Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
2.
Mol Cancer ; 10: 20, 2011 Feb 21.
Article in English | MEDLINE | ID: mdl-21338484

ABSTRACT

BACKGROUND: As well as inducing direct oncolysis, reovirus treatment of melanoma is associated with activation of innate and adaptive anti-tumour immune responses. RESULTS: Here we characterise the effects of conditioned media from reovirus-infected, dying human melanoma cells (reoTCM), in the absence of live virus, to address the immune bystander potential of reovirus therapy. In addition to RANTES, IL-8, MIP-1α and MIP-1ß, reovirus-infected melanoma cells secreted eotaxin, IP-10 and the type 1 interferon IFN-ß. To address the mechanisms responsible for the inflammatory composition of reoTCM, we show that IL-8 and IFN-ß secretion by reovirus-infected melanoma cells was associated with activation of NF-κB and decreased by pre-treatment with small molecule inhibitors of NF-κB and PKR; specific siRNA-mediated knockdown further confirmed a role for PKR. This pro-inflammatory milieu induced a chemotactic response in isolated natural killer (NK) cells, dendritic cells (DC) and anti-melanoma cytotoxic T cells (CTL). Following culture in reoTCM, NK cells upregulated CD69 expression and acquired greater lytic potential against tumour targets. Furthermore, melanoma cell-loaded DC cultured in reoTCM were more effective at priming adaptive anti-tumour immunity. CONCLUSIONS: These data demonstrate that the PKR- and NF-κB-dependent induction of pro-inflammatory molecules that accompanies reovirus-mediated killing can recruit and activate innate and adaptive effector cells, thus potentially altering the tumour microenvironment to support bystander immune-mediated therapy as well as direct viral oncolysis.


Subject(s)
Chemokines/biosynthesis , Immunity/immunology , Inflammation Mediators/metabolism , Melanoma/immunology , NF-kappa B/metabolism , Reoviridae/immunology , eIF-2 Kinase/metabolism , Adaptive Immunity/drug effects , Adaptive Immunity/immunology , Cell Line, Tumor , Chemokines/metabolism , Chemotaxis/drug effects , Culture Media, Conditioned/pharmacology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , Immunity/drug effects , Immunity, Innate/drug effects , Immunity, Innate/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Melanoma/enzymology , Melanoma/metabolism , Melanoma/virology , Phenotype , Reoviridae/drug effects , Reoviridae Infections/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
3.
J Immunol ; 183(7): 4312-21, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19734207

ABSTRACT

Oncolytic virotherapy may mediate antitumor effects via direct oncolysis or immune-mediated tumor regression. Although the ability of oncolytic viruses to generate adaptive antitumor immunity has been characterized, their interactions with the innate immune system are relatively unclear. Using a human in vitro system, this study investigates the innate immunological consequences of reovirus therapy and its potential to activate NK cell-mediated antitumor activity. Dendritic cells (DC) loaded with reovirus-infected human melanoma Mel888 cells (DC-MelReo), but not reovirus-infected tumor cells alone, induced IFN-gamma production within the NK cell population upon coculture with PBMC, in a cell-to-cell contact-dependent manner. DC-MelReo secreted the chemokines CCL2, 3, 4, 5, 7, 8, 11, and CXCL10; these culture supernatants induced NK cell chemotaxis. Coculture of DC-MelReo with purified NK cells induced reciprocal contact-dependent phenotypic DC maturation, while DC-MelReo elicited up-regulation of the activation marker CD69 on NK cells, in a partially contact and partially IL-12 dependent manner. Significantly, DC-MelReo induced NK cell cytotoxicity toward tumor cells by a type I IFN dependent mechanism. These data demonstrate that tumor infection by reovirus can act via DC to induce NK cell recruitment, activation, and cytotoxicity, along with reciprocal DC maturation. These findings suggest that reciprocal DC-NK cell interactions, following reovirus therapy, may play an important role in altering the immune milieu of the tumor microenvironment and mediating tumor regression.


Subject(s)
Cell Communication/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Melanoma, Experimental/immunology , Melanoma, Experimental/prevention & control , Reoviridae/immunology , Cell Line, Tumor , Cells, Cultured , Chemotaxis, Leukocyte/immunology , Coculture Techniques , Cytotoxicity, Immunologic , Dendritic Cells/pathology , Humans , Killer Cells, Natural/pathology , Melanoma, Experimental/virology , Oncolytic Virotherapy/methods , Reoviridae Infections/immunology , Reoviridae Infections/prevention & control , Reoviridae Infections/virology
4.
iScience ; 24(6): 102555, 2021 Jun 25.
Article in English | MEDLINE | ID: mdl-34142056

ABSTRACT

Glycogen synthase kinase-3 (GSK-3) is a positive regulator of PD-1 expression in CD8+ T cells and GSK-3 inhibition enhances T cell function and is effective in the control of tumor growth. GSK-3 has two co-expressed isoforms, GSK-3α and GSK-3ß. Using conditional gene targeting, we demonstrate that both isoforms contribute to T cell function to different degrees. Gsk3b-/- mice suppressed tumor growth to the same degree as Gsk3a/b-/- mice, whereas Gsk3a-/- mice behaved similarly to wild-type, revealing an important role for GSK-3ß in regulating T cell-mediated anti-tumor immunity. The individual GSK-3α and ß isoforms have differential effects on PD-1, IFNγ, and granzyme B expression and operate in synergy to control PD-1 expression and the infiltration of tumors with CD4 and CD8 T cells. Our data reveal a complex interplay of the GSK-3 isoforms in the control of tumor immunity and highlight non-redundant activity of GSK-3 isoforms in T cells, with implications for immunotherapy.

5.
JCI Insight ; 6(9)2021 05 10.
Article in English | MEDLINE | ID: mdl-33822775

ABSTRACT

T cell receptor (TCR) triggering by antigen results in metabolic reprogramming that, in turn, facilitates the exit of T cells from quiescence. The increased nutrient requirements of activated lymphocytes are met, in part, by upregulation of cell surface transporters and enhanced uptake of amino acids, fatty acids, and glucose from the environment. However, the role of intracellular pathways of amino acid biosynthesis in T cell activation is relatively unexplored. Asparagine is a nonessential amino acid that can be synthesized intracellularly through the glutamine-hydrolyzing enzyme asparagine synthetase (ASNS). We set out to define the requirements for uptake of extracellular asparagine and ASNS activity in CD8+ T cell activation. At early time points of activation in vitro, CD8+ T cells expressed little or no ASNS, and, as a consequence, viability and TCR-stimulated growth, activation, and metabolic reprogramming were substantially impaired under conditions of asparagine deprivation. At later time points (more than 24 hours of activation), TCR-induced mTOR-dependent signals resulted in ASNS upregulation that endowed CD8+ T cells with the capacity to function independently of extracellular asparagine. Thus, our data suggest that the coordinated upregulation of ASNS expression and uptake of extracellular asparagine is involved in optimal T cell effector responses.


Subject(s)
Asparagine/metabolism , Aspartate-Ammonia Ligase/metabolism , CD8-Positive T-Lymphocytes/metabolism , Lymphocyte Activation/physiology , Receptors, Antigen, T-Cell/metabolism , Animals , Aspartate-Ammonia Ligase/genetics , Cell Survival , In Vitro Techniques , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction
6.
Cancers (Basel) ; 13(23)2021 11 25.
Article in English | MEDLINE | ID: mdl-34885051

ABSTRACT

BACKGROUND: Previous data on glycogen synthase kinase 3 (GSK-3) inhibition in cancer models support a cytotoxic effect with selectivity for tumor cells compared to normal tissue but the effect of these inhibitors in glioma has not been widely studied. Here, we investigate their potential as cytotoxics in glioma. METHODS: We assessed the effect of pharmacologic GSK-3 inhibition on established (U87, U251) and patient-derived (GBM1, GBM4) glioblastoma (GBM) cell lines using cytotoxicity assays as well as undertaking a detailed investigation of the effect on cell cycle, mitosis, and centrosome biology. We also assessed drug uptake and efficacy of GSK-3 inhibition alone and in combination with radiation in xenograft models. RESULTS: Using the selective GSK-3 inhibitor AZD2858, we demonstrated single agent cytotoxicity in two patient-derived glioma cell lines (GBM1, GBM4) and two established cell lines (U251 and U87) with IC50 in the low micromolar range promoting centrosome disruption, failed mitosis, and S-phase arrest. Glioma xenografts exposed to AZD2858 also showed growth delay compared to untreated controls. Combined treatment with radiation increased the cytotoxic effect of clinical radiation doses in vitro and in orthotopic glioma xenografts. CONCLUSIONS: These data suggest that GSK-3 inhibition promotes cell death in glioma through disrupting centrosome function and promoting mitotic failure and that AZD2858 is an effective adjuvant to radiation at clinical doses.

7.
J Immunol ; 181(5): 3108-15, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18713981

ABSTRACT

In vivo, dendritic cells (DC) are programmed to orchestrate innate and adaptive immunity in response to pathogen-derived "danger" signals. Under particular circumstances, DC can also be directly cytotoxic against tumor cells, potentially allowing them to release tumor associated Ags from dying cells and then prime antitumor immunity against them. In this study, we describe the innate characteristics of DC (OK-DC) generated in vitro after exposure of immature human myeloid-derived DC to OK432, a penicillin-inactivated and lyophilized preparation of Streptococcus pyrogenes. OK-DC produced proinflammatory cytokines, stimulated autologous T cell proliferation and IFN-gamma secretion, expressed CCR7, and migrated in response to MIP-3beta. Moreover, OK-DC displayed strong, specific cytotoxicity toward tumor cell targets. This cytotoxicity was associated with novel, OK432-induced up-regulation of CD40L on the cell surface of OK-DC, and was absolutely dependent on expression of CD40 on the tumor targets. These data demonstrate that maturation of human DC with OK432, an adjuvant suitable for clinical use, induces direct tumor cell killing by DC, and describes a novel CD40/CD40L-mediated mechanism for specific DC antitumor cytotoxicity.


Subject(s)
CD40 Antigens/metabolism , CD40 Ligand/metabolism , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Neoplasms/immunology , Picibanil/pharmacology , CD40 Ligand/genetics , Cell Death , Dendritic Cells/drug effects , Humans , Neoplasms/pathology , Up-Regulation/drug effects
8.
Neuro Oncol ; 20(2): 203-214, 2018 01 22.
Article in English | MEDLINE | ID: mdl-29016820

ABSTRACT

Background: CNS tumors, including medulloblastoma and pediatric glioblastoma (pGBM) account for the majority of solid pediatric malignancies. There remains an unmet need to identify novel treatment approaches in poor prognosis and relapsed pediatric brain tumors, where therapeutic options are limited. Small-molecule B-cell lymphoma 2 (BCL-2) family inhibitors may enhance tumor cell killing when combined with conventional and targeted chemotherapeutic agents. We investigated the effect of disrupting BCL-2 and B cell lymphoma-extra large (BCL-XL) protein function using ABT-263, ABT-199 and WEHI-539 in medulloblastoma and pGBM cells following treatment with MLN8237, an Aurora kinase inhibitor under investigation as a novel agent for the treatment of malignant brain tumors. Methods: Tumor cell growth and viability were determined by MTT/WST-1 assays and flow cytometry. Effects on cell phenotype, cell cycle progression, and ploidy were determined by live cell imaging and DNA content analysis. Apoptosis was determined by annexin V/propidium iodide staining and time-lapse microscopy and confirmed by measuring caspase-3/7 activity and western blotting and by short interfering RNA (siRNA) knockdown of BCL-2 associated X protein/BCL-2 antagonist killer (BAX/BAK). Results: ABT-263, in combination with MLN8237, reduced mitotic slippage and polyploidy and promoted the elimination of mitotically defective cells via a BAX/BAK-dependent, caspase-mediated apoptotic pathway. The BCL-XL antagonist, WEHI-539, significantly augmented tumor cell killing when used in combination with MLN8237, as well as sensitized resistant brain tumor cells to a novel BAX activator, SMBA1. In addition, siRNA-mediated knockdown of BCL-XL sensitized pGBM and medulloblastoma cells to MLN8237 and mimicked the effect of combination drug treatment. Conclusion: Selective small-molecule inhibitors of BCL-XL may enhance the efficacy of MLN8237 and other targeted chemotherapeutic agents.


Subject(s)
Azepines/pharmacology , Glioblastoma/drug therapy , Medulloblastoma/drug therapy , Proto-Oncogene Proteins c-bcl-2/drug effects , Pyrimidines/pharmacology , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cerebellar Neoplasms/drug therapy , Humans
9.
Cancer Immunol Res ; 6(10): 1161-1173, 2018 10.
Article in English | MEDLINE | ID: mdl-30209061

ABSTRACT

Immunotherapy is showing promise for otherwise incurable cancers. Oncolytic viruses (OVs), developed as direct cytotoxic agents, mediate their antitumor effects via activation of the immune system. However, OVs also stimulate antiviral immune responses, including the induction of OV-neutralizing antibodies. Current dogma suggests that the presence of preexisting antiviral neutralizing antibodies in patients, or their development during viral therapy, is a barrier to systemic OV delivery, rendering repeat systemic treatments ineffective. However, we have found that human monocytes loaded with preformed reovirus-antibody complexes, in which the reovirus is fully neutralized, deliver functional replicative reovirus to tumor cells, resulting in tumor cell infection and lysis. This delivery mechanism is mediated, at least in part, by antibody receptors (in particular FcγRIII) that mediate uptake and internalization of the reovirus/antibody complexes by the monocytes. This finding has implications for oncolytic virotherapy and for the design of clinical OV treatment strategies. Cancer Immunol Res; 6(10); 1161-73. ©2018 AACR.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Melanoma, Experimental/therapy , Monocytes/immunology , Oncolytic Virotherapy , Oncolytic Viruses , Reoviridae , Animals , Cell Line , Chlorocebus aethiops , Female , Humans , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Mice, Inbred C57BL , Receptors, IgG/immunology
10.
PLoS One ; 8(5): e64051, 2013.
Article in English | MEDLINE | ID: mdl-23691145

ABSTRACT

Pediatric high grade glioma is refractory to conventional multimodal treatment, highlighting a need to develop novel efficacious therapies. We investigated tumor metabolism as a potential therapeutic target in a panel of diverse pediatric glioma cell lines (SF188, KNS42, UW479 and RES186) using metformin and 2-deoxyglucose. As a single agent, metformin had little effect on cell viability overall. SF188 cells were highly sensitive to 2-deoxyglucose however, combination of metformin with 2-deoxyglucose significantly reduced cell proliferation compared to either drug alone in all cell lines tested. In addition, the combination of the two agents was associated with a rapid decrease in cellular ATP and subsequent AMPK activation. However, increased cell death was only observed in select cell lines after prolonged exposure to the drug combination and was caspase independent. Anti-apoptotic BCL-2 family proteins have been indicated as mediators of resistance against metabolic stress. Therefore we sought to determine whether pharmacological inhibition of BCL-2/BCL-xL with ABT-263 could potentiate apoptosis in response to these agents. We found that ABT-263 increased sensitivity to 2-deoxyglucose and promoted rapid and extensive cell death in response to the combination of 2-deoxyglucose and metformin. Furthermore, cell death was inhibited by the pan-caspase inhibitor, z-VAD-FMK suggesting that ABT-263 potentiated caspase-dependent cell death in response to 2-deoxyglucose or its combination with metformin. Overall, these data provide support for the concept that targeting metabolic and anti-apoptotic pathways may be an effective therapeutic strategy in pediatric glioma.


Subject(s)
Aniline Compounds/pharmacology , Apoptosis/drug effects , Deoxyglucose/pharmacology , Glioma/drug therapy , Metformin/pharmacology , Sulfonamides/pharmacology , Adenosine Triphosphate/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Blotting, Western , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Child , Drug Synergism , Flow Cytometry , Glioma/metabolism , Humans , Immunohistochemistry , Statistics, Nonparametric
11.
Clin Cancer Res ; 15(13): 4374-4381, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19509134

ABSTRACT

PURPOSE: Reovirus is a naturally occurring oncolytic virus in clinical trials. Although tumor infection by reovirus can generate adaptive antitumor immunity, its therapeutic importance versus direct viral oncolysis is undefined. This study addresses the requirement for viral oncolysis and replication, and the relative importance of antitumor immunity and direct oncolysis in therapy. EXPERIMENTAL DESIGN: Nonantigen specific T cells loaded with reovirus were delivered i.v. to C57BL/6 and severe combined immunodeficient mice bearing lymph node and splenic metastases from the murine melanoma, B16ova, with assessment of viral replication, metastatic clearance by tumor colony outgrowth, and immune priming. Human cytotoxic lymphocyte priming assays were done with dendritic cells loaded with Mel888 cells before the addition of reovirus. RESULTS: B16ova was resistant to direct oncolysis in vitro, and failed to support reovirus replication in vitro or in vivo. Nevertheless, reovirus purged lymph node and splenic metastases in C57BL/6 mice and generated antitumor immunity. In contrast, reovirus failed to reduce tumor burden in severe combined immunodeficient mice bearing either B16ova or reovirus-sensitive B16tk metastases. In the human system, reovirus acted solely as an adjuvant when added to dendritic cells already loaded with Mel888, supporting priming of specific antitumor cytotoxic lymphocyte, in the absence of significant direct tumor oncolysis; UV-treated nonreplicating reovirus was similarly immunogenic. CONCLUSION: The immune response is critical in mediating the efficacy of reovirus, and does not depend upon direct viral oncolysis or replication. The findings are of direct relevance to fulfilling the potential of this novel anticancer agent.


Subject(s)
Cytotoxicity, Immunologic/physiology , Mammalian orthoreovirus 3/physiology , Melanoma, Experimental/therapy , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Virus Replication/physiology , Animals , Cell Line, Tumor , Cells, Cultured , Humans , Immunotherapy, Adoptive/methods , Mammalian orthoreovirus 3/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/virology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Oncolytic Viruses/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/transplantation , T-Lymphocytes, Cytotoxic/virology , Treatment Outcome , Virus Replication/immunology
12.
J Immunol ; 180(9): 6018-26, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18424722

ABSTRACT

Oncolytic viruses can exert their antitumor activity via direct oncolysis or activation of antitumor immunity. Although reovirus is currently under clinical investigation for the treatment of localized or disseminated cancer, any potential immune contribution to its efficacy has not been addressed. This is the first study to investigate the ability of reovirus to activate human dendritic cells (DC), key regulators of both innate and adaptive immune responses. Reovirus induced DC maturation and stimulated the production of the proinflammatory cytokines IFN-alpha, TNF-alpha, IL-12p70, and IL-6. Activation of DC by reovirus was not dependent on viral replication, while cytokine production (but not phenotypic maturation) was inhibited by blockade of PKR and NF-kappaB signaling. Upon coculture with autologous NK cells, reovirus-activated DC up-regulated IFN-gamma production and increased NK cytolytic activity. Moreover, short-term coculture of reovirus-activated DC with autologous T cells also enhanced T cell cytokine secretion (IL-2 and IFN-gamma) and induced non-Ag restricted tumor cell killing. These data demonstrate for the first time that reovirus directly activates human DC and that reovirus-activated DC stimulate innate killing by not only NK cells, but also T cells, suggesting a novel potential role for T cells in oncolytic virus-induced local tumor cell death. Hence reovirus recognition by DC may trigger innate effector mechanisms to complement the virus's direct cytotoxicity, potentially enhancing the efficacy of reovirus as a therapeutic agent.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate , Killer Cells, Natural/immunology , Mammalian orthoreovirus 3/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Cell Death/immunology , Cell Line, Tumor , Coculture Techniques , Cytokines/immunology , Dendritic Cells/virology , Humans , Immunity, Cellular , NF-kappa B/immunology , Signal Transduction/immunology , Time Factors , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL