Sei sulla pagina 1di 13

Gastroenterology 2015;149:1153–1162

Genetics and Molecular Pathogenesis of Gastric


Adenocarcinoma

GENOMICS
Patrick Tan1,2,3,4,7 Khay-Guan Yeoh5,6,7

1
Cancer and Stem Cell Biology Program, Duke-National University of Singapore Graduate Medical School; 2Genome Institute
of Singapore, Agency for Science, Technology, and Research; 3Cancer Science Institute of Singapore, National University of
Singapore; 4Cellular and Molecular Research, National Cancer Centre Singapore; 5Department of Medicine, Yong Loo Lin
School of Medicine, National University of Singapore; 6Department of Gastroenterology and Hepatology, National University
Health System; and 7Singapore Gastric Cancer Consortium, Singapore

Gastric cancer (GC) is globally the fifth most common were simultaneously profiled on multiple molecular plat-
cancer and third leading cause of cancer death. A complex forms to identify 4 genomic subtypes: CIN (chromosomal
disease arising from the interaction of environmental and instability), MSI (microsatellite instability), GS (genome
host-associated factors, key contributors to GC’s high stable), and EBV (Epstein-Barr virus).3 The comprehensive
mortality include its silent nature, late clinical presenta- nature of the TCGA study provides an invaluable resource
tion, and underlying biological and genetic heterogeneity. upon which to interpret other related GC findings.
Achieving a detailed molecular understanding of the
various genomic aberrations associated with GC will be
critical to improving patient outcomes. The recent years
has seen considerable progress in deciphering the genomic
Gastric Premalignancy and the Critical
landscape of GC, identifying new molecular components Role of Inflammation
such as ARID1A and RHOA, cellular pathways, and tissue The human stomach consists of the fundus, corpus or
populations associated with gastric malignancy and pro- body, and pyloric antrum. The gastric mucosa contains three
gression. The Cancer Genome Atlas (TCGA) project is a main types of glands: cardiac glands (containing mucus-
landmark in the molecular characterization of GC. Key producing foveolar cells), oxyntic glands (parietal cells and
challenges for the future will involve the translation of chief cells producing hydrochloric acid and pepsinogen
these molecular findings to clinical utility, by enabling respectively), and pyloric glands with mucus–secreting cells
novel strategies for early GC detection, and precision and endocrine G cells secreting gastrin. Chronic atrophic
therapies for individual GC patients. gastritis and intestinal metaplasia (IM) involving the gastric
mucosa are considered important steps in GC pathogenesis
(Figure 1).4 Mucosal atrophy is characterized by loss of
Keywords: Cancer Genetics; Cancer Genomics; Gastric Cancer.
glandular elements and replacement by metaplastic cells or
fibrosis, with concomitant hypochlorhydria. IM is a pre-
neoplastic lesion characterized by transformation of the
gastric mucosa into an intestinal-like phenotype, replete
G astric cancer (GC) is currently the third leading
cause of global cancer-related death, and particu-
larly prevalent in Asia. Despite a steadily declining inci-
with goblet cells and intestinal mucins. IM, associated with

dence, GC still causes more than 723,000 deaths a year.1


Abbreviations used in this paper: CIMP, CpG island methylation pheno-
Achieving a detailed molecular understanding of GC patho- type; CIN, Chromosomal instability; EBV, Epstein-Barr virus; EMT,
genesis is pivotal to improving patient outcomes for this epithelial-mesenchymal transition; GC, gastric cancer; GS, genome sta-
ble; Helicobacter pylori, H pylori; HDGC, hereditary diffuse gastric cancer;
complex disease. This is clearly exemplified by the TOGA IM, intestinal metaplasia; lincRNAs, long intergenic noncoding RNAs;
Phase III trial, where GC patients with HER2/neu receptor miRNAs, microRNAs; MSI, microsatellite instability; NGS, next-generation
sequencing; sCNAs, somatic copy number alterations; TFs, transcription
tyrosine kinase (RTK)-positive tumors experienced clinical factors; TCGA, The Cancer Genome Atlas.
benefit from chemotherapy plus trastuzumab (a HER2-
targeting antibody) relative to chemotherapy alone.2 In Most current article
this review, we have attempted to capture the latest ad-
© 2015 by the AGA Institute
vances in GC molecular genetics. Special attention is paid to 0016-5085/$36.00
the recent landmark TCGA study, where almost 300 GCs http://dx.doi.org/10.1053/j.gastro.2015.05.059
1154 Tan and Yeoh Gastroenterology Vol. 149, No. 5
GENOMICS

Figure 1. Cause and pathogenesis of intestinal-type GC. A summary of current knowledge of the cause and pathogenesis of
intestinal type GC is shown, including host and environmental factors as well as acquired molecular events.52,143–145 GC,
gastric cancer.

over-expression of the homeobox transcription factor and smoking.10 High salt intake, often due to traditional
CDX2,5 is currently classified into three subtypes: intestinal diets containing salted fish, is the best documented dietary
type, gastric type, and mixed gastric-intestinal type. The risk factor for atrophic gastritis.11 Recent data also suggests
latter, also known as incomplete IM, is considered to confer that iron deficiency may be a GC risk factor, as iron deple-
the highest risk for GC development. Injury to the gastric tion can accelerate the progression of carcinogenesis by
mucosa has also been observed to cause metaplastic augmenting H pylori virulence.12 EBV infection is recognized
changes with spasmolytic peptide expression. This phe- as an etiological agent in 5%–10% of GCs.13,14
nomena, termed spasmolytic peptide expressing metaplasia H pylori is the most significant environmental risk factor
(SPEM) or trefoil factor family 2 (TFF2) expressing meta- for GC and is recognized as a class I carcinogen by the
plasia, is induced after Helicobacter pylori (H pylori) infec- World Health Organization. Although more than 50% of the
tion and chronic gastritis.6 Spasmolytic peptide expressing world population is infected with H pylori, only 1%–2% of
metaplasia has also been implicated as a precursor event in infected people will develop GC in their lifetime.15 A major
GC progression.7 H pylori virulence factor is the cytotoxin-associated gene A
In most patients, frank GC is often preceded by several (CagA), within the bacterial cag pathogenicity island (cag-
decades of chronic gastric mucosal inflammation. Chronic PAI) which is injected into the cytoplasm of gastric
inflammation activates the NF-kB transcription factor, a key epithelial cells upon microbial colonization.16 In transgenic
mediator of tumor promotion.8 Chronic inflammation also mice, systemic expression of CagA has been shown to
causes increased oxidative stress, due to reactive oxygen induce gastric epithelial hyperplasia, gastric polyps, gastric
species and nitrosamines generated by leukocytes and mac- and intestinal adenocarcinomas.17 Genetic variations in
rophages which can damage proliferating cells. Moreover, the CagA, specifically present in Asian strains but not non-Asian
production of chemokines and cytokines may induce not only strains, are also associated with increased chronic gastritis,
leukocyte migration but also promote carcinogenesis. Exper- gastric ulcer and gastric adenocarcinoma in human pa-
imentally, mice with impaired immune systems such as severe tients.18 In gastric epithelial cells, CagA undergoes tyrosine
combined immune deficiency (SCID) or recombinase phosphorylation by Src kinase and activates SHP-2 (Src
activating gene (RAG2)-deficient mice are very susceptible to homology 2- containing tyrosine phosphatase). Activated
H pylori infection, but do not develop significant gastric dis- SHP-2 induces the Ras-ERK pathway, a key regulator of cell
ease.9 Such data further supports an important functional role growth, migration, and adhesion.19 CagA also disrupts tight
of the host immune system in GC pathogenesis. junctions20 and can target the PAR1/MARK kinase to alter
apical-basolateral cell polarity, ultimately causing disorga-
nization of the gastric mucosal architecture.21 CagA also has
Cause and Epidemiology of Gastric tyrosine phosphorylation-independent functions, including
Cancer interactions with the Met tyrosine kinase and E-cadherin.
Environmental factors play critical roles in GC patho- This latter interaction disrupts binding between E-cadherin
genesis, with major risk factors being H pylori infection, diet and b-catenin, leading to nuclear accumulation of b-catenin
October 2015 Pathogenesis of Gastric Adenocarcinoma 1155

and subsequent activation of b-catenin-dependent Furthermore, a combination of high-risk host genotypes


transcription.22 and high-risk H pylori genotypes greatly increased the
Several clinical studies have evaluated the utility of H probability of GC, up to 87-fold over baseline. These studies
pylori eradication for GC prevention. Randomized trials in highlight again the critical role of host immunity in deter-
high risk populations have suggested that H pylori eradica- mining GC outcomes following H pylori infection.36 Certain
tion is most effective in patient subgroups where prema- high risk GC genotypes may differ in different populations,
lignant lesions have not yet developed.23 For example, a H for example, a meta-analysis showed Asian high risk SNPs

