamous arette smoke) are different in each type of metaplasia.
metaplasia and facilitate the development of squamous The initiation and/or maintenance of oesophageal and
dysplasia13. Indeed, there may be a role for communi- gastric intestinal metaplasia and pancreatic ADM may
ties of microorganisms in oesophageal intestinal meta- share certain principles: activation of common cell sig-
plasia128 and, by extension, possibly in lung squamous nalling pathways (including mutant KRAS) and the roles
metaplasia and skin squamous metaplasia. of pro-inflammatory cells, immune cells and fibroblasts,
Pro-inflammatory and immunological cues influ- which may act in concert to promote metaplasia.
ence epithelial cell signalling and the induction of Intestinal metaplasia, whether in the oesophagus or
metaplasia. This mechanism has been established for the stomach, is a bona fide precursor to a continuum of low-
IL‑6–STAT3 pathway in Barrett oesophagus61 and pan- grade dysplasia to high-grade dysplasia to adenocarci-
creatic ADM129. Pancreatic ADM lesions are commonly noma82,87,147. Squamous metaplasia, likewise, can progress
associated with inflammation111. Immune cells in the to dysplasia and squamous cell carcinoma8. Pancreatic
microenvironment play important roles in metaplasia. ADM, at least in mice, is a precursor to PanIN and
For example, depletion of macrophages in L635‑induced pancreatic ductal adenocarcinoma148. Considering that
gastric SPEM prevents its development 130. Macrophage- metaplasia is a precursor to dysplasia and cancer, there
secreted inflammatory cytokines, such as tumour necro- is still a need to elucidate further the entire s­ pectrum of
sis factor (TNF) and CC motif chemokine 5 (CCL5), events that are responsible in this continuum.
play roles in pancreatic ADM, with contributions Screening for metaplasia and the metaplasia-­dysplasia
from macrophage-released matrix metalloproteinases continuum is possible when tissue access is possible.
(MMPs), such as MMP9, as well131,132. Recently, it was This is the case in the oesophagus and stomach, which
demonstrated that IL‑13 alters macrophage populations may be assessed by upper endoscopy, and in the cervix,
from an inflammatory macrophage subpopulation to which may be assessed by Pap smear. In each tissue, the
an alternatively activated macrophage subpopulation finding of low-grade dysplasia requires closer evaluation
in ADM133. The development of Barrett‑like metaplasia by the pathologist and closer monitoring by the clinician.
in the mouse requires the suppression of CD8+ T cell-­ The identification of patient groups at risk of dysplasia
dependent apoptosis of epithelial cells, which is probably and cancer enables the use of more elaborate screening

NATURE REVIEWS | CANCER VOLUME 17 | OCTOBER 2017 | 601


©
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

and surveillance strategies. Examples include genomic metaplasia-dysplasia-cancer states, with the possible
and transcriptome analyses that may be helpful in differ- exception of the progression of Barrett oesophageal dys-
ent tissue metaplastic states, as has been done in Barrett plasia to cancer. For the progression of Barrett oesopha­
oesophagus. Furthermore, cell-free DNA and circulating gus, one would imagine a reduction of the observed
epithelial and tumour cells may provide further infor- 0.12% frequency of progression6 if all patients could be
mation for subtyping patients in the future. Circulating captured; however, ablation is recommended only for
materials might identify those patients who are likely low-grade and high-grade dysplasia and not for meta-
to progress to dysplasia or who harbour dysplastic cells. plasia. Gastric intestinal metaplasia with low-grade dys­
Another at‑risk population group comprises individu- plasia requires close monitoring, and high-grade ­dysplasia
als with an inherited genetic predisposition to cancer. It requires endoscopic or surgical resection151.
does not appear that most cancers in inherited genetic Future research in metaplasia should continue to
syndromes evolve through metaplasia. However, a small focus or elaborate upon determining cell or cells of ori-
subset of Barrett o ­ esophagus cases may be heritable149,150. gin, carrying out new or expanded genomic analyses,
From a therapeutic viewpoint, ablation through developing more model systems (for example, 3D cul-
mechanical and chemical modalities in the cervix, blad- tures) and applying this knowledge to prevention and
der and skin is an approach to eradicating metaplas- therapy. Furthermore, given the nature of tissue meta-
tic lesions. It is difficult to estimate how many cancers plasia, the identification of common mechanisms and
would be averted with ablative strategies, as popula- pathways in different tissues may alleviate the barriers
tion-based calculations are lacking in almost all tissue to prevention and therapy.

1. Slack, J. M. & Tosh, D. Transdifferentiation and 17. Bellino, R. et al. Metaplastic breast carcinoma: radiofrequency ablation. Am. J. Gastroenterol. 112,
metaplasia—switching cell types. Curr. Opin. Genet. pathology and clinical outcome. Anticancer Res. 23, 87–94 (2017).
