American Association for Cancer Research
Browse
10780432ccr190733-sup-218231_2_supp_5651888_pkrmk1.docx (16.92 kB)

Table S1 from TFEB Mediates Immune Evasion and Resistance to mTOR Inhibition of Renal Cell Carcinoma via Induction of PD-L1

Download (16.92 kB)
journal contribution
posted on 2023-03-31, 21:02 authored by Cai Zhang, Yaqi Duan, Minghui Xia, Yuting Dong, Yufei Chen, Lu Zheng, Shuaishuai Chai, Qian Zhang, Zhengping Wei, Na Liu, Jing Wang, Chaoyang Sun, Zhaohui Tang, Xiang Cheng, Jie Wu, Guoping Wang, Fang Zheng, Arian Laurence, Bing Li, Xiang-Ping Yang

Patient characteristics

Funding

National Scientific Foundation of China

Tongji Medical College

History

ARTICLE ABSTRACT

Despite the FDA approval of mTOR inhibitors (mTORi) for the treatment of renal cell carcinoma (RCC), the benefits are relatively modest and the few responders usually develop resistance. We investigated whether the resistance to mTORi is due to upregulation of PD-L1 and the underlying molecular mechanism. The effects of transcription factor EB (TFEB) on RCC proliferation, apoptosis, and migration were evaluated. Correlation of TFEB with PD-L1 expression, as well as effects of mTOR inhibition on TFEB and PD-L1 expression, was assessed in human primary clear cell RCCs. The regulation of TFEB on PD-L1 was assessed by chromatin immunoprecipitation and luciferase reporter assay. The therapeutic efficacies of mTORi plus PD-L1 blockade were evaluated in a mouse model. The function of tumor-infiltrating CD8+ T cells was analyzed by flow cytometry. TFEB did not affect tumor cell proliferation, apoptosis, and migration. We found a positive correlation between TFEB and PD-L1 expression in RCC tumor tissues, primary tumor cells, and RCC cells. TFEB bound to PD-L1 promoter in RCCs and inhibition of mTOR led to enhanced TFEB nuclear translocation and PD-L1 expression. Simultaneous inhibition of mTOR and blockade of PD-L1 enhanced CD8+ cytolytic function and tumor suppression in a xenografted mouse model of RCC. These data revealed that TFEB mediates resistance to mTOR inhibition via induction of PD-L1 in human primary RCC tumors, RCC cells, and murine xenograft model. Our data provide a strong rationale to target mTOR and PD-L1 jointly as a novel immunotherapeutic approach for RCC treatment.