Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
2.
Sci Rep ; 13(1): 13524, 2023 08 19.
Article in English | MEDLINE | ID: mdl-37598239

ABSTRACT

The NLRP3 inflammasome is an intracellular, multiprotein complex that promotes the auto-catalytic activation of caspase-1 and the subsequent maturation and secretion of the pro-inflammatory cytokines, IL-1ß and IL-18. Persistent activation of the NLRP3 inflammasome has been implicated in the pathophysiology of a number of inflammatory and autoimmune diseases, including neuroinflammation, cardiovascular disease, non-alcoholic steatohepatitis, lupus nephritis and severe asthma. Here we describe the preclinical profile of JT002, a novel small molecule inhibitor of the NLRP3 inflammasome. JT002 potently reduced NLRP3-dependent proinflammatory cytokine production across a number of cellular assays and prevented pyroptosis, an inflammatory form of cell death triggered by active caspase-1. JT002 demonstrated in vivo target engagement at therapeutically relevant concentrations when orally dosed in mice and prevented body weight loss and improved inflammatory and fibrotic endpoints in a model of Muckle-Wells syndrome (MWS). In two distinct models of neutrophilic airway inflammation, JT002 treatment significantly reduced airway hyperresponsiveness and airway neutrophilia. These results provide a rationale for the therapeutic targeting of the NLRP3 inflammasome in severe asthma and point to the use of JT002 in a variety of inflammatory disorders.


Subject(s)
Cardiovascular Diseases , Lupus Nephritis , Animals , Mice , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Caspase 1
3.
J Pharmacol Exp Ther ; 386(2): 242-258, 2023 08.
Article in English | MEDLINE | ID: mdl-37308266

ABSTRACT

The NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome is a multiprotein complex and component of the innate immune system that is activated by exogenous and endogenous danger signals to promote activation of caspase-1 and the maturation and release of the proinflammatory cytokines interleukin (IL)-1ß and IL-18. Inappropriate activation of NLRP3 has been implicated in the pathophysiology of multiple inflammatory and autoimmune diseases, including cardiovascular disease, neurodegenerative diseases, and nonalcoholic steatohepatitis (NASH), thus increasing the clinical interest of this target. We describe in this study the preclinical pharmacologic, pharmacokinetic, and pharmacodynamic properties of a novel and highly specific NLRP3 inhibitor, JT001 (6,7-dihydro-5H-pyrazolo[5,1-b][1,3]oxazine-3-sulfonylurea). In cell-based assays, JT001 potently and selectively inhibited NLRP3 inflammasome assembly, resulting in the inhibition of cytokine release and the prevention of pyroptosis, a form of inflammatory cell death triggered by active caspase-1. Oral administration of JT001 to mice inhibited IL-1ß production in peritoneal lavage fluid at plasma concentrations that correlated with mouse in vitro whole blood potency. Orally administered JT001 was effective in reducing hepatic inflammation in three different murine models, including the Nlrp3A350V /+CreT model of Muckle-Wells syndrome (MWS), a diet-induced obesity NASH model, and a choline-deficient diet-induced NASH model. Significant reductions in hepatic fibrosis and cell damage were also observed in the MWS and choline-deficient models. Our findings demonstrate that blockade of NLRP3 attenuates hepatic inflammation and fibrosis and support the use of JT001 to investigate the role of NLRP3 in other inflammatory disease models. SIGNIFICANCE STATEMENT: Persistent inflammasome activation is the consequence of inherited mutations of NLRP3 and results in the development of cryopyrin-associated periodic syndromes associated with severe systemic inflammation. NLRP3 is also upregulated in nonalcoholic steatohepatitis, a metabolic chronic liver disease currently missing a cure. Selective and potent inhibitors of NLRP3 hold great promise and have the potential to overcome an urgent unmet need.


