Siglec-9 acts as an immune-checkpoint molecule on macrophages in glioblastoma, restricting T-cell priming and immunotherapy response

Nat Cancer. 2023 Sep;4(9):1273-1291. doi: 10.1038/s43018-023-00598-9. Epub 2023 Jul 17.

Abstract

Neoadjuvant immune-checkpoint blockade therapy only benefits a limited fraction of patients with glioblastoma multiforme (GBM). Thus, targeting other immunomodulators on myeloid cells is an attractive therapeutic option. Here, we performed single-cell RNA sequencing and spatial transcriptomics of patients with GBM treated with neoadjuvant anti-PD-1 therapy. We identified unique monocyte-derived tumor-associated macrophage subpopulations with functional plasticity that highly expressed the immunosuppressive SIGLEC9 gene and preferentially accumulated in the nonresponders to anti-PD-1 treatment. Deletion of Siglece (murine homolog) resulted in dramatically restrained tumor development and prolonged survival in mouse models. Mechanistically, targeting Siglece directly activated both CD4+ T cells and CD8+ T cells through antigen presentation, secreted chemokines and co-stimulatory factor interactions. Furthermore, Siglece deletion synergized with anti-PD-1/PD-L1 treatment to improve antitumor efficacy. Our data demonstrated that Siglec-9 is an immune-checkpoint molecule on macrophages that can be targeted to enhance anti-PD-1/PD-L1 therapeutic efficacy for GBM treatment.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • B7-H1 Antigen
  • CD8-Positive T-Lymphocytes / pathology
  • Glioblastoma* / genetics
  • Glioblastoma* / therapy
  • Humans
  • Immune Checkpoint Proteins / genetics
  • Immune Checkpoint Proteins / therapeutic use
  • Immunotherapy / methods
  • Macrophages / pathology
  • Mice

Substances

  • B7-H1 Antigen
  • Immune Checkpoint Proteins

Associated data

  • figshare/10.6084/m9.figshare.22434341