Recent clinical trials conducted at the Surgery Branch of the National Cancer Institute (NCI) has have found that adoptive transfer of autologous tumor reactive T cells into patients preconditioned with nonmyeloablative chemotherapy leads to significant durable objective clinical responses. T cells used in these studies were TIL harvested from melanoma lesions. There are two key problems that have limited its use outside the NCI. First, many patients don't have suitable tumors TIL harvest. Second, it is difficult to isolate and expand tumor reactive T cells to therapeutic numbers for most patients. New approaches for adoptive T cell transfer are needed if this approach will make it beyond the experimental stage. We published the first report demonstrating that it was feasible to redirect the reactivity of normal PBL-derived T cells using retroviral vectors encoding TCR genes isolated from a MART-1 reactive T cell clone. This opened the possibility of providing any patient with a source of autologous tumor-reactive T cells capable of recognizing any antigen so long as a TCR was available which could recognize that antigen. This MART-1 reactive TCR was the first used to treat patients with TCR gene modified T cells demonstrating the feasibility and safety of using TCR gene modified T cells in humans. However, this trial and three others had fewer objective clinical response compared to TIL suggesting TCR gene modified T cells are less effective that TIL. Based on these trials, it is clear there are critical differences between these two cells types. Over the years, the field of TCR gene transfer has focused on TCR affinity, TCR pairing, and TCR expression. The biology of T cells engineered with viral vectors has largely been unexplored. We therefore hypothesize that TCR gene modified T cells are fundamentally different than normal T cells in how they respond to environmental factors. We further hypothesize that a better understanding of how TCR transduced T cells are impacted by lymphopenia, T cell help, activation-induced cell death (AICD), costimulation, and immune suppression will lead to better TCR transduced T cells for patient treatment. Based on these hypotheses, we developed this Program to improve the function of TCR gene modified T cells through a series of laboratory comparing TCR transduced T cells to normal T cells bearing the same TCR. When complimented with clinical trials, our highly integrated Program will advance the field of TCR gene transfer leading to their improved therapeutic efficacy not only for patients with melanoma but for patients with other malignancies and chronic viral infections.

Public Health Relevance

The goal of this integrated and collaborative Program is to gain a better understanding of the biology of TCR gene modified T cells to enhance their therapeutic efficacy in vivo. Projects in this Program will study how lymphopenia, antigen stimulation, T cell help, cosfimulafion, and suppression influence the persistence and function of adoptively transferred TCR gene modified T cells. These studies will be conducted in mouse melanoma models and clinical trials in melanoma patients.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
5P01CA154778-05
Application #
8929161
Study Section
Special Emphasis Panel (ZCA1-RPRB-J (M1))
Program Officer
Song, Min-Kyung H
Project Start
2011-09-21
Project End
2016-08-31
Budget Start
2015-09-01
Budget End
2016-08-31
Support Year
5
Fiscal Year
2015
Total Cost
$3,406,082
Indirect Cost
$653,248
Name
Loyola University Chicago
Department
Surgery
Type
Schools of Medicine
DUNS #
791277940
City
Maywood
State
IL
Country
United States
Zip Code
60153
Spear, Timothy T; Wang, Yuan; Smith Jr, Thomas W et al. (2018) Altered Peptide Ligands Impact the Diversity of Polyfunctional Phenotypes in T Cell Receptor Gene-Modified T Cells. Mol Ther 26:996-1007
Wrangle, John M; Velcheti, Vamsidhar; Patel, Manish R et al. (2018) ALT-803, an IL-15 superagonist, in combination with nivolumab in patients with metastatic non-small cell lung cancer: a non-randomised, open-label, phase 1b trial. Lancet Oncol 19:694-704
Spear, Timothy T; Foley, Kendra C; Garrett-Mayer, Elizabeth et al. (2018) TCR modifications that enhance chain pairing in gene-modified T cells can augment cross-reactivity and alleviate CD8 dependence. J Leukoc Biol 103:973-983
Riley, Timothy P; Hellman, Lance M; Gee, Marvin H et al. (2018) T cell receptor cross-reactivity expanded by dramatic peptide-MHC adaptability. Nat Chem Biol 14:934-942
Nelson, Alexander; Cunha, Christina; Nishimura, Michael I et al. (2018) Activated human Foxp3+ regulatory T cells produce membrane-bound TNF. Cytokine 111:454-459
Knochelmann, Hannah M; Smith, Aubrey S; Dwyer, Connor J et al. (2018) CAR T Cells in Solid Tumors: Blueprints for Building Effective Therapies. Front Immunol 9:1740
Wrangle, John M; Patterson, Alicia; Johnson, C Bryce et al. (2018) IL-2 and Beyond in Cancer Immunotherapy. J Interferon Cytokine Res 38:45-68
Scheffel, Matthew J; Scurti, Gina; Wyatt, Megan M et al. (2018) N-acetyl cysteine protects anti-melanoma cytotoxic T cells from exhaustion induced by rapid expansion via the downmodulation of Foxo1 in an Akt-dependent manner. Cancer Immunol Immunother 67:691-702
Chiuzan, Cody; Garrett-Mayer, Elizabeth; Nishimura, Michael (2018) An adaptive dose-finding design based on both safety and immunologic responses in cancer clinical trials. Stat Biopharm Res 10:185-195
Chatterjee, Shilpak; Daenthanasanmak, Anusara; Chakraborty, Paramita et al. (2018) CD38-NAD+Axis Regulates Immunotherapeutic Anti-Tumor T Cell Response. Cell Metab 27:85-100.e8

Showing the most recent 10 out of 50 publications