Journal for ImmunoTherapy of Cancer (Aug 2024)

Nanoparticles targeting immune checkpoint protein VISTA induce potent antitumor immunity

  • Taylor J Moon,
  • Hieu Minh Ta,
  • Anubhuti Bhalotia,
  • Kai E Paulsen,
  • Diarmuid W Hutchinson,
  • Gabrielle M Arkema,
  • Andrew S Choi,
  • Michiko G Haynie,
  • Laolu Ogunnaike,
  • Margee Dever,
  • Li Lily Wang,
  • Efstathios Karathanasis

DOI
https://doi.org/10.1136/jitc-2024-008977
Journal volume & issue
Vol. 12, no. 8

Abstract

Read online

Background Immune checkpoint protein V-domain immunoglobulin suppressor of T cell activation (VISTA) controls antitumor immunity and is a valuable target for cancer immunotherapy. Previous mechanistic studies have indicated that VISTA impairs the toll-like receptor (TLR)-mediated activation of myeloid antigen-presenting cells, promoting the expansion of myeloid-derived suppressor cells, and suppressing tumor-reactive cytotoxic T cell function.Methods The aim of this study was to develop a dual-action lipid nanoparticle (dual-LNP) coloaded with VISTA-specific siRNA and TLR9 agonist CpG oligonucleotide. We used three murine preclinical tumor models, melanoma YUMM1.7, melanoma B16F10, and colon carcinoma MC38 to assess the functional synergy of the two cargoes of the dual LNP and therapeutic efficacy.Results The dual-LNP synergistically augmented antitumor immune responses and rejected large established tumors whereas LNPs containing VISTA siRNA or CpG alone were ineffective. In comparison with therapies using the soluble CpG and a VISTA-specific monoclonal antibody, the dual-LNP demonstrated superior therapeutic efficacy yet with reduced systemic inflammatory cytokine production. In three murine models, the dual-LNP treatment achieved a high cure rate. Tumor rejection was associated with influx of immune cells to tumor tissues, augmented dendritic cell activation, production of proinflammatory cytokines, and improved function of cytotoxic T cells.Conclusions Our studies show the dual-LNP ensured codelivery of its synergistic cargoes to tumor-infiltrating myeloid cells, leading to simultaneous silencing of VISTA and stimulation of TLR9. As a result, the dual-LNP drove a highly potent antitumor immune response that rejected large aggressive tumors, thus may be a promising therapeutic platform for treating immune-cold tumors.