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

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

Abstract

Purpose: Metastatic mucosal melanoma responds poorly to anti-programmed cell death-1 (PD-1) monotherapy. Vascular endothelial growth factor (VEGF) has been shown to play an important immunosuppressive role in the tumor microenvironment. The combination of VEGF inhibition and PD-1 blockade provides therapeutic opportunities for patients refractory to either therapy alone.

Patients and methods: We conducted a single-center, phase IB trial evaluating the safety and preliminary efficacy of toripalimab, a humanized immunoglobulin G4 monoclonal antibody against PD-1 in combination with the VEGF receptor inhibitor axitinib in patients with advanced melanoma, including patients with chemotherapy-naïve mucosal melanomas (88%). Patients received toripalimab at 1 or 3 mg/kg via intravenous infusion every 2 weeks, in combination with axitinib 5 mg orally twice a day, in a dose-escalation and cohort-expansion study until confirmed disease progression, unacceptable toxicity, or voluntary withdrawal. The primary objective was safety. Secondary objectives included efficacy, pharmacokinetics, pharmacodynamics, immunogenicity, and tumor tissue biomarkers.

Results: Thirty-three patients were enrolled. No dose-limiting toxicities were observed. Ninety-seven percent of patients experienced treatment-related adverse events (TRAEs). The most common TRAEs were mild (grade 1 or 2) and included diarrhea, proteinuria, hand and foot syndrome, fatigue, AST or ALT elevation, hypertension, hypo- or hyperthyroidism, and rash. Grade 3 or greater TRAEs occurred in 39.4% of patients. By the cutoff date, among 29 patients with chemotherapy-naïve mucosal melanoma, 14 patients (48.3%; 95% CI, 29.4% to 67.5%) achieved objective response, and the median progression-free survival time was 7.5 months (95% CI, 3.7 months to not reached) per Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1.

Conclusion: The combination of toripalimab plus axitinib was tolerable and showed promising antitumor activity in patients with treatment-naïve metastatic mucosal melanoma. Patients enrolled in this study were all Asian, and this combination therapy must be validated in a randomized phase III trial that includes a non-Asian population before it can become a standard of care.

