Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Publication year range
1.
Cell ; 186(21): 4567-4582.e20, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37794590

ABSTRACT

CRISPR-Cas9 genome editing has enabled advanced T cell therapies, but occasional loss of the targeted chromosome remains a safety concern. To investigate whether Cas9-induced chromosome loss is a universal phenomenon and evaluate its clinical significance, we conducted a systematic analysis in primary human T cells. Arrayed and pooled CRISPR screens revealed that chromosome loss was generalizable across the genome and resulted in partial and entire loss of the targeted chromosome, including in preclinical chimeric antigen receptor T cells. T cells with chromosome loss persisted for weeks in culture, implying the potential to interfere with clinical use. A modified cell manufacturing process, employed in our first-in-human clinical trial of Cas9-engineered T cells (NCT03399448), reduced chromosome loss while largely preserving genome editing efficacy. Expression of p53 correlated with protection from chromosome loss observed in this protocol, suggesting both a mechanism and strategy for T cell engineering that mitigates this genotoxicity in the clinic.


Subject(s)
CRISPR-Cas Systems , Chromosome Aberrations , Gene Editing , T-Lymphocytes , Humans , Chromosomes , CRISPR-Cas Systems/genetics , DNA Damage , Gene Editing/methods , Clinical Trials as Topic
2.
Annu Rev Immunol ; 32: 189-225, 2014.
Article in English | MEDLINE | ID: mdl-24423116

ABSTRACT

Adoptive immunotherapy, or the infusion of lymphocytes, is a promising approach for the treatment of cancer and certain chronic viral infections. The application of the principles of synthetic biology to enhance T cell function has resulted in substantial increases in clinical efficacy. The primary challenge to the field is to identify tumor-specific targets to avoid off-tumor, on-target toxicity. Given recent advances in efficacy in numerous pilot trials, the next steps in clinical development will require multicenter trials to establish adoptive immunotherapy as a mainstream technology.


Subject(s)
Immunotherapy, Adoptive , Neoplasms/immunology , Neoplasms/therapy , Virus Diseases/immunology , Virus Diseases/therapy , Adoptive Transfer , Animals , Antigens/genetics , Antigens/immunology , Biomarkers , Cell- and Tissue-Based Therapy , Gene Transfer Techniques , Genetic Therapy , Humans , Neoplasms/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transduction, Genetic , Virus Diseases/genetics
3.
Immunity ; 56(10): 2388-2407.e9, 2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37776850

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy targeting CD19 has achieved tremendous success treating B cell malignancies; however, some patients fail to respond due to poor autologous T cell fitness. To improve response rates, we investigated whether disruption of the co-inhibitory receptors CTLA4 or PD-1 could restore CART function. CRISPR-Cas9-mediated deletion of CTLA4 in preclinical models of leukemia and myeloma improved CAR T cell proliferation and anti-tumor efficacy. Importantly, this effect was specific to CTLA4 and not seen upon deletion of CTLA4 and/or PDCD1 in CAR T cells. Mechanistically, CTLA4 deficiency permitted unopposed CD28 signaling and maintenance of CAR expression on the T cell surface under conditions of high antigen load. In clinical studies, deletion of CTLA4 rescued the function of T cells from patients with leukemia that previously failed CAR T cell treatment. Thus, selective deletion of CTLA4 reinvigorates dysfunctional chronic lymphocytic leukemia (CLL) patient T cells, providing a strategy for increasing patient responses to CAR T cell therapy.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Receptors, Chimeric Antigen , Humans , Receptors, Antigen, T-Cell/metabolism , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , T-Lymphocytes , Immunotherapy, Adoptive , Antigens, CD19
4.
Nature ; 629(8010): 211-218, 2024 May.
Article in English | MEDLINE | ID: mdl-38600391

