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.
Cancer Cell ; 35(5): 738-751.e9, 2019 05 13.
Article in English | MEDLINE | ID: mdl-31085175

ABSTRACT

Ripretinib (DCC-2618) was designed to inhibit the full spectrum of mutant KIT and PDGFRA kinases found in cancers and myeloproliferative neoplasms, particularly in gastrointestinal stromal tumors (GISTs), in which the heterogeneity of drug-resistant KIT mutations is a major challenge. Ripretinib is a "switch-control" kinase inhibitor that forces the activation loop (or activation "switch") into an inactive conformation. Ripretinib inhibits all tested KIT and PDGFRA mutants, and notably is a type II kinase inhibitor demonstrated to broadly inhibit activation loop mutations in KIT and PDGFRA, previously thought only achievable with type I inhibitors. Ripretinib shows efficacy in preclinical cancer models, and preliminary clinical data provide proof-of-concept that ripretinib inhibits a wide range of KIT mutants in patients with drug-resistant GISTs.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Animals , CHO Cells , Cell Line , Cell Line, Tumor , Cricetulus , Drug Resistance, Neoplasm/genetics , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/genetics , HCT116 Cells , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Mutation/drug effects , Mutation/genetics
2.
Mol Cancer Ther ; 14(9): 2023-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26285778

ABSTRACT

Altiratinib (DCC-2701) was designed based on the rationale of engineering a single therapeutic agent able to address multiple hallmarks of cancer (1). Specifically, altiratinib inhibits not only mechanisms of tumor initiation and progression, but also drug resistance mechanisms in the tumor and microenvironment through balanced inhibition of MET, TIE2 (TEK), and VEGFR2 (KDR) kinases. This profile was achieved by optimizing binding into the switch control pocket of all three kinases, inducing type II inactive conformations. Altiratinib durably inhibits MET, both wild-type and mutated forms, in vitro and in vivo. Through its balanced inhibitory potency versus MET, TIE2, and VEGFR2, altiratinib provides an agent that inhibits three major evasive (re)vascularization and resistance pathways (HGF, ANG, and VEGF) and blocks tumor invasion and metastasis. Altiratinib exhibits properties amenable to oral administration and exhibits substantial blood-brain barrier penetration, an attribute of significance for eventual treatment of brain cancers and brain metastases.


Subject(s)
Aminopyridines/pharmacology , Anilides/pharmacology , Drug Resistance, Neoplasm , Neovascularization, Pathologic , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Receptor, TIE-2/antagonists & inhibitors , Tumor Microenvironment , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Aminopyridines/chemistry , Anilides/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Bevacizumab/chemistry , Bevacizumab/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Drug Design , Drug Therapy, Combination , Female , Hepatocyte Growth Factor/metabolism , Humans , Inhibitory Concentration 50 , Melanoma, Experimental , Mice , Models, Molecular , Molecular Conformation , Monocytes/drug effects , Monocytes/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/metabolism , Receptor, TIE-2/metabolism , Recombinant Proteins , Stromal Cells/drug effects , Stromal Cells/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
3.
J Biomol Screen ; 16(7): 724-33, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21521800

ABSTRACT

Transforming growth factor ß (TGF-ß) type I receptor (activin receptor-like kinase 5, ALK5) has been identified as a promising target for fibrotic diseases. To find a novel inhibitor of ALK5, the authors performed a high-throughput screen of a library of 420,000 compounds using dephosphorylated ALK5. From primary hits of 1521 compounds, 555 compounds were confirmed. In total, 124 compounds were then selected for follow-up based on their unique structures and other properties. Repeated concentration-response testing and final interference assays of the above compounds resulted in the discovery of a structurally novel ALK5 inhibitor (compound 8) (N-(thiophen 2-ylmethyl)-3-(3,4,5 trimethoxyphenyl)imidazo[1,2ß]pyridazin 6-amine) with a low IC(50) value of 0.7 µM. Compound 8 also inhibited the TGF-ß-induced nuclear translocation of SMAD with an EC(50) value of 0.8 µM. Kinetic analysis revealed that compound 8 inhibited ALK5 via mixed-type inhibition, suggesting that it may bind to ALK5 differently than other published adenosine triphosphate site inhibitors.


Subject(s)
High-Throughput Screening Assays , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Adenosine Diphosphate/metabolism , Cell Line, Tumor , Computer Simulation , Fluorescence Resonance Energy Transfer , Fluoroimmunoassay , Humans , Kinetics , Molecular Conformation , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Smad Proteins/metabolism , Small Molecule Libraries/pharmacology , Transforming Growth Factor beta/pharmacology
4.
Biochim Biophys Acta ; 1789(5): 422-31, 2009 May.
Article in English | MEDLINE | ID: mdl-19414071

ABSTRACT

Histone deacetylase 5 (HDAC5) represses expression of nuclear genes that promote cardiac hypertrophy. Agonism of a variety of G protein coupled receptors (GPCRs) triggers phosphorylation-dependent nuclear export of HDAC5 via the CRM1 nuclear export receptor, resulting in derepression of pro-hypertrophic genes. A cell-based high-throughput screen of a commercial compound collection was employed to identify compounds with the ability to preserve the nuclear fraction of GFP-HDAC5 in primary cardiomyocytes exposed to GPCR agonists. A hit compound potently inhibited agonist-induced GFP-HDAC5 nuclear export in cultured neonatal rat ventricular myocytes (NRVMs). A small set of related compounds was designed and synthesized to evaluate structure-activity relationship (SAR). The results demonstrated that inhibition of HDAC5 nuclear export was a result of compounds irreversibly reacting with a key cysteine residue in CRM1 that is required for its function. CRM1 inhibition by the compounds also resulted in potent suppression of cardiomyocyte hypertrophy. These studies define a novel class of anti-hypertrophic compounds that function through irreversible inhibition of CRM1-dependent nuclear export.


Subject(s)
Cardiomegaly/drug therapy , Histone Deacetylases/metabolism , Karyopherins/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Active Transport, Cell Nucleus/drug effects , Amides/pharmacology , Aniline Compounds/pharmacology , Animals , Cardiomegaly/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Histone Deacetylase Inhibitors , Histone Deacetylases/chemistry , Humans , Karyopherins/metabolism , Microscopy, Fluorescence , Myocytes, Cardiac/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , Structure-Activity Relationship , Exportin 1 Protein
5.
Assay Drug Dev Technol ; 6(6): 811-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19231942

ABSTRACT

For ultra-high-throughput screening, 10-30 nl of compound dissolved in 75% dimethyl sulfoxide (DMSO)/25% water (vol/vol) is spotted into 1,536- and 3,456-well ChemLib plates (Aurora Biotechnologies, Carlsbad, CA) and stored appropriately for a short time before screening. Although this practice eliminates the compound plating bottleneck, plated volumes of DMSO slowly evaporate from assay wells if plates are not properly stored in the interim. Since many assays are sensitive to DMSO concentrations, even slight evaporation may cause intra-plate variation and thus decrease assay quality. Using a cytochrome P450 3A4 Vivid Blue assay (Invitrogen, Carlsbad), we investigated the rate, pattern, and quantity of evaporation over a 1-year time frame to identify best practices for long-term (i.e., 6 months or greater) storage of assay-ready compound plates. Our findings regarding evaporation at plate edges indicate that nanospots preplated in ChemLib 1,536- or 3,456-well plates are best stored at -80 degrees C, in a bag, with or without the outer evaporation wells filled or at -20 degrees C, in a bag, with evaporation wells filled.


Subject(s)
Drug Evaluation, Preclinical/methods , Fluorometry/instrumentation , Fluorometry/methods , Preservation, Biological/methods , Biological Assay , Cytochrome P-450 CYP3A/metabolism , Dimethyl Sulfoxide/chemistry , Dimethyl Sulfoxide/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/trends , Fluorescent Dyes/chemistry , Humidity , Indicators and Reagents/chemistry , Nanospheres/analysis , Nanospheres/chemistry , Refrigeration , Robotics , Solvents/chemistry , Temperature , Time Factors , Volatilization
6.
J Biomol Screen ; 11(7): 736-42, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16928980

ABSTRACT

Elongation Factor P (EF-P) is an essential component of bacterial protein synthesis, enhancing the rate of translation by facilitating the addition of amino acids to the growing peptide chain. Using purified Staphylococcus aureus EF-P and a reconstituted Escherichia coli ribosomal system, an assay monitoring the addition of radiolabeled N-formyl methionine to biotinylated puromycin was developed. Reaction products were captured with streptavidin-coated scintillation proximity assay (SPA) beads and quantified by scintillation counting. Data from the assay were used to create a kinetic model of the reaction scheme. In this model, EF-P binding to the ribosome essentially doubled the rate of the ribosomal peptidyl transferase reaction. As described here, EF-P bound to the ribosomes with an apparent K(a) of 0.75 microM, and the substrates N-fMet-tRNA and biotinylated puromycin had apparent K(m)s of 19 microM and 0.5 microM, respectively. The assay was shown to be sensitive to a number of antibiotics known to target ribosomal peptide bond synthesis, such as chloramphenicol and puromycin, but not inhibitors that target other stages of protein synthesis, such as fusidic acid or thiostrepton.


Subject(s)
Anti-Bacterial Agents/analysis , Anti-Bacterial Agents/pharmacology , Drug Evaluation, Preclinical/methods , Microbial Sensitivity Tests , Peptide Elongation Factors/antagonists & inhibitors , Peptidyl Transferases/antagonists & inhibitors , Ribosomal Proteins/antagonists & inhibitors , Dose-Response Relationship, Drug , Escherichia coli/metabolism , Kinetics , Reproducibility of Results , Ribosomes/metabolism , Staphylococcus aureus/metabolism , Time Factors
7.
J Biomol Screen ; 8(3): 239-46, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12857377

ABSTRACT

High-throughput screening (HTS) has become an essential part of the drug discovery process. Due to the rising requirements for both data quality and quantity, along with increased screening cost and the demand to shorten the time for lead identification, increasing throughput and cost-effectiveness has become a necessity in the hit identification process. The authors present a multiplexed HTS for 2 nuclear receptors, the farnesoid X-activated receptor and the peroxisome proliferator-activated receptor delta in a viable cell-based reporter gene assay. The 2 nuclear receptors were individually transfected into human hepatoma cells, and the transient transfected cell lines were pooled for the multiplexed screen. Hits identified by the multiplexed screen are similar to those identified by the individual receptor screens. Furthermore, the multiplexed screen provides selectivity information if ligands selective for one and not the other receptor are one of the hit criteria. The data demonstrate that multiplexing nuclear receptors can be a simple, efficient, cost-effective, and reliable alternative to traditional HTS of individual targets without compromising data quality.


Subject(s)
Cell Nucleus/metabolism , Drug Design , Drug Evaluation, Preclinical/methods , Genes, Reporter , Cell Line, Tumor , Cell Survival , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Humans , Ligands , Luciferases/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Time Factors , Transcription Factors/metabolism , Transcriptional Activation , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...