Dev. 11, 581–586 (2001). 669–673 (2003). 32. Zeki, S. S. et al. Clonal selection and persistence in
2. Jopling, C., Boue, S. & Izpisua Belmonte, J. C. 18. Alam, M. & Ratner, D. Cutaneous squamous-cell dysplastic Barrett’s esophagus and intramucosal
Dedifferentiation, transdifferentiation and carcinoma. N. Engl. J. Med. 344, 975–983 (2001). cancers after failed radiofrequency ablation.
reprogramming: three routes to regeneration. 19. Buezo, G. F., Fernandez, J. F., Tello, E. D. & Am. J. Gastroenterol. 108, 1584–1592 (2013).
Nat. Rev. Mol. Cell Biol. 12, 79–89 (2011). Diez, A. G. Squamous metaplasia of sebaceous 33. Noto, J. M. & Peek, R. M. Jr. Helicobacter pylori:
3. Quinlan, J. M., Colleypriest, B. J., Farrant, M. & gland. J. Cutan. Pathol. 27, 298–300 (2000). an overview. Methods Mol. Biol. 921, 7–10 (2012).
Tosh, D. Epithelial metaplasia and the development of 20. Chen, X. et al. Oxidative damage in an esophageal 34. Petersen, C. P., Mills, J. C. & Goldenring, J. R.
cancer. Biochim. Biophys. Acta 1776, 10–21 (2007). adenocarcinoma model with rats. Carcinogenesis 21, Murine models of gastric corpus preneoplasia.
4. Slack, J. M. Metaplasia and transdifferentiation: from 257–263 (2000). Cell. Mol. Gastroenterol. Hepatol. 3, 11–26 (2017).
pure biology to the clinic. Nat. Rev. Mol. Cell Biol. 8, 21. Inayama, M., Hashimoto, N., Tokoro, T. & 35. Burclaff, J., Osaki, L. H., Liu, D., Goldenring, J. R. &
369–378 (2007). Shiozaki, H. Involvement of oxidative stress in Mills, J. C. Targeted apoptosis of parietal cells is
5. Sharma, P. et al. Dysplasia and cancer in a large experimentally induced reflux esophagitis and insufficient to induce metaplasia in stomach.
multicenter cohort of patients with Barrett’s esophageal cancer. Hepatogastroenterology 54, Gastroenterology 152, 762–766 (2017).
esophagus. Clin. Gastroenterol. Hepatol. 4, 566–572 761–765 (2007). 36. Amieva, M. & Peek, R. M. Jr. Pathobiology of
(2006). 22. Jenkins, G. J. et al. Deoxycholic acid at neutral and helicobacter pylori-induced gastric cancer.
One of a number of key studies to estimate the acid pH, is genotoxic to oesophageal cells through Gastroenterology 150, 64–78 (2016).
progression of Barrett oesophagus to dysplasia the induction of ROS: The potential role of anti- 37. Jeong, S. et al. Distinct metaplastic and inflammatory
and adenocarcinoma. oxidants in Barrett’s oesophagus. Carcinogenesis phenotypes in autoimmune and adenocarcinoma-
6. Hvid-Jensen, F., Pedersen, L., Drewes, A. M., 28, 136–142 (2007). associated chronic atrophic gastritis. United Eur.
Sorensen, H. T. & Funch-Jensen, P. Incidence of 23. Song, S., Guha, S., Liu, K., Buttar, N. S. & Gastroenterol. J. 5, 37–44 (2017).
adenocarcinoma among patients with Barrett’s Bresalier, R. S. COX‑2 induction by unconjugated 38. Correa, P., Piazuelo, M. B. & Wilson, K. T. Pathology of
esophagus. N. Engl. J. Med. 365, 1375–1383 bile acids involves reactive oxygen species-mediated gastric intestinal metaplasia: clinical implications. Am.
(2011). signalling pathways in Barrett’s oesophagus and J. Gastroenterol. 105, 493–498 (2010).
7. Leube, R. E. & Rustad, T. J. Squamous cell metaplasia oesophageal adenocarcinoma. Gut 56, 1512–1521 A comprehensive review on gastric intestinal
in the human lung: molecular characteristics of (2007). metaplasia.
epithelial stratification. Virchows Arch. B Cell Pathol. 24. Feng, C. et al. Diallyl disulfide suppresses the 39. Lordick, F. et al. Unmet needs and challenges in gastric
Incl Mol. Pathol. 61, 227–253 (1991). inflammation and apoptosis resistance induced by cancer: the way forward. Cancer Treat. Rev. 40,
8. Dotto, G. P. & Rustgi, A. K. Squamous cell cancers: DCA through ROS and the NF‑kappaB signaling 692–700 (2014).
a unified perspective on biology and genetics. pathway in human Barrett’s epithelial cells. 40. Pasechnikov, V., Chukov, S., Fedorov, E., Kikuste, I. &
Cancer Cell 29, 622–637 (2016). Inflammation 40, 818–831 (2017). Leja, M. Gastric cancer: prevention, screening and
9. Park, K. J. & Soslow, R. A. Current concepts in cervical 25. Feagins, L. A. et al. Mechanisms of oxidant early diagnosis. World J. Gastroenterol. 20,
pathology. Arch. Pathol. Lab Med. 133, 729–738 production in esophageal squamous cell and 13842–13862 (2014).
