In the previous project period of this grant, we investigated the transcriptional mechanisms of CD8+ T cell ?exhaustion?, a hyporesponsive state observed under conditions of sustained antigen stimulation in cancer and chronic viral infections. Exhausted T cells show decreased proliferation and cytokine production, and upregu- late inhibitory cell surface receptors including CTLA4, PD-1, LAG3 and TIM3. Antibodies to these receptors reverse T cell exhaustion, and their administration to cancer patients forms the basis for ?immune checkpoint blockade?, a strategy that has recently been remarkably successful in cancer immunotherapy. Combinations of antibodies to inhibitory receptors show greater efficacy than administration of individual antibodies alone, consistent with the fact that exhausted T cells typically express several inhibitory receptors. Thus understanding the molecular mechanisms that lead to inhibitory receptor expression and T cell ?exhaustion/ dysfunction? would complement and enhance the effects of therapies that target combinations of individual inhibitory receptors. We previously showed that the transcriptional programs of anergy and exhaustion are initiated by the transcription factor NFAT, acting in the absence of its partner AP-1 (Fos-Jun). In the course of these studies, we developed an engineered NFAT, CA-RIT-NFAT1, which induces the characteristic features of exhaustion when transduced into CD8+ T cells. To understand the biological implications of this hyporesponsive program, we used mouse models of anti-tumor responses that involving adoptive transfer of tumor-reactive or unreactive TCR- transgenic T cells or T cells bearing chimeric antigen receptors (CAR-T cells). Using these models, we identified Nr4a transcription factors, and other families of transcription factors, as ?exhaustion-related? target of NFAT. We further showed that tumor-infiltrating CD8+ T cells lacking all three Nr4a family members displayed a gene expression profile similar to that of activated T cells and rejected tumors more efficiently than control CD8+ T cells. In this application we will test the hypothesis that mouse and human CD4+ and CD8+ tumor-infiltrating T cells (TILs) are functionally silenced by a cell-intrinsic transcriptional program mediated, at least in part, by NFAT and Nr4a transcription factors, and other transcription factors induced by NFAT.
In Aim 1, we will define mechanistically the cell-intrinsic roles of Nr4a transcription factors in CD8+ T cell exhaustion. We will ask how Nr4a deletion in TILs overcomes exhaustion, enhances the effector phenotype and promotes tumor regression.
In Aim 2, we will examine the roles of other pertinent transcription factors in CD8+ T cell exhaustion, and define the kinetics with which the exhaustion program unfolds.
In Aim 3, we will examine the roles of NFAT and Nr4a in primary human T cells bearing exhaustion markers or transduced with CA-RIT-NFAT1, and in TILs isolated from human tumors. Our proposed studies will contribute to a broad mechanistic understanding of the transcriptional mechanisms operating in tumor-infiltrating immune cells, and may spark improved immunotherapies for cancer patients.

Public Health Relevance

One of the hallmarks of cancer is its ability to evade the immune system. The immune cells responsible for eliminating tumor cells, known as CD8 T-cells, are supposed to function by entering into tumors and destroying them; in many cases, however, they enter a hyporesponsive state known as ?exhaustion? where they infiltrate the tumor normally but fail to kill the tumor cells. Recent strategies for cancer immunotherapy are aimed at overcoming exhaustion so that the tumor-infiltrating CD8 T cells can effectively eliminate cancers. In this application we investigate two potential strategies for overcoming CD8 T cell exhaustion in tumor-infiltrating cells, and compare the features of exhausted T cells in humans and mice.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
2R01AI040127-26A1
Application #
9670689
Study Section
Cellular and Molecular Immunology - A Study Section (CMIA)
Program Officer
Lapham, Cheryl K
Project Start
1991-08-01
Project End
2023-08-31
Budget Start
2018-09-24
Budget End
2019-08-31
Support Year
26
Fiscal Year
2018
Total Cost
Indirect Cost
Name
La Jolla Institute
Department
Type
DUNS #
603880287
City
La Jolla
State
CA
Country
United States
Zip Code
92037
Hirve, Nupura; Rajanikanth, Vangipurapu; Hogan, Patrick G et al. (2018) Coiled-Coil Formation Conveys a STIM1 Signal from ER Lumen to Cytoplasm. Cell Rep 22:72-83
Scott-Browne, James P; Lio, Chan-Wang J; Rao, Anjana (2017) TET proteins in natural and induced differentiation. Curr Opin Genet Dev 46:202-208
Märklin, Melanie; Heitmann, Jonas S; Fuchs, Alexander R et al. (2017) NFAT2 is a critical regulator of the anergic phenotype in chronic lymphocytic leukaemia. Nat Commun 8:755
Spira, Avrum; Yurgelun, Matthew B; Alexandrov, Ludmil et al. (2017) Precancer Atlas to Drive Precision Prevention Trials. Cancer Res 77:1510-1541
Moffett, Howell F; Cartwright, Adam N R; Kim, Hye-Jung et al. (2017) The microRNA miR-31 inhibits CD8+ T cell function in chronic viral infection. Nat Immunol 18:791-799
Pereira, Renata M; Hogan, Patrick G; Rao, Anjana et al. (2017) Transcriptional and epigenetic regulation of T cell hyporesponsiveness. J Leukoc Biol 102:601-615
Zang, Shengbing; Li, Jia; Yang, Haiyan et al. (2017) Mutations in 5-methylcytosine oxidase TET2 and RhoA cooperatively disrupt T cell homeostasis. J Clin Invest 127:2998-3012
Yu, Bingfei; Zhang, Kai; Milner, J Justin et al. (2017) Epigenetic landscapes reveal transcription factors that regulate CD8+ T cell differentiation. Nat Immunol 18:573-582
Hogan, Patrick G (2017) Calcium-NFAT transcriptional signalling in T cell activation and T cell exhaustion. Cell Calcium 63:66-69
Mognol, Giuliana P; Spreafico, Roberto; Wong, Victor et al. (2017) Exhaustion-associated regulatory regions in CD8+ tumor-infiltrating T cells. Proc Natl Acad Sci U S A 114:E2776-E2785

Showing the most recent 10 out of 75 publications