Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2019

Open Access 01-12-2019 | Kidney Cancer | Research

Safety and clinical activity with an anti-PD-1 antibody JS001 in advanced melanoma or urologic cancer patients

Authors: Bixia Tang, Xieqiao Yan, Xinan Sheng, Lu Si, Chuanliang Cui, Yan Kong, Lili Mao, Bin Lian, Xue Bai, Xuan Wang, Siming Li, Li Zhou, Jiayi Yu, Jie Dai, Kai Wang, Jinwei Hu, Lihou Dong, Haifeng Song, Hai Wu, Hui Feng, Sheng Yao, Zhihong Chi, Jun Guo

Published in: Journal of Hematology & Oncology | Issue 1/2019

Login to get access

Abstract

Background

JS001, a humanized IgG4 monoclonal antibody against the programmed death-1 (PD-1) receptor, blocks the interaction of PD-1 with its ligands and promotes T cell activation in preclinical studies. This phase I study is designed to evaluate the safety, tolerability, and clinical activity of JS001 in advanced melanoma or urologic cancer patients who are refractory to standard systemic therapy.

Patients and methods

In the dose escalation cohorts, subjects initially received a single-dose, intravenous infusion of JS001, and were followed for 28 days followed by multi-dose infusions every 2 weeks. In the dose expansion cohorts, subjects received multi-dose infusions every 2 weeks. Clinical response was evaluated after each 8-week treatment cycle according to RECIST v1.1 criteria.

Results

Thirty-six subjects diagnosed with advanced melanoma (n = 22), urothelial cancer (UC) (n = 8), or renal cell cancer (RCC) (n = 6) were enrolled. Melanoma subjects included 14 acral and 4 mucosal subtypes. JS001 was well tolerated, and no dose-limiting toxicity was observed. By the safety data cutoff date, 100% of subjects had treatment-related adverse events (TRAE) with most adverse events being grade 1 or 2, and ≥ grade 3 TRAEs occurred in 36%. Among all 36 subjects, 1 confirmed complete response (acral melanoma), 7 confirmed partial responses (2 acral melanoma, 1 mucosal melanoma, 2 UC, and 2 RCC), and 10 stable disease were observed, for an objective response rate of 22.2% (95% CI, 10.1 to 39.2), and a disease control rate of 50.0% (95% CI, 32.9 to 67.1). Clinical responses were correlated with PD-L1 expression on tumor cells, the presence of tumor infiltrating lymphocytes (TIL), baseline tumor volume, ECOG performance status, serum LDH levels, high percentage of activated CD8+ T cells and CD3− CD16+ CD54+ NK cells in the peripheral blood as well as tumor mutational burden (TMB).

Conclusion

JS001 was well tolerated and demonstrated promising anti-tumor activity in UC and RCC as well as in previously underexplored acral and mucosal melanoma subtypes. Subjects with an immune-active profile in the tumor microenvironment or in peripheral blood responded favorably to JS001 treatment. The completion of the current phase I study has led to the initiation of the first prospective anti-PD-1 registration trial in Asia focusing on acral and mucosal melanoma subtypes, representative of the regional disease epidemiology.

Trial registration

Clinical Trial ID: NCT02836795, registered July 19, 2016, retrospectively registered.
Appendix
Available only for authorised users
Literature
1.
go back to reference Ahmadzadeh M, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114(8):1537–44.CrossRef Ahmadzadeh M, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114(8):1537–44.CrossRef
2.
go back to reference Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol. 2006;6(4):295–307.CrossRef Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol. 2006;6(4):295–307.CrossRef
3.
go back to reference Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol. 2008;8(6):467–77.CrossRef Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol. 2008;8(6):467–77.CrossRef
4.
go back to reference Blank C, Mackensen A. Contribution of the PD-L1/PD-1 pathway to T-cell exhaustion: an update on implications for chronic infections and tumor evasion. Cancer Immunol Immunother. 2007;56(5):739–45.CrossRef Blank C, Mackensen A. Contribution of the PD-L1/PD-1 pathway to T-cell exhaustion: an update on implications for chronic infections and tumor evasion. Cancer Immunol Immunother. 2007;56(5):739–45.CrossRef
5.
go back to reference Dong H, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8(8):793–800.CrossRef Dong H, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8(8):793–800.CrossRef
6.
go back to reference Agata Y, et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 1996;8(5):765–72.CrossRef Agata Y, et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 1996;8(5):765–72.CrossRef
7.
go back to reference Ishida Y, et al. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11(11):3887–95.CrossRef Ishida Y, et al. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11(11):3887–95.CrossRef
8.
go back to reference Freeman GJ, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192(7):1027–34.CrossRef Freeman GJ, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192(7):1027–34.CrossRef
9.
go back to reference Tsushima F, et al. Interaction between B7-H1 and PD-1 determines initiation and reversal of T-cell anergy. Blood. 2007;110(1):180–5.CrossRef Tsushima F, et al. Interaction between B7-H1 and PD-1 determines initiation and reversal of T-cell anergy. Blood. 2007;110(1):180–5.CrossRef
10.
go back to reference Zou W, Wolchok JD, Chen L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: mechanisms, response biomarkers, and combinations. Sci Transl Med. 2016;8(328):328rv4.CrossRef Zou W, Wolchok JD, Chen L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: mechanisms, response biomarkers, and combinations. Sci Transl Med. 2016;8(328):328rv4.CrossRef
11.
go back to reference Blank C, et al. PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Res. 2004;64(3):1140–5.CrossRef Blank C, et al. PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Res. 2004;64(3):1140–5.CrossRef
12.
go back to reference Hirano F, et al. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 2005;65(3):1089–96.PubMed Hirano F, et al. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 2005;65(3):1089–96.PubMed
13.
go back to reference Topalian SL, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014;32(10):1020–30.CrossRef Topalian SL, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014;32(10):1020–30.CrossRef
14.
go back to reference Hamid O, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369(2):134–44.CrossRef Hamid O, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369(2):134–44.CrossRef
15.
go back to reference Lo JA, Fisher DE. The melanoma revolution: from UV carcinogenesis to a new era in therapeutics. Science. 2014;346(6212):945–9.CrossRef Lo JA, Fisher DE. The melanoma revolution: from UV carcinogenesis to a new era in therapeutics. Science. 2014;346(6212):945–9.CrossRef
16.
go back to reference McLaughlin CC, et al. Incidence of noncutaneous melanomas in the U.S. Cancer. 2005;103(5):1000–7.CrossRef McLaughlin CC, et al. Incidence of noncutaneous melanomas in the U.S. Cancer. 2005;103(5):1000–7.CrossRef
17.
go back to reference Chi Z, et al. Clinical presentation, histology, and prognoses of malignant melanoma in ethnic Chinese: a study of 522 consecutive cases. BMC Cancer. 2011;11:85.CrossRef Chi Z, et al. Clinical presentation, histology, and prognoses of malignant melanoma in ethnic Chinese: a study of 522 consecutive cases. BMC Cancer. 2011;11:85.CrossRef
18.
go back to reference Furney SJ, et al. The mutational burden of acral melanoma revealed by whole-genome sequencing and comparative analysis. Pigment Cell Melanoma Res. 2014;27(5):835–8.CrossRef Furney SJ, et al. The mutational burden of acral melanoma revealed by whole-genome sequencing and comparative analysis. Pigment Cell Melanoma Res. 2014;27(5):835–8.CrossRef