(2009). Barrett’s cell lines. Am. J. Physiol. Gastrointest Liver 41. Hamashima, C. et al. The Japanese guidelines for
10. Regauer, S. & Reich, O. CK17 and p16 expression Physiol. 294, G411–G417 (2008). gastric cancer screening. Jpn J. Clin. Oncol. 38,
patterns distinguish (atypical) immature squamous 26. Federico, A., Morgillo, F., Tuccillo, C., Ciardiello, F. & 259–267 (2008).
metaplasia from high-grade cervical intraepithelial Loguercio, C. Chronic inflammation and oxidative 42. Choi, K. S. et al. Performance of gastric cancer
neoplasia (CIN III). Histopathology 50, 629–635 stress in human carcinogenesis. Int. J. Cancer 121, screening by endoscopy testing through the National
(2007). 2381–2386 (2007). Cancer Screening Program of Korea. Cancer Sci. 102,
11. Zsemlye, M. High-grade cervical dysplasia: 27. Rustgi, A. K. & El‑Serag, H. B. Esophageal 1559–1564 (2011).
pathophysiology, diagnosis, and treatment. Obstet. carcinoma. N. Engl. J. Med. 371, 2499–2509 43. Reichert, M. & Rustgi, A. K. Pancreatic ductal cells in
Gynecol. Clin. North Am. 35, 615–621 (2008). (2014). development, regeneration, and neoplasia. J. Clin.
12. Psyrri, A. & DiMaio, D. Human papillomavirus in A review article on oesophageal squamous cell Invest. 121, 4572–4578 (2011).
cervical and head-and-neck cancer. Nat. Clin. Pract. carcinoma and adenocarcinoma. 44. Basturk, O. et al. A revised classification system and
Oncol. 5, 24–31 (2008). 28. Spechler, S. J. & Souza, R. F. Barrett’s esophagus. recommendations from the baltimore consensus
13. Burd, E. M. Human papillomavirus and cervical N. Engl. J. Med. 371, 836–845 (2014). meeting for neoplastic precursor lesions in the
cancer. Clin. Microbiol. Rev. 16, 1–17 (2003). 29. Shaheen, N. J. et al. Radiofrequency ablation in pancreas. Am. J. Surg. Pathol. 39, 1730–1741
14. Raju, G. C. The histological and immunohistochemical Barrett’s esophagus with dysplasia. N. Engl. J. Med. (2015).
evidence of squamous metaplasia from the 360, 2277–2288 (2009). 45. Hegyi, P. & Petersen, O. H. The exocrine pancreas:
myoepithelial cells in the breast. Histopathology 17, A clinical trial that demonstrated efficacy of the acinar–ductal tango in physiology and
272–275 (1990). radiofrequency ablation of Barrett oesophagus pathophysiology. Rev. Physiol. Biochem. Pharmacol.
15. Behranwala, K. A., Nasiri, N., Abdullah, N., Trott, P. A. with dysplasia. 165, 1–30 (2013).
& Gui, G. P. Squamous cell carcinoma of the breast: 30. Schlottmann, F. & Patti, M. G. Current concepts in 46. Ying, H. et al. Genetics and biology of pancreatic ductal
clinico-pathologic implications and outcome. treatment of Barrett’s esophagus with and without adenocarcinoma. Genes Dev. 30, 355–385 (2016).
Eur. J. Surg. Oncol. 29, 386–389 (2003). dysplasia. J. Gastrointest. Surg. 21, 1354–1360 47. Hosoda, W. & Wood, L. D. Molecular genetics of
16. Wang, X. et al. Metaplastic carcinoma of the breast: (2017). pancreatic neoplasms. Surg. Pathol. Clin. 9, 685–703
p53 analysis identified the same point mutation in 31. Guthikonda, A. et al. Clinical outcomes following (2016).
the three histologic components. Mod. Pathol. 14, recurrence of intestinal metaplasia after successful 48. Strobel, O. et al. In vivo lineage tracing defines the
1183–1186 (2001). treatment of Barrett’s esophagus with role of acinar-to‑ductal transdifferentiation in

602 | OCTOBER 2017 | VOLUME 17 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

inflammatory ductal metaplasia. Gastroenterology 69. Leedham, S. J. et al. Individual crypt genetic 90. Que, J. et al. Multiple dose-dependent roles for Sox2
133, 1999–2009 (2007). heterogeneity and the origin of metaplastic glandular in the patterning and differentiation of anterior
This study used lineage tracing to demonstrate epithelium in human Barrett’s oesophagus. Gut 57, foregut endoderm. Development 134, 2521–2531
pancreatic ADM. 1041–1048 (2008). (2007).
