Abstract

TGF-β is secreted in the tumour microenvironment in a latent, inactive form bound to latency associated protein and activated by the integrin αV subunit. The activation of latent TGF-β by cancer-cell-expressed αV re-shapes the tumour microenvironment, and this could affect patient responses to PD-1-targeting therapy. Here we show, using multiplex immunofluorescence staining in cohorts of anti-PD-1 and anti-PD-L1-treated lung cancer patients, that decreased expression of cancer cell αV is associated with improved immunotherapy-related, progression-free survival, as well as with an increased density of CD8+CD103+ tumour-infiltrating lymphocytes. Mechanistically, tumour αV regulates CD8 T cell recruitment, induces CD103 expression on activated CD8+ T cells and promotes their differentiation to granzyme B-producing CD103+CD69+ resident memory T cells via autocrine TGF-β signalling. Thus, our work provides the underlying principle of targeting cancer cell αV for more efficient PD-1 checkpoint blockade therapy.

Highlights

  • TGF-β is secreted in the tumour microenvironment in a latent, inactive form bound to latency associated protein and activated by the integrin αV subunit

  • To investigate the impact of tumour αV expression on survival in patients with lung cancer, we used a retrospective cohort of 113 patients with treatment-naïve early-stage NSCLC15

  • We did not find any significant difference in overall survival (OS) of patients bearing αVlow and αVhigh tumours (Fig. 1b), with a hazard ratio (HR) = 1.05, 95% confidence interval (CI) 0.38−2.97, and median OS of 68 months

Read more

Summary

Introduction

TGF-β is secreted in the tumour microenvironment in a latent, inactive form bound to latency associated protein and activated by the integrin αV subunit. The quality of CD8+ tumour-infiltrating lymphocytes (TIL), especially their reactivity toward the cognate target, is directly associated with the efficacy of anti-PD-1 In this regard, expression of PD-1 on CD8+ TIL appeared to define clonally expanded tumour neoantigen-specific T cells detected in cancer patients[9,10]. Human non-small-cell lung cancer (NSCLC) CD103+CD8+ TRM cells frequently express the activation marker CD69 and a panel of T cell inhibitory receptors, including PD-114. They are enriched with tumour-reactive T lymphocytes able to kill autologous tumour cells upon blockade of PD-1 with neutralizing antibodies[15]. CD103+CD8+ TRM expands during anti-PD-1 immunotherapy, and their accumulation in tumours is associated with the improved outcome of anti-PD-(L)1-treated patients[14,18,19]

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.