GENOMICS
pylori eradication program in Matsu Island, Taiwan showed differ from those identified in the non-Asian population.37
that eradication reduces the incidence of atrophic gastritis Another identified host genetic factor for GC is prostate
and peptic ulcer but not intestinal metaplasia.24 In a meta- stem cell antgen (PSCA), which influences susceptibility to
analysis of 6 randomized controlled trials, H pylori eradi- diffuse-type GC and may play a role in gastric epithelial cell
cation reduced the risk of developing GC by RR of 0.65 (95% proliferation.38 PSCA genetic variants have also been
CI, 0.43–0.98).25 In Japan, which has high rates of GC recently confirmed as influencing GC in a wide variety of
prevalence and H pylori infection, the national health in- populations.39
surance system approved eradication therapy for H pylori- Hereditary GC syndromes are rare and account for 1-3%
related chronic gastritis in 2013.26 of GC, comprising mainly three types: hereditary diffuse
Smoking has been associated with a 1.5–2.5 fold gastric cancer (HDGC), gastric adenocarcinoma and prox-
increased risk of GC in both case-control and cohort studies, imal polyposis of the stomach (GAPPS) and familial intesti-
and this risk increases with the frequency and duration of nal gastric cancer (FIGC).40 HDGC is due to E-cadherin
smoking. Studies have reported that smokers have higher (CDH1) germline mutations,41 and patients with this con-
hazard ratios (HR) of GC in the cardia (HR, 2.86–4.10) dition are often managed with prophylactic gastrectomies.41
compared to distal portions of the stomach (HR, Besides CDH1, other recently identified candidate HDGC
1.52–1.94).27 Carcinogens in tobacco smoke, notably nitro- genes include CTNNA1, BRCA2 and STK11.42 GAPPS, first
samines and other nitroso compounds, may exert mutagenic reported in 2012, is characterized by autosomal dominant
effects, thereby increasing GC risk.28 Smoking also increases transmission of fundic gland polyposis (including dysplastic
the risk for precancerous lesions such as intestinal meta- lesions or adenocarcinoma or both) restricted to the prox-
plasia and dysplasia.29 imal stomach with no evidence of colorectal/duodenal pol-
EBV has been detected in 5%–10% of GC.13,14 A meta- yposis or other hereditary gastrointestinal cancer
analysis of 70 studies including 15,952 cases of GC syndromes.43 Its genetic cause is as yet unknown. GC has
demonstrated that EBV-associated GC predominantly arises also been associated with other hereditary cancer syn-
in men and is found in cardia and body sites more often dromes such as Lynch syndrome, hereditary nonpolyposis
than in the antrum.14 A major molecular feature of EBV- colorectal cancer syndrome, familial adenomatous polypo-
associated GCs is CpG island promoter methylation of sis, and Li-Fraumeni syndrome.40
cancer-related genes.30 Expression of EBV viral latent
membrane protein 2A (LMP2A) may be responsible for the
promotion of DNA methylation, by inducing STAT3 phos- Gastric Cancer Pathology
phorylation and subsequent transcription of DNA methyl- GC is morphologically, biologically, and genetically het-
transferase 1 (DNMT1). EBV also encodes a large number of erogeneous. Most GCs are adenocarcinomas, with lym-
microRNAs (miRNAs),31 mostly encoded in the BHRF1 and phoma, sarcomas, and carcinoids comprising less than 5%.
BART regions of the genome. EBV miRNAs can repress Adenocarcinomas are grouped by the Lauren classification
cellular proteins, including p53 up-regulated modulator of into intestinal and diffuse types.44,45 In intestinal type tu-
apoptosis (PUMA), DICER1, and BIM.32 A recent study mors, the malignant cells form gland-like structures, unlike
profiling 44 known EBV miRNAs in clinical samples from those in diffuse type tumors. Intestinal type GC is more
EBV-associated GC patients found that EBV-miR-BART4-5p common in high-risk regions while diffuse type GC is more
plays an important role in gastric carcinogenesis through common in low-risk areas. Diffuse type GC is more common
regulation of apoptosis.33 in young patients, in whom there is a female preponder-
ance,45 and behaves more aggressively than the intestinal
type.46 Besides the Lauren classification, other histopatho-
Gastric Cancer Host Genetics logical classifications for GC have been proposed. For
Besides environmental agents, GC pathogenesis also in- example, the WHO classification groups GC into 4 main
volves host genetic factors. One of the first host genetic types based on the predominant histological pattern:
factors identified in GC involved polymorphisms in the tubular, papillary, mucinous and signet ring cell.47
proinflammatory gene interleukin 1-b (IL-1 b).34 A more
recent study reported that combinations of single nucleotide
polymorphisms (SNPs) in immune-related genes [inter- Molecular Genetic Landscape of
leukin 1-b (IL-1 b), interleukin 1 receptor antagonist Gastric Cancer
(IL-1RN), tumor necrosis factor- a (TNF- a) and interleukin Studies from several groups over the past decade have
10 (IL-10)] conferred a manifold increased risk of devel- now produced a near-comprehensive catalogue of GC-
oping GC, but only in H pylori infected patients.35 associated “driver” alterations, including gene mutations,
1156 Tan and Yeoh Gastroenterology Vol. 149, No. 5
GENOMICS

Figure 2. Genomic fea-


tures of GC, inspired by
the classic Hallmarks of
Cancer report by Hanahan
and Weinberg.146 Details
of each genomic feature
are provided in the text.
GC, gastric cancer.

somatic copy number alterations (sCNAs), structural vari- chromosomal regions and focal gene regions. GCs have
ants, epigenetic changes, and transcriptional changes also been shown to exhibit mutations in other canonical
involving mRNAs and noncoding RNAs (ncRNAs) (Figure 2). oncogenes (KRAS, CTNNB1, PIK3CA) and tumor suppressor
Certain driver alterations can also be associated with spe- genes (SMAD4, APC).52 Reflecting the importance of RTK/
cific GC subtypes. For example, MSI is a genetic hypermu- RAS/MAPK signaling in GC, frequent mutations in the
tability phenomenon where microsatellite regions in tumor ERBB3 RTK and the ligand/RTK NRG1/ERBB4 genes have
genomes accumulate mutations due to defective DNA recently been reported.53,54 Some of these mutations
mismatch repair. Next-generation sequencing (NGS) studies appear to be enriched in specific GC subtypes, for example,
have shown that 15%–20% of GCs are characterized by MSI. EBV-positive GCs frequently exhibit PIK3CA mutations,3
For ease of presentation, our review has been structured to while diffuse-type GCs have been observed to exhibit
summarize each molecular level independently. frequent somatic mutations in CDH1.55
Recent NGS studies of GC have also highlighted two new
GC genes - ARID1A and RHOA. ARID1A, mutated in 10%–15%
Gene Mutations in Gastric Cancer of GCs,56,57 encodes a component of the SWI/SNF chromatin
Like most solid epithelial cancers, GCs are often char- remodeler complex. ARID1A mutations in GC are typically
acterized by somatically-acquired mutations in various inactivating (eg, frameshifts, nonsense mutations), consistent
genes. Current estimates based on NGS indicate that in each with ARID1A acting as a tumor suppressor. Functional anal-
individual GC (excluding hypermutated cases), there are ysis suggests that ARID1A plays a role in GC cell proliferation,
approximately 50 to 70 nonsynonymous mutations, a mu- through the control of cell-cycle regulators CCNE1 and
tation level comparable to colon and esophageal cancer.48 E2F1.57 RHOA was recently shown by multiple studies to
Mutated genes in GC can be broadly classified into three exhibit recurrent mutations in diffuse-type/genome-stable
categories: a) high-frequency drivers displaying a high-rate GCs.3,55,58 Unlike ARID1A where mutations are dispersed
of recurrence (>5%–10%) across multiple GCs; b) throughout the gene, the RHOA mutations are localized to an
low-frequency drivers that are recurrently mutated in the N-terminal hot-spot region (Tyr42, Arg5 and Gly17), and are
1%–10% range, but which still contribute to disease path- predicted to modulate downstream Rho signaling. Functional
ogenesis; and c) bystander/passenger mutations that arise analysis of these RHOA mutations suggest that they may
as a consequence of underlying mutational processes such impart resistance to anoikis, a form of programmed cell death
as CpG deamination,49 but which do not functionally occurring after cellular detachment from a solid substrate.55
contribute to tumorigenesis. Clinically, the discovery of RHOA hot-spot mutations is
Among high-frequency drivers, TP53 is the most particularly exciting, as it provides an inroad for the devel-
frequently mutated gene in GC, exhibiting aberrations in opment of new approaches to target diffuse-type GCs, tradi-
w50% of cases.50 Reflecting TP53’s cellular function as a tionally associated with extremely poor prognosis.
guardian of genomic integrity, TP53 mutated GCs often Besides high-frequency driver mutations, genomic
exhibit high levels of sCNAs3,51 involving both broad studies have also uncovered a “long tail” of low frequency
October 2015 Pathogenesis of Gastric Adenocarcinoma 1157

driver mutations in GC, such as the gastric mucin MUC6, checkpoint inhibitor genes, and are of particular relevance
BCOR (encoding a BCL6 corepressor), FAT4 (a proto- as targets of immunotherapy.3
cadherin), and RNF43, a Wnt pathway regulator.3,55,57 Another class of genes targeted by gene amplification in
Despite their lower mutation frequencies, these genes also GC are related to cell cycle control, including CCND1, CCNE1,
likely contribute to the ability of GCs to manifest different and CDK6. These cell-cycle genes are clinically relevant in
cancer hallmarks. For example, mutations in the antigen- two capacities. First, direct therapies are available for tar-
presenting genes HLA-B and B2M, observed in MSI- gets such as CDK6, while CCND1 and CCNE1, which encode

GENOMICS
positive GCs, may play a role in evading host anti-tumor cyclin subunits, may also be targetable through their
immune responses arising from the presentation of tumor cognate CDK partners (CDK4/6 for CCND1, CDK1/2 for
neo-antigens.3 A key challenge for the GC field will be to CCNE1). Second, coamplification of these cell-cycle regula-
develop systematic, high-throughput approaches to func- tors with other therapeutic targets, particularly those in the
tionally test the effects of these mutations, either singly or in RTK/RAS/MAPK pathway, may modulate responses to the
combination, on the GC cell. therapies targeting the latter. For example, CCNE1 amplifi-
NGS analyses of GCs, particularly at the whole-genome cations frequently occur together with HER2 amplifica-
level, are also providing insights into prominent pathways tions,67,70 a configuration that has been linked to acquired
and mutation signatures associated with GC develop- trastuzumab resistance in breast cancer.71 HER2/CCNE1
ment.55,59,60 For example, when all mutated genes (regard- coamplification has recently been validated as a mechanism
less of driver status) are considered in a collective pathway of primary resistance against lapatinib (a dual EGFR/HER2
analysis, cell adhesion and chromatin remodeling are inhibitor) in HER2-amplified GC.70 Knowledge of the
consistently highlighted as the top major disrupted path- amplification status of these cell-cycle related genes in in-
ways.56,57 Emerging evidence suggests that these processes dividual GCs may thus further improve our ability to stratify
are likely to be disrupted even in premalignant gastric tis- patients in clinical trials employing targeted agents.
sues and early GC.61,62 A survey of mutational “signatures”, A third class of GC-amplified genes corresponds to those
referring to mutations occurring in particular sequence involved in transcriptional regulation, such as GATA factors
contexts, has highlighted specific carcinogenic and muta- (GATA4, GATA6), KLF5, and MYC. Amplifications of GATA
tional processes active in GC, including microsatellite factors and KLF5 appear to be restricted to gastrointestinal,
instability, CpG age-associated deamination, and the acti- and possibly related hepatobiliary malignancies.72,73 These
vation of cytidine deaminases such as AID and transcription factors (TFs) are expressed in the developing
APOBEC3B.63–65 Emerging studies are now being performed and normal stomach,74 and may act as “lineage-survival”
to study patterns of GC tumor evolution and clonality, factors functioning to reawaken early developmental pro-
occurring either within the primary tumor, or between grams to drive GC tumorigenesis. Interestingly, recent
primary lesions and metastatic sites.54,66 Such results will chromatin immunoprecipitation (ChIP) sequencing studies
be crucial for identifying molecular events associated with provide evidence that these amplified TFs, may in fact
GC progression rather than initiation, and may suggest collaborate with one another to regulate common down-
improved strategies for managing patients with advanced stream promoters in genes associated with GC develop-
metastatic GC. ment.73 Another transcription factor amplified in GC is
OCT1, a regulator of ERK signaling.75 Although traditionally
considered to be “undruggable” by the pharmaceutical in-
sCNAs and Structural Variation in dustry, novel chemical biology technologies for disrupting
Gastric Cancer such oncogenic TFs are in development.76
sCNAs are another major mechanism by which onco- Besides amplifications, genomic deletions also
genes and tumor suppressor genes are selectively activated frequently occur in GC, involving tumor suppressor genes
or inactivated in GC. Several studies have reported genome- such as WWOX, RB1, PARK2, FHIT, and CDKN2A/B.67 So-
scale analyses to identify GC genes affected by sCNA,67,68 matic deletions in CDH1 have been associated with poor
highlighting specific genes targeted by this mechanism. patient prognosis,77 and genomic deletions involving
The most predominant class of genes frequently amplified nonprotein coding genes such as mir-101a have been re-
in GC are those related to RTK/RAS/MAPK signaling, ported, which can cause upregulation of the oncogenic his-
including HER2, EGFR, MET, FGFR2, and RAS genes (KRAS, tone methyltransferase EZH2.78 Interestingly, while most
and to a lesser extent NRAS).3,67 In total, about 30%–40% of genomic deletions are often interpreted as loss-of-function
GCs are likely to harbor a RTK/RAS/MAPK-related amplifi- events, a recent study has shown that the phosphodies-
cation, and therapies targeting these genes are in clinical terase PDE4D, frequently targeted by microdeletions in
trials, including trastuzumab (HER2), nimotuzumab (EGFR), several cancers including GC, is associated with a constitu-
AZD4547 (FGFR2), and onartuzumab (MET) (Table 1). tively active PDE4D isoform that promotes tumor growth.79
Another gene frequently amplified in GC that may intersect It is thus possible that, as sequencing technologies improve,
with the canonical RTK/RAS/MAPK pathway is VEGFA, a detailed fine mapping of genomic deletion break-points will
mediator of angiogenesis that is particularly noteworthy highlight similar unexpected examples, prompting the
given the role of anti-angiogenic therapy in GC.69 In the revisiting of these “deleted” genes as tumor suppressors.
TCGA study, EBV-positive GCs were also found to harbor Genomic rearrangements, either balanced or associated
frequent amplifications in PD-L1 and L2, which are immune with copy number changes, can also result in fusion genes,
1158 Tan and Yeoh Gastroenterology Vol. 149, No. 5

Table 1.GC Genomic Alterations as Therapeutic Targets

Gene Alteration Prevalence in GCa Representative Therapies Clinical Statusb

ERRB2 Amplification/overexpression 10%–20% Traztuzumab/trastuzumab Approved/Phase III


emtansine
VEGFR2 Overexpression w50% Ramucirumab Approved
VEGF Overexpression 40%–50% Bevacizumab/regorafenib Phase III (negative)/phase II
GENOMICS

EGFR Amplification/overexpression 6%–27% Cetuximab/nimotuzumab Phase II/III


MET Amplification/overexpression 5%–40% Onartuzumab/AMG337 Phase II/III
FGFR2 Amplification/overexpression 4%–12% AZD4547/dovitinib Phase II
ATM Loss (Protein) 60% Olaparib Phase III
PIK3CA Mutation 5%–10% Everolimus/GDC0068 Pre-clinical/early trials
CDK4/6 Amplification 6%–15% LEE001 Phase II
PD-L1/L2 Amplification/overexpression 15% of EBV-positive GC MPDL3280A Preclinical/phase I
MSI Mutation 15%–20% Pembrolizumab (anti-PD1) Phase II
ARID1A Mutation 8%–10% EZH2 inhibition Preclinical

GC, gastric cancer; EBV, Epstein-Barr virus; MSI, microsatellite Instability.


a
Prevalence rates incorporate data based on both genomic alterations and protein immunohistochemistry.
b
Most ongoing trials do not involve biomarker-selected GC patients.

creating new chimeric protein products (eg, BCR-ABL specific residues in histones are methylated, acetylated, or
in chronic myelogenous leukemia), or causing high expression phosphorylated. There is great interest in studying how
of one gene through hijacking of the partner’s promoter (eg, environmental risk factors for GC may modulate the gastric
TMPRRS2-ERG in prostate cancer). An RNA-sequencing study epigenome. For example, H pylori has been shown to infect
in 2010 revealed for the first time the presence of rare RAF- and induce widespread DNA methylation changes in stom-
fusion genes in GC and prostate cancers (eg, AGTRAP-BRAF), ach epithelial cells.86 Such “epigenetic field defects” are
and preclinical models expressing these RAF fusions were likely to contribute to an individual’s risk of developing GC.
shown sensitive to treatment by sorafinib, originally designed The epigenetic field model has recently been supported by a
as a RAF inhibitor.80 The second fusion gene identified in GC recent prospective clinical trial.87
was CD44-SLC1A2, which juxtaposes the SLC1A2 glutamate There is a rich body of GC literature relating promoter
transporter against the CD44 promoter.81 Tumors expressing DNA methylation to the transcriptional silencing of tumor
this gene fusion expressed high levels of SLC1A2 which may suppressor genes (CDH1, RUNX3, p16, and hMLH1).88
facilitate the acquisition of glutamate by the cancer cell to fuel Alterations in DNA methylation can also influence GC pro-
cancer metabolism. Subsequently, CD44-SLC1A2 and related gression and treatment response. For example, methylation-
APIP-SLC1A2 fusions have been observed in colon cancer, associated silencing of PLA2G2A, a secreted phospholipase,
indicating that such fusions may be restricted to may play a role in driving aggressive GC,89 and methylation
gastrointestinal-type cancers.82,83 ROS1 rearrangements in GC of SULF2 and BMP4 have been linked to chemotherapeutic
have been recently reported,84 and in the TCGA study, responses.90,91 Beyond individual genes, genome-wide
CLDN18-ARHGAP26 fusions were observed in genome-stable/ studies have also revealed large scale patterns of DNA
diffuse type GC. Interestingly, ARHGAP26 is a member of the methylation in GC. Multiple studies have confirmed that
Rho-effector pathway, and the CLN18-ARHGAP26 fusions are certain GCs can exhibit a CpG island methylation phenotype
mutually exclusive to RhoA mutations and CDH1 mutations, all (CIMP), characterized by genome-wide methylation of CpG
pointing to a common dependence on cell-adhesion related islands.92–94 Patients with CIMP type GCs tend to be
Rho signaling in diffuse-type GC.3 Indeed, recent functional younger, with less-differentiated tumors. Pathway analysis
data has confirmed that CLDN18-ARHGAP26 expression in reveals that genes targeted by CIMP in GC are enriched in
epithelial cells can reduce cell-cell and cell-extracellular ma- genes corresponding to PRC2-targets in embryonic stem
trix-adhesion, while contributing to epithelial-mesenchymal cells, suggesting a reawakening in CIMP tumors of a nascent
transition (EMT).85 Taken collectively, these examples attest stem cell program. Interestingly, in preclinical assays, CIMP-
to the presence of gene fusions in GC, which due to their positive GCs appear to exhibit heightened sensitivity to DNA
cancer-specific nature, may be exploited for improved di- methyltransferase inhibitors such as 5’aza.93
agnostics or therapeutics. As described by earlier studies and confirmed by TCGA,
EBV-positive GCs also exhibit exceptionally high DNA
methylation levels,3,30 Indeed, among all tumor types stud-
Epigenetic Alterations involving DNA ied by TCGA to date, EBV-positive GCs appear to exhibit the
Methylation and Chromatin highest DNA methylation levels, surpassing even CIMP tu-
Epigenetic aberrations are also emerging as important mors. It is possible that such hypermethylation may repre-
players in GC pathogenesis. These can occur at the level of sent a cellular reaction to viral infection. Emphasizing
DNA methylation, where methyl groups are attached to the importance of tumor immunity in this GC subtype,
cytosine bases at CG motifs; or histone modifications where EBV-positive GCs also typically exhibit a high lymphocytic
October 2015 Pathogenesis of Gastric Adenocarcinoma 1159

infiltrate,95 and (as described above) also frequently exhibit II GC patients.105 This is a highly clinically-relevant ques-
PD-L1 and PD-L2 amplification. Another recent study has tion, as Stage II patients identified as “poor prognosis” by
recently reported additional mutated genes in EBV-positive the gene signature might be prescribed adjuvant chemo-
GC, which may contribute to tumor neo-antigen formation in therapy, while “good prognosis” patients might be treated
this unique and intriguing cancer subtype.96 with surgery alone.
In contrast to DNA methylation, our understanding of Transcriptomic information can also be exploited as a
histone modifications in GC is still embryonic. An earlier powerful discovery method to identify deregulated path-

GENOMICS
study assessing patterns of the H3K37me3 repressive mark ways in GC. One study exploring this approach reported that
found over a hundred genes exhibiting H3k27me3 differ- GCs exhibiting combinations of activated oncogenic path-
ences between GCs and matched normal tissues.97 More ways were associated with poor prognosis relative to GCs
recently, a study employing a nano-scale chromatin immu- with activation of single pathways.106 Investigators have
noprecipitation techniques to profile multiple histone marks also used RNA-sequencing data of GCs from different
in GCs and matched normal tissues,98 found evidence of countries to identify the AMPK/HNF4a/Wnt5a pathway as
widespread alternative promoter usage and deregulated a targetable oncogenic pathway in early stage GC.107,108
enhancer elements, including an alternative isoform of the However, it should be noted that while transcriptomes are
MET RTK that produces a MET protein variant lacking the highly dynamic, almost all GC transcriptomic studies to date
SEMA N-terminal regulatory domain. Future work will be have relied upon analyzing GCs obtained at a single time-
focused on understanding the mechanisms underlying such point, usually upon surgical resection. There is thus great
regulated regulatory elements, and how they might be interest in understanding gene expression (and other mo-
harnessed for predictive or prognostic potential. lecular) changes in GCs during temporal progression or
treatment. Demonstrating the power of this temporal
approach, one highly notable study used paired endoscopic
Gastric Cancer Transcriptome—Gene biopsies to study gene expression changes in GC patients
Signatures and Alternative Splicing prior to and after developing resistance to 5-FU/platinum
Altered regulation of gene expression programs are therapies.109 They identified an acquired-resistance signa-
critical to the ability of tumors to express different cancer ture comprising genes related to cell survival (mTOR
hallmarks. In early 2000, early microarray studies already pathway), DNA repair, and embryonic stem cell biology.
revealed a staggering diversity of expression programs Data from such efforts, while still scanty in the literature,
operative in GC, associated with different histological sub- will prove vital in attempts to overcome chemotherapy
types, tumor status, and clinical traits.99–101 In the past resistance in GC.
decade, groups have deployed more mature transcriptomic Transcriptomic analysis is also revealing new molecular
technologies, on larger patient cohorts, to identify robust features of GC biology, above and beyond those provided
gene expression “signatures” that can subtype GC in a through the study of DNA. One especially exciting field of
clinically relevant fashion. Such GC transcriptome studies exploration is alternative splicing, where different transcript
have typically fallen into two categories: a) unsupervised, isoforms of the same gene may play distinct functional or
where GC subtypes are discovered based on molecular data oncogenic roles. One of most famous examples of alternative
in an unbiased fashion and then correlated to clinical data; splicing in GC involves the CD44 gene, which encodes a cell-
or b) supervised, where clinical data is directly correlated to surface glycoprotein that binds hyaluronic acid and medi-
molecular data to identify highly associated expression ates a diversity of cell-cell interactions. Recent data has
patterns. Among unsupervised studies, one report, starting revealed that GC stem cells express a specific CD44 tran-
from a panel of GC cell lines, identified two “intrinsic” script variant (CD44v8-10) that can be used to enrich for
subtypes of GC (G-INT/G-DIF), that when mapped to pri- tumor-initiating cells.110 Functional analysis of this variant
mary GCs correlated with Lauren’s classification, and was suggests that its role is to act as a copartner of the xCT
both prognostic and predictive to 5-FU related chemother- cysteine transporter, to regulate oxidative stress.111 Another
apies.102 This classification has recently been extended to recent example of alternative splicing in GC, revealed
three subtypes - proliferative, mesenchymal, and metabolic, through integrated exome/transcriptome analysis, involves
where cell lines with mesenchymal features were found to the selective use of the ZAK kinase isoform TV1 in tumors,
be more responsive to PIK3CA/mTOR inhibitor com- which is required to maintain cell proliferation in experi-
pounds.51 Other unsupervised classifications have been mental systems.112 Alternative splicing events can also
proposed, including a recent 4-subtype system by the Asian cause production of fusion genes in the absence of genome
Cancer Research Group (ACRG), a collaboration between rearrangements, through the process of “read-through”
academia and industry.103 In complement to these, groups transcription. One example of this process is PPP1R1B-
performing supervised analyses have also reported specific STARD3, involving two genes adjacent to one another in the
gene signatures for directly predicting GC patient prognosis. human genome, which fuses exon 6 of PPP1R1B to exon 2 of
One study reported a 6-gene signature that could be used to STARD3. Interestingly, functional analysis suggests that gene
compute an individualized “risk-score” for predicting GC products arising from such “read-through” fusions may be
prognosis.104 In another study from Korea, investigators enriched in gastric tumors related to matched normal con-
explored expression patterns of >1000 GCs to identify an 8- trols, and may contribute towards certain pro-oncogenic
gene signature capable of predicting the prognosis of Stage features.113
1160 Tan and Yeoh Gastroenterology Vol. 149, No. 5

Non-coding RNAs, miRNAs and Beyond reported in other tumor types, such as breast, lung and
Non-coding RNAs represent yet another active player esophageal cancers.131,132 The mechanisms underlying the
in GC pathogenesis, involving different RNA classes such role of the tumor stroma in supporting GC growth are
as miRNAs and long intergenic noncoding RNAs (lincR- likely multifactorial, ranging from providing a suitable
NAs). miRNAs are small RNAs (w22 nucleotides) that can extracellular matrix to maintain cancer cell growth, active
bind to target gene transcripts to induce the latter’s cell-cell interactions via cancer-associated fibroblasts
which can secrete growth-stimulatory molecules such as
GENOMICS

