Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes

Abstract

Gastric cancer, a leading cause of cancer-related deaths, is a heterogeneous disease. We aim to establish clinically relevant molecular subtypes that would encompass this heterogeneity and provide useful clinical information. We use gene expression data to describe four molecular subtypes linked to distinct patterns of molecular alterations, disease progression and prognosis. The mesenchymal-like type includes diffuse-subtype tumors with the worst prognosis, the tendency to occur at an earlier age and the highest recurrence frequency (63%) of the four subtypes. Microsatellite-unstable tumors are hyper-mutated intestinal-subtype tumors occurring in the antrum; these have the best overall prognosis and the lowest frequency of recurrence (22%) of the four subtypes. The tumor protein 53 (TP53)-active and TP53-inactive types include patients with intermediate prognosis and recurrence rates (with respect to the other two subtypes), with the TP53-active group showing better prognosis. We describe key molecular alterations in each of the four subtypes using targeted sequencing and genome-wide copy number microarrays. We validate these subtypes in independent cohorts in order to provide a consistent and unified framework for further clinical and preclinical translational research.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The four distinct subtypes in GC.
Figure 2: Molecular subtype and survival association.
Figure 3: Molecular alteration landscape of ACRG GC.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

References

  1. Jemal, A. et al. Global cancer statistics. CA Cancer J. Clin. 61, 69–90 (2011).

    Article  PubMed  Google Scholar 

  2. Edge, S. et al. (eds.). AJCC Cancer Staging Manual (Springer, New York, 2010).

  3. Lee, J. et al. Phase III trial comparing capecitabine plus cisplatin versus capecitabine plus cisplatin with concurrent capecitabine radiotherapy in completely resected gastric cancer with D2 lymph node dissection: the ARTIST trial. J. Clin. Oncol. 30, 268–273 (2012).

    Article  CAS  PubMed  Google Scholar 

  4. Kim, S. et al. An observational study suggesting clinical benefit for adjuvant postoperative chemoradiation in a population of over 500 cases after gastric resection with D2 nodal dissection for adenocarcinoma of the stomach. Int. J. Radiat. Oncol. Biol. Phys. 63, 1279–1285 (2005).

    Article  PubMed  Google Scholar 

  5. Bang, Y.J. et al. Adjuvant capecitabine and oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): a phase 3 open-label, randomised controlled trial. Lancet 379, 315–321 (2012).

    Article  CAS  PubMed  Google Scholar 

  6. Macdonald, J.S. et al. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N. Engl. J. Med. 345, 725–730 (2001).

    Article  CAS  PubMed  Google Scholar 

  7. Wang, G. et al. Comparison of global gene expression of gastric cardia and noncardia cancers from a high-risk population in China. PLoS ONE 8, e63826 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Shah, M.A. et al. Molecular classification of gastric cancer: a new paradigm. Clin. Cancer Res. 17, 2693–2701 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Tay, S.T. et al. A combined comparative genomic hybridization and expression microarray analysis of gastric cancer reveals novel molecular subtypes. Cancer Res. 63, 3309–3316 (2003).

    CAS  PubMed  Google Scholar 

  10. Cancer Genome Atlas Research Network. et al. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513, 202–209 (2014).

  11. Chen, X. et al. Variation in gene expression patterns in human gastric cancers. Mol. Biol. Cell 14, 3208–3215 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Tan, I.B. et al. Intrinsic subtypes of gastric cancer, based on gene expression pattern, predict survival and respond differently to chemotherapy. Gastroenterology 141, 476–485 (2011).

    Article  PubMed  Google Scholar 

  13. Cho, J.Y. et al. Gene expression signature-based prognostic risk score in gastric cancer. Clin. Cancer Res. 17, 1850–1857 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Deng, N. et al. A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets. Gut 61, 673–684 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. An, C. et al. Prognostic significance of CpG island methylator phenotype and microsatellite instability in gastric carcinoma. Clin. Cancer Res. 11, 656–663 (2005).

    CAS  PubMed  Google Scholar 

  16. Liu, Z. et al. Large-scale characterization of DNA methylation changes in human gastric carcinomas with and without metastasis. Clin. Cancer Res. 20, 4598–4612 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zouridis, H. et al. Methylation subtypes and large-scale epigenetic alterations in gastric cancer. Sci. Transl. Med. 4, 156ra140 (2012).

    Article  CAS  PubMed  Google Scholar 

  18. Wang, K. et al. Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat. Genet. 46, 573–582 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Kakiuchi, M. et al. Recurrent gain-of-function mutations of RHOA in diffuse-type gastric carcinoma. Nat. Genet. 46, 583–587 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. Liu, J. et al. Integrated exome and transcriptome sequencing reveals ZAK isoform usage in gastric cancer. Nat. Commun. 5, 3830 (2014).

    Article  CAS  PubMed  Google Scholar 

  21. Ooi, C.H. et al. Oncogenic pathway combinations predict clinical prognosis in gastric cancer. PLoS Genet. 5, e1000676 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Wu, Y. et al. Comprehensive genomic meta-analysis identifies intra-tumoural stroma as a predictor of survival in patients with gastric cancer. Gut 62, 1100–1111 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Wong, S.S. et al. Genomic landscape and genetic heterogeneity in gastric adenocarcinoma revealed by whole-genome sequencing. Nat. Commun. 5, 5477 (2014).

    Article  PubMed  Google Scholar 

  24. Jolliffe, I. Principal Component Analysis (Wiley Online Library, 2002).

  25. Loboda, A. et al. EMT is the dominant program in human colon cancer. BMC Med. Genomics 4, 9 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  26. CancerGenomeAtlasResearchNetwork. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487, 330–337 (2012).

  27. Coppola, D. et al. Unique ectopic lymph node-like structures present in human primary colorectal carcinoma are identified by immune gene array profiling. Am. J. Pathol. 179, 37–45 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Dai, H. et al. A cell proliferation signature is a marker of extremely poor outcome in a subpopulation of breast cancer patients. Cancer Res. 65, 4059–4066 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Cancer Genome Atlas Research Network. et al. Comprehensive molecular portraits of human breast tumours. Nature 490, 61–70 (2012).

  30. Benita, Y. et al. Gene enrichment profiles reveal T cell development, differentiation, and lineage-specific transcription factors including ZBTB25 as a novel NF-AT repressor. Blood 115, 5376–5384 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lee, J. et al. Nanostring-based multigene assay to predict recurrence for gastric cancer patients after surgery. PLoS ONE 9, e90133 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Mori, Y. et al. Instabilotyping reveals unique mutational spectra in microsatellite-unstable gastric cancers. Cancer Res. 62, 3641–3645 (2002).

    CAS  PubMed  Google Scholar 

  33. Mori, Y. et al. The impact of microsatellite instability on the molecular phenotype of colorectal tumors. Cancer Res. 63, 4577–4582 (2003).

    CAS  PubMed  Google Scholar 

  34. Lei, Z. et al. Identification of molecular subtypes of gastric cancer with different responses to PI3-kinase inhibitors and 5-fluorouracil. Gastroenterology 145, 554–565 (2013).

    Article  CAS  PubMed  Google Scholar 

  35. Russell, S. et al. Immune cell infiltration patterns and survival in head and neck squamous cell carcinoma. Head Neck Oncol. 5, 24 (2013).

    PubMed  Google Scholar 

  36. Sørlie, T. et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc. Natl. Acad. Sci. USA 98, 10869–10874 (2001).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Higgins, M.J. & Baselga, J. Targeted therapies for breast cancer. J. Clin. Invest. 121, 3797–3803 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Roepman, P. et al. Colorectal cancer intrinsic subtypes predict chemotherapy benefit, deficient mismatch repair and epithelial-to-mesenchymal transition. Int. J. Cancer. 134, 552–562 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Salazar, R. et al. Gene expression signature to improve prognosis prediction of stage II and III colorectal cancer. J. Clin. Oncol. 29, 17–24 (2011).

    Article  PubMed  Google Scholar 

  40. Li, T., Kung, H.J., Mack, P.C. & Gandara, D.R. Genotyping and genomic profiling of non-small-cell lung cancer: implications for current and future therapies. J. Clin. Oncol. 31, 1039–1049 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Cancer Genome Atlas Research Network. Integrated genomic characterization of endometrial carcinoma. Nature 497, 67–73 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ciriello, G. et al. Emerging landscape of oncogenic signatures across human cancers. Nat. Genet. 45, 1127–1133 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bang, Y.J. et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376, 687–697 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Barretina, J. et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483, 603–607 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Suraweera, N. et al. Evaluation of tumor microsatellite instability using five quasimonomorphic mononucleotide repeats and pentaplex PCR. Gastroenterology 123, 1804–1811 (2002).

    Article  CAS  PubMed  Google Scholar 

  46. Song, H.J. et al. Host inflammatory response predicts survival of patients with Epstein-Barr virus-associated gastric carcinoma. Gastroenterology 139, 84–92 (2010).

    Article  PubMed  Google Scholar 

  47. Rüschoff, J. et al. HER2 testing in gastric cancer: a practical approach. Mod. Pathol. 25, 637–650 (2012).

    Article  CAS  PubMed  Google Scholar 

  48. Greenman, C.D. et al. PICNIC: an algorithm to predict absolute allelic copy number variation with microarray cancer data. Biostatistics 11, 164–175 (2010).

    Article  PubMed  Google Scholar 

  49. Lee, J. et al. Prognostic model to predict survival following first-line chemotherapy in patients with metastatic gastric adenocarcinoma. Ann. Oncol. 18, 886–891 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Grambsch, P.M. & Therneau, T.M. Proportional hazards tests and diagnostics based on weighted residuals. Biometrika 81, 515–526 (1994).

    Article  Google Scholar 

Download references

Acknowledgements

The Asian Cancer Research Group (ACRG, http://www.asiancancerresearchgroup.org/about_us.html) is a non-profit consortium of pharmaceutical industry and academic medical centers and genomics companies dedicated to accelerating research, and ultimately improving treatment for patients affected with the most commonly-diagnosed cancers in Asia. ACRG seeks to increase and improve the knowledge of cancers prevalent in Asia by generating comprehensive genomic data sets and sharing them freely with the scientific community in order to accelerate drug discovery efforts. This study is primarily supported by the ACRG. Samsung Medical Center provided partial support through grant no. GF01140111 (K.-M.K. and J. Lee).

Author information

Authors and Affiliations

Authors

Contributions

J. Lee, C.R., A.A. and S.K. conceptualized and planned the study. M.G.C., T.S.S., J.H.L., S.T.K., W.K.K., S.H.P. and J.M.B. contributed to collection of surgical samples and associated clinical information. K.-M.K. and I.-G.D. conducted the pathology assessment. A.A., K.-M.K., A.L. and J. Lee coordinated the data generation and led the data analysis. L.G. and S.L. generated the targeted sequencing data. S.S.W. analyzed the sequencing data. J.G.J. and J.F. generated the Affymetrix gene expression and Affymetrix SNP6 data. R. Cristescu, M.N., J.C.T., K.Y., J.W., Y.G.Y., J. Liu and A.L. processed, analyzed and participated in discussions related to the genomics data. I.S. and S.-H.J. conducted the statistical analysis of the clinical data. P.T., J.H., R. Chen, X.S.Y., M.A. and D.H. participated in discussions, provided critical scientific input, analysis suggestions and logistical support toward the project. R. Cristescu, J. Lee, M.N., K.-M.K. and A.A. wrote the manuscript.

Corresponding authors

Correspondence to Andrey Loboda, Sung Kim or Amit Aggarwal.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10 and Supplementary Table 1 (PDF 1209 kb)

Supplementary Data 1

Clinical Information ACRG Cohort (XLS 216 kb)

Supplementary Data 2

Case Summary and Mean Median survival time (all cohorts) (XLSX 19 kb)

Supplementary Data 3

Summarized Clinical Information TCGA SINGAPORE SMC2 cohort (XLSX 23 kb)

Supplementary Data 4

Targeted sequencing data (XLSX 376 kb)

Supplementary Data 5

Gene Signatures (XLSX 20 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cristescu, R., Lee, J., Nebozhyn, M. et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med 21, 449–456 (2015). https://doi.org/10.1038/nm.3850

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3850

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer