Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Med Chem ; 66(9): 6354-6371, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37120845

ABSTRACT

A novel series of non-amidine-based C1s inhibitors have been explored. Starting from high-throughput screening hit 3, isoquinoline was replaced with 1-aminophthalazine to enhance C1s inhibitory activity while exhibiting good selectivity against other serine proteases. We first disclose a crystal structure of a complex of C1s and a small-molecule inhibitor (4e), which guided structure-based optimization around the S2 and S3 sites to further enhance C1s inhibitory activity by over 300-fold. Improvement of membrane permeability by incorporation of fluorine at the 8-position of 1-aminophthalazine led to identification of (R)-8 as a potent, selective, orally available, and brain-penetrable C1s inhibitor. (R)-8 significantly inhibited membrane attack complex formation induced by human serum in a dose-dependent manner in an in vitro assay system, proving that selective C1s inhibition blocked the classical complement pathway effectively. As a result, (R)-8 emerged as a valuable tool compound for both in vitro and in vivo assessment.


Subject(s)
Complement Activation , Complement C1s , Humans , Complement C1s/chemistry , Complement C1s/metabolism , Serine Endopeptidases/metabolism , Brain/metabolism
2.
J Med Chem ; 64(2): 1103-1115, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33404239

ABSTRACT

O-GlcNAcase (OGA) has received increasing attention as an attractive therapeutic target for tau-mediated neurodegenerative disorders; however, its role in these pathologies remains unclear. Therefore, potent chemical tools with favorable pharmacokinetic profiles are desirable to characterize this enzyme. Herein, we report the discovery of a potent and novel OGA inhibitor, compound 5i, comprising an aminopyrimidine scaffold, identified by virtual screening based on multiple methodologies combining structure-based and ligand-based approaches, followed by sequential optimization with a focus on ligand lipophilicity efficiency. This compound was observed to increase the level of O-GlcNAcylated protein in cells and display suitable pharmacokinetic properties and brain permeability. Crystallographic analysis revealed that the chemical series bind to OGA via characteristic hydrophobic interactions, which resulted in a high affinity for OGA with moderate lipophilicity. Compound 5i could serve as a useful chemical probe to help establish a proof-of-concept of OGA inhibition as a therapeutic target for the treatment of tauopathies.


Subject(s)
Acetylglucosamine/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , beta-N-Acetylhexosaminidases/antagonists & inhibitors , Animals , Brain/metabolism , Cell Line , Computer Simulation , Drug Design , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacokinetics , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Neuroprotective Agents/pharmacokinetics , Structure-Activity Relationship , Tauopathies/drug therapy
3.
Biochem Biophys Res Commun ; 523(3): 795-801, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31954521

ABSTRACT

The DEAD-box family of RNA helicases plays essential roles in both transcriptional and translational mRNA degradation; they unwind short double-stranded RNA by breaking the RNA-RNA interactions. Two DEAD-box RNA helicases, eukaryotic translation initiation factor 4A3 (eIF4A3) and DEAD-box helicase 3 (DDX3X), show high homology in the ATP-binding region and are considered key molecules for cancer progression. Several small molecules that target eIF4A3 and DDX3X have been reported to inhibit cancer cell growth; however, more potent compounds are required for cancer therapeutics, and there is a critical need for high-throughput assays to screen for RNA helicase inhibitors. In this study, we developed novel fluorescence resonance energy transfer-based high-throughput RNA helicase assays for eIF4A3 and DDX3X. Using these assays, we identified several eIF4A3 allosteric inhibitors whose inhibitory effect on eIF4A3 ATPase showed a strong correlation with inhibitory effect on helicase activity. From 102 compounds that exhibited eIF4A3 ATPase inhibition, we identified a selective DDX3X inhibitor, C1, which showed stronger inhibition of DDX3X than of eIF4A3. Small-molecule helicase inhibitors can be valuable for clarifying the molecular machinery of DEAD-box RNA helicases. The high-throughput quantitative assays established here should facilitate the evaluation of the helicase inhibitory activity of compounds.


