Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Pharmacology ; 106(3-4): 189-201, 2021.
Article in English | MEDLINE | ID: mdl-33621976

ABSTRACT

AIM: It has been reported that necrostatin-1 (Nec-1) is a specific necroptosis inhibitor that could attenuate programmed cell death induced by myocardial ischemia/reperfusion (I/R) injury. This study aimed to observe the effect and mechanism of novel Nec-1 analog (Z)-5-(3,5-dimethoxybenzyl)-2-imine-1-methylimidazolin-4-1 (DIMO) on myocardial I/R injury. METHODS: Male SD rats underwent I/R injury with or without different doses of DIMO (1, 2, or 4 mg/kg) treatment. Isolated neonatal rat cardiomyocytes were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) treatment with or without DIMO (0.1, 1, 10, or 100 µM). Myocardial infarction was measured by TTC staining. Cardiomyocyte injury was assessed by lactate dehydrogenase assay (LDH) and flow cytometry. Receptor-interacting protein 1 kinase (RIP1K) and autophagic markers were detected by co-immunoprecipitation and Western blotting analysis. Molecular docking of DIMO into the ATP binding site of RIP1K was performed using GLIDE. RESULTS: DIMO at doses of 1 or 2 mg/kg improved myocardial infarct size. However, the DIMO 4 mg/kg dose was ineffective. DIMO at the dose of 0.1 µM decreased LDH leakage and the ratio of PI-positive cells followed by OGD/R treatment. I/R or OGD/R increased RIP1K expression and in its interaction with RIP3K, as well as impaired myocardial autophagic flux evidenced by an increase in LC3-II/I ratio, upregulated P62 and Beclin-1, and activated cathepsin B and L. In contrast, DIMO treatment reduced myocardial cell death and reversed the above mentioned changes in RIP1K and autophagic flux caused by I/R and OGD/R. DIMO binds to RIP1K and inhibits RIP1K expression in a homology modeling and ligand docking. CONCLUSION: DIMO exerts cardioprotection against I/R- or OGD/R-induced injury, and its mechanisms may be associated with the reduction in RIP1K activation and restoration impaired autophagic flux.


Subject(s)
Autophagy/drug effects , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Imidazoles/chemistry , Indoles/chemistry , Myocardial Reperfusion Injury/prevention & control , Animals , Animals, Newborn , Beclin-1/metabolism , Cathepsin B/metabolism , Cathepsin L/metabolism , Cell Death/drug effects , Hemodynamics/drug effects , Male , Microtubule-Associated Proteins/metabolism , Molecular Docking Simulation , Myocardial Infarction/metabolism , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/drug effects , Necroptosis/drug effects , Primary Cell Culture , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Structural Homology, Protein
2.
Nat Chem Biol ; 13(4): 389-395, 2017 04.
Article in English | MEDLINE | ID: mdl-28135237

ABSTRACT

Polycomb repressive complex 2 (PRC2) is a regulator of epigenetic states required for development and homeostasis. PRC2 trimethylates histone H3 at lysine 27 (H3K27me3), which leads to gene silencing, and is dysregulated in many cancers. The embryonic ectoderm development (EED) protein is an essential subunit of PRC2 that has both a scaffolding function and an H3K27me3-binding function. Here we report the identification of A-395, a potent antagonist of the H3K27me3 binding functions of EED. Structural studies demonstrate that A-395 binds to EED in the H3K27me3-binding pocket, thereby preventing allosteric activation of the catalytic activity of PRC2. Phenotypic effects observed in vitro and in vivo are similar to those of known PRC2 enzymatic inhibitors; however, A-395 retains potent activity against cell lines resistant to the catalytic inhibitors. A-395 represents a first-in-class antagonist of PRC2 protein-protein interactions (PPI) for use as a chemical probe to investigate the roles of EED-containing protein complexes.


