Tumor immunotherapy using EBV-specific cytotoxic T lymphocytes (CTLs) has been successful for the treatment of EBV-associated lymphomas arising in the immunocompetent host. However, the majority of referred patients have EBV-negative lymphomas, precluding the use of virus-targeted T cells;also, CTLs do not eliminate tumors in all patients, likely because potentially immunogenic tumors arising in immunocompetent persons develop immune evasion strategies that subvert the effector function of the infused CTLs. These include downregulation of viral antigens, and the production of soluble immunomodulatory cytokines, such as 1L4, from malignant and infiltrating inhibitory cells, which limit the in vivo activity of adoptively transferred T cells. To extend T cell therapy to all patients with Hodgkin lymphoma (HL), regardless of tumor EBV status, we used the previous SPORE funding cycle to develop a strategy to reactivate and expand CTLs with specificity for the nonviral tumor-associated antigens (TAAs) Survivin SSX2, MAGE-A4, NY-ESO-1 and FRAME expressed by malignant cells. We reasoned that targeting multiple TAAs simultaneously would minimize tumor immune escape and therefore are assessing the safety and efficacy of these multiTAA-CTLs in vivo. We now propose, in Project 1, to address two remaining barriers to successful T cell therapy, namely (a) low target antigen expression, and (b) the immunosuppressive tumor microenvironment. Thus, our central hypotheses are that the efficacy ot immunotherapy for HL can be enhanced by combination with epigenetic drugs and by modifying infused T cells to retain Thi function in the Th2 cytokine milieu of the tumor. Hence, in Aim 1 of our renewal application, in a phase I clinical trial, we will generate and infuse multiTAA-targeted CTLs in combination with a hypomethylating agent known to upregulate T cell target antigen expression on malignant cells and sensitize them to immune-mediated destruction. The impact of this combination therapy on tumor immunogenicity, T cell antitumor activity and epitope spreading will be assessed in Aim 2. Finally, Aim 3 seeks to protect adoptively transferred multiTAA-targeted CTLs from the tumor microenvironment by engineering the T cells to express a chimeric cytokine receptor that can bind the Th2 cytokine 1L4 and convert its inhibitory signal to one that is activating. Although restricted to preclinical models initially.
Aim 3 will be clinically developed in the future. In summary, this approach combining drug therapy with tumorspecific CTLs may enable T cell immunotherapy to become more broadly applied to all patients with cancer.

Public Health Relevance

We have developed a personalized medicine to treat Hodgkin's lymphoma (HL) by taking immune cells (T cells) from patient blood, and training them to kill HL cells. These cells will then be administered with a drug which improves their ability to recognize the tumor. Finally we will protect the T cells from the inhibitory tumor environment by modifying them with a molecule that delivers a potent activating signal at the tumor site. We believe that this novel approach will be safe and effective for HL and ultimately for other cancers.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Specialized Center (P50)
Project #
2P50CA126752-06
Application #
8339657
Study Section
Special Emphasis Panel (ZCA1)
Project Start
Project End
Budget Start
2012-09-01
Budget End
2013-08-31
Support Year
6
Fiscal Year
2012
Total Cost
$248,378
Indirect Cost
$72,324
Name
Baylor College of Medicine
Department
Type
DUNS #
051113330
City
Houston
State
TX
Country
United States
Zip Code
77030
Brunetti, Lorenzo; Gundry, Michael C; Kitano, Ayumi et al. (2018) Highly Efficient Gene Disruption of Murine and Human Hematopoietic Progenitor Cells by CRISPR/Cas9. J Vis Exp :
Xiong, Wei; Chen, Yuhui; Kang, Xi et al. (2018) Immunological Synapse Predicts Effectiveness of Chimeric Antigen Receptor Cells. Mol Ther 26:963-975
Heslop, Helen E; Brenner, Malcolm K (2018) Seek and You Will Not Find: Ending the Hunt for Replication-Competent Retroviruses during Human Gene Therapy. Mol Ther 26:1-2
Hogstad, Brandon; Berres, Marie-Luise; Chakraborty, Rikhia et al. (2018) RAF/MEK/extracellular signal-related kinase pathway suppresses dendritic cell migration and traps dendritic cells in Langerhans cell histiocytosis lesions. J Exp Med 215:319-336
Mamonkin, Maksim; Mukherjee, Malini; Srinivasan, Madhuwanti et al. (2018) Reversible Transgene Expression Reduces Fratricide and Permits 4-1BB Costimulation of CAR T Cells Directed to T-cell Malignancies. Cancer Immunol Res 6:47-58
Velasquez, Mireya Paulina; Bonifant, Challice L; Gottschalk, Stephen (2018) Redirecting T cells to hematological malignancies with bispecific antibodies. Blood 131:30-38
Kalra, Mamta; Gerdemann, Ulrike; Luu, Jessica D et al. (2018) Epstein-Barr Virus (EBV)-derived BARF1 encodes CD4- and CD8-restricted epitopes as targets for T-cell immunotherapy. Cytotherapy :
Ngai, Ho; Tian, Gengwen; Courtney, Amy N et al. (2018) IL-21 Selectively Protects CD62L+ NKT Cells and Enhances Their Effector Functions for Adoptive Immunotherapy. J Immunol 201:2141-2153
Morita, Daisuke; Nishio, Nobuhiro; Saito, Shoji et al. (2018) Enhanced Expression of Anti-CD19 Chimeric Antigen Receptor in piggyBac Transposon-Engineered T Cells. Mol Ther Methods Clin Dev 8:131-140
Bollard, Catherine M; Tripic, Tamara; Cruz, Conrad Russell et al. (2018) Tumor-Specific T-Cells Engineered to Overcome Tumor Immune Evasion Induce Clinical Responses in Patients With Relapsed Hodgkin Lymphoma. J Clin Oncol 36:1128-1139

Showing the most recent 10 out of 270 publications