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

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.

[1]  R. Edwards,et al.  Analytical Concordance of PD-L1 Assays Utilizing Antibodies From FDA-Approved Diagnostics in Advanced Cancers: A Systematic Literature Review , 2021, JCO precision oncology.

[2]  A. Shoushtari Incorporating VEGF Blockade Into a Shifting Treatment Paradigm for Mucosal Melanoma. , 2021, Journal of Clinical Oncology.

[3]  Li Zhou,et al.  Randomized Phase II Study of Bevacizumab in Combination With Carboplatin Plus Paclitaxel in Patients With Previously Untreated Advanced Mucosal Melanoma. , 2021, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[4]  A. Goodman,et al.  The Challenges of Tumor Mutational Burden as an Immunotherapy Biomarker. , 2020, Cancer cell.

[5]  P. Ascierto,et al.  CheckMate 067: Long-term outcomes in patients with mucosal melanoma. , 2020 .

[6]  Haifeng Song,et al.  Safety, Efficacy, and Biomarker Analysis of Toripalimab in Previously Treated Advanced Melanoma: Results of the POLARIS-01 Multicenter Phase II Trial , 2020, Clinical Cancer Research.

[7]  K. Flaherty,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 , 2019, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[8]  Chao Li,et al.  “Interchangeability” of PD-L1 immunohistochemistry assays: a meta-analysis of diagnostic accuracy , 2019, Modern Pathology.

[9]  Yanhong Gu,et al.  A Phase Ib Study of Pembrolizumab as Second-Line Therapy for Chinese Patients With Advanced or Metastatic Melanoma (KEYNOTE-151) , 2019, Translational oncology.

[10]  C. Brennan,et al.  Tumor mutational load predicts survival after immunotherapy across multiple cancer types , 2019, Nature Genetics.

[11]  A. Ribas,et al.  Antitumour activity of pembrolizumab in advanced mucosal melanoma: a post-hoc analysis of KEYNOTE-001, 002, 006 , 2018, British Journal of Cancer.

[12]  M. Rebelatto,et al.  Consistency of tumor and immune cell programmed cell death ligand-1 expression within and between tumor blocks using the VENTANA SP263 assay , 2018, Diagnostic Pathology.

[13]  J. Reeves,et al.  Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma , 2018, Nature Medicine.

[14]  G. Getz,et al.  Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors , 2018, Nature Genetics.

[15]  Dai Fukumura,et al.  Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges , 2018, Nature Reviews Clinical Oncology.

[16]  F. Hodi,et al.  Monitoring immune-checkpoint blockade: response evaluation and biomarker development , 2017, Nature Reviews Clinical Oncology.

[17]  H. Kim,et al.  Oncogenic BRAF fusions in mucosal melanomas activate the MAPK pathway and are sensitive to MEK/PI3K inhibition or MEK/CDK4/6 inhibition , 2017, Oncogene.

[18]  Catherine A. Shang,et al.  Whole-genome landscapes of major melanoma subtypes , 2017, Nature.

[19]  J. Wolchok,et al.  Clinical features and response to systemic therapy in a historical cohort of advanced or unresectable mucosal melanoma , 2017, Melanoma research.

[20]  K. Savage,et al.  Efficacy and Safety of Nivolumab Alone or in Combination With Ipilimumab in Patients With Mucosal Melanoma: A Pooled Analysis. , 2017, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[21]  R. Sullivan,et al.  The efficacy of anti‐PD‐1 agents in acral and mucosal melanoma , 2016, Cancer.

[22]  J. Taube,et al.  Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy , 2016, Nature Reviews Cancer.

[23]  M. Valsecchi Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. , 2015, The New England journal of medicine.

[24]  J. Larkin,et al.  Pembrolizumab versus Ipilimumab in Advanced Melanoma. , 2015, The New England journal of medicine.

[25]  D. Schadendorf,et al.  Nivolumab in previously untreated melanoma without BRAF mutation. , 2015, The New England journal of medicine.

[26]  I. Mellman,et al.  Oncology meets immunology: the cancer-immunity cycle. , 2013, Immunity.

[27]  G. Stamp,et al.  Genome sequencing of mucosal melanomas reveals that they are driven by distinct mechanisms from cutaneous melanoma , 2013, The Journal of pathology.

[28]  H. Yoshiji,et al.  Simultaneous blockade of programmed death 1 and vascular endothelial growth factor receptor 2 (VEGFR2) induces synergistic anti‐tumour effect in vivo , 2013, Clinical and experimental immunology.

[29]  M. Postow,et al.  Mucosal Melanoma: Pathogenesis, Clinical Behavior, and Management , 2012, Current Oncology Reports.

[30]  D. McDermott,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 , 2011, Clinical Cancer Research.

[31]  C. Cui,et al.  Clinical presentation, histology, and prognoses of malignant melanoma in ethnic Chinese: A study of 522 consecutive cases , 2011, BMC Cancer.

[32]  D. Pinkel,et al.  Somatic activation of KIT in distinct subtypes of melanoma. , 2006, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[33]  J. Fridlyand,et al.  Distinct sets of genetic alterations in melanoma. , 2005, The New England journal of medicine.

[34]  Ahmedin Jemal,et al.  Incidence of noncutaneous melanomas in the U.S. , 2005, Cancer.

[35]  D. Schadendorf,et al.  Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. , 2019, The New England journal of medicine.

[36]  J. Lunceford,et al.  IFN- γ –related mRNA profile predicts clinical response to PD-1 blockade , 2017 .