The Preclinical Development and Clinical Monitoring Facility (PDCMF) of the Experimental Transplantation and Immunology Branch supports the development and implementation of new protocols involving adoptive immune cell therapies through preclinical development, translational implementation of clinical products and preservation and analysis of patient blood and tissues during clinical trials. PDCMF staff, working within the Cell Processing Service of the Department of Transfusion Medicine, have documented patient data and aliquots of cells for RCR/RCL testing for James Kochenderfer's Chimeric Antigen Receptor (CAR) therapies (protocols 10-C-0054, 14-C-0168, 16-C-0054, 17-C-0048), in fulfillment of FDA requirements. PDCMF staff have also helped to optimize the generation of cell products (Jin, et al J Transl Med 2018) and to prepare the IND application and preclinical validation to bring new cell products to clinical trial under GMP conditions. Notable contributions in 2018 include: 1) isolation of naive CD8 T cells and the cell culture conditions, including comparisons of TWS119 and CHR99021, necessary to produce Tscm to generate CAR products for the amended 10-C-0054; 2) trial runs and optimization of the Miltenyi Prodigy for the Cell Processing Service; 3) validation of reagents for a new anti-BCMA fully-human variable heavy chain (FHVH) CAR product (18-C-0125, PI: James Kochenderfer). As per our role in ZIC BC 010934, the PDCMF processes and preserves peripheral blood, marrow aspirates, and tumor biopsies, as well as the research aliquots of the starting and final cell products for this Project. We collaborate with the Cell Processing Service of DTM, the ETIB Flow Cytometry Facility, and ETIB T Cell Facility, and we routinely ship specimens to extramural labs for RCR/RCL testing and for a multicenter CRADA trial (16-C-0025; PI: James Kochenderfer). Our further contribution to the protocols can be distinguished on the basis of their principal investigators, James Kochenderfer and Christian Hinrichs. James Kochenderfer's clinical trials can be subdivided by the target molecules of their CAR constructs: CD30 (17-C-0048), CD19 (10-C-0054, 16-C-0054), and BCMA (14-C-0168 and upcoming 18-C-0125). While 17-C-0048 and 10-C-0054 are actively accruing patients and 16-C-0054 reports are under preparation, results from 14-C-0168 have been published (Brudno, et al JCO 2018), wherein we characterized the CAR+ cells in the infusion product and at their peak level in the blood. We incorporated several activation, differentiation, and exhaustion markers into a multiparameter flow cytometry panel and showed a decreased ratio of CD4:CD8 within CAR+ T cells, post-infusion. Furthermore, many circulating CAR+ CD8+ T cells appeared terminally differentiated. These findings may spur future studies regarding the ramifications for clinical response, persistence of the cells, and re-treatment or co-treatment options. Dr. Christian Hinrichs has initiated therapeutic trials for treatment of viral-induced tumors, either by checkpoint inhibitors of the PD-1/PD-L1 system (16-C-0160 and, in collaboration, 17-C-0125, PI: Mark Roschewski) or adoptive immunotherapy utilizing expanded T cells transduced to express a transgenic T cell receptor (16-C-0154 and 17-C-0116). The PDCMF has especially supported these protocols by the collection of tumor biopsies, in coordination with the ETIB T Cell Facility and with Dr. Hinrichs's researchers. The biopsies are sectioned and apportioned for downstream applications, such as viral genotyping, RNA-based analysis, and immunohistochemistry, with each requiring its own processing and storage conditions.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Scientific Cores Intramural Research (ZIC)
Project #
1ZICBC011029-11
Application #
9780225
Study Section
Project Start
Project End
Budget Start
Budget End
Support Year
11
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Basic Sciences
Department
Type
DUNS #
City
State
Country
Zip Code
Brudno, Jennifer N; Maric, Irina; Hartman, Steven D et al. (2018) T Cells Genetically Modified to Express an Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor Cause Remissions of Poor-Prognosis Relapsed Multiple Myeloma. J Clin Oncol 36:2267-2280
Ali, Syed Abbas; Shi, Victoria; Maric, Irina et al. (2016) T cells expressing an anti-B-cell-maturation-antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood :
Amarnath, Shoba; Foley, Jason E; Farthing, Don E et al. (2015) Bone marrow-derived mesenchymal stromal cells harness purinergenic signaling to tolerize human Th1 cells in vivo. Stem Cells 33:1200-12
Mossoba, Miriam E; Halverson, David C; Kurlander, Roger et al. (2015) High-Dose Sirolimus and Immune-Selective Pentostatin plus Cyclophosphamide Conditioning Yields Stable Mixed Chimerism and Insufficient Graft-versus-Tumor Responses. Clin Cancer Res 21:4312-20
Kochenderfer, James N; Dudley, Mark E; Carpenter, Robert O et al. (2013) Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood 122:4129-39
Carpenter, Robert O; Evbuomwan, Moses O; Pittaluga, Stefania et al. (2013) B-cell maturation antigen is a promising target for adoptive T-cell therapy of multiple myeloma. Clin Cancer Res 19:2048-60
Fowler, Daniel H; Mossoba, Miriam E; Steinberg, Seth M et al. (2013) Phase 2 clinical trial of rapamycin-resistant donor CD4+ Th2/Th1 (T-Rapa) cells after low-intensity allogeneic hematopoietic cell transplantation. Blood 121:2864-74
Hardy, Nancy M; Fellowes, Vicki; Rose, Jeremy J et al. (2012) Costimulated tumor-infiltrating lymphocytes are a feasible and safe alternative donor cell therapy for relapse after allogeneic stem cell transplantation. Blood 119:2956-9