Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Cell Rep ; 40(5): 111153, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35926468

ABSTRACT

Adoptive T cell therapies (ACT) have been curative for a limited number of cancer patients. The sensitization of cancer cells to T cell killing may expand the benefit of these therapies for more patients. To this end, we use a three-step approach to identify cancer genes that disfavor T cell immunity. First, we profile gene transcripts upregulated by cancer under selection pressure from T cell killing. Second, we identify potential tumor gene targets and pathways that disfavor T cell killing using signaling pathway activation libraries and genome-wide loss-of-function CRISPR-Cas9 screens. Finally, we implement pharmacological perturbation screens to validate these targets and identify BIRC2, ITGAV, DNPEP, BCL2, and ERRα as potential ACT-drug combination candidates. Here, we establish that BIRC2 limits antigen presentation and T cell recognition of tumor cells by suppressing IRF1 activity and provide evidence that BIRC2 inhibition in combination with ACT is an effective strategy to increase efficacy.


Subject(s)
Neoplasms , T-Lymphocytes , Antigen Presentation , CRISPR-Cas Systems/genetics , Humans , Neoplasms/genetics , Oncogenes , Systems Analysis
2.
Cancer Cell ; 37(6): 818-833.e9, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32516591

ABSTRACT

T cells are central to all currently effective cancer immunotherapies, but the characteristics defining therapeutically effective anti-tumor T cells have not been comprehensively elucidated. Here, we delineate four phenotypic qualities of effective anti-tumor T cells: cell expansion, differentiation, oxidative stress, and genomic stress. Using a CRISPR-Cas9-based genetic screen of primary T cells we measured the multi-phenotypic impact of disrupting 25 T cell receptor-driven kinases. We identified p38 kinase as a central regulator of all four phenotypes and uncovered transcriptional and antioxidant pathways regulated by p38 in T cells. Pharmacological inhibition of p38 improved the efficacy of mouse anti-tumor T cells and enhanced the functionalities of human tumor-reactive and gene-engineered T cells, paving the way for clinically relevant interventions.


Subject(s)
Breast Neoplasms/therapy , CRISPR-Cas Systems , Immunotherapy, Adoptive/methods , Melanoma, Experimental/therapy , Phenotype , T-Lymphocytes/transplantation , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Differentiation , Female , Genetic Engineering , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/immunology , p38 Mitogen-Activated Protein Kinases/genetics
3.
J Exp Med ; 216(11): 2619-2634, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31405895

ABSTRACT

Host conditioning has emerged as an important component of effective adoptive cell transfer-based immunotherapy for cancer. High levels of IL-1ß are induced by host conditioning, but its impact on the antitumor function of T cells remains unclear. We found that the administration of IL-1ß increased the population size and functionality of adoptively transferred T cells within the tumor. Most importantly, IL-1ß enhanced the ability of tumor-specific T cells to trigger the regression of large, established B16 melanoma tumors in mice. Mechanistically, we showed that the increase in T cell numbers was associated with superior tissue homing and survival abilities and was largely mediated by IL-1ß-stimulated host cells. In addition, IL-1ß enhanced T cell functionality indirectly via its actions on radio-resistant host cells in an IL-2- and IL-15-dependent manner. Our findings not only underscore the potential of provoking inflammation to enhance antitumor immunity but also uncover novel host regulations of T cell responses.


Subject(s)
Immunotherapy, Adoptive/methods , Interleukin-1beta/immunology , Lymphocyte Activation/immunology , Melanoma, Experimental/therapy , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Cell Line, Tumor , Cytokines/blood , Interleukin-1beta/metabolism , Interleukin-6/blood , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Tumor Necrosis Factor-alpha/blood
4.
Science ; 363(6434)2019 03 29.
Article in English | MEDLINE | ID: mdl-30923193

ABSTRACT

A paradox of tumor immunology is that tumor-infiltrating lymphocytes are dysfunctional in situ, yet are capable of stem cell-like behavior including self-renewal, expansion, and multipotency, resulting in the eradication of large metastatic tumors. We find that the overabundance of potassium in the tumor microenvironment underlies this dichotomy, triggering suppression of T cell effector function while preserving stemness. High levels of extracellular potassium constrain T cell effector programs by limiting nutrient uptake, thereby inducing autophagy and reduction of histone acetylation at effector and exhaustion loci, which in turn produces CD8+ T cells with improved in vivo persistence, multipotency, and tumor clearance. This mechanistic knowledge advances our understanding of T cell dysfunction and may lead to novel approaches that enable the development of enhanced T cell strategies for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immune Tolerance , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , Potassium/metabolism , Stem Cells/immunology , Acetyl Coenzyme A/metabolism , Acetylation , Animals , Autophagy/immunology , Caloric Restriction , Cell Differentiation/genetics , Epigenesis, Genetic , Histones/metabolism , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Tumor Microenvironment
5.
JCI Insight ; 2(23)2017 12 07.
Article in English | MEDLINE | ID: mdl-29212954

ABSTRACT

Adoptive immunotherapies using T cells genetically redirected with a chimeric antigen receptor (CAR) or T cell receptor (TCR) are entering mainstream clinical practice. Despite encouraging results, some patients do not respond to current therapies. In part, this phenomenon has been associated with infusion of reduced numbers of early memory T cells. Herein, we report that AKT signaling inhibition is compatible with CAR and TCR retroviral transduction of human T cells while promoting a CD62L-expressing central memory phenotype. Critically, this intervention did not compromise cell yield. Mechanistically, disruption of AKT signaling preserved MAPK activation and promoted the intranuclear localization of FOXO1, a transcriptional regulator of T cell memory. Consequently, AKT signaling inhibition synchronized the transcriptional profile for FOXO1-dependent target genes across multiple donors. Expression of an AKT-resistant FOXO1 mutant phenocopied the influence of AKT signaling inhibition, while addition of AKT signaling inhibition to T cells expressing mutant FOXO1 failed to further augment the frequency of CD62L-expressing cells. Finally, treatment of established B cell acute lymphoblastic leukemia was superior using anti-CD19 CAR-modified T cells transduced and expanded in the presence of an AKT inhibitor compared with conventionally grown T cells. Thus, inhibition of signaling along the PI3K/AKT axis represents a generalizable strategy to generate large numbers of receptor-modified T cells with an early memory phenotype and superior antitumor efficacy.


Subject(s)
Immunotherapy, Adoptive/methods , Proto-Oncogene Proteins c-akt/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocyte Subsets/immunology , Tissue Engineering/methods , Animals , Cell Differentiation , Female , Forkhead Box Protein O1/metabolism , Gene Expression Regulation/immunology , Humans , Immunologic Memory , L-Selectin/metabolism , Lymphocyte Activation/immunology , Mice, Inbred NOD , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/immunology , Transduction, Genetic/methods , Xenograft Model Antitumor Assays
6.
Nature ; 548(7669): 537-542, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28783722

ABSTRACT

Somatic gene mutations can alter the vulnerability of cancer cells to T-cell-based immunotherapies. Here we perturbed genes in human melanoma cells to mimic loss-of-function mutations involved in resistance to these therapies, by using a genome-scale CRISPR-Cas9 library that consisted of around 123,000 single-guide RNAs, and profiled genes whose loss in tumour cells impaired the effector function of CD8+ T cells. The genes that were most enriched in the screen have key roles in antigen presentation and interferon-γ signalling, and correlate with cytolytic activity in patient tumours from The Cancer Genome Atlas. Among the genes validated using different cancer cell lines and antigens, we identified multiple loss-of-function mutations in APLNR, encoding the apelin receptor, in patient tumours that were refractory to immunotherapy. We show that APLNR interacts with JAK1, modulating interferon-γ responses in tumours, and that its functional loss reduces the efficacy of adoptive cell transfer and checkpoint blockade immunotherapies in mouse models. Our results link the loss of essential genes for the effector function of CD8+ T cells with the resistance or non-responsiveness of cancer to immunotherapies.


Subject(s)
Genes, Essential/genetics , Immunotherapy , Neoplasms/genetics , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer , Animals , Antigen Presentation/genetics , Apelin/metabolism , Apelin Receptors/genetics , Apelin Receptors/metabolism , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Female , Genome/genetics , Histocompatibility Antigens Class I/immunology , Humans , Interferon-gamma/immunology , Janus Kinase 1/metabolism , Knowledge Bases , Melanoma/genetics , Melanoma/immunology , Melanoma/metabolism , Melanoma/therapy , Mice , Mutation , Neoplasms/immunology , Neoplasms/metabolism , Reproducibility of Results , T-Lymphocytes, Cytotoxic/metabolism
7.
Cell Metab ; 26(1): 94-109, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28683298

ABSTRACT

Cancer immunotherapy is an increasingly successful strategy for the treatment of patients who have advanced or conventional therapy-resistant cancers. T cells are key mediators of tumor destruction and their specificity for tumor-expressed antigens is of paramount importance, but other T cell-intrinsic qualities, such as durability, longevity, and functionality also play important roles in determining the efficacy of immunotherapy. The cellular energetic pathways that are utilized by T cells play a key role in regulating each of these qualities. Metabolic activity, which both regulates and is regulated by cellular signaling pathways and epigenetics, also profoundly influences the trajectories of T cell differentiation and fate. In this Review, we discuss how cell metabolism influences T cell anti-tumor activity, the metabolic qualities of highly-functional T cells, and strategies to modulate metabolism for improving the immune response to tumors.


Subject(s)
Immunotherapy/methods , Metabolic Networks and Pathways , Neoplasms/metabolism , Neoplasms/therapy , T-Lymphocytes/metabolism , Animals , Cell Differentiation , Cell Survival , Epigenesis, Genetic , Humans , Immunity, Cellular , Neoplasms/genetics , Neoplasms/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tumor Microenvironment
8.
Curr Opin Immunol ; 46: 14-22, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28412583

ABSTRACT

Immunotherapies designed to trigger T cell destruction of tumor cells can result in sustained and complete responses in patients whose cancers were resistant to available treatment options. Evidence suggests that powering the T cell response - how T cells generate energy - plays an important role in their effectiveness. Furthermore the metabolism of T cells can be modulated to improve their anti-cancer activities. In this review, we will discuss the key metabolic properties of anti-cancer T cells, along with potential strategies to enhance immunotherapy through the modulation of T cell metabolism.


Subject(s)
Energy Metabolism , Immunity , Neoplasms/immunology , Neoplasms/metabolism , Animals , Cell Differentiation/immunology , Humans , Immunotherapy/methods , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mitochondria/metabolism , Neoplasms/pathology , Neoplasms/therapy , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome , Tumor Microenvironment/immunology
9.
Cell Metab ; 24(5): 647-648, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27829132

ABSTRACT

A clear role for cellular metabolism in regulating immune cell function has recently been established. In a recent issue of Cell, Geiger et al. (2016) demonstrate a role for intracellular arginine abundance in T cells for the promotion of oxidative metabolism, increased cell viability, persistence, and in vivo antitumor response.


Subject(s)
Arginine , T-Lymphocytes/immunology , Cell Survival
10.
Oncoimmunology ; 5(9): e1014776, 2016.
Article in English | MEDLINE | ID: mdl-27757294

ABSTRACT

Pharmacologic inhibitors of the serine/threonine kinase Akt, initially aimed at deranged oncogenic pathways in tumors, have recently been shown to act as immunomodulators that markedly enhance the antitumor properties of T cells. Repurposing Akt inhibitors to improve antitumor immunity may be viewed as a manifestation of a larger paradigmatic shift in which hallmark characteristics of cancer (e.g., immune evasion), rather than merely causal features (e.g., somatic mutations) can be exploited for therapeutic benefit.

11.
Nature ; 537(7621): 539-543, 2016 09 22.
Article in English | MEDLINE | ID: mdl-27626381

ABSTRACT

Tumours progress despite being infiltrated by tumour-specific effector T cells. Tumours contain areas of cellular necrosis, which are associated with poor survival in a variety of cancers. Here, we show that necrosis releases intracellular potassium ions into the extracellular fluid of mouse and human tumours, causing profound suppression of T cell effector function. Elevation of the extracellular potassium concentration ([K+]e) impairs T cell receptor (TCR)-driven Akt-mTOR phosphorylation and effector programmes. Potassium-mediated suppression of Akt-mTOR signalling and T cell function is dependent upon the activity of the serine/threonine phosphatase PP2A. Although the suppressive effect mediated by elevated [K+]e is independent of changes in plasma membrane potential (Vm), it requires an increase in intracellular potassium ([K+]i). Accordingly, augmenting potassium efflux in tumour-specific T cells by overexpressing the potassium channel Kv1.3 lowers [K+]i and improves effector functions in vitro and in vivo and enhances tumour clearance and survival in melanoma-bearing mice. These results uncover an ionic checkpoint that blocks T cell function in tumours and identify potential new strategies for cancer immunotherapy.


Subject(s)
Cations, Monovalent/metabolism , Melanoma/immunology , Potassium/metabolism , T-Lymphocytes/immunology , Tumor Escape/immunology , Tumor Microenvironment/immunology , Animals , Humans , Immune Tolerance/immunology , Immunotherapy/methods , Kv1.3 Potassium Channel/metabolism , Male , Melanoma/metabolism , Melanoma/pathology , Melanoma/therapy , Membrane Potentials , Mice , Necrosis , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Survival Analysis , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases/metabolism
12.
Cell ; 166(5): 1117-1131.e14, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27565342

ABSTRACT

Cancer cells must evade immune responses at distant sites to establish metastases. The lung is a frequent site for metastasis. We hypothesized that lung-specific immunoregulatory mechanisms create an immunologically permissive environment for tumor colonization. We found that T-cell-intrinsic expression of the oxygen-sensing prolyl-hydroxylase (PHD) proteins is required to maintain local tolerance against innocuous antigens in the lung but powerfully licenses colonization by circulating tumor cells. PHD proteins limit pulmonary type helper (Th)-1 responses, promote CD4(+)-regulatory T (Treg) cell induction, and restrain CD8(+) T cell effector function. Tumor colonization is accompanied by PHD-protein-dependent induction of pulmonary Treg cells and suppression of IFN-γ-dependent tumor clearance. T-cell-intrinsic deletion or pharmacological inhibition of PHD proteins limits tumor colonization of the lung and improves the efficacy of adoptive cell transfer immunotherapy. Collectively, PHD proteins function in T cells to coordinate distinct immunoregulatory programs within the lung that are permissive to cancer metastasis. PAPERCLIP.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung/immunology , Oxygen/metabolism , Prolyl Hydroxylases/metabolism , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/enzymology , Glycolysis/immunology , Interferon-gamma/immunology , Lung/pathology , Lung Neoplasms/therapy , Lymphocyte Activation , Mice , Mice, Knockout , Neoplasm Metastasis , Neuropilin-1/metabolism , Prolyl Hydroxylases/genetics , T-Lymphocytes, Regulatory/enzymology , Th1 Cells/enzymology , Th1 Cells/immunology
13.
Nat Immunol ; 17(7): 851-860, 2016 07.
Article in English | MEDLINE | ID: mdl-27158840

ABSTRACT

T cell antigen receptor (TCR) signaling drives distinct responses depending on the differentiation state and context of CD8(+) T cells. We hypothesized that access of signal-dependent transcription factors (TFs) to enhancers is dynamically regulated to shape transcriptional responses to TCR signaling. We found that the TF BACH2 restrains terminal differentiation to enable generation of long-lived memory cells and protective immunity after viral infection. BACH2 was recruited to enhancers, where it limited expression of TCR-driven genes by attenuating the availability of activator protein-1 (AP-1) sites to Jun family signal-dependent TFs. In naive cells, this prevented TCR-driven induction of genes associated with terminal differentiation. Upon effector differentiation, reduced expression of BACH2 and its phosphorylation enabled unrestrained induction of TCR-driven effector programs.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/physiology , Transcription Factor AP-1/metabolism , Vaccinia virus/immunology , Vaccinia/immunology , Adaptive Immunity , Animals , Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/genetics , Cells, Cultured , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncogene Protein p65(gag-jun) , Signal Transduction/genetics , Transcription Factor AP-1/genetics
14.
J Clin Invest ; 126(2): 599-604, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26731475

ABSTRACT

The immune system has a powerful ability to recognize and kill cancer cells, but its function is often suppressed within tumors, preventing clearance of disease. Functionally diverse innate and adaptive cellular lineages either drive or constrain immune reactions within tumors. The transcription factor (TF) BACH2 regulates the differentiation of multiple innate and adaptive cellular lineages, but its role in controlling tumor immunity has not been elucidated. Here, we demonstrate that BACH2 is required to establish immunosuppression within tumors. Tumor growth was markedly impaired in Bach2-deficient mice and coincided with intratumoral activation of both innate and adaptive immunity. However, augmented tumor clearance in the absence of Bach2 was dependent upon the adaptive immune system. Analysis of tumor-infiltrating lymphocytes from Bach2-deficient mice revealed high frequencies of rapidly proliferating effector CD4+ and CD8+ T cells that expressed the inflammatory cytokine IFN-γ. Effector T cell activation coincided with a reduction in the frequency of intratumoral Foxp3+ Tregs. Mechanistically, BACH2 promoted tumor immunosuppression through Treg-mediated inhibition of intratumoral CD8+ T cells and IFN-γ. These findings demonstrate that BACH2 is a key component of the molecular program of tumor immunosuppression and identify therapeutic targets for the reversal of immunosuppression in cancer.


Subject(s)
Adaptive Immunity , Basic-Leucine Zipper Transcription Factors/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Innate , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/pathology , Interferon-gamma/genetics , Interferon-gamma/immunology , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/pathology , T-Lymphocytes, Regulatory/pathology
15.
J Clin Invest ; 126(1): 318-34, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26657860

ABSTRACT

Adoptive cell transfer (ACT) of purified naive, stem cell memory, and central memory T cell subsets results in superior persistence and antitumor immunity compared with ACT of populations containing more-differentiated effector memory and effector T cells. Despite a clear advantage of the less-differentiated populations, the majority of ACT trials utilize unfractionated T cell subsets. Here, we have challenged the notion that the mere presence of less-differentiated T cells in starting populations used to generate therapeutic T cells is sufficient to convey their desirable attributes. Using both mouse and human cells, we identified a T cell-T cell interaction whereby antigen-experienced subsets directly promote the phenotypic, functional, and metabolic differentiation of naive T cells. This process led to the loss of less-differentiated T cell subsets and resulted in impaired cellular persistence and tumor regression in mouse models following ACT. The T memory-induced conversion of naive T cells was mediated by a nonapoptotic Fas signal, resulting in Akt-driven cellular differentiation. Thus, induction of Fas signaling enhanced T cell differentiation and impaired antitumor immunity, while Fas signaling blockade preserved the antitumor efficacy of naive cells within mixed populations. These findings reveal that T cell subsets can synchronize their differentiation state in a process similar to quorum sensing in unicellular organisms and suggest that disruption of this quorum-like behavior among T cells has potential to enhance T cell-based immunotherapies.


Subject(s)
Immunologic Memory , Immunotherapy, Adoptive , T-Lymphocytes/immunology , Animals , Cell Differentiation , Fas Ligand Protein/physiology , Female , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/physiology , T-Lymphocytes/cytology , fas Receptor/physiology
16.
Cell Metab ; 23(1): 63-76, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26674251

ABSTRACT

Long-term survival and antitumor immunity of adoptively transferred CD8(+) T cells is dependent on their metabolic fitness, but approaches to isolate therapeutic T cells based on metabolic features are not well established. Here we utilized a lipophilic cationic dye tetramethylrhodamine methyl ester (TMRM) to identify and isolate metabolically robust T cells based on their mitochondrial membrane potential (ΔΨm). Comprehensive metabolomic and gene expression profiling demonstrated global features of improved metabolic fitness in low-ΔΨm-sorted CD8(+) T cells. Transfer of these low-ΔΨm T cells was associated with superior long-term in vivo persistence and an enhanced capacity to eradicate established tumors compared with high-ΔΨm cells. Use of ΔΨm-based sorting to enrich for cells with superior metabolic features was observed in CD8(+), CD4(+) T cell subsets, and long-term hematopoietic stem cells. This metabolism-based approach to cell selection may be broadly applicable to therapies involving the transfer of HSC or lymphocytes for the treatment of viral-associated illnesses and cancer.


Subject(s)
Lymphoid Progenitor Cells/physiology , Melanoma, Experimental/therapy , Membrane Potential, Mitochondrial , T-Lymphocyte Subsets/physiology , Animals , CD8-Positive T-Lymphocytes/physiology , Cell Line, Tumor , Cytokines/physiology , Hematopoietic Stem Cells/physiology , Humans , Lymphoid Progenitor Cells/transplantation , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Oxidative Stress , Stem Cell Transplantation , T-Lymphocyte Subsets/transplantation , Transcriptome
17.
Cell Mol Immunol ; 13(4): 502-13, 2016 07.
Article in English | MEDLINE | ID: mdl-25914936

ABSTRACT

To better elucidate epigenetic mechanisms that correlate with the dynamic gene expression program observed upon T-cell differentiation, we investigated the genomic landscape of histone modifications in naive and memory CD8(+) T cells. Using a ChIP-Seq approach coupled with global gene expression profiling, we generated genome-wide histone H3 lysine 4 (H3K4me3) and H3 lysine 27 (H3K27me3) trimethylation maps in naive, T memory stem cells, central memory cells, and effector memory cells in order to gain insight into how histone architecture is remodeled during T cell differentiation. We show that H3K4me3 histone modifications are associated with activation of genes, while H3K27me3 is negatively correlated with gene expression at canonical loci and enhancers associated with T-cell metabolism, effector function, and memory. Our results also reveal histone modifications and gene expression signatures that distinguish the recently identified T memory stem cells from other CD8(+) T-cell subsets. Taken together, our results suggest that CD8(+) lymphocytes undergo chromatin remodeling in a progressive fashion. These findings have major implications for our understanding of peripheral T-cell ontogeny and the formation of immunological memory.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Lineage/genetics , Cell Lineage/immunology , Epigenesis, Genetic , Animals , Chromatin Assembly and Disassembly/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Profiling , Histones/metabolism , Immunologic Memory/genetics , Lymphocyte Subsets/metabolism , Methylation , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Protein Processing, Post-Translational
18.
J Exp Med ; 212(12): 2095-113, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26527801

ABSTRACT

Improving the functional avidity of effector T cells is critical in overcoming inhibitory factors within the tumor microenvironment and eliciting tumor regression. We have found that Cish, a member of the suppressor of cytokine signaling (SOCS) family, is induced by TCR stimulation in CD8(+) T cells and inhibits their functional avidity against tumors. Genetic deletion of Cish in CD8(+) T cells enhances their expansion, functional avidity, and cytokine polyfunctionality, resulting in pronounced and durable regression of established tumors. Although Cish is commonly thought to block STAT5 activation, we found that the primary molecular basis of Cish suppression is through inhibition of TCR signaling. Cish physically interacts with the TCR intermediate PLC-γ1, targeting it for proteasomal degradation after TCR stimulation. These findings establish a novel targetable interaction that regulates the functional avidity of tumor-specific CD8(+) T cells and can be manipulated to improve adoptive cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Melanoma, Experimental/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cells, Cultured , Humans , Immune Tolerance/genetics , Immune Tolerance/immunology , Immunoblotting , Immunotherapy, Adoptive/methods , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , Phospholipase C gamma/immunology , Phospholipase C gamma/metabolism , Protein Binding/immunology , Reverse Transcriptase Polymerase Chain Reaction , Suppressor of Cytokine Signaling Proteins/deficiency , Suppressor of Cytokine Signaling Proteins/genetics , Transcriptome/genetics , Transcriptome/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
19.
Cell ; 162(6): 1206-8, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26359979

ABSTRACT

It is thought that cancer cells engage in Warburg metabolism to meet intrinsic biosynthetic requirements of cell growth and proliferation. Papers by Chang et al. and Ho et al. show that Warburg metabolism enables tumor cells to restrict glucose availability to T cells, suppressing anti-tumor immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Glycolysis , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma/immunology , Melanoma/therapy , Monitoring, Immunologic , Neoplasms/metabolism , Phosphoenolpyruvate/metabolism , Tumor Microenvironment , Animals
20.
Cancer Res ; 75(2): 296-305, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25432172

ABSTRACT

Adoptive cell therapy (ACT) using autologous tumor-infiltrating lymphocytes (TIL) results in complete regression of advanced cancer in some patients, but the efficacy of this potentially curative therapy may be limited by poor persistence of TIL after adoptive transfer. Pharmacologic inhibition of the serine/threonine kinase Akt has recently been shown to promote immunologic memory in virus-specific murine models, but whether this approach enhances features of memory (e.g., long-term persistence) in TIL that are characteristically exhausted and senescent is not established. Here, we show that pharmacologic inhibition of Akt enables expansion of TIL with the transcriptional, metabolic, and functional properties characteristic of memory T cells. Consequently, Akt inhibition results in enhanced persistence of TIL after adoptive transfer into an immunodeficient animal model and augments antitumor immunity of CD8 T cells in a mouse model of cell-based immunotherapy. Pharmacologic inhibition of Akt represents a novel immunometabolomic approach to enhance the persistence of antitumor T cells and improve the efficacy of cell-based immunotherapy for metastatic cancer.


Subject(s)
Immunotherapy, Adoptive/methods , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/therapy , Melanoma/therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Humans , Immunologic Memory , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/pathology , Melanoma/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Proto-Oncogene Proteins c-akt/immunology , Random Allocation , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...