Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
ChemMedChem ; 14(24): 2093-2101, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31697454

ABSTRACT

We identified novel potent inhibitors of p38 mitogen-activated protein (MAP) kinase using a structure-based design strategy, beginning with lead compound, 3-(butan-2-yl)-6-(2,4-difluoroanilino)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one (1). To enhance the inhibitory activity of 1 against production of tumor necrosis factor-α (TNF-α) in human whole blood (hWB) cell assays, we designed and synthesized hybrid compounds in which the imidazo[4,5-b]pyridin-2-one core was successfully linked with the p-methylbenzamide fragment. Among the compounds evaluated, 3-(3-tert-butyl-2-oxo-2,3-dihydro-1H-imidazo[4,5-b]pyridin-6-yl)-4-methyl-N-(1-methyl-1H-pyrazol-3-yl)benzamide (25) exhibited potent p38 inhibition, superior suppression of TNF-α production in hWB cells, and also significant in vivo efficacy in a rat model of collagen-induced arthritis (CIA). In this paper, we report the discovery of potent, selective, and orally bioavailable imidazo[4,5-b]pyridin-2-one-based p38 MAP kinase inhibitors.


Subject(s)
Arthritis, Experimental/drug therapy , Drug Design , Imidazoles/pharmacology , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Arthritis, Experimental/chemically induced , Cell Line , Collagen , Crystallography, X-Ray , Disease Models, Animal , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Models, Molecular , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Rats , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Appl Opt ; 58(19): 5288-5293, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31503627

ABSTRACT

We present mechanically tunable Tm/Ho-codoped fiber lasers based on multimode interference in single mode-multimode-single-mode structures. The intermediate multimode fiber (MMF) is separated into two segments with a liquid core in between. The mode field diameter of the single-mode fiber is designed to 13 µm for the MMF having a core diameter of 105 µm. The tunable range is attained to 64 nm by extending the effective length of the MMF. The laser wavelength is tuned over the ranges of 1809-1866 nm and 1866-1930 nm for the initial MMF lengths of 35.3 and 34.5 mm, respectively.

3.
ChemMedChem ; 14(10): 1022-1030, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30945818

ABSTRACT

We identified a lead series of p38 mitogen-activated protein kinase inhibitors using a structure-based design strategy from high-throughput screening of hit compound 1. X-ray crystallography of 1 with the kinase showed an infrequent flip of the peptide bond between Met109 and Gly110, which was considered to lead to high kinase selectivity. Our structure-based design strategy was to conduct scaffold transformation of 1 with maintenance of hydrogen bond interactions with the flipped hinge backbone of the enzyme. In accordance with this strategy, we focused on scaffold transformation to identify imidazo[4,5-b]pyridin-2-one derivatives as potent inhibitors of the p38 MAP kinase. Of the compounds evaluated, 21 was found to be a potent inhibitor of the p38 MAP kinase, lipopolysaccharide-induced tumor necrosis factor-α (TNF-α) production in human monocytic leukemia cells, and TNF-α-induced production of interleukin-8 in human whole blood cells. Herein we describe the discovery of potent and orally bioavailable imidazo[4,5-b]pyridin-2-one-based p38 MAP kinase inhibitors that suppressed cytokine production in a human whole blood cell-based assay.


Subject(s)
Antineoplastic Agents/chemistry , Imidazoles/chemistry , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Pyridones/chemistry , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Blood Cells , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Hydrogen Bonding , Imidazoles/chemical synthesis , Imidazoles/pharmacokinetics , Interleukin-8/metabolism , Lipopolysaccharides/chemistry , Models, Molecular , Molecular Structure , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Pyridones/pharmacokinetics , Rats , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism
4.
Bioorg Med Chem ; 26(3): 647-660, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29291937

ABSTRACT

We identified novel potent inhibitors of p38 MAP kinase using structure-based design strategy. X-ray crystallography showed that when p38 MAP kinase is complexed with TAK-715 (1) in a co-crystal structure, Phe169 adopts two conformations, where one interacts with 1 and the other shows no interaction with 1. Our structure-based design strategy shows that these two conformations converge into one via enhanced protein-ligand hydrophobic interactions. According to the strategy, we focused on scaffold transformation to identify imidazo[1,2-b]pyridazine derivatives as potent inhibitors of p38 MAP kinase. Among the herein described and evaluated compounds, N-oxide 16 exhibited potent inhibition of p38 MAP kinase and LPS-induced TNF-α production in human monocytic THP-1 cells, and significant in vivo efficacy in rat collagen-induced arthritis models. In this article, we report the discovery of potent, selective and orally bioavailable imidazo[1,2-b]pyridazine-based p38 MAP kinase inhibitors with pyridine N-oxide group.


Subject(s)
Drug Design , Protein Kinase Inhibitors/chemical synthesis , Pyridazines/chemistry , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Arthritis/drug therapy , Arthritis/etiology , Cell Line , Disease Models, Animal , Enzyme Activation/drug effects , Female , Humans , Molecular Dynamics Simulation , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Structure, Tertiary , Pyridazines/pharmacology , Pyridazines/therapeutic use , Rats , Rats, Inbred Lew , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Neurochem Res ; 42(11): 3279-3288, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28780732

ABSTRACT

Irregular N-methyl-D-aspartate receptor (NMDAR) function is one of the main hypotheses employed to facilitate understanding of the underlying disease state of schizophrenia. Although direct agonism of the NMDAR has not yielded promising therapeutics, advances have been made by modulating the NMDAR co-agonist site which is activated by glycine and D-serine. One approach to activate the co-agonist site is to increase synaptic D-serine levels through inhibition of D-amino acid oxidase (DAO), the major catabolic clearance pathway for this and other D-amino acids. A number of DAO inhibitors have been developed but most have not entered clinical trials. One exception to this is sodium benzoate which has demonstrated efficacy in small trials of schizophrenia and Alzheimer's disease. Herein we provide data on the effect of sodium benzoate and an optimised Takeda compound, PGM030756 on ex vivo DAO enzyme occupancy and cerebellar D-serine levels in mice. Both compounds achieve high levels of enzyme occupancy; although lower doses of PGM030756 (1, 3 and 10 mg/kg) were required to achieve this compared to sodium benzoate (300, 1000 mg/kg). Cerebellar D-serine levels were increased by both agents with a delay of approximately 6 h after dosing before the peak effect was achieved. Our data and methods may be useful in understanding the effects of sodium benzoate that have been seen in clinical trials of schizophrenia and Alzheimer's disease and to support the potential clinical assessment of other DAO inhibitors, such as PGM030756, which demonstrate good enzyme occupancy and D-serine increases following administration of low oral doses.


Subject(s)
Cerebellum/metabolism , Chlorobenzenes/pharmacology , D-Amino-Acid Oxidase/antagonists & inhibitors , D-Amino-Acid Oxidase/metabolism , Enzyme Inhibitors/pharmacology , Pyridazines/pharmacology , Serine/metabolism , Sodium Benzoate/pharmacology , Administration, Oral , Animals , Biomarkers/metabolism , Chlorobenzenes/administration & dosage , Chlorobenzenes/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Humans , Male , Mice , Mice, Inbred C57BL , Pyridazines/administration & dosage , Pyridazines/chemistry , Sodium Benzoate/administration & dosage , Sodium Benzoate/chemistry
6.
Biochem Biophys Res Commun ; 488(2): 393-399, 2017 06 24.
Article in English | MEDLINE | ID: mdl-28501621

ABSTRACT

Prolyl-tRNA synthetase (PRS) is a member of the aminoacyl-tRNA synthetase family of enzymes and catalyzes the synthesis of prolyl-tRNAPro using ATP, l-proline, and tRNAPro as substrates. An ATP-dependent PRS inhibitor, halofuginone, was shown to suppress autoimmune responses, suggesting that the inhibition of PRS is a potential therapeutic approach for inflammatory diseases. Although a few PRS inhibitors have been derivatized from natural sources or substrate mimetics, small-molecule human PRS inhibitors have not been reported. In this study, we discovered a novel series of pyrazinamide PRS inhibitors from a compound library using pre-transfer editing activity of human PRS enzyme. Steady-state biochemical analysis on the inhibitory mode revealed its distinctive characteristics of inhibition with proline uncompetition and ATP competition. The binding activity of a representative compound was time-dependently potentiated by the presence of l-proline with Kd of 0.76 nM. Thermal shift assays demonstrated the stabilization of PRS in complex with l-proline and pyrazinamide PRS inhibitors. The binding mode of the PRS inhibitor to the ATP site of PRS enzyme was elucidated using the ternary complex crystal structure with l-proline. The results demonstrated the different inhibitory and binding mode of pyrazinamide PRS inhibitors from preceding halofuginone. Furthermore, the PRS inhibitor inhibited intracellular protein synthesis via a different mode than halofuginone. In conclusion, we have identified a novel drug-like PRS inhibitor with a distinctive binding mode. This inhibitor was effective in a cellular context. Thus, the series of PRS inhibitors are considered to be applicable to further development with differentiation from preceding halofuginone.


