This project will address the therapy of mesothelin expressing tumors by developing and testing engineered? T cells with potent antitumor cytotoxicity. Mesothelin is a tumor-associated antigen that is frequently over? expressed on mesothelioma, non-small cell lung cancer, pancreatic and ovarian cancers. The strategy to be? used is the """"""""T-body"""""""" approach, which employs genetically reprogrammed, patient-derived lymphocytes? transfected with a novel chimeric receptor that contains combinations of the signal transduction domains of? 4-1BB (CD137), CD28, and CD3zeta as well as anti-mesothelin scFv (anti-meso-CD28-41BB-zeta). The central? hypothesis to be tested is that previous trials of adoptive therapy for cancer have used insufficient numbers? of cytotoxic T lymphocytes (CTL) that have shown inadequate engraftment, persistence and effector? function to self antigens. Presently, we are the only laboratory in the world that is actively testing lentiviral? modified T cells in the clinic, and in that trial we have demonstrated safety and prolonged lentiviral gene? transfer. The following three specific aims will test the hypothesis that engineered human T cells expressing? an anti-mesothelin-CD28-41BB-zeta chimeric receptor will have potent antitumor activity in vitro and in vivo by:? (1) developing and optimizing the anti-meso scFv vector. The avidity and the cytosolic signaling modules will? be optimized to obtain highly efficient lentiviral vectors that retarget T cells to specifically kill tumor cells that? express mesothelin at low effector to target ratios in vitro; (2) carrying out in vitro experiments to optimize? the effector functions of anti-mesothelin scFv CD28-41BB-zeta T bodies. Experiments will determine optimal? conditions for redirected T cell serial killing, cytokine production and proliferation, and compare this to? natural MHC restricted CTLs; and (3) performing in vivo experiments in immunodeficient NOD/SCID/beta2null? mice xenografted with human tumors that express mesothelin. These experiments will test the hypothesis? that vectors with high affinity scFv receptors and 4-1BB and CD28 signaling modules will have the most? potent anti-tumor effects. Finally, the engraftment, persistence and antitumor effects of chimeric T cells? given by intravenous and intraperitoneal routes will be compared using bioluminescence imaging. In? summary, an outstanding team of basic and translational scientists has been assembled that will develop? and test a universal T cell receptor to target some of the most common and drug resistant tumors.? Lay Description. A common reason for failure of immunotherapy of epithelial tumors is that the? immune system does not generate sufficient numbers of T cells to eradicate the tumor cells. It is now? possible to use lentiviral vector technology to engineer T cells with potent and specific antitumor effects.? This project will evaluate engineered T cells that target mesothelin that is overexpressed on both uncommon? tumors such as mesothelioma and a variety of commonly lethal tumors including pancreatic, ovarian and? non-small cell lung carcinoma.?

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
5P01CA066726-12
Application #
7664442
Study Section
Special Emphasis Panel (ZCA1)
Project Start
Project End
Budget Start
2008-08-01
Budget End
2009-07-31
Support Year
12
Fiscal Year
2008
Total Cost
$169,269
Indirect Cost
Name
University of Pennsylvania
Department
Type
DUNS #
042250712
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
Aggarwal, Charu; Haas, Andrew R; Metzger, Susan et al. (2018) Phase I Study of Intrapleural Gene-Mediated Cytotoxic Immunotherapy in Patients with Malignant Pleural Effusion. Mol Ther 26:1198-1205
Moon, Edmund K; Wang, Liang-Chuan S; Bekdache, Kheng et al. (2018) Intra-tumoral delivery of CXCL11 via a vaccinia virus, but not by modified T cells, enhances the efficacy of adoptive T cell therapy and vaccines. Oncoimmunology 7:e1395997
Klampatsa, Astero; Haas, Andrew R; Moon, Edmund K et al. (2017) Chimeric Antigen Receptor (CAR) T Cell Therapy for Malignant Pleural Mesothelioma (MPM). Cancers (Basel) 9:
Moon, Edmund K; Ranganathan, Raghuveer; Eruslanov, Evgeniy et al. (2016) Blockade of Programmed Death 1 Augments the Ability of Human T Cells Engineered to Target NY-ESO-1 to Control Tumor Growth after Adoptive Transfer. Clin Cancer Res 22:436-47
Liu, X; Barrett, D M; Jiang, S et al. (2016) Improved anti-leukemia activities of adoptively transferred T cells expressing bispecific T-cell engager in mice. Blood Cancer J 6:e430
Liu, Xiaojun; Ranganathan, Raghuveer; Jiang, Shuguang et al. (2016) A Chimeric Switch-Receptor Targeting PD1 Augments the Efficacy of Second-Generation CAR T Cells in Advanced Solid Tumors. Cancer Res 76:1578-90
O'Hara, Mark; Stashwick, Caitlin; Haas, Andrew R et al. (2016) Mesothelin as a target for chimeric antigen receptor-modified T cells as anticancer therapy. Immunotherapy 8:449-60
Sterman, Daniel H; Alley, Evan; Stevenson, James P et al. (2016) Pilot and Feasibility Trial Evaluating Immuno-Gene Therapy of Malignant Mesothelioma Using Intrapleural Delivery of Adenovirus-IFN? Combined with Chemotherapy. Clin Cancer Res 22:3791-800
Newick, Kheng; O'Brien, Shaun; Sun, Jing et al. (2016) Augmentation of CAR T-cell Trafficking and Antitumor Efficacy by Blocking Protein Kinase A Localization. Cancer Immunol Res 4:541-51
Andy, Uduak U; Harvie, Heidi S; Smith, Ariana L et al. (2015) Validation of a self-administered instrument to measure adherence to anticholinergic drugs in women with overactive bladder. Neurourol Urodyn 34:424-8

Showing the most recent 10 out of 85 publications