transcript or protein downregulation. Studies from mul-


tiple groups have now produced a growing list of >200 IL-6, and providing a physical barrier inhibiting chemo-
miRNAs with potential roles in GC development, pro- therapy from reaching target malignant cells.133 Major
gression, and treatment response.114,115 Examples of pathways implicated in the tumor stroma include TGFb
oncogenic miRNAs include miR-21, miR-27a, and mir- signaling and EMT-related signaling events, pointing to
130b; which are overexpressed in GC tumors and cell complex interactions between cancer cells and their
lines relative to normal controls.116,117 Oncogenic miRNAs nonmalignant neighboring cellular populations.130,133
typically function to target and downregulate tumor Studies are now emerging at attempting to target the tu-
suppressor genes, for example, mir-21 has been reported mor stroma for therapy.134
to target the tumor suppressor genes PTEN and Another intra-tumor cellular population vitally
PDCD4,116,118 while mir-130b has been shown to regulate important to GC development are cells regulating tumor
RUNX3.117 Reciprocally, tumor suppressor miRNAs such vasculature and immunity. GCs have been shown to
as mir-375, mir-486, mir-29c, and mir-101 are down- secrete pro-angiogenic compounds such as VEGF, CXCL1,
regulated in GC, typically by DNA methylation or genomic and Ang-2 to stimulate blood vessel formation.133 In GC,
loss, and are required to silence genes with oncogenic multiple clinical trials targeting various aspects of the
potential such as PDK1, OLFM4, ITGB1, and EZH2 tumor angiogenesis axis have been conducted. In the
respectively.78,119,120,121 Some miRNAs may also AVAGAST trial, the combination of bevacizumab (an
contribute to GC pathogenesis by functioning as key antibody targeting VEGFA) and chemotherapy did not
components of oncogenic signaling pathways, as shown extend overall survival compared to patients treated
by recent data implicating mir-365 in PTEN/Akt with chemotherapy alone.69 Recently however, the
signaling,122 or by responding to external carcinogens (H RAINBOW and REGARD trials evaluating ramucirumab, a
pylori) to stimulate cellular proliferation (eg, mir-210).123 VEGFR2-targeting antibody, reported an improvement in
Work is also ongoing to characterize the role of miRNAs overall survival in patients with advanced GC.135,136
in tumor progression and drug resistance, with numerous Interestingly, for at least two of these studies (AVA-
studies reporting miRNA expression patterns associated GAST and RAINBOW), subsequent subset and biomarker
with patient outcome.124,125 For example, miRNAs in the analysis revealed that GC patients from non-Asian
mir-200 family play a key role in regulating EMT via EMT countries tended to receive clinical benefit from the
regulators such as ZEB1, and mir-200 downregulaton is addition of bevacizumab, compared to patients from
associated with poor prognosis.126 Asian countries. It is thus possible that GC populations in
In contrast to miRNAs, our current knowledge of the role different countries (specifically Asian and non-Asian
of lincRNAs in GC is still immature, however recent studies countries) may respond differently to anti-angiogenic
examining expression of lincRNAs such as HOTAIR and and potentially other therapies.
GAPLINC in GC suggest an important role for this RNA class Finally, immune cells of many different types are also
in GC development and progression.127,128 Identifying the frequently observed in primary GCs, and certain subtypes
most prevalent deregulated lincRNAs in GC, and deciphering of GC (eg, EBV-positive GC and MSI-positive GC) are well-
their functional and mechanistic roles in regulating gene known to be associated with a high lymphocytic infil-
expression and cancer behavior, will clearly be highly fertile trate. Depending on their specific identities and immune
area for future GC research. functions, such tumor-infiltrating immune cells may play
pro- or anti-tumorigenic roles. For example, GCs with high
levels of regulatory T cells (FOXP3-positive) have been
reported to exhibit good prognosis,137 while those with
Beyond the Cancer Cell: Contributions high levels of IL-17 producing CD8-positive T cells (cyto-
From the Tumor Microenvironment toxic T cells) are associated with poor prognosis.138 The
Gastric tumors are composed not just of cancer cells role of tumor immunity in cancer progression is extremely
but also other cell populations including stromal cells, broad, and readers are directed to other excellent reviews
immune cells, and blood vessels. Far from being innocent dedicated to this subject.139 Interestingly, two recent
bystanders, studies are now demonstrating that these studies have also reported that Asian and non-Asian GC
“noncancer” compartments are also likely to play impor- populations may differ in their regulation of tumor im-
tant roles in GC disease progression and aggressiveness. munity factors at the genetic and gene expression level,
Several studies have now confirmed that GCs with a high which may impact region-specific effects on therapy
stromal content, determined either by gene expression outcome and prognosis.140,141 Given the intimate rela-
analysis or histopathological evaluation, are associated tionship between immune cells and tumor vasculature, it
with poor prognosis.129,130 Similar findings have also been will be interesting to test if such differences might
October 2015 Pathogenesis of Gastric Adenocarcinoma 1161

contribute to the above-mentioned geography-specific 5. Almeida R, Silva E, Santos-Silva F, et al. Expression of


clinical trial outcomes, or if they may impact patient se- intestine-specific transcription factors, CDX1 and CDX2,
lection in future trials evaluating different types of immune in intestinal metaplasia and gastric carcinomas. J Pathol
checkpoint inhibitors. 2003;199:36–40.
6. Goldenring JR, Nomura S. Differentiation of the gastric
mucosa III. Animal models of oxyntic atrophy and
Conclusions and Challenges metaplasia. Am J Physiol Gastrointest Liver Physiol

GENOMICS
2006;291:G999–1004.
Our knowledge of GC molecular pathogenesis has
considerably evolved over the years, but much remains 7. Goldenring JR, Nam KT. Oxyntic atrophy, metaplasia,
and gastric cancer. Dev Diff Dis 2010;96:117–131.
unknown. In terms of basic understanding, the role of novel
molecular entities such as lincRNAs and chromatin alter- 8. Karin M. Inflammation and cancer: the long reach of Ras.
Nat Med 2005;11:20–21.
ations in GC remains to be unraveled. DNA sequencing
studies have also revealed a complex community of non- 9. Eaton KA, Logan SM, Baker PE, et al. Helicobacter pylori
with a truncated lipopolysaccharide O chain fails to
cultivatable microbiota in the human stomach beyond H
induce gastritis in SCID mice injected with splenocytes
pylori and EBV, and it will be critical to understand how the
from wild-type C57BL/6J mice. Infect Immun 2004;
gastric “microbiome” may interact with both H pylori and
72:3925–3931.
the host immune response to influence gastric carcinogen-
10. Lochhead P, El-Omar EM. Gastric cancer. Br Med Bull
esis.142 In the early detection field, molecular findings may
2008;85:87–100.
facilitate new approaches for diagnosing GC early, by iden-
11. Tsugane S, Sasazuki S. Diet and the risk of gastric
tifying high-risk individuals through the molecular charac-
cancer: review of epidemiological evidence. Gastric
terization of preneoplastic lesions or even blood-based
Cancer 2007;10:75–83.
biomarker assays. Finally, therapeutic strategies remain to
12. Noto JM, Gaddy JA, Lee JY, et al. Iron deficiency ac-
be developed to target specific GC subtypes (particularly
celerates Helicobacter pylori-induced carcinogenesis in
diffuse type GC), based upon their somatic driver alterations rodents and humans. J Clin Invest 2013;123:479–492.
or tumor-associated cell compartments (eg, stromal cells
13. Iwakiri D TK. Epstein-Barr virus and gastric cancers. In:
and tumor-infiltrating immune cells). The next five years ES R, ed. Epstein-Barr virus. Norfolk, VA: Caister Aca-
will undoubtedly see a further explosion of findings in the demic Press, 2005:157–169.
GC field, which will be essential to combat this deadly 14. Murphy G, Pfeiffer R, Camargo MC, et al. Meta-analysis
disease. shows that prevalence of Epstein-Barr virus-positive
gastric cancer differs based on sex and anatomic loca-
tion. Gastroenterology 2009;137:824–833.
Supplementary Material 15. Peleteiro B, Bastos A, Ferro A, et al. Prevalence of Hel-
NOTE: The first 50 references associated with this article icobacter pylori infection worldwide: a systematic review
are available below in print. The remaining references of studies with national coverage. Dig Dis Sci 2014;
accompanying this article are available online only with the 59:1698–1709.
electronic version of this article. To access the supplemen- 16. Odenbreit S, Puls J, Sedlmaier B, et al. Translocation of
tary material accompanying this article, visit the online Helicobacter pylori CagA into gastric epithelial cells by
version of Gastroenterology at www.gastrojournal.org, and type IV secretion. Science 2000;287:1497–1500.
at http://dx.doi.org/10.1053/j.gastro.2015.05.059. 17. Ohnishi N, Yuasa H, Tanaka S, et al. Transgenic
expression of Helicobacter pylori CagA induces
gastrointestinal and hematopoietic neoplasms in
References mouse. Proc Natl Acad Sci U S A 2008;105:
1. Ferlay JSI, Ervik M, Dikshit R, et al. International Agency 1003–1008.
for Research on Cancer, Cancer Incidence, and Mortality 18. Higashi H, Tsutsumi R, Fujita A, et al. Biological activity
Worldwide. Lyon, France: IARC CancerBase no. 11, of the Helicobacter pylori virulence factor CagA is
2013. determined by variation in the tyrosine phosphorylation
2. Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastu- sites. Proc Natl Acad Sci U S A 2002;99:14428–14433.
zumab in combination with chemotherapy versus 19. Higashi H, Tsutsumi R, Muto S, et al. SHP-2 tyrosine
chemotherapy alone for treatment of HER2-positive phosphatase as an intracellular target of Helicobacter
advanced gastric or gastro-oesophageal junction can- pylori CagA protein. Science 2002;295:683–686.
cer (ToGA): a phase 3, open-label, randomised controlled 20. Amieva MR, Vogelmann R, Covacci A, et al. Disruption of
trial. Lancet 2010;376:687–697. the epithelial apical-junctional complex by Helicobacter
3. Cancer Genome Atlas Research N. Comprehensive pylori CagA. Science 2003;300:1430–1434.
molecular characterization of gastric adenocarcinoma. 21. Saadat I, Higashi H, Obuse C, et al. Helicobacter pylori
Nature 2014;513:202–209. CagA targets PAR1/MARK kinase to disrupt epithelial
4. Correa P. Human gastric carcinogenesis: a multistep and cell polarity. Nature 2007;447:330.
multifactorial process—first American Cancer Society 22. Murata-Kamiya N, Kurashima Y, Teishikata Y, et al.
Award Lecture on cancer epidemiology and prevention. Helicobacter pylori CagA interacts with E-cadherin and
Cancer Res 1992;52:6735–6740. deregulates the beta-catenin signal that promotes in-
1162 Tan and Yeoh Gastroenterology Vol. 149, No. 5