49. De Lisle, R. C. & Logsdon, C. D. Pancreatic acinar cells 70. Garman, K. S. et al. Ductal metaplasia in oesophageal 91. Kim, B. M., Buchner, G., Miletich, I., Sharpe, P. T.
in culture: expression of acinar and ductal antigens in a submucosal glands is associated with inflammation & Shivdasani, R. A. The stomach mesenchymal
growth-related manner. Eur. J. Cell Biol. 51, 64–75 and oesophageal adenocarcinoma. Histopathology transcription factor Barx1 specifies gastric epithelial
(1990). 67, 771–782 (2015). identity through inhibition of transient Wnt signaling.
50. Githens, S. et al. Mouse pancreatic acinar/ductular 71. Kruger, L. et al. Ductular and proliferative response of Dev. Cell 8, 611–622 (2005).
tissue gives rise to epithelial cultures that are esophageal submucosal glands in a porcine model of 92. Chen, Z., Fillmore, C. M., Hammerman, P. S., Kim, C. F.
morphologically, biochemically, and functionally esophageal injury and repair. Am J. Physiol. & Wong, K. K. Non-small-cell lung cancers: a
indistinguishable from interlobular duct cell cultures. Gastrointest. Liver Physiol. http://dx.doi.org/10.1152/ heterogeneous set of diseases. Nat. Rev. Cancer 14,
In Vitro Cell. Dev. Biol. Anim. 30A, 622–635 (1994). ajpgi.00036.2017 (2017). 535–546 (2014).
51. Rooman, I., Heremans, Y., Heimberg, H. & Bouwens, L. 72. Goldenring, J. R., Nam, K. T. & Mills, J. C. The origin of 93. Daniely, Y. et al. Critical role of p63 in the
Modulation of rat pancreatic acinoductal pre-neoplastic metaplasia in the stomach: chief cells development of a normal esophageal and
transdifferentiation and expression of PDX‑1 in vitro. emerge from the Mist. Exp. Cell Res. 317, tracheobronchial epithelium. Am. J. Physiol. Cell
Diabetologia 43, 907–914 (2000). 2759–2764 (2011). Physiol. 287, C171–C181 (2004).
52. Sphyris, N., Logsdon, C. D. & Harrison, D. J. Improved This article presents an overview of SPEM. 94. Gontan, C. et al. Sox2 is important for two crucial
retention of zymogen granules in cultured murine 73. Lennerz, J. K. et al. The transcription factor MIST1 processes in lung development: branching
pancreatic acinar cells and induction of acinar–ductal is a novel human gastric chief cell marker whose morphogenesis and epithelial cell differentiation.
transdifferentiation in vitro. Pancreas 30, 148–157 expression is lost in metaplasia, dysplasia, and Dev. Biol. 317, 296–309 (2008).
(2005). carcinoma. Am. J. Pathol. 177, 1514–1533 (2010). 95. Eda, A. et al. Aberrant expression of CDX2 in Barrett’s
53. Houbracken, I. et al. Lineage tracing evidence for 74. Mills, J. C. & Goldenring, J. R. Metaplasia in the epithelium and inflammatory esophageal mucosa.
transdifferentiation of acinar to duct cells and plasticity stomach arises from gastric chief cells. Cell. Mol. J. Gastroenterol. 38, 14–22 (2003).
of human pancreas. Gastroenterology 141, 731–741 Gastroenterol. Hepatol. 4, 85–88 (2017). 96. Phillips, R. W., Frierson, H. F. Jr & Moskaluk, C. A.
(2011). 75. Hayakawa, Y. et al. Mist1 expressing gastric stem cells Cdx2 as a marker of epithelial intestinal differentiation
54. Davis, M. A. & Reynolds, A. B. Blocked acinar maintain the normal and neoplastic gastric epithelium in the esophagus. Am. J. Surg. Pathol. 27,
development, E‑cadherin reduction, and intraepithelial and are supported by a perivascular stem cell niche. 1442–1447 (2003).
neoplasia upon ablation of p120‑catenin in the mouse Cancer Cell 28, 800–814 (2015). 97. Silberg, D. G. et al. Cdx2 ectopic expression induces
salivary gland. Dev. Cell 10, 21–31 (2006). 76. Goldenring, J. R. & Nam, K. T. Oxyntic atrophy, gastric intestinal metaplasia in transgenic mice.
