Skip to main content
Log in

The Molecular Landscape of Gastric Cancers for Novel Targeted Therapies from Real-World Genomic Profiling

  • Original Research Article
  • Published:
Targeted Oncology Aims and scope Submit manuscript

Abstract

Background

Panel-based comprehensive genomic profiling is used in clinical practice worldwide; however, large real-world datasets of patients with advanced gastric cancer are not well known.

Objective

We investigated what differences exist in clinically relevant alterations for molecularly defined or age-stratified subgroups.

Methods

This was a collaborative biomarker study of a real-world dataset from comprehensive genomic profiling testing (Foundation Medicine, Inc.). Hybrid capture was carried out on at least 324 cancer-related genes and select introns from 31 genes frequently rearranged in cancer. Overall, 4634 patients were available for analyses and were stratified by age (≥ 40/< 40 years), microsatellite instability status, tumor mutational burden status (high 10 ≥ /low < 10 Muts/Mb), Epstein–Barr virus status, and select gene alterations. We analyzed the frequency of alterations with a chi-square test with Yate’s correction.

Results

Genes with frequent alterations included TP53 (60.1%), ARID1A (19.6%), CDKN2A (18.2%), KRAS (16.6%), and CDH1 (15.8%). Differences in comprehensive genomic profiling were observed according to molecularly defined or age-stratified subgroups. Druggable genomic alterations were detected in 31.4% of patients; ATM (4.4%), BRAF V600E (0.4%), BRCA1 (1.5%), BRCA2 (2.9%), ERBB2 amplification (9.2%), IDH1 (0.2%), KRAS G12C (0.7%), microsatellite instability-high (4.8%), NTRK1/2/3 fusion (0.13%), PIK3CA mutation (11.4%), and tumor mutational burden-high (9.4%). CDH1 alterations and MET amplification were significantly more frequent in patients aged < 40 years (27.7 and 6.2%) than in those aged ≥ 40 years (14.7 and 4.0%).

Conclusions

Real-world datasets from clinical panel testing revealed the genomic landscape in gastric cancer by subgroup. These findings provide insights for the current therapeutic strategies and future development of treatments in gastric cancer.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7–33.

    Article  PubMed  Google Scholar 

  2. The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.

    Article  Google Scholar 

  3. Nakamura Y, Kawazoe A, Lordick F, et al. Biomarker-targeted therapies for advanced-stage gastric and gastro-oesophageal junction cancers: an emerging paradigm. Nat Rev Clin Oncol. 2021;18:473–87.

    Article  PubMed  Google Scholar 

  4. Liu Y, Sethi NS, Hinoue T, et al. Comparative molecular analysis of gastrointestinal adenocarcinomas. Cancer Cell. 2018;33:721–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Totoki Y, Saito-Adachi M, Shiraishi Y, et al. Multiancestry genomic and transcriptomic analysis of gastric cancer. Nat Genet. 2023;55:581–94.

    Article  CAS  PubMed  Google Scholar 

  6. Ichikawa H, Nagahashi M, Shimada Y, et al. Actionable gene-based classification toward precision medicine in gastric cancer. Genome Med. 2017;9:93.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Takeda M, Takahama T, Sakai K, et al. Clinical application of the FoundationOne CDx assay to therapeutic decision-making for patients with advanced solid tumors. Oncologist. 2021;26:e588–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Inagaki C, Maeda D, Hatake K, et al. Clinical utility of next-generation sequencing-based panel testing under the universal health-care system in Japan: a retrospective analysis at a single university hospital. Cancers (Basel). 2021;13:1121.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Takeda H, Imoto K, Umemoto K, et al. Clinical utility of genomic profiling tests in patients with advanced gastrointestinal cancers. Target Oncol. 2022;17:177–85.

