Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
PLoS One ; 8(7): e67583, 2013.
Article in English | MEDLINE | ID: mdl-23844038

ABSTRACT

Mitogen-Activated Protein Kinase (MAPK) pathway activation has been implicated in many types of human cancer. BRAF mutations that constitutively activate MAPK signalling and bypass the need for upstream stimuli occur with high prevalence in melanoma, colorectal carcinoma, ovarian cancer, papillary thyroid carcinoma, and cholangiocarcinoma. In this report we characterize the novel, potent, and selective BRAF inhibitor, dabrafenib (GSK2118436). Cellular inhibition of BRAF(V600E) kinase activity by dabrafenib resulted in decreased MEK and ERK phosphorylation and inhibition of cell proliferation through an initial G1 cell cycle arrest, followed by cell death. In a BRAF(V600E)-containing xenograft model of human melanoma, orally administered dabrafenib inhibited ERK activation, downregulated Ki67, and upregulated p27, leading to tumor growth inhibition. However, as reported for other BRAF inhibitors, dabrafenib also induced MAPK pathway activation in wild-type BRAF cells through CRAF (RAF1) signalling, potentially explaining the squamous cell carcinomas and keratoacanthomas arising in patients treated with BRAF inhibitors. In addressing this issue, we showed that concomitant administration of BRAF and MEK inhibitors abrogated paradoxical BRAF inhibitor-induced MAPK signalling in cells, reduced the occurrence of skin lesions in rats, and enhanced the inhibition of human tumor xenograft growth in mouse models. Taken together, our findings offer preclinical proof of concept for dabrafenib as a specific and highly efficacious BRAF inhibitor and provide evidence for its potential clinical benefits when used in combination with a MEK inhibitor.


Subject(s)
Melanoma/drug therapy , Melanoma/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Female , Humans , Imidazoles/administration & dosage , Melanoma/pathology , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mutation , Oximes/administration & dosage , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
2.
ACS Med Chem Lett ; 4(3): 358-62, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-24900673

ABSTRACT

Hyperactive signaling of the MAP kinase pathway resulting from the constitutively active B-Raf(V600E) mutated enzyme has been observed in a number of human tumors, including melanomas. Herein we report the discovery and biological evaluation of GSK2118436, a selective inhibitor of Raf kinases with potent in vitro activity in oncogenic B-Raf-driven melanoma and colorectal carcinoma cells and robust in vivo antitumor and pharmacodynamic activity in mouse models of B-Raf(V600E) human melanoma. GSK2118436 was identified as a development candidate, and early clinical results have shown significant activity in patients with B-Raf mutant melanoma.

4.
Mol Cancer Ther ; 7(9): 2876-83, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18790768

ABSTRACT

Recent studies have shown that there is a considerable heterogeneity in the response of melanoma cell lines to MEK and BRAF inhibitors. In the current study, we address whether dysregulation of cyclin-dependent kinase 4 (CDK4) and/or cyclin D1 contribute to the BRAF inhibitor resistance of melanoma cells. Mutational screening identified a panel of melanoma cell lines that harbored both a BRAF V600E mutation and a CDK4 mutation: K22Q (1205Lu), R24C (WM39, WM46, and SK-Mel-28), and R24L (WM902B). Pharmacologic studies showed that the presence of a CDK4 mutation did not alter the sensitivity of these cell lines to the BRAF inhibitor. The only cell line with significant BRAF inhibitor resistance was found to harbor both a CDK4 mutation and a CCND1 amplification. Array comparative genomic hybridization analysis showed that CCND1 was amplified in 17% of BRAF V600E-mutated human metastatic melanoma samples, indicating the clinical relevance of this finding. As the levels of CCND1 amplification in cell lines are lower than those seen in clinical specimens, we overexpressed cyclin D1 alone and in the presence of CDK4 in a drug-sensitive melanoma line. Cyclin D1 overexpression alone increased resistance and this was enhanced when cyclin D1 and CDK4 were concurrently overexpressed. In conclusion, increased levels of cyclin D1, resulting from genomic amplification, may contribute to the BRAF inhibitor resistance of BRAF V600E-mutated melanomas, particularly when found in the context of a CDK4 mutation/overexpression.


Subject(s)
Cyclin D1/metabolism , Drug Resistance, Neoplasm/drug effects , Imidazoles/pharmacology , Melanoma/genetics , Mutation/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Amino Acid Substitution/drug effects , Base Sequence , Cell Line, Tumor , Cyclin D1/genetics , Cyclin-Dependent Kinase 4/genetics , DNA Mutational Analysis , Gene Amplification/drug effects , Glutamic Acid/genetics , Humans , Melanoma/metabolism , Molecular Sequence Data , Valine/genetics
5.
Cancer Res ; 68(14): 5743-52, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18632627

ABSTRACT

Although many melanomas harbor either activating mutations in BRAF or NRAS, there remains a substantial, yet little known, group of tumors without either mutation. Here, we used a genomic strategy to define a novel group of melanoma cell lines with co-overexpression of cyclin-dependent kinase 4 (CDK4) and KIT. Although this subgroup lacked any known KIT mutations, they had high phospho-KIT receptor expression, indicating receptor activity. Quantitative PCR confirmed the existence of a similar KIT/CDK4 subgroup in human melanoma samples. Pharmacologic studies showed the KIT/CDK4-overexpressing subgroup to be resistant to BRAF inhibitors but sensitive to imatinib in both in vitro and in vivo melanoma models. Mechanistically, imatinib treatment led to increased apoptosis and G(1) phase cell cycle arrest associated with the inhibition of phospho-ERK and increased expression of p27(KIP). Other melanoma cell lines, which retained some KIT expression but lacked phospho-KIT, were not sensitive to imatinib, suggesting that KIT expression alone is not predictive of response. We suggest that co-overexpression of KIT/CDK4 is a potential mechanism of oncogenic transformation in some BRAF/NRAS wild-type melanomas. This group of melanomas may be a subpopulation for which imatinib or other KIT inhibitors may constitute optimal therapy.


Subject(s)
Cyclin-Dependent Kinase 4/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Animals , Gene Expression Profiling , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Spheroids, Cellular/metabolism , Tumor Cells, Cultured/metabolism , ras Proteins/metabolism
6.
Gastroenterology ; 134(7): 1981-93, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18439605

ABSTRACT

BACKGROUND & AIMS: Esophageal squamous cell carcinoma (ESCC) is known to be a highly angiogenic tumor. Here, we investigated the role of the stromal fibroblasts in the ESCC-induced angiogenic response using a novel 3-dimensional model. METHODS: A novel assay was developed where cocultures of ESCC and esophageal fibroblasts induced human microvascular endothelial cell (HMVEC) vascular network formation in a 3-dimensional collagen gel. Biochemical studies showed that the ESCC-induced activation of the fibroblasts was required to induce vascular network formation via a transforming growth factor (TGF)-beta and vascular endothelial growth factor (VEGF)-dependent pathway. RESULTS: Conditioned media from a panel of 4 ESCC lines transdifferentiated normal esophageal fibroblasts into myofibroblasts via TGF-beta signaling. The presence of fibroblasts was essential for efficient HMVEC network formation, and the addition of ESCC cells to these cultures greatly enhanced the angiogenic process. The role of TGF-beta in this process was shown by the complete inhibition of network formation following TGF-beta inhibitor treatment. Finally, we showed that ESCC-derived TGF-beta regulates angiogenesis through the release of VEGF from the fibroblasts and that the VEGF release was blocked following TGF-beta inhibition. CONCLUSIONS: This study shows the essential role of fibroblasts in the ESCC angiogenic-induced response and suggests that the pharmacologic targeting of the TGF-beta signaling axis could be of therapeutic benefit in this deadly disease.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Endothelial Cells/metabolism , Esophageal Neoplasms/metabolism , Fibroblasts/metabolism , Neovascularization, Pathologic/metabolism , Paracrine Communication , Benzamides/pharmacology , Benzodioxoles/pharmacology , Carcinoma, Squamous Cell/blood supply , Cell Line, Tumor , Cell Transdifferentiation , Coculture Techniques , Collagen/metabolism , Culture Media, Conditioned/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Esophageal Neoplasms/blood supply , Esophagus/embryology , Esophagus/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Gels , Humans , Imidazoles/pharmacology , Microcirculation/metabolism , Paracrine Communication/drug effects , Pyrazoles/pharmacology , Pyridines/pharmacology , Transfection , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/metabolism , Vascular Endothelial Growth Factor A/metabolism
7.
Anal Bioanal Chem ; 390(1): 343-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17985120

ABSTRACT

No universal approach has been reported for specific monitoring of the catalytic activity of a wide range of kinases in cells. The present study describes an original platform for detecting the autonomous activity of serine/threonine kinases in cells through the introduction of expression vectors encoding modified substrate kinase fusion proteins. The surrogate substrate used consists of the p53 tumor suppressor protein fused with individual kinase domains (Chk1, DYRK3, and Cdk5) at its carboxy-terminal through four tandem Gly-Gly-Gly-Gly-Ser repeats. After transfection into cells, phosphorylation of the p53 moiety could be specifically induced by the catalytic activity of kinases contained in the fusion protein. Moreover, p53 phosphorylation was significantly blocked when a kinase-inactive mutant was used as the fusion partner instead of the active kinase. Using this system, the cell-based evaluation of several Cdk5 inhibitors was demonstrated. Thus, this approach provides a novel platform for the specific, cell-based screening of inhibitors of a wide prospective range of protein kinases and is of tremendous potential for drug discovery efforts.


Subject(s)
Cyclin-Dependent Kinase 5/analysis , Cyclin-Dependent Kinase 5/metabolism , Protein Kinases/analysis , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/metabolism , Animals , Cell Line , Checkpoint Kinase 1 , Chlorocebus aethiops , Cyclin-Dependent Kinase 5/genetics , Humans , Phosphorylation , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
J Biomol Screen ; 12(6): 809-17, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17517898

ABSTRACT

Checkpoint kinase 1 (Chk1), a serine/threonine kinase, plays an important role in DNA damage checkpoint control and is an attractive target for cancer treatment. To develop a Chk1-specific cell-based assay, stable clones were established in which Chk1 kinase domain fused at its N-terminus with p53 through 4 tandem repeats of Gly-Gly-Gly-Gly-Ser was expressed in an inducible manner. Chk1 kinase specificity of the phosphorylation of fused p53 was confirmed by the experiments with a kinase-inactive Chk1. Only in the presence of an inducer molecule was phosphorylation of p53 at Ser-15 in the stable clones induced. Furthermore, its assay performance proved acceptable for high-throughput screening applications, judging from the Z' factor values (> 0.77). Finally, the cell-based assay thus established yielded structure-activity relationship data for a small set of test inhibitors of Chk1 within cells. Collectively, these results demonstrate that the established cell-based assay provides a novel and highly sensitive cellular platform for Chk1 inhibitor discovery.


Subject(s)
Drug Evaluation, Preclinical/methods , Gene Targeting/methods , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Cell Line , Checkpoint Kinase 1 , Feasibility Studies , Gene Expression Regulation , Genes, Reporter , Genes, p53 , HeLa Cells , Humans , Inhibitory Concentration 50 , Models, Biological , Phosphorylation , Protein Kinases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection
9.
Cancer Res ; 66(23): 11100-5, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17145850

ABSTRACT

Oncogenic BRAF alleles are both necessary and sufficient for cellular transformation, suggesting that chemical inhibition of the activated mutant protein kinase may reverse the tumor phenotype. Here, we report the characterization of SB-590885, a novel triarylimidazole that selectively inhibits Raf kinases with more potency towards B-Raf than c-Raf. Crystallographic analysis revealed that SB-590885 stabilizes the oncogenic B-Raf kinase domain in an active configuration, which is distinct from the previously reported mechanism of action of the multi-kinase inhibitor, BAY43-9006. Malignant cells expressing oncogenic B-Raf show selective inhibition of mitogen-activated protein kinase activation, proliferation, transformation, and tumorigenicity when exposed to SB-590885, whereas other cancer cell lines and normal cells display variable sensitivities or resistance to similar treatment. These studies support the validation of oncogenic B-Raf as a target for cancer therapy and provide the first evidence of a correlation between the expression of oncogenic BRAF alleles and a positive response to a selective B-Raf inhibitor.


Subject(s)
Imidazoles/therapeutic use , Neoplasms/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Alleles , Animals , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Crystallization , Crystallography, X-Ray , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , HT29 Cells , Humans , Imidazoles/chemistry , Mice , Mice, Nude , Models, Molecular , Molecular Structure , Mutation/genetics , Neoplasms/enzymology , Neoplasms/pathology , Phosphorylation/drug effects , Protein Conformation/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/genetics , Xenograft Model Antitumor Assays
10.
Proc Natl Acad Sci U S A ; 103(20): 7625-30, 2006 May 16.
Article in English | MEDLINE | ID: mdl-16684877

ABSTRACT

Heat shock protein (Hsp)90 is emerging as an important therapeutic target for the treatment of cancer. Two analogues of the Hsp90 inhibitor geldanamycin are currently in clinical trials. Geldanamycin (GA) and its analogues have been reported to bind purified Hsp90 with low micromolar potency, in stark contrast to their low nanomolar antiproliferative activity in cell culture and their potent antitumor activity in animal models. Several models have been proposed to account for the approximately 100-fold-greater potency in cell culture, including that GA analogues bind with greater affinity to a five-protein Hsp90 complex than to Hsp90 alone. We have determined that GA and the fluorescent analogue BODIPY-GA (BDGA) both demonstrate slow, tight binding to purified Hsp90. BDGA, used to characterize the kinetics of ligand-Hsp90 interactions, was found to bind Hsp90alpha with k(off) = 2.5 x 10(-3) min(-1), t(1/2) = 4.6 h, and Ki* = 10 nM. It was found that BDGA binds to a functional multiprotein Hsp90 complex with kinetics and affinity identical to that of Hsp90 alone. Also, BDGA binds to Hsp90 from multiple cell lysates in a time-dependent manner with similar kinetics. Therefore, our results indicate that the high potency of GA in cell culture and in vivo can be accounted for by its time-dependent, tight binding to Hsp90 alone. In the broader context, these studies highlight the essentiality of detailed biochemical characterization of drug-target interactions for the effective translation of in vitro pharmacology to cellular and in vivo efficacy.


Subject(s)
Antibiotics, Antineoplastic , HSP90 Heat-Shock Proteins , Quinones , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/metabolism , Benzoquinones , Boron Compounds/chemistry , Boron Compounds/metabolism , Cells, Cultured , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic , Protein Binding , Quinones/chemistry , Quinones/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...