Subject(s)
Antineoplastic Agents/pharmacology , Indans/pharmacology , Polycomb Repressive Complex 2/antagonists & inhibitors , Sulfonamides/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Indans/chemistry , Models, Molecular , Molecular Structure , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Protein Binding/drug effects , Structure-Activity Relationship , Sulfonamides/chemistry , Tumor Cells, Cultured
3.
Bioorg Med Chem Lett ; 27(7): 1576-1583, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28254486

ABSTRACT

Herein we disclose SAR studies of a series of dimethylamino pyrrolidines which we recently reported as novel inhibitors of the PRC2 complex through disruption of EED/H3K27me3 binding. Modification of the indole and benzyl moieties of screening hit 1 provided analogs with substantially improved binding and cellular activities. This work culminated in the identification of compound 2, our nanomolar proof-of-concept (PoC) inhibitor which provided on-target tumor growth inhibition in a mouse xenograft model. X-ray crystal structures of several inhibitors bound in the EED active-site are also discussed.


Subject(s)
Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/metabolism , Pyrrolidines/pharmacology , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Ligands , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Polycomb Repressive Complex 2/chemistry , Protein Binding , Pyrrolidines/chemical synthesis , Pyrrolidines/chemistry , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Xenograft Model Antitumor Assays
4.
Bioorg Med Chem ; 25(1): 166-174, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28340987

ABSTRACT

A series of curcumin derivatives as potent dual inhibitors of xanthine oxidase (XOD) and urate transporter 1 (URAT1) was discovered as anti-hyperuricemic agents. These compounds proved efficient effects on anti-hyperuricemic activity and uricosuric activity in vivo. More importantly, some of them exhibited proved efficient effects on inhibiting XOD activity and suppressing uptake of uric acid via URAT1 in vitro. Especially, the treatment of 4d was demonstrated to improve uric acid over-production and under-excretion in oxonate-induced hyperuricemic mice through regulating XOD activity and URAT1 expression. Docking study was performed to elucidate the potent XOD inhibition of 4d. Compound 4d may serve as a tool compound for further design of anti-hyperuricemic drugs targeting both XOD and URAT1.


Subject(s)
Curcumin/analogs & derivatives , Curcumin/therapeutic use , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Hyperuricemia/drug therapy , Organic Anion Transporters/antagonists & inhibitors , Organic Cation Transport Proteins/antagonists & inhibitors , Xanthine Oxidase/antagonists & inhibitors , Animals , Curcumin/pharmacology , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Hyperuricemia/metabolism , Male , Mice , Models, Molecular , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism , Uric Acid/metabolism , Xanthine Oxidase/metabolism
5.
Bioorg Med Chem ; 23(13): 3221-7, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26003342

ABSTRACT

A type of novel rhodanine-based 4-anilinoquinazoline, which designed the combination between quinazoline as the backbone and various substituted biological rhodanine groups as the side chain, have been synthesized, and their antiproliferative activities were also evaluated firstly. These compounds displayed good antiproliferative activity and EGFR-TK inhibitory activity. Among them, compound 8d showed good inhibitory activity (IC50=2.7µM for Hep G2, IC50=3.1µM for A549) and molecular docking of 8d into EGFR TK active site was also performed, this inhibitor well fitting the active site might well explain its excellent inhibitory activity.


Subject(s)
Aniline Compounds/chemical synthesis , Antineoplastic Agents/chemical synthesis , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Quinazolines/chemical synthesis , Rhodanine/chemistry , Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Humans , Molecular Docking Simulation , Protein Binding , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Structure-Activity Relationship
7.
Expert Opin Ther Pat ; 34(8): 611-625, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38889204

ABSTRACT

INTRODUCTION: The P2Y14 receptor (P2Y14R), a member of the G protein-coupled receptor family, is activated by extracellular nucleotides. Due to its involvement in inflammatory, immunological and other associated processes, P2Y14R has emerged as a promising therapeutic target. Despite lacking a determined three-dimensional crystal structure, the homology modeling technique based on closely related P2Y receptors' crystallography has been extensively utilized for developing active compounds targeting P2Y14R. Recent discoveries have unveiled numerous highly effective and subtype-specific P2Y14R inhibitors. This study presents an overview of the latest advancements in P2Y14R inhibitors. AREAS COVERED: This review presents an overview of the advancements in P2Y14R inhibitor research over the past five years, encompassing new patents, journal articles, and highlighting the therapeutic prospects inherent in these compounds. EXPERT OPINION: The recent revelation of the vast potential of P2Y14R inhibitors has led to the development of novel compounds that exhibit promising capabilities for the treatment of sterile inflammation of the kidney, potentially diabetes, and asthma. Despite being a relatively nascent class of compounds, certain members have already exhibited their capacity to surmount specific challenges posed by conventional P2Y14R inhibitors. Targeting P2Y14R through small molecules may present a promising therapeutic strategy for effectively managing diverse inflammatory diseases.


Subject(s)
Drug Development , Inflammation , Patents as Topic , Humans , Animals , Inflammation/drug therapy , Inflammation/physiopathology , Drug Design , Asthma/drug therapy , Asthma/physiopathology , Diabetes Mellitus/drug therapy , Receptors, Purinergic P2
8.
J Med Chem ; 67(8): 6456-6494, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38574366

ABSTRACT

Dysregulation of IL17A drives numerous inflammatory and autoimmune disorders with inhibition of IL17A using antibodies proven as an effective treatment. Oral anti-IL17 therapies are an attractive alternative option, and several preclinical small molecule IL17 inhibitors have previously been described. Herein, we report the discovery of a novel class of small molecule IL17A inhibitors, identified via a DNA-encoded chemical library screen, and their subsequent optimization to provide in vivo efficacious inhibitors. These new protein-protein interaction (PPI) inhibitors bind in a previously undescribed mode in the IL17A protein with two copies binding symmetrically to the central cavities of the IL17A homodimer.


Subject(s)
DNA , Drug Discovery , Interleukin-17 , Small Molecule Libraries , Interleukin-17/metabolism , Interleukin-17/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , DNA/metabolism , DNA/chemistry , Humans , Animals , Structure-Activity Relationship , Protein Binding , Mice
9.
Bioorg Med Chem ; 21(2): 388-94, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23245570

ABSTRACT

A type of novel α,ß-unsaturated cyclohexanone analogous, which designed based on the curcumin core structure, have been discovered as potential EGFR inhibitors. These compounds exhibit potent antiproliferative activity in two human tumor cell lines (Hep G2 and B16-F10). Among them, compounds I(3) and I(12) displayed the most potent EGFR inhibitory activity (IC(50) = 0.43 µM and 1.54 µM, respectively). Molecular docking of I(12) into EGFR TK active site was also performed. This inhibitor nicely fitting the active site might well explain its excellent inhibitory activity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Curcumin/analogs & derivatives , Cyclohexanones/chemistry , Drug Design , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Binding Sites , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Curcumin/chemical synthesis , Curcumin/toxicity , ErbB Receptors/metabolism , Hep G2 Cells , Humans , Molecular Docking Simulation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/toxicity , Structure-Activity Relationship
10.
Bioorg Med Chem ; 21(19): 6084-91, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23962660

ABSTRACT

Two new series of new compounds containing a 6-amino-substituted group or 6-acrylamide-substituted group linked to a 4-anilinoquinazoline nucleus have been discovered as potential EGFR inhibitors. These compounds proved efficient effects on antiproliferative activity and EGFR-TK inhibitory activity. Especially, N(6)-((5-bromothiophen-2-yl)methyl)-N(4)-(3-chlorophenyl)quinazoline-4,6-diamine (5e), showed the most potent inhibitory activity (IC50=3.11µM for Hep G2, IC50=0.82µM for A549). The EGFR molecular docking model suggested that the new compound is nicely bound to the region of EGFR, and cell morphology by Hoechst stain experiment suggested that these compounds efficiently induced apoptosis of A549 cells.


Subject(s)
Aniline Compounds/chemical synthesis , Antineoplastic Agents/chemical synthesis , Drug Discovery , ErbB Receptors/antagonists & inhibitors , Quinazolines/chemical synthesis , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Molecular Structure , Protein Binding/drug effects , Quinazolines/chemistry , Quinazolines/pharmacology , Structure-Activity Relationship
11.
Bioorg Med Chem ; 20(14): 4194-200, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22739087

ABSTRACT

A series of novel rhodanine-based acylsulfonamide derivatives were designed, synthesized, and evaluated as small-molecule inhibitors of anti-apoptotic Bcl-2 protein. These compounds exhibit potent antiproliferative activity in three human tumor cell lines (Hep G2, PC-3 and B16-F10). Among them, the most potent compounds 10 and 11 bind to Bcl-2 with a K(i) of 20 and 25 nM, respectively. Docking studies demonstrated that these two compounds orient similarly at the binding site of Bcl-2, and the calculated binding affinities (Glide XP score) of compound 10 is more negative than that of compound 11. The binding interactions of compounds with high binding affinity to Bcl-2 protein were analyzed.


Subject(s)
Drug Design , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Rhodanine/chemistry , Sulfonamides/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Computer Simulation , Hep G2 Cells , Humans , Protein Binding/drug effects , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/metabolism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology
12.
Bioorg Med Chem ; 20(1): 317-23, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22112541

ABSTRACT

A type of novel 4,6-substituted-(diaphenylamino)quinazolines, which designed based on the 4-(phenylamino)quinazoline moiety, have been discovered as potential EGFR inhibitors. These compounds displayed good antiproliferative activity and EGFR-TK inhibitory activity. Especially, 4-((4-(3-bromophenylamino)quinazolin-6-ylamino)methyl)phenol (5b), showed the most potent inhibitory activity (IC(50)=0.28µM for Hep G2, IC(50)=0.59µM for A16-F10 and IC(50)=0.87µM for EGFR) and effectively induces apoptosis in a dose-dependent manner in the Hep G2 cell line. Molecular docking of 5b into EGFR TK active site was also performed. This inhibitor nicely fitting the active site might well explain its excellent inhibitory activity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Drug Design , ErbB Receptors/antagonists & inhibitors , Phenols/chemical synthesis , Quinazolines/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Binding Sites , Computer Simulation , Crystallography, X-Ray , ErbB Receptors/metabolism , Hep G2 Cells , Humans , Molecular Conformation , Phenols/chemistry , Phenols/pharmacology , Protein Structure, Tertiary , Quinazolines/chemical synthesis , Quinazolines/pharmacology
13.
J Enzyme Inhib Med Chem ; 27(5): 708-14, 2012 Oct.
Article in English | MEDLINE | ID: mdl-21985377

ABSTRACT

Two series of urea and thiourea derivatives (1a-11a, 1b-11b) have been synthesized; all the 22 compounds were reported for the first time. Their anti-proliferative activities against the melanoma cell line B16-F10 were evaluated. Among the compounds tested, compound 6b exhibited the most potent activity in melanoma cells growth inhibition (IC(50) = 0.33 µM). The bioassay tests showed that anti-proliferative activities of these novel compounds were possibly caused by inhibition of ERK1/2 phosphorylation level. Therefore, compound 6b can be a potential anti-melanoma agent and an inhibitor of ERK1/2 phosphorylation deserving further research.


Subject(s)
Melanoma, Experimental/drug therapy , Urea/chemical synthesis , Urea/pharmacology , Animals , Biological Assay , Cell Line, Tumor , Drug Screening Assays, Antitumor , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Inhibitory Concentration 50 , MAP Kinase Signaling System , Magnetic Resonance Spectroscopy , Melanoma, Experimental/pathology , Mice , Phosphorylation , Spectrometry, Mass, Electrospray Ionization , Thiourea/chemical synthesis , Thiourea/pharmacology , Thiourea/therapeutic use , Urea/therapeutic use
14.
Expert Opin Ther Pat ; 32(9): 1027-1042, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35914778

