Blockade of co-inhibitory ?checkpoint? molecules, PD-1 and CTLA-4, has induced impressive clinical responses in advanced tumors; yet only in a subset of patients. Limited success with checkpoint blockade therapy suggests other cell extrinsic or intrinsic mechanisms may be dampening an effective immune response. Cytotoxic CD8+ T cells (CTL) encountering chronic antigen and environmental stressors (e.g. hypoxia, nutrient deprivation) can be driven to a terminally differentiated state marked by hyporesponsiveness and epigenetic, transcriptional, and metabolic dysfunction; a cell fate known as ?exhaustion?. However, it remains unclear whether terminally exhausted CTL (Texh) are simply non-functional or instead possess tolerogenic or suppressive properties. Our lab has conducted transcriptional profiling of tumor-infiltrating CD8+PD-1int (progenitor exhausted) CTL versus CD8+PD-1hiTim-3+ Texh, revealing that exhausted cells express a pattern of genes associated with immune regulation. Among these is the ectonucleosidase, CD39, responsible for hydrolyzation of extracellular ATP (eATP) to AMP. eATP is released upon T cell activation or cell death and induces proinflammatory cascades in numerous cell types. Because depletion of eATP can limit T cell activation, CD39 expression on Texh is likely to directly influence effector functions and tolerance in the tumor microenvironment. Indeed, when sorted directly from tumor, Texh, but not progenitor exhausted CTL, induce marked suppression of T cell effector responses. Strong preliminary in vitro data has revealed a HIF1a-dependent mechanism for CD39 expression in CTL. In cancer patients, disruption of tumor hypoxia through metformin treatment or VEGFR inhibition improves response to immunotherapy. Thus, we hypothesize that tumor hypoxia-driven CD39 expression on Texh represents a major barrier to the anti-tumor immune response. Our data support a model that as CTL progress to terminal exhaustion, hypoxic exposure upregulates CD39, providing Texh a mechanism to suppress proinflammatory processes. These findings suggest that Texh are deleterious to anti-tumor immunity and may need to be drastically reprogrammed or deleted in order to alleviate the immunosuppressive tumor microenvironment.

Public Health Relevance

The most impactful immunotherapeutic strategies in cancer have targeted inhibitory pathways that prevent tumor killing by inflammatory immune cells; yet only a subset of patients treated progress to clinical remission. A major barrier to successful tumor clearance following immunotherapy is T cell exhaustion, a dysfunctional state marked by decreased responsiveness and metabolic instability. Our work highlights a suppressor role of exhausted T cells, directly regulating anti-tumor immune functions and potentially restricting successful tumor clearance.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Individual Predoctoral NRSA for M.D./Ph.D. Fellowships (ADAMHA) (F30)
Project #
5F30CA247034-02
Application #
10076548
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Bian, Yansong
Project Start
2019-12-06
Project End
2023-12-05
Budget Start
2020-12-06
Budget End
2021-12-05
Support Year
2
Fiscal Year
2021
Total Cost
Indirect Cost
Name
University of Pittsburgh
Department
Microbiology/Immun/Virology
Type
Schools of Medicine
DUNS #
004514360
City
Pittsburgh
State
PA
Country
United States
Zip Code
15213