Ford et al. generated cancer-associated fibroblast (CAF)-rich murine models of lung, breast, and colorectal cancers to show that CAFs promote tumor growth and resistance to immunotherapy (anti-cancer vaccination and anti-PD-1) by suppressing CD8+ T cell infiltration in the tumors. CD8+ T cells from CAF-rich tumors upregulated CTLA-4 and were predominantly localized at tumor margins. NOX4 or TGFβ1 inhibition prevented CAF differentiation, but only NOX4 inhibition normalized CAFs into a quiescent fibroblast-like state, promoted CD8+ T cell infiltration, and re-sensitized CAF-rich tumors to anti-cancer vaccination and anti-PD-1 therapy.

Contributed by Shishir Pant

Determining mechanisms of resistance to PD-1/PD-L1 immune checkpoint immunotherapy is key to developing new treatment strategies. Cancer-associated fibroblasts (CAF) have many tumor-promoting functions and promote immune evasion through multiple mechanisms, but as yet, there are no CAF-specific inhibitors clinically available. Here we generated CAF-rich murine tumor models (TC1, MC38, 4T1) to investigate how CAF influence the immune microenvironment and affect response to different immunotherapy modalities (anti-cancer vaccination; TC1, [HPV E7 DNA vaccine];PD-1, MC38) and found that CAFs broadly suppressed response by specifically excluding CD8+ T-cells from tumors (not CD4+ T-cells or macrophages); CD8+ T-cell exclusion was similarly present in CAF-rich human tumors. RNA sequencing of CD8+ T-cells from CAF-rich murine tumors and immunochemistry analysis of human tumors identified significant upregulation of CTLA-4 in the absence of other exhaustion markers; inhibiting CTLA-4 with a non-depleting antibody overcame the CD8+ T-cell exclusion effect without affecting T-regs. We then examined the potential for CAF targeting, focusing on the ROS-producing enzyme NOX4, which is upregulated by CAF in many human cancers, and compared this to TGF-062;1 inhibition, a key regulator of the CAF phenotype. siRNA knockdown or pharmacological inhibition (GKT137831 [Setanaxib]) of NOX4 'normalized' CAF to a quiescent phenotype and promoted intratumoral CD8+T-cell infiltration, overcoming the exclusion effect; TGF-062;1 inhibition could prevent, but not reverse, CAF differentiation. Finally, NOX4 inhibition restored immunotherapy response in CAF-rich tumors. These findings demonstrate that CAF-mediated immunotherapy resistance can be effectively overcome through NOX4 inhibition, and could improve outcome in a broad range of cancers.

Author Info: (1) Cancer sciences, University of Southampton. (2) Cancer Sciences Unit, Faculty of Medicine, University of Southampton. (3) Cancer Sciences Division, School of Medicine, Universi

Author Info: (1) Cancer sciences, University of Southampton. (2) Cancer Sciences Unit, Faculty of Medicine, University of Southampton. (3) Cancer Sciences Division, School of Medicine, University of Southampton. (4) Genkyotex S.A. (5) Screening and Biotechnology, Genkyotex Suisse SA. (6) Genkyotex (Switzerland). (7) Cancer Sciences, University of Southampton. (8) University of Southampton. (9) Pathology, University Hospital Southampton. (10) Vccine Discovery, La Jolla Institute for Immunology. (11) Cancer Science Unit, University of Southampton. (12) Princess Margaret Cancer Centre. (13) School of Biosciences, University of Kent. (14) ENT, Poole Hospital NHS Foundation Trust. (15) 9420 Athena Circle, La Jolla Institute For Allergy & Immunology. (16) Cancer Sciences Division, University of Southampton. (17) Cancer Sciences Unit, Faculty of Medicine, University of Southampton. (18) Cancer Sciences, University of Southampton School of Medicine. (19) Cancer Sciences Unit, University of Southampton, Faculty of Medicine g.thomas@soton.ac.uk.