Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cancer Res ; 81(12): 3374-3386, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33837043

ABSTRACT

Screening for sensitizers of cancer cells to TRAIL-mediated apoptosis identified a natural product of the 17ß-hydroxywithanolide (17-BHW) class, physachenolide C (PCC), as a promising hit. In this study, we show that PCC was also able to sensitize melanoma and renal carcinoma cells to apoptosis in response not only to TRAIL, but also to the synthetic polynucleotide poly I:C, a viral mimetic and immune activator, by reducing levels of antiapoptotic proteins cFLIP and Livin. Both death receptor and TLR3 signaling elicited subsequent increased assembly of a proapoptotic ripoptosome signaling complex. Administration of a combination of PCC and poly I:C in human M14 melanoma xenograft and a syngeneic B16 melanoma model provided significant therapeutic benefit as compared with individual agents. In addition, PCC enhanced melanoma cell death in response to activated human T cells in vitro and in vivo in a death ligand-dependent manner. Biochemical mechanism-of-action studies established bromo and extraterminal domain (BET) proteins as major cellular targets of PCC. Thus, by targeting of BET proteins to reduce antiapoptotic proteins and enhance caspase-8-dependent apoptosis of cancer cells, PCC represents a unique agent that can potentially be used in combination with various immunotherapeutic approaches to promote tumor regression and improve outcome. SIGNIFICANCE: These findings demonstrate that PCC selectively sensitizes cancer cells to immune-mediated cell death, potentially improving the efficacy of cancer immunotherapies. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/12/3374/F1.large.jpg.


Subject(s)
Biological Products/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Immunotherapy/methods , Melanoma, Experimental/drug therapy , Poly I-C/pharmacology , Transcription Factors/antagonists & inhibitors , Withanolides/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Cell Proliferation , Drug Therapy, Combination , Female , Humans , Interferon Inducers/pharmacology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Int J Cancer ; 132(4): 785-94, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-22821831

ABSTRACT

MIF is a proinflammatory cytokine and is implicated in cancer. A higher MIF level is found in many human cancer and cancer-prone inflammatory diseases, including chronic pancreatitis and pancreatic cancer. We tested the hypothesis that MIF contributes to pancreatic cancer aggressiveness and predicts disease outcome in resected cases. Consistent with our hypothesis we found that an elevated MIF mRNA expression in tumors was significantly associated with poor outcome in resected cases. Multivariate Cox-regression analysis further showed that MIF is independently associated with patients' survival (HR = 2.26, 95% CI = 1.17-4.37, p = 0.015). Mechanistic analyses revealed that MIF overexpression decreased E-cadherin and increased vimentin mRNA and protein levels in pancreatic cancer cell lines, consistent with the features of epithelial-to-mesenchymal transition (EMT). Furthermore, MIF-overexpression significantly increased ZEB1/2 and decreased miR-200b expression, while shRNA-mediated inhibition of MIF increased E-cadherin and miR-200b expression, and reduced the expression of ZEB1/2 in Panc1 cells. Re-expression of miR-200b in MIF overexpressing cells restored the epithelial characteristics, as indicated by an increase in E-cadherin and decrease in ZEB1/2 and vimentin expression. A reduced sensitivity to the chemotherapeutic drug, gemcitabine, occurred in MIF-overexpressing cells. Indicative of an increased malignant potential, MIF over-expressing cells showed significant increase in their invasion ability in vitro, and tumor growth and metastasis in an orthotopic xenograft mouse model. These results support a role of MIF in disease aggressiveness, indicating its potential usefulness as a candidate target for designing improved treatment in pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Epithelial-Mesenchymal Transition/genetics , Macrophage Migration-Inhibitory Factors/genetics , Pancreatic Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Macrophage Migration-Inhibitory Factors/metabolism , Male , Mice, Nude , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prognosis , RNA Interference , Transplantation, Heterologous , Gemcitabine
3.
Cancer Invest ; 30(5): 343-63, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22571338

ABSTRACT

Using two MYCN transgenic mouse strains, we established 10 transplantable neuroblastoma cell lines via serial orthotopic passage in the adrenal gland. Tissue arrays demonstrate that by histochemistry, vascularity, immunohistochemical staining for neuroblastoma markers, catecholamine analysis, and concurrent cDNA microarray analysis, there is a close correspondence between the transplantable lines and the spontaneous tumors. Several genes closely associated with the pathobiology and immune evasion of neuroblastoma, novel targets that warrant evaluation, and decreased expression of tumor suppressor genes are demonstrated. These studies describe a unique and generalizable approach to expand the utility of transgenic models of spontaneous tumor, providing new tools for preclinical investigation.


Subject(s)
Drug Discovery , Gene Expression Profiling , Neuroblastoma/pathology , Animals , Apoptosis , Cell Line, Tumor , Cyclin-Dependent Kinase 4/analysis , Genes, Tumor Suppressor , High-Throughput Screening Assays , Humans , Mice , Mice, Inbred C57BL , N-Myc Proto-Oncogene Protein , Neoplasm Transplantation , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/ultrastructure , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Principal Component Analysis , Tissue Array Analysis
4.
PLoS One ; 7(3): e33303, 2012.
Article in English | MEDLINE | ID: mdl-22428016

ABSTRACT

The liver is an immunologically unique organ containing tolerogenic dendritic cells (DC) that maintain an immunosuppressive microenvironment. Although systemic IL-12 administration can improve responses to tumors, the effects of IL-12-based treatments on DC, in particular hepatic DC, remain incompletely understood. In this study, we demonstrate systemic IL-12 administration induces a 2-3 fold increase in conventional, but not plasmacytoid, DC subsets in the liver. Following IL-12 administration, hepatic DC became more phenotypically and functionally mature, resembling the function of splenic DC, but differed as compared to their splenic counterparts in the production of IL-12 following co-stimulation with toll-like receptor (TLR) agonists. Hepatic DCs from IL-12 treated mice acquired enhanced T cell proliferative capabilities similar to levels observed using splenic DCs. Furthermore, IL-12 administration preferentially increased hepatic T cell activation and IFNγ expression in the RENCA mouse model of renal cell carcinoma. Collectively, the data shows systemic IL-12 administration enables hepatic DCs to overcome at least some aspects of the inherently suppressive milieu of the hepatic environment that could have important implications for the design of IL-12-based immunotherapeutic strategies targeting hepatic malignancies and infections.


Subject(s)
Carcinoma, Renal Cell/immunology , Dendritic Cells/drug effects , Interleukin-12/pharmacology , Liver/immunology , Animals , Dendritic Cells/immunology , Flow Cytometry , Interferon-gamma/metabolism , Interleukin-12/administration & dosage , Liver/cytology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Statistics, Nonparametric , T-Lymphocytes/immunology
5.
Cancer Res ; 71(7): 2718-27, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21324921

ABSTRACT

Obesity is a risk factor for development of certain cancers but the basis for this risk is unclear. In this study, we developed a novel mouse model that demonstrates directly how lipogenic phenotypes commonly associated with diet-induced metabolic syndromes can influence hepatic cancer development. Activated AKT and ß-catenin (AKT/CAT) genes were hydrodynamically codelivered using the Sleeping Beauty transposon to initiate liver tumorigenesis. AKT/CAT and MET/CAT combination induced microscopic tumor foci by 4 weeks, whereas no tumorigenesis resulted from delivery of AKT, MET, or CAT alone. Primary AKT/CAT tumor cells were steatotic (fatty) hepatocellular adenomas which progressed to hepatocellular carcinomas (HCC) upon in vivo passage, whereas primary MET/CAT tumors emerged directly as frank HCC. Conversion of AKT/CAT tumor cells to frank HCC during passage was associated with induction of the human HCC marker α-fetoprotein and the stem cell marker CD133. Using hierarchical clustering and gene set enrichment analysis, we compared the primary murine AKT/CAT and MET/CAT tumors to a panel of 53 human HCCs and determined that these two mouse models could be stratified as distinct subtypes associated in humans with poor clinical prognosis. The chief molecular networks identified in primary and passaged AKT/CAT tumors were steatosis and lipid metabolic pathways, respectively. Our findings show how coactivation of the AKT and CAT pathways in hepatocytes can efficiently model development of a lipogenic tumor phenotype. Furthermore, we believe that our approach could speed the dissection of microenvironmental factors responsible for driving steatotic-neoplastic transformation to frank carcinoma, through genetic modification of existing immunodefined transgenic models.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic/metabolism , Liver Neoplasms/metabolism , Oncogene Protein v-akt/metabolism , beta Catenin/metabolism , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA Transposable Elements , Enzyme Activation , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Oncogenes , Proto-Oncogene Proteins c-met/metabolism
6.
J Immunol ; 186(2): 838-47, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21148802

ABSTRACT

The fate of invariant NKT (iNKT) cells following activation remains controversial and unclear. We systemically examined how iNKT cells are regulated following TCR-dependent and -independent activation with α-galactosylceramide (αGC) or IL-18 plus IL-12, respectively. Our studies reveal activation by αGC or IL-18 plus IL-12 induced transient depletion of iNKT cells exclusively in the liver that was independent of caspase 3-mediated apoptosis. The loss of iNKT cells was followed by repopulation and expansion of phenotypically distinct cells via different mechanisms. Liver iNKT cell expansion following αGC, but not IL-18 plus IL-12, treatment required an intact spleen and IFN-γ. Additionally, IL-18 plus IL-12 induced a more prolonged expansion of liver iNKT cells compared with αGC. iNKT cells that repopulate the liver following αGC had higher levels of suppressive receptors PD-1 and Lag3, whereas those that repopulate the liver following IL-18 plus IL-12 had increased levels of TCR and ICOS. In contrast to acute treatment that caused a transient loss of iNKT cells, chronic αGC or IL-18 plus IL-12 treatment caused long-term systemic loss requiring an intact thymus for repopulation of the liver. This report reveals a previously undefined role for the liver in the depletion of activated iNKT cells. Additionally, TCR-dependent and -independent activation differentially regulate iNKT cell distribution and phenotype. These results provide new insights for understanding how iNKT cells are systemically regulated following activation.


Subject(s)
Cell Differentiation/immunology , Liver/immunology , Liver/metabolism , Lymphocyte Activation/immunology , Lymphocyte Depletion , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/physiology , Animals , Caspase 3/metabolism , Galactosylceramides/physiology , Immunophenotyping , Interleukin-12/physiology , Interleukin-18/physiology , Liver/cytology , Lymphocyte Depletion/methods , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , Spleen/cytology , Spleen/immunology , Spleen/metabolism
7.
Proc Natl Acad Sci U S A ; 106(46): 19455-60, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19892741

ABSTRACT

Treatment of mice bearing orthotopic, metastatic tumors with anti-CD40 antibody resulted in only partial, transient anti-tumor effects whereas combined treatment with IL-2/anti-CD40, induced tumor regression. The mechanisms for these divergent anti-tumor responses were examined by profiling tumor-infiltrating leukocyte subsets and chemokine expression within the tumor microenvironment after immunotherapy. IL-2/anti-CD40, but not anti-CD40 alone, induced significant infiltration of established tumors by NK and CD8(+) T cells. To further define the role of chemokines in leukocyte recruitment into tumors, we evaluated anti-tumor responses in mice lacking the chemokine receptor, CCR2. The anti-tumor effects and leukocyte recruitment mediated by anti-CD40 alone, were completely abolished in CCR2(-/-) mice. In contrast, IL-2/anti-CD40-mediated leukocyte recruitment and reductions in primary tumors and metastases were maintained in CCR2(-/-) mice. Treatment of mice with IL-2/anti-CD40, but not anti-CD40 alone, also caused an IFN-gamma-dependent increase in the expression of multiple Th1 chemokines within the tumor microenvironment. Interestingly, although IL-2/anti-CD40 treatment increased Tregs in the spleen, it also caused a coincident IFN-gamma-dependent reduction in CD4(+)/FoxP3(+) Tregs, myeloid-derived suppressor cells and Th2 chemokine expression specifically within the tumor microenvironment that was not observed after treatment with anti-CD40 alone. Similar effects were observed using IL-15 in combination with anti-CD40. Taken together, our data demonstrate that IL-2/anti-CD40, but not anti-CD40 alone, can preferentially reduce the overall immunosuppressive milieu within the tumor microenvironment. These results suggest that the use of anti-CD40 in combination with IL-2 or IL-15 may hold substantially more promise for clinical cancer treatment than anti-CD40 alone.


Subject(s)
Antibodies/therapeutic use , CD40 Antigens/agonists , Immunosuppression Therapy/methods , Interleukin-2/therapeutic use , Neoplasms/therapy , Animals , Arginase/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Chemokine CCL5/biosynthesis , Chemokine CXCL9/biosynthesis , Chemokines/biosynthesis , Chemokines, CC/biosynthesis , Drug Synergism , Lymphocytes, Tumor-Infiltrating/immunology , Macrophage Inflammatory Proteins/biosynthesis , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Receptors, CCR2/biosynthesis , Receptors, CCR2/genetics , Receptors, Cytokine/biosynthesis
8.
J Immunol ; 182(7): 4328-38, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19299733

ABSTRACT

IL-27 exerts antitumor activity in murine orthotopic neuroblastoma, but only partial antitumor effect in disseminated disease. This study demonstrates that combined treatment with IL-2 and IL-27 induces potent antitumor activity in disseminated neuroblastoma metastasis. Complete durable tumor regression was achieved in 90% of mice bearing metastatic TBJ-IL-27 tumors treated with IL-2 compared with only 40% of mice bearing TBJ-IL-27 tumors alone and 0% of mice bearing TBJ-FLAG tumors with or without IL-2 treatment. Comparable antitumor effects were achieved by IL-27 protein produced upon hydrodynamic IL-27 plasmid DNA delivery when combined with IL-2. Although delivery of IL-27 alone, or in combination with IL-2, mediated pronounced regression of neuroblastoma metastases in the liver, combined delivery of IL-27 and IL-2 was far more effective than IL-27 alone against bone marrow metastases. Combined exposure to IL-27 produced by tumor and IL-2 synergistically enhances the generation of tumor-specific CTL reactivity. Potentiation of CTL reactivity by IL-27 occurs via mechanisms that appear to be engaged during both the initial sensitization and effector phase. Potent immunologic memory responses are generated in mice cured of their disseminated disease by combined delivery of IL-27 and IL-2, and depletion of CD8(+) ablates the antitumor efficacy of this combination. Moreover, IL-27 delivery can inhibit the expansion of CD4(+)CD25(+)Foxp3(+) regulatory and IL-17-expressing CD4(+) cells that are otherwise observed among tumor-infiltrating lymphocytes from mice treated with IL-2. These studies demonstrate that IL-27 and IL-2 synergistically induce complete tumor regression and long-term survival in mice bearing widely metastatic neuroblastoma tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/immunology , Interleukin-2/immunology , Interleukins/immunology , Lymphocyte Activation/drug effects , Neuroblastoma/immunology , T-Lymphocytes, Cytotoxic/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bone Marrow Neoplasms/drug therapy , Bone Marrow Neoplasms/secondary , Drug Synergism , Flow Cytometry , Interferon-gamma/immunology , Interleukin-2/administration & dosage , Interleukins/administration & dosage , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Neuroblastoma/drug therapy , Neuroblastoma/secondary , T-Lymphocytes, Cytotoxic/immunology
9.
Nat Med ; 13(3): 354-60, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17334371

ABSTRACT

Protective cell-mediated immune responses in cancer are critically dependent on T-helper type 1 (T(H)1) cytokines such as interferon-gamma (IFN-gamma). We have previously shown that the combination of CD40 stimulation and interleukin-2 (IL-2) leads to synergistic antitumor responses in several models of advanced metastatic disease. We now report that after this treatment and other immunotherapy regimens, the CD4+ T-cell population, in contrast to CD8+ T cells, did not significantly increase but rather exhibited a substantial level of apoptosis that was dependent on IFN-gamma. Mice immunized with tumor cells and treated with an immunotherapy regimen that was initially protective were later unable to mount effective memory responses compared with immunized mice not receiving immunotherapy. Immunotherapy given to tumor-bearing Ifngr-/- mice resulted in restoration of secondary responses. Thus, although immunotherapeutic regimens inducing strong IFN-gamma responses can lead to successful early antitumor efficacy, they may also impair the development of durable antitumor responses.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Carcinoma, Renal Cell/immunology , Immunologic Memory , Immunotherapy, Active , Interferon-gamma/physiology , Kidney Neoplasms/immunology , Melanoma, Experimental/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/prevention & control , Cell Line , Cells, Cultured , Clonal Deletion/genetics , Clonal Deletion/immunology , Female , Immunologic Memory/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/prevention & control , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
10.
Cancer Res ; 66(22): 11005-12, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17108139

ABSTRACT

The use of interleukin-18 (IL-18) together with IL-12 induced high levels of IFN-gamma in tumor-bearing mice and regression of liver tumors that was abolished in IFN-gamma((-/-)) mice. Natural killer (NK) and NKT cells were the major producers of IFN-gamma in the livers of mice treated with IL-18 and/or IL-12. Liver NK cells were significantly increased by treatment with IL-18/IL-12, whereas the degree of liver NKT cell TCR detection was diminished by this treatment. Reduction of NK cells with anti-asGM1 decreased the antitumor activity of IL-18/IL-12 therapy and revealed NK cells to be an important component for tumor regression in the liver. In contrast, the antitumor effects of both IL-18 and IL-12 were further increased in CD1d((-/-)) mice, which lack NKT cells. Our data, therefore, show that the antitumor activity induced in mice by IL-18/IL-12 is NK and IFN-gamma dependent and is able to overcome an endogenous immunosuppressive effect of NKT cells in the liver microenvironment. These results suggest that immunotherapeutic approaches that enhance NK cell function while eliminating or altering NKT cells could be effective in the treatment of cancer in the liver.


Subject(s)
Interleukin-12/pharmacology , Interleukin-18/pharmacology , Killer Cells, Natural/immunology , Liver/immunology , T-Lymphocytes/immunology , Animals , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Killer Cells, Natural/drug effects , Liver/cytology , Liver/drug effects , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/secondary , Liver Neoplasms, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, SCID , Recombinant Proteins/pharmacology , T-Lymphocytes/drug effects
11.
J Immunol ; 176(10): 6302-12, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16670342

ABSTRACT

Human neuroblastomas possess several mechanisms of self-defense that may confer an ability to resist apoptosis and contribute to the observed difficulty in treating these tumors in the clinical setting. These molecular alterations may include defects in proapoptotic genes as well as the overexpression of prosurvival factors, such as Akt among others. As a key regulator of the turnover of proteins that modulate the cell cycle and mechanisms of apoptosis, the proteasome could serve as an important target for the treatment of neuroblastoma. The present studies provide the first evidence that bortezomib, a newly approved inhibitor of proteasome function, inhibits phosphorylation of Akt, induces the translocation of proapoptotic Bid, and potently enhances the apoptosis of murine neuroblastoma tumor cells in vitro. Furthermore, in that inhibitors of the Akt pathway can sensitize otherwise resistant TBJ/Neuro-2a cells to apoptosis induced by IFN-gamma plus TNF-alpha, we hypothesized that bortezomib also could sensitize these cells to IFN-gamma plus TNF-alpha. We demonstrate for the first time that bortezomib not only up-regulates the expression of receptors for IFN-gamma and TNF-alpha on both TBJ neuroblastoma and EOMA endothelial cell lines, but also markedly enhances the sensitivity of these cells to apoptosis induced by IFN-gamma plus TNF-alpha in vitro. Furthermore, bortezomib enhances the in vivo antitumor efficacy of IFN-gamma/TNF-alpha-inducing cytokines, including both IL-2 and IL-12 in mice bearing well-established primary and/or metastatic TBJ neuroblastoma tumors. Collectively, these studies suggest that bortezomib could be used therapeutically to enhance the proapoptotic and overall antitumor activity of systemic cytokine therapy in children with advanced neuroblastoma.


Subject(s)
Apoptosis/immunology , Cytokines/therapeutic use , Neuroblastoma/enzymology , Neuroblastoma/therapy , Proteasome Inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/physiology , Disease Models, Animal , Growth Inhibitors/pharmacology , Interleukin-12/physiology , Interleukin-12/therapeutic use , Interleukin-2/physiology , Interleukin-2/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Male , Mice , Mice, Inbred A , Neuroblastoma/drug therapy , Neuroblastoma/secondary , Pyrazines/pharmacology
12.
J Immunother ; 29(2): 151-64, 2006.
Article in English | MEDLINE | ID: mdl-16531816

