Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters

Publication year range
1.
J Pharmacol Exp Ther ; 359(1): 194-206, 2016 10.
Article in English | MEDLINE | ID: mdl-27432892

ABSTRACT

The renal outer medullary potassium (ROMK) channel, located at the apical surface of epithelial cells in the thick ascending loop of Henle and cortical collecting duct, contributes to salt reabsorption and potassium secretion, and represents a target for the development of new mechanism of action diuretics. This idea is supported by the phenotype of antenatal Bartter's syndrome type II associated with loss-of-function mutations in the human ROMK channel, as well as, by cardiovascular studies of heterozygous carriers of channel mutations associated with type II Bartter's syndrome. Although the pharmacology of ROMK channels is still being developed, channel inhibitors have been identified and shown to cause natriuresis and diuresis, in the absence of any significant kaliuresis, on acute oral dosing to rats or dogs. Improvements in potency and selectivity have led to the discovery of MK-7145 [5,5'-((1R,1'R)-piperazine-1,4-diylbis(1-hydroxyethane-2,1-diyl))bis(4-methylisobenzofuran-1(3H)-one)], a potential clinical development candidate. In spontaneously hypertensive rats, oral dosing of MK-7145 causes dose-dependent lowering of blood pressure that is maintained during the entire treatment period, and that displays additive/synergistic effects when administered in combination with hydrochlorothiazide or candesartan, respectively. Acute or chronic oral administration of MK-7145 to normotensive dogs led to dose-dependent diuresis and natriuresis, without any significant urinary potassium losses or changes in plasma electrolyte levels. Elevations in bicarbonate and aldosterone were found after 6 days of dosing. These data indicate that pharmacological inhibition of ROMK has potential as a new mechanism for the treatment of hypertension and/or congestive heart failure. In addition, Bartter's syndrome type II features are manifested on exposure to ROMK inhibitors.


Subject(s)
Bartter Syndrome/physiopathology , Benzofurans/pharmacology , Blood Pressure/drug effects , Phenotype , Piperazines/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Animals , Bartter Syndrome/drug therapy , Benzimidazoles/pharmacology , Benzofurans/therapeutic use , Biphenyl Compounds , Dogs , Dose-Response Relationship, Drug , Drug Synergism , Female , HEK293 Cells , Humans , Hydrochlorothiazide/pharmacology , Male , Piperazines/therapeutic use , Potassium Channel Blockers/therapeutic use , Rats , Tetrazoles/pharmacology
2.
Bioorg Med Chem Lett ; 26(9): 2339-43, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27017115

ABSTRACT

Following the discovery of small molecule acyl piperazine ROMK inhibitors and their initial preclinical validation as a novel diuretic agent, our group set out to discover new ROMK inhibitors with reduced risk for QT effects, suitable for further pharmacological experiments in additional species. Several strategies for decreasing hERG affinity while maintaining ROMK inhibition were investigated and are described herein. The most promising candidate, derived from the newly discovered 4-N-heteroaryl acetyl series, improved functional hERG/ROMK ratio by >10× over the previous lead. In vivo evaluation demonstrated comparable diuretic effects in rat with no detectable QT effects at the doses evaluated in an in vivo dog model.


Subject(s)
ERG1 Potassium Channel/physiology , Heterocyclic Compounds/pharmacology , Piperazines/pharmacology , Heterocyclic Compounds/chemistry , Piperazines/chemistry , Structure-Activity Relationship
3.
Bioorg Med Chem Lett ; 26(23): 5695-5702, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27839686

ABSTRACT

Following the discovery of small molecule acyl piperazine ROMK inhibitors, the acyl octahydropyrazino[2,1-c][1,4]oxazine series was identified. This series displays improved ROMK/hERG selectivity, and as a consequence, the resulting ROMK inhibitors do not evoke QTc prolongation in an in vivo cardiovascular dog model. Further efforts in this series led to the discovery of analogs with improved pharmacokinetic profiles. This new series also retained comparable ROMK potency compared to earlier leads.


Subject(s)
Oxazines/chemistry , Oxazines/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Animals , Diuresis/drug effects , Dogs , Heart Failure/drug therapy , Humans , Hypertension/drug therapy , Macaca mulatta , Oxazines/pharmacokinetics , Potassium Channels, Inwardly Rectifying/metabolism , Rats, Sprague-Dawley , Transcriptional Regulator ERG/antagonists & inhibitors , Transcriptional Regulator ERG/metabolism
4.
J Magn Reson Imaging ; 41(6): 1622-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25168165

ABSTRACT

BACKGROUND: To characterize regional kidney sodium response by MRI following NKCC2 inhibition. METHODS: Regional renal sodium signals were monitored noninvasively using (23) Na-MRI at 9.4T with a temporal resolution of 1.5 min in anesthetized rats (N = 14). A mild NKCC2 inhibition was induced using a slow intravenous furosemide infusion. Time course of sodium signal was modeled as an exponential transient with a single characteristic time constant. RESULTS: Under normal physiological conditions, the renal sodium signals in medullary and cortical regions were stable and found to respond differently to furosemide challenge. Furosemide infusion at 1.2 mg/kg/h (N = 7) increased sodium signal in the cortex by 40 ± 6% (P < 7 × 10(-5) ) whereas decreased in the medulla by 29 ± 2% (P < 3 × 10(-6) ) with different temporal kinetics. The characteristic time constants of the change were determined to be: 8 ± 2 and 70 ± 10 min for medulla and cortex. Also, the medullary change occurred 9(±3) times faster than cortical independent of furosemide infusion rate up to 35 mg/kg/h. CONCLUSION: The pharmacological effects in terms of regional kidney sodium signal changes induced by NKCC2 inhibition are region-specific and highly predictable. Using noninvasive sodium MRI, we obtained regional renal sodium kinetics data sets in response to a low dose furosemide infusion in normal rats.


Subject(s)
Diuretics/pharmacology , Furosemide/pharmacology , Kidney Cortex/drug effects , Kidney Medulla/drug effects , Magnetic Resonance Imaging/methods , Sodium/metabolism , Animals , Male , Rats , Rats, Sprague-Dawley
5.
J Pharmacol Exp Ther ; 348(1): 153-64, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24142912

ABSTRACT

The renal outer medullary potassium (ROMK) channel, which is located at the apical membrane of epithelial cells lining the thick ascending loop of Henle and cortical collecting duct, plays an important role in kidney physiology by regulating salt reabsorption. Loss-of-function mutations in the human ROMK channel are associated with antenatal type II Bartter's syndrome, an autosomal recessive life-threatening salt-wasting disorder with mild hypokalemia. Similar observations have been reported from studies with ROMK knockout mice and rats. It is noteworthy that heterozygous carriers of Kir1.1 mutations associated with antenatal Bartter's syndrome have reduced blood pressure and a decreased risk of developing hypertension by age 60. Although selective ROMK inhibitors would be expected to represent a new class of diuretics, this hypothesis has not been pharmacologically tested. Compound A [5-(2-(4-(2-(4-(1H-tetrazol-1-yl)phenyl)acetyl)piperazin-1-yl)ethyl)isobenzofuran-1(3H)-one)], a potent ROMK inhibitor with appropriate selectivity and characteristics for in vivo testing, has been identified. Compound A accesses the channel through the cytoplasmic side and binds to residues lining the pore within the transmembrane region below the selectivity filter. In normotensive rats and dogs, short-term oral administration of compound A caused concentration-dependent diuresis and natriuresis that were comparable to hydrochlorothiazide. Unlike hydrochlorothiazide, however, compound A did not cause any significant urinary potassium losses or changes in plasma electrolyte levels. These data indicate that pharmacologic inhibition of ROMK has the potential for affording diuretic/natriuretic efficacy similar to that of clinically used diuretics but without the dose-limiting hypokalemia associated with the use of loop and thiazide-like diuretics.


Subject(s)
Diuresis/drug effects , Diuresis/physiology , Natriuresis/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Dogs , Dose-Response Relationship, Drug , Female , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Male , Natriuresis/physiology , Rats , Rats, Sprague-Dawley
7.
J Pharmacol Exp Ther ; 344(2): 407-16, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23161216

ABSTRACT

The voltage-gated potassium channels Kv2.1 and Kv2.2 are highly expressed in pancreatic islets, yet their contribution to islet hormone secretion is not fully understood. Here we investigate the role of Kv2 channels in pancreatic islets using a combination of genetic and pharmacologic approaches. Pancreatic ß-cells from Kv2.1(-/-) mice possess reduced Kv current and display greater glucose-stimulated insulin secretion (GSIS) relative to WT ß-cells. Inhibition of Kv2.x channels with selective peptidyl [guangxitoxin-1E (GxTX-1E)] or small molecule (RY796) inhibitors enhances GSIS in isolated wild-type (WT) mouse and human islets, but not in islets from Kv2.1(-/-) mice. However, in WT mice neither inhibitor improved glucose tolerance in vivo. GxTX-1E and RY796 enhanced somatostatin release in isolated human and mouse islets and in situ perfused pancreata from WT and Kv2.1(-/-) mice. Kv2.2 silencing in mouse islets by adenovirus-small hairpin RNA (shRNA) specifically enhanced islet somatostatin, but not insulin, secretion. In mice lacking somatostatin receptor 5, GxTX-1E stimulated insulin secretion and improved glucose tolerance. Collectively, these data show that Kv2.1 regulates insulin secretion in ß-cells and Kv2.2 modulates somatostatin release in δ-cells. Development of selective Kv2.1 inhibitors without cross inhibition of Kv2.2 may provide new avenues to promote GSIS for the treatment of type 2 diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Shab Potassium Channels/metabolism , Somatostatin/metabolism , Adult , Animals , Arthropod Proteins , Benzamides/pharmacology , Cells, Cultured , Electrophysiological Phenomena , Female , Glucose/pharmacology , Humans , Insulin Secretion , Insulin-Secreting Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Patch-Clamp Techniques , Peptides/pharmacology , Potassium Channel Blockers/pharmacology , Protein Binding , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Shab Potassium Channels/antagonists & inhibitors , Shab Potassium Channels/genetics , Spider Venoms/pharmacology , Young Adult
8.
Bioorg Med Chem Lett ; 23(12): 3640-5, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23652221

ABSTRACT

A series of benzazepinones were synthesized and evaluated for block of Nav1.7 sodium channels. Compound 30 from this series displayed potent channel block, good selectivity versus other targets, and dose-dependent oral efficacy in a rat model of neuropathic pain.


Subject(s)
Benzazepines/pharmacology , Neuralgia/drug therapy , Sodium Channel Blockers/pharmacology , Animals , Disease Models, Animal , Rats
9.
Bioorg Med Chem Lett ; 23(21): 5829-32, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24075732

ABSTRACT

A sub-class of distinct small molecule ROMK inhibitors were developed from the original lead 1. Medicinal chemistry endeavors led to novel ROMK inhibitors with good ROMK functional potency and improved hERG selectivity. Two of the described ROMK inhibitors were characterized for the first in vivo proof-of-concept biology studies, and results from an acute rat diuresis model confirmed the hypothesis that ROMK inhibitors represent new mechanism diuretic and natriuretic agents.


Subject(s)
Benzofurans/chemistry , Benzofurans/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Animals , Benzofurans/pharmacokinetics , Diuresis/drug effects , Drug Discovery , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Potassium Channels, Inwardly Rectifying/metabolism , Rats , Rats, Sprague-Dawley , Tetrazoles/chemistry , Tetrazoles/pharmacokinetics , Tetrazoles/pharmacology
10.
Mol Pharmacol ; 81(4): 567-77, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22241372

ABSTRACT

High-conductance calcium-activated potassium (Maxi-K) channels are present in smooth muscle where they regulate tone. Activation of Maxi-K channels causes smooth muscle hyperpolarization and shortening of action-potential duration, which would limit calcium entry through voltage-dependent calcium channels leading to relaxation. Although Maxi-K channels appear to indirectly mediate the relaxant effects of a number of agents, activators that bind directly to the channel with appropriate potency and pharmacological properties useful for proof-of-concept studies are not available. Most agents identified to date display significant polypharmacy that severely compromises interpretation of experimental data. In the present study, a high-throughput, functional, cell-based assay for identifying Maxi-K channel agonists was established and used to screen a large sample collection (>1.6 million compounds). On the basis of potency and selectivity, a family of tetrahydroquinolines was further characterized. Medicinal chemistry efforts afforded identification of compound X, from which its two enantiomers, Y and Z, were resolved. In in vitro assays, Z is more potent than Y as a channel activator. The same profile is observed in tissues where the ability of either agent to relax precontracted smooth muscles, via a potassium channel-dependent mechanism, is demonstrated. These data, taken together, suggest that direct activation of Maxi-K channels represents a mechanism to be explored for the potential treatment of a number of diseases associated with smooth muscle hyperexcitability.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/physiology , Muscle, Smooth/physiology , Animals , CHO Cells , Chromatography, Liquid , Cricetinae , Cricetulus , Large-Conductance Calcium-Activated Potassium Channels/agonists , Magnetic Resonance Spectroscopy , Mass Spectrometry , Muscle Relaxation
11.
Mol Pharmacol ; 81(3): 488-97, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22188924

ABSTRACT

Biological, genetic, and clinical evidence provide validation for N-type calcium channels (Ca(V)2.2) as therapeutic targets for chronic pain. A state-dependent Ca(V)2.2 inhibitor may provide an improved therapeutic window over ziconotide, the peptidyl Ca(V)2.2 inhibitor used clinically. Supporting this notion, we recently reported that in preclinical models, the state-dependent Ca(V)2 inhibitor (3R)-5-(3-chloro-4-fluorophenyl)-3-methyl-3-(pyrimidin-5-ylmethyl)-1-(1H-1,2,4-triazol-3-yl)-1,3-dihydro-2H-indol-2-one (TROX-1) has an improved therapeutic window compared with ziconotide. Here we characterize TROX-1 inhibition of Cav2.2 channels in more detail. When channels are biased toward open/inactivated states by depolarizing the membrane potential under voltage-clamp electrophysiology, TROX-1 inhibits Ca(V)2.2 channels with an IC(50) of 0.11 µM. The voltage dependence of Ca(V)2.2 inhibition was examined using automated electrophysiology. TROX-1 IC(50) values were 4.2, 0.90, and 0.36 µM at -110, -90, and -70 mV, respectively. TROX-1 displayed use-dependent inhibition of Ca(V)2.2 with a 10-fold IC(50) separation between first (27 µM) and last (2.7 µM) pulses in a train. In a fluorescence-based calcium influx assay, TROX-1 inhibited Ca(V)2.2 channels with an IC(50) of 9.5 µM under hyperpolarized conditions and 0.69 µM under depolarized conditions. Finally, TROX-1 potency was examined across the Ca(V)2 subfamily. Depolarized IC(50) values were 0.29, 0.19, and 0.28 µM by manual electrophysiology using matched conditions and 1.8, 0.69, and 1.1 µM by calcium influx for Ca(V)2.1, Ca(V)2.2, and Ca(V)2.3, respectively. Together, these in vitro data support the idea that a state-dependent, non-subtype-selective Ca(V)2 channel inhibitor can achieve an improved therapeutic window over the relatively state-independent Ca(V)2.2-selective inhibitor ziconotide in preclinical models of chronic pain.


Subject(s)
Calcium Channel Blockers/chemistry , Calcium Channels, N-Type/drug effects , Indoles/chemistry , Triazoles/chemistry , Calcium Channel Blockers/pharmacology , Cell Line , Humans , Indoles/pharmacology , Inhibitory Concentration 50 , Membrane Potentials/drug effects , Patch-Clamp Techniques , Triazoles/pharmacology
12.
Mol Pharmacol ; 80(6): 959-64, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21948463

ABSTRACT

Identification of selective ion channel inhibitors represents a critical step for understanding the physiological role that these proteins play in native systems. In particular, voltage-gated potassium (K(V)2) channels are widely expressed in tissues such as central nervous system, pancreas, and smooth muscle, but their particular contributions to cell function are not well understood. Although potent and selective peptide inhibitors of K(V)2 channels have been characterized, selective small molecule K(V)2 inhibitors have not been reported. For this purpose, high-throughput automated electrophysiology (IonWorks Quattro; Molecular Devices, Sunnyvale, CA) was used to screen a 200,000-compound mixture (10 compounds per sample) library for inhibitors of K(V)2.1 channels. After deconvolution of 190 active samples, two compounds (A1 and B1) were identified that potently inhibit K(V)2.1 and the other member of the K(V)2 family, K(V)2.2 (IC(50), 0.1-0.2 µM), and that possess good selectivity over K(V)1.2 (IC(50) >10 µM). Modeling studies suggest that these compounds possess a similar three-dimensional conformation. Compounds A1 and B1 are >10-fold selective over Na(V) channels and other K(V) channels and display weak activity (5-9 µM) on Ca(V) channels. The biological activity of compound A1 on native K(V)2 channels was confirmed in electrophysiological recordings of rat insulinoma cells, which are known to express K(V)2 channels. Medicinal chemistry efforts revealed a defined structure-activity relationship and led to the identification of two compounds (RY785 and RY796) without significant Ca(V) channel activity. Taken together, these newly identified channel inhibitors represent important tools for the study of K(V)2 channels in biological systems.


Subject(s)
Drug Discovery/methods , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Shab Potassium Channels/antagonists & inhibitors , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Rats , Shab Potassium Channels/physiology , Structure-Activity Relationship
14.
Proc Natl Acad Sci U S A ; 105(32): 11140-5, 2008 Aug 12.
Article in English | MEDLINE | ID: mdl-18682566

ABSTRACT

Niemann-Pick C1-like protein (NPC1L1) mediates the absorption of dietary cholesterol in the proximal region of the intestine, a process that is blocked by cholesterol absorption inhibitors (CAIs), including ezetimibe (EZE). Using a proteomic approach, we demonstrate that NPC1L1 is the protein to which EZE and its analogs bind. Next, we determined the site of interaction of EZE analogs with NPC1L1 by exploiting the different binding affinities of mouse and dog NPC1L1 for the radioligand analog of EZE, [(3)H]AS. Chimeric and mutational studies indicate that high-affinity binding of [(3)H]AS to dog NPC1L1 depends on molecular determinants present in a 61-aa region of a large extracellular domain (loop C), where Phe-532 and Met-543 appear to be key contributors. These data suggest that the [(3)H]AS-binding site resides in the intestinal lumen and are consistent with preclinical data demonstrating in vivo efficacy of a minimally bioavailable CAI. Furthermore, these determinants of [(3)H]AS binding lie immediately adjacent to a hotspot of human NPC1L1 polymorphisms correlated with hypoabsorption of cholesterol. These observations, taken together with the recently described binding of cholesterol to the N terminus (loop A) of the close NPC1L1 homologue, NPC1, may provide a molecular basis for understanding EZE inhibition of NPC1L1-mediated cholesterol absorption. Specifically, EZE binding to an extracellular site distinct from where cholesterol binds prevents conformational changes in NPC1L1 that are necessary for the translocation of cholesterol across the membrane.


Subject(s)
Anticholesteremic Agents/pharmacology , Azetidines/pharmacology , Cholesterol, Dietary/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Animals , Binding Sites/drug effects , Binding Sites/genetics , Biological Transport, Active/drug effects , Biological Transport, Active/genetics , Cell Line , Cell Membrane/genetics , Cell Membrane/metabolism , Dogs , Ezetimibe , Intestinal Absorption/drug effects , Intestinal Absorption/genetics , Membrane Transport Proteins/genetics , Mice , Mutation , Polymorphism, Genetic , Protein Binding/drug effects , Protein Binding/genetics , Protein Structure, Secondary/genetics , Protein Structure, Tertiary/genetics , Proteomics/methods
15.
J Pharmacol Exp Ther ; 334(2): 545-55, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20439438

ABSTRACT

Voltage-gated calcium channel (Ca(v))2.2 (N-type calcium channels) are key components in nociceptive transmission pathways. Ziconotide, a state-independent peptide inhibitor of Ca(v)2.2 channels, is efficacious in treating refractory pain but exhibits a narrow therapeutic window and must be administered intrathecally. We have discovered an N-triazole oxindole, (3R)-5-(3-chloro-4-fluorophenyl)-3-methyl-3-(pyrimidin-5-ylmethyl)-1-(1H-1,2,4-triazol-3-yl)-1,3-dihydro-2H-indol-2-one (TROX-1), as a small-molecule, state-dependent blocker of Ca(v)2 channels, and we investigated the therapeutic advantages of this compound for analgesia. TROX-1 preferentially inhibited potassium-triggered calcium influx through recombinant Ca(v)2.2 channels under depolarized conditions (IC(50) = 0.27 microM) compared with hyperpolarized conditions (IC(50) > 20 microM). In rat dorsal root ganglion (DRG) neurons, TROX-1 inhibited omega-conotoxin GVIA-sensitive calcium currents (Ca(v)2.2 channel currents), with greater potency under depolarized conditions (IC(50) = 0.4 microM) than under hyperpolarized conditions (IC(50) = 2.6 microM), indicating state-dependent Ca(v)2.2 channel block of native as well as recombinant channels. TROX-1 fully blocked calcium influx mediated by a mixture of Ca(v)2 channels in calcium imaging experiments in rat DRG neurons, indicating additional block of all Ca(v)2 family channels. TROX-1 reversed inflammatory-induced hyperalgesia with maximal effects equivalent to nonsteroidal anti-inflammatory drugs, and it reversed nerve injury-induced allodynia to the same extent as pregabalin and duloxetine. In contrast, no significant reversal of hyperalgesia was observed in Ca(v)2.2 gene-deleted mice. Mild impairment of motor function in the Rotarod test and cardiovascular functions were observed at 20- to 40-fold higher plasma concentrations than required for analgesic activities. TROX-1 demonstrates that an orally available state-dependent Ca(v)2 channel blocker may achieve a therapeutic window suitable for the treatment of chronic pain.


Subject(s)
Analgesics/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/physiology , Indoles/pharmacology , Triazoles/pharmacology , Analgesics/adverse effects , Analgesics/pharmacokinetics , Animals , Baroreflex/drug effects , Biological Availability , Calcium Channel Blockers/adverse effects , Calcium Channel Blockers/pharmacokinetics , Calcium Channels, N-Type/genetics , Calcium Channels, R-Type/physiology , Cation Transport Proteins/physiology , Cell Line , Dogs , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Hyperalgesia/drug therapy , Hypotension, Orthostatic/chemically induced , Indoles/adverse effects , Indoles/pharmacokinetics , Male , Mice , Mice, Knockout , Neurons/drug effects , Neurons/physiology , Pain/drug therapy , Pain/etiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Triazoles/adverse effects , Triazoles/pharmacokinetics
17.
Bioorg Med Chem Lett ; 20(18): 5536-40, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20709552

ABSTRACT

Voltage-gated sodium channels have been shown to play a critical role in neuropathic pain. With a goal to develop potent peripherally active sodium channel blockers, a series of low molecular weight biaryl substituted imidazoles, oxazoles, and thiazole carboxamides were identified with good in vitro and in vivo potency.


Subject(s)
Neuralgia/drug therapy , Oxazoles/therapeutic use , Sodium Channel Blockers/therapeutic use , Sodium Channels/metabolism , Thiazoles/therapeutic use , Animals , Dogs , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Imidazoles/pharmacology , Imidazoles/therapeutic use , Microsomes, Liver/metabolism , NAV1.7 Voltage-Gated Sodium Channel , Oxazoles/chemistry , Oxazoles/metabolism , Oxazoles/pharmacology , Rats , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/metabolism , Sodium Channel Blockers/pharmacology , Thiazoles/chemistry , Thiazoles/metabolism , Thiazoles/pharmacology
19.
Bioorg Med Chem Lett ; 19(18): 5334-8, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19683443

ABSTRACT

A series of novel isoxazole voltage gated sodium channel blockers have been synthesized and evaluated. Substitutions on the benzylic position of benzamide were investigated to determine their effect on Na(v)1.7 inhibitory potency. The spirocyclobutyl substitution had the most significant enhancement on Na(v)1.7 inhibitory activity.


Subject(s)
Isoxazoles/therapeutic use , Pain/drug therapy , Sodium Channel Blockers/therapeutic use , Sodium Channels/metabolism , Animals , Cell Line , Chronic Disease , Humans , Isoxazoles/chemistry , Isoxazoles/pharmacology , Pain/immunology , Rats , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacology , Spinal Nerves/drug effects , Structure-Activity Relationship
20.
Bioorg Med Chem Lett ; 19(18): 5329-33, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19674896

ABSTRACT

Analogs of the previously reported voltage gated sodium channel blocker CDA54 were prepared in which one of the amide functions was replaced with aromatic and non-aromatic heterocycles. Replacement of the amide with an aromatic heterocycle resulted in significant loss of sodium channel blocking activity, while non-aromatic heterocycle replacements were well tolerated.


Subject(s)
Isoxazoles/chemistry , Isoxazoles/pharmacology , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacology , Animals , Isoxazoles/therapeutic use , Models, Molecular , Molecular Structure , Pain/drug therapy , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/therapeutic use , Spinal Nerves/drug effects , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL