OncoImmunology (Dec 2023)

Tumor-intrinsic RGS1 potentiates checkpoint blockade response via ATF3-IFNGR1 axis

  • Baojun Wang,
  • Bo Jiang,
  • Lin Du,
  • Wenyuan Chen,
  • Qing Zhang,
  • Wei Chen,
  • Meng Ding,
  • Wenmin Cao,
  • Jie Gao,
  • Yongming Deng,
  • Yao Fu,
  • Yan Li,
  • Yonglong Xiao,
  • Wenli Diao,
  • Hongqian Guo

DOI
https://doi.org/10.1080/2162402X.2023.2279800
Journal volume & issue
Vol. 12, no. 1

Abstract

Read online

ABSTRACTBackground Non-responsiveness is a major barrier in current cancer immune checkpoint blockade therapies, and the mechanism has not been elucidated yet. Therefore, it is necessary to discover the mechanism and biomarkers of tumor immunotherapeutic resistance.Methods Bioinformatics analysis was performed based on CD8+ T cell infiltration in multiple tumor databases to screen out genes related to anti-tumor immunity. Associations between Regulator of G-protein signaling 1 (RGS1) and IFNγ-STAT1 signaling, and MHCI antigen presentation pathway were examined by RT-qPCR, western blotting, and flow cytometry. The modulatory mechanisms of RGS1 were investigated via CHIP-qPCR and dual-luciferase assay. The clinical and therapeutic implications of RGS1 were comprehensively investigated using tumor cell lines, mouse models, and clinical samples receiving immunotherapy.Results RGS1 was identified as the highest gene positively correlated with immunogenicity among RGS family. Inhibition of RGS1 in neoplastic cells dampened anti-tumor immune response and elicited resistance to immunotherapy in both renal and lung murine subcutaneous tumors. Mechanistically, RGS1 enhanced the binding of activating transcription factor 3 (ATF3) to the promoter of interferon gamma receptor 1 (IFNGR1), activated STAT1 and the subsequent expression of IFNγ-inducible genes, especially CXCL9 and MHC class I (MHCI), thereby influenced CD8+ T cell infiltration and antigen presentation and processing. Clinically, lower expression level of RGS1 was associated with resistance of PD1 inhibition therapy and shortened progression-free survival among 21 NSCLC patients receiving immunotherapy.Conclusions Together, these findings uncover a novel mechanism that elicits immunotherapy resistance and highlight the function of tumor-intrinsic RGS1, which brings new insights for future strategies to sensitize anti-PD1 immunotherapy.

Keywords