Whole stromal fibroblast signature is linked to specific chemokine and immune infiltration patterns and to improved survival in NSCLC

Oncoimmunology. 2023 Nov 28;12(1):2274130. doi: 10.1080/2162402X.2023.2274130. eCollection 2023.

Abstract

Cancer associated fibroblasts (CAF) are known to orchestrate multiple components of the tumor microenvironment, whereas the influence of the whole stromal-fibroblast compartment is less understood. Here, an extended stromal fibroblast signature was investigated to define its impact on immune cell infiltration. The lung cancer adenocarcinoma (LUAD) data set of the cancer genome atlas (TCGA) was used to test whole stroma signatures and cancer-associated fibroblast signatures for their impact on prognosis. 3D cell cultures of the NSCLC cancer cell line A549 together with the fibroblast cell line SV80 were used in combination with infiltrating peripheral blood mononuclear cells (PBMC) for in-vitro investigations. Immune cell infiltration was assessed via flow cytometry, chemokines were analyzed by immunoassays and RNA microarrays. Results were confirmed in specimens from NSCLC patients by flow cytometry or immunohistochemistry as well as in the TCGA data set. The TCGA analyses correlated the whole stromal-fibroblast signature with an improved outcome, whereas no effect was found for the CAF signatures. In 3D microtumors, the presence of fibroblasts induced infiltration of B cells and CD69+CD4+ T cells, which was linked to an increased expression of CCL13 and CXCL16. The stroma/lymphocyte interaction was confirmed in NSCLC patients, as stroma-rich tumors displayed an elevated B cell count and survival in the local cohort and the TCGA data set. A whole stromal fibroblast signature was associated with an improved clinical outcome in lung adenocarcinoma and in vitro and in vivo experiments suggest that this signature increases B and T cell recruitment via induction of chemokines.

Keywords: CAF; cancer; co-culture; immune cells; infiltration; microenvironment.

MeSH terms

  • Adenocarcinoma of Lung* / pathology
  • Carcinoma, Non-Small-Cell Lung* / genetics
  • Carcinoma, Non-Small-Cell Lung* / pathology
  • Chemokines / genetics
  • Chemokines / metabolism
  • Fibroblasts / metabolism
  • Fibroblasts / pathology
  • Humans
  • Leukocytes, Mononuclear / metabolism
  • Lung Neoplasms* / genetics
  • Lung Neoplasms* / pathology
  • Tumor Microenvironment

Substances

  • Chemokines

Grants and funding

This work was funded by means of a PhD grant of the Austrian Society of Haematology and Oncology (OeGHO, www.oegho.at) for SK. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.