Abstract

Abstract Introduction: CD73 mediates the final dephosphorylation in the conversion of extracellular ATP to adenosine, a metabolite which signals through the A2 family of receptors. Like PD-1, adenosine appears to be part of a negative feedback loop to limit immune activation and prevent excessive inflammation in the context of tissue damage. However, intratumoral hypoxia likely drives excessive CD73-mediated adenosine generation and prevents optimal anti-tumor immune responses. We show that intratumoral adenosine levels are sufficient to drive pervasive suppression of multiple immune subsets in vitro and an orally bioavailable small molecule can prevent adenosine generation in vitro and within the tumor in vivo. Methods We examined the effects of adenosine signaling on multiple immune cell subsets through in vitro functional assays. We interrogated T cell activation, proliferation, and cytolytic activity, as well as NK cell function in vitro. Furthermore, we investigated and identified functional targets of adenosine signaling within T cells using transcriptomic and protein expression analyses. In addition, we performed in vitro myeloid differentiation assays to determine the effect of adenosine signaling on myeloid lineage maturation. Finally, we explored the activity of a novel, orally bioavailable inhibitor of CD73 in vitro and assessed its ability to rescue T cell activation and suppress AMP conversion to adenosine within the tumor microenvironment. Results and Conclusions We found that T cell-expressed CD73 was sufficient to drive adenosine generation from AMP in vitro, resulting in suppression of anti-CD3/CD28-induced T cell activation and proliferation and a reduced capacity to kill cognate antigen-expressing tumor cells. Adenosine-exposed T cells displayed decreased induction of markers associated with activation at both the mRNA and protein level, including Ki67, ICOS, and PD-1, while showing higher expression of naïve-associated CD73. Interestingly, we also observed a third subset of markers which were uniquely induced by adenosine. In addition to its activity on T cells, adenosine signaling caused functional suppression of NK cells in the presence of target Yac-1 cells. In the myeloid compartment, NECA, a stable analog of adenosine, prevented in vitro differentiation of CD103+ cross-presenting dendritic cells, a population which provides a critical stimulus to tumor-infiltrating T cells. Additionally, NECA impaired macrophage expression of CD80, a canonical M1 marker and mediator of T cell costimulation, in both the presence and absence of M1-polarizing IFNγ and LPS. Finally, we showed that a novel, orally bioavailable CD73 inhibitor was able to effectively inhibit AMP to adenosine conversion both in vitro and in vivo, while an anti-CD73 antibody had incomplete effects. Taken together, an orally bioavailable small molecule inhibitor of CD73 represents a potential therapeutic approach to reverse immunosuppression within the tumor microenvironment. Citation Format: Todd C Metzger, Brian R Blank, Brenda Chan, Chelsea Chen, Yuping Chen, Xiaohui Du, Frank L Duong, Valeria R. Fantin, Lori S Friedman, Jared T Moore, Daqing Sun, Jessica Sun, Dena Sutimantanapi, Qiuping Ye, Natalie Yuen, Tatiana Zavorotinskaya. Intratumoral immunosuppression is reversed by blocking adenosine production with an oral inhibitor of CD73 [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr LB-A19. doi:10.1158/1535-7163.TARG-19-LB-A19

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