Second-line nivolumab with paclitaxel in biomarker-enriched gastric cancer: not all biomarkers are created equal but is the question even relevant anymore?
Editorial Commentary

Second-line nivolumab with paclitaxel in biomarker-enriched gastric cancer: not all biomarkers are created equal but is the question even relevant anymore?

Joan R. Choo1 ORCID logo, Geoffrey Ku2 ORCID logo

1Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore; 2Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA

Correspondence to: Geoffrey Ku, MD, MBA. Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 300 E. 66th Street, Rm 1035, New York, NY, 10065, USA. Email: kug@mskcc.org.

Comment on: Lee CK, Lee JB, Park SJ, et al. Second-line chemoimmunotherapy with nivolumab and paclitaxel in immune-related biomarkerenriched advanced gastric cancer: a multicenter phase Ib/II study. Gastric Cancer 2024;27:118-30.


Keywords: Gastric cancer (GC); biomarkers; immunotherapy


Received: 25 February 2024; Accepted: 05 June 2024; Published online: 21 June 2024.

doi: 10.21037/actr-24-15


Incorporating immune-checkpoint inhibition (ICI) via anti-programmed cell death 1 (PD-1) antibodies in the initial treatment of advanced gastric cancer (GC) has led to improved survival, particularly in patients whose tumors express programmed death-ligand 1 (PD-L1) (1,2). However, there has been no demonstrable benefit for ICIs in the second-line setting in immunotherapy-naïve patients—an increasingly rare situation.

In the US, the relevance of this question has diminished, given blanket approvals of first-line ICI plus chemotherapy in any patient with advanced esophageal, gastroesophageal junction (GEJ) or gastric adenocarcinoma, HER2-negative (3) and -positive (4). Ironically, one contemporary population that this question applies to are patients with Claudin18.2 positive tumors who, in the near future, are expected to receive zolbetuximab with chemotherapy (5,6).

As we know from the global KEYNOTE-061 (7), and similarly seen in the East Asian KEYNOTE-063 (8) study, single agent anti-PD-1 therapy in the 2nd-line setting is not preferred to chemotherapy—overall survival (OS) was not superior [9.1 vs. 8.3 months, hazard ratio (HR) 0.82, 95% confidence interval (CI): 0.66–1.03] and progression-free survival (PFS) was shorter [1.5 vs. 4.1 months, HR 1.27, 95% CI: 1.03–1.57] in patients treated with pembrolizumab compared to paclitaxel. However, the survival curves crossed after 8 months with delayed but sustained benefit seen in a subset of patients treated with pembrolizumab (7). This phenomenon has been observed in multiple other studies of anti-PD-1/anti-PD-L1 inhibitors in various tumors (9-11) and highlights the need for both biomarker-driven strategies and combination treatment approaches to overcome the delayed antitumor response of single agent ICI.

In Gastric Cancer, Lee and colleagues reported their findings from the phase 2 K-umbrella-03 study which evaluated the combination of nivolumab and paclitaxel in immune biomarker-enriched GC patients (12). The Authors selected patients with PD-L1-positive, Epstein-Barr virus (EBV)-positive or deficient mismatch repair (dMMR) HER2-negative GC who had previously received first-line chemotherapy with a fluoropyrimidine/platinum doublet. For added context, this was a substudy of the larger K-umbrella study of 4 biomarker cohorts: (I) epidermal growth factor receptor (EGFR) overexpressed [immunohistochemistry (IHC) 2+/3+] cohort for treatment with a pan-ERBB inhibitor; (II) PTEN loss/null cohort for treatment with a PIK3Cb inhibitor; (III) nivolumab cohort; and (IV) patients without predefined biomarkers for standard of care treatment (13). The investigators are certainly to be congratulated on the design of what at the time was a state-of-the-art umbrella study designed to simultaneously test multiple compelling hypotheses.

Unfortunately, K-umbrella-03 was ultimately a negative study that did not achieve its primary objective of improved PFS with the nivolumab/paclitaxel combination in this immune biomarker-enriched population. The null hypothesis of the study was a median PFS of 2.9 months, while the alternative hypothesis was a median PFS of 4.4 months, based on the RAINBOW study of paclitaxel with or without ramucirumab (14). The median PFS was 3.9 months (range, 2.7–5.5 months) with a median OS of 11.2 months (range, 6.0–18.7 months) and an objective response rate (ORR) of 23.3%. Although this was a negative study, there were once again glimmers of activity: in those who responded to nivolumab-paclitaxel, the median duration of response was 16.7 months. Exploratory analyses suggested that baseline interleukin-1 receptor antagonist levels could help predict for response to the treatment strategy.

Several factors impact the interpretability and relevance of the results from this study. Lee and colleagues enrolled a heterogenous group of patients with GC who were positive for at least 1 of 3 immune biomarkers with dramatically differential degrees of benefit from immunotherapy. Patients with PD-L1 Combined Positive Score (CPS) ≥1 tumors made up 95.8% of the study group. The tumor subtypes that we would expect to benefit dramatically from ICI—dMMR and EBV-positive tumors—only made up 4.2% and 10.4% respectively of patients enrolled, or 8 out of the 48 patients. These small numbers make it difficult even to draw definitive conclusions about the benefit of this combination in these patients. In fact, the authors found no significant association between mismatch repair (MMR) status and survival within their study population. Of course, there is little doubt about the role of immunotherapy in dMMR tumors; deficient MMR status is a widely accepted predictive biomarker and pan-tumor indication for treatment with ICI (15). Specifically in dMMR GC, ICI has been shown to be active regardless of the line of therapy it is administered (16).

Meanwhile, EBV-positive GC, which is characterised by intra-/peri-tumoral immune cell infiltration with genomic amplification of the locus containing genes encoding PD-L1 and PD-L2 (17), was previously described to have a remarkable ORR of 100% to ICI in a small cohort of six patients (18). However, findings from recent studies have been more mixed: no responses to camrelizumab were seen in six EBV-positive GC patients who had failed two prior lines of chemotherapy (19); 1 of 4 patients treated with toripalimab had a response (20) and; 1 of 3 EBV-positive patients treated with either pembrolizumab or nivolumab responded (21). In K-umbrella-03, an ORR of 33% (1 of 3 patients with measurable disease) was observed in EBV-positive patients treated with nivolumab/paclitaxel. Taken together, these data suggest that EBV positivity is an imperfect biomarker with apparent significant heterogeneity. Larger prospective studies or even retrospective series are needed to clarify its role as an independent predictive biomarker for immunotherapy benefit.

Since results from the RAINBOW trial were published in 2014, ramucirumab/paclitaxel has become the global standard of care in the 2nd-line setting in a biomarker-unselected population, demonstrating improved median OS (9.6 vs. 7.4 months, HR 0.807, 95% CI: 0.678–0.962), improved PFS (4.4 vs. 2.9 months, HR 0.635, 95% CI: 0.536–0.752) and higher ORR (27% vs. 16%) compared to paclitaxel (14). As such, more relevant studies have built on this standard.

The benefit of adding nivolumab to ramucirumab/paclitaxel was evaluated by Nakajima and colleagues, who conducted the single-arm phase 2 UMIN-CTR trial that enrolled ICI-naïve patients without biomarker enrichment (22). This regimen was associated with an ORR of 37.2%, median PFS of 5.1 months, and median OS of 13.1 months. Patients with PD-L1 CPS ≥1 tumors seemed to do better than those with PD-L1 CPS <1 tumors (median OS 13.8 vs. 8.0 months). Therefore, the findings of this study and K-umbrella-03 support the notion that adding ICI to paclitaxel (with or without ramucirumab) to an ICI-naive population in the 2nd-line setting may be beneficial but it is challenging to tease out the patients who truly benefit.

A more contemporary and relevant iteration of the Nakajima study is the planned SWOG S2303 study (NCT06203600), which is anticipated to open shortly in the US. This is a randomized phase II/III study that will address the benefit of adding nivolumab to ramucirumab/paclitaxel as 2nd-line therapy in patients with PD-L1 CPS ≥1 GEJ/GC, whose disease has previously progressed on 1st-line ICI plus chemotherapy. Essentially, the question is whether there is a benefit to continuing an ICI beyond progression into the next line of therapy.

Finally, the field has also moved on from the evaluation of an anti-PD-1/PD-L1 antibody alone with chemotherapy. Several promising novel combinations being tested in completed or ongoing global 1st-line phase 3 trials include the use of lenvatinib/pembrolizumab with chemotherapy in the LEAP-015 trial (NCT04662710) and the addition of anti-TIGIT antibodies alongside anti-PD-1 and chemotherapy in the STAR-221 trial (NCT05568095). The outcomes of these studies are eagerly anticipated.


Acknowledgments

Funding: None.


Footnote

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-24-15/prf

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://actr.amegroups.com/article/view/10.21037/actr-24-15/coif). J.R.C. has received travel support from AstraZeneca. G.K. has received research funding to his institution from AstraZeneca, BMS, CARsgen, Daiichi Sankyo, I-MAB, Jazz, Merck, Oncolys, Pieris, Triumvira and Zymeworks; consulting fees from Astellas, AstraZeneca, Bayer, BMS, Daiichi Sankyo, I-MAB, Jazz, Merck, Pieris and Zymeworks; travel support from Dava Oncology and I-MAB. The authors have no other 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/.