55. Ishiyama, N. et al. Dynamic and static interactions metaplasia, and gastric cancer. Prog. Mol. Biol. Transl Gastroenterology 122, 689–696 (2002).
between p120 catenin and E‑cadherin regulate the Sci. 96, 117–131 (2010). This study showed that expression of CDX2 in the
stability of cell-cell adhesion. Cell 141, 117–128 77. Barker, N., Bartfeld, S. & Clevers, H. Tissue-resident mouse stomach results in gastric intestinal
(2010). adult stem cell populations of rapidly self-renewing metaplasia.
56. Kourtidis, A., Ngok, S. P. & Anastasiadis, P. Z. p120 organs. Cell Stem Cell 7, 656–670 (2010). 98. Gao, N., White, P. & Kaestner, K. H. Establishment
catenin: an essential regulator of cadherin stability, 78. Leushacke, M. et al. Lgr5‑expressing chief cells drive of intestinal identity and epithelial-mesenchymal
adhesion-induced signaling, and cancer progression. epithelial regeneration and cancer in the oxyntic signaling by Cdx2. Dev. Cell 16, 588–599 (2009).
Prog. Mol. Biol. Transl Sci. 116, 409–432 (2013). stomach. Nat. Cell. Biol. 19, 774–786 (2017). This study showed that conditional knockout of
57. Gutierrez-Gonzalez, L. & Wright, N. A. Biology of 79. Nam, K. T. et al. Spasmolytic polypeptide-expressing Cdx2 in the mouse intestine results in loss of
intestinal metaplasia in 2008: more than a simple metaplasia (SPEM) in the gastric oxyntic mucosa does intestinal identity and in squamous metaplasia.
phenotypic alteration. Dig. Liver Dis. 40, 510–522 not arise from Lgr5‑expressing cells. Gut 61, 99. Coskun, M., Troelsen, J. T. & Nielsen, O. H. The role
(2008). 1678–1685 (2012). of CDX2 in intestinal homeostasis and inflammation.
58. Evans, J. A. & McDonald, S. A. The complex, clonal, 80. Hayakawa, Y., Fox, J. G. & Wang, T. C. Isthmus stem Biochim. Biophys. Acta 1812, 283–289 (2011).
and controversial nature of Barrett’s esophagus. cells are the origins of metaplasia in the gastric corpus. 100. Gao, N. & Kaestner, K. H. Cdx2 regulates endo-
Adv. Exp. Med. Biol. 908, 27–40 (2016). Cell. Mol. Gastroenterol. Hepatol. 4, 89–94 (2017). lysosomal function and epithelial cell polarity.
59. Fitzgerald, R. C. et al. British Society of 81. Brembeck, F. H. et al. The mutant K‑ras oncogene Genes Dev. 24, 1295–1305 (2010).
Gastroenterology guidelines on the diagnosis and causes pancreatic periductal lymphocytic infiltration 101. Pan, F. C. et al. Spatiotemporal patterns of
management of Barrett’s oesophagus. Gut 63, 7–42 and gastric mucous neck cell hyperplasia in transgenic multipotentiality in Ptf1a‑expressing cells during
(2014). mice. Cancer Res. 63, 2005–2009 (2003). pancreas organogenesis and injury-induced facultative
60. Spechler, S. J. et al. A summary of the 2016 James W. 82. Lavery, D. L. et al. Evolution of oesophageal restoration. Development 140, 751–764 (2013).
Freston conference of the american gastroenterological adenocarcinoma from metaplastic columnar 102. Pin, C. L., Rukstalis, J. M., Johnson, C. &
association intestinal metaplasia in the esophagus and epithelium without goblet cells in Barrett’s Konieczny, S. F. The bHLH transcription factor Mist1
stomach: origins, differences, similarities and oesophagus. Gut 65, 907–913 (2016). is required to maintain exocrine pancreas cell
significance. Gastroenterology 153, e6‑e13 (2017). 83. Nicholson, A. M. et al. Barrett’s metaplasia glands are organization and acinar cell identity. J. Cell Biol. 155,
This article comprises a compendium of summaries clonal, contain multiple stem cells and share a 519–530 (2001).
from a conference on intestinal metaplasia in the common squamous progenitor. Gut 61, 1380–1389 103. Kopp, J. L. et al. Identification of Sox9‑dependent
oesophagus and stomach. (2012). acinar-to‑ductal reprogramming as the principal
61. Quante, M. et al. Bile acid and inflammation activate This is an example of a study in human tissues mechanism for initiation of pancreatic ductal
gastric cardia stem cells in a mouse model of Barrett- showing that Barrett metaplasia is clonally evolved adenocarcinoma. Cancer Cell 22, 737–750 (2012).
