Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Cell Rep Med ; 5(7): 101628, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38986621

ABSTRACT

Chimeric antigen receptor T cells (CART) targeting lymphocyte antigens can induce T cell fratricide and require additional engineering to mitigate self-damage. We demonstrate that the expression of a chimeric antigen receptor (CAR) targeting CD5, a prominent pan-T cell antigen, induces rapid internalization and complete loss of the CD5 protein on T cells, protecting them from self-targeting. Notably, exposure of healthy and malignant T cells to CD5.CART cells induces similar internalization of CD5 on target cells, transiently shielding them from cytotoxicity. However, this protection is short-lived, as sustained activity of CD5.CART cells in patients with T cell malignancies results in full ablation of CD5+ T cells while sparing healthy T cells naturally lacking CD5. These results indicate that continuous downmodulation of the target antigen in CD5.CART cells produces effective fratricide resistance without undermining their on-target cytotoxicity.


Subject(s)
CD5 Antigens , Receptors, Chimeric Antigen , CD5 Antigens/metabolism , Humans , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Cytotoxicity, Immunologic , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Immunotherapy, Adoptive/methods , Animals , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology
2.
Mol Ther ; 31(1): 24-34, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36086817

ABSTRACT

Chimeric antigen receptor (CAR)-mediated targeting of T lineage antigens for the therapy of blood malignancies is frequently complicated by self-targeting of CAR T cells or their excessive differentiation driven by constant CAR signaling. Expression of CARs targeting CD7, a pan-T cell antigen highly expressed in T cell malignancies and some myeloid leukemias, produces robust fratricide and often requires additional mitigation strategies, such as CD7 gene editing. In this study, we show fratricide of CD7 CAR T cells can be fully prevented using ibrutinib and dasatinib, the pharmacologic inhibitors of key CAR/CD3ζ signaling kinases. Supplementation with ibrutinib and dasatinib rescued the ex vivo expansion of unedited CD7 CAR T cells and allowed regaining full CAR-mediated cytotoxicity in vitro and in vivo on withdrawal of the inhibitors. The unedited CD7 CAR T cells persisted long term and mediated sustained anti-leukemic activity in two mouse xenograft models of human T cell acute lymphoblastic leukemia (T-ALL) by self-selecting for CD7-, fratricide-resistant CD7 CAR T cells that were transcriptionally similar to control CD7-edited CD7 CAR T cells. Finally, we showed feasibility of cGMP manufacturing of unedited autologous CD7 CAR T cells for patients with CD7+ malignancies and initiated a phase I clinical trial (ClinicalTrials.gov: NCT03690011) using this approach. These results indicate pharmacologic inhibition of CAR signaling enables generating functional CD7 CAR T cells without additional engineering.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Receptors, Chimeric Antigen , Mice , Animals , Humans , T-Lymphocytes , Immunotherapy, Adoptive/methods , Dasatinib/metabolism , Feasibility Studies , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism
3.
JCI Insight ; 52019 04 02.
Article in English | MEDLINE | ID: mdl-30938714

ABSTRACT

BACKGROUND: Subgroups of patients with relapsed or refractory (R/R) chronic lymphocytic leukemia (CLL) exhibit suboptimal outcomes after standard therapies, including oral kinase inhibitors. We and others have previously reported on safety and efficacy of autologous CD19-targeted CAR T-cells for these patients; here we report safety and long-term follow-up of CAR T-cell therapy with or without conditioning chemotherapy for patients with R/R CLL and indolent B-cell non-Hodgkin lymphoma (B-NHL). METHODS: We conducted a phase 1 clinical trial investigating CD19-targeted CAR T-cells incorporating a CD28 costimulatory domain (19-28z). Seventeen of 20 patients received conditioning chemotherapy prior to CAR T-cell infusion. Five patients with CLL received ibrutinib at the time of autologous T-cell collection and/or CAR T-cell administration. RESULTS: This analysis included 16 patients with R/R CLL and 4 patients with R/R indolent B-NHL. Cytokine release syndrome (CRS) was observed in all 20 patients but grades 3 and 4 CRS and neurological events were uncommon (10% for each). Ex vivo expansion of T-cells and proportions of CD4+/CD8+ CAR T-cells with CD62L+CD127+ immunophenotype were significantly greater in patients on ibrutinib at leukapheresis. Three of 12 evaluable CLL patients receiving conditioning chemotherapy achieved CR (two had minimal residual disease-negative CR). All patients achieving CR remained progression-free at median follow-up of 53 months. CONCLUSION: Conditioning chemotherapy and 19-28z CAR T-cells were acceptably tolerated across investigated dose levels in heavily pretreated patients with R/R CLL and indolent B-NHL, and a subgroup of patients achieved durable CR. Ibrutinib therapy may modulate autologous T-cell phenotype. TRIAL REGISTRATION: ClinicalTrials.gov NCT00466531. FUNDING: Juno Therapeutics.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cytokine Release Syndrome/epidemiology , Immunotherapy, Adoptive/methods , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Lymphoma, B-Cell/therapy , Neoplasm Recurrence, Local/therapy , Transplantation Conditioning/methods , Adenine/analogs & derivatives , Adult , Aged , Antigens, CD19/immunology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chemotherapy, Adjuvant/adverse effects , Chemotherapy, Adjuvant/methods , Cytokine Release Syndrome/immunology , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Follow-Up Studies , Humans , Immunotherapy, Adoptive/adverse effects , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/mortality , Male , Middle Aged , Neoadjuvant Therapy/adverse effects , Neoadjuvant Therapy/methods , Neoplasm Recurrence, Local/immunology , Piperidines , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Receptors, Chimeric Antigen/immunology , Transplantation Conditioning/adverse effects , Transplantation, Autologous/adverse effects , Transplantation, Autologous/methods
4.
Cancer Cell ; 35(3): 473-488.e6, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30889381

ABSTRACT

Chimeric antigen receptor (CAR) T cells provide great efficacy in B cell malignancies. However, improved CAR T cell therapies are still needed. Here, we engineered tumor-targeted CAR T cells to constitutively express the immune-stimulatory molecule CD40 ligand (CD40L) and explored efficacy in different mouse leukemia/lymphoma models. We observed that CD40L+ CAR T cells circumvent tumor immune escape via antigen loss through CD40/CD40L-mediated cytotoxicity and induction of a sustained, endogenous immune response. After adoptive cell transfer, the CD40L+ CAR T cells displayed superior antitumor efficacy, licensed antigen-presenting cells, enhanced recruitment of immune effectors, and mobilized endogenous tumor-recognizing T cells. These effects were absent in Cd40-/- mice and provide a rationale for the use of CD40L+ CAR T cells in cancer treatment.


Subject(s)
CD40 Ligand/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , Animals , Antigen-Presenting Cells/immunology , CD40 Ligand/genetics , Cell Line, Tumor , Humans , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Receptors, Chimeric Antigen/genetics , Tumor Escape , Xenograft Model Antitumor Assays
5.
Nat Biotechnol ; 36(9): 847-856, 2018 10.
Article in English | MEDLINE | ID: mdl-30102295

ABSTRACT

The efficacy of chimeric antigen receptor (CAR) T cell therapy against poorly responding tumors can be enhanced by administering the cells in combination with immune checkpoint blockade inhibitors. Alternatively, the CAR construct has been engineered to coexpress factors that boost CAR-T cell function in the tumor microenvironment. We modified CAR-T cells to secrete PD-1-blocking single-chain variable fragments (scFv). These scFv-secreting CAR-T cells acted in both a paracrine and autocrine manner to improve the anti-tumor activity of CAR-T cells and bystander tumor-specific T cells in clinically relevant syngeneic and xenogeneic mouse models of PD-L1+ hematologic and solid tumors. The efficacy was similar to or better than that achieved by combination therapy with CAR-T cells and a checkpoint inhibitor. This approach may improve safety, as the secreted scFvs remained localized to the tumor, protecting CAR-T cells from PD-1 inhibition, which could potentially avoid toxicities associated with systemic checkpoint inhibition.


Subject(s)
Programmed Cell Death 1 Receptor/immunology , Receptors, Chimeric Antigen/immunology , Single-Chain Antibodies/immunology , T-Lymphocytes/immunology , Animals , Humans , Mice , Tumor Microenvironment , Xenograft Model Antitumor Assays
6.
Cell Rep ; 23(7): 2130-2141, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768210

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has proven clinically beneficial against B cell acute lymphoblastic leukemia and non-Hodgkin's lymphoma. However, suboptimal clinical outcomes have been associated with decreased expansion and persistence of adoptively transferred CAR T cells, antigen-negative relapses, and impairment by an immunosuppressive tumor microenvironment. Improvements in CAR T cell design are required to enhance clinical efficacy, as well as broaden the applicability of this technology. Here, we demonstrate that interleukin-18 (IL-18)-secreting CAR T cells exhibit enhanced in vivo expansion and persistence and significantly increase long-term survival in syngeneic mouse models of both hematological and solid malignancies. In addition, we demonstrate that IL-18-secreting CAR T cells are capable of modulating the tumor microenvironment, as well as enhancing an effective endogenous anti-tumor immune response. IL-18-secreting CAR T cells represent a promising strategy to enhance the clinical outcomes of adoptive T cell therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Immune System/metabolism , Immunotherapy , Neoplasms/drug therapy , T-Lymphocytes/immunology , Animals , Antineoplastic Agents/therapeutic use , Autocrine Communication/drug effects , B-Lymphocytes/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Humans , Immune System/drug effects , Immunotherapy, Adoptive , Interleukin-18/metabolism , Mice, Inbred C57BL , Mice, SCID , Neoplasms/immunology , Neoplasms/pathology , Signal Transduction/drug effects , T-Lymphocytes/drug effects , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
7.
Mol Ther ; 23(4): 769-78, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25582824

ABSTRACT

Adoptive cell therapy with genetically modified T cells expressing a chimeric antigen receptor (CAR) is a promising therapy for patients with B-cell acute lymphoblastic leukemia. However, CAR-modified T cells (CAR T cells) have mostly failed in patients with solid tumors or low-grade B-cell malignancies including chronic lymphocytic leukemia with bulky lymph node involvement. Herein, we enhance the antitumor efficacy of CAR T cells through the constitutive expression of CD40 ligand (CD40L, CD154). T cells genetically modified to constitutively express CD40L (CD40L-modified T cells) demonstrated increased proliferation and secretion of proinflammatory TH1 cytokines. Further, CD40L-modified T cells augmented the immunogenicity of CD40(+) tumor cells by the upregulated surface expression of costimulatory molecules (CD80 and CD86), adhesion molecules (CD54, CD58, and CD70), human leukocyte antigen (HLA) molecules (Class I and HLA-DR), and the Fas-death receptor (CD95). Additionally, CD40L-modified T cells induced maturation and secretion of the proinflammatory cytokine interleukin-12 by monocyte-derived dendritic cells. Finally, tumor-targeted CD19-specific CAR/CD40L T cells exhibited increased cytotoxicity against CD40(+) tumors and extended the survival of tumor-bearing mice in a xenotransplant model of CD19(+) systemic lymphoma. This preclinical data supports the clinical application of CAR T cells additionally modified to constitutively express CD40L with anticipated enhanced antitumor efficacy.


Subject(s)
CD40 Ligand/metabolism , Immunotherapy , Lymphoma, Follicular/therapy , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Heterografts , Humans , Immunophenotyping , Lymphoma, Follicular/immunology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...