Comprehensive molecular characterization of gastric adenocarcinoma

Cancer Genome Atlas Research Network

Abstract

Gastric cancer is a leading cause of cancer deaths, but analysis of its molecular and clinical characteristics has been complicated by histological and aetiological heterogeneity. Here we describe a comprehensive molecular evaluation of 295 primary gastric adenocarcinomas as part of The Cancer Genome Atlas (TCGA) project. We propose a molecular classification dividing gastric cancer into four subtypes: tumours positive for Epstein-Barr virus, which display recurrent PIK3CA mutations, extreme DNA hypermethylation, and amplification of JAK2, CD274 (also known as PD-L1) and PDCD1LG2 (also known as PD-L2); microsatellite unstable tumours, which show elevated mutation rates, including mutations of genes encoding targetable oncogenic signalling proteins; genomically stable tumours, which are enriched for the diffuse histological variant and mutations of RHOA or fusions involving RHO-family GTPase-activating proteins; and tumours with chromosomal instability, which show marked aneuploidy and focal amplification of receptor tyrosine kinases. Identification of these subtypes provides a roadmap for patient stratification and trials of targeted therapies.

Conflict of interest statement

The author declare no competing financial interests.

Figures

Figure 1. Molecular subtypes of gastric cancer.
Figure 1. Molecular subtypes of gastric cancer.
a, Gastric cancer cases are divided into subtypes: Epstein–Barr virus (EBV)-positive (red), microsatellite instability (MSI, blue), genomically stable (GS, green) and chromosomal instability (CIN, light purple) and ordered by mutation rate. Clinical (top) and molecular data (top and bottom) from 227 tumours profiled with all six platforms are depicted. b, A flowchart outlines how tumours were classified into molecular subtypes. c, Differences in clinical and histological characteristics among subtypes with subtypes coloured as in a, b. The plot of patient age at initial diagnosis shows the median, 25th and 75th percentile values (horizontal bar, bottom and top bounds of the box), and the highest and lowest values within 1.5 times the interquartile range (top and bottom whiskers, respectively). GE, gastroesophageal. PowerPoint slide
Figure 2. Molecular characteristics of EBV-positive gastric…
Figure 2. Molecular characteristics of EBV-positive gastric cancers.
a, The heatmap represents unsupervised clustering of DNA methylation at CpG sites for 295 tumours into four clusters: EBV-CIMP (n = 28), Gastric-CIMP (n = 77), cluster 3 (n = 73) and cluster 4 (n = 117). Profiles for non-malignant gastric mucosa are to the left of the tumours. b, The proportion of tumours harbouring PIK3CA mutation in the molecular subtypes with mutations at sites noted recurrently in this data set or in the COSMIC database marked separately (top). Locations of PIK3CA mutations with the subtype of the sample with each mutation colour-coded (bottom). PowerPoint slide
Figure 3. Significantly mutated genes in non-hypermutated…
Figure 3. Significantly mutated genes in non-hypermutated gastric cancer.
a, Bars represent somatic mutation rate for the 215 samples with synonymous and non-synonymous mutation rates distinguished by colour. b, Significantly mutated genes, identified by MutSigCV, are ranked by the q value (right) with samples grouped by subtype. Mutation colour indicates the class of mutation. PowerPoint slide
Figure 4. RHOA and ARHGAP6/26 somatic genomic…
Figure 4. RHOA and ARHGAP6/26 somatic genomic alterations are recurrent in genomically stable gastric cancer.
a, Missense mutations in the GTPase RHOA, including residues Y42 and D59, linked via hydrogen bond (red arc). b, Mutated regions (coloured as in panel a) mapped on the structures of RHOA and ROCK1. c, A schematic of CLDN18–ARHGAP26 translocation is shown for the fusion transcript and predicted fusion protein. SH3 denotes SRC homology 3 domain. d, The frequency of RHOA and CDH1 mutations, CLDN18ARHGAP6 or ARHGAP26 fusions are shown across gastric cancer subtypes. e, RHOA mutations and CLDN18ARHGAP6 or ARHGAP26 fusions are mutually exclusive in genomically stable tumours. PowerPoint slide
Figure 5. Integrated molecular description of gastric…
Figure 5. Integrated molecular description of gastric cancer.
a, Mutations, copy-number changes and translocations for select genes are shown across samples organized by molecular subtypes. Mutations that are recurrent in this data set or in the COSMIC repository are distinguished by colour. Alteration frequencies are expressed as a percentage of all cases. b, Alterations in RTK/RAS and RTK/PI(3)K signalling pathways across molecular subtypes. Red denotes predicted activation; blue denotes predicted inactivation. c, The heatmap shows NCI-PID pathways that are significantly elevated (red) or decreased (blue) in each of the four subtypes as compared with non-malignant gastric mucosa. PowerPoint slide
Figure 6. Key features of gastric cancer…
Figure 6. Key features of gastric cancer subtypes.
This schematic lists some of the salient features associated with each of the four molecular subtypes of gastric cancer. Distribution of molecular subtypes in tumours obtained from distinct regions of the stomach is represented by inset charts. PowerPoint slide