like metaplasia. Cancer Cell 21, 36–51 (2012). and contains multipotential stem cells and that In this article, the important role of SOX9 in
This article presents genetic in vivo lineage-tracing division may occur by fission. pancreatic ADM and PanIN is elucidated.
evidence for the cell of origin for Barrett‑like 84. Gutierrez-Gonzalez, L. et al. The clonal origins of 104. Reichert, M. et al. The Prrx1 homeodomain
metaplasia. dysplasia from intestinal metaplasia in the human transcription factor plays a central role in pancreatic
62. Wang, X. et al. Residual embryonic cells as precursors stomach. Gastroenterology 140, 1251–1260 (2011). regeneration and carcinogenesis. Genes Dev. 27,
of a Barrett’s‑like metaplasia. Cell 145, 1023–1035 85. McDonald, S. A. et al. Mechanisms of field 288–300 (2013).
(2011). cancerization in the human stomach: the expansion 105. Habbe, N. et al. Spontaneous induction of murine
63. Celli, J. et al. Heterozygous germline mutations in the and spread of mutated gastric stem cells. pancreatic intraepithelial neoplasia (mPanIN) by
p53 homolog p63 are the cause of EEC syndrome. Cell Gastroenterology 134, 500–510 (2008). acinar cell targeting of oncogenic Kras in adult mice.
99, 143–153 (1999). 86. Pan, Q. et al. Identification of lineage-uncommitted, Proc. Natl Acad. Sci. USA 105, 18913–18918
64. McDonald, S. A., Lavery, D., Wright, N. A. & long-lived, label-retaining cells in healthy human (2008).
Jansen, M. Barrett oesophagus: lessons on its origins esophagus and stomach, and in metaplastic 106. Means, A. L. et al. Pancreatic epithelial plasticity
from the lesion itself. Nat. Rev. Gastroenterol. Hepatol. esophagus. Gastroenterology 144, 761–770 (2013). mediated by acinar cell transdifferentiation and
12, 50–60 (2015). 87. McDonald, S. A., Graham, T. A., Lavery, D. L., generation of nestin-positive intermediates.
65. Lavery, D. L. et al. The stem cell organisation, and the Wright, N. A. & Jansen, M. The Barrett’s gland in Development 132, 3767–3776 (2005).
proliferative and gene expression profile of Barrett’s phenotype space. Cell. Mol. Gastroenterol. Hepatol. 1, 107. Zhu, L., Shi, G., Schmidt, C. M., Hruban, R. H.
epithelium, replicates pyloric-type gastric glands. Gut 41–54 (2015). & Konieczny, S. F. Acinar cells contribute to the
63, 1854–1863 (2014). 88. Liu, K. et al. Sox2 cooperates with inflammation- molecular heterogeneity of pancreatic intraepithelial
66. Vega, M. E. et al. Inhibition of Notch signaling mediated Stat3 activation in the malignant neoplasia. Am. J. Pathol. 171, 263–273 (2007).
enhances transdifferentiation of the esophageal transformation of foregut basal progenitor cells. 108. Miyamoto, Y. et al. Notch mediates TGF alpha-
squamous epithelium towards a Barrett’s‑like Cell Stem Cell 12, 304–315 (2013). induced changes in epithelial differentiation during
metaplasia via KLF4. Cell Cycle 13, 3857–3866 This study showed that SOX2 is critical for pancreatic tumorigenesis. Cancer Cell 3, 565–576
(2014). oesophageal and forestomach tissue identity (2003).
67. Minacapelli, C. D. et al. Barrett’s metaplasia develops and patterning. 109. Kawaguchi, Y. et al. The role of the transcriptional
from cellular reprograming of esophageal squamous 89. Giroux, V. et al. Long-lived keratin 15+ esophageal regulator Ptf1a in converting intestinal to pancreatic
epithelium due to gastroesophageal reflux. progenitor cells contribute to homeostasis and progenitors. Nat. Genet. 32, 128–134 (2002).
Am. J. Physiol. Gastrointest Liver Physiol. 312, regeneration. J. Clin. Invest. 127, 2378–2391 110. Miyatsuka, T. et al. Persistent expression of PDX‑1
G615–G622 (2017). (2017). in the pancreas causes acinar-to‑ductal metaplasia
68. Sarosi, G. et al. Bone marrow progenitor cells In this study, based on in vivo lineage tracing, a through Stat3 activation. Genes Dev. 20, 1435–1440
contribute to esophageal regeneration and metaplasia subset of oesophageal basal cells is characterized (2006).
in a rat model of Barrett’s esophagus. Dis. Esophagus as being long-lived progenitor cells that contribute 111. Halbrook, C. J. et al. Mitogen-activated protein kinase
21, 43–50 (2008). to tissue regeneration. kinase activity maintains acinar-to‑ductal metaplasia

NATURE REVIEWS | CANCER VOLUME 17 | OCTOBER 2017 | 603


©
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

and is required for organ regeneration in pancreatitis. 126. Ross-Innes, C. S. et al. Whole-genome sequencing acinar cell targeting of mutant Kras in transgenic
Cell. Mol. Gastroenterol. Hepatol. 3, 99–118 (2017). provides new insights into the clonal architecture mice. Cancer Res. 63, 2016–2019 (2003).