Subject(s)
Non-alcoholic Fatty Liver Disease , Mice , Animals , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Proteins , Pyrin Domain , Liver Cirrhosis/drug therapy , Liver Cirrhosis/prevention & control , Caspase 1/metabolism , Inflammation , Choline/adverse effects , Interleukin-1beta/metabolism
4.
Anticancer Drugs ; 34(10): 1122-1131, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37067993

ABSTRACT

Histone lysine demethylase 4 (KDM4) is an epigenetic regulator that facilitates the transition between transcriptionally silent and active chromatin states by catalyzing the removal of methyl groups on histones H3K9, H3K36, and H1.4K26. KDM4 overamplification or dysregulation has been reported in various cancers and has been shown to drive key processes linked to tumorigenesis, such as replicative immortality, evasion of apoptosis, metastasis, DNA repair deficiency, and genomic instability. KDM4 also plays a role in epigenetic regulation of cancer stem cell renewal and has been linked to more aggressive disease and poorer clinical outcomes. The KDM4 family is composed of four main isoforms (KDM4A-D) that demonstrate functional redundancy and cross-activity; thus, selective inhibition of one isoform appears to be ineffective and pan-inhibition targeting multiple KDM4 isoforms is required. Here, we describe TACH101, a novel, small-molecule pan-inhibitor of KDM4 that selectively targets KDM4A-D with no effect on other KDM families. TACH101 demonstrated potent antiproliferative activity in cancer cell lines and organoid models derived from various histologies, including colorectal, esophageal, gastric, breast, pancreatic, and hematological malignancies. In vivo , potent inhibition of KDM4 led to efficient tumor growth inhibition and regression in several xenograft models. A reduction in the population of tumor-initiating cells was observed following TACH101 treatment. Overall, these observations demonstrate the broad applicability of TACH101 as a potential anticancer agent and support its advancement into clinical trials.


Subject(s)
Histone Demethylases , Neoplasms , Humans , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histone Demethylases/therapeutic use , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Epigenesis, Genetic , Neoplasms/drug therapy , Neoplasms/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/therapeutic use
5.
J Med Chem ; 65(21): 14721-14739, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36279149

ABSTRACT

Inappropriate activation of the NLRP3 inflammasome has been implicated in multiple inflammatory and autoimmune diseases. Herein, we aimed to develop novel NLRP3 inhibitors that could minimize the risk of drug-induced liver injury. Lipophilic ligand efficiency was used as a guiding metric to identify a series of 6,7-dihydro-5H-pyrazolo[5,1-b][1,3]oxazinesulfonylureas. A leading compound from this series was advanced into safety studies in cynomolgus monkeys, and renal toxicity, due to compound precipitation, was observed. To overcome this obstacle, we focused on improving the solubility of our compounds, specifically by introducing basic amine substituents into the scaffold. This led to the identification of GDC-2394, a potent and selective NLRP3 inhibitor, with an in vitro and in vivo safety profile suitable for advancement into human clinical trials.


Subject(s)
NLR Family, Pyrin Domain-Containing 3 Protein , Oxazines , Animals , Humans , Oxazines/pharmacology , Oxazines/therapeutic use , Inflammasomes , Sulfonamides/pharmacology , Macaca fascicularis
6.
J Med Chem ; 63(23): 14522-14529, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33034194

ABSTRACT

Histone demethylase LSDl (KDMlA) belongs to the flavin adenine dinucleotide (FAD) dependent family of monoamine oxidases and is vital in regulation of mammalian biology. Dysregulation and overexpression of LSD1 are hallmarks of a number of human diseases, particularly cancers that are characterized as morphologically poorly differentiated. As such, inhibitors of LSD1 have potential to be beneficial as a cancer therapy. The most clinically advanced inhibitors of LSDl are covalent inhibitors derived from tranylcypromine (TCP). Herein, we report the discovery of a novel series of reversible and selective LSDl inhibitors. Exploration of structure-activity relationships (SARs) and optimization of ADME properties resulted in the identification of clinical candidate CC-90011. CC-90011 exhibits potent on-target induction of cellular differentiation in acute myeloid leukemia (AML) and small cell lung cancer (SCLC) cell lines, and antitumor efficacy in patient-derived xenograft (PDX) SCLC models. CC-90011 is currently in phase 2 trials in patients with first line, extensive stage SCLC (ClinicalTrials.gov identifier: NCT03850067).