testinal transdifferentiation in gastric epithelial cells. gastrointestinal cancer in white individuals. Gastroen-
Oncogene 2007;26:4617–4626. terology 2011;140:435–441.
23. Wong BC, Lam SK, Wong WM, et al. Helicobacter pylori 40. Oliveira C, Pinheiro H, Figueiredo J, et al. Familial
eradication to prevent gastric cancer in a high-risk region gastric cancer: genetic susceptibility, pathology, and
of China: a randomized controlled trial. JAMA 2004; implications for management. Lancet Oncol 2015;16:
291:187–194. e60–e70.
24. Lee YC, Chen TH, Chiu HM, et al. The benefit of mass 41. Huntsman DG, Carneiro F, Lewis FR, et al. Early gastric
GENOMICS

eradication of Helicobacter pylori infection: a cancer in young, asymptomatic carriers of germ-line E-


community-based study of gastric cancer prevention. cadherin mutations. N Engl J Med 2001;344:1904–1909.
Gut 2013;62:676–682. 42. Hansford S, Kaurah P, Li-Chang H, et al. Hereditary
25. Fuccio L, Zagari RM, Eusebi LH, et al. Meta-analysis: can Diffuse Gastric Cancer Syndrome: CDH1 Mutations and
Helicobacter pylori eradication treatment reduce the risk Beyond. JAMA Oncol 2015;1:23–32.
for gastric cancer? Ann Intern Med 2009;151:121–128. 43. Worthley DL, Phillips KD, Wayte N, et al. Gastric
26. Shiota S, Yamaoka Y. Strategy for the treatment of adenocarcinoma and proximal polyposis of the stomach
Helicobacter pylori infection. Curr Pharm Des 2014; (GAPPS): a new autosomal dominant syndrome. Gut
20:4489–4500. 2012;61:774–779.
27. Nomura AM, Wilkens LR, Henderson BE, et al. The associ- 44. Lauren P. The two histological main types of gastric
ation of cigarette smoking with gastric cancer: the multi- carcinoma: diffuse and so-called intestinal-type carci-
ethnic cohort study. Cancer Causes Control 2012;23:51–58. noma. An attempt at a histo-clinical classification. Acta
28. Dyke GW, Craven JL, Hall R, et al. Smoking-related DNA Pathol Microbiol Scand 1965;64:31–49.
adducts in human gastric cancers. Int J Cancer 1992; 45. Kaneko S, Yoshimura T. Time trend analysis of gastric
52:847–850. cancer incidence in Japan by histological types, 1975-
29. Kneller RW, You WC, Chang YS, et al. Cigarette smoking 1989. Br J Cancer 2001;84:400–405.
and other risk factors for progression of precancerous 46. Adachi Y, Yasuda K, Inomata M, et al. Pathology and
stomach lesions. J Natl Cancer Inst 1992;84:1261–1266. prognosis of gastric carcinoma: well versus poorly
30. Kaneda A, Matsusaka K, Aburatani H, et al. Epstein-Barr differentiated type. Cancer 2000;89:1418–1424.
virus infection as an epigenetic driver of tumorigenesis. 47. Bosman FT. WHO classification of tumours of the
Cancer Res 2012;72:3445–3450. digestive system. Lyon, Paris: IARC Press, 2010.
31. Pfeffer S, Zavolan M, Grasser FA, et al. Identification of 48. Lawrence MS, Stojanov P, Polak P, et al. Mutational
virus-encoded microRNAs. Science 2004;304:734–736. heterogeneity in cancer and the search for new cancer-
32. Marquitz AR, Mathur A, Nam CS, et al. The Epstein-Barr associated genes. Nature 2013;499:214–218.
Virus BART microRNAs target the pro-apoptotic protein 49. Gonzalgo ML, Jones PA. Mutagenic and epigenetic ef-
Bim. Virology 2011;412:392–400. fects of DNA methylation. Mutat Res 1997;386:107–118.
33. Shinozaki-Ushiku A, Kunita A, Isogai M, et al. Profiling of 50. Hanazono K, Natsugoe S, Stein HJ, et al. Distribution of
virus-encoded microRNAs in Epstein-Barr virus-associ- p53 mutations in esophageal and gastric carcinomas and
ated gastric carcinoma and their roles in gastric carci- the relationship with p53 expression. Oncol Rep 2006;
nogenesis. J Virol 2015;89:5581–5591. 15:821–824.
34. El-Omar EM, Carrington M, Chow WH, et al. Interleukin-1
polymorphisms associated with increased risk of gastric
Received March 25, 2015. Accepted May 20, 2015.
cancer. Nature 2000;404:398–402.
35. El-Omar EM, Rabkin CS, Gammon MD, et al. Increased Reprint requests
Address requests for reprints to: Patrick Tan, Cancer and Stem Cell Biology
risk of noncardia gastric cancer associated with proin- Program, Duke-NUS Graduate Medical School, 8 College Road, Singapore
flammatory cytokine gene polymorphisms. Gastroenter- 169857. e-mail: gmstanp@duke-nus.edu.sg; fax: (þ65) 6226 5294; or Khay
ology 2003;124:1193–1201. Guan Yeoh, Department of Medicine, Yong Loo Lin School of Medicine, 1E
Kent Ridge Road, NUHS Tower Block, Level 11, Singapore 119228. e-mail:
36. Figueiredo C, Machado JC, Pharoah P, et al. Heli- khay_guan_yeoh@nuhs.edu.sg; fax: (þ65) 6778 5743.
cobacter pylori and interleukin 1 genotyping: an oppor-
Acknowledgements
tunity to identify high-risk individuals for gastric We thank Beatrice Tan, Jennie Wong, Tze Wei Chua, and Feng Zhu for
carcinoma. J Natl Cancer Inst 2002;94:1680–1687. assistance with manuscript preparation and Wei Peng Yong and Matthew Ng
37. Persson C, Canedo P, Machado JC, et al. Polymorphisms for valuable feedback. We thank Prof Teh Ming and Dr Supriya Srivastava,
Dept of Pathology, National University of Singapore for supplying the
in inflammatory response genes and their association with histopathology images for Figure 1. Due to length restrictions, we apologize
gastric cancer: A HuGE systematic review and meta-an- to those clinicians and scientists in the GC community whose work we may
have inadvertently missed.
alyses. Am J Epidemiol 2011;173:259–270.
38. Study Group of Millennium Genome Project for C, Conflicts of interest
Sakamoto H, Yoshimura K, et al. Genetic variation in The authors disclose no conflicts.
PSCA is associated with susceptibility to diffuse-type Funding
gastric cancer. Nat Genet 2008;40:730–740. This work was supported by the Singapore Gastric Cancer Consortium, funded
through the National Medical Research Council/National Research Foundation
39. Lochhead P, Frank B, Hold GL, et al. Genetic variation in Translational and Clinical Research Flagship Program (NMRC/TCR/009-NUHS/
the prostate stem cell antigen gene and upper 2013).
October 2015 Pathogenesis of Gastric Adenocarcinoma 1162.e1