ABSTRACT

A major limitation of chimeric antigen receptor (CAR) T cell therapies is the poor persistence of these cells in vivo1. The expression of memory-associated genes in CAR T cells is linked to their long-term persistence in patients and clinical efficacy2-6, suggesting that memory programs may underpin durable CAR T cell function. Here we show that the transcription factor FOXO1 is responsible for promoting memory and restraining exhaustion in human CAR T cells. Pharmacological inhibition or gene editing of endogenous FOXO1 diminished the expression of memory-associated genes, promoted an exhaustion-like phenotype and impaired the antitumour activity of CAR T cells. Overexpression of FOXO1 induced a gene-expression program consistent with T cell memory and increased chromatin accessibility at FOXO1-binding motifs. CAR T cells that overexpressed FOXO1 retained their function, memory potential and metabolic fitness in settings of chronic stimulation, and exhibited enhanced persistence and tumour control in vivo. By contrast, overexpression of TCF1 (encoded by TCF7) did not enforce canonical memory programs or enhance the potency of CAR T cells. Notably, FOXO1 activity correlated with positive clinical outcomes of patients treated with CAR T cells or tumour-infiltrating lymphocytes, underscoring the clinical relevance of FOXO1 in cancer immunotherapy. Our results show that overexpressing FOXO1 can increase the antitumour activity of human CAR T cells, and highlight memory reprogramming as a broadly applicable approach for optimizing therapeutic T cell states.


Subject(s)
Forkhead Box Protein O1 , Immunologic Memory , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , T-Lymphocytes , Animals , Humans , Mice , Cell Line, Tumor , Chromatin/metabolism , Chromatin/genetics , Forkhead Box Protein O1/metabolism , Gene Editing , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/cytology
5.
Nature ; 602(7897): 503-509, 2022 02.
Article in English | MEDLINE | ID: mdl-35110735

ABSTRACT

The adoptive transfer of T lymphocytes reprogrammed to target tumour cells has demonstrated potential for treatment of various cancers1-7. However, little is known about the long-term potential and clonal stability of the infused cells. Here we studied long-lasting CD19-redirected chimeric antigen receptor (CAR) T cells in two patients with chronic lymphocytic leukaemia1-4 who achieved a complete remission in 2010. CAR T cells remained detectable more than ten years after infusion, with sustained remission in both patients. Notably, a highly activated CD4+ population emerged in both patients, dominating the CAR T cell population at the later time points. This transition was reflected in the stabilization of the clonal make-up of CAR T cells with a repertoire dominated by a small number of clones. Single-cell profiling demonstrated that these long-persisting CD4+ CAR T cells exhibited cytotoxic characteristics along with ongoing functional activation and proliferation. In addition, longitudinal profiling revealed a population of gamma delta CAR T cells that prominently expanded in one patient concomitant with CD8+ CAR T cells during the initial response phase. Our identification and characterization of these unexpected CAR T cell populations provide novel insight into the CAR T cell characteristics associated with anti-cancer response and long-term remission in leukaemia.


Subject(s)
CD4-Positive T-Lymphocytes , Immunotherapy, Adoptive , Leukemia , Receptors, Chimeric Antigen , Antigens, CD19/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Separation , Humans , Leukemia/immunology , Leukemia/therapy , Receptors, Chimeric Antigen/immunology , Time Factors
6.
Semin Immunol ; 70: 101840, 2023 11.
Article in English | MEDLINE | ID: mdl-37729825

ABSTRACT

Population aging, a pervasive global demographic trend, is anticipated to challenge health and social systems worldwide. This phenomenon is due to medical advancements enabling longer lifespans, with 20% of the US population soon to be over 65 years old. Consequently, there will be a surge in age-related diseases. Senescence, characterized by the loss of biological maintenance and homeostasis at molecular and cellular levels, either correlates with or directly causes age-related phenotypic changes. Decline of the immune system is a critical factor in the senescence process, with cancer being a primary cause of death in elderly populations. Chimeric antigen receptor (CAR) T cell therapy, an innovative approach, has demonstrated success mainly in pediatric and young adult hematological malignancies but remains largely ineffective for diseases affecting older populations, such as late-in-life B cell malignancies and most solid tumor indications. This limitation arises because CAR T cell efficacy heavily relies on the fitness of the patient-derived starting T cell material. Numerous studies suggest that T cell senescence may be a key driver of CAR T cell deficiency. This review examines correlates and underlying factors associated with favorable CAR T cell outcomes and explores potential experimental and clinically actionable strategies for T cell rejuvenation.


Subject(s)
Neoplasms , Receptors, Antigen, T-Cell , Adolescent , Humans , Child , Aged , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes , Immunotherapy, Adoptive , Aging
7.
Nat Immunol ; 14(6): 593-602, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23603793

ABSTRACT

We found upregulation of expression of the microRNA miR-155 in primary effector and effector memory CD8(+) T cells, but low miR-155 expression in naive and central memory cells. Antiviral CD8(+) T cell responses and viral clearance were impaired in miR-155-deficient mice, and this defect was intrinsic to CD8(+) T cells, as miR-155-deficient CD8(+) T cells mounted greatly diminished primary and memory responses. Conversely, miR-155 overexpression augmented antiviral CD8(+) T cell responses in vivo. Gene-expression profiling showed that miR-155-deficient CD8(+) T cells had enhanced type I interferon signaling and were more susceptible to interferon's antiproliferative effect. Inhibition of the type I interferon-associated transcription factors STAT1 or IRF7 resulted in enhanced responses of miR-155-deficient CD8(+) T cells in vivo. We have thus identified a previously unknown role for miR-155 in regulating responsiveness to interferon and CD8(+) T cell responses to pathogens in vivo.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interferons/immunology , MicroRNAs/immunology , Signal Transduction/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Flow Cytometry , Gene Expression Profiling , Gene Regulatory Networks/genetics , Gene Regulatory Networks/immunology , Immunoblotting , Immunologic Memory/genetics , Immunologic Memory/immunology , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Regulatory Factor-7/metabolism , Interferons/metabolism , Interferons/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , Signal Transduction/genetics , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/immunology , Suppressor of Cytokine Signaling Proteins/metabolism
8.
Immunity ; 44(2): 380-90, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26885860

ABSTRACT

Chimeric antigen receptors (CARs) redirect T cell cytotoxicity against cancer cells, providing a promising approach to cancer immunotherapy. Despite extensive clinical use, the attributes of CAR co-stimulatory domains that impact persistence and resistance to exhaustion of CAR-T cells remain largely undefined. Here, we report the influence of signaling domains of coreceptors CD28 and 4-1BB on the metabolic characteristics of human CAR T cells. Inclusion of 4-1BB in the CAR architecture promoted the outgrowth of CD8(+) central memory T cells that had significantly enhanced respiratory capacity, increased fatty acid oxidation and enhanced mitochondrial biogenesis. In contrast, CAR T cells with CD28 domains yielded effector memory cells with a genetic signature consistent with enhanced glycolysis. These results provide, at least in part, a mechanistic insight into the differential persistence of CAR-T cells expressing 4-1BB or CD28 signaling domains in clinical trials and inform the design of future CAR T cell therapies.


Subject(s)
CD28 Antigens/metabolism , CD8-Positive T-Lymphocytes/physiology , Cancer Vaccines/immunology , Immunotherapy , Neoplasms/therapy , Receptors, Antigen, T-Cell/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , CD28 Antigens/genetics , Cell Respiration , Cells, Cultured , Glycolysis , Humans , Immunologic Memory , Lipid Metabolism , Mitochondria/metabolism , Neoplasms/immunology , Receptor Cross-Talk , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , Signal Transduction/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
10.
Nature ; 558(7709): 307-312, 2018 06.
Article in English | MEDLINE | ID: mdl-29849141

ABSTRACT

Cancer immunotherapy based on genetically redirecting T cells has been used successfully to treat B cell malignancies1-3. In this strategy, the T cell genome is modified by integration of viral vectors or transposons encoding chimaeric antigen receptors (CARs) that direct tumour cell killing. However, this approach is often limited by the extent of expansion and persistence of CAR T cells4,5. Here we report mechanistic insights from studies of a patient with chronic lymphocytic leukaemia treated with CAR T cells targeting the CD19 protein. Following infusion of CAR T cells, anti-tumour activity was evident in the peripheral blood, lymph nodes and bone marrow; this activity was accompanied by complete remission. Unexpectedly, at the peak of the response, 94% of CAR T cells originated from a single clone in which lentiviral vector-mediated insertion of the CAR transgene disrupted the methylcytosine dioxygenase TET2 gene. Further analysis revealed a hypomorphic mutation in this patient's second TET2 allele. TET2-disrupted CAR T cells exhibited an epigenetic profile consistent with altered T cell differentiation and, at the peak of expansion, displayed a central memory phenotype. Experimental knockdown of TET2 recapitulated the potency-enhancing effect of TET2 dysfunction in this patient's CAR T cells. These findings suggest that the progeny of a single CAR T cell induced leukaemia remission and that TET2 modification may be useful for improving immunotherapies.


Subject(s)
5-Methylcytosine/metabolism , Antigens, CD19/immunology , Dioxygenases/genetics , Immunotherapy/methods , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Adoptive Transfer , Aged , Alleles , Cell Differentiation , Clinical Trials as Topic , Clone Cells/cytology , Clone Cells/immunology , Dioxygenases/metabolism , Epigenesis, Genetic , HEK293 Cells , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Mutation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transgenes
11.
Mol Ther ; 31(8): 2309-2325, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37312454

ABSTRACT

Multiple clinical studies have treated mesothelin (MSLN)-positive solid tumors by administering MSLN-directed chimeric antigen receptor (CAR) T cells. Although these products are generally safe, efficacy is limited. Therefore, we generated and characterized a potent, fully human anti-MSLN CAR. In a phase 1 dose-escalation study of patients with solid tumors, we observed two cases of severe pulmonary toxicity following intravenous infusion of this product in the high-dose cohort (1-3 × 108 T cells per m2). Both patients demonstrated progressive hypoxemia within 48 h of infusion with clinical and laboratory findings consistent with cytokine release syndrome. One patient ultimately progressed to grade 5 respiratory failure. An autopsy revealed acute lung injury, extensive T cell infiltration, and accumulation of CAR T cells in the lungs. RNA and protein detection techniques confirmed low levels of MSLN expression by benign pulmonary epithelial cells in affected lung and lung samples obtained from other inflammatory or fibrotic conditions, indicating that pulmonary pneumocyte and not pleural expression of mesothelin may lead to dose-limiting toxicity. We suggest patient enrollment criteria and dosing regimens of MSLN-directed therapies consider the possibility of dynamic expression of mesothelin in benign lung with a special concern for patients with underlying inflammatory or fibrotic conditions.


Subject(s)
Mesothelin , Neoplasms , Humans , GPI-Linked Proteins/genetics , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Neoplasms/therapy , T-Lymphocytes
13.
Proc Natl Acad Sci U S A ; 117(10): 5442-5452, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32094195

ABSTRACT

Chimeric antigen receptor (CAR)-T immunotherapy has yielded impressive results in several B cell malignancies, establishing itself as a powerful means to redirect the natural properties of T lymphocytes. In this strategy, the T cell genome is modified by the integration of lentiviral vectors encoding CAR that direct tumor cell killing. However, this therapeutic approach is often limited by the extent of CAR-T cell expansion in vivo. A major outstanding question is whether or not CAR-T integration itself enhances the proliferative competence of individual T cells by rewiring their regulatory landscape. To address this question, it is critical to define the identity of an individual CAR-T cell and simultaneously chart where the CAR-T vector integrates into the genome. Here, we report the development of a method called EpiVIA (https://github.com/VahediLab/epiVIA) for the joint profiling of the chromatin accessibility and lentiviral integration site analysis at the population and single-cell levels. We validate our technique in clonal cells with previously defined integration sites and further demonstrate the ability to measure lentiviral integration sites and chromatin accessibility of host and viral genomes at the single-cell resolution in CAR-T cells. We anticipate that EpiVIA will enable the single-cell deconstruction of gene regulation during CAR-T therapy, leading to the discovery of cellular factors associated with durable treatment.


Subject(s)
Chromatin , Epigenesis, Genetic , Immunotherapy, Adoptive , Single-Cell Analysis/methods , T-Lymphocytes , Virus Integration/genetics , Clone Cells , Genetic Testing , Genome, Human , Humans , Lentivirus , Proviruses
14.
Mol Cancer ; 21(1): 78, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35303871

ABSTRACT

Chimeric Antigen Receptor (CAR) T-cells represent a breakthrough in personalized cancer therapy. In this strategy, synthetic receptors comprised of antigen recognition, signaling, and costimulatory domains are used to reprogram T-cells to target tumor cells for destruction. Despite the success of this approach in refractory B-cell malignancies, optimal potency of CAR T-cell therapy for many other cancers, particularly solid tumors, has not been achieved. Factors such as T-cell exhaustion, lack of CAR T-cell persistence, cytokine-related toxicities, and bottlenecks in the manufacturing of autologous products have hampered the safety, effectiveness, and availability of this approach. With the ease and accessibility of CRISPR-Cas9-based gene editing, it is possible to address many of these limitations. Accordingly, current research efforts focus on precision engineering of CAR T-cells with conventional CRISPR-Cas9 systems or novel editors that can install desired genetic changes with or without introduction of a double-stranded break (DSB) into the genome. These tools and strategies can be directly applied to targeting negative regulators of T-cell function, directing therapeutic transgenes to specific genomic loci, and generating reproducibly safe and potent allogeneic universal CAR T-cell products for on-demand cancer immunotherapy. This review evaluates several of the ongoing and future directions of combining next-generation CRISPR-Cas9 gene editing with synthetic biology to optimize CAR T-cell therapy for future clinical trials toward the establishment of a new cancer treatment paradigm.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , CRISPR-Cas Systems , Gene Editing , Humans , Immunotherapy, Adoptive , Neoplasms/drug therapy , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes
15.
Mol Ther ; 29(2): 626-635, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33186691