112. Shi, C. et al. KRAS2 mutations in human pancreatic of Barrett’s esophagus and esophageal 142. Guerra, C. et al. Chronic pancreatitis is essential for
acinar–ductal metaplastic lesions are limited to those adenocarcinoma. Nat. Genet. 47, 1038–1046 induction of pancreatic ductal adenocarcinoma by
with PanIN: implications for the human pancreatic (2015). K‑Ras oncogenes in adult mice. Cancer Cell 11,
cancer cell of origin. Mol. Cancer Res. 7, 230–236 A parallel study to Ref. 125 on deep DNA 291–302 (2007).
(2009). sequencing of Barrett oesophagus lesions. 143. Choi, E., Hendley, A. M., Bailey, J. M., Leach, S. D.
113. Lo, H. G. et al. A single transcription factor is 127. Silva, T. C. et al. hTERT, MYC and TP53 deregulation & Goldenring, J. R. Expression of activated ras in
sufficient to induce and maintain secretory cell in gastric preneoplastic lesions. BMC Gastroenterol. gastric chief cells of mice leads to the full spectrum
architecture. Genes Dev. 31, 154–171 (2017). 12, 85 (2012). of metaplastic lineage transitions. Gastroenterology
114. Capoccia, B. J. et al. The ubiquitin ligase Mindbomb 128. Yang, L. et al. Inflammation and intestinal metaplasia 150, 918–930 (2016).
1 coordinates gastrointestinal secretory cell of the distal esophagus are associated with 144. Schmidt, M. K. et al. c‑Myc overexpression is strongly
maturation. J. Clin. Invest. 123, 1475–1491 (2013). alterations in the microbiome. Gastroenterology associated with metaplasia-dysplasia-
115. Zhu, L. et al. Inhibition of Mist1 homodimer 137, 588–597 (2009). adenocarcinoma sequence in the esophagus.
formation induces pancreatic acinar-to‑ductal 129. Fukuda, A. et al. Stat3 and MMP7 contribute to Dis. Esophagus 20,
metaplasia. Mol. Cell. Biol. 24, 2673–2681 (2004). pancreatic ductal adenocarcinoma initiation and 212–216 (2007).
116. Weis, V. G. et al. Maturity and age influence chief progression. Cancer Cell 19, 441–455 (2011). 145. de Souza, C. R. et al. MYC deregulation in gastric
cell ability to transdifferentiate into metaplasia. 130. Petersen, C. P. et al. Macrophages promote cancer and its clinicopathological implications.
Am. J. Physiol. Gastrointest Liver Physiol. 312, progression of spasmolytic polypeptide-expressing PLoS ONE 8, e64420 (2013).
G67–G76 (2017). metaplasia after acute loss of parietal cells. 146. Lantuejoul, S., Salameire, D., Salon, C. &
117. Karki, A. et al. Silencing Mist1 gene expression is Gastroenterology 146, 1727–1738 (2014). Brambilla, E. Pulmonary preneoplasia—sequential
essential for recovery from acute pancreatitis. 131. Liou, G. Y. et al. Macrophage-secreted cytokines drive molecular carcinogenetic events. Histopathology 54,
PLoS ONE 10, e0145724 (2015). pancreatic acinar-to‑ductal metaplasia through NF‑κB 43–54 (2009).
118. Vasseur, S. et al. Structural and functional and MMPs. J. Cell Biol. 202, 563–577 (2013). 147. Hayakawa, Y., Sethi, N., Sepulveda, A. R., Bass, A. J.
characterization of the mouse p8 gene: promotion of 132. Liou, G. Y. & Storz, P. Inflammatory macrophages in & Wang, T. C. Oesophageal adenocarcinoma and
transcription by the CAAT-enhancer binding protein pancreatic acinar cell metaplasia and initiation of gastric cancer: should we mind the gap? Nat. Rev.
alpha (C/EBPalpha) and C/EBPbeta trans-acting pancreatic cancer. Oncoscience 2, 247–251 (2015). Cancer 16, 305–318 (2016).