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Organic Chemicals/pharmacology , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Humans , Organic Chemicals/chemistry , Structure-Activity Relationship
7.
Bioorg Med Chem Lett ; 29(1): 103-106, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30409536

ABSTRACT

The histone demethylase LSD1 is a key enzyme in the epigenetic regulation of gene transcription. Here we present our efforts to discover small molecule reversible inhibitors of LSD1 as an attractive approach to treat hematologic malignancies and certain solid tumors. Using structure-based drug design, we designed and synthesized a novel series of heteroaromatic imidazole inhibitors that demonstrate potent inhibition of the demethylase activity and low nanomolar cell-based activity. This novel LSD1 inhibitor series was further optimized by attenuating the hERG inhibition and improving oral bioavailability.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Imidazoles/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Histone Demethylases/metabolism , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 28(9): 1490-1494, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29627262

ABSTRACT

Histone lysine demethylases (KDMs) play a key role in epigenetic regulation and KDM5A and KDM5B have been identified as potential anti-cancer drug targets. Using structural information from known KDM4 and KDM5 inhibitors, a potent series of pyrazolylpyridines was designed. Structure-activity relationship (SAR) exploration resulted in the identification of compound 33, an orally available, potent inhibitor of KDM5A/5B with promising selectivity. Potent cellular inhibition as measured by levels of tri-methylated H3K4 was demonstrated with compound 33 in the breast cancer cell line ZR-75-1.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Retinoblastoma-Binding Protein 2/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Female , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , MCF-7 Cells , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Models, Molecular , Molecular Structure , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Retinoblastoma-Binding Protein 2/metabolism , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 28(10): 1811-1816, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29657099

ABSTRACT

The bromodomain and extra-terminal (BET) family of epigenetic proteins has attracted considerable attention in drug discovery given its involvement in regulating gene transcription. Screening a focused small molecule library based on the bromodomain pharmacophore resulted in the identification of 2-methylisoquinoline-1-one as a novel BET bromodomain-binding motif. Structure guided SAR exploration resulted in >10,000-fold potency improvement for the BRD4-BD1 bromodomain. Lead compounds exhibited excellent potencies in both biochemical and cellular assays in MYC-dependent cell lines. Compound 36 demonstrated good physicochemical properties and promising exposure levels in exploratory PK studies.


Subject(s)
Drug Design , Isoquinolines/chemistry , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Binding Sites , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Humans , Inhibitory Concentration 50 , Isoquinolines/chemical synthesis , Isoquinolines/pharmacology , Molecular Dynamics Simulation , Nuclear Proteins/metabolism , Protein Structure, Tertiary , Structure-Activity Relationship , Transcription Factors/metabolism
10.
Cancer Res ; 77(21): 5900-5912, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28883001

ABSTRACT

Traditional treatments for breast cancer fail to address therapy-resistant cancer stem-like cells that have been characterized by changes in epigenetic regulators such as the lysine demethylase KDM4. Here, we describe an orally available, selective and potent KDM4 inhibitor (QC6352) with unique preclinical characteristics. To assess the antitumor properties of QC6352, we established a method to isolate and propagate breast cancer stem-like cells (BCSC) from individual triple-negative tumors resected from patients after neoadjuvant chemotherapy. Limiting-dilution orthotopic xenografts of these BCSCs regenerated original patient tumor histology and gene expression. QC6352 blocked BCSC proliferation, sphere formation, and xenograft tumor formation. QC6352 also abrogated expression of EGFR, which drives the growth of therapy-resistant triple-negative breast cancer cells. Our findings validate a unique BCSC culture system for drug screening and offer preclinical proof of concept for KDM4 inhibition as a new strategy to treat triple-negative breast cancer. Cancer Res; 77(21); 5900-12. ©2017 AACR.


