Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Nat Immunol ; 22(6): 746-756, 2021 06.
Article in English | MEDLINE | ID: mdl-34031618

ABSTRACT

T cell exhaustion presents one of the major hurdles to cancer immunotherapy. Among exhausted CD8+ tumor-infiltrating lymphocytes, the terminally exhausted subset contributes directly to tumor cell killing owing to its cytotoxic effector function. However, this subset does not respond to immune checkpoint blockades and is difficult to be reinvigorated with restored proliferative capacity. Here, we show that a half-life-extended interleukin-10-Fc fusion protein directly and potently enhanced expansion and effector function of terminally exhausted CD8+ tumor-infiltrating lymphocytes by promoting oxidative phosphorylation, a process that was independent of the progenitor exhausted T cells. Interleukin-10-Fc was a safe and highly efficient metabolic intervention that synergized with adoptive T cell transfer immunotherapy, leading to eradication of established solid tumors and durable cures in the majority of treated mice. These findings show that metabolic reprogramming by upregulating mitochondrial pyruvate carrier-dependent oxidative phosphorylation can revitalize terminally exhausted T cells and enhance the response to cancer immunotherapy.


Subject(s)
Immunotherapy, Adoptive/methods , Interleukin-10/pharmacology , Neoplasms/therapy , Oxidative Phosphorylation/drug effects , T-Lymphocytes, Cytotoxic/drug effects , Animals , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Cell Line, Tumor , Combined Modality Therapy/methods , Disease Models, Animal , Drug Synergism , Female , HEK293 Cells , Half-Life , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin Fc Fragments/therapeutic use , Interleukin-10/therapeutic use , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Neoplasms/immunology , Neoplasms/pathology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Receptors, Interleukin-10/metabolism , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
2.
Nat Immunol ; 20(2): 206-217, 2019 02.
Article in English | MEDLINE | ID: mdl-30664764

ABSTRACT

Immune checkpoint blockade therapy has shifted the paradigm for cancer treatment. However, the majority of patients lack effective responses due to insufficient T cell infiltration in tumors. Here we show that expression of mitochondrial uncoupling protein 2 (UCP2) in tumor cells determines the immunostimulatory feature of the tumor microenvironment (TME) and is positively associated with prolonged survival. UCP2 reprograms the immune state of the TME by altering its cytokine milieu in an interferon regulatory factor 5-dependent manner. Consequently, UCP2 boosts the conventional type 1 dendritic cell- and CD8+ T cell-dependent anti-tumor immune cycle and normalizes the tumor vasculature. Finally we show, using either a genetic or pharmacological approach, that induction of UCP2 sensitizes melanomas to programmed cell death protein-1 blockade treatment and elicits effective anti-tumor responses. Together, this study demonstrates that targeting the UCP2 pathway is a potent strategy for alleviating the immunosuppressive TME and overcoming the primary resistance of programmed cell death protein-1 blockade.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Melanoma, Experimental/immunology , Skin Neoplasms/immunology , Tumor Microenvironment/immunology , Uncoupling Protein 2/immunology , Animals , Antineoplastic Agents, Immunological/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Drug Resistance, Neoplasm/immunology , Female , Humans , Immunotherapy/methods , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Melanoma, Experimental/mortality , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Skin Neoplasms/blood supply , Skin Neoplasms/drug therapy , Skin Neoplasms/mortality , Survival Analysis , Treatment Outcome , Uncoupling Protein 2/genetics , Uncoupling Protein 2/metabolism
3.
Nat Immunol ; 20(4): 515-516, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30862953

ABSTRACT

In the version of this article initially published, the bars were not aligned with the data points or horizontal axis labels in Fig. 5d, and the labels along each horizontal axis of Fig. 5j-l indicating the presence (+) or absence (-) of doxycycline (Dox) were incorrectly included with the labels below that axis. Also, the right vertical bar above Fig. 7b linking 'P = 0.0001' to the key was incorrect; the correct comparison is αPD-1 versus Dox + αPD-1. Similarly, the right vertical bar above Fig. 7e linking 'P = 0.0002' to the key was incorrect; the correct comparison is αPD-1 versus Rosig + αPD-1. The errors have been corrected in the HTML and PDF versions of the article.

4.
J Immunol ; 193(4): 1759-65, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25015820

ABSTRACT

Invariant NKT (iNKT) cells play critical roles in bridging innate and adaptive immunity. The Raptor containing mTOR complex 1 (mTORC1) has been well documented to control peripheral CD4 or CD8 T cell effector or memory differentiation. However, the role of mTORC1 in iNKT cell development and function remains largely unknown. By using mice with T cell-restricted deletion of Raptor, we show that mTORC1 is selectively required for iNKT but not for conventional T cell development. Indeed, Raptor-deficient iNKT cells are mostly blocked at thymic stage 1-2, resulting in a dramatic decrease of terminal differentiation into stage 3 and severe reduction of peripheral iNKT cells. Moreover, residual iNKT cells in Raptor knockout mice are impaired in their rapid cytokine production upon αGalcer challenge. Bone marrow chimera studies demonstrate that mTORC1 controls iNKT differentiation in a cell-intrinsic manner. Collectively, our data provide the genetic evidence that iNKT cell development and effector functions are under the control of mTORC1 signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Differentiation/immunology , Multiprotein Complexes/genetics , Natural Killer T-Cells/cytology , TOR Serine-Threonine Kinases/genetics , Animals , Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , CD8-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Immunologic Memory , Interferon-gamma/biosynthesis , Lectins, C-Type/biosynthesis , Lymphocyte Activation/immunology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Regulatory-Associated Protein of mTOR , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/biosynthesis
5.
Cell Metab ; 34(5): 731-746.e9, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35452600

ABSTRACT

Glycolysis, including both lactate fermentation and pyruvate oxidation, orchestrates CD8+ T cell differentiation. However, how mitochondrial pyruvate metabolism and uptake controlled by the mitochondrial pyruvate carrier (MPC) impact T cell function and fate remains elusive. We found that genetic deletion of MPC drives CD8+ T cell differentiation toward a memory phenotype. Metabolic flexibility induced by MPC inhibition facilitated acetyl-coenzyme-A production by glutamine and fatty acid oxidation that results in enhanced histone acetylation and chromatin accessibility on pro-memory genes. However, in the tumor microenvironment, MPC is essential for sustaining lactate oxidation to support CD8+ T cell antitumor function. We further revealed that chimeric antigen receptor (CAR) T cell manufacturing with an MPC inhibitor imprinted a memory phenotype and demonstrated that infusing MPC inhibitor-conditioned CAR T cells resulted in superior and long-lasting antitumor activity. Altogether, we uncover that mitochondrial pyruvate uptake instructs metabolic flexibility for guiding T cell differentiation and antitumor responses.


Subject(s)
Memory T Cells , Monocarboxylic Acid Transporters , Lactates , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Pyruvic Acid/metabolism
6.
Cell Mol Immunol ; 18(7): 1761-1771, 2021 07.
Article in English | MEDLINE | ID: mdl-32055005

ABSTRACT

Memory CD8 T cells can provide long-term protection against tumors, which depends on their enhanced proliferative capacity, self-renewal and unique metabolic rewiring to sustain cellular fitness. Specifically, memory CD8 T cells engage oxidative phosphorylation and fatty acid oxidation to fulfill their metabolic demands. In contrast, tumor-infiltrating lymphocytes (TILs) display severe metabolic defects, which may underlie their functional decline. Here, we show that overexpression of proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), the master regulator of mitochondrial biogenesis (MB), favors CD8 T cell central memory formation rather than resident memory generation. PGC-1α-overexpressing CD8 T cells persist and mediate more robust recall responses to bacterial infection or peptide vaccination. Importantly, CD8 T cells with enhanced PGC-1α expression provide stronger antitumor immunity in a mouse melanoma model. Moreover, TILs overexpressing PGC-1α maintain higher mitochondrial activity and improved expansion when rechallenged in a tumor-free host. Altogether, our findings indicate that enforcing mitochondrial biogenesis promotes CD8 T cell memory formation, metabolic fitness, and antitumor immunity in vivo.


Subject(s)
Cancer Vaccines , Animals , CD8-Positive T-Lymphocytes/metabolism , Mice , Mitochondria/metabolism , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Vaccines, Subunit
7.
Front Immunol ; 10: 294, 2019.
Article in English | MEDLINE | ID: mdl-30863405

ABSTRACT

Cross-presenting Xcr1+CD8α DCs are attractive APCs to target for therapeutic cancer vaccines, as they are able to take up and process antigen from dying tumor cells for their MHCI-restricted presentation to CD8 T cells. To this aim, we developed fusion proteins made of the Xcr1 ligand Xcl1 fused to an OVA synthetic long peptide (SLP) and IgG1 Fc fragment. We demonstrated the specific binding and uptake of the Xcl1 fusion proteins by Xcr1+ DCs. Most importantly, their potent adjuvant effect on the H-2Kb/OVA specific T cell response was associated with a sustained tumor control even against the poorly immunogenic B16-OVA melanoma tumor. The increased tumor protection correlated with higher tumor infiltration of antigen-specific CD8+ T cells, increased IFNγ production and degranulation potential. Altogether, these results demonstrate that therapeutic cancer vaccines may be greatly improved by the combination of SLP antigen and Xcl1 fusion proteins.


Subject(s)
Cancer Vaccines/immunology , Chemokines, C/immunology , Dendritic Cells/immunology , Melanoma, Experimental/therapy , Ovalbumin/immunology , Recombinant Fusion Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CHO Cells , Cancer Vaccines/administration & dosage , Chemokines, C/genetics , Chemokines, C/metabolism , Cricetinae , Cricetulus , Dendritic Cells/metabolism , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/genetics , Ovalbumin/metabolism , Peptide Fragments/genetics , Peptide Fragments/immunology , Peptide Fragments/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vaccination/methods , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
8.
J Immunother Cancer ; 6(1): 71, 2018 07 13.
Article in English | MEDLINE | ID: mdl-30005714

ABSTRACT

Adoptive transfer of T cells transduced with Chimeric Antigen Receptors (CAR) are now FDA-approved for the treatment of B-cell malignancies. Yet, the functionality of the endogenous TCR in CART cells has not been fully assessed. Here, we demonstrate that CART cells progressively upregulate Fas, FasL, DR5 and TRAIL, which result in their programmed cell death, independently of antigen-mediated TCR or CAR activation. CART cell apoptosis occurs even when the CAR contains a single (co-)activatory domain such as CD3ζ, CD28 or 4-1BB. Importantly, the dominant role of the Fas and DR5 pathways in CART cell apoptosis is demonstrated by the significant rescue of CART cells upon in vivo blockade by combined Fas-Fc and DR5-Fc recombinant proteins. These observations are of crucial importance for the long-term persistence of CART cells and for the development of new applications including the combined TCR and CAR activation against solid tumors.


Subject(s)
Immunotherapy, Adoptive , Melanoma, Experimental/therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Skin Neoplasms/therapy , fas Receptor/immunology , Animals , Cell Death , Fas Ligand Protein/immunology , Female , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Receptors, Chimeric Antigen/immunology , Skin Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/immunology , Tumor Burden
9.
J Immunother Cancer ; 6(1): 92, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30253808

ABSTRACT

After publication of this article [1], it was noticed that 3 authors were missed from the author list.

10.
Cell Rep ; 14(5): 1206-1217, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26804903

ABSTRACT

Upon infection, antigen-specific naive CD8 T cells are activated and differentiate into short-lived effector cells (SLECs) and memory precursor cells (MPECs). The underlying signaling pathways remain largely unresolved. We show that Rictor, the core component of mammalian target of rapamycin complex 2 (mTORC2), regulates SLEC and MPEC commitment. Rictor deficiency favors memory formation and increases IL-2 secretion capacity without dampening effector functions. Moreover, mTORC2-deficient memory T cells mount more potent recall responses. Enhanced memory formation in the absence of mTORC2 was associated with Eomes and Tcf-1 upregulation, repression of T-bet, enhanced mitochondrial spare respiratory capacity, and fatty acid oxidation. This transcriptional and metabolic reprogramming is mainly driven by nuclear stabilization of Foxo1. Silencing of Foxo1 reversed the increased MPEC differentiation and IL-2 production and led to an impaired recall response of Rictor KO memory T cells. Therefore, mTORC2 is a critical regulator of CD8 T cell differentiation and may be an important target for immunotherapy interventions.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Forkhead Transcription Factors/metabolism , Immunologic Memory , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Nucleus/metabolism , Forkhead Box Protein O1 , Immunologic Memory/genetics , Interleukin-2/biosynthesis , Mechanistic Target of Rapamycin Complex 2 , Mice, Inbred C57BL , Mice, Knockout , Rapamycin-Insensitive Companion of mTOR Protein , T-Box Domain Proteins/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL