CD8+ T cell priming that is required for curative intratumorally anchored anti-4-1BB immunotherapy is constrained by Tregs

Nat Commun. 2024 Mar 1;15(1):1900. doi: 10.1038/s41467-024-45625-0.

Abstract

Although co-stimulation of T cells with agonist antibodies targeting 4-1BB (CD137) improves antitumor immune responses in preclinical studies, clinical success has been limited by on-target, off-tumor activity. Here, we report the development of a tumor-anchored ɑ4-1BB agonist (ɑ4-1BB-LAIR), which consists of a ɑ4-1BB antibody fused to the collagen-binding protein LAIR. While combination treatment with an antitumor antibody (TA99) shows only modest efficacy, simultaneous depletion of CD4+ T cells boosts cure rates to over 90% of mice. Mechanistically, this synergy depends on ɑCD4 eliminating tumor draining lymph node regulatory T cells, resulting in priming and activation of CD8+ T cells which then infiltrate the tumor microenvironment. The cytotoxic program of these newly primed CD8+ T cells is then supported by the combined effect of TA99 and ɑ4-1BB-LAIR. The combination of TA99 and ɑ4-1BB-LAIR with a clinically approved ɑCTLA-4 antibody known for enhancing T cell priming results in equivalent cure rates, which validates the mechanistic principle, while the addition of ɑCTLA-4 also generates robust immunological memory against secondary tumor rechallenge. Thus, our study establishes the proof of principle for a clinically translatable cancer immunotherapy.

MeSH terms

  • 4-1BB Ligand / immunology
  • Animals
  • Antibodies
  • Antineoplastic Agents*
  • CD8-Positive T-Lymphocytes
  • Immunotherapy
  • Mice
  • Neoplasms* / immunology
  • Neoplasms* / therapy
  • T-Lymphocytes, Regulatory*
  • Tumor Microenvironment

Substances

  • Antibodies
  • Antineoplastic Agents
  • 4-1BB Ligand