Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Neuro Oncol ; 16(7): 914-23, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24470545

ABSTRACT

BACKGROUND: Grades II and III gliomas have unpredictable rates of progression, making management decisions difficult. Currently, several clinical and radiological characteristics are utilized to predict progression and survival but collectively are suboptimal. METHODS: In this study, we analyzed a set of 108 nonenhancing hemispheric grade II-III gliomas. Demographic variables, including patient age, tumor diameter, extent of resection, and performance status, were combined with molecular data (IDH mutation status [mIDH], 1p/19q codeletion, PTEN deletion, and EGFR amplification). A complete dataset for all variables was compiled for 70 of the 108 patients. Both univariable and multivariable analyses were performed to determine whether the molecular data singly or in combination offer advantages over tumor type and grade for prediction of overall survival (OS) and/or progression-free rate (PFR). RESULTS: Patient age, clinical variables (tumor diameter, extent of resection, performance status), and pathology (tumor type and grade) were not predictive of OS or PFR. IDH mutation status alone was predictive of longer OS and PFR for the entire group of tumors; 1p/19q deletion alone was predictive of OS but not PFR. In the multivariable analysis, none of the clinical or demographic factors were predictive of OS or PFR. IDH mutation status, 1p/19q codeletion, and PTEN deletion were predictive of OS (P = .003, P = .005, P = .02, respectively). Both mIDH (P < .001) and the interaction term of 1p/19q and PTEN (P < .001) were found to be predictive of PFR. CONCLUSIONS: We conclude that the combination of mIDH, 1p/19q codeletion, and PTEN deletion may be particularly effective in discriminating good prognosis from poor prognosis hemispheric gliomas. We propose that such a scheme merits testing on larger prospective cohorts. Should our findings be confirmed, routine clinical analysis of hemispheric gliomas for mIDH, 1p/19q codeletion, and PTEN deletion would be justified.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioma/genetics , Glioma/pathology , Adult , Brain Neoplasms/mortality , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 19/genetics , DNA Mutational Analysis , Disease Progression , Disease-Free Survival , Female , Genotype , Glioma/mortality , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Isocitrate Dehydrogenase/genetics , Kaplan-Meier Estimate , Male , Middle Aged , Mutation , Neoplasm Grading , PTEN Phosphohydrolase/genetics , Polymerase Chain Reaction , Prognosis , Proportional Hazards Models , Sequence Deletion , Tissue Array Analysis
2.
J Immunother ; 30(1): 64-74, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17198084

ABSTRACT

Recombinant interleukin-2 (rIL-2) is a pleiotropic cytokine that activates select immune effector cell responses associated with antitumor activity, including antibody-dependent cellular cytotoxicity (ADCC). Rituximab is an anti-CD20 monoclonal antibody that activates ADCC in non-Hodgkin lymphoma (NHL). The ability of rIL-2 to augment rituximab-dependent tumor responses was investigated. The efficacy of rIL-2 in combination with rituximab was evaluated in 2 NHL tumor xenograft models: the CD20hi, rituximab-sensitive, low-grade Daudi model and the CD20lo, aggressive, rituximab-resistant Namalwa model. Combination of rIL-2 plus rituximab was synergistic in a rituximab-sensitive Daudi tumor model, as evidenced by significant tumor regressions and increased time to tumor progression, compared with rIL-2 and rituximab single agents. In contrast, rituximab-resistant Namalwa tumors were responsive to single-agent rIL-2 and showed an increased response when combined with rituximab. Using in vitro killing assays, rIL-2 was shown to enhance activity of rituximab by activating ADCC and lymphokine-activated killer activity. Additionally, the activity of rIL-2 plus rituximab F(ab')2 was similar to that of rIL-2 alone, indicating a critical role for immunoglobulin G1 Fc-FcgammaR-effector responses in mediating ADCC. Antiproliferative and apoptotic tumor responses, along with an influx of immune effector cells, were observed by immunohistochemistry. Collectively, the data suggest that rIL-2 mediates potent tumoricidal activity against NHL tumors, in part, through activation and trafficking of monocytes and natural killer cells to tumors. These data support the mechanistic and therapeutic rationale for combination of rIL-2 with rituximab in NHL clinical trials and for single-agent rIL-2 in rituximab-resistant NHL patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Interleukin-2/pharmacology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Murine-Derived , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, CD20/immunology , Drug Synergism , Female , Humans , Immunoglobulin Fc Fragments/immunology , Interleukin-2/administration & dosage , Interleukin-2/immunology , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Monocytes/immunology , Rituximab , Xenograft Model Antitumor Assays
3.
Antimicrob Agents Chemother ; 50(10): 3260-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17005803

ABSTRACT

The lack of a robust small-animal model for hepatitis C virus (HCV) has hindered the discovery and development of novel drug treatments for HCV infections. We developed a reproducible and easily accessible xenograft mouse efficacy model in which HCV RNA replication is accurately monitored in vivo by real-time, noninvasive whole-body imaging of gamma-irradiated SCID mice implanted with a mouse-adapted luciferase replicon-containing Huh-7 cell line (T7-11). The model was validated by demonstrating that both a small-molecule NS3/4A protease inhibitor (BILN 2061) and human alpha interferon (IFN-alpha) decreased HCV RNA replication and that treatment withdrawal resulted in a rebound in replication, which paralleled clinical outcomes in humans. We further showed that protease inhibitor and IFN-alpha combination therapy was more effective in reducing HCV RNA replication than treatment with each compound alone and supports testing in humans. This robust mouse efficacy model provides a powerful tool for rapid evaluation of potential anti-HCV compounds in vivo as part of aggressive drug discovery efforts.


Subject(s)
Antiviral Agents/pharmacology , Carbamates/pharmacology , Disease Models, Animal , Hepacivirus/drug effects , Interferon-alpha/pharmacology , Macrocyclic Compounds/pharmacology , Quinolines/pharmacology , Thiazoles/pharmacology , Virus Replication/drug effects , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/therapeutic use , Carbamates/administration & dosage , Carbamates/therapeutic use , Cell Line, Tumor/transplantation , Drug Evaluation, Preclinical , Female , Hepatitis C/virology , Humans , Interferon-alpha/administration & dosage , Interferon-alpha/therapeutic use , Macrocyclic Compounds/administration & dosage , Macrocyclic Compounds/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Quinolines/administration & dosage , Quinolines/therapeutic use , Thiazoles/administration & dosage , Thiazoles/therapeutic use
4.
Clin Cancer Res ; 12(16): 4908-15, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16914579

ABSTRACT

PURPOSE: The ectopically expressed and deregulated fibroblast growth factor receptor 3 (FGFR3) results from a t(4;14) chromosomal translocation that occurs in approximately 15% of multiple myeloma (MM) patients and confers a particularly poor prognosis. This study assesses the antimyeloma activity of CHIR-258, a small-molecule inhibitor of multiple receptor tyrosine kinases that is currently in phase I trials, in a newly developed FGFR3-driven preclinical MM animal model. EXPERIMENTAL DESIGN: We developed an orthotopic MM model in mice using a luciferase-expressing human KMS-11-luc line that expresses mutant FGFR3 (Y373C). The antimyeloma activity of CHIR-258 was evaluated at doses that inhibited FGFR3 signaling in vivo in this FGFR3-driven animal model. RESULTS: Noninvasive bioluminescence imaging detected MM lesions in nearly all mice injected with KMS-11-luc cells, which were mainly localized in the spine, skull, and pelvis, resulting in frequent development of paralysis. Daily oral administration of CHIR-258 at doses that inhibited FGFR3 signaling in KMS-11-luc tumors in vivo resulted in a significant inhibition of KMS-11-luc tumor growth, which translated into a significant improvement in animal survival. CONCLUSIONS: Our data provide a relevant preclinical basis for clinical trials of CHIR-258 in FGFR3-positive MM patients.


Subject(s)
Benzimidazoles/pharmacology , Multiple Myeloma/drug therapy , Quinolones/pharmacology , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/biosynthesis , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Growth Processes/drug effects , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, SCID , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Multiple Myeloma/enzymology , Phosphorylation/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/blood , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 11(14): 5281-91, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-16033847

ABSTRACT

PURPOSE: Fms-like tyrosine kinase 3 (FLT3) encodes a receptor tyrosine kinase (RTK) for which activating mutations have been identified in a proportion of acute myelogenous leukemia (AML) patients and associated with poor clinical prognosis. Given the relevance of FLT3 mutations in AML, we investigated the activity of CHIR-258, an orally active, multitargeted small molecule, with potent activity against FLT3 kinase and class III, IV, and V RTKs involved in endothelial and tumor cell proliferation in AML models. EXPERIMENTAL DESIGN: CHIR-258 was tested on two human leukemic cell lines in vitro and in vivo with differing FLT3 mutational status [MV4;11 cells express FLT3 internal tandem duplications (ITD) versus RS4;11 cells with wild-type (WT) FLT3]. RESULTS: Antiproliferative activity of CHIR-258 against MV4;11 was approximately 24-fold greater compared with RS4;11, indicating more potent inhibition against cells with constitutively activated FLT3 ITD. Dose-dependent down modulation of receptor phosphorylation and downstream signaling [signal transducer and activator of transcription 5 (STAT5) and extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase] in MV4;11 cells with CHIR-258 confirmed the molecular mechanism of action. Target modulation of phospho-FLT3, phospho-STAT5, and phospho-ERK in MV4;11 tumors was achieved at biologically active doses of CHIR-258. Tumor regressions and eradication of AML cells from the bone marrow were shown in s.c. and bone marrow engraftment leukemic xenograft models. Tumor responses were characterized by decreased cellular proliferation and positive immunohistochemical staining for active caspase-3 and cleaved poly(ADP-ribose) polymerase, suggesting cell death was mediated in part via apoptosis. CONCLUSIONS: Our data indicate that CHIR-258 may be an effective therapy in FLT3-associated AML and warrants clinical trials.


Subject(s)
Benzimidazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Proto-Oncogene Proteins/genetics , Quinolones/pharmacology , Receptor Protein-Tyrosine Kinases/genetics , Animals , Cell Proliferation , DNA Mutational Analysis , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Immunohistochemistry , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/veterinary , Mice , Mice, SCID , Neoplasm Transplantation , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Tandem Repeat Sequences , Transplantation, Heterologous , Tumor Cells, Cultured , fms-Like Tyrosine Kinase 3
6.
Clin Cancer Res ; 11(10): 3633-41, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15897558

ABSTRACT

PURPOSE: To evaluate the therapeutic and biological effects of CHIR-258, an orally bioavailable, potent inhibitor of class III-V receptor tyrosine kinases, in colon cancer models. EXPERIMENTAL DESIGN: The pharmacologic activity of CHIR-258 was characterized by monitoring target modulation as well as by evaluating the antitumor and antiangiogenic effects in human colon xenograft models. RESULTS: CHIR-258 inhibits vascular endothelial growth factor receptor 1/2, fibroblast growth factor receptor 1/3, and platelet-derived growth factor receptor beta (PDGFRbeta) and shows both antitumor and antiangiogenic activities in vivo. Treatment of KM12L4a human colon cancer cells with CHIR-258 resulted in a dose-dependent inhibition of vascular endothelial growth factor receptor 1 and PDGFRbeta phosphorylation and reduction of phosphorylated extracellular signal-regulated kinase (ERK) levels, indicating modulation of target receptors and downstream signaling. In vivo administration of CHIR-258 resulted in significant tumor growth inhibition and tumor regressions, including large, established tumors (500-1,000 mm(3)). Immunohistochemical analysis showed a reduction of phosphorylated PDGFRbeta and phosphorylated ERK in tumor cells after oral dosing with CHIR-258 compared with control tumors. These changes were accompanied by decreased tumor cell proliferation rate and reduced intratumoral microvessel density. CHIR-258 inhibited the phosphorylation of PDGFRbeta and ERK phosphorylation in tumors within 2 hours following dosing and the inhibitory activity was sustained for >24 hours. Significant antitumor activity was observed with intermittent dosing schedules, indicating a sustained biological activity. CONCLUSION: These studies provide evidence that biological activity of CHIR-258 in tumors correlates with efficacy and aids in the identification of potential biomarkers of this multitargeted receptor tyrosine kinase inhibitor. CHIR-258 exhibits properties that make it a promising candidate for clinical development in a variety of solid and hematologic malignancies.


Subject(s)
Benzimidazoles/pharmacology , Biomarkers, Tumor/analysis , Colonic Neoplasms/drug therapy , Growth Substances/biosynthesis , Neovascularization, Pathologic , Quinolones/pharmacology , Animals , Benzimidazoles/pharmacokinetics , Cell Proliferation , Colonic Neoplasms/pathology , Colonic Neoplasms/veterinary , Female , Humans , Mice , Phosphorylation , Proto-Oncogene Proteins c-sis/metabolism , Quinolones/pharmacokinetics , Transplantation, Heterologous , Vascular Endothelial Growth Factor Receptor-1/metabolism
7.
Cancer Res ; 63(12): 3145-53, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12810642

ABSTRACT

To evaluate whether beta-catenin signaling has a role in the regulation of angiogenesis in colon cancer, a series of angiogenesis-related gene promoters was analyzed for beta-catenin/TCF binding sites. Strikingly, the gene promoter of human vascular endothelial growth factor (VEGF, or VEGF-A) contains seven consensus binding sites for beta-catenin/TCF. Analysis of laser capture microdissected human colon cancer tissue indicated a direct correlation between up-regulation of VEGF-A expression and adenomatous polyposis coli (APC) mutational status (activation of beta-catenin signaling) in primary tumors. In metastases, this correlation was not observed. Analysis by immunohistochemistry of intestinal polyps in mice heterozygous for the multiple intestinal neoplasia gene (Min/+) at 5 months revealed an increase and redistribution of VEGF-A in proximity to those cells expressing nuclear beta-catenin with a corresponding increase in vessel density. Transfection of normal colon epithelial cells with activated beta-catenin up-regulated levels of VEGF-A mRNA and protein by 250-300%. When colon cancer cells with elevated beta-catenin levels were treated with beta-catenin antisense oligodeoxynucleotides, VEGF-A expression was reduced by more than 50%. Taken together, our observations indicate a close link between beta-catenin signaling and the regulation of VEGF-A expression in colon cancer.


Subject(s)
Adenocarcinoma/genetics , Colonic Neoplasms/genetics , Cytoskeletal Proteins/physiology , Endothelial Growth Factors/biosynthesis , Gene Expression Regulation, Neoplastic/physiology , Intercellular Signaling Peptides and Proteins/biosynthesis , Lymphokines/biosynthesis , Neoplasm Proteins/biosynthesis , Promoter Regions, Genetic/genetics , Trans-Activators/physiology , Adenocarcinoma/blood supply , Adenocarcinoma/etiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/metabolism , Animals , Binding Sites , Colon/metabolism , Colonic Neoplasms/blood supply , Colonic Neoplasms/etiology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytoskeletal Proteins/genetics , Endothelial Growth Factors/genetics , Fibroblast Growth Factor 2/analysis , Gene Expression Regulation, Neoplastic/drug effects , Genes, APC , Genes, ras , Growth Substances/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intestinal Mucosa/metabolism , Lymphokines/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neoplasm Proteins/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Recombinant Fusion Proteins/physiology , Signal Transduction , Subcellular Fractions/chemistry , Trans-Activators/genetics , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...