Skip to main content

Advertisement

Log in

Immunotherapy in Gastroesophageal Cancers: Current Evidence and Ongoing Trials

  • Upper Gastrointestinal Cancers (JD Berlin, Section Editor)
  • Published:
Current Treatment Options in Oncology Aims and scope Submit manuscript

Opinion statement

Data supporting the use of immunotherapy in the treatment of gastroesophageal cancer continues to evolve. The promising results from adjuvant immunotherapy and trials combining immunotherapy plus chemotherapy in the 1L setting have led to broad US FDA approvals. Among the PD-L1 negative subgroups, the magnitude of benefit is diminished; effective therapy for this population remains an unmet need. A detailed biologic understanding of the PD-L1 negative (and low) population represents a barrier to developing effective combination therapies, although combination angiogenesis inhibitors and immunotherapy look encouraging. Early phase clinical trials, particularly with pembrolizumab plus lenvatinib (EPOC 1706), demonstrated a clear signal independent of PD-L1, and a confirmatory phase III trial of pembrolizumab plus lenvatinib is planned. Conceptually, it is important to think of immune checkpoint inhibitor therapy as targeted therapy, most active in clearly defined biomarker-selected populations. Pre-planned analyses have reliably shown a clear trend toward a greater magnitude of benefit in patients with higher PD-L1 expression, particularly CPS ≥ 5 and ≥ 10. Whether there is a linear relationship at higher cutoffs is not well known, though it likely represents smaller and smaller populations. Although beyond the scope of this clinically oriented review, recognition of the spatial and temporal heterogeneity in PD-L1 expression is important and repeat testing from progression samples across lines of therapy should be considered. Questions about additional predictive biomarkers, particularly plasma-derived, remain. Responses by tumor histology and location also differ, and special attention to these factors as well as MSI-H, HER2, and EBV subgroups in future trials is warranted. Questions regarding the incorporation of immunotherapy after progression on 1L immunotherapy plus chemotherapy combinations will arise as these combinations are used more frequently, and this represents a key area of future investigation. Overall, the role of immunotherapy continues to expand in GEA, and we welcome any additional tools for this difficult-to-treat group of cancers.

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.

Similar content being viewed by others

Data Availability

Not applicable.

Code Availability

Not applicable.

References and Recommended Reading

  1. Torre LA, et al. Global cancer incidence and mortality rates and trends—an update. Cancer Epidemiol Biomarkers Prev. 2016;25(1):16–27.

    Article  PubMed  Google Scholar 

  2. Shah MA. Update on metastatic gastric and esophageal cancers. J Clin Oncol. 2015;33(16):1760–9.

    Article  CAS  PubMed  Google Scholar 

  3. Dong H, et al. B7–H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat Med. 1999;5(12):1365–9.

    Article  CAS  PubMed  Google Scholar 

  4. Freeman GJ, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192(7):1027–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol. 2005;23:515–48.

    Article  PubMed  CAS  Google Scholar 

  6. Nishimura H, et al. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999;11(2):141–51.

    Article  CAS  PubMed  Google Scholar 

  7. Dong H, et al. Tumor-associated B7–H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8(8):793–800.

    Article  CAS  PubMed  Google Scholar 

  8. Iwai Y, et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci USA. 2002;99(19):12293–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Peggs KS, et al. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med. 2009;206(8):1717–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Khunger M, et al. Programmed cell death 1 (PD-1) ligand (PD-L1) expression in solid tumors as a predictive biomarker of benefit from PD-1/PD-L1 axis inhibitors: a systematic review and meta-analysis. JCO Precis Oncol. 2017;1:1–15.

    PubMed  Google Scholar 

  12. Zhang M, et al. The clinicopathological and prognostic significance of PD-L1 expression in gastric cancer: a meta-analysis of 10 studies with 1,901 patients. Sci Rep. 2016;6:37933.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Huang B, et al. The expression status and prognostic significance of programmed cell death 1 ligand 1 in gastrointestinal tract cancer: a systematic review and meta-analysis. Onco Targets Ther. 2015;8:2617–25.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Dai C, et al. Prognostic and predictive values of PD-L1 expression in patients with digestive system cancer: a meta-analysis. Onco Targets Ther. 2017;10:3625–34.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Gu L, et al. PD-L1 and gastric cancer prognosis: a systematic review and meta-analysis. PLoS ONE. 2017. https://doi.org/10.1371/journal.pone.0182692.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Liu YX, et al. Prognostic significance of PD-L1 expression in patients with gastric cancer in East Asia: a meta-analysis. Onco Targets Ther. 2016;9:2649–54.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Wu P, et al. PD-L1 and survival in solid tumors: a meta-analysis. PLoS ONE. 2015. https://doi.org/10.1371/journal.pone.0131403.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Xu F, et al. Clinicopathologic significance and prognostic value of B7 homolog 1 in gastric cancer: a systematic review and meta-analysis. Medicine (Baltimore). 2015. https://doi.org/10.1097/MD.0000000000001911.

    Article  PubMed Central  Google Scholar 

  19. Zhang W, et al. Induction of PD-L1 expression by epidermal growth factor receptor-mediated signaling in esophageal squamous cell carcinoma. Onco Targets Ther. 2017;10:763–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016;17(12):e542–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Antonia SJ, Ozguroglu M. Durvalumab in stage III non-small-cell lung cancer. N Engl J Med. 2018;378(9):869–70.

    PubMed  Google Scholar 

  22. Schmid P, et al. Pembrolizumab for early triple-negative breast cancer. N Engl J Med. 2020;382(9):810–21.

    Article  CAS  PubMed  Google Scholar 

  23. Kelly RJ, et al. LBA9_PR Adjuvant nivolumab in resected esophageal or gastroesophageal junction cancer (EC/GEJC) following neoadjuvant chemoradiation therapy (CRT): first results of the CheckMate 577 study. Ann Oncol. 2020;31:S1193–4.

    Article  Google Scholar 

  24. Bang YJ, et al. KEYNOTE-585: Phase III study of perioperative chemotherapy with or without pembrolizumab for gastric cancer. Future Oncol. 2019;15(9):943–52.

    Article  CAS  PubMed  Google Scholar 

  25. Mamdani H, et al. Safety and efficacy of durvalumab following trimodality therapy for locally advanced esophageal and GEJ adenocarcinoma: early efficacy results from Big Ten Cancer Research Consortium study. J Clin Oncol. 2019;37(4_suppl):5–5.

    Article  Google Scholar 

  26. N.C.C. Network. Esophageal and esophagogastric junction cancers (Version 1.2021). January 1, 2021]; Available from: https://www.nccn.org/professionals/physician_gls/pdf/esophageal.pdf.

  27. Kato K, et al. LBA8_PR Pembrolizumab plus chemotherapy versus chemotherapy as first-line therapy in patients with advanced esophageal cancer: the phase 3 KEYNOTE-590 study. Ann Oncol. 2020;31:S1192–3.

    Article  Google Scholar 

  28. Shitara K, et al. Efficacy and safety of pembrolizumab or pembrolizumab plus chemotherapy vs chemotherapy alone for patients with first-line, advanced gastric cancer: the KEYNOTE-062 phase 3 randomized clinical trial. JAMA Oncol. 2020;6(10):1571–80.

    Article  PubMed  Google Scholar 

  29. Moehler M, et al. LBA6_PR Nivolumab (nivo) plus chemotherapy (chemo) versus chemo as first-line (1L) treatment for advanced gastric cancer/gastroesophageal junction cancer (GC/GEJC)/esophageal adenocarcinoma (EAC): first results of the CheckMate 649 study. Ann Oncol. 2020;31:S1191.

    Article  Google Scholar 

  30. Boku N, et al. LBA7_PR Nivolumab plus chemotherapy versus chemotherapy alone in patients with previously untreated advanced or recurrent gastric/gastroesophageal junction (G/GEJ) cancer: ATTRACTION-4 (ONO-4538-37) study. Ann Oncol. 2020;31:S1192.

    Article  Google Scholar 

  31. Kato K, et al. KEYNOTE-590: phase III study of first-line chemotherapy with or without pembrolizumab for advanced esophageal cancer. Future Oncol. 2019;15(10):1057–66.

    Article  CAS  PubMed  Google Scholar 

  32. Moehler MH, et al. CheckMate 649: a randomized, multicenter, open-label, phase III study of nivolumab (NIVO) + ipilimumab (IPI) or nivo + chemotherapy (CTX) versus CTX alone in patients with previously untreated advanced (Adv) gastric (G) or gastroesophageal junction (GEJ) cancer. J Clin Oncol. 2018. https://doi.org/10.1200/JCO.2018.36.4_suppl.TPS192.

    Article  Google Scholar 

  33. Network, N.C.C., Gastric cancer (Version 1.2021).

  34. Charalambous H, et al. P1.01–12 switch maintenance pembrolizumab in patients with metastatic non small cell lung cancer (SWIPE). J Thorac Oncol. 2018;13(10):S463–4.

    Article  Google Scholar 

  35. Ciuleanu T, et al. Maintenance pemetrexed plus best supportive care versus placebo plus best supportive care for non-small-cell lung cancer: a randomised, double-blind, phase 3 study. Lancet. 2009;374(9699):1432–40.

    Article  CAS  PubMed  Google Scholar 

  36. Paz-Ares LG, et al. PARAMOUNT: final overall survival results of the phase III study of maintenance pemetrexed versus placebo immediately after induction treatment with pemetrexed plus cisplatin for advanced nonsquamous non-small-cell lung cancer. J Clin Oncol. 2013;31(23):2895–902.

    Article  CAS  PubMed  Google Scholar 

  37. Chung HC, et al. Avelumab (anti-PD-L1) as first-line switch-maintenance or second-line therapy in patients with advanced gastric or gastroesophageal junction cancer: phase 1b results from the JAVELIN Solid Tumor trial. J Immunother Cancer. 2019;7(1):30.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Moehler M, et al. Phase III trial of avelumab maintenance after first-line induction chemotherapy versus continuation of chemotherapy in patients with gastric cancers: results from JAVELIN Gastric 100. J Clin Oncol. 2020. https://doi.org/10.1200/JCO.20.00892.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Shitara K, et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet. 2018;392(10142):123–33.

    Article  CAS  PubMed  Google Scholar 

  40. Fuchs CS, et al. Pembrolizumab versus paclitaxel for previously treated patients with PD-L1–positive advanced gastric or gastroesophageal junction cancer (GC): update from the phase III KEYNOTE-061 trial. J Clin Oncol. 2020;38(15_suppl):4503–4503.

    Article  Google Scholar 

  41. Wainberg ZA, et al. Efficacy of pembrolizumab monotherapy for advanced gastric/gastroesophageal junction cancer with programmed death ligand 1 combined positive score >/=10. Clin Cancer Res. 2021. https://doi.org/10.1158/1078-0432.CCR-20-2980.

    Article  PubMed  Google Scholar 

  42. Kojima T, et al. Pembrolizumab versus chemotherapy as second-line therapy for advanced esophageal cancer: phase III KEYNOTE-181 study. J Clin Oncol. 2019. https://doi.org/10.1200/JCO.2019.37.4_suppl.2.

    Article  Google Scholar 

  43. FDA/CDER. FDA approves pembrolizumab for advanced esophageal squamous cell cancer. [cited 2021 January 1]; Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-pembrolizumab-advanced-esophageal-squamous-cell-cancer.

  44. Kato K, et al. Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019;20(11):1506–17.

    Article  CAS  PubMed  Google Scholar 

  45. FDA/CDER. FDA approves nivolumab for esophageal squamous cell carcinoma. [cited 2021 January 1]; Available from: https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-nivolumab-esophageal-squamous-cell-carcinoma.

  46. Kang YK, et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;390(10111):2461–71.

    Article  CAS  PubMed  Google Scholar 

  47. Gravalos C, Jimeno A. HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol. 2008;19(9):1523–9.

    Article  CAS  PubMed  Google Scholar 

  48. Hofmann M, et al. Assessment of a HER2 scoring system for gastric cancer: results from a validation study. Histopathology. 2008;52(7):797–805.

    Article  CAS  PubMed  Google Scholar 

  49. Tanner M, et al. Amplification of HER-2 in gastric carcinoma: association with Topoisomerase IIalpha gene amplification, intestinal type, poor prognosis and sensitivity to trastuzumab. Ann Oncol. 2005;16(2):273–8.

    Article  CAS  PubMed  Google Scholar 

  50. Janjigian YY, et al. First-line pembrolizumab (P), trastuzumab (T), capecitabine (C) and oxaliplatin (O) in HER2-positive metastatic esophagogastric adenocarcinoma (mEGA). J Clin Oncol. 2019. https://doi.org/10.1200/JCO.2019.37.4_suppl.62.

    Article  PubMed  Google Scholar 

  51. Chung HC, et al. KEYNOTE-811 pembrolizumab plus trastuzumab and chemotherapy for HER2+ metastatic gastric or gastroesophageal junction cancer (mG/GEJc): a double-blind, randomized, placebo-controlled phase III study. J Clin Oncol. 2020. https://doi.org/10.1200/JCO.2020.38.4_suppl.TPS463.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Rha SY, et al. A multi-institutional phase Ib/II trial of first-line triplet regimen (pembrolizumab, trastuzumab, chemotherapy) for HER2-positive advanced gastric and gastroesophageal junction cancer (PANTHERA Trial): molecular profiling and clinical update. J Clin Oncol. 2021. https://doi.org/10.1200/JCO.2021.39.3_suppl.218.

    Article  PubMed  Google Scholar 

  53. Catenacci DVT, et al. Margetuximab plus pembrolizumab in patients with previously treated, HER2-positive gastro-oesophageal adenocarcinoma (CP-MGAH22-05): a single-arm, phase 1b–2 trial. Lancet Oncol. 2020;21(8):1066–76.

    Article  CAS  PubMed  Google Scholar 

  54. Cristescu R, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21(5):449–56.

    Article  CAS  PubMed  Google Scholar 

  55. Derks S, et al. Abundant PD-L1 expression in Epstein-Barr Virus-infected gastric cancers. Oncotarget. 2016;7(22):32925–32.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Ma C, et al. Programmed death-ligand 1 expression is common in gastric cancer associated with Epstein-Barr virus or microsatellite instability. Am J Surg Pathol. 2016;40(11):1496–506.

    Article  PubMed  Google Scholar 

  57. Kim ST, et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat Med. 2018;24(9):1449–58.

    Article  CAS  PubMed  Google Scholar 

  58. Mishima S, et al. Clinicopathological and molecular features of responders to nivolumab for patients with advanced gastric cancer. J Immunother Cancer. 2019;7(1):24.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Liu X, Meltzer SJ. Gastric cancer in the era of precision medicine. Cell Mol Gastroenterol Hepatol. 2017;3(3):348–58.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Rodriquenz MG, et al. MSI and EBV positive gastric cancer’s subgroups and their link with novel immunotherapy. J Clin Med. 2020;9(5):1427.

    Article  CAS  PubMed Central  Google Scholar 

  61. Baniak N, et al. Gastric biomarkers: a global review. World J Surg Oncol. 2016;14(1):212.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Mathiak M, et al. Clinicopathologic characteristics of microsatellite instable gastric carcinomas revisited: urgent need for standardization. Appl Immunohistochem Mol Morphol. 2017;25(1):12–24.

    Article  PubMed  Google Scholar 

  63. Choi YY, et al. Microsatellite instability and programmed cell death-ligand 1 expression in stage II/III gastric cancer: post hoc analysis of the CLASSIC randomized controlled study. Ann Surg. 2019;270(2):309–16.

    Article  PubMed  Google Scholar 

  64. Smyth EC, et al. Mismatch repair deficiency, microsatellite instability, and survival: an exploratory analysis of the medical research council adjuvant gastric infusional chemotherapy (MAGIC) trial. JAMA Oncol. 2017;3(9):1197–203.

    Article  PubMed  Google Scholar 

  65. Trials.Gov, C. Phase II trial of neoadjuvant pembrolizumab for patients with early stage gastroesophageal adenocarcinoma [cited 2021 January 1]; Available from: https://clinicaltrials.gov/ct2/show/NCT04089904.

  66. Hong MH, et al. A phase II trial of preoperative chemoradiotherapy and pembrolizumab for locally advanced esophageal squamous cell carcinoma (ESCC). J Clin Oncol. 2019;37(15_suppl):4027–4027.

    Article  Google Scholar 

  67. Kelly RJ, et al. Neoadjuvant nivolumab plus concurrent chemoradiation in stage II/III esophageal/gastroesophageal junction cancer. I Clin Oncol. 2019;37(4_suppl):142–142.

    Google Scholar 

  68. Greally M, et al. Phase Ib/II trial of durvalumab and chemoradiation (CRT) with carboplatin/paclitaxel for esophageal and gastroesophageal junction (GEJ) adenocarcinoma. J Clin Oncol. 2018. https://doi.org/10.1200/JCO.2018.36.4_suppl.172.

    Article  Google Scholar 

  69. Mamdani H, et al. Safety and efficacy of durvalumab following multimodality therapy for locally advanced esophageal and GEJ adenocarcinoma: two-year follow-up results from Big Ten Cancer Research Consortium study. J Clin Oncol. 2020. https://doi.org/10.1200/JCO.2020.38.4_suppl.404.

    Article  Google Scholar 

  70. Bang YJ, et al. Phase III, randomised trial of avelumab versus physician’s choice of chemotherapy as third-line treatment of patients with advanced gastric or gastro-oesophageal junction cancer: primary analysis of JAVELIN Gastric 300. Ann Oncol. 2018;29(10):2052–60.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Fuchs CS, et al. Safety and efficacy of pembrolizumab monotherapy in patients with previously treated advanced gastric and gastroesophageal junction cancer: phase 2 clinical KEYNOTE-059 trial. JAMA Oncol. 2018. https://doi.org/10.1001/jamaoncol.2018.0013.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Shah MA, et al. Efficacy and safety of pembrolizumab for heavily pretreated patients with advanced, metastatic adenocarcinoma or squamous cell carcinoma of the esophagus: the phase 2 KEYNOTE-180 study. JAMA Oncol. 2019;5(4):546–50.

    Article  PubMed  Google Scholar 

Download references

Funding

AGA Research Foundation’s AGA-Gastric Cancer Foundation Ben Feinstein Memorial Research Scholar Award in Gastric Cancer—AGA2020-13-02 (SJK), and Stand-Up-2-Cancer Gastric Cancer Interception Award (SJK).

Author information

Authors and Affiliations

Authors

Contributions

J.H. and K.P. performed the primary literature search and prepared the manuscript. S.J.K. conceived the concept for this manuscript and provided meaningful feedback and guidance on the manuscript. J.G., M.C., M.M., and A.P. critically revised the work.

Corresponding author

Correspondence to Samuel Klempner MD.

Ethics declarations

Conflict of interest

Jasmine Huynh declares that she has no conflict of interest. Kanishka Patel declares that she has no conflict of interest. Jun Gong has received compensation for service as a consultant from Exelixis, QED Therapeutics, Elsevier, Basilea, and HalioDx. May Cho has received compensation for service on advisory boards for Amgen, Eisai, Taiho, Astellas, Exelixis, Ipsen, Seagen, QED Therapeutics, AstraZeneca, Basilea, and Genentech/Roche, and serves on the speaker’s bureau for Pfizer and Taiho. Midhun Malla is supported, in part, by a grant from the NIH National Institute of General Medical Sciences (Grant #5U54GM104942-05); and has received compensation for service as a consultant/advisor from AstraZeneca, QED Therapeutics, Omni Health, and Curio Science. Aparna Parikh has received institutional research funding from PureTech Health, PMV Pharmaceuticals, Plexxikon, Takeda, Bristol-Myers Squibb, and Novartis; has received compensation for service as a consultant/advisor from Natera, Foundation Medicine, PureTech Health, Checkmate Pharmaceuticals, Eli Lilly, and Pfizer; serves on the DSMC for Roche; and holds equity in C2i Genomics. Samuel Klempner has received compensation for service as a consultant/advisor from Eli Lilly, Bristol-Myers Squibb, Merck, Astellas, Daiichi-Sankyo, Natera, and Pieris, and owns stock/equity in Turning Point Therapeutics.

Ethical Approval

Not applicable.

Consent to Participate

Not applicable.

Consent for Publication

Not applicable.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on Upper Gastrointestinal Cancers

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huynh, J., Patel, K., Gong, J. et al. Immunotherapy in Gastroesophageal Cancers: Current Evidence and Ongoing Trials. Curr. Treat. Options in Oncol. 22, 100 (2021). https://doi.org/10.1007/s11864-021-00893-6

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s11864-021-00893-6

Keywords

Navigation