Toripalimab plus axitinib in patients with metastatic mucosal melanoma: 3-year survival update and biomarker analysis

Siming Li, Xiaowen Wu, Xieqiao Yan, Li Zhou, Zhihong Chi, Lu Si, Chuanliang Cui, Bixia Tang, Lili Mao, Bin Lian, Xuan Wang, Xue Bai, Jie Dai, Yan Kong, Xiongwen Tang, Hui Feng, Sheng Yao, Keith T Flaherty, Jun Guo, Xinan Sheng, Siming Li, Xiaowen Wu, Xieqiao Yan, Li Zhou, Zhihong Chi, Lu Si, Chuanliang Cui, Bixia Tang, Lili Mao, Bin Lian, Xuan Wang, Xue Bai, Jie Dai, Yan Kong, Xiongwen Tang, Hui Feng, Sheng Yao, Keith T Flaherty, Jun Guo, Xinan Sheng

Abstract

Background: Mucosal melanoma is an aggressive melanoma subtype with poor response to antiprogrammed cell death-1 (PD-1) monotherapy. Axitinib in combination with toripalimab, a humanized IgG4 mAb against PD-1, showed a promising response rate in patients with metastatic mucosal melanoma (MM) in a phase Ib study. Here, we report the updated overall survival (OS), duration of response (DoR), and biomarker analysis results.

Methods: Patients with advanced MM received toripalimab 1 or 3 mg/kg intravenously every 2 weeks combined with axitinib 5 mg orally two times per day until disease progression or unacceptable toxicity. Tumor programmed cell death ligand-1 (PD-L1) expression, tumor mutational burden (TMB), and gene expression profile (GEP) by messenger RNA sequencing were evaluated for correlation with survival.

Results: As of April 2, 2021, the median follow-up was 42.5 months. Among 29 chemotherapy-naïve patients with metastatic MM, the median OS was 20.7 months (95% CI 9.7 to 32.7 months); the median progression-free survival (PFS) was 7.5 months (95% CI 3.8 to 14.8 months); and the median DoR was 13.4 months (95% CI 5.5 to 20.6 months). The OS rates of 1, 2, and 3 years were 62.1%, 44.8%, and 31.0%, respectively. Biomarker analysis found that PD-L1 expression and TMB level were not associated with survival benefits. In contrast, a 12-GEP signature correlated with improved PFS (17.7 vs 5.7 months, p=0.0083) and OS (35.6 vs 17.6 months, p=0.039).

Conclusions: The 3-year survival update confirmed the antitumor activity and long-term survival benefit of the toripalimab plus axitinib combination in patients with advanced MM. The 12-gene GEP is of value in predicting the outcomes of vascular endothelial growth factor receptor-tyrosine kinase inhibitor and PD-1 blockade combination therapy, but requires further validation.

Trial registration numbers: NCT03086174.

Keywords: biomarkers; combination; drug therapy; immunotherapy; melanoma; tumor.

Conflict of interest statement

Competing interests: JG is a member of the advisory board/consultant of MSD, Roche, Pfizer, Bayer, Novartis, Simcere, Shanghai Junshi Bioscience, and Oriengene. KF serves on the board of directors of Loxo Oncology, Clovis Oncology, Strata Oncology, and Vivid Biosciences; on the corporate advisory boards of X4 Pharmaceuticals and PIC Therapeutics; on the scientific advisory boards of Sanofi, Amgen, Asana, Adaptimmune, Fount, Aeglea, Array BioPharma, Shattuck Labs, Arch Oncology, Tolero, Apricity, Oncoceutics, Fog Pharma, Neon Therapeutics, and Tvardi; and as a consultant to Novartis, Genentech, BMS, Merck, Takeda, Verastem, Checkmate, Boston Biomedical, Pierre Fabre, Cell Medica, and Debiopharm. XT, HF, and SY are employed by Shanghai Junshi Bioscience. The rest of the authors have no disclosures of potential conflicts of interest.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY. Published by BMJ.

Figures

Figure 1
Figure 1
(A) Waterfall plot. Maximal change of tumor size from baseline assessed by investigator per RECIST V.1.1 (n=33). The length of the bar represents maximal decrease or minimal increase in target lesion(s). (B) Spider plot. Change in individual tumor burden over time from baseline assessed by investigator per RECIST V.1.1 (n=33). PD, progressive disease; PD-L1, programmed cell death ligand-1; PR, partial response; RECIST, Response Evaluation Criteria in Solid Tumors; SD, stable disease; TMB, tumor mutational burden.
Figure 2
Figure 2
(A) DoR by RECIST V.1.1. (B) PFS by Response Evaluation Criteria in Solid Tumors V.1.1. (C) OS of 29 patients with chemotherapy-naïve mucosal melanoma. Probability of survival is shown at indicated time points. Censored patients are marked with a vertical line in the graph. Numbers of patients at risk at indicated time points are shown below the x-axis. DoR, duration of response; OS, overall survival; PFS, progression-free survival.
Figure 3
Figure 3
Forest plot. Subgroup analysis of overall survival of 29 patients with chemotherapy-naïve mucosal melanoma. The scale is months (0–50) in the forest plot. ECOG, Eastern Cooperative Oncology Group; GEP, gene expression profile; LDH, lactate dehydrogenase; NA, not applicable; OS, overall survival; PD-L1, programmed cell death ligand-1; TMB, Tumor mutational burden.
Figure 4
Figure 4
PFS and OS by biomarkers include (A, B) PD-L1, (C, D) TMB, and (E, F) GEP. GEP, gene expression profile; OS, overall survival; PFS, progression-free survival; PD-L1, programmed cell death ligand-1; TMB, tumor mutational burden