factors involves a C/EBP cis-acting element and other 133. Liou, G. Y. et al. The presence of interleukin‑13 at 148. Hezel, A. F., Kimmelman, A. C., Stanger, B. Z.,
regions of the promoter. Biochem. J. 2, 377–383 pancreatic ADM/PanIN lesions alters macrophage Bardeesy, N. & Depinho, R. A. Genetics and biology of
(1999). populations and mediates pancreatic tumorigenesis. pancreatic ductal adenocarcinoma. Genes Dev. 20,
119. Zenilman, M. E., Tuchman, D., Zheng, Q., Levine, J. Cell Rep. 19, 1322–1333 (2017). 1218–1249 (2006).
& Delany, H. Comparison of reg I and reg III levels 134. Kong, J. et al. Immature myeloid progenitors 149. Orloff, M. et al. Germline mutations in MSR1, ASCC1,
during acute pancreatitis in the rat. Ann. Surg. 232, promote disease progression in a mouse model of and CTHRC1 in patients with Barrett esophagus and
646–652 (2000). Barrett’s‑like metaplasia. Oncotarget 6, esophageal adenocarcinoma. JAMA 306, 410–419
120. Ramsey, V. G. et al. The maturation of mucus- 32980–33005 (2015). (2011).
secreting gastric epithelial progenitors into digestive- 135. Liu, X. et al. Genetic ablation of Smoothened in 150. Sun, X. et al. Linkage and related analyses of Barrett’s
enzyme secreting zymogenic cells requires Mist1. pancreatic fibroblasts increases acinar–ductal esophagus and its associated adenocarcinomas.
Development 134, 211–222 (2007). metaplasia. Genes Dev. 30, 1943–1955 (2016). Mol. Genet. Genom. Med. 4, 407–419 (2016).
121. Nomura, S. et al. Alterations in gastric mucosal In this study, the importance of SHH signalling 151. Pittayanon, R. et al. The risk of gastric cancer in
lineages induced by acute oxyntic atrophy in wild- from stromal fibroblasts in pancreatic ADM is patients with gastric intestinal metaplasia in 5‑year
type and gastrin-deficient mice. Am. J. Physiol. demonstrated. follow‑up. Aliment. Pharmacol. Ther. 46, 40–45
Gastrointest Liver Physiol. 288, G362–G375 136. Pasca di Magliano, M. et al. Hedgehog/Ras (2017).
(2005). interactions regulate early stages of pancreatic
122. Kaz, A. M., Grady, W. M., Stachler, M. D. & cancer. Genes Dev. 20, 3161–3173 (2006). Acknowledgements
Bass, A. J. Genetic and epigenetic alterations in 137. Wang, D. H. et al. Aberrant epithelial-mesenchymal This work was funded in part through the National Cancer
Barrett’s esophagus and esophageal Hedgehog signaling characterizes Barrett’s Institute and the American Cancer Society. The authors apol-
adenocarcinoma. Gastroenterol. Clin. North Am. 44, metaplasia. Gastroenterology 138, 1810–1822 ogize in advance if all relevant references were not able to
473–489 (2015). (2010). be cited.
123. Kaz, A. M. et al. DNA methylation profiling in 138. Konstantinou, D., Bertaux-Skeirik, N. & Zavros, Y.
Barrett’s esophagus and esophageal adenocarcinoma Hedgehog signaling in the stomach. Curr. Opin. Author contributions
reveals unique methylation signatures and molecular Pharmacol. 31, 76–82 (2016). A.K.R. and V.G. researched the data for the article, wrote the
subclasses. Epigenetics 6, 1403–1412 (2011). 139. Rustgi, A. K. Pancreatic fibroblasts smoothen their article and reviewed and/or edited the manuscript before its
124. Buas, M. F. et al. Germline variation in inflammation- activities via AKT‑GLI2‑TGFα. Genes Dev. 30, submission. A.K.R. made substantial contributions to the
related pathways and risk of Barrett’s oesophagus 1911–1912 (2016). discussions of the content.
and oesophageal adenocarcinoma. Gut http://dx.doi. 140. Pitarresi, J. R. et al. Stromal ETS2 regulates
org/10.1136/gutjnl-2016-311622 (2016). chemokine production and immune cell recruitment Competing interests statement
125. Stachler, M. D. et al. Paired exome analysis of during acinar-to‑ductal metaplasia. Neoplasia 18, The authors declare no competing interests.
Barrett’s esophagus and adenocarcinoma. Nat. 541–552 (2016).
Genet. 47, 1047–1055 (2015). 141. Grippo, P. J., Nowlin, P. S., Demeure, M. J., Publisher’s note
In this study, the important role of mutant TP53 in Longnecker, D. S. & Sandgren, E. P. Preinvasive Springer Nature remains neutral with regard to jurisdictional
Barrett oesophagus is demonstrated. pancreatic neoplasia of ductal phenotype induced by claims in published maps and institutional affiliations.

604 | OCTOBER 2017 | VOLUME 17 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