SLAMF7 Signaling Reprograms T Cells toward Exhaustion in the Tumor Microenvironment

J Immunol. 2021 Jan 1;206(1):193-205. doi: 10.4049/jimmunol.2000300. Epub 2020 Dec 7.

Abstract

T cell exhaustion represents one of the most pervasive strategies tumors employ to circumvent the immune system. Although repetitive, cognate TCR signaling is recognized as the primary driving force behind this phenomenon, and it remains unknown what other forces drive T cell exhaustion in the tumor microenvironment (TME). In this study, we show that activation of the self-ligand SLAMF7 immune receptor on T cells induced STAT1 and STAT3 phosphorylation, expression of multiple inhibitory receptors, and transcription factors associated with T cell exhaustion. Analysis of The Cancer Genome Atlas revealed that SLAMF7 transcript levels were strongly correlated with various inhibitory receptors and that high SLAMF7 expression was indicative of poor survival in clear cell renal cell carcinoma (ccRCC). Targeted reanalysis of a CyTOF dataset, which profiled the TME in 73 ccRCC patients, revealed cell-type-specific SLAMF7 expression patterns, strong correlations between exhausted T cells and SLAMF7+ tumor-associated macrophages (TAMs), and a unique subset of SLAMF7highCD38high TAMs. These SLAMF7highCD38high TAMs showed the strongest correlations with exhausted T cells and were an independent prognostic factor in ccRCC. Confirmatory ex vivo coculture studies validated that SLAMF7-SLAMF7 interactions between murine TAMs and CD8+ T cells induce expression of multiple inhibitory receptors. Finally, mice lacking SLAMF7 show restricted growth of B16-F10 tumors, and CD8+ T cells from these mice express less PD-1 and TOX and exhibited an impaired ability to progress through the exhaustion developmental trajectory to terminal exhaustion. These findings suggest that SLAMF7 might play an important role in modulating T cell function in the TME.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Carcinoma, Renal Cell / immunology
  • Carcinoma, Renal Cell / metabolism*
  • Carcinoma, Renal Cell / mortality
  • Cells, Cultured
  • Cellular Reprogramming
  • Female
  • Gene Expression Regulation, Neoplastic
  • Humans
  • Immune Tolerance
  • Kidney Neoplasms / immunology
  • Kidney Neoplasms / metabolism*
  • Kidney Neoplasms / mortality
  • Male
  • Melanoma / immunology
  • Melanoma / metabolism*
  • Melanoma, Experimental
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Neoplasms, Experimental
  • Signal Transduction
  • Signaling Lymphocytic Activation Molecule Family / genetics
  • Signaling Lymphocytic Activation Molecule Family / metabolism*
  • Skin Neoplasms / immunology
  • Skin Neoplasms / metabolism*
  • Survival Analysis
  • T-Lymphocytes / immunology*
  • Tumor Microenvironment

Substances

  • SLAMF7 protein, human
  • Signaling Lymphocytic Activation Molecule Family
  • Slamf7 protein, mouse