67. Deng N, Goh LK, Wang H, et al. A comprehensive survey


Supplementary References
of genomic alterations in gastric cancer reveals sys-
51. Lei Z, Tan IB, Das K, et al. Identification of molecular tematic patterns of molecular exclusivity and co-
subtypes of gastric cancer with different responses to occurrence among distinct therapeutic targets. Gut
PI3-kinase inhibitors and 5-fluorouracil. Gastroenter- 2012;61:673–684.
ology 2013;145:554–565.
68. Dulak AM, Schumacher SE, van Lieshout J, et al.
52. Grabsch HI, Tan P. Gastric cancer pathology and un- Gastrointestinal adenocarcinomas of the esophagus,
derlying molecular mechanisms. Dig Surg 2013; stomach, and colon exhibit distinct patterns of genome
30:150–158. instability and oncogenesis. Cancer Res 2012;
53. Jaiswal BS, Kljavin NM, Stawiski EW, et al. Oncogenic 72:4383–4393.
ERBB3 mutations in human cancers. Cancer Cell 2013; 69. Ohtsu A, Shah MA, Van Cutsem E, et al. Bevacizumab in
23:603–617. combination with chemotherapy as first-line therapy in
54. Chen K, Yang D, Li X, et al. Mutational landscape of advanced gastric cancer: a randomized, double-blind,
gastric adenocarcinoma in Chinese: implications for placebo-controlled phase III study. J Clin Oncol 2011;
prognosis and therapy. Proc Natl Acad Sci U S A 2015; 29:3968–3976.
112:1107–1112. 70. Kim J, Fox C, Peng S, et al. Preexisting oncogenic
55. Wang K, Yuen ST, Xu J, et al. Whole-genome events impact trastuzumab sensitivity in ERBB2-
sequencing and comprehensive molecular profiling amplified gastroesophageal adenocarcinoma. J Clin
identify new driver mutations in gastric cancer. Nat Invest 2014;124:5145–5158.
Genet 2014;46:573–582. 71. Scaltriti M, Eichhorn PJ, Cortes J, et al. Cyclin E ampli-
56. Wang K, Kan J, Yuen ST, et al. Exome sequencing fication/overexpression is a mechanism of trastuzumab
identifies frequent mutation of ARID1A in molecular resistance in HER2þ breast cancer patients. Proc Natl
subtypes of gastric cancer. Nat Genet 2011; Acad Sci U S A 2011;108:3761–3766.
43:1219–1223. 72. Sulahian R, Casey F, Shen J, et al. An integrative analysis
57. Zang ZJ, Cutcutache I, Poon SL, et al. Exome reveals functional targets of GATA6 transcriptional regu-
sequencing of gastric adenocarcinoma identifies recur- lation in gastric cancer. Oncogene 2014;33:5637–5648.
rent somatic mutations in cell adhesion and chromatin 73. Chia NY, Deng N, Das K, et al. Regulatory crosstalk
remodeling genes. Nat Genet 2012;44:570–574. between lineage-survival oncogenes KLF5, GATA4 and
58. Kakiuchi M, Nishizawa T, Ueda H, et al. Recurrent gain- GATA6 cooperatively promotes gastric cancer develop-
of-function mutations of RHOA in diffuse-type gastric ment. Gut 2015;64:707–719.
carcinoma. Nat Genet 2014;46:583–587. 74. Haveri H, Westerholm-Ormio M, Lindfors K, et al. Tran-
59. Nagarajan N, Bertrand D, Hillmer AM, et al. Whole- scription factors GATA-4 and GATA-6 in normal and
genome reconstruction and mutational signatures in neoplastic human gastrointestinal mucosa. BMC Gas-
gastric cancer. Genome Biol 2012;13:R115. troenterol 2008;8:9.
60. Wong SS, Kim KM, Ting JC, et al. Genomic landscape 75. Qian J, Kong X, Deng N, et al. OCT1 is a determinant of
and genetic heterogeneity in gastric adenocarcinoma synbindin-related ERK signalling with independent
revealed by whole-genome sequencing. Nat Commun prognostic significance in gastric cancer. Gut 2015;
2014;5:5477. 64:37–48.
61. Takeshima H, Niwa T, Takahashi T, et al. Frequent 76. Yan C, Higgins PJ. Drugging the undruggable: tran-
involvement of chromatin remodeler alterations in gastric scription therapy for cancer. Biochim Biophys Acta 2013;
field cancerization. Cancer Lett 2015;357:328–338. 1835:76–85.
62. Kim TM, Jung SH, Kim MS, et al. The mutational burdens 77. Corso G, Carvalho J, Marrelli D, et al. Somatic mutations
and evolutionary ages of early gastric cancers are and deletions of the E-cadherin gene predict poor sur-
comparable to those of advanced gastric cancers. vival of patients with gastric cancer. J Clin Oncol 2013;
J Pathol 2014;234:365–374. 31:868–875.
63. Yoon K, Lee S, Han TS, et al. Comprehensive genome- 78. Carvalho J, van Grieken NC, Pereira PM, et al. Lack of
and transcriptome-wide analyses of mutations associ- microRNA-101 causes E-cadherin functional deregula-
ated with microsatellite instability in Korean gastric tion through EZH2 up-regulation in intestinal gastric
cancers. Genome Res 2013;23:1109–1117. cancer. J Pathol 2012;228:31–44.
64. Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signa- 79. Lin DC, Xu L, Ding LW, et al. Genomic and functional
tures of mutational processes in human cancer. Nature characterizations of phosphodiesterase subtype 4D in
2013;500:415–421. human cancers. Proc Natl Acad Sci U S A 2013;
65. Matsumoto Y, Marusawa H, Kinoshita K, et al. Heli- 110:6109–6114.
cobacter pylori infection triggers aberrant expression of 80. Palanisamy N, Ateeq B, Kalyana-Sundaram S, et al.
activation-induced cytidine deaminase in gastric Rearrangements of the RAF kinase pathway in prostate
epithelium. Nat Med 2007;13:470–476. cancer, gastric cancer and melanoma. Nat Med 2010;
66. Nadauld LD, Garcia S, Natsoulis G, et al. Metastatic tu- 16:793–798.
mor evolution and organoid modeling implicate TGFBR2 81. Tao J, Deng NT, Ramnarayanan K, et al. CD44-SLC1A2
as a cancer driver in diffuse gastric cancer. Genome Biol gene fusions in gastric cancer. Sci Transl Med 2011;
2014;15:428. 3:77ra30.
1162.e2 Tan and Yeoh Gastroenterology Vol. 149, No. 5

82. Giacomini CP, Sun S, Varma S, et al. Breakpoint analysis associated cryptic promoters and somatically acquired
of transcriptional and genomic profiles uncovers novel regulatory elements. Nat Commun 2014;5:4361.
gene fusions spanning multiple human cancer types. 99. Tay ST, Leong SH, Yu K, et al. A combined comparative
PLoS Genet 2013;9:e1003464. genomic hybridization and expression microarray anal-
83. Shinmura K, Kato H, Igarashi H, et al. CD44–SLC1A2 ysis of gastric cancer reveals novel molecular subtypes.
Fusion Transcripts in Primary Colorectal Cancer. Pathol Cancer Res 2003;63:3309–3316.
Oncol Res 2015;21:759–764. 100.Boussioutas A, Li H, Liu J, et al. Distinctive patterns of
84. Lee J, Lee SE, Kang SY, et al. Identification of ROS1 gene expression in premalignant gastric mucosa and
rearrangement in gastric adenocarcinoma. Cancer 2013; gastric cancer. Cancer Res 2003;63:2569–2577.
119:1627–1635. 101.Chen X, Leung SY, Yuen ST, et al. Variation in gene
85. Yao F, Kausalya JP, Sia YY, et al. Recurrent fusion genes expression patterns in human gastric cancers. Mol Biol
in gastric cancer: CLDN18-ARHGAP26 induces loss of Cell 2003;14:3208–3215.
epithelial integrity. Cell Rep 2015;12:272–285. 102.Tan IB, Ivanova T, Lim KH, et al. Intrinsic subtypes of
86. Niwa T, Tsukamoto T, Toyoda T, et al. Inflammatory gastric cancer, based on gene expression pattern, pre-
processes triggered by Helicobacter pylori infection dict survival and respond differently to chemotherapy.
cause aberrant DNA methylation in gastric epithelial Gastroenterology 2011;141:476–485.
cells. Cancer Res 2010;70:1430–1440. 103.Cristescu R, Lee J, Nebozhyn M, et al. Molecular anal-
87. Asada K, Nakajima T, Shimazu T, et al. Demonstration of ysis of gastric cancer identifies discrete subtypes asso-
the usefulness of epigenetic cancer risk prediction by a ciated with distinct clinical characteristics and survival
multicentre prospective cohort study. Gut 2015; outcomes: the ACRG (Asian Cancer Research Group)
64:388–396. study. Nature Medicine 2015;21:449–456.
88. Nakamura J, Tanaka T, Kitajima Y, et al. Methylation- 104.Cho JY, Lim JY, Cheong JH, et al. Gene expression
mediated gene silencing as biomarkers of gastric cancer: signature-based prognostic risk score in gastric cancer.
a review. World J Gastroenterol 2014;20:11991–12006. Clin Cancer Res 2011;17:1850–1857.
89. Ganesan K, Ivanova T, Wu Y, et al. Inhibition of gastric 105.Lee J, Sohn I, Do IG, et al. Nanostring-based multigene
cancer invasion and metastasis by PLA2G2A, a novel assay to predict recurrence for gastric cancer patients
beta-catenin/TCF target gene. Cancer Res 2008; after surgery. PLoS One 2014;9:e90133.
68:4277–4286. 106.Ooi CH, Ivanova T, Wu J, et al. Oncogenic pathway
90. Wang L, Xie L, Wang J, et al. Correlation between the combinations predict clinical prognosis in gastric cancer.
methylation of SULF2 and WRN promoter and the iri- PLoS Genet 2009;5:e1000676.
notecan chemosensitivity in gastric cancer. BMC Gas- 107.Chang HR, Nam S, Kook MC, et al. HNF4alpha is a
troenterol 2013;13:173. therapeutic target that links AMPK to WNT signalling in
91. Ivanova T, Zouridis H, Wu Y, et al. Integrated epi- early-stage gastric cancer. Gut 2014 [Epub ahead of
genomics identifies BMP4 as a modulator of cisplatin print]; pii: gutjnl-2014-307918.
sensitivity in gastric cancer. Gut 2013;62:22–33. 108.Kim YH, Liang H, Liu X, et al. AMPKalpha modulation in
92. Toyota M, Ahuja N, Suzuki H, et al. Aberrant methylation cancer progression: multilayer integrative analysis of the
in gastric cancer associated with the CpG island meth- whole transcriptome in Asian gastric cancer. Cancer Res
ylator phenotype. Cancer Res 1999;59:5438–5442. 2012;72:2512–2521.
93. Zouridis H, Deng N, Ivanova T, et al. Methylation sub- 109.Kim HK, Choi IJ, Kim CG, et al. A gene expression
types and large-scale epigenetic alterations in gastric signature of acquired chemoresistance to cisplatin and
cancer. Sci Transl Med 2012;4:156ra140. fluorouracil combination chemotherapy in gastric cancer
94. Matsusaka K, Kaneda A, Nagae G, et al. Classification of patients. PLoS One 2011;6:e16694.
Epstein-Barr virus-positive gastric cancers by definition 110.Lau WM, Teng E, Chong HS, et al. CD44v8-10 is a
of DNA methylation epigenotypes. Cancer Res 2011; cancer-specific marker for gastric cancer stem cells.
71:7187–7197. Cancer Res 2014;74:2630–2641.
95. Grogg KL, Lohse CM, Pankratz VS, et al. Lymphocyte- 111.Ishimoto T, Nagano O, Yae T, et al. CD44 variant regu-
rich gastric cancer: associations with Epstein-Barr virus, lates redox status in cancer cells by stabilizing the xCT
microsatellite instability, histology, and survival. Mod subunit of system xc(-) and thereby promotes tumor
Pathol 2003;16:641–651. growth. Cancer Cell 2011;19:387–400.
96. Liang Q, Yao X, Tang S, et al. Integrative identification of 112.Liu J, McCleland M, Stawiski EW, et al. Integrated exome
Epstein-Barr virus-associated mutations and epigenetic and transcriptome sequencing reveals ZAK isoform us-
alterations in gastric cancer. Gastroenterology 2014; age in gastric cancer. Nat Commun 2014;5:3830.
147:1350–1362. 113.Yun SM, Yoon K, Lee S, et al. PPP1R1B-STARD3
97. Zhang L, Zhong K, Dai Y, et al. Genome-wide analysis of chimeric fusion transcript in human gastric cancer pro-
histone H3 lysine 27 trimethylation by ChIP-chip in motes tumorigenesis through activation of PI3K/AKT
gastric cancer patients. J Gastroenterol 2009; signaling. Oncogene 2014;33:5341–5347.
44:305–312. 114.Song JH, Meltzer SJ. MicroRNAs in pathogenesis,
98. Muratani M, Deng N, Ooi WF, et al. Nanoscale chromatin diagnosis, and treatment of gastroesophageal cancers.
profiling of gastric adenocarcinoma reveals cancer- Gastroenterology 2012;143:35–47.
October 2015 Pathogenesis of Gastric Adenocarcinoma 1162.e3

115.Song S, Ajani JA. The role of microRNAs in cancers of outcome, and highlight inflammatory pathways in
the upper gastrointestinal tract. Nat Rev Gastroenterol digestive cancers. Proc Natl Acad Sci U S A 2010;
Hepatol 2013;10:109–118. 107:2177–2182.
116.Lu Z, Liu M, Stribinskis V, et al. MicroRNA-21 promotes 132.Rajski M, Zanetti-Dallenbach R, Vogel B, et al. IGF-I
cell transformation by targeting the programmed cell induced genes in stromal fibroblasts predict the clinical
death 4 gene. Oncogene 2008;27:4373–4379. outcome of breast and lung cancer patients. BMC Med
117.Lai KW, Koh KX, Loh M, et al. MicroRNA-130b regulates 2010;8:1.
the tumour suppressor RUNX3 in gastric cancer. Eur J 133.Quail DF, Joyce JA. Microenvironmental regulation of
Cancer 2010;46:1456–1463. tumor progression and metastasis. Nat Med 2013;
118.Zhang BG, Li JF, Yu BQ, et al. microRNA-21 promotes 19:1423–1437.
tumor proliferation and invasion in gastric cancer by 134.Merchant JL, Saqui-Salces M. Inhibition of Hedgehog
targeting PTEN. Oncol Rep 2012;27:1019–1026. signaling in the gastrointestinal tract: targeting the can-
119.Tsukamoto Y, Nakada C, Noguchi T, et al. MicroRNA- cer microenvironment. Cancer Treat Rev 2014;40:12–21.
375 is downregulated in gastric carcinomas and regu- 135.Wilke H, Muro K, Van Cutsem E, et al. Ramucirumab plus
lates cell survival by targeting PDK1 and 14-3-3zeta. paclitaxel versus placebo plus paclitaxel in patients with
Cancer Res 2010;70:2339–2349. previously treated advanced gastric or gastro-
120.Oh HK, Tan AL, Das K, et al. Genomic loss of miR-486 oesophageal junction adenocarcinoma (RAINBOW): a
regulates tumor progression and the OLFM4 anti- double-blind, randomised phase 3 trial. Lancet Oncol
apoptotic factor in gastric cancer. Clin Cancer Res 2011; 2014;15:1224–1235.
17:2657–2667. 136.Fuchs CS, Tomasek J, Yong CJ, et al. Ramucirumab
121.Han TS, Hur K, Xu G, et al. MicroRNA-29c mediates monotherapy for previously treated advanced gastric or
initiation of gastric carcinogenesis by directly targeting gastro-oesophageal junction adenocarcinoma (RE-
ITGB1. Gut 2015;64:203–214. GARD): an international, randomised, multicentre,
122.Guo SL, Ye H, Teng Y, et al. Akt-p53-miR-365-cyclin D1/ placebo-controlled, phase 3 trial. Lancet 2014;383:31–39.
cdc25A axis contributes to gastric tumorigenesis 137.Haas M, Dimmler A, Hohenberger W, et al. Stromal
induced by PTEN deficiency. Nat Commun 2013;4:2544. regulatory T-cells are associated with a favourable
123.Kiga K, Mimuro H, Suzuki M, et al. Epigenetic silencing prognosis in gastric cancer of the cardia. BMC Gastro-
of miR-210 increases the proliferation of gastric epithe- enterol 2009;9:65.
lium during chronic Helicobacter pylori infection. Nat 138.Zhuang Y, Peng LS, Zhao YL, et al. CD8(þ) T cells that
Commun 2014;5:4497. produce interleukin-17 regulate myeloid-derived sup-
124.Li X, Zhang Y, Zhang Y, et al. Survival prediction of pressor cells and are associated with survival time of
gastric cancer by a seven–microRNA signature. Gut patients with gastric cancer. Gastroenterology 2012;
2010;59:579–585. 143:951–962.
125.Ueda T, Volinia S, Okumura H, et al. Relation between 139.Fridman WH, Pages F, Sautes-Fridman C, et al. The
microRNA expression and progression and prognosis of immune contexture in human tumours: impact on clinical
gastric cancer: a microRNA expression analysis. Lancet outcome. Nat Rev Cancer 2012;12:298–306.
Oncol 2010;11:136–146. 140.Lin SJ, Gagnon-Bartsch JA, Tan IB, et al. Signatures of
126.Song F, Yang D, Liu B, et al. Integrated microRNA tumour immunity distinguish Asian and non-Asian
network analyses identify a poor-prognosis subtype of gastric adenocarcinomas. Gut 2014 [Epub ahead of
gastric cancer characterized by the miR-200 family. Clin print]; pii: gutjnl-2014-308252.
Cancer Res 2014;20:878–889. 141.Sunakawa Y, Stremitzer S, Cao S, et al. Association of
127.Okugawa Y, Toiyama Y, Hur K, et al. Metastasis-asso- variants in genes encoding for macrophage-related
ciated long non-coding RNA drives gastric cancer functions with clinical outcome in patients with
development and promotes peritoneal metastasis. locoregional gastric cancer. Ann Oncol 2015;
Carcinogenesis 2014;35:2731–2739. 26:332–339.
128.Hu Y, Wang J, Qian J, et al. Long noncoding RNA 142.Abreu MT, Peek RM Jr. Gastrointestinal malignancy and
GAPLINC regulates CD44-dependent cell invasiveness the microbiome. Gastroenterology 2014;146:1534–1546.
and associates with poor prognosis of gastric cancer. 143.Yasui W, Oue N, Aung PP, et al. Molecular-pathological
Cancer Res 2014;74:6890–6902. prognostic factors of gastric cancer: a review. Gastric
129.Busuttil RA, George J, Tothill RW, et al. A signature Cancer 2005;8:86–94.
predicting poor prognosis in gastric and ovarian cancer 144.Wroblewski LE, Peek RM Jr, Wilson KT. Helicobacter
represents a coordinated macrophage and stromal pylori and gastric cancer: factors that modulate disease
response. Clin Cancer Res 2014;20:2761–2772. risk. Clin Microbiol Rev 2010;23:713–739.
130.Wu Y, Grabsch H, Ivanova T, et al. Comprehensive 145.Loh M, Koh KX, Yeo BH, et al. Meta-analysis of genetic
genomic meta-analysis identifies intra-tumoural stroma polymorphisms and gastric cancer risk: variability in
as a predictor of survival in patients with gastric cancer. associations according to race. Eur J Cancer 2009;
Gut 2013;62:1100–1111. 45:2562–2568.
131.Saadi A, Shannon NB, Lao-Sirieix P, et al. Stromal genes 146.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell
discriminate preinvasive from invasive disease, predict 2000;100:57–70.

Potrebbero piacerti anche