References

    1. Ferlay, J. et al. GLOBOCAN 2012 v1.0, cancer incidence and mortality worldwide. IARC CancerBase 11, , accessed on January 15, 2014 (2013)
    1. Laurén P. The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. Acta Pathol., Microbiol. Scand. 1965;64:31–49. doi: 10.1111/apm.1965.64.1.31.
    1. WHO Classification of Tumours of the Digestive System 4th edn. (International Agency for Research on Cancer, 2010)
    1. Uemura N, et al. Helicobacter pylori infection and the development of gastric cancer. N. Engl. J. Med. 2001;345:784–789. doi: 10.1056/NEJMoa001999.
    1. Bertuccio P, et al. Recent patterns in gastric cancer: a global overview. International J. Cancer. 2009;125:666–673. doi: 10.1002/ijc.24290.
    1. Richards FM, et al. Germline E-cadherin gene (CDH1) mutations predispose to familial gastric cancer and colorectal cancer. Hum. Mol. Genet. 1999;8:607–610. doi: 10.1093/hmg/8.4.607.
    1. Keller G, et al. Analysis for microsatellite instability and mutations of the DNA mismatch repair gene hMLH1 in familial gastric cancer. International J Cancer. 1996;68:571–576. doi: 10.1002/(SICI)1097-0215(19961127)68:5<571::AID-IJC3>;2-W.
    1. Toyota M, et al. Aberrant methylation in gastric cancer associated with the CpG island methylator phenotype. Cancer Res. 1999;59:5438–5442.
    1. Tan IB, et al. Intrinsic subtypes of gastric cancer, based on gene expression pattern, predict survival and respond differently to chemotherapy. Gastroenterology. 2011;141:476–485. doi: 10.1053/j.gastro.2011.04.042.
    1. Lei Z, et al. Identification of molecular subtypes of gastric cancer with different responses to PI3-kinase inhibitors and 5-fluorouracil. Gastroenterology. 2013;145:554–565. doi: 10.1053/j.gastro.2013.05.010.
    1. Boussioutas A, et al. Distinctive patterns of gene expression in premalignant gastric mucosa and gastric cancer. Cancer Res. 2003;63:2569–2577.
    1. Wang K, et al. Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer. Nature Genet. 2011;43:1219–1223. doi: 10.1038/ng.982.
    1. Shen R, Olshen AB, Ladanyi M. Integrative clustering of multiple genomic data types using a joint latent variable model with application to breast and lung cancer subtype analysis. Bioinformatics. 2009;25:2906–2912. doi: 10.1093/bioinformatics/btp543.
    1. Murphy G, Pfeiffer R, Camargo MC, Rabkin CS. Meta-analysis shows that prevalence of Epstein-Barr virus-positive gastric cancer differs based on sex and anatomic location. Gastroenterology. 2009;137:824–833. doi: 10.1053/j.gastro.2009.05.001.
    1. Matsusaka K, et al. Classification of Epstein–Barr virus-positive gastric cancers by definition of DNA methylation epigenotypes. Cancer Res. 2011;71:7187–7197. doi: 10.1158/0008-5472.CAN-11-1349.
    1. Geddert H, Zur Hausen A, Gabbert HE, Sarbia M. EBV-infection in cardiac and non-cardiac gastric adenocarcinomas is associated with promoter methylation of p16, p14 and APC, but not hMLH1. Anal. Cell. Pathol. 2010;33:143–149. doi: 10.1155/2010/453764.
    1. Lee J, et al. High-throughput mutation profiling identifies frequent somatic mutations in advanced gastric adenocarcinoma. PLoS ONE. 2012;7:e38892. doi: 10.1371/journal.pone.0038892.
    1. Sukawa Y, et al. Alterations in the human epidermal growth factor receptor 2-phosphatidylinositol 3-kinase-v-Akt pathway in gastric cancer. World J. Gastroenterology. 2012;18:6577–6586. doi: 10.3748/wjg.v18.i45.6577.
    1. Strong MJ, et al. Differences in gastric carcinoma microenvironment stratify according to EBV infection intensity: implications for possible immune adjuvant therapy. PLoS Pathog. 2013;9:e1003341. doi: 10.1371/journal.ppat.1003341.
    1. The Cancer Genome Atlas Network Comprehensive molecular characterization of human colon and rectal cancer. Nature487, 330–337 (2012)
    1. Levine Douglas A. Integrated genomic characterization of endometrial carcinoma. Nature. 2013;497(7447):67–73. doi: 10.1038/nature12113.
    1. Lawrence MS, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499:214–218. doi: 10.1038/nature12213.
    1. Bernal M, Ruiz-Cabello F, Concha A, Paschen A, Garrido F. Implication of the β2-microglobulin gene in the generation of tumor escape phenotypes. Cancer Immunol. Immunother. 2012;61:1359–1371. doi: 10.1007/s00262-012-1321-6.
    1. Greulich H, et al. Functional analysis of receptor tyrosine kinase mutations in lung cancer identifies oncogenic extracellular domain mutations of ERBB2. Proc. Natl Acad. Sci. USA. 2012;109:14476–14481. doi: 10.1073/pnas.1203201109.
    1. Grossmann V, et al. Whole-exome sequencing identifies somatic mutations of BCOR in acute myeloid leukemia with normal karyotype. Blood. 2011;118:6153–6163. doi: 10.1182/blood-2011-07-365320.
    1. Pugh TJ, et al. Medulloblastoma exome sequencing uncovers subtype-specific somatic mutations. Nature. 2012;488:106–110. doi: 10.1038/nature11329.
    1. Dulak AM, et al. Exome and whole-genome sequencing of esophageal adenocarcinoma identifies recurrent driver events and mutational complexity. Nature Genet. 2013;45:478–486. doi: 10.1038/ng.2591.
    1. Ridley AJ, et al. Cell migration: integrating signals from front to back. Science. 2003;302:1704–1709. doi: 10.1126/science.1092053.
    1. Thumkeo D, Watanabe S, Narumiya S. Physiological roles of Rho and Rho effectors in mammals. Eur. J. Cell Biol. 2013;92:303–315. doi: 10.1016/j.ejcb.2013.09.002.
    1. Aznar S, et al. Simultaneous tyrosine and serine phosphorylation of STAT3 transcription factor is involved in Rho A GTPase oncogenic transformation. Mol. Biol. Cell. 2001;12:3282–3294. doi: 10.1091/mbc.12.10.3282.
    1. Yu H, Jove R. The STATs of cancer–new molecular targets come of age. Nature Rev. Cancer. 2004;4:97–105. doi: 10.1038/nrc1275.
    1. Palomero T, et al. Recurrent mutations in epigenetic regulators, RHOA and FYN kinase in peripheral T cell lymphomas. Nature Genet. 2014;46:166–170. doi: 10.1038/ng.2873.
    1. Sakata-Yanagimoto M, et al. Somatic RHOA mutation in angioimmunoblastic T cell lymphoma. Nature Genet. 2014;46:171–175. doi: 10.1038/ng.2872.
    1. Doherty GJ, et al. The endocytic protein GRAF1 is directed to cell-matrix adhesion sites and regulates cell spreading. Mol. Biol. Cell. 2011;22:4380–4389. doi: 10.1091/mbc.e10-12-0936.
    1. Joneson T, White MA, Wigler MH, Bar-Sagi D. Stimulation of membrane ruffling and MAP kinase activation by distinct effectors of RAS. Science. 1996;271:810–812. doi: 10.1126/science.271.5250.810.
    1. Türeci O, et al. Claudin-18 gene structure, regulation, and expression is evolutionary conserved in mammals. Gene. 2011;481:83–92. doi: 10.1016/j.gene.2011.04.007.
    1. Chen BJ, et al. PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies. Clinical Cancer Res. 2013;19:3462–3473. doi: 10.1158/1078-0432.CCR-13-0855.
    1. Green MR, et al. Constitutive AP-1 activity and EBV infection induce PD-L1 in Hodgkin lymphomas and posttransplant lymphoproliferative disorders: implications for targeted therapy. Clinical Cancer Res. 2012;18:1611–1618. doi: 10.1158/1078-0432.CCR-11-1942.
    1. The Cancer Genome Atlas Network Comprehensive molecular characterization of human colon and rectal cancer. Nature487, 330–337 (2012)
    1. Fuchs CS, et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet. 2014;383:31–39. doi: 10.1016/S0140-6736(13)61719-5.
    1. Schaefer CF, et al. PID: the pathway interaction database. Nucleic Acids Res. 2009;37:D674–D679. doi: 10.1093/nar/gkn653.

Source: PubMed

3
Tilaa