ABSTRACT

Neuroblastoma is one of the most common solid tumors in children. The prognosis of patients with advanced neuroblastoma is poor overall despite standard therapeutic modalities and has stimulated substantial interest in the potential role for biologics such as immunotherapeutic and/or antiangiogenic agents for the treatment of neuroblastoma. To facilitate preclinical investigation of the efficacy and mechanisms of action of new biologic agents for the treatment of neuroblastoma, a comprehensive panel of disease-specific fluorescence-based model systems has been developed by our group to image the growth, neovascularization, metastasis, and apoptosis of neuroblastoma tumors. These model systems use fluorescent proteins to monitor cytokine-induced alterations in the growth and metastasis of neuroblastoma and allow for monitoring and/or quantitation of even minimal residual disease that is localized to visceral organ sites such as the liver, lung, and/or bone marrow. Further, based on the differential spectra of red fluorescent protein, green fluorescent protein (GFP), and agents such as 4'-6-diamidino-2-phenylindole (DAPI) (blue) and fluorescein isothiocyanate-dextran (green), multicolor systems have now been established by our group that allow for combined assessment of parameters, including the macroscopic relation of tumors to their associated vasculature and, within tissue sections, simultaneous quantitation of tumor neovascularization and evaluation of therapy-induced apoptosis within the tumor and vascular endothelial compartments. Further, by engineering cells to express specific mediators of apoptosis that have been linked to GFP (ie, BID-EGFP), these systems can also be used to dissect mechanisms by which neuroblastoma cells are induced to undergo apoptosis in vitro as well as in vivo. Collectively, these model systems provide important tools for investigation of the biology of neuroblastoma tumors and evaluation of mechanisms that mediate the regression of these tumors in response to novel therapeutic agents, including cytokines such as interleukin-12.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Diagnostic Imaging/methods , Interleukin-12/administration & dosage , Neuroblastoma/diagnosis , Neuroblastoma/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cytokines/administration & dosage , Cytokines/therapeutic use , Dextrans , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescence , Green Fluorescent Proteins , Indoles , Luminescent Proteins , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Neuroblastoma/blood supply , Neuroblastoma/pathology , Red Fluorescent Protein
13.
J Natl Cancer Inst ; 98(3): 190-202, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16449679

ABSTRACT

BACKGROUND: Patients with advanced neuroblastoma have a poor prognosis. The antiapoptotic protein Akt has been implicated as a possible mediator of the resistance of human neuroblastoma cells to apoptosis; the proapoptotic protein Bid, is inhibited by activated Akt. Neuroblastoma has demonstrated responsiveness to immunotherapeutic approaches in preclinical studies, prompting investigation of new therapeutic strategies based on potentiation of the host immune response, including the use of systemic cytokines. METHODS: We examined the antitumor efficacy and mechanisms of action of the central immunoregulatory cytokine interleukin-12 (IL-12) in mice bearing established orthotopic neuroblastoma tumors derived from murine TBJ and Neuro-2a cells. Cohorts of mice (10 mice/group) bearing established orthotopic neuroblastoma tumors were injected intraperitoneally with IL-12 or vehicle and monitored for survival. IL-12-induced apoptosis within the tumor microenvironment was investigated using ribonuclease protection assays, nuclear staining, and electron microscopy. Protein expression was determined via Western blot analysis and enzyme-linked immunosorbent assays. Confocal microscopy was used to examine the distribution of overexpressed Bid-enhanced green fluorescent protein fusion protein (Bid-EGFP) in TBJ cells. All statistical tests were two-sided. RESULTS: IL-12 induced complete tumor regression and long-term survival of 8 (80%) of 10 mice bearing established neuroblastoma tumors compared with 1 (10%) of 10 control mice (P = .0055) and profound tumor cell apoptosis in vivo despite the fact that TBJ and Neuro-2a cells were resistant to receptor-mediated apoptosis in vitro. These cells expressed high levels of phosphorylated Akt, a key prosurvival molecule, and Akt inhibitors sensitized neuroblastoma cells to apoptosis mediated by IL-12-inducible cytokines including tumor necrosis factor-alpha and interferon-gamma in vitro. IL-12 increased the expression of proapoptotic genes and decreased Akt phosphorylation within established TBJ tumors in conjunction with activation and subcellular translocation of Bid. CONCLUSIONS: Our results suggest that IL-12 overcomes a potentially critical mechanism of tumor self-defense in vivo by inhibiting Akt activity and imply that IL-12 may possess unique therapeutic activity against tumors that express high levels of activated Akt.


Subject(s)
Antineoplastic Agents/pharmacology , Interleukin-12/pharmacology , Neuroblastoma/drug therapy , Neuroblastoma/enzymology , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Adrenal Gland Neoplasms/drug therapy , Adrenal Gland Neoplasms/enzymology , Animals , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , Blotting, Western , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Fluorescent Dyes , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Green Fluorescent Proteins/drug effects , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Indoles , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Mice , Microscopy, Confocal , Neoplasm Transplantation , Phosphorylation/drug effects , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
14.
J Immunol ; 173(12): 7170-82, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15585838

ABSTRACT

We have shown previously that IFN-gamma-inducing cytokines such as IL-12 can mediate potent antitumor effects against murine solid tumors. IL-27 is a newly described IL-12-related cytokine that potentiates various aspects of T and/or NK cell function. We hypothesized that IL-27 might also mediate potent antitumor activity in vivo. TBJ neuroblastoma cells engineered to overexpress IL-27 demonstrated markedly delayed growth compared with control mice, and complete durable tumor regression was observed in >90% of mice bearing either s.c. or orthotopic intra-adrenal tumors, and 40% of mice bearing induced metastatic disease. The majority of mice cured of their original TBJ-IL-27 tumors were resistant to tumor rechallenge. Furthermore, TBJ-IL-27 tumors were heavily infiltrated by CD8(+) T cells, and draining lymph node-derived lymphocytes from mice bearing s.c. TBJ-IL-27 tumors are primed to proliferate more readily when cultured ex vivo with anti-CD3/anti-CD28 compared with lymphocytes from mice bearing control tumors, and to secrete higher levels of IFN-gamma. In addition, marked enhancement of local IFN-gamma gene expression and potent up-regulation of cell surface MHC class I expression are noted within TBJ-IL-27 tumors compared with control tumors. Functionally, these alterations occur in conjunction with the generation of tumor-specific CTL reactivity in mice bearing TBJ-IL-27 tumors, and the induction of tumor regression via mechanisms that are critically dependent on CD8(+), but not CD4(+) T cells or NK cells. Collectively, these studies suggest that IL-27 could be used therapeutically to potentiate the host antitumor immune response in patients with malignancy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interleukins/therapeutic use , Neuroblastoma/secondary , Neuroblastoma/therapy , Adjuvants, Immunologic/biosynthesis , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/therapeutic use , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Cell Movement/immunology , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/biosynthesis , Immunologic Memory , Injections, Intravenous , Injections, Subcutaneous , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukins/biosynthesis , Interleukins/genetics , Liver Neoplasms/immunology , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Male , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, SCID , Molecular Sequence Data , Neoplasm Transplantation/immunology , Neuroblastoma/genetics , Neuroblastoma/immunology , Transfection , Up-Regulation/immunology
15.
Cancer Res ; 63(17): 5320-8, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14500363

ABSTRACT

von Hippel-Lindau (VHL) disease is a multisystem inherited cancer syndrome characterized by the development of highly vascular tumors including hemangioblastomas of the retina and central nervous system, pheochromocytomas, and clear cell renal carcinoma, which result from somatic inactivation of the wild-type VHL allele in cells harboring a germ-line VHL mutation. Homozygous inactivation of the VHL gene in mice resulted in embryonic lethality. To produce a mouse model that closely mimics human VHL disease and avoids embryonic lethality, we used Cre/lox site-specific recombination technology. We generated mice carrying conditional VHL alleles and a cre transgene under the control of the human beta-actin promoter, which directs cre expression in a mosaic pattern in multiple organs. VHL(f/d)/Cre mice developed multiple, hepatic hemangiomas that led to premature death, as well as angiectasis and angiogenesis in multiple organs. Interestingly, testes of male VHL(f/d)/Cre mice were unusually small with severely reduced sperm count resulting in infertility. Loss of pVHL function in this VHL conditional knockout mouse model results in an extensive abnormal vascular phenotype in multiple mouse organs, which will provide a useful animal model for testing potential antiangiogenic therapies for VHL disease treatment. Importantly, the phenotypic defects in sperm development observed in these mice support a novel role for VHL in spermatogenesis. This VHL conditional knockout mouse model will provide an in vivo system for studying the functional requirement of the VHL gene in reproductive biology.


