Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Nat Commun ; 14(1): 1459, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36927729

ABSTRACT

There has been considerable scientific effort dedicated to understanding the biologic consequence and therapeutic implications of aberrant tryptophan metabolism in brain tumors and neurodegenerative diseases. A majority of this work has focused on the upstream metabolism of tryptophan; however, this has resulted in limited clinical application. Using global metabolomic profiling of patient-derived brain tumors, we identify the downstream metabolism of tryptophan and accumulation of quinolinate (QA) as a metabolic node in glioblastoma and demonstrate its critical role in promoting immune tolerance. QA acts as a metabolic checkpoint in glioblastoma by inducing NMDA receptor activation and Foxo1/PPARγ signaling in macrophages, resulting in a tumor supportive phenotype. Using a genetically-engineered mouse model designed to inhibit production of QA, we identify kynureninase as a promising therapeutic target to revert the potent immune suppressive microenvironment in glioblastoma. These findings offer an opportunity to revisit the biologic consequence of this pathway as it relates to oncogenesis and neurodegenerative disease and a framework for developing immune modulatory agents to further clinical gains in these otherwise incurable diseases.


Subject(s)
Biological Products , Brain Neoplasms , Glioblastoma , Neurodegenerative Diseases , Mice , Animals , Glioblastoma/genetics , Tryptophan/metabolism , Quinolinic Acid/metabolism , PPAR gamma/metabolism , Neurodegenerative Diseases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Macrophages/metabolism , Brain Neoplasms/pathology , Immune Tolerance , Biological Products/metabolism , Tumor Microenvironment
2.
Cancers (Basel) ; 14(22)2022 Nov 11.
Article in English | MEDLINE | ID: mdl-36428642

ABSTRACT

Glioblastoma (GBM) represents an aggressive and immune-resistant cancer. Preclinical investigations have identified anti-tumor activity of a ketogenic diet (KD) potentially being used to target GBM's glycolytic phenotype. Since immune cells in the microenvironment have a similar reliance upon nutrients to perform their individual functions, we sought to determine if KD influenced the immune landscape of GBM. Consistent with previous publications, KD improved survival in GBM in an immune-competent murine model. Immunophenotyping of tumors identified KD-influenced macrophage polarization, with a paradoxical 50% increase in immune-suppressive M2-like-macrophages and a decrease in pro-inflammatory M1-like-macrophages. We recapitulated KD in vitro using a modified cell culture based on metabolomic profiling of serum in KD-fed mice, mechanistically linking the observed changes in macrophage polarization to PPARγ-activation. We hypothesized that parallel increases in M2-macrophage polarization tempered the therapeutic benefit of KD in GBM. To test this, we performed investigations combining KD with the CSF-1R inhibitor (BLZ945), which influences macrophage polarization. The combination demonstrated a striking improvement in survival and correlative studies confirmed BLZ945 normalized KD-induced changes in macrophage polarization. Overall, KD demonstrates antitumor activity in GBM; however, its efficacy is attenuated by promoting an immunosuppressive phenotype in macrophages. Combinatorial strategies designed to modulate macrophage polarization represent a rational approach to improve the anti-tumor activity of KD in GBM.

3.
In Vivo ; 35(1): 119-129, 2021.
Article in English | MEDLINE | ID: mdl-33402457

ABSTRACT

AIM: To use inhibition of colony-stimulating factor-1 receptor (CSF-1R) to target tumor-associated macrophages (TAMs) and improve the efficacy of radiotherapy in glioblastoma (GBM). MATERIALS AND METHODS: The CSF-1R inhibitor BLZ-945 was used to examine the impact of CSF-1R inhibition on M2 polarization in vitro. Using an orthotopic, immunocompetent GBM model, mice were treated with vehicle, RT, BLZ-945, or RT plus BLZ-945. RESULTS: BLZ-945 reduced M2 polarization in vitro. BLZ-945 alone did not improve median overall survival (mOS=29 days) compared to control mice (mOS=27 days). RT improved survival (mOS=45 days; p=0.02), while RT plus BLZ-945 led to the longest survival (mOS=not reached; p=0.005). Resected tumors had a relatively large population of M2 TAMs in GBM at baseline, which was increased in response to RT. BLZ-945 reduced RT-induced M2 infiltration. CONCLUSION: Inhibition of CSF-1R improved response to RT in the treatment of GBM and may represent a promising strategy to improve RT-induced antitumor immune responses.


Subject(s)
Glioblastoma , Animals , Colony-Stimulating Factors , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Macrophages , Mice , Receptor Protein-Tyrosine Kinases
4.
Mol Cancer Ther ; 19(7): 1415-1422, 2020 07.
Article in English | MEDLINE | ID: mdl-32430486

ABSTRACT

Glioblastoma is the most common primary malignant brain tumor in adults. Despite aggressive treatment, outcomes remain poor with few long-term survivors. Therefore, considerable effort is being made to identify novel therapies for this malignancy. Targeting tumor metabolism represents a promising therapeutic strategy and activation of fatty acid oxidation (FAO) has been identified as a central metabolic node contributing toward gliomagenesis. Perhexiline is a compound with a long clinical track record in angina treatment and commonly described as an FAO inhibitor. We therefore sought to determine whether this compound might be repurposed to serve as a novel therapy in glioblastoma. Perhexiline demonstrated potent in vitro cytotoxicity, induction of redox stress and apoptosis in a panel of glioblastoma cell lines. However, the antitumor activity of perhexiline was distinct when compared with the established FAO inhibitor etomoxir. By evaluating mitochondrial respiration and lipid dynamics in glioblastoma cells following treatment with perhexiline, we confirmed this compound did not inhibit FAO in our models. Using in silico approaches, we identified FYN as a probable target of perhexiline and validated the role of this protein in perhexiline sensitivity. We extended studies to patient samples, validating the potential of FYN to serve as therapeutic target in glioma. When evaluated in vivo, perhexiline demonstrated the capacity to cross the blood-brain barrier and antitumor activity in both flank and orthotopic glioblastoma models. Collectively, we identified potent FYN-dependent antitumor activity of perhexiline in glioblastoma, thereby, representing a promising agent to be repurposed for the treatment of this devastating malignancy.


Subject(s)
Brain Neoplasms/drug therapy , Calcium Channel Blockers/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/drug therapy , Perhexiline/pharmacology , Proto-Oncogene Proteins c-fyn/metabolism , Animals , Apoptosis , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Oxidation-Reduction , Proto-Oncogene Proteins c-fyn/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Cell Death Dis ; 11(4): 253, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32312953

ABSTRACT

Despite advances in molecularly characterizing glioblastoma (GBM), metabolic alterations driving its aggressive phenotype are only beginning to be recognized. Integrative cross-platform analysis coupling global metabolomic and gene expression profiling on patient-derived glioma identified fatty acid ß-oxidation (FAO) as a metabolic node in GBM. We determined that the biologic consequence of enhanced FAO is directly dependent upon tumor microenvironment. FAO serves as a metabolic cue to drive proliferation in a ß-HB/GPR109A dependent autocrine manner in nutrient favorable conditions, while providing an efficient, alternate source of ATP only in nutrient unfavorable conditions. Rational combinatorial strategies designed to target these dynamic roles FAO plays in gliomagenesis resulted in necroptosis-mediated metabolic synthetic lethality in GBM. In summary, we identified FAO as a dominant metabolic node in GBM that provides metabolic plasticity, allowing these cells to adapt to their dynamic microenvironment. Combinatorial strategies designed to target these diverse roles FAO plays in gliomagenesis offers therapeutic potential in GBM.


Subject(s)
Brain Neoplasms/metabolism , Cell Plasticity/physiology , Fatty Acids/metabolism , Glioblastoma/metabolism , Tumor Microenvironment/physiology , Brain Neoplasms/pathology , Cell Line, Tumor , Gene Expression Profiling/methods , Glioblastoma/pathology , Humans , Metabolomics/methods , Oxidation-Reduction , Phenotype , Tumor Microenvironment/genetics
6.
Cancer Immunol Immunother ; 68(7): 1107-1120, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31119318

ABSTRACT

Glioblastoma (GBM) is one of the most aggressive tumors. Numerous studies in the field of immunotherapy have focused their efforts on identifying various pathways linked with tumor-induced immunosuppression. Recent research has demonstrated that metabolic reprogramming in a tumor can contribute towards immune tolerance. To begin to understand the interface between metabolic remodeling and the immune-suppressive state in GBM, we performed a focused, integrative analysis coupling metabolomics with gene-expression profiling in patient-derived GBM (n = 80) and compared them to low-grade astrocytoma (LGA; n = 28). Metabolic intermediates of tryptophan, arginine, prostaglandin, and adenosine emerged as immuno-metabolic nodes in GBM specific to the mesenchymal and classical molecular subtypes of GBM. Integrative analyses emphasized the importance of downstream metabolism of several of these metabolic pathways in GBM. Using CIBERSORT to analyze immune components from the transcriptional profiles of individual tumors, we demonstrated that tryptophan and adenosine metabolism resulted in an accumulation of Tregs and M2 macrophages, respectively, and was recapitulated in mouse models. Furthermore, we extended these findings to preclinical models to determine their potential utility in defining the biologic and/or immunologic consequences of the identified metabolic programs. Collectively, through integrative analysis, we uncovered multifaceted ways by which metabolic reprogramming may contribute towards immune tolerance in GBM, providing the framework for further investigations designed to determine the specific immunologic consequence of these metabolic programs and their therapeutic potential.


Subject(s)
Brain Neoplasms/immunology , Glioblastoma/immunology , Metabolic Networks and Pathways/immunology , Metabolome/immunology , Adenosine/metabolism , Adult , Animals , Brain/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Gene Expression Profiling , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Macrophages/immunology , Macrophages/metabolism , Metabolomics/methods , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tryptophan/metabolism
7.
J Biol Chem ; 294(23): 9198-9212, 2019 06 07.
Article in English | MEDLINE | ID: mdl-30971427

ABSTRACT

Adoptive transfer of tumor epitope-reactive T cells has emerged as a promising strategy to control tumor growth. However, chronically-stimulated T cells expanded for adoptive cell transfer are susceptible to cell death in an oxidative tumor microenvironment. Because oxidation of cell-surface thiols also alters protein functionality, we hypothesized that increasing the levels of thioredoxin (Trx), an antioxidant molecule facilitating reduction of proteins through cysteine thiol-disulfide exchange, in T cells will promote their sustained antitumor function. Using pre-melanosome protein (Pmel)-Trx1 transgenic mouse-derived splenic T cells, flow cytometry, and gene expression analysis, we observed here that higher Trx expression inversely correlated with reactive oxygen species and susceptibility to T-cell receptor restimulation or oxidation-mediated cell death. These Trx1-overexpressing T cells exhibited a cluster of differentiation 62Lhi (CD62Lhi) central memory-like phenotype with reduced glucose uptake (2-NBDGlo) and decreased effector function (interferon γlo). Furthermore, culturing tumor-reactive T cells in the presence of recombinant Trx increased the dependence of T cells on mitochondrial metabolism and improved tumor control. We conclude that strategies for increasing the antioxidant capacity of antitumor T cells modulate their immunometabolic phenotype leading to improved immunotherapeutic control of established tumors.


Subject(s)
T-Lymphocytes/metabolism , Thioredoxins/metabolism , Animals , Antioxidants/metabolism , Cell Line, Tumor , Glucose Transporter Type 1/metabolism , L-Selectin/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Oxidative Stress , Phenotype , Reactive Oxygen Species/metabolism , Receptors, Antigen, T-Cell/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thioredoxins/genetics , Tumor Microenvironment , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/metabolism
8.
Neuro Oncol ; 21(3): 337-347, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30476237

ABSTRACT

BACKGROUND: Although considerable progress has been made in understanding molecular alterations driving gliomagenesis, the diverse metabolic programs contributing to the aggressive phenotype of glioblastoma remain unclear. The aim of this study was to define and provide molecular context to metabolic reprogramming driving gliomagenesis. METHODS: Integrative cross-platform analyses coupling global metabolomic profiling with genomics in patient-derived glioma (low-grade astrocytoma [LGA; n = 28] and glioblastoma [n = 80]) were performed. Identified programs were then metabolomically, genomically, and functionally evaluated in preclinical models. RESULTS: Clear metabolic programs were identified differentiating LGA from glioblastoma, with aberrant lipid, peptide, and amino acid metabolism representing dominant metabolic nodes associated with malignant transformation. Although the metabolomic profiles of glioblastoma and LGA appeared mutually exclusive, considerable metabolic heterogeneity was observed in glioblastoma. Surprisingly, integrative analyses demonstrated that O6-methylguanine-DNA methyltransferase methylation and isocitrate dehydrogenase mutation status were equally distributed among glioblastoma metabolic profiles. Transcriptional subtypes, on the other hand, tightly clustered by their metabolomic signature, with proneural and mesenchymal tumor profiles being mutually exclusive. Integrating these metabolic phenotypes with gene expression analyses uncovered tightly orchestrated and highly redundant transcriptional programs designed to support the observed metabolic programs by actively importing these biochemical substrates from the microenvironment, contributing to a state of enhanced metabolic heterotrophy. These findings were metabolomically, genomically, and functionally recapitulated in preclinical models. CONCLUSION: Despite disparate molecular pathways driving the progression of glioblastoma, metabolic programs designed to maintain its aggressive phenotype remain conserved. This contributes to a state of enhanced metabolic heterotrophy supporting survival in diverse microenvironments implicit in this malignancy.


Subject(s)
Amino Acids/metabolism , Astrocytoma/metabolism , Brain Neoplasms/metabolism , Carcinogenesis , Glioblastoma/metabolism , Lipid Metabolism , Metabolomics , Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cellular Reprogramming , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Gene Expression Profiling , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Mutation , Neoplasm Grading , Peptides/metabolism , Tumor Suppressor Proteins/genetics
9.
Clin Cancer Res ; 24(15): 3632-3643, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29691296

ABSTRACT

Purpose: Immune checkpoint inhibitors designed to revert tumor-induced immunosuppression have emerged as potent anticancer therapies. Tryptophan metabolism represents an immune checkpoint, and targeting this pathway's rate-limiting enzyme IDO1 is actively being investigated clinically. Here, we studied the intermediary metabolism of tryptophan metabolism in glioblastoma and evaluated the activity of the IDO1 inhibitor GDC-0919, both alone and in combination with radiation (RT).Experimental Design: LC/GC-MS and expression profiling was performed for metabolomic and genomic analyses of patient-derived glioma. Immunocompetent mice were injected orthotopically with genetically engineered murine glioma cells and treated with GDC-0919 alone or combined with RT. Flow cytometry was performed on isolated tumors to determine immune consequences of individual treatments.Results: Integrated cross-platform analyses coupling global metabolomic and gene expression profiling identified aberrant tryptophan metabolism as a metabolic node specific to the mesenchymal and classical subtypes of glioblastoma. GDC-0919 demonstrated potent inhibition of this node and effectively crossed the blood-brain barrier. Although GDC-0919 as a single agent did not demonstrate antitumor activity, it had a strong potential for enhancing RT response in glioblastoma, which was further augmented with a hypofractionated regimen. RT response in glioblastoma involves immune stimulation, reflected by increases in activated and cytotoxic T cells, which was balanced by immune checkpoint reactivation, reflected by an increase in IDO1 expression and regulatory T cells (Treg). GDC-0919 mitigated RT-induced Tregs and enhanced T-cell activation.Conclusions: Tryptophan metabolism represents a metabolic node in glioblastoma, and combining RT with IDO1 inhibition enhances therapeutic response by mitigating RT-induced immunosuppression. Clin Cancer Res; 24(15); 3632-43. ©2018 AACR.


Subject(s)
Cell Cycle Checkpoints/immunology , Enzyme Inhibitors/administration & dosage , Glioblastoma/drug therapy , Imidazoles/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoles/pharmacology , Tryptophan/metabolism , Animals , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/immunology , Glioblastoma/immunology , Glioblastoma/pathology , Glioblastoma/radiotherapy , Humans , Imidazoles/therapeutic use , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Indoles/therapeutic use , Metabolomics , Mice , Radiotherapy/adverse effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tryptophan/antagonists & inhibitors , Xenograft Model Antitumor Assays
10.
J Neurooncol ; 134(3): 531-539, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28567588

ABSTRACT

Radiation therapy plays a central role in the management of glioblastoma. Although primarily thought of as modality to provide local tumor control through DNA damage, the capacity of ionizing radiation to modulate tumor immune response has long been recognized. The recent emergence of clinically active immunotherapies offers exciting potential for harnessing the immune modulatory effects or radiation through combinatorial strategies designed to enhance clinical outcomes. In this Review, we provide background describing the unique immune environment within the central nervous system, how ionizing radiation may modulate the tumor immune response, preclinical and clinical data testing the combination of radiation and immune modulating agents, and highlight some of the current challenges in extending these findings clinically.


Subject(s)
Central Nervous System Neoplasms/therapy , Glioblastoma/therapy , Immunotherapy , Radiotherapy , Animals , Central Nervous System Neoplasms/immunology , Combined Modality Therapy , Glioblastoma/immunology , Humans
11.
Neuro Oncol ; 19(12): 1599-1606, 2017 Nov 29.
Article in English | MEDLINE | ID: mdl-28541485

ABSTRACT

BACKGROUND: Glioblastoma represents an archetypal example of a heterogeneous malignancy. To understand the diverse molecular consequences of this complex tumor ecology, we analyzed RNA-seq data generated from commonly identified intratumoral structures in glioblastoma enriched using laser capture microdissection. METHODS: Raw gene-level values of fragments per kilobase of transcript per million reads mapped and the associated clinical data were acquired from the publicly available Ivy Glioblastoma Atlas Project database and analyzed using MetaboAnalyst (v3.0). The database includes gene expression data generated from multiple structural features commonly identified in glioblastoma enriched by laser capture microdissection. RESULTS: We uncovered a relationship between subtype heterogeneity in glioblastoma and its unique tumor microenvironment, with infiltrating cells harboring a proneural signature while the mesenchymal subtype was enriched in perinecrotic regions. When evaluating the tumors' transcriptional profiles in the context of their derived structural regions, there was a relatively small amount of intertumoral heterogeneity in glioblastoma, with individual regions from different tumors clustering tightly together. Analyzing the transcriptional profiles in the context of evolutionary progression identified unique cellular programs associated with specific phases of gliomagenesis. Mediators of cell signaling and cell cycle progression appear to be critical events driving proliferation in the tumor core, while in addition to a multiplex strategy for promoting angiogenesis and/or an immune-tolerant environment, transformation to perinecrotic zones involved global metabolic alterations. CONCLUSION: These findings suggest that intratumoral heterogeneity in glioblastoma is a conserved, predictable consequence to its complex microenvironment, and combinatorial approaches designed to target these unequivocally present tumor biomes may lead to therapeutic gains.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/pathology , Evolution, Molecular , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , High-Throughput Nucleotide Sequencing/methods , Tumor Microenvironment/genetics , Brain Neoplasms/genetics , Gene Expression Profiling , Glioblastoma/genetics , Humans , Prognosis
12.
Neuro Oncol ; 19(10): 1308-1315, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28541512

ABSTRACT

The fields of tumor metabolism and immune oncology have both independently received considerable attention over the last several years. The majority of research in tumor metabolism has largely focused on the Warburg effect and its resulting biologic consequences, including energy and macromolecule production. However, recent investigations have identified elegant, multifaceted strategies by which alterations in tumor metabolism can also contribute to a potent tolerogenic immune environment. One of the most notable is increased tryptophan metabolism through activation of indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO). However, this pathway represents one of numerous metabolic pathways that may modulate the immune system. For example, metabolites associated with aerobic glycolysis, adenosine, arginine, and prostaglandin metabolism have all been implicated in cancer-mediated immune tolerance and represent attractive therapeutic targets. In this review, we will provide an overview of the emerging interface between these 2 timely areas of cancer research and provide an overview of strategies currently being tested to target these next-generation metabolic immune checkpoints.


Subject(s)
Glioblastoma/immunology , Signal Transduction/immunology , Tryptophan Oxygenase/metabolism , Tryptophan/metabolism , Glioblastoma/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenine/immunology , Kynurenine/metabolism , Tryptophan/immunology , Tryptophan Oxygenase/immunology
13.
Oncotarget ; 7(43): 69371-69383, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27750220

ABSTRACT

Advancements in adoptive cell transfer therapy (ACT) has led to the use of T cells engineered with tumor specific T cell receptors, which after rapid expansion can be obtained in sufficient numbers for treating patients. However, due to massive proliferation these cells are close to replicative senescence, exhibit exhausted phenotype, and also display increased susceptibility to activation induced cell death. We have previously shown that tumor reactive T cells undergo caspase-independent cell death upon TCR restimulation with cognate antigen, which involves reactive oxygen species and c-jun N-terminal kinase. Herein, we show that a large fraction of the human melanoma epitope tyrosinase reactive TCR transduced T cells that exhibit effector memory (TEM) phenotype and undergo programmed necrosis, or necroptosis, upon TCR restimulation. As compared to the T central memory (TCM) subsets, the TEM subset displayed an increased expression of genes involved in necroptotic cell death, and a necrotic phenotype upon TCR restimulation as confirmed by electron microscopy. Higher expression of receptor-interacting kinases (RIPK) that mediate necroptosis was also observed in the TEM fraction. Further, the TEM cells were rescued from undergoing necroptosis when pretreated with necroptotic inhibitor NecroX2 before TCR restimulation. Importantly, NecroX2 pretreated tumor reactive T cells also exhibited better tumor control and increased in vivo persistence when adoptively-transferred to treat subcutaneously established murine melanoma B16-F10. Thus, it is likely that the outcome of ACT could be vastly improved by interfering with the necroptotic cell death pathway in activated tumor reactive T cells used in immunotherapy.


Subject(s)
Apoptosis/immunology , CD8-Positive T-Lymphocytes/transplantation , Immunotherapy, Adoptive/methods , Melanoma, Experimental/therapy , Receptors, Antigen, T-Cell/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , L-Selectin/immunology , L-Selectin/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Necrosis/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
14.
Cancer Res ; 76(18): 5229-5240, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27466285

ABSTRACT

Repetitive stimulation of T-cell receptor (TCR) with cognate antigen results in robust proliferation and expansion of the T cells, and also imprints them with replicative senescence signatures. Our previous studies have shown that life-span and antitumor function of T cells can be enhanced by inhibiting reactive oxygen species (ROS) or intervening with ROS-dependent JNK activation that leads to its activation-induced cell death. Because tumor suppressor protein p53 is also a redox active transcription factor that regulates cellular ROS generation that triggers downstream factor-mediating apoptosis, we determined if p53 levels could influence persistence and function of tumor-reactive T cells. Using h3T TCR transgenic mice, with human tyrosinase epitope-reactive T cells developed on p53 knockout (KO) background, we determined its role in regulating antitumor T-cell function. Our data show that as compared with h3T cells, h3T-p53 KO T cells exhibited enhanced glycolytic commitment that correlated with increased proliferation, IFNγ secretion, cytolytic capacity, expression of stemness gene signature, and decreased TGF-ß signaling. This increased effector function correlated to the improved control of subcutaneously established murine melanoma after adoptive transfer of p53-KO T cells. Pharmacological inhibition of human TCR-transduced T cells using a combination of p53 inhibitors also potentiated the T-cell effector function and improved persistence. Thus, our data highlight the key role of p53 in regulating the tumor-reactive T-cell response and that targeting this pathway could have potential translational significance in adoptive T-cell therapy. Cancer Res; 76(18); 5229-40. ©2016 AACR.


Subject(s)
Melanoma/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Suppressor Protein p53/immunology , Adoptive Transfer , Animals , Disease Models, Animal , Flow Cytometry , Heterografts , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Receptors, Antigen, T-Cell/immunology
15.
Oncoimmunology ; 4(1): e985942, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25949871

ABSTRACT

Improving persistence and sustained function of effector CD8+ T cell response is key for achieving significant tumor control in adoptive T cell immunotherapy protocols. Our recent report shows that high anti-oxidant property is central to potent anti-tumor effector T cells, and directly correlates to CD62Lhi central memory, low glycolytic and low mitochondrial membrane potential phenotype, all of which may be linked and contribute to better tumor control.

16.
Cancer Immunol Immunother ; 64(5): 539-49, 2015 May.
Article in English | MEDLINE | ID: mdl-25676709

ABSTRACT

Mouse CD8(+) T cells conditioned with interleukin (IL)-12 ex vivo mediate the potent regression of established melanoma when transferred into lymphodepleted mice. However, the quantitative and qualitative changes induced by IL-12 in the responding mouse CD8(+) T cells have not been well defined. Moreover, the mechanisms by which IL-12-conditioning impacts human CD8(+) T cells, and how such cells might be expanded prior to infusion into patients is not known. We found that ex vivo IL-12-conditioning of mouse CD8(+) T cells led to a tenfold-100-fold increase in persistence and anti-tumor efficacy upon adoptive transfer into lymphodepleted mice. The enhancing effect of IL-12 was associated with maintenance of functional avidity. Importantly, in the context of ongoing ACT clinical trials, human CD8(+) T cells genetically modified with a tyrosinase-specific T cell receptor (TCR) exhibited significantly enhanced functional activity when conditioned with IL-12 as indicated by heightened granzyme B expression and elevated peptide-specific CD107a degranulation. This effect was sustainable despite the 20 days of in vitro cellular expansion required to expand cells over 1,000-fold allowing adequate cell numbers for administration to cancer patients. Overall, these findings support the efficacy and feasibility of ex vivo IL-12-conditioning of TCR-modified human CD8(+) T cells for adoptive transfer and cancer therapy.


Subject(s)
Adoptive Transfer , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/transplantation , Interleukin-12/pharmacology , Melanoma/therapy , Receptors, Antigen, T-Cell/immunology , Skin Neoplasms/therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation , Granzymes/biosynthesis , Humans , Interleukin-12/immunology , Lymphocyte Depletion , Lysosomal-Associated Membrane Protein 1/metabolism , Melanoma/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin Neoplasms/immunology
17.
Cancer Res ; 74(21): 6048-59, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25205101

ABSTRACT

T cells of the T helper (Th)17 subset offer promise in adoptive T-cell therapy for cancer. However, current protocols for ex vivo programming of Th17 cells, which include TGFß exposure, increase the expression of CD39 and CD73, two cell surface ATP ectonucleotidases that reduce T-cell effector functions and promote immunosuppression. Here, we report that ATP-mediated suppression of IFNγ production by Th17 cells can be overcome by genetic ablation of CD73 or by using IL1ß instead of TGFß to program Th17 cells ex vivo. Th17 cells cultured in IL1ß were also highly polyfunctional, expressing high levels of effector molecules and exhibiting superior short-term control of melanoma in mice, despite reduced stem cell-like properties. TGFß addition at low doses that did not upregulate CD73 expression but induced stemness properties drastically improved the antitumor effects of IL1ß-cultured Th17 cells. Effector properties of IL1ß-dependent Th17 cells were likely related to their high glycolytic capacity, since ex vivo programming in pyruvate impaired glycolysis and antitumor effects. Overall, we show that including TGFß in ex vivo cultures used to program Th17 cells blunts their immunotherapeutic potential and demonstrate how this potential can be more fully realized for adoptive T-cell therapy.


Subject(s)
5'-Nucleotidase/biosynthesis , Immunotherapy, Adoptive , Interleukin-1beta/genetics , Neoplasms/therapy , 5'-Nucleotidase/immunology , Animals , Antigens, CD/biosynthesis , Antigens, CD/immunology , Apyrase/biosynthesis , Apyrase/immunology , Cell Line, Tumor , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/immunology , Gene Expression Regulation, Neoplastic , Humans , Immune Tolerance/immunology , Interleukin-1beta/metabolism , Mice , Neoplasms/immunology , Neoplasms/pathology , Th17 Cells/immunology , Th17 Cells/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
18.
Cancer Res ; 74(21): 6036-6047, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25164014

ABSTRACT

Ex vivo-expanded CD8(+) T cells used for adoptive immunotherapy generally acquire an effector memory-like phenotype (TEM cells). With regard to therapeutic applications, two undesired features of this phenotype in vivo are limited persistence and reduced antitumor efficacy, relative to CD8(+) T cells with a central memory-like phenotype (TCM cells). Furthermore, there is incomplete knowledge about all the differences between TEM and TCM cells that may influence tumor treatment outcomes. Given that TCM cells survive relatively longer in oxidative tumor microenvironments, we investigated the hypothesis that TCM cells possess relatively greater antioxidative capacity than TEM cells. Here, we report that TCM cells exhibit a relative increase compared with TEM cells in the expression of cell surface thiols, a key target of cellular redox controls, along with other antioxidant molecules. Increased expression of redox regulators in TCM cells inversely correlated with the generation of reactive oxygen and nitrogen species, proliferative capacity, and glycolytic enzyme levels. Notably, T-cell receptor-transduced T cells pretreated with thiol donors, such as N-acetyl cysteine or rapamycin, upregulated thiol levels and antioxidant genes. A comparison of antitumor CD8(+) T-cell populations on the basis of surface thiol expression showed that thiol-high cells persisted longer in vivo and exerted superior tumor control. Our results suggest that higher levels of reduced cell surface thiols are a key characteristic of T cells that can control tumor growth and that profiling this biomarker may have benefits to adoptive T-cell immunotherapy protocols.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Immunotherapy, Adoptive , Neoplasms/immunology , Sulfhydryl Compounds/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell- and Tissue-Based Therapy , Humans , Immunologic Memory/immunology , Lymphocyte Activation , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology
19.
PLoS One ; 9(2): e89392, 2014.
Article in English | MEDLINE | ID: mdl-24586745

ABSTRACT

We have recently demonstrated the characterization of human tyrosinase TCR bearing h3T-A2 transgenic mouse model, which exhibits spontaneous autoimmune vitiligo and retinal dysfunction. The purpose of current study was to determine the role of T cells and IFN-γ in retina dysfunction and retinal ganglion cell (RGC) death using this model. RGC function was measured by pattern electroretinograms (ERGs) in response to contrast reversal of patterned visual stimuli. RGCs were visualized by fluorogold retrograde-labeling. Expression of CD3, IFN-γ, GFAP, and caspases was measured by immunohistochemistry and Western blotting. All functional and structural changes were measured in 12-month-old h3T-A2 mice and compared with age-matched HLA-A2 wild-type mice. Both pattern-ERGs (42%, p = 0.03) and RGC numbers (37%, p = 0.0001) were reduced in h3T-A2 mice when compared with wild-type mice. The level of CD3 expression was increased in h3T-A2 mice (h3T-A2: 174 ± 27% vs. HLA-A2: 100%; p = 0.04). The levels of effector cytokine IFN-γ were also increased significantly in h3T-A2 mice (h3T-A2: 189 ± 11% vs. HLA-A2: 100%; p = 0.023). Both CD3 and IFN-γ immunostaining were increased in nerve fiber (NF) and RGC layers of h3T-A2 mice. In addition, we have seen a robust increase in GFAP staining in h3T-A2 mice (mainly localized to NF layer), which was substantially reduced in IFN-γ ((-/-)) knockout h3T-A2 mice. We also have seen an up-regulation of caspase-3 and -9 in h3T-A2 mice. Based on our data we conclude that h3T-A2 transgenic mice exhibit visual defects that are mostly associated with the inner retinal layers and RGC function. This novel h3T-A2 transgenic mouse model provides opportunity to understand RGC pathology and test neuroprotective strategies to rescue RGCs.


Subject(s)
Interferon-gamma/metabolism , Monophenol Monooxygenase/metabolism , Receptors, Antigen, T-Cell/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Interferon-gamma/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monophenol Monooxygenase/genetics , Receptors, Antigen, T-Cell/genetics , Retina/cytology , Retina/metabolism
20.
Antioxid Redox Signal ; 18(12): 1497-534, 2013 Apr 20.
Article in English | MEDLINE | ID: mdl-22938635

ABSTRACT

Reactive oxygen species (ROS) are thought to have effects on T-cell function and proliferation. Low concentrations of ROS in T cells are a prerequisite for cell survival, and increased ROS accumulation can lead to apoptosis/necrosis. The cellular redox state of a T cell can also affect T-cell receptor signaling, skewing the immune response. Various T-cell subsets have different redox statuses, and this differential ROS susceptibility could modulate the outcome of an immune response in various disease states. Recent advances in T-cell redox signaling reveal that ROS modulate signaling cascades such as the mitogen-activated protein kinase, phosphoinositide 3-kinase (PI3K)/AKT, and JAK/STAT pathways. Also, tumor microenvironments, chronic T-cell stimulation leading to replicative senescence, gender, and age affect T-cell susceptibility to ROS, thereby contributing to diverse immune outcomes. Antioxidants such as glutathione, thioredoxin, superoxide dismutase, and catalase balance cellular oxidative stress. T-cell redox states are also regulated by expression of various vitamins and dietary compounds. Changes in T-cell redox regulation may affect the pathogenesis of various human diseases. Many strategies to control oxidative stress have been employed for various diseases, including the use of active antioxidants from dietary products and pharmacologic or genetic engineering of antioxidant genes in T cells. Here, we discuss the existence of a complex web of molecules/factors that exogenously or endogenously affect oxidants, and we relate these molecules to potential therapeutics.


Subject(s)
Oxidative Stress , T-Lymphocytes/physiology , Animals , Autoimmune Diseases/metabolism , Cardiovascular Diseases/metabolism , Gene Expression Regulation , Health , Humans , Inflammation/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/physiology , Neoplasms/metabolism , Oxidation-Reduction , Parkinson Disease/metabolism , Skin Diseases/metabolism , Transcription Factors/physiology , Virus Diseases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...