American Association for Cancer Research
Browse
23266066cir180098-sup-197301_2_supp_5032190_pf27rc.pdf (20.2 MB)

Supplementary Figures from FAP Delineates Heterogeneous and Functionally Divergent Stromal Cells in Immune-Excluded Breast Tumors

Download (20.2 MB)
journal contribution
posted on 2023-04-03, 23:20 authored by Viviana Cremasco, Jillian L. Astarita, Angelo L. Grauel, Shilpa Keerthivasan, Kenzie MacIsaac, Matthew C. Woodruff, Michael Wu, Lotte Spel, Stephen Santoro, Zohreh Amoozgar, Tyler Laszewski, Sara Cruz Migoni, Konstantin Knoblich, Anne L. Fletcher, Martin LaFleur, Kai W. Wucherpfennig, Ellen Pure, Glenn Dranoff, Michael C. Carroll, Shannon J. Turley

Supplementary Figures 1-8

Funding

NIH

Barr Foundation

American Cancer Society

Cancer Research Institute

Science Foundation

History

ARTICLE ABSTRACT

Cancer-associated fibroblasts (CAFs) are generally associated with poor clinical outcome. CAFs support tumor growth in a variety of ways and can suppress antitumor immunity and response to immunotherapy. However, a precise understanding of CAF contributions to tumor growth and therapeutic response is lacking. Discrepancies in this field of study may stem from heterogeneity in the composition and function of fibroblasts in the tumor microenvironment. Furthermore, it remains unclear whether CAFs directly interact with and suppress T cells. Here, mouse and human breast tumors were used to examine stromal cells expressing fibroblast activation protein (FAP), a surface marker for CAFs. Two discrete populations of FAP+ mesenchymal cells were identified on the basis of podoplanin (PDPN) expression: a FAP+PDPN+ population of CAFs and a FAP+PDPN− population of cancer-associated pericytes (CAPs). Although both subsets expressed extracellular matrix molecules, the CAF transcriptome was enriched in genes associated with TGFβ signaling and fibrosis compared with CAPs. In addition, CAFs were enriched at the outer edge of the tumor, in close contact with T cells, whereas CAPs were localized around vessels. Finally, FAP+PDPN+ CAFs suppressed the proliferation of T cells in a nitric oxide–dependent manner, whereas FAP+PDPN− pericytes were not immunosuppressive. Collectively, these findings demonstrate that breast tumors contain multiple populations of FAP-expressing stromal cells of dichotomous function, phenotype, and location.

Usage metrics

    Cancer Immunology Research

    Licence

    Exports

    RefWorks
    BibTeX
    Ref. manager
    Endnote
    DataCite
    NLM
    DC