How do we improve chemoimmunotherapy in gastroesophageal adenocarcinoma?
Editorial Commentary

How do we improve chemoimmunotherapy in gastroesophageal adenocarcinoma?

Jane E. Rogers1, Jaffer A. Ajani2

1Department of Pharmacy Clinical Program, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA; 2Department of Gastrointestinal Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA

Correspondence to: Jane E. Rogers, PharmD. Department of Pharmacy Clinical Program, University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA. Email: jerogers@mdanderson.org; Jaffer A. Ajani, MD. Department of Gastrointestinal Medical Oncology, University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA. Email: jajani@mdanderson.org.

Comment on: An M, Mehta A, Min BH, et al. Early Immune Remodeling Steers Clinical Response to First-Line Chemoimmunotherapy in Advanced Gastric Cancer. Cancer Discov 2024;14:766-85.


Keywords: Gastric neoplasms; programmed death-1 (PD-1); programmed death ligand-1 (PD-L1); immunotherapy


Received: 31 July 2024; Accepted: 11 December 2024; Published online: 07 February 2025.

doi: 10.21037/actr-24-137


Gastroesophageal adenocarcinoma (GEAC) continues to need advancements in management. Building on immunotherapy activity in the refractory setting, many studies using either an anti-programmed death-1 (anti-PD-1) or anti-programmed death ligand-1 (anti-PD-L1) immune checkpoint agent, in combination mostly with cytotoxics, have been reported recently in patients with advanced or localized GEAC. Representative studies among these in the advanced setting are KEYNOTE-811, CHECKMATE-649, ATTRACTION-4, KEYNOTE062, KEYNOTE-859, ORIENT-16, JAVLIN Gastric 100, and RATIONALE-305 (1-8) (Table 1). Modest and heterogenous survival improvement from the immune checkpoint addition to fluoropyrimidine plus platinum is seen (1-9). These results are not as robust as those in other tumors because a smaller number of patients seems to benefit. This is either due to the inherent tumor microenvironment (TME) composition of GEAC (that we ignore entirely in the clinic) or heterogeneity of the epithelial cells [for example, chromosomal instability (CIN) that confers resistance to cytotoxics and immunotherapy]. An elaborate molecular definition of GEAC tumors is lacking. Bulk clinical observations suggest that not all patients will benefit from the addition of immune checkpoint therapy, as seen in CHECKMATE-649, an international phase 3 trial, which studied the addition of nivolumab, an anti-PD-1 agent, to upfront fluoropyrimidine plus platinum in the front-line advanced GEAC setting (2). CHECKMATE-649 showed a distinction in outcomes based on PD-L1 combined positive score (CPS) score. Those with a CPS score of ≥5 were those that derived benefit from the addition of anti-PD-1 therapy. Some of these patients will benefit a lot as there is prolonged survival and duration of response that can be seen. Similarly, in ORIENT-16, a phase 3 Chinese study, sintilimab, an anti-PD-1 agent, was added to front-line fluoropyrimidine plus platinum therapy. A more dramatic benefit was seen in advanced gastric adenocarcinoma (GAC) PD-L1 CPS ≥5 reported an objective response rate (ORR) of 63.6% and a median duration of response of 9.8 months (6). The phase 3 RATIONALE-305 showed similar activity in PD-L1 positive patients [tumor area positivity (TAP) score >5] with tislelizumab, an anti-PD-1 agent, plus investigator choice chemotherapy. ORR was 50.4% with a duration of response of 9 months. This result again highlights more impact in those with higher PD-L1 scoring. The same was seen in KEYNOTE-811 in relation to PD-L1 CPS positivity as the benefit of the addition of pembrolizumab was only in those with a CPS score ≥1 (1). With results showing ~40–50% not having shrinkage and with a duration of response of only ~9 months (2,6,8), there are intrinsic and acquired immune resistance pathways present in GEAC.

Table 1

Outcomes of front-line immunotherapy in GAC

Trial Population Treatment design Outcomes
KEYNOTE-811 HER2-positive LA or metastatic GAC or GEJ Pembrolizumab plus trastuzumab plus chemotherapy vs. trastuzumab plus chemotherapy Median PFS: 10 vs. 8.1 months, P=0.0002
Median OS: 20 vs. 16.8 months, P=0.084
ORR: 72.6% vs. 59.8%
PD-L1 CPS ≥1
     ⬥ Median PFS: 10.8 vs. 7.2 months (did not differ if CPS <1)
     ⬥ Median OS: 20.5 vs. 15.6 months (not improved if CPS <1)
CHECKMATE-649 HER2-negative LA or metastatic GAC or GEJ or esophageal adenocarcinoma Nivolumab plus chemotherapy vs. nivolumab plus ipilimumab vs. chemotherapy All patients
     ⬥ Median OS: 13.8 vs. 11.6 months, P=0.0002
     ⬥ Median PFS: 7.7 vs. 6.9 months
     ⬥ ORR: 60% vs. 45%
PD-L1 CPS ≥5
     ⬥ Median OS: 14.4 vs. 11.1 months, P<0.0001
     ⬥ Median PFS: 7.7 vs. 6.1 months, P<0.0001
     ⬥ ORR
ATTRACTION-4 HER2-negative LA or metastatic GAC or GEJ Nivolumab plus chemotherapy vs. placebo plus chemotherapy Median OS: 17.5 vs. 17.2 months
Median PFS: 10.5 vs. 8.3 months
ORR: 57% vs. 48%
KEYNOTE-062 LA or metastatic GAC or GEJ with PD-L1 CPS ≥1 Pembrolizumab vs. pembrolizumab with chemotherapy vs. placebo with chemotherapy PD-L1 CPS ≥1 (pembrolizumab vs. chemotherapy)
     ⬥ Median OS: 10.6 vs. 11.1 months
     ⬥ Median PFS: 2 vs. 6.4 months
PD-L1 CPS ≥10 (pembrolizumab vs. chemotherapy)
     ⬥ Median OS: 17.4 vs. 10.8 months
     ⬥ Median PFS: 2.9 vs. 6.1 months
     ⬥ ORR: 25% vs. 37.8%
KEYNOTE-859 HER2-negative LA or metastatic GAC or GEJ Pembrolizumab with chemotherapy vs. placebo with chemotherapy All patients
     ⬥ Median OS: 12.9 vs. 11.5 months, P<0.0001
     ⬥ Median PFS: 6.9 vs. 5.6 months
     ⬥ ORR: 51% vs. 42%
PD-L1 CPS ≥1
     ⬥ Median OS: 13 vs. 11.4 months, P<0.0001
     ⬥ Median PFS: 6.9 vs. 5.6 months
     ⬥ ORR: 52% vs. 43%
PD-L1 CPS ≥10
     ⬥ Median OS: 15.7 vs. 11.8 months, P<0.0001
     ⬥ Median PFS: 8.1 vs. 5.6 months
     ⬥ ORR: 61% vs. 43%
ORIENT-16 LA or metastatic GAC or GEJ Sintilimab plus chemotherapy vs. placebo plus chemotherapy All patients
     ⬥ Median OS: 15.2 vs. 12.3 months, P=0.009
     ⬥ Median PFS: 7.1 vs. 5.7 months, P=0.77
     ⬥ ORR: 58.2% vs. 48.4%, P=0.02
PD-L1 CPS ≥5
     ⬥ Median OS: 18.4 vs. 12.9 months, P=0.002
     ⬥ Median PFS: 7.7 vs. 5.8 months, P<0.001
     ⬥ ORR: 63.6% vs. 49.4%, P=0.008
JAVLIN GASTRIC-100 HER-2-negative LA or metastatic GAC or GEJ Avelumab vs. continued chemotherapy All patients
     ⬥ Median OS: 10.4 vs. 10.9 months, P=0.1779
Patients without progressive disease after 12 weeks of first-line chemotherapy      ⬥ Median PFS: 3.2 vs. 4.4 months
     ⬥ ORR: 13.3% vs. 14.4%
PD-L1 CPS ≥1
     ⬥ Median OS: 14.9 vs. 11.6 months
     ⬥ Median PFS: 4.3 vs. 5.1 months
RATIONALE-305 HER-2-negative LA or metastatic GAC or GEJ Tislelizumab plus chemotherapy vs. placebo plus chemotherapy PD-L1 TAP score ≥5%
     ⬥ Median OS: 17.2 vs. 12.6 months, P=0.006
     ⬥ Median PFS: 7.2 vs. 5.9 months, P<0.001
     ⬥ ORR: 50% vs. 43%, P=0.08
All patients
     ⬥ Median OS: 15 vs. 12.9 months, P=0.001
     ⬥ Median PFS: 6.9 vs. 6.2 months
     ⬥ ORR: 47% vs. 41%

GAC, gastric adenocarcinoma; HER2, human epidermal growth factor receptor-2; LA, locally advanced; GEJ, gastroesophageal junction adenocarcinoma; PD-L1, programmed death ligand-1; CPS, combined positive score; PFS, progression-free survival; OS, overall survival; ORR, objective response rate.

Similarly, several studies in the refractory and rechallenge setting are underway but again, none considering TME or overall molecular/immune sub-typing of GEAC. Such examples are: the PARAMUNE, INTEGRATE, LEAP, DURIGAST-PRODIGE, and CAP 02 Re-challenge trials (9-14).

Localized GEAC has also been recently investigated for its share of immune checkpoint additions in studies such as DANTE/IKF-S633, KEYNOTE 585, MATTERHORN, ATTRACTION-5, and VESTIGE (15-19). However, data is still maturing and whether this will have a robust advantage to the patients with localized GEAC is conflicting. Meaningful pathological complete response (pCR) improvement has been reported; however, time will be needed to gather survival data and pCR will correlate. The only trial currently with survival endpoints reported is the KEYNOTE 585. Final analysis of KEYNOTE 585 reported event-free survival 44.4 vs. 25.5 months, median overall survival 71.8 vs. 55.7 months, and pCR rate 13.4% vs. 2%; however, primary endpoint for event-free survival did not meet prespecified statistical significance (16). Whether certain subgroups would have survival benefits remains unanswered. As with the DANTE trial, PD-L1 CPS ≥1 was reported at 58% of the population and therefore may not be a reliable marker in the localized setting (15). All these studies have ignored the driver epithelial oncogenes and the machinery responsible for immune suppression. Current basic questions necessary to move the needle include (I) what specific patterns of innate and acquired resistance to immunotherapy or other therapy are present in GEAC; (II) what would change a “cold” TME to a host favoring TME?; and (III) what novel biomarkers, perhaps, in the blood could trigger customized therapeutics. Obtaining actionable biomarkers is necessary and how these will be determined remains unanswered as serial biopsies are not always feasible and/or patient preferred in standard practice. Additionally, how accurate blood and tissue analysis align along with how to incorporate these in clinical practice remain key areas to answer these questions.

We must applaud An and colleagues who conducted a very difficult phase 2 trial of advanced GEAC patients (n=47) (9). An and colleagues conducted a prospective single-arm sequential chemoimmunotherapy trial in patients with metastatic GEAC. The researchers performed serial sampling for deep correlative studies to build upon the authors’ prior work (serial biopsies in GEAC patients on chemotherapy do demonstrate early TME remodeling). Serial biopsies were taken from the primary tumor (day 1 before cycle 1, day 1 before cycle 2, and day 1 before cycle 7). They made an attempt to systemically study the TME before treatment and following treatment. They also tried to parse out the TME changes with cytotoxic agents versus the addition of pembrolizumab. Obtaining a clear picture of the TME holds answers on how tumor cells behave particularly after exposure to treatment to help us understand how the cancer progresses and/or responds and how to remodel the TME to make it harder for tumor growth. However, when it comes to investigating patients, the specimen collection and precisely repeating collection from the same area remains a challenge. Nevertheless, An and colleagues were able to generate wealth of information that we never had before. All patients were Korean which could impact extrapolation to the Western population as often GEAC has shown differences in epidemiology, treatment, and survival between Asian and Western countries but this may not be that important and can be sorted out in the future. The authors’ objectives were to validate prior results of TME remodeling after exposure to fluoropyrimidine plus platinum therapy and to additionally understand the contribution of pembrolizumab on standard chemotherapy, and to identify candidate pathways for future exploration.

The authors described important differences of pro-immunity and anti-immunity features seen in slow progressors and fast progressors. Slow progressors were found to have increased expression of representative genes of immunogenic cell death after a single cycle of chemotherapy alone, and a larger proportion showed remodeled TME toward immune enriched environments compared to fast progressors. The authors found T cell expansion after anti-PD-1 in slow progressors leading to questions of whether early chemotherapy induced TME remodeling determines whether anti-PD-1 will enhance response or improve prognosis. Further compelling information observed was that the CD8 T cell expansion (naive, memory, and exhausted cells) showed slow progressors following anti-PD-1 therapy. CD8 effector subsets were found in responders. The authors describe potential ways of depriving the TME of immunosuppressive signals or stimulating the induction with pro-immunity hubs. More studies similar to the one performed by An and colleagues are needed to understand how more patients can derive advantage from the current armamentarium.


AcknowledgmentsOther Section

None.


FootnoteOther Section

Provenance and Peer Review: This article was commissioned by the editorial office, AME Clinical Trials Review. The article has undergone external peer review.

Peer Review File: Available at https://actr.amegroups.com/article/view/10.21037/actr-24-137/prf

Funding: None.

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://actr.amegroups.com/article/view/10.21037/actr-24-137/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


ReferencesOther Section

  1. Janjigian YY, Kawazoe A, Bai Y, et al. Pembrolizumab plus trastuzumab and chemotherapy for HER2-positive gastric or gastro-oesophageal junction adenocarcinoma: interim analyses from the phase 3 KEYNOTE-811 randomised placebo-controlled trial. Lancet 2023;402:2197-208. [Crossref] [PubMed]
  2. Janjigian YY, Ajani JA, Moehler M, et al. First-Line Nivolumab Plus Chemotherapy for Advanced Gastric, Gastroesophageal Junction, and Esophageal Adenocarcinoma: 3-Year Follow-Up of the Phase III CheckMate 649 Trial. J Clin Oncol 2024;42:2012-20. [Crossref] [PubMed]
  3. Kang YK, Chen LT, Ryu MH, et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (ATTRACTION-4): a randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2022;23:234-47. [Crossref] [PubMed]
  4. Shitara K, Van Cutsem E, Bang YJ, 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;6:1571-80. [Crossref] [PubMed]
  5. Rha SY, Oh DY, Yañez P, et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for HER2-negative advanced gastric cancer (KEYNOTE-859): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol 2023;24:1181-95. [Crossref] [PubMed]
  6. Xu J, Jiang H, Pan Y, et al. Sintilimab Plus Chemotherapy for Unresectable Gastric or Gastroesophageal Junction Cancer: The ORIENT-16 Randomized Clinical Trial. JAMA 2023;330:2064-74. [Crossref] [PubMed]
  7. Moehler M, Dvorkin M, Boku N, 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 2021;39:966-77. [Crossref] [PubMed]
  8. Qiu MZ, Oh DY, Kato K, et al. Tislelizumab plus chemotherapy versus placebo plus chemotherapy as first line treatment for advanced gastric or gastro-oesophageal junction adenocarcinoma: RATIONALE-305 randomised, double blind, phase 3 trial. BMJ 2024;385:e078876. [Crossref] [PubMed]
  9. An M, Mehta A, Min BH, et al. Early Immune Remodeling Steers Clinical Response to First-Line Chemoimmunotherapy in Advanced Gastric Cancer. Cancer Discov 2024;14:766-85. [Crossref] [PubMed]
  10. Saeed A, Colby S, Oberstein PE, et al. SWOG S2303: Randomized phase II/III trial of 2nd line nivolumab + paclitaxel + ramucirumab versus paclitaxel + ramucirumab in patients with PD-L1 CPS ≥1 advanced gastric and esophageal adenocarcinoma (PARAMUNE). J Clin Oncol 2024;42:abstr TPS430.
  11. Pavlakis N, Shitara K, Sjoquist KM, et al. INTEGRATE IIa: A randomised, double-blind, phase III study of regorafenib versus placebo in refractory advanced gastrooesophageal cancer (AGOC)—A study led by the Australasian Gastro-intestinal Trials Group (AGITG). J Clin Oncol 2023;41:abstr LBA294.
  12. Yanez PE, Ben-Aharon I, Rojas C, et al. First-line lenvatinib plus pembrolizumab plus chemotherapy versus chemotherapy in advanced/metastatic gastroesophageal adenocarcinoma (LEAP-015): safety run-in results. J Clin Oncol 2023;41:abstr 411.
  13. Tougeron D, Dahan L, Hajbi FE, et al. The PRODIGE 59-DURIGAST trial: A randomized phase II study evaluating FOLFIRI plus durvalumab and FOLFIRI plus durvalumab plus tremelimumab in second-line treatment of patients with advanced gastric or gastro-esophageal junction adenocarcinoma. J Clin Oncol 2022; 40:abstr 4036.
  14. Meng X, Wang J, Xia J, et al. Camrelizumab plus apatinib after progression on immune checkpoint inhibitors in patients with advanced esophageal squamous cell carcinoma (CAP 02 Re-challenge): a single arm multicenter phase 2 trial. J Clin Oncol 2024;42:abstr 364.
  15. Lorenzen S, Götze TO, Thuss-Patience P, et al. Perioperative Atezolizumab Plus Fluorouracil, Leucovorin, Oxaliplatin, and Docetaxel for Resectable Esophagogastric Cancer: Interim Results From the Randomized, Multicenter, Phase II/III DANTE/IKF-s633 Trial. J Clin Oncol 2024;42:410-20. [Crossref] [PubMed]
  16. Shitara K, Rha SY, Wyrwicz LS, et al. LBA3 Final analysis of the phase III KEYNOTE-585 study of pembrolizumab plus chemotherapy vs chemotherapy as perioperative therapy in locally advanced gastric and gastroesophageal junction cancer. Ann Oncol 2024;35:S213. [Crossref]
  17. Janjigian YY, Al-Batran SE, Wainberg ZA, et al. LBA73 Pathological complete response (pCR) to durvalumab plus 5-fluorouracil, leucovorin, oxaliplatin, and docetaxel (FLOT): in resectable gastric and gastroesophageal junction cancer (GC/GEJC): interim results of the global phase III MATTERHORN study. Ann Oncol 2023;34:S1315-6. [Crossref]
  18. Kang YK, Terashima M, Kim YW, et al. Adjuvant nivolumab plus chemotherapy versus placebo plus chemotherapy for stage III gastric or gastro-oesophageal junction cancer after gastrectomy with D2 or more extensive lymph-node dissection (ATTRACTION-5): a randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Gastroenterol Hepatol 2024;9:705-17. [Crossref] [PubMed]
  19. Smyth E, Mauer M, Cella C, et al. O-6 EORTC 1707 VESTIGE: adjuvant immunotherapy in patients with resected gastroesophageal adenocarcinoma (GEA) following preoperative chemotherapy with high risk for recurrence (ypN+ and /or R1) an open-label randomized controlled phase II study. Ann Oncol 2023;34:S182-3. [Crossref]
doi: 10.21037/actr-24-137
Cite this article as: Rogers JE, Ajani JA. How do we improve chemoimmunotherapy in gastroesophageal adenocarcinoma? AME Clin Trials Rev 2025;3:9.

Download Citation