ABSTRACT

INTRODUCTION: Lysine-specific demethylase 1 (LSD1), which belongs to the demethylase of non-histone proteins, is believed to promote cancer cell proliferation and metastasis by modifying histones. LSD1 dysfunction may play a key role in a variety of cancers, such as acute myeloid leukemia and non-small cell lung cancer, indicating that LSD1 is a promising epigenetic target for cancer therapy. Many different types of small molecule LSD1 inhibitors have been developed and shown to inhibit tumor cell proliferation, invasion, and migration, providing a new treatment strategy for solid tumors. AREAS COVERED: This review summarizes the progress of LSD1 inhibitor research in the last four years, including selected new patents and article publications, as well as the therapeutic potential of these compounds. EXPERT OPINION: Natural products offer a promising prospect for developing novel potent LSD1 inhibitors, as structural design and activity of irreversible and reversible inhibitors have been continuously optimized since the discovery of the LSD1 target in 2004. The use of 'microtubule-binding agents' and 'dual-agent combination' has recently become a new anticancer technique, reducing the resistance and adverse reactions of traditional drugs. Several microtubule-binding drugs have been used successfully in clinical practice, providing structural scaffolds and new ideas for the development of safer drugs.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Enzyme Inhibitors/pharmacology , Histone Demethylases , Histones/chemistry , Histones/metabolism , Humans , Patents as Topic
15.
J Org Chem ; 76(17): 7048-55, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21736359

ABSTRACT

Cortisol and the glucocorticoid receptor (GR) signaling pathway has been linked to the development of diabetes and metabolic syndrome. In vivo, 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) catalyzes the conversion of inactive cortisone to its active form, cortisol. Existing clinical data have supported 11ß-HSD1 as a valid therapeutic target for type 2 diabetes. In our research program, (R)-1,1,1-trifluoro-2-(3-((R)-4-(4-fluoro-2-(trifluoromethyl)phenyl)-2-methylpiperazin-1-ylsulfonyl)phenyl)propan-2-ol (HSD-016) was discovered to be a potent, selective, and efficacious 11ß-HSD1 inhibitor and advanced as a clinical candidate. Herein, a reliable and scalable synthesis of HSD-016 is described. Key transformations include an asymmetric synthesis of a chiral tertiary alcohol via Sharpless dihydroxylation, epoxide formation, and subsequent mild reduction. This route ensured multikilogram quantities of HSD-016 necessary for clinical studies.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/chemical synthesis , Piperazines/chemical synthesis , Propanols/chemical synthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1/chemistry , Administration, Oral , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Molecular Structure , Piperazines/chemistry , Piperazines/pharmacology , Propanols/chemistry , Propanols/pharmacology
16.
Bioorg Med Chem ; 19(15): 4454-9, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21742506

ABSTRACT

ß-ketoacyl-acyl carrier protein synthase III (FabH) catalyzes the initial step of fatty acid biosynthesis via a type II fatty acid synthase in most bacteria. The important role of this essential enzyme combined with its unique structural features and ubiquitous occurrence in bacteria has made it an attractive new target for the development of new FabH inhibitors. We first used a structure-based approach to develop 24 new vinylogous carbamates (4a-15a, 4b-15b) that target FabH for the development of new antibiotics in this paper. Potent FabH inhibitory and selective anti- Gram-negative bacteria activities were observed in most of these vinylogous carbamates. Especially, compound 6a and 7a showed the most potent FabH inhibitory activity with IC50 of 2.6 and 3.3 µM, respectively. Docking simulation was performed to position compound 6a into the Escherichia coli FabH active site and the possible binding conformation of compounds has been proposed. The biological data and molecular docking indicated that compounds 6a and 7a were potent inhibitors of E. coli FabH as antibiotics deserving further research.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Carbamates/chemistry , Carbamates/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/enzymology , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Anti-Bacterial Agents/chemical synthesis , Carbamates/chemical synthesis , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/enzymology , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/enzymology , Humans , Models, Molecular , Structure-Activity Relationship
17.
J Enzyme Inhib Med Chem ; 26(1): 37-45, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20583855

ABSTRACT

A series of salicylanilide derivatives (compounds 1-32) were synthesised by reacting substituted salicylic acids and anilines. The chemical structures of these compounds were determined by (1)H-NMR, electrospray ionisation mass spectrometry (ESI-MS) and elemental analysis. The compounds were assayed for their antiproliferative activities against the Hep-G2 cell line by the 3-(4,5-dimethylthylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method. Among the compounds tested, 22 and 28 showed the most favouable antiproliferative activities with 50% inhibitory concentration (IC(50)) values of 1.7 and 1.3 µM, respectively, which were comparable to the positive control of 5-fluorouracil (IC(50)=1.8 µM). A solid-phase ELISA assay was also performed to evaluate the ability of compounds 1-32 to inhibit the autophosphorylation of the epidermal growth factor receptor tyrosine kinase (EGFR TK). Docking simulations of 22 and 28 were carried out to illustrate the binding mode of the molecule into the EGFR active site, and the result suggested that both compounds 22 and 28 could bind the EGFR kinase well.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Salicylanilides/pharmacology , Aniline Compounds/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Fluorouracil/pharmacology , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Protein Binding , Salicylanilides/chemical synthesis , Salicylanilides/metabolism , Salicylates/chemistry , Structure-Activity Relationship , Tetrazolium Salts/analysis , Thiazoles/analysis
18.
Bioorg Med Chem Lett ; 20(22): 6653-6, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20943391

ABSTRACT

Vascular endothelial growth factor receptor-2 (VEGFR-2) plays a crucial role in the process of cancer angiogenesis. A series of quinoline amide derivatives were prepared and found to be good inhibitors of VEGFR-2. The inhibitory activities were investigated against VEGFR-2 kinase and human umbilical vein endothelial cells (HUVEC) in vitro. Compound 6 (5-chloro-2-hydroxy-N-(quinolin-8-yl)benzamide) exhibited the most potent inhibitory activity (IC(50)=3.8 and 5.5 nM for VEGFR-2 kinase and HUVEC, respectively). Docking simulation supported the initial pharmacophoric hypothesis and suggested a common mode of interaction at the ATP-binding site of VEGFR-2, which demonstrates that compound 6 is a potential agent for cancer therapy deserving further researching.


Subject(s)
Amides/chemistry , Quinolines/chemical synthesis , Quinolines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Cells, Cultured , Drug Design , Humans , Inhibitory Concentration 50 , Models, Molecular , Quinolines/chemistry
19.
Bioorg Med Chem Lett ; 20(6): 2025-8, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20153183

ABSTRACT

Beta-ketoacyl-acyl carrier protein synthase III (FabH) catalyzes the initial step of fatty acid biosynthesis via a type II fatty acid synthase in most bacteria. The important role of this essential enzyme combined with its unique structural features and ubiquitous occurrence in bacteria has made it an attractive new target for the development of new FabH inhibitors. The synthesis and biological evaluation halide-deoxybenzoins derivatives are described in this Letter. Potent FabH inhibitory and selective anti-Gram-negative bacteria activities were observed in deoxybenzoin derivatives. Furthermore, compound 19 was able to reduce the ECE-induced IL-8 production in gastric mucosal cells significantly. Based on the biological data and molecular docking, compound 19 is a potential FabH inhibitor and anti-inflammatory agent deserving further research.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Benzoin/analogs & derivatives , Anti-Inflammatory Agents/chemical synthesis , Benzoin/chemical synthesis , Benzoin/chemistry , Benzoin/pharmacology , Drug Design , Enzyme-Linked Immunosorbent Assay , Models, Molecular
20.
Bioorg Med Chem ; 18(13): 4606-14, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20627597

ABSTRACT

Two series of pyrazole derivatives designing for potential EGFR kinase inhibitors have been discovered. Some of them exhibited significant EGFR inhibitory activity. Compound 3-(3,4-dimethylphenyl)-5-(4-methoxyphenyl)-4,5-dihydro-1H-pyrazole-1-carbothioamide (C5) displayed the most potent EGFR inhibitory activity with IC50 of 0.07 µM, which was comparable to the positive control erlotinib. Docking simulation was performed to position compound C5 into the EGFR active site to determine the probable binding model. Antiproliferative assay results indicating that some of the pyrazole derivatives own high antiproliferative activity against MCF-7. Compound C5 showed significant antiproliferative activity against MCF-7 with IC50 of 0.08 µM. Therefore, compound C5 with potent inhibitory activity in tumor growth inhibition would be a potential anticancer agent.


Subject(s)
Antineoplastic Agents/chemistry , Pyrazoles/chemistry , Thioamides/chemistry , Thiourea/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Binding Sites , Cell Line, Tumor , Computer Simulation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Neoplasms/drug therapy , Pyrazoles/chemical synthesis , Pyrazoles/therapeutic use , Thioamides/chemical synthesis , Thioamides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL