Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Cell ; 187(5): 1278-1295.e20, 2024 Feb 29.
Article En | MEDLINE | ID: mdl-38387457

CRISPR technologies have begun to revolutionize T cell therapies; however, conventional CRISPR-Cas9 genome-editing tools are limited in their safety, efficacy, and scope. To address these challenges, we developed multiplexed effector guide arrays (MEGA), a platform for programmable and scalable regulation of the T cell transcriptome using the RNA-guided, RNA-targeting activity of CRISPR-Cas13d. MEGA enables quantitative, reversible, and massively multiplexed gene knockdown in primary human T cells without targeting or cutting genomic DNA. Applying MEGA to a model of CAR T cell exhaustion, we robustly suppressed inhibitory receptor upregulation and uncovered paired regulators of T cell function through combinatorial CRISPR screening. We additionally implemented druggable regulation of MEGA to control CAR activation in a receptor-independent manner. Lastly, MEGA enabled multiplexed disruption of immunoregulatory metabolic pathways to enhance CAR T cell fitness and anti-tumor activity in vitro and in vivo. MEGA offers a versatile synthetic toolkit for applications in cancer immunotherapy and beyond.


Metabolic Engineering , T-Lymphocytes , Humans , Gene Expression Profiling , Metabolic Engineering/methods , RNA , Transcriptome
2.
Cancer Cell ; 42(2): 266-282.e8, 2024 02 12.
Article En | MEDLINE | ID: mdl-38278150

Adenosine (Ado) mediates immune suppression in the tumor microenvironment and exhausted CD8+ CAR-T cells express CD39 and CD73, which mediate proximal steps in Ado generation. Here, we sought to enhance CAR-T cell potency by knocking out CD39, CD73, or adenosine receptor 2a (A2aR) but observed only modest effects. In contrast, overexpression of Ado deaminase (ADA-OE), which metabolizes Ado to inosine (INO), induced stemness and enhanced CAR-T functionality. Similarly, CAR-T cell exposure to INO augmented function and induced features of stemness. INO induced profound metabolic reprogramming, diminishing glycolysis, increasing mitochondrial and glycolytic capacity, glutaminolysis and polyamine synthesis, and reprogrammed the epigenome toward greater stemness. Clinical scale manufacturing using INO generated enhanced potency CAR-T cell products meeting criteria for clinical dosing. These results identify INO as a potent modulator of CAR-T cell metabolism and epigenetic stemness programming and deliver an enhanced potency platform for cell manufacturing.


Inosine , T-Lymphocytes , Humans , T-Lymphocytes/metabolism
3.
bioRxiv ; 2023 Apr 25.
Article En | MEDLINE | ID: mdl-37162847

Adenosine (Ado) mediates immune suppression in the tumor microenvironment and exhausted CD8+ CAR T cells mediate Ado-induced immunosuppression through CD39/73-dependent Ado production. Knockout of CD39, CD73 or A2aR had modest effects on exhausted CAR T cells, whereas overexpression of Ado deaminase (ADA), which metabolizes Ado to inosine (INO), induced stemness features and potently enhanced functionality. Similarly, and to a greater extent, exposure of CAR T cells to INO augmented CAR T cell function and induced hallmark features of T cell stemness. INO induced a profound metabolic reprogramming, diminishing glycolysis and increasing oxidative phosphorylation, glutaminolysis and polyamine synthesis, and modulated the epigenome toward greater stemness. Clinical scale manufacturing using INO generated enhanced potency CAR T cell products meeting criteria for clinical dosing. These data identify INO as a potent modulator of T cell metabolism and epigenetic stemness programming and deliver a new enhanced potency platform for immune cell manufacturing.

4.
Science ; 378(6620): eabn5647, 2022 Nov 11.
Article En | MEDLINE | ID: mdl-36356142

T cells are the major arm of the immune system responsible for controlling and regressing cancers. To identify genes limiting T cell function, we conducted genome-wide CRISPR knockout screens in human chimeric antigen receptor (CAR) T cells. Top hits were MED12 and CCNC, components of the Mediator kinase module. Targeted MED12 deletion enhanced antitumor activity and sustained the effector phenotype in CAR- and T cell receptor-engineered T cells, and inhibition of CDK8/19 kinase activity increased expansion of nonengineered T cells. MED12-deficient T cells manifested increased core Meditator chromatin occupancy at transcriptionally active enhancers-most notably for STAT and AP-1 transcription factors-and increased IL2RA expression and interleukin-2 sensitivity. These results implicate Mediator in T cell effector programming and identify the kinase module as a target for enhancing potency of antitumor T cell responses.


Cyclin C , Mediator Complex , Neoplasms , Receptors, Chimeric Antigen , T-Lymphocytes , Humans , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinases/metabolism , Mediator Complex/genetics , T-Lymphocytes/immunology , Transcription Factors/genetics , Genome-Wide Association Study , Cyclin C/genetics , Genetic Testing , Immunotherapy, Adoptive , Neoplasms/immunology , Neoplasms/therapy
5.
Nat Med ; 28(9): 1860-1871, 2022 09.
Article En | MEDLINE | ID: mdl-36097223

Approximately 60% of patients with large B cell lymphoma treated with chimeric antigen receptor (CAR) T cell therapies targeting CD19 experience disease progression, and neurotoxicity remains a challenge. Biomarkers associated with resistance and toxicity are limited. In this study, single-cell proteomic profiling of circulating CAR T cells in 32 patients treated with CD19-CAR identified that CD4+Helios+ CAR T cells on day 7 after infusion are associated with progressive disease and less severe neurotoxicity. Deep profiling demonstrated that this population is non-clonal and manifests hallmark features of T regulatory (TReg) cells. Validation cohort analysis upheld the link between higher CAR TReg cells with clinical progression and less severe neurotoxicity. A model combining expansion of this subset with lactate dehydrogenase levels, as a surrogate for tumor burden, was superior for predicting durable clinical response compared to models relying on each feature alone. These data credential CAR TReg cell expansion as a novel biomarker of response and toxicity after CAR T cell therapy and raise the prospect that this subset may regulate CAR T cell responses in humans.


Neurotoxicity Syndromes , Receptors, Chimeric Antigen , Antigens, CD19 , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Lactate Dehydrogenases , Neurotoxicity Syndromes/etiology , Proteomics , Receptors, Antigen, T-Cell
6.
Cell ; 185(10): 1745-1763.e22, 2022 05 12.
Article En | MEDLINE | ID: mdl-35483375

Regulatable CAR platforms could circumvent toxicities associated with CAR-T therapy, but existing systems have shortcomings including leakiness and attenuated activity. Here, we present SNIP CARs, a protease-based platform for regulating CAR activity using an FDA-approved small molecule. Design iterations yielded CAR-T cells that manifest full functional capacity with drug and no leaky activity in the absence of drug. In numerous models, SNIP CAR-T cells were more potent than constitutive CAR-T cells and showed diminished T cell exhaustion and greater stemness. In a ROR1-based CAR lethality model, drug cessation following toxicity onset reversed toxicity, thereby credentialing the platform as a safety switch. In the same model, reduced drug dosing opened a therapeutic window that resulted in tumor eradication in the absence of toxicity. SNIP CARs enable remote tuning of CAR activity, which provides solutions to safety and efficacy barriers that are currently limiting progress in using CAR-T cells to treat solid tumors.


Neoplasms , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive/methods , Neoplasms/drug therapy , Neoplasms/pathology , Peptide Hydrolases , Receptors, Antigen, T-Cell , T-Lymphocytes/pathology
7.
Science ; 372(6537)2021 04 02.
Article En | MEDLINE | ID: mdl-33795428

T cell exhaustion limits immune responses against cancer and is a major cause of resistance to chimeric antigen receptor (CAR)-T cell therapeutics. Using murine xenograft models and an in vitro model wherein tonic CAR signaling induces hallmark features of exhaustion, we tested the effect of transient cessation of receptor signaling, or rest, on the development and maintenance of exhaustion. Induction of rest through enforced down-regulation of the CAR protein using a drug-regulatable system or treatment with the multikinase inhibitor dasatinib resulted in the acquisition of a memory-like phenotype, global transcriptional and epigenetic reprogramming, and restored antitumor functionality in exhausted CAR-T cells. This work demonstrates that rest can enhance CAR-T cell efficacy by preventing or reversing exhaustion, and it challenges the notion that exhaustion is an epigenetically fixed state.


Dasatinib/pharmacology , Epigenesis, Genetic , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic , Down-Regulation , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenome , Female , Hepatocyte Nuclear Factor 1-alpha/metabolism , High Mobility Group Proteins/metabolism , Humans , Immunologic Memory , Lymphocyte Activation , Lymphoid Enhancer-Binding Factor 1/metabolism , Male , Mice , Neoplasms, Experimental/therapy , Protein Domains , Protein Stability , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/immunology , Signal Transduction , T-Lymphocytes/metabolism , Transcription, Genetic , Xenograft Model Antitumor Assays
8.
Clin Cancer Res ; 27(4): 1058-1068, 2021 02 15.
Article En | MEDLINE | ID: mdl-33087332

PURPOSE: Immunomonitoring of chimeric antigen receptor (CAR) T cells relies primarily on their quantification in the peripheral blood, which inadequately quantifies their biodistribution and activation status in the tissues. Noninvasive molecular imaging of CAR T cells by PET is a promising approach with the ability to provide spatial, temporal, and functional information. Reported strategies rely on the incorporation of reporter transgenes or ex vivo biolabeling, significantly limiting the application of CAR T-cell molecular imaging. In this study, we assessed the ability of antibody-based PET (immunoPET) to noninvasively visualize CAR T cells. EXPERIMENTAL DESIGN: After analyzing human CAR T cells in vitro and ex vivo from patient samples to identify candidate targets for immunoPET, we employed a syngeneic, orthotopic murine tumor model of lymphoma to assess the feasibility of in vivo tracking of CAR T cells by immunoPET using the 89Zr-DFO-anti-ICOS tracer, which we have previously reported. RESULTS: Analysis of human CD19-CAR T cells during activation identified the Inducible T-cell COStimulator (ICOS) as a potential target for immunoPET. In a preclinical tumor model, 89Zr-DFO-ICOS mAb PET-CT imaging detected significantly higher signal in specific bone marrow-containing skeletal sites of CAR T-cell-treated mice compared with controls. Importantly, administration of ICOS-targeting antibodies at tracer doses did not interfere with CAR T-cell persistence and function. CONCLUSIONS: This study highlights the potential of ICOS-immunoPET imaging for monitoring of CAR T-cell therapy, a strategy readily applicable to both commercially available and investigational CAR T cells.See related commentary by Volpe et al., p. 911.


Immunotherapy, Adoptive/methods , Inducible T-Cell Co-Stimulator Protein/metabolism , Lymphoma, Large B-Cell, Diffuse/therapy , T-Lymphocytes/transplantation , Animals , Biological Products/therapeutic use , Cell Line, Tumor , Coculture Techniques , Datasets as Topic , Disease Models, Animal , Humans , Inducible T-Cell Co-Stimulator Protein/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , Mice , Mice, Transgenic , Molecular Imaging/methods , Positron Emission Tomography Computed Tomography , Positron-Emission Tomography/methods , RNA-Seq , Receptors, Chimeric Antigen/immunology , Retrospective Studies , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
9.
PLoS One ; 12(6): e0179606, 2017.
Article En | MEDLINE | ID: mdl-28662050

