The Journal of Clinical Investigation (Jan 2023)

Canagliflozin primes antitumor immunity by triggering PD-L1 degradation in endocytic recycling

  • Ling Ding,
  • Xi Chen,
  • Wenxin Zhang,
  • Xiaoyang Dai,
  • Hongjie Guo,
  • Xiaohui Pan,
  • Yanjun Xu,
  • Jianguo Feng,
  • Meng Yuan,
  • Xiaomeng Gao,
  • Jian Wang,
  • Xiaqing Xu,
  • Sicheng Li,
  • Honghai Wu,
  • Ji Cao,
  • Qiaojun He,
  • Bo Yang

Journal volume & issue
Vol. 133, no. 1

Abstract

Read online

Understanding the regulatory mechanisms of PD-L1 expression in tumors provides key clues for improving immune checkpoint blockade efficacy or developing novel oncoimmunotherapy. Here, we showed that the FDA-approved sodium-glucose cotransporter-2 (SGLT2) inhibitor canagliflozin dramatically suppressed PD-L1 expression and enhanced T cell–mediated cytotoxicity. Mechanistic study revealed that SGLT2 colocalized with PD-L1 at the plasma membrane and recycling endosomes and thereby prevented PD-L1 from proteasome-mediated degradation. Canagliflozin disturbed the physical interaction between SGLT2 and PD-L1 and subsequently allowed the recognition of PD-L1 by Cullin3SPOP E3 ligase, which triggered the ubiquitination and proteasome-mediated degradation of PD-L1. In mouse models and humanized immune-transformation models, either canagliflozin treatment or SGLT2 silencing significantly reduced PD-L1 expression and limited tumor progression — to a level equal to the PD-1 mAb — which was correlated with an increase in the activity of antitumor cytotoxic T cells. Notably, prolonged progression-free survival and overall survival curves were observed in the group of PD-1 mAb–treated patients with non–small cell lung cancer with high expression of SGLT2. Therefore, our study identifies a regulator of cell surface PD-L1, provides a ready-to-use small-molecule drug for PD-L1 degradation, and highlights a potential therapeutic target to overcome immune evasion by tumor cells.

Keywords