Journal for ImmunoTherapy of Cancer (Mar 2022)

Enhanced antigen cross-presentation in human colorectal cancer-associated fibroblasts through upregulation of the lysosomal protease cathepsin S

  • Peter A van Veelen,
  • Jacques Neefjes,
  • Sjoerd H van der Burg,
  • Thorbald van Hall,
  • Marten Visser,
  • Linda de Bruin,
  • Lukas JAC Hawinkels,
  • James CH Hardwick,
  • Saskia J Santegoets,
  • Tom J Harryvan,
  • Léonie Plug,
  • Lisa Griffioen,
  • Arend Mulder,
  • Gerbrand J van der Heden van Noort,
  • Marlieke LM Jongsma,
  • Miranda H Meeuwsen,
  • Emmanuel JHJ Wiertz,
  • Els ME Verdegaal

DOI
https://doi.org/10.1136/jitc-2021-003591
Journal volume & issue
Vol. 10, no. 3

Abstract

Read online

Background Cross-presentation of exogenous antigens in HLA-class I molecules by professional antigen presenting cells (APCs) is crucial for CD8+ T cell function. Recent murine studies show that several non-professional APCs, including cancer-associated fibroblasts (CAFs) also possess this capacity. Whether human CAFs are able to cross-present exogenous antigen, which molecular pathways are involved in this process and how this ultimately affects tumor-specific CD8+ T cell function is unknown.Methods In this study, we investigated the ability of human colorectal cancer (CRC)-derived CAFs to cross-present neoantigen-derived synthetic long peptides (SLPs), corresponding to tumor-derived mutant peptides, and how this affects tumor-specific T-cell function. Processing of the SLP was studied by targeting components of the cross-presentation machinery through CRISPR/Cas9 and siRNA-mediated genetic ablation to identify the key molecules involved in fibroblast-mediated cross-presentation. Multispectral flow cytometry and killing assays were performed to study the effect of fibroblast cross-presentation on T cell function.Results Here, we show that human CRC-derived CAFs display an enhanced capacity to cross-present neoantigen-derived SLPs when compared with normal colonic fibroblasts. Cross-presentation of antigens by fibroblasts involved the lysosomal protease cathepsin S. Cathepsin S expression by CAFs was detected in situ in human CRC tissue, was upregulated in ex vivo cultured CRC-derived CAFs and showed increased expression in normal fibroblasts after exposure to CRC-conditioned medium. Cognate interaction between CD8+ T cells and cross-presenting CAFs suppressed T cell function, reflected by decreased cytotoxicity, reduced activation (CD137) and increased exhaustion (TIM3, LAG3 and CD39) marker expression.Conclusion These data indicate that CAFs may directly suppress tumor-specific T cell function in an antigen-dependent fashion in human CRC.