BACKGROUND: Arsenic and cadmium are environmental pollutants, and although the evidence for adverse immune effects after prenatal arsenic and cadmium exposures is increasing, little is known about the underlying immunological mechanisms. METHODS: We investigated the relationship between prenatal arsenic and cadmium exposures and a variety of T cell subpopulations measured in cord blood for 63 participants in the New Hampshire Birth Cohort Study. Post-partum toenail concentrations of arsenic and cadmium were used as an estimate of maternal exposure during pregnancy. The characteristics of cord blood proportions of T lymphocytes and subpopulations (expression of markers for Th1, Th2, Th17, Th1Th17, induced and natural regulatory T cells and NKTs) are presented. RESULTS: In regression analyses, maternal arsenic exposure levels were inversely associated with cord blood T helper memory cells (-21%, 95% CI: -36%, -3%) and the association was found to be stronger in females. They were also inversely associated with activated T helper memory cells, particularly in males (-26%, 95% CI: -43%, -3%). Similarly, inverse associations were observed between cadmium exposure levels and activated T helper memory cells (-16%, 95% CI: -30%, -1%) and also for T helper memory cells in females (-20%, 95% CI: -35%, -3%). CONCLUSION: The results suggest that prenatal exposures to relatively low levels of arsenic and cadmium may contribute to altered distribution of T cell populations at birth. These changes in theory, could have contributed to the previously reported immunosuppressive effects observed later in infancy/childhood.


Arsenic/toxicity , Cadmium/toxicity , Environmental Exposure , Fetal Blood/cytology , T-Lymphocyte Subsets/drug effects , Adult , Arsenic/analysis , Cadmium/analysis , Female , Humans , Immunophenotyping , Nails/chemistry , Pregnancy , T-Lymphocyte Subsets/immunology , Young Adult
10.
Clin Immunol ; 155(2): 188-97, 2014 Dec.
Article En | MEDLINE | ID: mdl-25229165

Arsenic has wide-ranging effects on human health and there is evidence that it alters the immune response by influencing CD4+/CD8+ T cell ratios, IL-2 cytokine levels, and the expression of immune-response genes. We investigated the impact of in utero environmental arsenic exposure on immune development and function in newborns participating in a pregnancy cohort in New Hampshire, U.S., where arsenic levels have exceeded the current EPA maximum contaminant level of 10 µg/L. Our results showed that maternal urinary arsenic concentrations were inversely related to absolute total CD45RA+ CD4+ cord blood CD69+ T cell counts (N=116, p=0.04) and positively associated with CD45RA+ CD69- CD294+ cell counts (p=0.01). In placental samples (N=70), higher in utero urinary arsenic concentrations were positively associated with the expression of IL1ß (p=0.03). These data provide evidence that relatively low-level arsenic exposure in utero may alter the fetal immune system and lead to immune dysregulation.


Arsenic/adverse effects , Immune System/drug effects , Immune System/physiology , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects/immunology , Adult , Female , Fetal Blood/cytology , Gene Expression , Humans , Immunophenotyping , Infant, Newborn , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lymphocyte Activation/immunology , Male , New Hampshire , Phenotype , Placenta/immunology , Placenta/metabolism , Pregnancy , Risk Factors , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
11.
Proc Natl Acad Sci U S A ; 105(13): 5111-6, 2008 Apr 01.
Article En | MEDLINE | ID: mdl-18364393

Recombinant HIV-Tat (Tat) induces extensive apoptosis in peripheral blood mononuclear cells (PBMCs) cultured in typical CO2 incubators, which are equilibrated with air (21% O2). However, as we show here, Tat apoptosis induction fails in PBMCs cultured at physiological oxygen levels (5% O2). Under these conditions, Tat induces PBMCs to divide, efficiently primes them for HIV infection, and supports virus production by the infected cells. Furthermore, Tat takes only 2 h to prime PBMCs under these conditions. In contrast, PHA/IL-2, which is widely used to prime cells for HIV infection, takes 2-3 days. These findings strongly recommend culturing primary cells at physiological oxygen levels. In addition, they suggest HIV-Tat as a key regulator of HIV disease progression.


Cell Culture Techniques/methods , Gene Products, tat/pharmacology , HIV Infections/immunology , Lymphocytes/drug effects , Oxygen/pharmacology , Apoptosis/drug effects , Cell Division/drug effects , Disease Progression , Gene Products, tat/chemistry , Gene Products, tat/genetics , Gene Products, tat/metabolism , HIV/drug effects , HIV/physiology , HIV Infections/pathology , HIV Infections/virology , Humans , Interleukin-2/pharmacology , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/pathology , Oxygen/metabolism , Time Factors , Virus Replication/drug effects
...