Targeting nucleic acid sensors in tumor cells to reprogram biogenesis and RNA cargo of extracellular vesicles for T cell-mediated cancer immunotherapy

Cell Rep Med. 2023 Sep 19;4(9):101171. doi: 10.1016/j.xcrm.2023.101171. Epub 2023 Aug 31.

Abstract

Tumor-derived extracellular vesicles (EVs) have been associated with immune evasion and tumor progression. We show that the RNA-sensing receptor RIG-I within tumor cells governs biogenesis and immunomodulatory function of EVs. Cancer-intrinsic RIG-I activation releases EVs, which mediate dendritic cell maturation and T cell antitumor immunity, synergizing with immune checkpoint blockade. Intact RIG-I, autocrine interferon signaling, and the GTPase Rab27a in tumor cells are required for biogenesis of immunostimulatory EVs. Active intrinsic RIG-I signaling governs composition of the tumor EV RNA cargo including small non-coding stimulatory RNAs. High transcriptional activity of EV pathway genes and RIG-I in melanoma samples associate with prolonged patient survival and beneficial response to immunotherapy. EVs generated from human melanoma after RIG-I stimulation induce potent antigen-specific T cell responses. We thus define a molecular pathway that can be targeted in tumors to favorably alter EV immunomodulatory function. We propose "reprogramming" of tumor EVs as a personalized strategy for T cell-mediated cancer immunotherapy.

Keywords: RIG-I; RNA; STING; cancer immunotherapy; cancer resistance; extracellular vesicles; innate immunity; nucleic acid receptors; personalized therapy.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Humans
  • Immunotherapy
  • Melanoma* / genetics
  • Melanoma* / therapy
  • Nucleic Acids*
  • RNA
  • RNA, Neoplasm
  • T-Lymphocytes

Substances

  • RNA
  • Nucleic Acids
  • RNA, Neoplasm