19.
go back to reference Cho J, et al. Treatment outcome of PD-1 immune checkpoint inhibitor in Asian metastatic melanoma patients: correlative analysis with PD-L1 immunohistochemistry. Investig New Drugs. 2016;34(6):677–84.CrossRef Cho J, et al. Treatment outcome of PD-1 immune checkpoint inhibitor in Asian metastatic melanoma patients: correlative analysis with PD-L1 immunohistochemistry. Investig New Drugs. 2016;34(6):677–84.CrossRef
20.
go back to reference Fu J, 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(5):710–8.CrossRef Fu J, 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(5):710–8.CrossRef
21.
go back to reference Wang MH, et al. 33A model based adjustment for tumor mutational burden across different tumor types [J]. Annals of Oncology. 2017;28(suppl_7). Wang MH, et al. 33A model based adjustment for tumor mutational burden across different tumor types [J]. Annals of Oncology. 2017;28(suppl_7).
22.
go back to reference Taube JM, et al. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med. 2012;4(127):127ra37.CrossRef Taube JM, et al. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med. 2012;4(127):127ra37.CrossRef
23.
go back to reference Abdul Karim L, et al. Collaborative multi-institutional experience in comparing PD-L1 immunohistochemistry assays: concordance of SP142 and 22C3 immunoreactivity. Appl Immunohistochem Mol Morphol. 2018;26(2):e22.PubMed Abdul Karim L, et al. Collaborative multi-institutional experience in comparing PD-L1 immunohistochemistry assays: concordance of SP142 and 22C3 immunoreactivity. Appl Immunohistochem Mol Morphol. 2018;26(2):e22.PubMed
24.
go back to reference Eroglu Z, et al. High response rate to PD-1 blockade in desmoplastic melanomas. Nature. 2018;553(7688):347–50.CrossRef Eroglu Z, et al. High response rate to PD-1 blockade in desmoplastic melanomas. Nature. 2018;553(7688):347–50.CrossRef
25.
go back to reference Hellmann MD, et al. Tumor mutational burden and efficacy of nivolumab monotherapy and in combination with ipilimumab in small-cell lung cancer. Cancer Cell. 2018;33(5):853–861 e4.CrossRef Hellmann MD, et al. Tumor mutational burden and efficacy of nivolumab monotherapy and in combination with ipilimumab in small-cell lung cancer. Cancer Cell. 2018;33(5):853–861 e4.CrossRef
26.
go back to reference Hayward NK, et al. Whole-genome landscapes of major melanoma subtypes. Nature. 2017;545(7653):175–80.CrossRef Hayward NK, et al. Whole-genome landscapes of major melanoma subtypes. Nature. 2017;545(7653):175–80.CrossRef
27.
go back to reference Nakamura Y, Fujisawa Y. Diagnosis and management of acral lentiginous melanoma. Curr Treat Options in Oncol. 2018;19(8):42.CrossRef Nakamura Y, Fujisawa Y. Diagnosis and management of acral lentiginous melanoma. Curr Treat Options in Oncol. 2018;19(8):42.CrossRef
28.
go back to reference Navazio AS, et al. EMSY copy number variation in male breast cancers characterized for BRCA1 and BRCA2 mutations. Breast Cancer Res Treat. 2016;160(1):181–6.CrossRef Navazio AS, et al. EMSY copy number variation in male breast cancers characterized for BRCA1 and BRCA2 mutations. Breast Cancer Res Treat. 2016;160(1):181–6.CrossRef
29.
go back to reference Kong Y, et al. Frequent genetic aberrations in the CDK4 pathway in acral melanoma indicate the potential for CDK4/6 inhibitors in targeted therapy. Clin Cancer Res. 2017;23(22):6946–57.CrossRef Kong Y, et al. Frequent genetic aberrations in the CDK4 pathway in acral melanoma indicate the potential for CDK4/6 inhibitors in targeted therapy. Clin Cancer Res. 2017;23(22):6946–57.CrossRef
30.
go back to reference Cui C, Tang B, Guo J. Chemotherapy, biochemotherapy and anti-VEGF therapy in metastatic mucosal melanoma. Chin Clin Oncol. 2014;3(3):36.PubMed Cui C, Tang B, Guo J. Chemotherapy, biochemotherapy and anti-VEGF therapy in metastatic mucosal melanoma. Chin Clin Oncol. 2014;3(3):36.PubMed
Metadata
Title
Safety and clinical activity with an anti-PD-1 antibody JS001 in advanced melanoma or urologic cancer patients
Authors
Bixia Tang
Xieqiao Yan
Xinan Sheng
Lu Si
Chuanliang Cui
Yan Kong
Lili Mao
Bin Lian
Xue Bai
Xuan Wang
Siming Li
Li Zhou
Jiayi Yu
Jie Dai
Kai Wang
Jinwei Hu
Lihou Dong
Haifeng Song
Hai Wu
Hui Feng
Sheng Yao
Zhihong Chi
Jun Guo
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2019
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-018-0693-2

Other articles of this Issue 1/2019

Journal of Hematology & Oncology 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine