Tislelizumab in Asian patients with previously treated locally advanced or metastatic urothelial carcinoma

Dingwei Ye, Jiyan Liu, Aiping Zhou, Qing Zou, Hanzhong Li, Cheng Fu, Hailong Hu, Jian Huang, Shaoxing Zhu, Jie Jin, Lulin Ma, Jianming Guo, Jun Xiao, Se Hoon Park, Dahong Zhang, Xiusong Qiu, Yuanyuan Bao, Lilin Zhang, Wei Shen, Feng Bi, Dingwei Ye, Jiyan Liu, Aiping Zhou, Qing Zou, Hanzhong Li, Cheng Fu, Hailong Hu, Jian Huang, Shaoxing Zhu, Jie Jin, Lulin Ma, Jianming Guo, Jun Xiao, Se Hoon Park, Dahong Zhang, Xiusong Qiu, Yuanyuan Bao, Lilin Zhang, Wei Shen, Feng Bi

Abstract

Tislelizumab, an anti-programmed death protein-1 (PD-1) monoclonal antibody, was engineered to minimize binding to the FcγR on macrophages to abrogate antibody-dependent phagocytosis, a mechanism of T-cell clearance and potential resistance to anti-PD-1 therapy. This single-arm phase 2 trial (NCT04004221/CTR20170071) assessed the safety, tolerability, and efficacy of tislelizumab in patients with PD-L1-positive urothelial carcinoma who progressed during/following platinum-containing therapy and had no prior PD-(L)1 inhibitor treatment. Patients were considered PD-L1 positive if ≥ 25% of tumor/immune cells expressed PD-L1 when using the VENTANA™ PD-L1 (SP263) assay. The primary endpoint was objective response rate by independent review committee. As of September 16, 2019, 113 patients had a median study follow-up time of 9.4 mo. Most patients (76%) had visceral metastases, including 24% with liver and 23% with bone metastases. Among 104 efficacy-evaluable patients, confirmed objective response rate was 24% (95% confidence interval, 16, 33), including 10 complete and 15 partial responses. Median duration of response was not reached. Among 25 responders, 17/25 (68%) had ongoing responses. Median progression-free survival and overall survival times were 2.1 and 9.8 mo, respectively. The most common treatment-related adverse events were anemia (27%) and pyrexia (19%). Anemia (7%) and hyponatremia (5%) were the only grade 3-4 treatment-related adverse events and occurred in ≥ 5% of patients. Three investigator-assessed deaths were considered to be possibly related to study treatment (hepatic failure, n = 2; respiratory arrest, n = 1). Tislelizumab demonstrated meaningful clinical benefits in patients with previously treated locally advanced or metastatic PD-L1-positive urothelial carcinoma and had a manageable safety profile.

Keywords: Asian population; immunotherapy; programed death protein-1; tislelizumab; urothelial carcinoma.

Conflict of interest statement

Dingwei Ye, Jiyan Liu, Aiping Zhou, Qing Zou, Hanzhong Li, Cheng Fu, Hailong Hu, Jian Huang, Shaoxing Zhu, Jie Jin, Lulin Ma, Jianming Guo, Jun Xiao, Se Hoon Park, Dahong Zhang, and Feng Bi have nothing to disclose. Xiusong Qiu, Yuanyuan Bao, Lilin Zhang, and Wei Shen are employees of BeiGene, Ltd. The study was designed under the responsibility of BeiGene, Ltd, in conjunction with the steering committee. The study was funded, and tislelizumab was provided by, BeiGene Ltd. Additionally, BeiGene, Ltd. collected and analyzed the data and contributed to the interpretation of the study. All authors had full access to all of the data in the study and had final responsibility for the decision to submit for publication.

© 2020 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.

Figures

FIGURE 1
FIGURE 1
Patient disposition
FIGURE 2
FIGURE 2
Best percent change in sum of target lesion diameters from baseline per independent review committee in efficacy‐evaluable patients with programmed death ligand 1‐positive urothelial carcinoma
FIGURE 3
FIGURE 3
Time and duration of confirmed responses per RECIST v1.1 by independent review committee. Gray bars represent the duration of response
FIGURE 4
FIGURE 4
Objective response rates (ORRs) per RECIST v1.1 by independent review committee by population subgroup. Abbreviations: CI, confidence interval; ECOG, Eastern Cooperative Oncology Group; IC, immune cell; PD‐L1, programmed death ligand 1; TC, tumor cell
FIGURE 5
FIGURE 5
Kaplan‐Meier plot of overall survival (OS) (safety analysis set). Abbreviation: CI, confidence interval
FIGURE 6
FIGURE 6
Kaplan‐Meier plot of PFS per RECIST version 1.1 by IRC (efficacy‐evaluable set). Abbreviations: CI, confidence interval; IRC, independent review committee; PFS, progression‐free survival; RECIST v1.1, Response Evaluation Criteria in Solid Tumors version 1.1.

References

    1. Apetoh L, Ladoire S, Coukos G, Ghiringhelli F. Combining immunotherapy and anticancer agents: the right path to achieve cancer cure? Ann Oncol. 2015;26(9):1813‐1823.
    1. Gartrell BA, He T, Sharma J, Sonpavde G. Update of systemic immunotherapy for advanced urothelial carcinoma. Urol Oncol. 2017;35(12):678‐686.
    1. Dreicer R, Manola J, Roth BJ, et al. Phase III trial of methotrexate, vinblastine, doxorubicin, and cisplatin versus carboplatin and paclitaxel in patients with advanced carcinoma of the urothelium. Cancer. 2004;100(8):1639‐1645.
    1. Sternberg CN, de Mulder PH, Schornagel JH, et al. Randomized Phase III Trial of High–Dose‐Intensity Methotrexate, Vinblastine, Doxorubicin, and Cisplatin (MVAC) Chemotherapy and Recombinant Human Granulocyte Colony‐Stimulating Factor Versus Classic MVAC in Advanced Urothelial Tract Tumors: European Organization for Research and Treatment of Cancer Protocol No. 30924. J Clin Oncol. 2001;19(10):2638‐2646.
    1. De Santis M, Bellmunt J, Mead G, et al. Randomized phase II/III trial assessing gemcitabine/carboplatin and methotrexate/carboplatin/vinblastine in patients with advanced urothelial cancer who are unfit for cisplatin‐based chemotherapy: EORTC study 30986. J Clin Oncol. 2012;30(2):191‐199.
    1. Sonpavde G. PD‐1 and PD‐L1 inhibitors as salvage therapy for urothelial carcinoma. N Engl J Med. 2017;376(11):1073‐1074.
    1. Dahan R, Sega E, Engelhardt J, Selby M, Korman AJ, Ravetch JV. FcgammaRs modulate the anti‐tumor activity of antibodies targeting the PD‐1/PD‐L1 axis. Cancer Cell. 2015;28(3):285‐295.
    1. Mahoney KM, Freeman GJ, McDermott DF. The next immune‐checkpoint inhibitors: PD‐1/PD‐L1 blockade in melanoma. Clin Ther. 2015;37(4):764‐782.
    1. Wang X, Teng F, Kong L, Yu J. PD‐L1 expression in human cancers and its association with clinical outcomes. Onco Targets Ther. 2016;9:5023‐5039.
    1. Siefker‐Radtke A, Curti B. Immunotherapy in metastatic urothelial carcinoma: focus on immune checkpoint inhibition. Nat Rev Urol. 2018;15(2):112‐124.
    1. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti‐PD‐1 antibody in cancer. N Engl J Med. 2012;366(26):2443‐2454.
    1. Zhou TC, Sankin AI, Porcelli SA, Perlin DS, Schoenberg MP, Zang X. A review of the PD‐1/PD‐L1 checkpoint in bladder cancer: From mediator of immune escape to target for treatment. Urol Oncol. 2017;35(1):14‐20.
    1. Zhang T, Song X, Xu L, et al. The binding of an anti‐PD‐1 antibody to FcgammaRIota has a profound impact on its biological functions. Cancer Immunol Immunother. 2018;67(7):1079‐1090.
    1. Lee SH, Lee HT, Lim H, Kim Y, Park UB, Heo Y‐S. Crystal structure of PD‐1 in complex with an antibody‐drug tislelizumab used in tumor immune checkpoint therapy. Biochem Biophys Res Commun. 2020;527(1):226‐231.
    1. Lee A, Keam SJ. Tislelizumab: first approval. Drugs. 2020;80(6):617‐624.
    1. Feng Y, Hong Y, Sun H, et al. The molecular binding mechanism of tislelizumab, an investigational anti‐PD‐1 antibody, is differentiated from pembrolizumab and nivolumab. Presented at the 110th Annual Meeting of the American Association for Cancer Research, Atlanta, GA. March 29‐April 3, 2019; Abstract 2382.
    1. Guo J, Yuan Y, Bai Y, et al.Tislelizumab in Chinese patients with melanoma, urothelial carcinoma (UC), and renal cell carcinoma (RCC). Presented at 22nd Annual Chinese Society for Clinical Oncology Congress; September 18–22, 2019; Xiamen, China.
    1. Sandhu S, Hill A, Gan H, et al. Tislelizumab, an anti‐PD‐1 antibody, in patients with urothelial carcinoma (UC): results from an ongoing phase I/II study. Ann Oncol. 2018;29(Suppl 10):X28.
    1. Desai J, Deva S, Lee JS, et al. Phase IA/IB study of single‐agent tislelizumab, an investigational anti‐PD‐1 antibody, in solid tumors. J Immunother Cancer. 2020;8(1):e000453.
    1. Shen L, Guo J, Zhang Q, et al. Tislelizumab in Chinese patients with advanced solid tumors: an open‐label, non‐comparative, phase 1/2 study. J Immunother Cancer. 2020;8(1):e000437.
    1. Ye D, Liu J, Zhou A, et al. First report of efficacy and safety from a phase 2 trial of tislelizumab, an anti‐pd‐1 antibody, for the treatment of pd‐l1+ locally advanced or metastatic urothelial carcinoma in Asian patients. Ann Oncol. 2019;30(Suppl 5):V367.
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228‐247.
    1. Zajac M, Boothman AM, Ben Y, et al. Analytical validation and clinical utility of an immunohistochemical programmed death ligand‐1 diagnostic assay and combined tumor and immune cell scoring algorithm for durvalumab in urothelial carcinoma. Arch Pathol Lab Med. 2019;143(6):722‐731.
    1. Powles T, Necchi A, Rosen G, Hariharan S, Ab A. Anti‐programmed cell death 1/ligand 1 (PD‐1/PD‐L1) antibodies for the treatment of urothelial carcinoma: State of the art and future development. Clin Genitourin Cancer. 2018;16(2):117‐129.
    1. Fradet Y, Bellmunt J, Vaughn DJ, et al. Randomized phase III KEYNOTE‐045 trial of pembrolizumab versus paclitaxel, docetaxel, or vinflunine in recurrent advanced urothelial cancer: results of >2 years of follow‐up. Ann Oncol. 2019;30(6):970‐976.

Source: PubMed

3
Abonneren