Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
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.
J Med Chem ; 60(10): 4403-4423, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28471663

ABSTRACT

LOXL2 catalyzes the oxidative deamination of ε-amines of lysine and hydroxylysine residues within collagen and elastin, generating reactive aldehydes (allysine). Condensation with other allysines or lysines drives the formation of inter- and intramolecular cross-linkages, a process critical for the remodeling of the ECM. Dysregulation of this process can lead to fibrosis, and LOXL2 is known to be upregulated in fibrotic tissue. Small-molecules that directly inhibit LOXL2 catalytic activity represent a useful option for the treatment of fibrosis. Herein, we describe optimization of an initial hit 2, resulting in identification of racemic-trans-(3-((4-(aminomethyl)-6-(trifluoromethyl)pyridin-2-yl)oxy)phenyl)(3-fluoro-4-hydroxypyrrolidin-1-yl)methanone 28, a potent irreversible inhibitor of LOXL2 that is highly selective over LOX and other amine oxidases. Oral administration of 28 significantly reduced fibrosis in a 14-day mouse lung bleomycin model. The (R,R)-enantiomer 43 (PAT-1251) was selected as the clinical compound which has progressed into healthy volunteer Phase 1 trials, making it the "first-in-class" small-molecule LOXL2 inhibitor to enter clinical development.


Subject(s)
Amino Acid Oxidoreductases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Administration, Oral , Amino Acid Oxidoreductases/metabolism , Animals , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Fibrosis , Halogenation , Humans , Lung/drug effects , Lung/enzymology , Lung/pathology , Lung Diseases/drug therapy , Lung Diseases/enzymology , Lung Diseases/pathology , Male , Methylation , Mice, Inbred C57BL , Models, Molecular , Pyridines/administration & dosage , Pyridines/therapeutic use , Structure-Activity Relationship
5.
J Pharmacol Exp Ther ; 360(1): 1-13, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27754931

ABSTRACT

Autotaxin (ATX) is a secreted glycoprotein that converts lysophosphatidylcholine (LPC) to the bioactive phospholipid lysophosphatidic acid (LPA) and is the major enzyme generating circulating LPA. Inhibition of LPA signaling has profound antifibrotic effects in multiple organ systems, including lung, kidney, skin, and peritoneum. However, other LPA-generating pathways exist, and the role of ATX in localized tissue LPA production and fibrosis remains unclear and controversial. In this study, we describe the preclinical pharmacologic, pharmacokinetic, and pharmacodynamic properties of a novel small-molecule ATX inhibitor, PAT-505 [3-((6-chloro-2-cyclopropyl-1-(1-ethyl-1H-pyrazol-4-yl)-7-fluoro-1H-indol-3-yl) thio)-2-fluorobenzoic acid sodium salt]. PAT-505 is a potent, selective, noncompetitive inhibitor that displays significant inhibition of ATX activity in plasma and liver tissue after oral administration. When dosed therapeutically in a Stelic Mouse Animal Model of nonalcoholic steatohepatitis (NASH), PAT-505 treatment resulted in a small but significant improvement in fibrosis with only minor improvements in hepatocellular ballooning and hepatic inflammation. In a choline-deficient, high-fat diet model of NASH, therapeutic treatment with PAT-505 robustly reduced liver fibrosis with no significant effect on steatosis, hepatocellular ballooning, or inflammation. These data demonstrate that inhibiting autotaxin is antifibrotic and may represent a novel therapeutic approach for the treatment of multiple fibrotic liver diseases, including NASH.


Subject(s)
Enzyme Inhibitors/pharmacology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/enzymology , Phosphoric Diester Hydrolases/metabolism , Piperazines/pharmacology , Animals , Disease Models, Animal , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Mice , Piperazines/pharmacokinetics , Piperazines/therapeutic use
6.
Mol Pharmacol ; 88(6): 982-92, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26371182

ABSTRACT

Autotaxin (ATX) is a secreted enzyme that hydrolyzes lysophosphatidylcholine to lysophosphatidic acid (LPA). LPA is a bioactive phospholipid that regulates diverse biological processes, including cell proliferation, migration, and survival/apoptosis, through the activation of a family of G protein-coupled receptors. The ATX-LPA pathway has been implicated in many pathologic conditions, including cancer, fibrosis, inflammation, cholestatic pruritus, and pain. Therefore, ATX inhibitors represent an attractive strategy for the development of therapeutics to treat a variety of diseases. Mouse and rat ATX have been crystallized previously with LPA or small-molecule inhibitors bound. Here, we present the crystal structures of human ATX in complex with four previously unpublished, structurally distinct ATX inhibitors. We demonstrate that the mechanism of inhibition of each compound reflects its unique interactions with human ATX. Our studies may provide a basis for the rational design of novel ATX inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/metabolism , Animals , Cell Line, Tumor , Crystallization , HEK293 Cells , Humans , Mice , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship
7.
Br J Clin Pharmacol ; 75(3): 779-90, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22803688

ABSTRACT

AIM: To assess the pharmacokinetics, pharmacodynamics, safety and tolerability of the 5-lipoxygenase-activating protein inhibitor, GSK2190915, after oral dosing in two independent phase I studies, one in Western European and one in Japanese subjects, utilizing different formulations. METHOD: Western European subjects received single (50-1000 mg) or multiple (10-450 mg) oral doses of GSK2190915 or placebo in a dose-escalating manner. Japanese subjects received three of four GSK2190915 doses (10-200 mg) plus placebo once in a four period crossover design. Blood samples were collected for GSK2190915 concentrations and blood and urine were collected to measure leukotriene B4 and leukotriene E4, respectively, as pharmacodynamic markers of drug activity. RESULTS: There was no clear difference in adverse events between placebo and active drug-treated subjects in either study. Maximum plasma concentrations of GSK2190915 and area under the curve increased in a dose-related manner and mean half-life values ranged from 16-34 h. Dose-dependent inhibition of blood leukotriene B4 production was observed and near complete inhibition of urinary leukotriene E4 excretion was shown at all doses except the lowest dose. The EC50 values for inhibition of LTB4 were 85 nM and 89 nM in the Western European and Japanese studies, respectively. CONCLUSION: GSK2190915 is well-tolerated with pharmacokinetics and pharmacodynamics in Western European and Japanese subjects that support once daily dosing for 24 h inhibition of leukotrienes. Doses of ≥50 mg show near complete inhibition of urinary leukotriene E4 at 24 h post-dose, whereas doses of ≥150 mg are required for 24 h inhibition of blood LTB4.


Subject(s)
5-Lipoxygenase-Activating Protein Inhibitors , Indoles , Leukotriene E4/blood , Pentanoic Acids , 5-Lipoxygenase-Activating Protein Inhibitors/adverse effects , 5-Lipoxygenase-Activating Protein Inhibitors/pharmacokinetics , 5-Lipoxygenase-Activating Protein Inhibitors/pharmacology , Administration, Oral , Adolescent , Adult , Aged , Area Under Curve , Asian People , Biomarkers/blood , Biomarkers/urine , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Indoles/adverse effects , Indoles/pharmacokinetics , Indoles/pharmacology , Leukotriene B4/blood , Leukotriene B4/urine , Leukotriene E4/urine , Male , Middle Aged , Pentanoic Acids/adverse effects , Pentanoic Acids/pharmacokinetics , Pentanoic Acids/pharmacology , White People , Young Adult
8.
J Pharmacol Exp Ther ; 338(1): 290-301, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21487069

ABSTRACT

The prostaglandin D(2) (PGD(2)) receptor type 2 (DP2) is a G protein-coupled receptor that has been shown to be involved in a variety of allergic diseases, including allergic rhinitis, asthma, and atopic dermatitis. In this study, we describe the preclinical pharmacological and pharmacokinetic properties of the small-molecule DP2 antagonist [2'-(3-benzyl-1-ethyl-ureidomethyl)-6-methoxy-4'-trifluoromethyl-biphenyl-3-yl]-acetic acid (AM211). We determine that AM211 has high affinity for human, mouse, rat, and guinea pig DP2 and it shows selectivity over other prostanoid receptors and enzymes. Antagonist activity of AM211 at the DP2 receptor was confirmed by inhibition of PGD(2)-stimulated guanosine 5'-O-[γ-thio]triphosphate binding to membranes expressing human DP2. A basophil activation assay and a whole-blood assay of eosinophil shape change were used to demonstrate the ability of AM211 to potently antagonize PGD(2)-stimulated functional responses in relevant human cells and in the context of a physiologically relevant environment. AM211 exhibits good oral bioavailability in rats and dogs and dose-dependently inhibits 13,14-dihydro-15-keto-PGD(2)-induced leukocytosis in a guinea pig pharmacodynamic assay. AM211 demonstrates efficacy in two animal models of allergic inflammation, including an ovalbumin-induced lung inflammation model in guinea pigs and an ovalbumin-induced mouse model of allergic rhinitis. AM211 represents a potent and selective antagonist of DP2 that may be used clinically to evaluate the role of DP2 in T helper 2-driven allergic inflammatory diseases.


Subject(s)
Disease Models, Animal , Methylurea Compounds/therapeutic use , Phenylacetates/therapeutic use , Prostaglandin Antagonists/therapeutic use , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Rhinitis, Allergic, Perennial/drug therapy , Adult , Animals , Dogs , Female , Guinea Pigs , HEK293 Cells , Humans , Hypersensitivity/drug therapy , Hypersensitivity/immunology , Hypersensitivity/metabolism , Male , Methylurea Compounds/chemistry , Methylurea Compounds/pharmacology , Mice , Mice, Inbred BALB C , Phenylacetates/chemistry , Phenylacetates/pharmacology , Pneumonia/drug therapy , Pneumonia/immunology , Pneumonia/metabolism , Prostaglandin Antagonists/chemistry , Prostaglandin Antagonists/pharmacology , Protein Binding/physiology , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/immunology , Receptors, Prostaglandin/metabolism , Rhinitis, Allergic, Perennial/immunology , Rhinitis, Allergic, Perennial/metabolism
9.
Eur J Pharmacol ; 640(1-3): 211-8, 2010 Aug 25.
Article in English | MEDLINE | ID: mdl-20519143

ABSTRACT

We evaluated the in vivo pharmacological properties of AM803 3-[3-tert-butylsulfanyl-1-[4-(6-ethoxy-pyridin-3-yl)-benzyl]-5-(5-methyl-pyridin-2-ylmethoxy)-1H-indol-2-yl]-2,2-dimethyl-propionic acid, a selective five-lipoxygenase-activating protein (FLAP) inhibitor, using rat and mouse models of acute inflammation. Oral administration of AM803 (1 mg/kg) resulted in sustained inhibition of ex vivo ionophore-challenged whole blood LTB4 biosynthesis with >90% inhibition for up to 12 h and an EC50 of approximately 7 nM. When rat lungs were challenged in vivo with calcium-ionophore, AM803 inhibited LTB4 and cysteinyl leukotriene (CysLT) production with ED50s of 0.12 mg/kg and 0.37 mg/kg, respectively. The inhibition measured 16 h following a single oral dose of 3 mg/kg was 86% and 41% for LTB4 and CysLTs, respectively. In an acute inflammation setting, AM803 dose-dependently reduced LTB4, CysLTs, plasma protein extravasation and neutrophil influx induced by peritoneal zymosan injection. Finally, AM803 increased survival time in mice exposed to a lethal intravenous injection of platelet activating factor (PAF). The magnitude of effect was similar to that of an inhibitor of five-lipoxygenase (5-LO) and LTA4 hydrolase but superior to a leukotriene CysLT1 receptor antagonist. In summary, AM803 is a novel, potent and selective FLAP inhibitor that has excellent pharmacodynamic properties in vivo and is effective in animal models of acute inflammation and in a model of lethal shock.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Indoles/pharmacology , Inflammation/metabolism , Membrane Proteins/antagonists & inhibitors , Pentanoic Acids/pharmacology , Propionates/pharmacology , 5-Lipoxygenase-Activating Proteins , Animals , Chronic Disease , Cysteine/biosynthesis , Disease Models, Animal , Female , Humans , Indoles/pharmacokinetics , Indoles/therapeutic use , Inflammation/drug therapy , Leukotriene B4/biosynthesis , Leukotrienes/biosynthesis , Lung/drug effects , Lung/metabolism , Male , Mice , Pentanoic Acids/pharmacokinetics , Pentanoic Acids/therapeutic use , Platelet Activating Factor/pharmacology , Propionates/pharmacokinetics , Propionates/therapeutic use , Rats , Substrate Specificity , Zymosan/pharmacology
10.
J Pharmacol Exp Ther ; 332(3): 764-75, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19996299

ABSTRACT

Prostaglandin D(2) (PGD(2)) is one of a family of biologically active lipids derived from arachidonic acid via the action of COX-1 and COX-2. PGD(2) is released from mast cells and binds primarily to two G protein-coupled receptors, namely DP1 and DP2, the latter also known as chemoattractant receptor-homologous molecule expressed on Th2 cells. DP2 is predominantly expressed on eosinophils, Th2 cells, and basophils, but it is also expressed to a lesser extent on monocytes, mast cells, and epithelial cells. Interaction of PGD(2) and its active metabolites with DP2 results in cellular chemotaxis, degranulation, up-regulation of adhesion molecules, and cytokine production. Chronic obstructive pulmonary disease (COPD) is a chronic progressive inflammatory disease characterized by elevated lung neutrophils, macrophages, and CD8+ T lymphocytes and mucus hypersecretion. Cigarette smoke contributes to the etiology of COPD and was used here as a provoking agent in a murine model of COPD. In an acute model, {2'-[(cyclopropanecarbonyl-ethyl-amino)-methyl]-6-methoxy-4'-trifluoro-methyl-biphenyl-3-yl}-acetic acid, sodium salt (AM156) and (5-{2-[(benzoyloxycarbonyl-ethyl-amino)-methyl]-4-trifluoromethyl-phenyl}-pyridin-3-yl)-acetic acid, sodium salt) (AM206), potent DP2 receptor antagonists, dose-dependently inhibited influx of neutrophils and lymphocytes to smoke-exposed airways. In a subchronic model, AM156 and AM206 inhibited neutrophil and lymphocyte trafficking to the airways. Furthermore, AM156 and AM206 treatment inhibited mucus cell metaplasia and prevented the thickening of the airway epithelial layer induced by cigarette smoke. These data suggest that DP2 receptor antagonism may represent a novel therapy for COPD or other conditions characterized by neutrophil influx, mucus hypersecretion, and airway remodeling.


Subject(s)
Lung/drug effects , Mucus/metabolism , Pulmonary Disease, Chronic Obstructive/prevention & control , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Respiratory Mucosa/drug effects , Smoking/adverse effects , Animals , Benzylamines/pharmacokinetics , Benzylamines/pharmacology , Cell Line , Cell Movement , Female , Guinea Pigs , Humans , In Vitro Techniques , Inflammation/immunology , Inflammation/metabolism , Inflammation/prevention & control , Lung/immunology , Lung/pathology , Lymphocytes/drug effects , Lymphocytes/immunology , Male , Metaplasia , Mice , Mice, Inbred BALB C , Neutrophils/drug effects , Neutrophils/immunology , Niacin/analogs & derivatives , Niacin/pharmacokinetics , Niacin/pharmacology , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/pathology , Respiratory Mucosa/pathology
11.
J Pharmacol Exp Ther ; 331(3): 1042-50, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19749079

ABSTRACT

Leukotrienes (LTs) are proinflammatory lipid mediators synthesized by the conversion of arachidonic acid (AA) to LTA(4) by the enzyme 5-lipoxygenase (5-LO) in the presence of 5-LO-activating protein (FLAP). 3-[3-tert-Butylsulfanyl-1-[4-(6-methoxy-pyridin-3-yl)-benzyl]-5-(pyridin-2-ylmethoxy)-1H-indol-2-yl]-2,2-dimethyl-propionic acid (AM103) is a novel selective FLAP inhibitor in development for the treatment of respiratory conditions such as asthma. In a rat ex vivo whole-blood calcium ionophore-induced LTB(4) assay, AM103 (administered orally at 1 mg/kg) displayed >50% inhibition for up to 6 h with a calculated EC(50) of approximately 60 nM. When rat lung was challenged in vivo with calcium ionophore, AM103 inhibited LTB(4) and cysteinyl leukotriene (CysLT) production with ED(50) values of 0.8 and 1 mg/kg, respectively. In this model, the EC(50) derived from plasma AM103 was approximately 330 nM for inhibition of both LTB(4) and CysLT. In an acute inflammation setting, AM103 displayed dose-dependent inhibition of LTB(4), CysLT, and plasma protein extravasation induced by peritoneal zymosan injection. In a model of chronic lung inflammation using ovalbumin-primed and challenged BALB/c mice, AM103 reduced the concentrations of eosinophil peroxidase, CysLTs, and interleukin-5 in the bronchoalveolar lavage fluid. Finally, AM103 increased survival time in mice exposed to a lethal intravenous injection of platelet-activating factor. In summary, AM103 is a novel, potent and selective FLAP inhibitor that has excellent pharmacodynamic properties in vivo and is effective in animal models of acute and chronic inflammation and in a model of lethal shock.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carrier Proteins/antagonists & inhibitors , Indoles/pharmacology , Inflammation/drug therapy , Membrane Proteins/antagonists & inhibitors , Propionates/pharmacology , 5-Lipoxygenase-Activating Proteins , Acute Disease , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Asthma/drug therapy , Asthma/enzymology , Asthma/metabolism , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Extravasation of Diagnostic and Therapeutic Materials/drug therapy , Extravasation of Diagnostic and Therapeutic Materials/enzymology , Extravasation of Diagnostic and Therapeutic Materials/metabolism , Female , Humans , Indoles/therapeutic use , Inflammation/enzymology , Inflammation/metabolism , Leukotriene B4/biosynthesis , Leukotriene B4/blood , Male , Mice , Mice, Inbred BALB C , Pneumonia/drug therapy , Pneumonia/enzymology , Pneumonia/metabolism , Propionates/therapeutic use , Rats , Rats, Sprague-Dawley , Zymosan
12.
Bioorg Med Chem Lett ; 14(8): 1913-6, 2004 Apr 19.
Article in English | MEDLINE | ID: mdl-15050626

ABSTRACT

A series of N-acridin-9-yl-butane-1,4-diamines were found to be high-affinity ligands of the alpha(2)delta subunit of voltage gated calcium channels. The SAR studies of butane-1,4-diamine side chain resulted in the identification of compound 10 (IC(50)=9 nM), which is more potent than gabapentin (IC(50)=27 nM). Partial saturation of the acridine ring was also pursued and provided a compound with higher binding affinity than 1.


Subject(s)
Amines , Calcium Channels/physiology , Cyclohexanecarboxylic Acids , Diamines/chemistry , Diamines/pharmacology , Protein Subunits/physiology , gamma-Aminobutyric Acid , Acetates/pharmacology , Binding, Competitive/drug effects , Calcium Channels/drug effects , Diamines/chemical synthesis , Gabapentin , Ligands , Molecular Structure , Protein Binding/drug effects , Protein Binding/physiology , Protein Subunits/drug effects , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...