The long-term objective is to use animal models to find mechanisms and approaches whereby adoptively transferred T cells transduced with mutant neoantigen-specific TCRs can eradicate solid cancers in patients without destroying normal tissues. Tumors in patients have been present at least for months or years and are usually at least 1cm in diameter at time of diagnosis. Therefore, large and long-established solid tumors expressing autochthonous (untransfected) mutant neoantigens are the focus of this proposal. A key element being explored is the destruction of the immunosuppressive stromal tumor microenvironment by transfer of MHC Class II restricted neoantigen-specific TCR-transduced CD4+ T cells (CD4+TCR). These T cells cause tumor destruction followed by long-term arrest of tumor growth. A second key element being explored is that CD4+TCR T cells form 4-cell-type clusters with MHC Class I restricted neoantigen-specific TCR-transduced CD8+ T cells (CD8+TCR) which are attracted to the cluster by recognizing the cross-presented neoantigen on stromal CD11b+ tumor-associated macrophages. Finally, intraclonal non-heritable heterogeneity is explored as novel mechanism of escape of cancers despite cancer cells seemingly lacking mutant neoantigen-negative variants.
Aim 1 is to determine the requirements and mechanisms for CD4+TCR T cells to destroy, without CD8+ T cells, advanced solid tumors and subsequently arrest their growth. It will be tested whether targeting tumor stroma is essential for this CD4+TCR-mediated destruction/arrest and whether these effects are independent of direct cancer cell recognition. Furthermore, it will be examined whether therapeutic CD4+TCRs can be isolated from tumor-infiltrating lymphocytes (TILs) that are failing to reject the cancer. Finally, it will be tested whether appropriate lysosomal processing of mutant neoantigen is a decisive predictor for CD4+TCR- mediated tumor destruction.
Aim 2 is to determine whether 4-cell-type clusters at the effector phase are essential for cancer eradication by CD4+TCRs and CD8+TCRs. It will be analyzed whether direct cancer cell recognition by the CD8+TCR is required for cancer eradication. Furthermore, it will be tested whether, for synergistic cancer cell destruction, the two neoantigens must be released from the same cancer cell because this would allow the CD8+TCR T cells to recognize the cross-presented neoantigen on the same CD11b+ stromal cell also being recognized by the CD4+TCR T cells. Antigen-specific 4-cell-type cluster formation will be quantified and modeled in vitro using longitudinal time-lapse imaging. Finally, will be determined whether cancers brought into equilibrium by CD4+TCRs can still be eradicated by subsequent treatment with CD8+TCR transduced T cells.
Aim 3 is to determine mechanisms of escape and the molecular nature of CD8+TCR recognized antigens that can be used to eradicate cancers in synergy with CD4+TCRs. It will be examined whether cloned cancer cells seemingly lacking antigen-negative variants nevertheless escape by the presence of cancer cells with low mutant mRNA expression due to intraclonal heterogeneity.

Public Health Relevance

The long-term objective is to develop therapeutic approaches for mutation-specific TCRs to eradicate cancer in patients. The immediate objective is to utilize in vitro assays and animal models that mimic human cancers in order to maximize translational potential. Results obtained by experimentation in animals will suggest new approaches and provide a blueprint for making mutation-specific TCR treatment effective in humans with advanced tumors.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
2R01CA022677-39
Application #
10051921
Study Section
Transplantation, Tolerance, and Tumor Immunology Study Section (TTT)
Program Officer
Mccarthy, Susan A
Project Start
1978-02-01
Project End
2025-06-30
Budget Start
2020-07-01
Budget End
2021-06-30
Support Year
39
Fiscal Year
2020
Total Cost
Indirect Cost
Name
University of Chicago
Department
Pathology
Type
Schools of Medicine
DUNS #
005421136
City
Chicago
State
IL
Country
United States
Zip Code
60637
Arina, Ainhoa; Karrison, Theodore; Galka, Eva et al. (2017) Transfer of Allogeneic CD4+ T Cells Rescues CD8+ T Cells in Anti-PD-L1-Resistant Tumors Leading to Tumor Eradication. Cancer Immunol Res 5:127-136
Kammertoens, Thomas; Friese, Christian; Arina, Ainhoa et al. (2017) Tumour ischaemia by interferon-? resembles physiological blood vessel regression. Nature 545:98-102
Posey Jr, Avery D; Schwab, Robert D; Boesteanu, Alina C et al. (2016) Engineered CAR T Cells Targeting the Cancer-Associated Tn-Glycoform of the Membrane Mucin MUC1 Control Adenocarcinoma. Immunity 44:1444-54
Arina, Ainhoa; Idel, Christian; Hyjek, Elizabeth M et al. (2016) Tumor-associated fibroblasts predominantly come from local and not circulating precursors. Proc Natl Acad Sci U S A 113:7551-6
Leisegang, Matthias; Engels, Boris; Schreiber, Karin et al. (2016) Eradication of Large Solid Tumors by Gene Therapy with a T-Cell Receptor Targeting a Single Cancer-Specific Point Mutation. Clin Cancer Res 22:2734-43
Blankenstein, Thomas; Leisegang, Matthias; Uckert, Wolfgang et al. (2015) Targeting cancer-specific mutations by T cell receptor gene therapy. Curr Opin Immunol 33:112-9
Binder, David C; Schreiber, Hans (2014) Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors--letter. Cancer Res 74:632; discussion 635
Arina, Ainhoa; Schreiber, Karin; Binder, David C et al. (2014) Adoptively transferred immune T cells eradicate established tumors despite cancer-induced immune suppression. J Immunol 192:1286-93
Binder, David C; Engels, Boris; Arina, Ainhoa et al. (2013) Antigen-specific bacterial vaccine combined with anti-PD-L1 rescues dysfunctional endogenous T cells to reject long-established cancer. Cancer Immunol Res 1:123-33
Liu, Rebecca Berlant; Engels, Boris; Schreiber, Karin et al. (2013) IL-15 in tumor microenvironment causes rejection of large established tumors by T cells in a noncognate T cell receptor-dependent manner. Proc Natl Acad Sci U S A 110:8158-63

Showing the most recent 10 out of 75 publications