Subject(s)
Disease Models, Animal , Hemangioma/genetics , Liver Neoplasms, Experimental/genetics , Neovascularization, Pathologic/genetics , Spermatogenesis/genetics , Transcription Factors , von Hippel-Lindau Disease/genetics , Actins/genetics , Alleles , Animals , DNA Nucleotidyltransferases/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Female , Gene Silencing , Hemangioma/blood supply , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Liver Neoplasms, Experimental/blood supply , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Promoter Regions, Genetic , Recombinases , Recombination, Genetic , Up-Regulation
16.
J Immunol ; 169(8): 4467-74, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12370382

ABSTRACT

IFN-gamma is a critical component of the endogenous and many cytokine-induced antitumor immune responses. In this study we have shown that the combination of IL-18 and IL-2 (IL-18/IL-2) synergistically enhances IFN-gamma production both in vitro and in vivo, and synergizes in vivo to induce complete durable regression of well-established 3LL tumors in >80% of treated mice. We have observed a nascent, but ineffective, host immune response against 3LL that depends on endogenous IFN-gamma and IL-12 production and the Fas/Fas ligand (Fas-L) pathway. The combined administration of IL-18/IL-2 engages this endogenous response to induce tumor regression via a mechanism that is independent of NK and NKT cells or IL-12, but is critically dependent on CD8(+) T cells, IFN-gamma, and the Fas/Fas-L pathway. These studies demonstrate the importance of IFN-gamma as well as the Fas/Fas-L pathway in both endogenous and cytokine-driven antitumor immune responses engaged by IL-18/IL-2 and provide preclinical impetus for clinical investigation of this potent anti-tumor combination.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/immunology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/prevention & control , Interferon-gamma/biosynthesis , Membrane Glycoproteins/physiology , fas Receptor/metabolism , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , B-Lymphocytes/immunology , Carcinoma, Lewis Lung/pathology , Cells, Cultured , Drug Synergism , Fas Ligand Protein , Injections, Intraperitoneal , Interferon-gamma/physiology , Interleukin-12/physiology , Interleukin-18/administration & dosage , Interleukin-18/pharmacology , Interleukin-2/administration & dosage , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Remission Induction , Signal Transduction/immunology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocyte Subsets/immunology , fas Receptor/physiology
17.
J Immunol ; 168(7): 3484-92, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11907109

ABSTRACT

Kidney cancer is a devastating disease; however, biological therapies have achieved some limited success. The murine renal cancer Renca has been used as a model for developing new preclinical approaches to the treatment of renal cell carcinoma. Successful cytokine-based approaches require CD8(+) T cells, but the exact mechanisms by which T cells mediate therapeutic benefit have not been completely identified. After successful biological therapy of Renca in BALB/c mice, we generated CTLs in vitro using mixed lymphocyte tumor cultures. These CTL mediated tumor-specific H-2K(d)-restricted lysis and production of IFN-gamma, TNF-alpha, and Fas ligand (FasL) in response to Renca. CTL used both granule- and FasL-mediated mechanisms to lyse Renca, although granule-mediated killing was the predominant lytic mechanism in vitro. The cytokines IFN-gamma and TNF-alpha increased the sensitivity of Renca cells to CTL lysis by both granule- and FasL-mediated death pathways. Adoptive transfer of these anti-Renca CTL into tumor-bearing mice cured most mice of established experimental pulmonary metastases, and successfully treated mice were immune to tumor rechallenge. Interestingly, we were able to establish Renca-specific CTL from mice gene targeted for perforin (pfp(-/-)) mice. Although these pfp(-/-) CTL showed reduced cytotoxic activity against Renca, their IFN-gamma production in the presence of Renca targets was equivalent to that of wild-type CTL, and adoptive transfer of pfp(-/-) CTL was as efficient as wild-type CTL in causing regression of established Renca pulmonary metastases. Therefore, although granule-mediated killing is of paramount importance for CTL-mediated lysis in vitro, some major in vivo effector mechanisms clearly are independent of perforin.


Subject(s)
Carcinoma, Renal Cell/therapy , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/immunology , Kidney Neoplasms/therapy , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/toxicity , T-Lymphocytes, Cytotoxic/immunology , Animals , Antineoplastic Agents/toxicity , Apoptosis/genetics , Apoptosis/immunology , Apoptosis Regulatory Proteins , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Cell Line, Transformed , Cytotoxicity, Immunologic/genetics , Epitopes, T-Lymphocyte/administration & dosage , Fas Ligand Protein , Immunotherapy, Adoptive/methods , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/physiology , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Ligands , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Lymphocyte Activation/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Lymphocyte Function-Associated Antigen-1/physiology , Melanoma, Experimental/immunology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Neoplasm Transplantation , Perforin , Pore Forming Cytotoxic Proteins , T-Lymphocytes, Cytotoxic/transplantation , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/toxicity , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...