ABSTRACT

MazF is an Escherichia coli-derived endoribonuclease that selectively cleaves ACA sequences of mRNA prevalent in HIV. We administered a single infusion of autologous CD4 T lymphocytes modified to express a Tat-dependent MazF transgene to 10 HIV-infected individuals (six remaining on antiretroviral therapy [ART]; four undergoing treatment interruption post-infusion) in order to provide a population of HIV-resistant immune cells. In participants who remained on ART, increases in CD4 and CD8 T cell counts of ~200 cells/mm3 each occurred within 2 weeks of infusion and persisted for at least 6 months. Modified cells were detectable for several months in the blood and trafficked to gastrointestinal lymph tissue. HIV-1 Tat introduced ex vivo to the modified CD4+ T cells induced MazF expression in both pre- and post-infusion samples, and MazF expression was detected in vivo post-viral-rebound during ATI. One participant experienced mild cytokine release syndrome. In sum, this study of a single infusion of MazF-modified CD4 T lymphocytes demonstrated safety of these cells, distribution to lymph tissue and maintenance of Tat-inducible MazF endoribonuclease activity, as well as sustained elevation of blood CD4 and CD8 T cell counts. Future studies to assess effects on viremia and latent proviral reservoir are warranted.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Endoribonucleases/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/physiology , tat Gene Products, Human Immunodeficiency Virus/metabolism , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endoribonucleases/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Genetic Therapy , HIV Infections/metabolism , HIV Infections/therapy , Viral Load , Virus Replication
16.
Blood ; 134(1): 44-58, 2019 07 04.
Article in English | MEDLINE | ID: mdl-31076448

ABSTRACT

In chronic lymphocytic leukemia (CLL), acquired T-cell dysfunction impedes development of effective immunotherapeutic strategies, through as-yet unresolved mechanisms. We have previously shown that CD8+ T cells in CLL exhibit impaired activation and reduced glucose uptake after stimulation. CD8+ T cells in CLL patients are chronically exposed to leukemic B cells, which potentially impacts metabolic homeostasis resulting in aberrant metabolic reprogramming upon stimulation. Here, we report that resting CD8+ T cells in CLL have reduced intracellular glucose transporter 1 (GLUT1) reserves, and have an altered mitochondrial metabolic profile as displayed by increased mitochondrial respiration, membrane potential, and levels of reactive oxygen species. This coincided with decreased levels of peroxisome proliferator-activated receptor γ coactivator 1-α, and in line with that, CLL-derived CD8+ T cells showed impaired mitochondrial biogenesis upon stimulation. In search of a therapeutic correlate of these findings, we analyzed mitochondrial biogenesis in CD19-directed chimeric antigen receptor (CAR) CD8+ T cells prior to infusion in CLL patients (who were enrolled in NCT01747486 and NCT01029366 [https://clinicaltrials.gov]). Interestingly, in cases with a subsequent complete response, the infused CD8+ CAR T cells had increased mitochondrial mass compared with nonresponders, which positively correlated with the expansion and persistence of CAR T cells. Our findings demonstrate that GLUT1 reserves and mitochondrial fitness of CD8+ T cells are impaired in CLL. Therefore, boosting mitochondrial biogenesis in CAR T cells might improve the efficacy of CAR T-cell therapy and other emerging cellular immunotherapies.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Immunotherapy, Adoptive , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mitochondria/metabolism , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Male , Middle Aged , Organelle Biogenesis , Receptors, Chimeric Antigen
18.
Am J Hematol ; 94(S1): S3-S9, 2019 05.
Article in English | MEDLINE | ID: mdl-30680780

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy represents a major advancement in personalized cancer treatment. In this strategy, a patient's own T cells are genetically engineered to express a synthetic receptor that binds a tumor antigen. CAR T cells are then expanded for clinical use and infused back into the patient's body to attack and destroy chemotherapy-resistant cancer. Dramatic clinical responses and high rates of complete remission have been observed in the setting of CAR T-cell therapy of B-cell malignancies. This resulted in two recent FDA approvals of CAR T cells directed against the CD19 protein for treatment of acute lymphoblastic leukemia and diffuse large B-cell lymphoma. Thus, CAR T cells are arguably one of the first successful examples of synthetic biology and personalized cellular cancer therapy to become commercially available. In this review, we introduce the concept of using CAR T cells to break immunological tolerance to tumors, highlight several challenges in the field, discuss the utility of biomarkers in the context of predicting clinical responses, and offer prospects for developing next-generation CAR T cell-based approaches that will improve outcome.