    Article  PubMed  Google Scholar 

  10. Umemoto K, Yamamoto H, Oikawa R, et al. The molecular landscape of pancreatobiliary cancers for novel targeted therapies from real-world genomic profiling. J Natl Cancer Inst. 2022;114:1279–86.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Kanda M, Terashima M, Kinoshita T, et al. A multi-institutional study to evaluate the feasibility of next-generation sequencing and genomic analysis using formalin-fixed, paraffin-embedded biopsies of gastric cancer. Gastric Cancer. 2023;26:108–15.

    Article  CAS  PubMed  Google Scholar 

  12. Setia N, Wang CX, Lager A, et al. Morphologic and molecular analysis of early-onset gastric cancer. Cancer. 2020;127:103–14.

    Article  PubMed  Google Scholar 

  13. Zhou Q, Tao F, Qiu L, et al. Somatic alteration characteristics of early-onset gastric cancer. J Oncol. 2022;2022:1498053.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Milbury CA, Creeden J, Yip WK, et al. Clinical and analytical validation of FoundationOne®CDx, a comprehensive genomic profiling assay for solid tumors. PLoS ONE. 2022;17: e0264138.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Woodhouse R, Li M, Hughes J, et al. Clinical and analytical validation of FoundationOne Liquid CDx, a novel 324-Gene cfDNA-based comprehensive genomic profiling assay for cancers of solid tumor origin. PLoS ONE. 2020;15: e0237802.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9:34.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Trabucco SE, Gowen K, Maund SL, et al. A novel next-generation sequencing approach to detecting microsatellite instability and pan-tumor characterization of 1000 microsatellite instability-high cases in 67,000 patient samples. J Mol Diagn. 2019;21:1053–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Frampton GM, Fichtenholtz A, Otto GA, et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 2013;31:1023–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lechner M, Frampton GM, Fenton T, et al. Targeted next-generation sequencing of head and neck squamous cell carcinoma identifies novel genetic alterations in HPV+ and HPV− tumors. Genome Med. 2013;5:49.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Sun JX, He Y, Scanford E, et al. A computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal. PLoS Comput Biol. 2018;14:e1005965.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Coleman RL, Oza AM, Lorusso D, et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;390:1949–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Huang X, Wojtowicz D, Przytycka TM. Detecting presence of mutational signatures in cancer with confidence. Bioinformatics. 2018;34:330–7.

    Article  CAS  PubMed  Google Scholar 

  23. Franzese N, Fan J, Sharan R, et al. ScalpelSig designs targeted genomic panels from data to detect activity of mutational signatures. J Comput Biol. 2022;29:56–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Momozawa Y, Sasai R, Usui Y, et al. Expansion of cancer risk profile for BRCA1 and BRCA2 pathogenic variants. JAMA Oncol. 2022;8:871–8.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Marabelle A, Le DT, Ascierto PA, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol. 2020;38:1–10.

    Article  CAS  PubMed  Google Scholar 

  26. Graf RP, Fisher V, Creeden J, et al. Real-world validation of TMB and microsatellite instability as predictive biomarkers of immune checkpoint inhibitor effectiveness in advanced gastroesophageal cancer. Cancer Res Commun. 2022;2:1037–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chao J, Fuchs CS, Shitara K, et al. Assessment of pembrolizumab therapy for the treatment of microsatellite instability-high gastric or gastroesophageal junction cancer among patients in the KEYNOTE-059, KEYNOTE-061, and KEYNOTE-062 clinical trials. JAMA Oncol. 2021;7:895–902.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Shitara K, Özgüroglu M, Bang YJ, et al. Molecular determinants of clinical outcomes with pembrolizumab versus paclitaxel in a randomized, open-label, phase III trial in patients with gastroesophageal adenocarcinoma. Ann Oncol. 2021;32:1127–36.

    Article  CAS  PubMed  Google Scholar 

  29. Wu YM, Cieslik M, Lonigro RJ, et al. Inactivation of CDK12 delineates a distinct immunogenic class of advanced prostate cancer. Cell. 2018;173:1770-82.e14.

    Article  CAS  PubMed  Google Scholar 

  30. Sokol ES, Pavlick D, Frampton GM, et al. Pan-cancer analysis of CDK12 loss-of-function alterations and their association with the focal tandem-duplicator phenotype. Oncologist. 2019;12:1526–33.

