There is an unmet medical need for the therapy of leukemia and lymphoma of B cell origin as most adult patients remain incurable. The most potent antitumor effect mediated by the immune system occurs in the setting of allogeneic stem cell transplantation (HSCT). However, most patients are not eligible for this therapy. The objective of this project is to engineer autologous lymphocytes to endow them with the features that would render them equally powerful as allogeneic lymphocytes, and therefore avoid the toxicity and make the therapy routinely available to patients who relapse after initial therapy. Based on recent pilot results in patients with chronic lymphocytic leukemia (CLL), our long term goal is to develop a novel therapy using cell transfer therapy of engineered T cells expressing chimeric antigen receptors (CARs) that bind CD19 for the treatment of B cell malignancies. The key questions currently facing the field are how to: (1) enhance the potency and specificity of autologous T cells so that HSCT can be avoided, and (2) develop mechanisms to increase the persistence of CAR T cells so that they are resistant to the immunosuppressive tumor microenvironment. CARs are an attractive approach to address these issues, because they are off the shelf and HLAindependent. However, in order to establish clinical proof of concept that will be required to justify randomized clinical trials, it will be necessary to optimize vector design, as seemingly small changes in CAR design can have major effects on the antitumor potency of the engineered T cells. Prior preclinical and clinical studies have shown safety of the CAR approach, but efficacy has been disappointing, in part due to poor in vivo survival of CAR expressing cells. Since these early studies utilized CARs containing only a 1st generation single CD3ζ signaling chain, our hypothesis, based on strong pre-clinical data and early phase I clinical results is that our 2nd generation CARs that harbor additional costimulatory domains will augment survival and/or function of CAR-expressing cells in lymphodepleted patients.
Our specific aims are to conduct a phase IIa study to: (1) Test the safety, feasibility and tolerability of 3rd generation anti-CD19 CARs in patients with advanced chronic lymphocytic leukemia; 2) Determine the persistence and function of the CAR T cells in patients by measuring the engraftment and trafficking of CAR T cells to the tumor microenvironment, and the duration of B cell aplasia as a proof of mechanism. Other studies will determine safety of the lentiviral vector. Together, these studies will test the central hypothesis that costimulatory signaling domains will provide a selective survival advantage to CAR T cells in patient with CLL, providing proof of mechanism for this new approach. Our approach addresses a clear unmet medical need, as the only curative approach for most patients with relapsed leukemia is HSCT; our CARs could conceivably replace the allogeneic HSCT with an autologous CAR approach, and thereby decrease toxicity and expense associated with transplantation. Finally, the scientific principles evaluated are broadly applicable to other cancer therapies.

Public Health Relevance

In this project a form of personalized medicine using a customized immune system will be tested for patients with advanced chronic lymphocytic leukemia: the patient is first treated with FDA approved chemotherapy and then infused with patient T lymphocytes that have been altered in the laboratory to have redirected specificity for a molecule on the surface of the tumor cells, a process that endows the T cells with the capability to kill tumor cells. The results in a pilot phase I trial indicate that the redirected T cells have potent antitumor effects in patients. The studies in this project will establish further the safety and feasibility of the procedure, and will provide essential information on the mechanism of action of this promising new therapy in a phase IIa clinical trial in patients with advanced chronic lymphocytic leukemia.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA165206-04
Application #
8787677
Study Section
Clinical Oncology Study Section (CONC)
Program Officer
Merritt, William D
Project Start
2012-03-12
Project End
2016-12-31
Budget Start
2015-01-01
Budget End
2015-12-31
Support Year
4
Fiscal Year
2015
Total Cost
$454,032
Indirect Cost
$170,262
Name
University of Pennsylvania
Department
Pathology
Type
Schools of Medicine
DUNS #
042250712
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
Fraietta, Joseph A; Nobles, Christopher L; Sammons, Morgan A et al. (2018) Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature 558:307-312
June, Carl H; O'Connor, Roddy S; Kawalekar, Omkar U et al. (2018) CAR T cell immunotherapy for human cancer. Science 359:1361-1365
Fraietta, Joseph A; Lacey, Simon F; Orlando, Elena J et al. (2018) Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med 24:563-571
Porter, David; Frey, Noelle; Wood, Patricia A et al. (2018) Grading of cytokine release syndrome associated with the CAR T cell therapy tisagenlecleucel. J Hematol Oncol 11:35
Fesnak, Andrew D; Hanley, Patrick J; Levine, Bruce L (2017) Considerations in T Cell Therapy Product Development for B Cell Leukemia and Lymphoma Immunotherapy. Curr Hematol Malig Rep 12:335-343
Schuster, Stephen J; Svoboda, Jakub; Chong, Elise A et al. (2017) Chimeric Antigen Receptor T Cells in Refractory B-Cell Lymphomas. N Engl J Med 377:2545-2554
Fitzgerald, Julie C; Weiss, Scott L; Maude, Shannon L et al. (2017) Cytokine Release Syndrome After Chimeric Antigen Receptor T Cell Therapy for Acute Lymphoblastic Leukemia. Crit Care Med 45:e124-e131
Bhoj, Vijay G; Arhontoulis, Dimitrios; Wertheim, Gerald et al. (2016) Persistence of long-lived plasma cells and humoral immunity in individuals responding to CD19-directed CAR T-cell therapy. Blood 128:360-70
Chen, Fang; Teachey, David T; Pequignot, Edward et al. (2016) Measuring IL-6 and sIL-6R in serum from patients treated with tocilizumab and/or siltuximab following CAR T cell therapy. J Immunol Methods 434:1-8
Fesnak, Andrew D; June, Carl H; Levine, Bruce L (2016) Engineered T cells: the promise and challenges of cancer immunotherapy. Nat Rev Cancer 16:566-81

Showing the most recent 10 out of 22 publications