Subject(s)
DEAD-box RNA Helicases/antagonists & inhibitors , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Small Molecule Libraries/pharmacology , DEAD-box RNA Helicases/metabolism , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Enzyme Assays/methods , Eukaryotic Initiation Factor-4A/metabolism , High-Throughput Screening Assays , Humans , Small Molecule Libraries/chemistry
4.
J Med Chem ; 62(20): 9175-9187, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31550153

ABSTRACT

Dysferlinopathies, which are muscular diseases caused by mutations in the dysferlin gene, remain serious medical problems due to the lack of therapeutic agents. Herein, we report the design, synthesis, and structure-activity relationships of a 2,6-disubstituted 3H-imidazo[4,5-b]pyridine series, which was identified from the phenotypic screening of chemicals that increase the level of dysferlin in myocytes differentiated from patient-derived induced pluripotent stem cells (iPSCs). Optimization studies with cell-based phenotypic assay led to the identification of a highly potent compound, 19, with dysferlin elevation effects at double-digit nanomolar concentrations. In addition, the molecular target of our chemical series was identified as tubulin, through a tubulin polymerization assay and a competitive binding assay using a photoaffinity labeling probe.


Subject(s)
Imidazoles/chemistry , Muscular Dystrophies, Limb-Girdle/drug therapy , Pyridines/chemistry , Tubulin Modulators/therapeutic use , Binding Sites , Cell Differentiation , Cell Proliferation/drug effects , Drug Design , Dysferlin/metabolism , Hep G2 Cells , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Molecular Docking Simulation , Muscular Dystrophies, Limb-Girdle/pathology , MyoD Protein/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Protein Structure, Tertiary , Pyridines/pharmacology , Pyridines/therapeutic use , Structure-Activity Relationship , Tubulin/chemistry , Tubulin/metabolism , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
5.
Oncotarget ; 9(26): 18480-18493, 2018 Apr 06.
Article in English | MEDLINE | ID: mdl-29719619

ABSTRACT

Protein arginine methyltransferase (PRMT) 4 (also known as coactivator-associated arginine methyltransferase 1; CARM1) is involved in a variety of biological processes and is considered as a candidate oncogene owing to its overexpression in several types of cancer. Selective PRMT4 inhibitors are useful tools for clarifying the molecular events regulated by PRMT4 and for validating PRMT4 as a therapeutic target. Here, we report the discovery of TP-064, a potent, selective, and cell-active chemical probe of human PRMT4 and its co-crystal structure with PRMT4. TP-064 inhibited the methyltransferase activity of PRMT4 with high potency (half-maximal inhibitory concentration, IC50 < 10 nM) and selectivity over other PRMT family proteins, and reduced arginine dimethylation of the PRMT4 substrates BRG1-associated factor 155 (BAF155; IC50= 340 ± 30 nM) and Mediator complex subunit 12 (MED12; IC50 = 43 ± 10 nM). TP-064 treatment inhibited the proliferation of a subset of multiple myeloma cell lines, with affected cells arrested in G1 phase of the cell cycle. TP-064 and its negative control (TP-064N) will be valuable tools to further investigate the biology of PRMT4 and the therapeutic potential of PRMT4 inhibition.

6.
Bioorg Med Chem ; 26(2): 470-482, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29258712

ABSTRACT

A series of tetrahydroisoquinoline derivatives were designed, synthesized, and evaluated for their potential as novel orally efficacious retinoic acid receptor-related orphan receptor-gamma t (RORγt) inverse agonists for the treatment of Th17-driven autoimmune diseases. We carried out cyclization of the phenylglycinamide core by structure-based drug design and successfully identified a tetrahydroisoquinoline carboxylic acid derivative 14 with good biochemical binding and cellular reporter activity. Interestingly, the combination of a carboxylic acid tether and a central fused bicyclic ring was crucial for optimizing PK properties, and the compound 14 showed significantly improved PK profile. Successive optimization of the carboxylate tether led to the discovery of compound 15 with increased inverse agonistic activity and an excellent PK profile. Oral treatment of mice with compound 15 robustly and dose-dependently inhibited IL-17A production in an IL23-induced gene expression assay.


Subject(s)
Drug Discovery , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Tetrahydroisoquinolines/pharmacology , Administration, Oral , Animals , Crystallography, X-Ray , Cytokines/biosynthesis , Dose-Response Relationship, Drug , Humans , Injections, Intradermal , Interleukin-23/administration & dosage , Interleukin-23/pharmacology , Jurkat Cells , Male , Mice , Mice, Inbred BALB C , Models, Animal , Models, Molecular , Molecular Structure , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Structure-Activity Relationship , Tetrahydroisoquinolines/administration & dosage , Tetrahydroisoquinolines/chemistry
7.
Bioorg Med Chem Lett ; 27(15): 3565-3571, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28579121

ABSTRACT

We report a design strategy to obtain potent DPP-4 inhibitors by incorporating salt bridge formation with Lys554 in the S1' pocket. By applying the strategy to the previously identified templates, quinoline 4 and pyridines 16a, 16b, and 17 have been identified as subnanomolar or nanomolar inhibitors of human DPP-4. Docking studies suggested that a hydrophobic interaction with Tyr547 as well as the salt bridge interaction is important for the extremely high potency. The design strategy would be useful to explore a novel design for DPP-4 inhibitors having a distinct structure with a unique binding mode.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Quinolines/chemistry , Quinolines/pharmacology , Animals , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl Peptidase 4/chemistry , Dipeptidyl Peptidase 4/metabolism , Drug Design , Female , Glucose Tolerance Test , Humans , Molecular Docking Simulation , Rats, Sprague-Dawley , Rats, Wistar , Structure-Activity Relationship
8.
Bioorg Med Chem ; 25(14): 3768-3779, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28571972

ABSTRACT

A lead compound A was identified previously as an stearoyl coenzyme A desaturase (SCD) inhibitor during research on potential treatments for obesity. This compound showed high SCD1 binding affinity, but a poor pharmacokinetic (PK) profile and limited chemical accessibility, making it suboptimal for use in anticancer research. To identify potent SCD1 inhibitors with more promising PK profiles, we newly designed a series of 'non-spiro' 4, 4-disubstituted piperidine derivatives based on molecular modeling studies. As a result, we discovered compound 1a, which retained moderate SCD1 binding affinity. Optimization around 1a was accelerated by analyzing Hansch-Fujita and Hammett constants to obtain 4-phenyl-4-(trifluoromethyl)piperidine derivative 1n. Fine-tuning of the azole moiety of 1n led to compound 1o (T-3764518), which retained nanomolar affinity and exhibited an excellent PK profile. Reflecting the good potency and PK profile, orally administrated compound 1o showed significant pharmacodynamic (PD) marker reduction (at 0.3mg/kg, bid) in HCT116 mouse xenograft model and tumor growth suppression (at 1mg/kg, bid) in 786-O mouse xenograft model. In conclusion, we identified a new series of SCD1 inhibitors, represented by compound 1o, which represents a promising new chemical tool suitable for the study of SCD1 biology as well as the potential development of novel anticancer therapies.


Subject(s)
Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemical synthesis , Oxadiazoles/chemical synthesis , Pyridazines/chemical synthesis , Stearoyl-CoA Desaturase/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Colonic Neoplasms/drug therapy , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , HCT116 Cells , Humans , Inhibitory Concentration 50 , Mice , Mice, Inbred BALB C , Mice, Nude , Microsomes, Liver/metabolism , Oxadiazoles/pharmacokinetics , Oxadiazoles/therapeutic use , Oxadiazoles/toxicity , Piperidines/chemistry , Piperidines/metabolism , Piperidines/pharmacology , Protein Binding , Pyridazines/pharmacokinetics , Pyridazines/therapeutic use , Pyridazines/toxicity , Spiro Compounds/chemistry , Stearoyl-CoA Desaturase/metabolism , Structure-Activity Relationship , Transplantation, Heterologous
9.
Bioorg Med Chem ; 24(22): 5771-5780, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27687967

ABSTRACT

The action of the aspartyl protease renin is the rate-limiting initial step of the renin-angiotensin-aldosterone system. Therefore, renin is a particularly promising target for blood pressure as well as onset and progression of cardiovascular and renal diseases. New pyrimidine derivatives 5-14 were designed in an attempt to enhance the renin inhibitory activity of compound 3 identified by our previous fragment-based drug design approach. Introduction of a basic amine essential for interaction with the two aspartic acids in the catalytic site and optimization of the S1/S3 binding elements including an induced-fit structural change of Leu114 ('Leu-in' to 'Leu-out') by a rational structure-based drug design approach led to the discovery of N-(piperidin-3-yl)pyrimidine-5-carboxamide 14, a 65,000-fold more potent renin inhibitor than compound 3. Surprisingly, this remarkable enhancement in the inhibitory activity of compound 14 has been achieved by the overall addition of only seven heavy atoms to compound 3. Compound 14 demonstrated excellent selectivity over other aspartyl proteases and moderate oral bioavailability in rats.


Subject(s)
Drug Design , Piperidines/pharmacology , Protease Inhibitors/pharmacology , Pyrimidines/pharmacology , Renin/antagonists & inhibitors , Animals , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Piperidines/chemical synthesis , Piperidines/chemistry , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Rats , Rats, Sprague-Dawley , Renin/blood , Structure-Activity Relationship
10.
Bioorg Med Chem ; 24(22): 6066-6074, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27720325

ABSTRACT

A novel approach was conducted for fragment-based lead discovery and applied to renin inhibitors. The biochemical screening of a fragment library against renin provided the hit fragment which showed a characteristic interaction pattern with the target protein. The hit fragment bound only to the S1, S3, and S3SP (S3 subpocket) sites without any interactions with the catalytic aspartate residues (Asp32 and Asp215 (pepsin numbering)). Prior to making chemical modifications to the hit fragment, we first identified its essential binding sites by utilizing the hit fragment's substructures. Second, we created a new and smaller scaffold, which better occupied the identified essential S3 and S3SP sites, by utilizing library synthesis with high-throughput chemistry. We then revisited the S1 site and efficiently explored a good building block attaching to the scaffold with library synthesis. In the library syntheses, the binding modes of each pivotal compound were determined and confirmed by X-ray crystallography and the library was strategically designed by structure-based computational approach not only to obtain a more active compound but also to obtain informative Structure Activity Relationship (SAR). As a result, we obtained a lead compound offering synthetic accessibility as well as the improved in vitro ADMET profiles. The fragments and compounds possessing a characteristic interaction pattern provided new structural insights into renin's active site and the potential to create a new generation of renin inhibitors. In addition, we demonstrated our FBDD strategy integrating highly sensitive biochemical assay, X-ray crystallography, and high-throughput synthesis and in silico library design aimed at fragment morphing at the initial stage was effective to elucidate a pocket profile and a promising lead compound.


Subject(s)
Drug Discovery , Protease Inhibitors/pharmacology , Renin/antagonists & inhibitors , Animals , CHO Cells , Cell Survival/drug effects , Cricetulus , Crystallography, X-Ray , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Models, Molecular , Molecular Structure , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Renin/metabolism , Structure-Activity Relationship
11.
J Med Chem ; 56(23): 9635-45, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24215352

ABSTRACT

Mcl-1 and Bcl-xL are crucial regulators of apoptosis, therefore dual inhibitors of both proteins could serve as promising new anticancer drugs. To design Mcl-1/Bcl-xL dual inhibitors, we performed structure-guided analyses of the corresponding selective Mcl-1 and Bcl-xL inhibitors. A cocrystal structure of a pyrazolo[1,5-a]pyridine derivative with Mcl-1 protein was successfully determined and revealed the protein-ligand binding mode. The key structure for Bcl-xL inhibition was further confirmed through the substructural analysis of ABT-263, a representative Bcl-xL/Bcl-2/Bcl-w inhibitor developed by Abbott Laboratories. On the basis of the structural data from this analysis, we designed hybrid compounds by tethering the Mcl-1 and Bcl-xL inhibitors together. The results of X-ray crystallographic analysis of hybrid compound 10 in complexes with both Mcl-1 and Bcl-xL demonstrated its binding mode with each protein. Following further optimization, compound 11 showed potent Mcl-1/Bcl-xL dual inhibitory activity (Mcl-1, IC50 = 0.088 µM; and Bcl-xL, IC50 = 0.0037 µM).


Subject(s)
Apoptosis/drug effects , Biphenyl Compounds/chemical synthesis , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Sulfonamides/chemical synthesis , bcl-X Protein/antagonists & inhibitors , Aniline Compounds/pharmacology , Apoptosis Regulatory Proteins/metabolism , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Crystallography, X-Ray , Drug Design , Humans , Models, Molecular , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology
12.
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
13.
Bioorg Med Chem ; 21(15): 4714-29, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23755884

ABSTRACT

For the purpose of discovering novel type-II inhibitors of vascular endothelial growth factor receptor 2 (VEGFR2) kinase, we designed and synthesized 5,6-fused heterocyclic compounds bearing a anilide group. A co-crystal structure analysis of imidazo[1,2-b]pyridazine derivative 2 with VEGFR2 revealed that the N1-nitrogen of imidazo[1,2-b]pyridazine core interacts with the backbone NH group of Cys919. To retain this essential interaction, we designed a series of imidazo[1,2-a]pyridine, [1,2,4]triazolo[1,5-a]pyridine, thiazolo[5,4-b]pyridine, and 1,3-benzothiazole derivatives maintaining a ring nitrogen as hydrogen bond acceptor (HBA) at the corresponding position. All compounds thus designed displayed strong inhibitory activity against VEGFR2 kinase, and the [1,2,4]triazolo[1,5-a]pyridine 13d displayed favorable physicochemical properties. Furthermore, 13d inhibited VEGFR2 kinase with slow dissociation kinetics and also inhibited platelet-derived growth factor receptor (PDGFR) kinases. Oral administration of 13d showed potent anti-tumor efficacy in DU145 and A549 xenograft models in nude mice.


Subject(s)
Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Design , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Kinetics , Lung Neoplasms/drug therapy , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Prostatic Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Pyridines/chemistry , Pyridines/pharmacokinetics , Random Allocation , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry , Triazoles/pharmacology , Vascular Endothelial Growth Factor Receptor-2/chemistry , Xenograft Model Antitumor Assays
14.
Bioorg Med Chem Lett ; 22(14): 4769-72, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22677317

ABSTRACT

Spiro-pyrazolidinedione derivatives without quaternary chiral center were discovered by structure-based drug design and characterized as potent acetyl-CoA carboxylase (ACC) inhibitors. The high metabolic stability of the spiro-pyrazolo[1,2-a]pyridazine scaffold and enhancement of the activity by incorporation of a 7-methoxy group on the benzothiophene core successfully led to the identification of compound 4c as an orally bioavailable and highly potent ACC inhibitor. Oral administration of 4c significantly decreased the values of the respiratory quotient in rats, indicating the stimulation of fatty acid oxidation.


Subject(s)
Acetyl-CoA Carboxylase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Pyrazoles/chemistry , Spiro Compounds/chemistry , Animals , Enzyme Inhibitors/pharmacology , Humans , Microsomes/drug effects , Microsomes/enzymology , Models, Molecular , Molecular Structure , Pyrazoles/pharmacology , Rats , Spiro Compounds/pharmacology , Stereoisomerism , Structure-Activity Relationship
15.
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.

16.
Bioorg Med Chem ; 19(21): 6261-73, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21975069

ABSTRACT

Melanin-concentrating hormone receptor 1 (MCHR1) antagonists have been studied as potential agents for the treatment of obesity. Initial structure-activity relationship studies of in-house hit compound 1a and subsequent optimization studies resulted in the identification of tetrahydroisoquinoline derivative 23, 1-(2-acetyl-1,2,3,4-tetrahydroisoquinolin-7-yl)-4-[4-(4-chlorophenyl)piperidin-1-yl]butan-1-one, as a potent hMCHR1 antagonist. A homology model of hMCHR1 suggests that these compounds interact with Asn 294 and Asp 123 in the binding site of hMCHR1 to enhance binding affinity. Oral administration of compound 23 dose-dependently reduced food intake in diet-induced obesity (DIO)-F344 rats.


Subject(s)
Anti-Obesity Agents/chemistry , Anti-Obesity Agents/pharmacology , Benzazepines/chemistry , Benzazepines/pharmacology , Obesity/drug therapy , Receptors, Pituitary Hormone/antagonists & inhibitors , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacokinetics , Benzazepines/chemical synthesis , Benzazepines/pharmacokinetics , CHO Cells , Cricetinae , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Dynamics Simulation , Obesity/metabolism , Protein Binding , Rats , Rats, Inbred F344 , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship
17.
Bioorg Med Chem ; 19(18): 5539-52, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21856163

ABSTRACT

Human melanin-concentrating hormone receptor 1 (hMCHR1) antagonists are promising targets for obesity treatment. We identified the tetrahydronaphthalene derivative 1a with modest binding affinity for hMCHR1 by screening an in-house G protein-coupled receptor (GPCR) ligand library. We synthesized a series of 6-aminomethyl-5,6,7,8-tetrahydronaphthalenes and evaluated their activity as hMCHR1 antagonists. Modification of the biphenylcarbonylamino group revealed that the biphenyl moiety played a crucial role in the interaction of the antagonist with the receptor. The stereoselective effect of the chiral center on binding affinity generated the novel 6-aminomethyl-7,8-dihydronaphthalene scaffold without a chiral center. Optimization of the amino group led to the identification of a potent antagonist 2s (4'-fluoro-N-[6-(1-pyrrolidinylmethyl)-7,8-dihydro-2-naphthalenyl][1,1'-biphenyl]-4-carboxamide), which significantly inhibited the nocturnal food intake in rats after oral administration. Pharmacokinetic analysis confirmed that 2s had good oral bioavailability and brain penetrance. This antagonist appears to be a viable lead compound that can be used to develop a promising therapy for obesity.


Subject(s)
Drug Discovery , Receptors, Somatostatin/antagonists & inhibitors , Tetrahydronaphthalenes/chemical synthesis , Tetrahydronaphthalenes/pharmacology , Animals , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Female , Humans , Ligands , Male , Mice , Mice, Inbred Strains , Mice, Obese , Models, Molecular , Molecular Structure , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Tetrahydronaphthalenes/chemistry
18.
Bioorg Med Chem ; 19(15): 4482-98, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21741847

ABSTRACT

Dipeptidyl peptidase IV (DPP-4) inhibition is a validated therapeutic option for type 2 diabetes, exhibiting multiple antidiabetic effects with little or no risk of hypoglycemia. In our studies involving non-covalent DPP-4 inhibitors, a novel series of quinoline-based inhibitors were designed based on the co-crystal structure of isoquinolone 2 in complex with DPP-4 to target the side chain of Lys554. Synthesis and evaluation of designed compounds revealed 1-[3-(aminomethyl)-4-(4-methylphenyl)-2-(2-methylpropyl)quinolin-6-yl]piperazine-2,5-dione (1) as a potent, selective, and orally active DPP-4 inhibitor (IC50=1.3 nM) with long-lasting ex vivo activity in dogs and excellent antihyperglycemic effects in rats. A docking study of compound 1 revealed a hydrogen-bonding interaction with the side chain of Lys554, suggesting this residue as a potential target site useful for enhancing DPP-4 inhibition.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/therapeutic use , Quinolines/chemistry , Quinolines/therapeutic use , Animals , Caco-2 Cells , Cell Line , Dipeptidyl Peptidase 4/chemistry , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dogs , Female , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Lysine/metabolism , Quinolines/pharmacokinetics , Quinolines/pharmacology , Rats , Rats, Wistar
19.
Bioorg Med Chem ; 19(16): 4953-70, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21764322

ABSTRACT

The design, synthesis, and structure-activity relationships of a new class of potent and orally active non-peptide dipeptidyl peptidase IV (DPP-4) inhibitors, 3-aminomethyl-1,2-dihydro-4-phenyl-1-isoquinolones, are described. We hypothesized that the 4-phenyl group of the isoquinolone occupies the S1 pocket of the enzyme, the 3-aminomethyl group forms an electrostatic interaction with the S2 pocket, and the introduction of a hydrogen bond donor onto the 6- or 7-substituent provides interaction with the hydrophilic region of the enzyme. Based on this hypothesis, intensive research focused on developing new non-peptide DPP-4 inhibitors has been carried out. Among the compounds designed in this study, we identified 2-[(3-aminomethyl-2-(2-methylpropyl)-1-oxo-4-phenyl-1,2-dihydro-6-isoquinolinyl)oxy]acetamide (35a) as a potent, selective, and orally bioavailable DPP-4 inhibitor, which exhibited in vivo efficacy in diabetic model rats. Finally, X-ray crystallography of 35a in a complex with the enzyme validated our hypothesized binding mode and identified Lys554 as a new target-binding site available for DPP-4 inhibitors.


Subject(s)
Dipeptidyl Peptidase 4/drug effects , Dipeptidyl-Peptidase IV Inhibitors/chemical synthesis , Hypoglycemic Agents/chemical synthesis , Isoquinolines/chemical synthesis , Administration, Oral , Animals , Blood Glucose , Caco-2 Cells , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl Peptidase 4/analysis , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/analysis , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/drug effects , Drug Design , Female , Glucose Tolerance Test , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Isoquinolines/administration & dosage , Isoquinolines/pharmacology , Isoquinolines/therapeutic use , Molecular Targeted Therapy , Peptides/metabolism , Quinolones/administration & dosage , Quinolones/chemical synthesis , Quinolones/chemistry , Quinolones/pharmacology , Quinolones/therapeutic use , Rats , Rats, Wistar , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...