    Article  Google Scholar 

  31. Liu H, Liu K, Dong Z. Targeting CDK12 for cancer therapy: function, mechanism, and drug discovery. Cancer Res. 2021;81:18–26.

    Article  CAS  PubMed  Google Scholar 

  32. Zhou X, Ji Y, Zhou J. Multiple strategies to develop small molecular KRAS directly bound inhibitors. Molecules. 2023;28:3615.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Saito M, Kono K. Landscape of EBV-positive gastric cancer. Gastric Cancer. 2021;24:983–9.

    Article  CAS  PubMed  Google Scholar 

  34. Gu Y, Zhang P, Wang J, et al. Somatic ARID1A mutation stratifies patients with gastric cancer to PD-1 blockade and adjuvant chemotherapy. Cancer Immunol Immunother. 2023;72:1199–208.

    Article  CAS  PubMed  Google Scholar 

  35. Carneiro F. Familial and hereditary gastric cancer, an overview. Best Pract Res Clin Gastroenterol. 2022;58–59: 101800.

    Article  PubMed  Google Scholar 

  36. Tanaka Y, Chiwaki F, Kojima S, et al. Multi-omic profiling of peritoneal metastases in gastric cancer identifies molecular subtypes and therapeutic vulnerabilities. Nat Cancer. 2021;2:962–77.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Hiroyuki Yamamoto.

Ethics declarations

Funding

No external funding was used in the preparation of this article.

Conflict of interest

Kumiko Umemoto has received honoraria from Chugai Pharmaceutical. Naoki Izawa has recived honoraria form Chugai Pharmaceutical. Jay A. Moore and Ethan S. Sokol are employees of Foundation Medicine, Inc. and shareholders in Roche. Yu Sunakawa has recived honoraria and research funding from Chugai Pharmaceutical. Hiroyuki Yamamoto, Hiroyuki Arai, Ritsuko Oikawa, Hiroyuki Takeda, Takuro Mizukami, Yohei Kubota, Ayako Doi, Yoshiki Horie, Takashi Ogura, and Naoki Izawa have no conflicts of interest that are directly relevant to the content of this article.

Ethics approval

Approval for this study, including a waiver of informed consent and a Health Insurance Portability and Accountability Act waiver of authorization, was obtained from the Western Institutional Review Board (protocol #20152817). This study was also approved by the Ethics Committee of St. Marianna University School of Medicine.

Consent to participate

Not applicable.

Consent for publication

Not applicable.

Availability of data and material

The sequencing data generated in this study are derived from clinical samples. The data supporting the findings of this study are provided within the paper and its supplementary files. All supplementary information accompanying the different analyses and figures presented in this study are provided in the ESM. Because of Health Insurance Portability and Accountability Act requirements, we are not consented to share individualized patient genomic data that contain potentially identifying or sensitive patient information. Foundation Medicine, Inc. is committed to collaborative data analysis, and we have well-established and widely utilized mechanisms by which investigators can query our core genomic database of > 600,000 de-identified sequenced cancers to obtain aggregated datasets. Requests for collaborative datashares can be made by contacting the corresponding author(s) and completing a study review committee form. Once approved, investigators are required to sign a data transfer agreement. Written proposals are considered at monthly meetings and data transfer agreements expire 18 months from execution of the agreement.

Code availability

Not applicable.

Author contributions

HY designed the study, performed the analyses, and interpreted the data. RO performed the analyses and interpreted the data. YS designed the study and interpreted the data. JM and ES collected the data, performed the analyses, and interpreted the data. All authors contributed to the writing and approved the final manuscript.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (PDF 580 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yamamoto, H., Arai, H., Oikawa, R. et al. The Molecular Landscape of Gastric Cancers for Novel Targeted Therapies from Real-World Genomic Profiling. Targ Oncol (2024). https://doi.org/10.1007/s11523-024-01052-1

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s11523-024-01052-1

Navigation