Abstract

Adenosine is an immunosuppressive factor that limits anti-tumor immunity through the suppression of multiple immune subsets including T cells via activation of the adenosine A2A receptor (A2AR). Using both murine and human chimeric antigen receptor (CAR) T cells, here we show that targeting A2AR with a clinically relevant CRISPR/Cas9 strategy significantly enhances their in vivo efficacy, leading to improved survival of mice. Effects evoked by CRISPR/Cas9 mediated gene deletion of A2AR are superior to shRNA mediated knockdown or pharmacological blockade of A2AR. Mechanistically, human A2AR-edited CAR T cells are significantly resistant to adenosine-mediated transcriptional changes, resulting in enhanced production of cytokines including IFNγ and TNF, and increased expression of JAK-STAT signaling pathway associated genes. A2AR deficient CAR T cells are well tolerated and do not induce overt pathologies in mice, supporting the use of CRISPR/Cas9 to target A2AR for the improvement of CAR T cell function in the clinic.

Highlights

  • Adenosine is an immunosuppressive factor that limits anti-tumor immunity through the suppression of multiple immune subsets including T cells via activation of the adenosine A2A receptor (A2AR)

  • chimeric antigen receptor (CAR) T cells were stimulated with an anti-CAR antibody in the presence or absence of NECA, an adenosine mimetic, as well as SCH58261, a selective A2AR antagonist

  • In the context of cells activated through the CAR, transcriptional changes induced by NECA were fully reversible with addition of the A2AR antagonist SCH58261, demonstrating that adenosine acts on CAR T-cell effector function principally through A2AR activation (Fig. 1B)

Read more

Summary

Introduction

Adenosine is an immunosuppressive factor that limits anti-tumor immunity through the suppression of multiple immune subsets including T cells via activation of the adenosine A2A receptor (A2AR) Using both murine and human chimeric antigen receptor (CAR) T cells, here we show that targeting A2AR with a clinically relevant CRISPR/Cas[9] strategy significantly enhances their in vivo efficacy, leading to improved survival of mice. Approved for the treatment of relapsed B-cell acute lymphoblastic leukemia and aggressive lymphoma following remarkable clinical response rates and defined curative potential[3] These effects have not been recapitulated in the solid tumor setting, where CAR T cells are faced with additional barriers such as tumor antigen heterogeneity, the requirement to traffic to the tumor site, and an immunosuppressive, hypoxic tumor microenvironment[1,2]. This work has set the scene for alternative strategies to target this pathway, including therapeutics directed toward the upstream ectoenzymes CD73, CD39, and CD38, or the upstream Hypoxia-HIF-1α axis itself[8,10] signifying the clinical interest in targeting this pathway[13]

Methods
Results
Conclusion
Full Text
Published version (Free)

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