Subject(s)
Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Chimeric Antigen/therapeutic use , Antigens, CD19/immunology , Antigens, CD19/therapeutic use , Biomarkers , Humans , Immune Tolerance
19.
Mol Ther ; 26(7): 1855-1866, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29807781

ABSTRACT

Cancer has an impressive ability to evolve multiple processes to evade therapies. While immunotherapies and vaccines have shown great promise, particularly in certain solid tumors such as prostate cancer, they have been met with resistance from tumors that use a multitude of mechanisms of immunosuppression to limit effectiveness. Prostate cancer, in particular, secretes transforming growth factor ß (TGF-ß) as a means to inhibit immunity while allowing for cancer progression. Blocking TGF-ß signaling in T cells increases their ability to infiltrate, proliferate, and mediate antitumor responses in prostate cancer models. We tested whether the potency of chimeric antigen receptor (CAR) T cells directed to prostate-specific membrane antigen (PSMA) could be enhanced by the co-expression of a dominant-negative TGF-ßRII (dnTGF-ßRII). Upon expression of the dominant-negative TGF-ßRII in CAR T cells, we observed increased proliferation of these lymphocytes, enhanced cytokine secretion, resistance to exhaustion, long-term in vivo persistence, and the induction of tumor eradication in aggressive human prostate cancer mouse models. Based on our observations, we initiated a phase I clinical trial to assess these CAR T cells as a novel approach for patients with relapsed and refractory metastatic prostate cancer (ClinicalTrials.gov: NCT03089203).


Subject(s)
Antigens, Surface/metabolism , Cell Proliferation/physiology , Glutamate Carboxypeptidase II/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Chimeric Antigen/metabolism , Receptors, Transforming Growth Factor beta/metabolism , T-Lymphocytes/metabolism , Cell Line , Cell Line, Tumor , Humans , Lymphocyte Activation/physiology , Male , Middle Aged , PC-3 Cells , Prostate/metabolism , Prostate/pathology , T-Lymphocytes/pathology , Transforming Growth Factor beta/metabolism
20.
Blood ; 127(9): 1117-27, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26813675

ABSTRACT

Anti-CD19 chimeric antigen receptor (CAR) T-cell therapy is highly promising but requires robust T-cell expansion and engraftment. A T-cell defect in chronic lymphocytic leukemia (CLL) due to disease and/or therapy impairs ex vivo expansion and response to CAR T cells. To evaluate the effect of ibrutinib treatment on the T-cell compartment in CLL as it relates to CAR T-cell generation, we examined the phenotype and function of T cells in a cohort of CLL patients during their course of treatment with ibrutinib. We found that ≥5 cycles of ibrutinib therapy improved the expansion of CD19-directed CAR T cells (CTL019), in association with decreased expression of the immunosuppressive molecule programmed cell death 1 on T cells and of CD200 on B-CLL cells. In support of these findings, we observed that 3 CLL patients who had been treated with ibrutinib for ≥1 year at the time of T-cell collection had improved ex vivo and in vivo CTL019 expansion, which correlated positively together and with clinical response. Lastly, we show that ibrutinib exposure does not impair CAR T-cell function in vitro but does improve CAR T-cell engraftment, tumor clearance, and survival in human xenograft models of resistant acute lymphocytic leukemia and CLL when administered concurrently. Our collective findings indicate that ibrutinib enhances CAR T-cell function and suggest that clinical trials with combination therapy are warranted. Our studies demonstrate that improved T-cell function may also contribute to the efficacy of ibrutinib in CLL. These trials were registered at www.clinicaltrials.gov as #NCT01747486, #NCT01105247, and #NCT01217749.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Adenine/analogs & derivatives , Administration, Oral , Aged , Animals , Antigens, CD/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cytotoxicity, Immunologic/drug effects , Demography , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Gene Transfer Techniques , Humans , Immunosuppression Therapy , K562 Cells , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Mice , Middle Aged , Piperidines , Programmed Cell Death 1 Receptor/metabolism , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , T-Lymphocytes/drug effects , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL