Loss of RBMS1 promotes anti-tumor immunity through enabling PD-L1 checkpoint blockade in triple-negative breast cancer

Cell Death Differ. 2022 Nov;29(11):2247-2261. doi: 10.1038/s41418-022-01012-0. Epub 2022 May 10.

Abstract

Immunotherapy has been widely utilized in multiple tumors, however, its efficacy in the treatment of triple-negative breast cancers (TNBC) is still being challenged. Meanwhile, functions and mechanisms of RNA binding proteins in regulating immunotherapy for TNBC remain largely elusive. Here we reported that the RNA binding protein RBMS1 is prevalent among immune-cold TNBC. Through a systematic shRNA-mediated screen, we found depletion of RBMS1 significantly reduced the level of programmed death ligand 1 (PD-L1) in TNBC. Clinically, RBMS1 was increased in breast cancer and its level was positively correlated to that of PD-L1. RBMS1 ablation stimulated cytotoxic T cell mediated anti-tumor immunity. Mechanistically, RBMS1 regulated the mRNA stability of B4GALT1, a newly identified glycosyltransferase of PD-L1. Depletion of RBMS1 destabilized the mRNA of B4GALT1, inhibited the glycosylation of PD-L1 and promoted the ubiquitination and subsequent degradation of PD-L1. Importantly, combination of RBMS1 depletion with CTLA4 immune checkpoint blockade or CAR-T treatment enhanced anti-tumor T-cell immunity both in vitro and in vivo. Together, our findings provided a new immunotherapeutic strategy against TNBC by targeting the immunosuppressive RBMS1.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Antibodies / therapeutic use
  • B7-H1 Antigen* / genetics
  • B7-H1 Antigen* / metabolism
  • DNA-Binding Proteins / metabolism
  • Humans
  • Immunotherapy
  • RNA, Small Interfering / therapeutic use
  • RNA-Binding Proteins
  • Triple Negative Breast Neoplasms* / metabolism

Substances

  • B7-H1 Antigen
  • Antibodies
  • RNA, Small Interfering
  • RBMS1 protein, human
  • DNA-Binding Proteins
  • RNA-Binding Proteins