Subject(s)
Adenosine Triphosphate/metabolism , Amino Acyl-tRNA Synthetases/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/pharmacology , Proline/metabolism , Pyrazinamide/pharmacology , Amino Acyl-tRNA Synthetases/metabolism , Binding Sites/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , Molecular Structure , Pyrazinamide/chemical synthesis , Pyrazinamide/chemistry , Structure-Activity Relationship
7.
Mol Cancer Ther ; 16(7): 1269-1278, 2017 07.
Article in English | MEDLINE | ID: mdl-28341789

ABSTRACT

Receptor tyrosine kinase therapies have proven to be efficacious in specific cancer patient populations; however, a significant limitation of tyrosine kinase inhibitor (TKI) treatment is the emergence of resistance mechanisms leading to a transient, partial, or complete lack of response. Combination therapies using agents with synergistic activity have potential to improve response and reduce acquired resistance. Chemoreagent or TKI treatment can lead to increased expression of hepatocyte growth factor (HGF) and/or MET, and this effect correlates with increased metastasis and poor prognosis. Despite MET's role in resistance and cancer biology, MET TKI monotherapy has yielded disappointing clinical responses. In this study, we describe the biological activity of a selective, oral MET TKI with slow off-rate and its synergistic antitumor effects when combined with an anti-HGF antibody. We evaluated the combined action of simultaneously neutralizing HGF ligand and inhibiting MET kinase activity in two cancer xenograft models that exhibit autocrine HGF/MET activation. The combination therapy results in additive antitumor activity in KP4 pancreatic tumors and synergistic activity in U-87MG glioblastoma tumors. Pharmacodynamic characterization of biomarkers that correlate with combination synergy reveal that monotherapies induce an increase in the total MET protein, whereas combination therapy significantly reduces total MET protein levels and phosphorylation of 4E-BP1. These results hold promise that dual targeting of HGF and MET by combining extracellular ligand inhibitors with intracellular MET TKIs could be an effective intervention strategy for cancer patients who have acquired resistance that is dependent on total MET protein. Mol Cancer Ther; 16(7); 1269-78. ©2017 AACR.


Subject(s)
Glioblastoma/drug therapy , Hepatocyte Growth Factor/genetics , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-met/genetics , Small Molecule Libraries/administration & dosage , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Drug Synergism , Glioblastoma/genetics , Hepatocyte Growth Factor/antagonists & inhibitors , Humans , Mice , Phosphoproteins/genetics , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Xenograft Model Antitumor Assays
8.
Environ Pollut ; 209: 46-52, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26629645

ABSTRACT

Understanding the mechanisms of (137)Cs movement across different ecosystems is crucial for projecting the environmental impact and management of nuclear contamination events. Here, we report differential movement of (137)Cs in adjacent forest and stream ecosystems. The food webs of the forest and stream ecosystems in our study were similar, in that they were both dominated by detrital-based food webs and the basal energy source was terrestrial litter. However, the concentration of (137)Cs in stream litter was significantly lower than in forest litter, the result of (137)Cs leaching from litter in stream water. The difference in (137)Cs concentrations between the two types of litter was reflected in the (137)Cs concentrations in the animal community. While the importance of (137)Cs fallout and the associated transfer to food webs has been well studied, research has been primarily limited to cases in a single ecosystem. Our results indicate that there are differences in the flow of (137)Cs through terrestrial and aquatic ecosystems, and that (137)Cs concentrations are reduced in both basal food resources and higher trophic animals in aquatic systems, where primary production is subsidized by a neighboring terrestrial ecosystem.


Subject(s)
Cesium Radioisotopes/analysis , Ecosystem , Rivers/chemistry , Trees/chemistry , Animals , Food Chain , Forests
9.
Kyobu Geka ; 67(13): 1183-5, 2014 Dec.
Article in Japanese | MEDLINE | ID: mdl-25434547

ABSTRACT

A 37-year-old woman, who had undergone surgery of atrial septal defect (ASD) at 12-year-old, developed bradycardia and referred to our hospital. Transthoracic echocardiography revealed high echoic tumor in the right atrium. The image of the tumor was of low intensity by T2 weighted magnetic resonance imaging (MRI) and floating mass with a stalk to the right atrium in cine MRI. She underwent tumor resection under cardiopulmonary bypass. Histopathologilal examination of the tumor was calcified amorphous tumor. The postoperative course was uneventful.


Subject(s)
Calcinosis/surgery , Heart Atria/surgery , Heart Neoplasms/surgery , Adult , Calcinosis/etiology , Female , Heart Atria/pathology , Heart Neoplasms/complications , Heart Neoplasms/pathology , Humans , Magnetic Resonance Imaging , Tomography, X-Ray Computed
10.
Nature ; 513(7516): 124-7, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25043059

ABSTRACT

Human GPR40 receptor (hGPR40), also known as free fatty-acid receptor 1 (FFAR1), is a G-protein-coupled receptor that binds long-chain free fatty acids to enhance glucose-dependent insulin secretion. Novel treatments for type-2 diabetes mellitus are therefore possible by targeting hGPR40 with partial or full agonists. TAK-875, or fasiglifam, is an orally available, potent and selective partial agonist of hGPR40 receptor, which reached phase III clinical trials for the potential treatment of type-2 diabetes mellitus. Data from clinical studies indicate that TAK-875, which is an ago-allosteric modulator of hGPR40 (ref. 3), demonstrates improved glycaemic control and low hypoglycaemic risk in diabetic patients. Here we report the crystal structure of hGPR40 receptor bound to TAK-875 at 2.3 Å resolution. The co-complex structure reveals a unique binding mode of TAK-875 and suggests that entry to the non-canonical binding pocket most probably occurs via the lipid bilayer. The atomic details of the extensive charge network in the ligand binding pocket reveal additional interactions not identified in previous studies and contribute to a clear understanding of TAK-875 binding to the receptor. The hGPR40-TAK-875 structure also provides insights into the plausible binding of multiple ligands to the receptor, which has been observed in radioligand binding and Ca(2+) influx assay studies. Comparison of the transmembrane helix architecture with other G-protein-coupled receptors suggests that the crystallized TAK-875-bound hGPR40 complex is in an inactive-like state.


Subject(s)
Benzofurans/chemistry , Benzofurans/pharmacology , Drug Partial Agonism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Sulfones/chemistry , Sulfones/pharmacology , Allosteric Regulation/drug effects , Benzofurans/metabolism , Binding Sites , Crystallography, X-Ray , Diabetes Mellitus, Type 2/drug therapy , Humans , Ligands , Lipid Bilayers/metabolism , Models, Molecular , Receptors, G-Protein-Coupled/metabolism , Structural Homology, Protein , Sulfones/metabolism , Surface Properties
11.
Mol Membr Biol ; 31(5): 168-75, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25068810

ABSTRACT

Elucidating the detailed mechanism of activation of membrane protein receptors and their ligand binding is essential for structure-based drug design. Membrane protein crystal structure analysis successfully aids in understanding these fundamental molecular interactions. However, protein crystal structure analysis of the G-protein-coupled receptor (GPCR) remains challenging, even for the class of GPCRs which have been included in the majority of structure analysis reports among membrane proteins, due to the substantial instability of these receptors when extracted from lipid bilayer membranes. It is known that increased thermostability tends to decrease conformational flexibility, which contributes to the generation of diffraction quality crystals. However, this is still not straightforward, and significant effort is required to identify thermostabilized mutants that are optimal for crystallography. To address this issue, a versatile screening platform based on a label-free ligand binding assay combined with transient overexpression in virus-like particles was developed. This platform was used to generate thermostabilized GPR40 [also known as free fatty acid receptor 1 (FFAR1)] for fasiglifam (TAK-875). This demonstrated that the thermostabilized mutant GPR40 (L42A/F88A/G103A/Y202F) was successfully used for crystal structure analysis.


Subject(s)
Benzofurans/chemistry , Membrane Proteins/chemistry , Receptors, G-Protein-Coupled/chemistry , Sulfones/chemistry , Benzofurans/metabolism , Cell Line , Humans , Ligands , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Proteins/metabolism , Mutation , Protein Binding , Protein Stability , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Staining and Labeling , Sulfones/metabolism , Temperature
12.
J Med Chem ; 57(12): 5459-63, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24847974

ABSTRACT

Catechol O-methyl transferase belongs to the diverse family of S-adenosyl-l-methionine transferases. It is a target involved in the treatment of Parkinson's disease. Here we present a fragment-based screening approach to discover noncatechol derived COMT inhibitors which bind at the SAM binding pocket. We describe the identification and characterization of a series of highly ligand efficient SAM competitive bisaryl fragments (LE = 0.33-0.58). We also present the first SAM-competitive small-molecule COMT co-complex crystal structure.


Subject(s)
Catechol O-Methyltransferase Inhibitors , S-Adenosylmethionine/metabolism , Animals , Binding Sites , Catechol O-Methyltransferase/chemistry , Humans , Kinetics , Mice , Models, Molecular , Protein Conformation , Pyrazoles/chemistry , Rats , S-Adenosylmethionine/chemistry , Structure-Activity Relationship , Thiazoles/chemistry , Triazoles/chemistry
13.
Bioorg Med Chem ; 21(24): 7686-98, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24216091

ABSTRACT

To identify compounds with potent antitumor efficacy for various human cancers, we aimed to synthesize compounds that could inhibit c-mesenchymal epithelial transition factor (c-Met) and vascular endothelial growth factor receptor 2 (VEGFR2) kinases. We designed para-substituted inhibitors by using co-crystal structural information from c-Met and VEGFR2 in complex with known inhibitors. This led to the identification of compounds 3a and 3b, which were capable of suppressing both c-Met and VEGFR2 kinase activities. Further optimization resulted in pyrazolone and pyridone derivatives, which could form intramolecular hydrogen bonds to enforce a rigid conformation, thereby producing potent inhibition. One compound of particular note was the imidazo[1,2-a]pyridine derivative (26) bearing a 6-methylpyridone ring, which strongly inhibited both c-Met and VEGFR2 enzyme activities (IC50=1.9, 2.2 nM), as well as proliferation of c-Met-addicted MKN45 cells and VEGF-stimulated human umbilical vein endothelial cells (IC50=5.0, 1.8 nM). Compound 26 exhibited dose-dependent antitumor efficacy in vivo in MKN45 (treated/control ratio [T/C]=4%, po, 5mg/kg, once-daily) and COLO205 (T/C=13%, po, 15 mg/kg, once-daily) mouse xenograft models.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Heterocyclic Compounds, 2-Ring/pharmacology , Niacinamide/analogs & derivatives , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Heterocyclic Compounds, 2-Ring/chemistry , Heterocyclic Compounds, 2-Ring/metabolism , Humans , Mice , Mice, Inbred BALB C , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Niacinamide/chemistry , Niacinamide/metabolism , Niacinamide/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Proto-Oncogene Proteins c-met/metabolism , Pyridines/chemistry , Pyridines/metabolism , Solubility , Structure-Activity Relationship , Vascular Endothelial Growth Factor Receptor-2/metabolism
14.
Bioorg Med Chem ; 21(8): 2333-2345, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23498918

ABSTRACT

Vascular endothelial growth factor (VEGF) plays important roles in tumor angiogenesis, and the inhibition of its signaling pathway is considered an effective therapeutic option for the treatment of cancer. In this study, we describe the design, synthesis, and biological evaluation of 2-acylamino-6-phenoxy-imidazo[1,2-b]pyridazine derivatives. Hybridization of two distinct imidazo[1,2-b]pyridazines 1 and 2, followed by optimization led to the discovery of N-[5-({2-[(cyclopropylcarbonyl)amino]imidazo[1,2-b]pyridazin-6-yl}oxy)-2-methylphenyl]-1,3-dimethyl-1H-pyrazole-5-carboxamide (23a, TAK-593) as a highly potent VEGF receptor 2 kinase inhibitor with an IC50 value of 0.95 nM. The compound 23a strongly suppressed proliferation of VEGF-stimulated human umbilical vein endothelial cells with an IC50 of 0.30 nM. Kinase selectivity profiling revealed that 23a inhibited platelet-derived growth factor receptor kinases as well as VEGF receptor kinases. Oral administration of 23a at 1 mg/kg bid potently inhibited tumor growth in a mouse xenograft model using human lung adenocarcinoma A549 cells (T/C=8%).


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Aminoimidazole Carboxamide/chemistry , Aminoimidazole Carboxamide/pharmacology , Animals , Disease Models, Animal , Female , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred AKR , Mice, Nude , Models, Molecular , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Rats , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
15.
Bioorg Med Chem Lett ; 22(24): 7326-9, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23147077

ABSTRACT

Imidazo[1,2-a]pyridine derivatives were designed, synthesized, and evaluated as inhibitors of the apoptosis signal-regulating kinase 1 (ASK1). These were based on a benzothiazole derivative that was discovered from high-throughput screening of our compound library. As a result, we identified potent, selective, and orally bioavailable ASK1 inhibitors for wide range of therapeutic targets.


Subject(s)
Drug Design , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , High-Throughput Screening Assays , MAP Kinase Kinase Kinase 5/metabolism , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyridines/chemical synthesis , Pyridines/chemistry , Rats , Structure-Activity Relationship
16.
ACS Med Chem Lett ; 3(4): 342-6, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-24900475

ABSTRACT

We present a straightforward process for the discovery of novel back pocket-binding fragment molecules against protein tyrosine kinases. The approach begins by screening against the nonphosphorylated target kinase with subsequent counterscreening of hits against the phosphorylated enzyme. Back pocket-binding fragments are inactive against the phosphorylated kinase. Fragment molecules are of insufficient size to span both regions of the ATP binding pocket; thus, the outcome is binary (back pocket-binding or hinge-binding). Next, fragments with the appropriate binding profile are assayed in combination with a known hinge-binding fragment and subsequently with a known back pocket-binding fragment. Confirmation of back pocket-binding by Yonetani-Theorell plot analysis progresses candidate fragments to crystallization trials. The method is exemplified by a fragment screening campaign against vascular endothelial growth factor receptor 2, and a novel back pocket-binding fragment is presented.

18.
Bioorg Med Chem ; 18(20): 7260-73, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20833055

ABSTRACT

We synthesized a series of pyrrolo[3,2-d]pyrimidine derivatives and evaluated their application as type-II inhibitors of vascular endothelial growth factor receptor 2 (VEGFR2) kinase. Incorporation of a diphenylurea moiety at the C4-position of the pyrrolo[3,2-d]pyrimidine core via an oxygen linker resulted in compounds that were potent inhibitors of VEGFR2 kinase. Of these derivatives, compound 20d showed the strongest inhibition of VEGF-stimulated proliferation of human umbilical vein endothelial cells (HUVEC). The co-crystal structure of 20d and VEGFR2 revealed that 20d binds to the inactive form of VEGFR2. Further studies indicated that 20d inhibited VEGFR2 kinase with slow dissociation kinetics and also inhibited PDGFR and Tie-2 kinases. Oral administration of the hydrochloride salt of 20d at 3mg/kg/day showed potent inhibition of tumor growth in a DU145 human prostate cancer cell xenograft nude mouse model.


Subject(s)
Antineoplastic Agents/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Phenylurea Compounds/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Pyrimidines/chemistry , Pyrroles/chemistry , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Binding Sites , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cells, Cultured , Crystallography, X-Ray , Drug Design , Humans , Mice , Mice, Nude , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacology , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/toxicity , Protein Structure, Tertiary , Pyrimidines/chemical synthesis , Pyrimidines/toxicity , Pyrroles/chemical synthesis , Pyrroles/toxicity , Receptor, TIE-2/antagonists & inhibitors , Receptor, TIE-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
19.
Bioorg Med Chem ; 18(20): 7150-63, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20833551

ABSTRACT

We have recently reported the discovery of pyrrolo[3,2-d]pyrimidine derivatives 1a and 1b as potent triple inhibitors of vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR), and Tie-2 kinases. To identify compounds having strong inhibitory activity against fibroblast growth factor receptor (FGFR) kinase, further modification was conducted using the co-crystal structure analysis of VEGFR2 and 1b. Among the compounds synthesized, urea derivative 11l having a piperazine moiety on the terminal benzene ring showed strong inhibitory activity against FGFR1 kinase as well as VEGFR2 kinase. A binding model of 11l complexed with VEGFR2 suggested that the piperazine moiety forms additional interactions with Ile1025 and His1026.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/chemistry , Phenylurea Compounds/chemistry , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemistry , Pyrroles/chemistry , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Urea/analogs & derivatives , Binding Sites , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cells, Cultured , Computer Simulation , Crystallography, X-Ray , Drug Design , Humans , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Receptor, TIE-2/antagonists & inhibitors , Receptor, TIE-2/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Structure-Activity Relationship , Urea/chemical synthesis , Urea/pharmacology
20.
Chem Commun (Camb) ; 46(10): 1763-5, 2010 Mar 14.
Article in English | MEDLINE | ID: mdl-20177642

ABSTRACT

Arynes are found to be facilely inserted into bis(pinacolato)diboron by using a platinum-isocyanide catalyst, affording diverse 1,2-diborylarenes, which can be converted into o-terphenyls via Suzuki-Miyaura coupling reaction.

SELECTION OF CITATIONS
SEARCH DETAIL