References

  1. Janjigian YY, Smit EF, Groen HJ, et al. Dual inhibition of EGFR with afatinib and cetuximab in kinase inhibitor-resistant EGFR-mutant lung cancer with and without T790M mutations. Cancer Discov 2014;4:1036-45. [Crossref] [PubMed]
  2. 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]
  3. FDA. Full prescribing information for nivolumab 2021. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/125554s091lbl.pdf
  4. FDA. Full prescribing information for pembrolizumab 2023. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/125514s148lbl.pdf
  5. Shah MA, Shitara K, Ajani JA, et al. Zolbetuximab plus CAPOX in CLDN18.2-positive gastric or gastroesophageal junction adenocarcinoma: the randomized, phase 3 GLOW trial. Nat Med 2023;29:2133-41. [Crossref] [PubMed]
  6. Shitara K, Lordick F, Bang YJ, et al. Zolbetuximab plus mFOLFOX6 in patients with CLDN18.2-positive, HER2-negative, untreated, locally advanced unresectable or metastatic gastric or gastro-oesophageal junction adenocarcinoma (SPOTLIGHT): a multicentre, randomised, double-blind, phase 3 trial. Lancet 2023;401:1655-68. [Crossref] [PubMed]
  7. Shitara K, Özgüroğlu M, Bang YJ, 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;392:123-33. [Crossref] [PubMed]
  8. Chung HC, Kang YK, Chen Z, et al. Pembrolizumab versus paclitaxel for previously treated advanced gastric or gastroesophageal junction cancer (KEYNOTE-063): A randomized, open-label, phase 3 trial in Asian patients. Cancer 2022;128:995-1003. [Crossref] [PubMed]
  9. Ferris RL, Blumenschein G Jr, Fayette J, et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N Engl J Med 2016;375:1856-67. [Crossref] [PubMed]
  10. Mok TSK, Wu YL, Kudaba I, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet 2019;393:1819-30. [Crossref] [PubMed]
  11. Bellmunt J, de Wit R, Vaughn DJ, et al. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N Engl J Med 2017;376:1015-26. [Crossref] [PubMed]
  12. Lee CK, Lee JB, Park SJ, et al. Second-line chemoimmunotherapy with nivolumab and paclitaxel in immune-related biomarker-enriched advanced gastric cancer: a multicenter phase Ib/II study. Gastric Cancer 2024;27:118-30. [Crossref] [PubMed]
  13. Lee CK, Kim HS, Jung M, et al. Open-Label, Multicenter, Randomized, Biomarker-Integrated Umbrella Trial for Second-Line Treatment of Advanced Gastric Cancer: K-Umbrella Gastric Cancer Study. J Clin Oncol 2024;42:348-57. [Crossref] [PubMed]
  14. Wilke H, Muro K, Van Cutsem E, et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol 2014;15:1224-35. [Crossref] [PubMed]
  15. Le DT, Uram JN, Wang H, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med 2015;372:2509-20. [Crossref] [PubMed]
  16. 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. [Crossref] [PubMed]
  17. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014;513:202-9. [Crossref] [PubMed]
  18. Kim ST, Cristescu R, Bass AJ, et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat Med 2018;24:1449-58. [Crossref] [PubMed]
  19. Sun YT, Guan WL, Zhao Q, et al. PD-1 antibody camrelizumab for Epstein-Barr virus-positive metastatic gastric cancer: a single-arm, open-label, phase 2 trial. Am J Cancer Res 2021;11:5006-15. [PubMed]
  20. Wang F, Wei XL, Wang FH, et al. Safety, efficacy and tumor mutational burden as a biomarker of overall survival benefit in chemo-refractory gastric cancer treated with toripalimab, a PD-1 antibody in phase Ib/II clinical trial NCT02915432. Ann Oncol 2019;30:1479-86. [Crossref] [PubMed]
  21. Kubota Y, Kawazoe A, Sasaki A, et al. The Impact of Molecular Subtype on Efficacy of Chemotherapy and Checkpoint Inhibition in Advanced Gastric Cancer. Clin Cancer Res 2020;26:3784-90. [Crossref] [PubMed]
  22. Nakajima TE, Kadowaki S, Minashi K, et al. Multicenter Phase I/II Study of Nivolumab Combined with Paclitaxel Plus Ramucirumab as Second-line Treatment in Patients with Advanced Gastric Cancer. Clin Cancer Res 2021;27:1029-36. [Crossref] [PubMed]
doi: 10.21037/actr-24-15
Cite this article as: Choo JR, Ku G. Second-line nivolumab with paclitaxel in biomarker-enriched gastric cancer: not all biomarkers are created equal but is the question even relevant anymore? AME Clin Trials Rev 2024;2:38.

Download Citation