Adoptive T cell immunotherapy strives to harness the power and specificity of the immune system to cure cancer in human patients. This approach relies on the transfer of tumor-reactive CD8+ T cells into patients, but the anticipated clinical benefits have not been achieved due to the induction of T cell tolerance. Peripheral tolerance is a multifaceted process, characterized by compromised proliferation, impaired effector function, and deletion of autoreactive T cells. Efforts to overcome tolerance for improved immunotherapy have been hampered because the T cell intrinsic pathways that regulate whether tumor/self-reactive T cells are directed toward tolerance versus immunity have yet to be defined. Using a pre-clinical mouse model of T cell tolerance, we found that Listeria monocytogenes vaccination at the time of adoptive CD8+ T cell transfer prevents tolerance. Specifically, transferred tumor/self-reactive T cells acquired effector function, displayed increased in vivo persistence, and provided a survival benefit to tumor-bearing hosts receiving adoptive T cell immunotherapy. T cell rescue corresponded with expression of the transcription factors T-bet and Eomesodermin (Eomes), both of which failed to be induced under tolerizing conditions in the absence of vaccination. T-bet and Eomes have a defined role in dictating type-I (IFN-? mediated) immune responses. Aside from their role in promoting IFN-? expression, these transcription factors also influence expression of several other genes that encode effector molecules important for CD8+ T cell trafficking or control of infected or malignant cells, includin CXCR3, granzyme B, perforin, and FasL. Our data suggest that T-bet and Eomes may be fundamental to the decision of T cell immunity versus tolerance, and as such, we propose to decipher the role of these transcription factors in this important fate decision. To this end, we wll utilize mice with genetic modulation of T-bet and Eomes in peripheral tumor/self-reactive CD8+ T cells as tools to dissect the role of these transcription factors during induction of T cell tolerance and under conditions of vaccine-mediated T cell rescue.
Specific Aim I will focus on the individual and collective contributions of T-bet and Eomes to vaccine-mediated rescue of peripheral CD8+ T cell tolerance. We will evaluate effector cytokine production, T cell persistence, and transcript and protein production for T-bet or Eomes target genes in T cells rendered unable to upregulate T-bet, Eomes, or both;or T cells with constitutive T-bet expression.
Specific Aim II will then focus on characterizing the specific contributions of these molecules to providing anti-tumor immunity within a tolerizing environment. Survival of tumor-bearing hosts will be used as a readout for therapeutic efficacy of adoptive immunotherapy with CD8+ T cells rendered genetically unable to express T-bet, Eomes or both;or T cells with constitutive T-bet expression. Identifying the contributions of T-bet and Eomes to T cell dysfunction during CD8+ T cell tolerance and adoptive immunotherapy may lead to improvements in T cell-based therapies for patients with cancer.

Public Health Relevance

Adoptive T cell immunotherapy strives to cure cancer by transfer of large numbers of tumor-specific T cells into human patients, but the anticipated clinical benefits of this therapy have yet to be achieved due to impaired function or deletion of transferred T cells. This project will identify T cell intrinsic molecular pathways involved in the regulation of T cell dysfunction and define strategies to enhance T cell responses during adoptive immunotherapy. Results from these studies will provide new insight that may foster translation of improved immunotherapies for the benefit of patients with malignant disease.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Individual Predoctoral NRSA for M.D./Ph.D. Fellowships (ADAMHA) (F30)
Project #
5F30CA180375-02
Application #
8693610
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Damico, Mark W
Project Start
2013-07-01
Project End
2016-06-30
Budget Start
2014-07-01
Budget End
2015-06-30
Support Year
2
Fiscal Year
2014
Total Cost
Indirect Cost
Name
Saint Louis University
Department
Microbiology/Immun/Virology
Type
Schools of Medicine
DUNS #
City
Saint Louis
State
MO
Country
United States
Zip Code
63103
Berrien-Elliott, Melissa M; Yuan, Jinyun; Swier, Lauryn E et al. (2015) Checkpoint blockade immunotherapy relies on T-bet but not Eomes to induce effector function in tumor-infiltrating CD8+ T cells. Cancer Immunol Res 3:116-24
Jackson, Stephanie R; Yuan, Jinyun; Berrien-Elliott, Melissa M et al. (2014) Inflammation programs self-reactive CD8+ T cells to acquire T-box-mediated effector function but does not prevent deletional tolerance. J Leukoc Biol 96:397-410
Jackson, Stephanie R; Yuan, Jinyun; Teague, Ryan M (2014) Targeting CD8+ T-cell tolerance for cancer immunotherapy. Immunotherapy 6:833-52
Jackson, Stephanie R; Berrien-Elliott, Melissa; Yuan, Jinyun et al. (2014) Neuropilin-1 expression is induced on tolerant self-reactive CD8+ T cells but is dispensable for the tolerant phenotype. PLoS One 9:e110707