Subject(s)
Cell Proliferation/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Neoplastic Stem Cells/metabolism , Triple Negative Breast Neoplasms/genetics , Animals , Cell Proliferation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice, Inbred NOD , Mice, SCID , Molecular Structure , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , RNA Interference , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
ACS Med Chem Lett ; 8(8): 869-874, 2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28835804

ABSTRACT

Histone lysine demethylases (KDMs) play a vital role in the regulation of chromatin-related processes. Herein, we describe our discovery of a series of potent KDM4 inhibitors that are both cell permeable and antiproliferative in cancer models. The modulation of histone H3K9me3 and H3K36me3 upon compound treatment was verified by homogeneous time-resolved fluorescence assay and by mass spectroscopy detection. Optimization of the series using structure-based drug design led to compound 6 (QC6352), a potent KDM4 family inhibitor that is efficacious in breast and colon cancer PDX models.

12.
J Med Chem ; 54(2): 510-24, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21186796

ABSTRACT

The discovery of two classes of heterocyclic dipeptidyl peptidase IV (DPP-4) inhibitors, pyrimidinones and pyrimidinediones, is described. After a single oral dose, these potent, selective, and noncovalent inhibitors provide sustained reduction of plasma DPP-4 activity and lowering of blood glucose in animal models of diabetes. Compounds 13a, 27b, and 27j were selected for development.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/chemical synthesis , Pyrimidinones/chemical synthesis , Animals , Binding Sites , Biological Availability , Crystallography, X-Ray , Cytochrome P-450 Enzyme Inhibitors , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dogs , Macaca fascicularis , Models, Molecular , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship
13.
J Med Chem ; 51(15): 4632-40, 2008 Aug 14.
Article in English | MEDLINE | ID: mdl-18620382

ABSTRACT

Inhibition of the vascular endothelial growth factor (VEGF) signaling pathway has emerged as one of the most promising new approaches for cancer therapy. We describe herein the key steps starting from an initial screening hit leading to the discovery of pazopanib, N(4)-(2,3-dimethyl-2H-indazol-6-yl)-N(4)-methyl-N(2)-(4-methyl-3-sulfonamidophenyl)-2,4-pyrimidinediamine, a potent pan-VEGF receptor (VEGFR) inhibitor under clinical development for renal-cell cancer and other solid tumors.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Sulfonamides/chemistry , Sulfonamides/pharmacology , Animals , Cells, Cultured , Crystallography, X-Ray , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Humans , Indazoles , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Mice , Models, Molecular , Molecular Structure , Neoplasms/blood supply , Neoplasms/drug therapy , Neoplasms/enzymology , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Receptors, Vascular Endothelial Growth Factor/chemistry , Receptors, Vascular Endothelial Growth Factor/metabolism , Sulfonamides/therapeutic use , Xenograft Model Antitumor Assays
14.
Mol Cancer Ther ; 6(7): 2012-21, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17620431

ABSTRACT

With the development of targeted therapeutics, especially for small-molecule inhibitors, it is important to understand whether the observed in vivo efficacy correlates with the modulation of desired/intended target in vivo. We have developed a small-molecule inhibitor of all three vascular endothelial growth factor (VEGF) receptors (VEGFR), platelet-derived growth factor receptor, and c-Kit tyrosine kinases, pazopanib (GW786034), which selectively inhibits VEGF-induced endothelial cell proliferation. It has good oral exposure and inhibits angiogenesis and tumor growth in mice. Because bolus administration of the compound results in large differences in C(max) and C(trough), we investigated the effect of continuous infusion of a VEGFR inhibitor on tumor growth and angiogenesis. GW771806, which has similar enzyme and cellular profiles to GW786034, was used for these studies due to higher solubility requirements for infusion studies. Comparing the pharmacokinetics by two different routes of administration (bolus p.o. dosing and continuous infusion), we showed that the antitumor and antiangiogenic activity of VEGFR inhibitors is dependent on steady-state concentration of the compound above a threshold. The steady-state concentration required for these effects is consistent with the concentration required for the inhibition of VEGF-induced VEGFR2 phosphorylation in mouse lungs. Furthermore, the steady-state concentration of pazopanib determined from preclinical activity showed a strong correlation with the pharmacodynamic effects and antitumor activity in the phase I clinical trial.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Indazoles/pharmacology , Indazoles/pharmacokinetics , Neovascularization, Pathologic/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , Sulfonamides/pharmacology , Sulfonamides/pharmacokinetics , Sulfones/pharmacology , Sulfones/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Cell Line, Tumor , Cell-Free System , Cornea/pathology , Dose-Response Relationship, Drug , Female , Fibroblast Growth Factor 2/pharmacology , Humans , Indazoles/administration & dosage , Indazoles/blood , Inhibitory Concentration 50 , Mice , Mice, Nude , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/blood , Pyrimidines/administration & dosage , Pyrimidines/blood , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Sulfonamides/administration & dosage , Sulfonamides/blood , Sulfones/administration & dosage , Sulfones/blood , Vascular Endothelial Growth Factor A/pharmacology
15.
Anesthesiology ; 107(1): 60-6, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17585216

