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 T cell therapies through preclinical development, translational implementation of clinical products and preservation and analysis of patient blood and tissues during clinical trials. Five novel protocols involving adoptive transfer of T cells have been implemented in recent years as a result of this process. (1) In 04-C-0055 Arm 4A, Daniel Fowler initially utilized expanded donor-derived CD4 helper cells grown for 12 days in IL-4 and rapamycin (T.Rapa.12) to enhance donor engraftment and reduce GVHD. The Preclinical Service supported implementation of clinical trial Arms 4B, utilizing a shorter 6 day expansion period to generate cells with increased anti-tumor potency (T.Rapa.6) and Arm 4C to implement an altered schedule of post transplant rapamycin. Following completion of each trial arm, we have characterized the early immune reconstitution following infusion of the T.Rapa product and have assessed the T cell receptor repertoire diversity of the infused T.Rapa cells and the transplant recipients at day 60 through spectratyping. We have assessed lymphokine production capacity post transplant to evaluate the extent and durability of the cytokine shift resulting from the infusion of Th2-rapa cells These studies have been reported in 2013 (Fowler et al, Blood, 2013). (2) The second novel product involves expansion of autologous T cells (both CD4 and CD8) in the presence of IFNalpha and rapamycin (T1.rapa), to generate a cell product that potentially has activity against residual myeloma plasma cells. In protocol 11-C-0016 (P.I. Claude Sportes/Daniel Fowler), patients with undergo an autologous transplant followed by infusion of T1.Rapa cells;through expanding the scale of product manufacturing, serial infusions of T1.Rapa cells are currently being implemented. Clinical production of these cells was developed through our staff in Cell Processing. Following initiation of this trial, we have monitored serial changes in lymphocyte populations in blood and in bone marrow and have provided plasma assays of inflammatory cytokines to monitor patient responses at each dose escalation. (3) We have supported the implementation of the first trial of the use of donor-derived anti-CD19 Chimeric Antigen Receptor (CAR) T cells in patients with relapsed or persistent lymphoma following allogeneic transplant (Protocol 10-C-0052, P.I. James Kochenderfer). We have tracked the presence of CAR T cells following adoptive transfer and have characterized expression of markers of activation and anergy. These studies have demonstrated the expansion of anti-CD19 CAR T cells in the blood concurrent with the onset of anti-tumor activity approximately one week after adoptive transfer (Kochenderfer et al, Blood 2013). (4) Furthermore, we have completed the translational implementation of a new CAR construct with activity against the BCMA receptor expressed on myeloma cells (Carpenter et al, Clin Cancer Res, 2013). We worked closely with the P.I. to establish the procedures for transfection of the CAR construct and expansion of cells, support functional assays of CAR T cell interferon production and validated the final GMP grade construct for use in a newly initiated trial. (5)Finally we are using cellular and molecular assays to assess a novel therapy (09-C-0224, P.I.: Nancy Hardy) utilizing irradiation of selected tumor sites followed by donor lymphocyte infusions (a standard method of immune therapy). Monitoring focuses on whether the irradiation and subsequent localized deaths of tumor cells has produced activation of T effectors and antigen presenting cells and increased anti-tumor immune activation. We have developed a multiplex RNA quantitation assay (Nanostring) using a custom panel of probes we designed to assess upregulation of genes induced by interferon (IFN) and genes responding to cell damage. Although developed to monitor changes in circulating monocytes in graft versus host disease (GVHD) (described in Project ZIC BC 010934), we have determined that these genes are upregulated following donor lymphocyte infusions and infusions of expanded donor T cells. We are implementing these assays on blood and tumor tissue after adoptive immune therapy.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Scientific Cores Intramural Research (ZIC)
Project #
1ZICBC011029-07
Application #
8938452
Study Section
Project Start
Project End
Budget Start
Budget End
Support Year
7
Fiscal Year
2014
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