Figures

FIG 1.
FIG 1.
(A) Maximal change of tumor size from baseline assessed by investigator per Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 (N = 33). The length of the bar represents maximal decrease or minimal increase in target lesion(s). (B) Change in individual tumor burden over time from baseline assessed by investigator per RECIST version 1.1 (N = 33). (C) Exposure and duration of response per RECIST version 1.1 (N = 33). (*) Patient with target lesion(s) reduction of more than 30% but with new lesion(s) or progression of nontarget lesion(s). (†) Unconfirmed partial response classified as stable disease. Mbp, million base pairs; PD-L1, programmed death ligand-1; TMB, tumor mutational burden.
FIG 2.
FIG 2.
Progression-free survival by (A) Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 and (B) Immune-Related RECIST (irRECIST) and (C) overall survival 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. NR, not reached.
FIG 3.
FIG 3.
Clinical response in relation to tumor programmed death ligand-1 (PD-L1) expression and tumor mutational burden (TMB) in patients with chemotherapy-naïve mucosal melanoma. (A) PD-L1–positive status was defined as the presence of membrane staining of any intensity in 1% or more of tumor cells or immune cells by SP263 immunohistochemistry (IHC) staining. TMB was calculated by summing up somatic mutations within the coding regions by whole-exon sequencing. A TMB of 6 mutations per million base pairs (Mbp) was the cutoff value. (B) Progression-free survival per Immune-Related Response Evaluation Criteria in Solid Tumors (irRECIST) of PD-L1–positive and PD-L1–negative patients. (C) Progression-free survival per irRECIST of patients with TMB of 6 mutations/Mbp or greater and patients with TMB of less than 6 mutations/Mbp. PD-L1–positive status was defined as the presence of membrane staining of any intensity in 1% or more of tumor cells or immune cells by SP263 IHC staining. 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. HR, hazard ratio; NR, not reached.
FIG 4.
FIG 4.
Inflammatory signature and correlation with clinical response. RNA sequencing and expression profiling of tumor biopsies were acquired from 24 patients with chemotherapy-naïve mucosal melanoma. The expression signatures of six inflammation-related genes (IL-6, CXCL1, CXCL2, CXCL3, CXCL8, and PTGS2), six angiogenesis-related genes (VEGFA, KDR, ESM1, PECAM1, ANGPTL4, and CD34), or 12 selected immune- or angiogenesis-related genes (CD274/PD-L1, CXCR6, CD27, CXCL9, IDO1, TIGIT, PDCD1LG2/PD-L2, LAG3, ANGPTL5, ANGPTL6, CD34, and KDR) were evaluated. To obtain a single score for the signature for each sample, the mean expression of the genes composing the signature was calculated. (A) The scores between patients with objective response (partial response [PR]; n = 13) per Immune-Related Response Evaluation Criteria in Solid Tumors (irRECIST) and patients with stable disease (SD) or progressive disease (PD; n = 11) were compared (P < .001). (B) The scores between patients achieving clinical benefit (PR or SD; n = 21) and patients with PD (n = 3) were compared (P < .001). (C) Associations between tumor mutational burden (TMB) and inflammation and angiogenesis gene expression profiles were also evaluated by Spearman correlation. Median TMB and median gene expression profiling scores are shown as dashed lines in the figure. r and P values of the correlations are provided. Mbp, million base pairs.
FIG A1.
FIG A1.
Design of phase IB combination study of toripalimab plus axitinib in treating patients with mucosal melanoma and clinical efficacy summary. DLT, dose-limiting toxicity; IV, intravenous; PD, progressive disease; PR, partial response; SD, stable disease.
FIG A2.
FIG A2.
CONSORT diagram for phase IB combination study of toripalimab with axitinib in treating patients with mucosal melanoma.
FIG A3.
FIG A3.
Serum concentration of toripalimab over 56 days in the combination study.
FIG A4.
FIG A4.
Kaplan-Meier plots of median overall survival of (A) Patients who are PD-L1 positive versus PD-L1 negative. (B) TMB ≥ 6 mutations/Mbp versus TMB

FIG A5.

The correlation of clinical response…

FIG A5.

The correlation of clinical response with inflammation or angiogenesis signature panel scores. McDermott…

FIG A5.
The correlation of clinical response with inflammation or angiogenesis signature panel scores. McDermott et al. inflammation panel: IL-6, CXCL1, CXCL2, CXCL3, CXCL8, and PTGS2; McDermott et al. angiogenesis panel: VEGFA, KDR, ESM1, PECAM1, ANGPTL4, and CD34; Cristescu et al. inflammation panel: IDO1, CXCL10, CXCL9, HLADRA, STAT1, and IFN-g. Responders are those with complete or partial response; nonresponders are those with stable or progressive disease.

FIG A6.

Genetic alternations and frequencies identified…

FIG A6.

Genetic alternations and frequencies identified by whole exome sequencing (WES) from 31 available…

FIG A6.
Genetic alternations and frequencies identified by whole exome sequencing (WES) from 31 available patients. Patients were grouped by clinical responses. PD, progressive disease; PR, partial response; SD, stable disease.
All figures (10)
FIG A5.
FIG A5.
The correlation of clinical response with inflammation or angiogenesis signature panel scores. McDermott et al. inflammation panel: IL-6, CXCL1, CXCL2, CXCL3, CXCL8, and PTGS2; McDermott et al. angiogenesis panel: VEGFA, KDR, ESM1, PECAM1, ANGPTL4, and CD34; Cristescu et al. inflammation panel: IDO1, CXCL10, CXCL9, HLADRA, STAT1, and IFN-g. Responders are those with complete or partial response; nonresponders are those with stable or progressive disease.
FIG A6.
FIG A6.
Genetic alternations and frequencies identified by whole exome sequencing (WES) from 31 available patients. Patients were grouped by clinical responses. PD, progressive disease; PR, partial response; SD, stable disease.

References

    1. Chang AE, Karnell LH, Menck HR. The National Cancer Data Base report on cutaneous and noncutaneous melanoma: A summary of 84,836 cases from the past decade—The American College of Surgeons Commission on Cancer and the American Cancer Society. Cancer. 1998;83:1664–1678.
    1. Chan KK, Chan RC, Ho RS, et al. Clinical patterns of melanoma in Asians: 11-year experience in a tertiary referral center. Ann Plast Surg. 2016;77(suppl 1):S6–S11.
    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.
    1. Hayward NK, Wilmott JS, Waddell N, et al. Whole-genome landscapes of major melanoma subtypes. Nature. 2017;545:175–180.
    1. Wang X, Si L, Guo J. Treatment algorithm of metastatic mucosal melanoma. Linchuang Zhongliuxue Zazhi. 2014;3:38.
    1. Lian B, Cui CL, Zhou L, et al. The natural history and patterns of metastases from mucosal melanoma: An analysis of 706 prospectively-followed patients. Ann Oncol. 2017;28:868–873.
    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.
    1. Curtin JA, Fridlyand J, Kageshita T, et al. Distinct sets of genetic alterations in melanoma. N Engl J Med. 2005;353:2135–2147.
    1. Curtin JA, Busam K, Pinkel D, et al. Somatic activation of KIT in distinct subtypes of melanoma. J Clin Oncol. 2006;24:4340–4346.
    1. Kong Y, Si L, Zhu Y, et al. Large-scale analysis of KIT aberrations in Chinese patients with melanoma. Clin Cancer Res. 2011;17:1684–1691.
    1. Si L, Kong Y, Xu X, et al. Prevalence of BRAF V600E mutation in Chinese melanoma patients: Large scale analysis of BRAF and NRAS mutations in a 432-case cohort. Eur J Cancer. 2012;48:94–100.
    1. Bai X, Mao LL, Chi ZH, et al. BRAF inhibitors: Efficacious and tolerable in BRAF-mutant acral and mucosal melanoma. Neoplasma. 2017;64:626–632.
    1. Carvajal RD, Antonescu CR, Wolchok JD, et al. KIT as a therapeutic target in metastatic melanoma. JAMA. 2011;305:2327–2334.
    1. Guo J, Si L, Kong Y, et al. Phase II, open-label, single-arm trial of imatinib mesylate in patients with metastatic melanoma harboring c-Kit mutation or amplification. J Clin Oncol. 2011;29:2904–2909.
    1. Hodi FS, Corless CL, Giobbie-Hurder A, et al. Imatinib for melanomas harboring mutationally activated or amplified KIT arising on mucosal, acral, and chronically sun-damaged skin. J Clin Oncol. 2013;31:3182–3190.
    1. Si L, Zhang X, Shu Y, et al: KEYNOTE-151: A phase 1b study of second-line pembrolizumab (Pembro) for Chinese patients (Pts) with advanced or metastatic melanoma. Ann Oncol 29:viii442-viii466, 2018 (suppl 8)
    1. Fu J, Wang F, Dong LH, et al. Preclinical evaluation of the efficacy, pharmacokinetics and immunogenicity of JS-001, a programmed cell death protein-1 (PD-1) monoclonal antibody. Acta Pharmacol Sin. 2017;38:710–718.
    1. Chi Z, Tang B, Sheng X, et al: A phase II study of JS001, a humanized PD-1 mAb, in patients with advanced melanoma in China. J Clin Oncol 36, 2018 (suppl 15; abstr 9539)
    1. Simonetti O, Lucarini G, Rubini C, et al. Clinical and prognostic significance of survivin, AKT and VEGF in primary mucosal oral melanoma. Anticancer Res. 2015;35:2113–2120.
    1. Cui C, Tang B, Guo J. Chemotherapy, biochemotherapy and anti-VEGF therapy in metastatic mucosal melanoma. Linchuang Zhongliuxue Zazhi. 2014;3:36.
    1. Johnson BF, Clay TM, Hobeika AC, et al. Vascular endothelial growth factor and immunosuppression in cancer: Current knowledge and potential for new therapy. Expert Opin Biol Ther. 2007;7:449–460.
    1. Voron T, Colussi O, Marcheteau E, et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J Exp Med. 2015;212:139–148.
    1. Dirkx AE, oude Egbrink MG, Castermans K, et al. Anti-angiogenesis therapy can overcome endothelial cell anergy and promote leukocyte-endothelium interactions and infiltration in tumors. FASEB J. 2006;20:621–630.
    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;172:500–506.
    1. Tang B, Yan X, Sheng X, et al. Safety and clinical activity with an anti-PD-1 antibody JS001 in advanced melanoma or urologic cancer patients. J Hematol Oncol. 2019;12:7.
    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.
    1. Samstein RM, Lee CH, Shoushtari AN, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51:202–206.
    1. Cristescu R, Mogg R, Ayers M, et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science. 2018;362:eaar3593.
    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;24:749–757.
    1. Yi JH, Yi SY, Lee HR, et al. Dacarbazine-based chemotherapy as first-line treatment in noncutaneous metastatic melanoma: Multicenter, retrospective analysis in Asia. Melanoma Res. 2011;21:223–227.
    1. Kaufman HL, Margolin K, Sullivan R. Management of metastatic melanoma in 2018. JAMA Oncol. 2018;4:857–858.
    1. Kim KB, Alrwas A. Treatment of KIT-mutated metastatic mucosal melanoma. Chin Clin Oncol. 2014;3:35.
    1. Beadling C, Jacobson-Dunlop E, Hodi FS, et al. KIT gene mutations and copy number in melanoma subtypes. Clin Cancer Res. 2008;14:6821–6828.
    1. Postow MA, Luke JJ, Bluth MJ, et al. Ipilimumab for patients with advanced mucosal melanoma. Oncologist. 2013;18:726–732.
    1. Alexander M, Mellor JD, McArthur G, et al. Ipilimumab in pretreated patients with unresectable or metastatic cutaneous, uveal and mucosal melanoma. Med J Aust. 2014;201:49–53.
    1. Del Vecchio M, Di Guardo L, Ascierto PA, et al. Efficacy and safety of ipilimumab 3 mg/kg in patients with pretreated, metastatic, mucosal melanoma. Eur J Cancer. 2014;50:121–127.
    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;35:226–235.
    1. Eroglu Z, Zaretsky JM, Hu-Lieskovan S, et al. High response rate to PD-1 blockade in desmoplastic melanomas. Nature. 2018;553:347–350.
    1. Hellmann MD, Callahan MK, Awad MM, et al. Tumor mutational burden and efficacy of nivolumab monotherapy and in combination with ipilimumab in small-cell lung cancer. Cancer Cell 33:853-861.e4, 2018 .
    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;127:2930–2940.
    1. Atkins MB, Plimack ER, Puzanov I, et al. Axitinib in combination with pembrolizumab in patients with advanced renal cell cancer: A non-randomised, open-label, dose-finding, and dose-expansion phase 1b trial. Lancet Oncol. 2018;19:405–415.
    1. Lee C-H, Makker V, Rasco DW, et al. Lenvatinib + pembrolizumab in patients with renal cell carcinoma: Updated results. J Clin Oncol. 2018;36(suppl 15; abstr 4560)
    1. Weber JS, Hodi FS, Wolchok JD, et al. Safety profile of nivolumab monotherapy: A pooled analysis of patients with advanced melanoma. J Clin Oncol. 2017;35:785–792.

Source: PubMed

3
Předplatit