References

    1. Shoushtari AN, Bluth MJ, Goldman DA, et al. . Clinical features and response to systemic therapy in a historical cohort of advanced or unresectable mucosal melanoma. Melanoma Res 2017;27:57–64. 10.1097/CMR.0000000000000306
    1. McLaughlin CC, Wu X-C, Jemal A, et al. . Incidence of noncutaneous melanomas in the U.S. Cancer 2005;103:1000–7. 10.1002/cncr.20866
    1. Chi Z, Li S, Sheng X, et al. . Clinical presentation, histology, and prognoses of malignant melanoma in ethnic Chinese: a study of 522 consecutive cases. BMC Cancer 2011;11:85. 10.1186/1471-2407-11-85
    1. Shoushtari AN. Incorporating VEGF blockade into a shifting treatment paradigm for mucosal melanoma. J Clin Oncol 2021;39:JCO2003523:867–9. 10.1200/JCO.20.03523
    1. Furney SJ, Turajlic S, Stamp G, et al. . Genome sequencing of mucosal melanomas reveals that they are driven by distinct mechanisms from cutaneous melanoma. J Pathol 2013;230:261–9. 10.1002/path.4204
    1. Curtin JA, Busam K, Pinkel D, et al. . Somatic activation of KIT in distinct subtypes of melanoma. J Clin Oncol 2006;24:4340–6. 10.1200/JCO.2006.06.2984
    1. Kim HS, Jung M, Kang HN, et al. . Oncogenic BRAF fusions in mucosal melanomas activate the MAPK pathway and are sensitive to MEK/PI3K inhibition or MEK/CDK4/6 inhibition. Oncogene 2017;36:3334–45. 10.1038/onc.2016.486
    1. Curtin JA, Fridlyand J, Kageshita T, et al. . Distinct sets of genetic alterations in melanoma. N Engl J Med 2005;353:2135–47. 10.1056/NEJMoa050092
    1. Hayward NK, Wilmott JS, Waddell N, et al. . Whole-genome landscapes of major melanoma subtypes. Nature 2017;545:175–80. 10.1038/nature22071
    1. Tang B, Chi Z, Chen Y, et al. . Safety, efficacy, and biomarker analysis of toripalimab in previously treated advanced melanoma: results of the POLARIS-01 multicenter phase II trial. Clin Cancer Res 2020;26:4250–9. 10.1158/1078-0432.CCR-19-3922
    1. Si L, Zhang X, Shu Y, et al. . A phase Ib study of pembrolizumab as second-line therapy for Chinese patients with advanced or metastatic melanoma (KEYNOTE-151). Transl Oncol 2019;12:828–35. 10.1016/j.tranon.2019.02.007
    1. Hamid O, Robert C, Ribas A, et al. . Antitumour activity of pembrolizumab in advanced mucosal melanoma: a post-hoc analysis of KEYNOTE-001, 002, 006. Br J Cancer 2018;119:670–4. 10.1038/s41416-018-0207-6
    1. Robert C, Long GV, Brady B, et al. . Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 2015;372:320–30. 10.1056/NEJMoa1412082
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. . Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med 2015;373:23–34. 10.1056/NEJMoa1504030
    1. Robert C, Schachter J, Long GV, et al. . Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med 2015;372:2521–32. 10.1056/NEJMoa1503093
    1. D'Angelo SP, Larkin J, Sosman JA, et al. . Efficacy and safety of nivolumab alone or in combination with ipilimumab in patients with mucosal melanoma: a pooled analysis. J Clin Oncol 2017;35:226–35. 10.1200/JCO.2016.67.9258
    1. Shoushtari AN, Wagstaff J, Ascierto PA, Butler MO, et al. . CheckMate 067: long-term outcomes in patients with mucosal melanoma. JCO 2020;38:10019. 10.1200/JCO.2020.38.15_suppl.10019
    1. Yan X, Sheng X, Chi Z, et al. . Randomized phase II study of bevacizumab in combination with carboplatin plus paclitaxel in patients with previously untreated advanced mucosal melanoma. J Clin Oncol 2021;39:881–9. 10.1200/JCO.20.00902
    1. 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. 10.1111/cei.12069
    1. Sheng X, Yan X, Chi Z, et al. . Axitinib in combination with toripalimab, a humanized immunoglobulin G4 monoclonal antibody against programmed cell death-1, in patients with metastatic mucosal melanoma: an open-label phase IB trial. J Clin Oncol 2019;37:2987–99. 10.1200/JCO.19.00210
    1. Scorer P, Scott M, Lawson N, et al. . Consistency of tumor and immune cell programmed cell death ligand-1 expression within and between tumor blocks using the VENTANA SP263 assay. Diagn Pathol 2018;13:47. 10.1186/s13000-018-0725-9
    1. McDermott DF, Huseni MA, Atkins MB, et al. . Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat Med 2018;24:749–57. 10.1038/s41591-018-0053-3
    1. Ayers M, Lunceford J, Nebozhyn M, et al. . IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 2017;127:2930–40. 10.1172/JCI91190
    1. Samstein RM, Lee C-H, Shoushtari AN, et al. . Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet 2019;51:202–6. 10.1038/s41588-018-0312-8
    1. Postow MA, Hamid O, Carvajal RD. Mucosal melanoma: pathogenesis, clinical behavior, and management. Curr Oncol Rep 2012;14:441–8. 10.1007/s11912-012-0244-x
    1. Fruehauf J, Lutzky J, McDermott D, et al. . Multicenter, phase II study of axitinib, a selective second-generation inhibitor of vascular endothelial growth factor receptors 1, 2, and 3, in patients with metastatic melanoma. Clin Cancer Res 2011;17:7462–9. 10.1158/1078-0432.CCR-11-0534
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. . Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 2019;381:1535–46. 10.1056/NEJMoa1910836
    1. Shoushtari AN, Munhoz RR, Kuk D, et al. . The efficacy of anti-PD-1 agents in acral and mucosal melanoma. Cancer 2016;122:3354–62. 10.1002/cncr.30259
    1. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity 2013;39:1–10. 10.1016/j.immuni.2013.07.012
    1. Fukumura D, Kloepper J, Amoozgar Z, et al. . Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat Rev Clin Oncol 2018;15:325–40. 10.1038/nrclinonc.2018.29
    1. Prince EA, Sanzari JK, Pandya D, et al. . Analytical concordance of PD-L1 assays utilizing antibodies from FDA-approved diagnostics in advanced cancers: a systematic literature review. JCO Precis Oncol 2021;5:953–73. 10.1200/PO.20.00412
    1. Nishino M, Ramaiya NH, Hatabu H, et al. . Monitoring immune-checkpoint blockade: response evaluation and biomarker development. Nat Rev Clin Oncol 2017;14:655–68. 10.1038/nrclinonc.2017.88
    1. Topalian SL, Taube JM, Anders RA, et al. . Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer 2016;16:275–87. 10.1038/nrc.2016.36
    1. Torlakovic E, Lim HJ, Adam J, et al. . "Interchangeability" of PD-L1 immunohistochemistry assays: a meta-analysis of diagnostic accuracy. Mod Pathol 2020;33:4–17. 10.1038/s41379-019-0327-4
    1. Jardim DL, Goodman A, de Melo Gagliato D, et al. . The challenges of tumor mutational burden as an immunotherapy biomarker. Cancer Cell 2021;39:154–73. 10.1016/j.ccell.2020.10.001
    1. Miao D, Margolis CA, Vokes NI, et al. . Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors. Nat Genet 2018;50:1271–81. 10.1038/s41588-018-0200-2

Source: PubMed

3
Suscribir