Combination and precision: ingredients for success in advanced gastric cancer
Editorial Commentary

Combination and precision: ingredients for success in advanced gastric cancer

Eric Mehlhaff1, Nataliya V. Uboha1,2

1Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA; 2Carbone Cancer Center, University of Wisconsin, Madison, WI, USA

Correspondence to: Nataliya V. Uboha, MD, PhD. Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, Section of Hematology & Oncology, University of Wisconsin, 600 Highland Ave, CSC K4/518, Madison, WI 53792, USA; Carbone Cancer Center, University of Wisconsin, Madison, WI, USA. Email: nvuboha@medicine.wisc.edu.

Comment on: Kang YK, Ryu MH, Di Bartolomeo M, et al. Rivoceranib, a VEGFR-2 inhibitor, monotherapy in previously treated patients with advanced or metastatic gastric or gastroesophageal junction cancer (ANGEL study): an international, randomized, placebo-controlled, phase 3 trial. Gastric Cancer 2024;27:375-86.


Keywords: Rivoceranib; gastric cancer; biomarker; antiangiogenic agent; tyrosine kinase inhibitor (TKI)


Received: 27 April 2024; Accepted: 17 July 2024; Published online: 12 August 2024.

doi: 10.21037/actr-24-53


Patients with metastatic gastric cancer have poor prognosis with overall survival (OS) remaining less than 2 years despite recent updates in standard practice (1). While there have been significant advances in first- and second-line settings, developing novel therapies for more advanced disease has been challenging. By incorporating more effective therapies earlier in the disease course, a growing number of patients may be candidates for systemic treatment options beyond the second line, highlighting the critical need for drug development. Currently, trifluridine/tipiracil is the only agent approved in the United States for patients whose disease progressed on two prior lines of therapy. This approval was based on the results from a global phase 3 study, TAGS, that randomized patients who have received at least two prior lines of therapies to either oral trifluridine/tipiracil or placebo. Treatment with trifluridine/tipiracil resulted in significant improvement in OS [median OS 5.7 vs. 3.6 months; hazard ratio (HR) 0.69] (2). Efficacy was sustained regardless of prognostic factors, such as performance status, number of previous treatment lines, HER2 status, number of metastatic sites and age.

The ANGEL study evaluated activity of rivoceranib, a selective VEGFR2 tyrosine kinase inhibitor (TKI), in a patient population similar to that enrolled in the TAGS study (3). Antiangiogenic agents have established activity in the management of this disease. Ramucirumab [a monoclonal antibody against vascular endothelial growth factor receptor-2 (VEGFR2)], in combination with paclitaxel, is a preferred second-line regimen in a biomarker non-select patient population (4). However, the results of studies with antiangiogenic agents have been mixed, likely affected by patient selection, treatment setting, and tumor heterogeneity (5,6). Small molecule TKIs targeting vascular endothelial growth factor (VEGF) signaling have been under active investigation in this disease for several years but are yet to yield actionable results. In the ANGEL study, participants (n=460) were randomized 2:1 to receive either rivoceranib plus best supportive care (BSC) or placebo plus BSC. Among intention-to-treat population, primary endpoint of median OS was not statistically different between the rivoceranib and placebo cohorts [5.78 vs. 5.13 months; HR 0.93, 95% confidence interval (CI): 0.74–1.15]. Improvements were observed with secondary outcomes of median PFS (2.83 vs. 1.77 months; HR 0.58, 95% CI: 0.45–0.79) and objective response rate (ORR) (6.87% vs. 0%), though margin of benefit was minimal. Rivoceranib had expected toxicities that are typical for other TKIs targeting angiogenesis, including anorexia, hypertension, proteinuria, diarrhea, and palmar-plantar erythrodysesthesia syndrome. While the study results are disappointing, several interesting observations can be made about the presented data. The study was able to enroll patients with disease progression after several lines of therapy, and it helped demonstrate that earlier use of active regimens may positively impact the ability to receive treatment in later-line settings. This phenomenon, unfortunately, was not observed in recent phase 3 trials, where only about 50% of patients were able to receive subsequent therapies after progression on the first-line treatment (1,7,8). Therefore, the ANGEL study also enrolled patients who had favorable disease to allow clinical trial participation very late in the course of their illness. The latter may, at least in part, explain better than expected outcomes of the control group with OS of 5.13 months in the whole study population and 4.73 months in those with ≥4 lines of therapy. Although further lines of therapy can affect OS results, both experimental and control groups had a similar number of patients receive subsequent treatments. It is also important to note recent results from a similar study, phase 3 INTEGRATE IIa (NCT02773524), that compared regorafenib, a TKI directed against angiogenic, stromal, and oncogenic kinases, to placebo in an analogous setting. Although INTEGRATE IIa demonstrated a statistically significant improvement in median OS (4.5 vs. 4.0 months; HR 0.70), it is questionable whether these results translate into relevant clinical benefit (9).

While single agent TKIs have little potential in advanced gastric cancer, combination strategies do hold promise in this disease. Several prior studies demonstrated synergism between antiangiogenic agents and immune checkpoint inhibitors. Inhibition of both programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) and VEGF pathways may enhance T-cell mobilization and enable anti-tumor activity within tumor microenvironment (10). In the phase 1b REGONIVO trial, treatment with regorafenib and nivolumab resulted in 40% response rate among 25 evaluable patients (11). Lenvatinib and pembrolizumab demonstrated promising activity as well (12). Ongoing phase 3 LEAP-015 trial is investigating combination of lenvatinib and pembrolizumab with chemotherapy in the first-line setting (NCT04662710).

Treatments for advanced gastric cancer are increasingly becoming more personalized, and biomarker-based approaches are now widely accepted. In advanced disease, treatment selection based on HER2 status, PD-L1 (combined positive score; CPS), microsatellite instability (MSI) status or mismatch repair (MMR) protein expression has become standard practice (7,8,13-15). These approaches have been shown to improve patient outcomes. Trastuzumab deruxtecan, a HER2-directed antibody drug conjugate, is the only targeted agent approved for use in late lines (16,17). Hopefully more biomarker-based strategies can be utilized in late lines to help mitigate unnecessary toxicities in pretreated patients, as well as address an unmet need for improved treatment efficacy. Importantly, there are several new biomarkers that are under active investigations across treatment lines. Overexpression of CLDN18.2, a tight junction protein which is normally expressed on gastric epithelial cells, has been found in close to a third of advanced gastroesophageal cancers (1,18). Treatment with zolbetuximab, a chimeric immunoglobulin G1 (IgG1) monoclonal antibody targeted against CLDN18.2, in combination with chemotherapy resulted in OS improvement in treatment-naïve, HER2-negative, locally advanced or metastatic gastric or gastroesophageal adenocarcinoma patients in two phase 3 trials (1,19). Claudin-directed bispecific antibodies, drug conjugates and even cellular therapies are in the developmental pipeline (20). Bemarituzumab, a monoclonal antibody against FGFR2b, demonstrated positive results in the randomized phase 2 FIGHT trial (NCT03694522) in combination with chemotherapy for tumors with FGFR2 overexpression by IHC or gene amplification (21). Ongoing phase 3 studies FORTITUDE-101 (NCT05052801) and FORTITUDE-102 (NCT05111626) will further validate this positive signal, and numerous trials are looking fibroblast growth factor receptor (FGFR) inhibition with TKIs.

There are several opportunities for combination strategies with TKIs, and some are already under active investigations as discussed earlier. Management of advanced gastric cancer requires further utilization of biomarker-based paradigms. Appropriate patient selection is key to both maximize benefits and minimize unnecessary treatment toxicities.


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-24-53/prf

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://actr.amegroups.com/article/view/10.21037/actr-24-53/coif). N.V.U. reports receiving grants from the University of Wisconsin, Carbone Cancer Center (P30 CA014520), research support from Arcus/Gilead, Ipsen; and consulting for Astellas, AZ, Pfizer, BMS, Ipsen, Elevation Oncology, Merck, BeiGene, Eisai, Arcus, holding stock or stock options in Exact Sciences, Natera. The other author has 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/.


References

  1. 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. Erratum in: Lancet 2023;402:290 Erratum in: Lancet 2024;403:30. [Crossref] [PubMed]
  2. Shitara K, Doi T, Dvorkin M, et al. Trifluridine/tipiracil versus placebo in patients with heavily pretreated metastatic gastric cancer (TAGS): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2018;19:1437-48. [Crossref] [PubMed]
  3. Kang YK, Ryu MH, Di Bartolomeo M, et al. Rivoceranib, a VEGFR-2 inhibitor, monotherapy in previously treated patients with advanced or metastatic gastric or gastroesophageal junction cancer (ANGEL study): an international, randomized, placebo-controlled, phase 3 trial. Gastric Cancer 2024;27:375-86. [Crossref] [PubMed]
  4. Wilke H, Van Cutsem E, Cheul Oh S, et al. RAINBOW: A global, phase 3, randomized, double-blind study of ramucirumab plus paclitaxel versus placebo plus paclitaxel in the treatment of metastatic gastric adenocarcinoma following disease progression on first-line platinum- and fluoropyrimidine-containing combination therapy: Results of a multiple Cox regression analysis adjusting for prognostic factors. J Clin Oncol 2014;32:abstr LBA7.
  5. Ohtsu A, Shah MA, Van Cutsem E, et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a randomized, double-blind, placebo-controlled phase III study. J Clin Oncol 2011;29:3968-76. [Crossref] [PubMed]
  6. Fuchs CS, Shitara K, Di Bartolomeo M, et al. Ramucirumab with cisplatin and fluoropyrimidine as first-line therapy in patients with metastatic gastric or junctional adenocarcinoma (RAINFALL): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol 2019;20:420-35. [Crossref] [PubMed]
  7. Janjigian YY, Shitara K, Moehler M, et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial. Lancet 2021;398:27-40. [Crossref] [PubMed]
  8. 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]
  9. Lam LL, Pavlakis N, Shitara K, et al. INTEGRATE II: randomised phase III controlled trials of regorafenib containing regimens versus standard of care in refractory Advanced Gastro-Oesophageal Cancer (AGOC): a study by the Australasian Gastro-Intestinal Trials Group (AGITG). BMC Cancer 2023;23:180. [Crossref] [PubMed]
  10. Yasuda S, Sho M, Yamato I, et al. Simultaneous blockade of programmed death 1 and vascular endothelial growth factor receptor 2 (VEGFR2) induces synergistic anti-tumour effect in vivo. Clin Exp Immunol 2013;172:500-6. [Crossref] [PubMed]
  11. Fukuoka S, Hara H, Takahashi N, et al. Regorafenib Plus Nivolumab in Patients With Advanced Gastric or Colorectal Cancer: An Open-Label, Dose-Escalation, and Dose-Expansion Phase Ib Trial (REGONIVO, EPOC1603). J Clin Oncol 2020;38:2053-61. [Crossref] [PubMed]
  12. Kawazoe A, Fukuoka S, Nakamura Y, et al. Lenvatinib plus pembrolizumab in patients with advanced gastric cancer in the first-line or second-line setting (EPOC1706): an open-label, single-arm, phase 2 trial. Lancet Oncol 2020;21:1057-65. [Crossref] [PubMed]
  13. Bang YJ, Van Cutsem E, Feyereislova A, 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 2010;376:687-97. [Crossref] [PubMed]
  14. Maio M, Ascierto PA, Manzyuk L, et al. Pembrolizumab in microsatellite instability high or mismatch repair deficient cancers: updated analysis from the phase II KEYNOTE-158 study. Ann Oncol 2022;33:929-38. [Crossref] [PubMed]
  15. 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]
  16. Shitara K, Bang YJ, Iwasa S, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Gastric Cancer. N Engl J Med 2020;382:2419-30. [Crossref] [PubMed]
  17. Van Cutsem E, di Bartolomeo M, Smyth E, et al. Trastuzumab deruxtecan in patients in the USA and Europe with HER2-positive advanced gastric or gastroesophageal junction cancer with disease progression on or after a trastuzumab-containing regimen (DESTINY-Gastric02): primary and updated analyses from a single-arm, phase 2 study. Lancet Oncol 2023;24:744-756. [Crossref] [PubMed]
  18. Kubota Y, Kawazoe A, Mishima S, et al. Comprehensive clinical and molecular characterization of claudin 18.2 expression in advanced gastric or gastroesophageal junction cancer. ESMO Open 2023;8:100762. Erratum in: ESMO Open 2024;9:102232. [Crossref] [PubMed]
  19. 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]
  20. Mehlhaff E, Miller D, Ebben JD, et al. Targeted Agents in Esophagogastric Cancer Beyond Human Epidermal Growth Factor Receptor-2. Hematol Oncol Clin North Am 2024;38:659-75. [Crossref] [PubMed]
  21. Wainberg ZA, Enzinger PC, Kang YK, et al. Bemarituzumab in patients with FGFR2b-selected gastric or gastro-oesophageal junction adenocarcinoma (FIGHT): a randomised, double-blind, placebo-controlled, phase 2 study. Lancet Oncol 2022;23:1430-40. [Crossref] [PubMed]
doi: 10.21037/actr-24-53
Cite this article as: Mehlhaff E, Uboha NV. Combination and precision: ingredients for success in advanced gastric cancer. AME Clin Trials Rev 2024;2:52.

Download Citation