ABSTRACT

BACKGROUND: A new benzodiazepine derivative, CNS 7056, has been developed to permit a superior sedative profile to current agents, i.e., more predictable fast onset, short duration of sedative action, and rapid recovery profile. This goal has been achieved by rendering the compound susceptible to metabolism via esterases. The authors now report on the profile of CNS 7056 in vitro and in vivo. METHODS: The affinity of CNS 7056 and its carboxylic acid metabolite, CNS 7054, for benzodiazepine receptors and their selectivity profiles were evaluated using radioligand binding. The activity of CNS 7056 and midazolam at subtypes (alpha1beta2gamma2, alpha2beta2gamma2, alpha3beta2gamma2, alpha5beta2gamma2) of the gamma-aminobutyric acid type A (GABAA) receptor was evaluated using the whole cell patch clamp technique. The activity of CNS 7056 at brain benzodiazepine receptors in vivo was measured in rats using extracellular electrophysiology in the substantia nigra pars reticulata. The sedative profile was measured in rodents using the loss of righting reflex test. RESULTS: CNS 7056 bound to brain benzodiazepine sites with high affinity. The carboxylic acid metabolite, CNS 7054, showed around 300 times lower affinity. CNS 7056 and CNS 7054 (10 mum) showed no affinity for a range of other receptors. CNS 7056 enhanced GABA currents in cells stably transfected with subtypes of the GABAA receptor. CNS 7056, like midazolam and other classic benzodiazepines, did not show clear selectivity between subtypes of the GABAA receptor. CNS 7056 (intravenous) caused a dose-dependent inhibition of substantia nigra pars reticulata neuronal firing and recovery to baseline firing rates was reached rapidly. CNS 7056 (intravenous) induced loss of the righting reflex in rodents. The duration of loss of righting reflex was short (< 10 min) and was inhibited by pretreatment with flumazenil. CONCLUSIONS: CNS 7065 is a high-affinity and selective ligand for the benzodiazepine site on the GABAA receptor. CNS 7056 does not show selectivity between GABAA receptor subtypes. CNS 7056 is a potent sedative in rodents with a short duration of action. Inhibition of substantia nigra pars reticulata firing and the inhibition of the effects of CNS 7056 by flumazenil show that it acts at the brain benzodiazepine receptor.


Subject(s)
Benzodiazepines/pharmacology , Hypnotics and Sedatives/pharmacology , Animals , Benzodiazepines/pharmacokinetics , Binding, Competitive/drug effects , Cell Line , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Electrophysiology , Extracellular Space/drug effects , Extracellular Space/metabolism , Flunitrazepam/pharmacokinetics , Humans , Hypnotics and Sedatives/pharmacokinetics , In Vitro Techniques , Ion Channels/drug effects , Ion Channels/metabolism , Membranes/drug effects , Membranes/metabolism , Mice , Midazolam/pharmacology , Postural Balance/drug effects , Radioligand Assay , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, GABA-A/drug effects , Substantia Nigra/drug effects , Swine , Swine, Miniature , Transfection , gamma-Aminobutyric Acid/physiology
16.
J Med Chem ; 50(10): 2297-300, 2007 May 17.
Article in English | MEDLINE | ID: mdl-17441705

ABSTRACT

Alogliptin is a potent, selective inhibitor of the serine protease dipeptidyl peptidase IV (DPP-4). Herein, we describe the structure-based design and optimization of alogliptin and related quinazolinone-based DPP-4 inhibitors. Following an oral dose, these noncovalent inhibitors provide sustained reduction of plasma DPP-4 activity and a lowering of blood glucose in animal models of diabetes. Alogliptin is currently undergoing phase III trials in patients with type 2 diabetes.


Subject(s)
Dipeptidyl Peptidase 4/chemistry , Dipeptidyl-Peptidase IV Inhibitors , Hypoglycemic Agents/chemical synthesis , Piperidines/chemical synthesis , Pyrimidinones/chemical synthesis , Quinazolinones/chemical synthesis , Uracil/analogs & derivatives , Animals , Binding Sites , Blood Glucose/analysis , Cytochrome P-450 Enzyme Inhibitors , Diabetes Mellitus, Experimental/drug therapy , Dogs , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/drug effects , Female , Glucose Tolerance Test , Haplorhini , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Molecular , Piperidines/pharmacokinetics , Piperidines/pharmacology , Pyrimidinones/pharmacokinetics , Pyrimidinones/pharmacology , Quinazolinones/pharmacokinetics , Quinazolinones/pharmacology , Rats , Rats, Wistar , Structure-Activity Relationship , Uracil/chemical synthesis , Uracil/pharmacokinetics , Uracil/pharmacology
18.
J Med Chem ; 48(5): 1610-9, 2005 Mar 10.
Article in English | MEDLINE | ID: mdl-15743202

ABSTRACT

A series of derivatives of 2-anilino-5-phenyloxazole (5) has been identified as inhibitors of VEGFR2 kinase. Herein we describe the structure-activity relationship (SAR) of this novel template. Optimization of both aryl rings led to very potent inhibitors at both the enzymatic and cellular levels. Oxazole 39 had excellent solubility and good oral PK when dosed as the bis-mesylate salt and demonstrated moderate in vivo efficacy against HT29 human colon tumor xenografts. X-ray crystallography confirmed the proposed binding mode, and comparison of oxazoles 39 and 46 revealed interesting differences in orientation of 2-pyridyl and 3-pyridyl rings, respectively, attached at the meta position of the 5-phenyl ring.


Subject(s)
Angiogenesis Inhibitors/chemical synthesis , Aniline Compounds/chemical synthesis , Oxazoles/chemical synthesis , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Adenosine Triphosphate/chemistry , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Aniline Compounds/pharmacokinetics , Aniline Compounds/pharmacology , Animals , Binding Sites , Cell Proliferation/drug effects , Cells, Cultured , Crystallography, X-Ray , Dogs , Humans , Ligands , Male , Mice , Models, Molecular , Oxazoles/pharmacokinetics , Oxazoles/pharmacology , Rats , Rats, Sprague-Dawley , Solubility , Structure-Activity Relationship , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/chemistry , Xenograft Model Antitumor Assays
19.
J Org Chem ; 68(10): 4093-5, 2003 May 16.
Article in English | MEDLINE | ID: mdl-12737599

ABSTRACT

An efficient and regioselective synthesis of 2-methyl-2H-indazoles and 2-ethyl-2H-indazoles using trimethyloxonium tetrafluoroborate or triethyloxonium hexafluorophosphate is reported.

20.
Bioorg Med Chem Lett ; 12(21): 3215-8, 2002 Nov 04.
Article in English | MEDLINE | ID: mdl-12372537

ABSTRACT

The synthesis and evaluation of novel ultrashort-acting benzodiazepine (USA BZD) agonists is described. A BZD scaffold was modified by incorporation of amino acids and derivatives. The propionate side chain of glutamic acid tethers an enzymatically labile functionality where the metabolite carboxylic acid displays markedly reduced BZD receptor affinity. The USA BZDs were characterized by full agonism profiles. Copyright2000 Elsevier Science Ltd.


Subject(s)
Benzodiazepines/chemical synthesis , Benzodiazepines/pharmacology , GABA-A Receptor Agonists , Animals , Benzodiazepines/pharmacokinetics , Chromatography, High Pressure Liquid , Humans , Postural Balance/drug effects , Rats , Receptors, GABA-A/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...