Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Med Chem ; 59(10): 4926-47, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27077528

ABSTRACT

Transient receptor potential vanilloid 3 (TRPV3) is a Ca(2+)- and Na(+)-permeable channel with a unique expression pattern. TRPV3 is found in both neuronal and non-neuronal tissues, including dorsal root ganglia, spinal cord, and keratinocytes. Recent studies suggest that TRPV3 may play a role in inflammation, pain sensation, and skin disorders. TRPV3 studies have been challenging, in part due to a lack of research tools such as selective antagonists. Herein, we provide the first detailed report on the development of potent and selective TRPV3 antagonists featuring a pyridinyl methanol moiety. Systematic optimization of pharmacological, physicochemical, and ADME properties of original lead 5a resulted in identification of a novel and selective TRPV3 antagonist 74a, which demonstrated a favorable preclinical profile in two different models of neuropathic pain as well as in a reserpine model of central pain.


Subject(s)
Cyclobutanes/chemical synthesis , Cyclobutanes/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , TRPV Cation Channels/antagonists & inhibitors , Calcium/metabolism , Cyclobutanes/chemistry , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Molecular Conformation , Pyridines/chemistry , Structure-Activity Relationship , TRPV Cation Channels/metabolism
2.
ACS Med Chem Lett ; 6(6): 641-4, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-26101566

ABSTRACT

The discovery of a novel peripherally acting and selective Cav3.2 T-type calcium channel blocker, ABT-639, is described. HTS hits 1 and 2, which have poor metabolic stability, were optimized to obtain 4, which has improved stability and oral bioavailability. Modification of 4 to further improve ADME properties led to the discovery of ABT-639. Following oral administration, ABT-639 produces robust antinociceptive activity in experimental pain models at doses that do not significantly alter psychomotor or hemodynamic function in the rat.

3.
J Med Chem ; 57(17): 7412-24, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25100568

ABSTRACT

The synthesis and characterization of a series of selective, orally bioavailable 1-(chroman-4-yl)urea TRPV1 antagonists is described. Whereas first-generation antagonists that inhibit all modes of TRPV1 activation can elicit hyperthermia, the compounds disclosed herein do not elevate core body temperature in preclinical models and only partially block acid activation of TRPV1. Advancing the SAR of this series led to the eventual identification of (R)-1-(7-chloro-2,2-bis(fluoromethyl)chroman-4-yl)-3-(3-methylisoquinolin-5-yl)urea (A-1165442, 52), an analogue that possesses excellent pharmacological selectivity, has a favorable pharmacokinetic profile, and demonstrates good efficacy against osteoarthritis pain in rodents.


Subject(s)
Analgesics/chemistry , Body Temperature/drug effects , TRPV Cation Channels/antagonists & inhibitors , Urea/chemistry , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Area Under Curve , Body Temperature/physiology , Dogs , Dose-Response Relationship, Drug , Drug Discovery , HEK293 Cells , Humans , Isoquinolines/chemistry , Isoquinolines/pharmacokinetics , Isoquinolines/pharmacology , Metabolic Clearance Rate , Models, Chemical , Molecular Structure , Rats , Structure-Activity Relationship , TRPV Cation Channels/chemistry , TRPV Cation Channels/metabolism , Urea/analogs & derivatives , Urea/pharmacokinetics , Urea/pharmacology
4.
Biochem Pharmacol ; 89(4): 536-44, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24726441

ABSTRACT

Activation of T-type Ca²âº channels contributes to nociceptive signaling by facilitating action potential bursting and modulation of membrane potentials during periods of neuronal hyperexcitability. The role of T-type Ca²âº channels in chronic pain is supported by gene knockdown studies showing that decreased Ca(v)3.2 channel expression results in the loss of low voltage-activated (LVA) currents in dorsal root ganglion (DRG) neurons and attenuation of neuropathic pain in the chronic constriction injury (CCI) model. ABT-639 is a novel, peripherally acting, selective T-type Ca²âº channel blocker. ABT-639 blocks recombinant human T-type (Ca(v)3.2) Ca²âº channels in a voltage-dependent fashion (IC50 = 2 µM) and attenuates LVA currents in rat DRG neurons (IC50 = 8 µM). ABT-639 was significantly less active at other Ca²âº channels (e.g. Ca(v)1.2 and Ca(v)2.2) (IC50 > 30 µM). ABT-639 has high oral bioavailability (%F = 73), low protein binding (88.9%) and a low brain:plasma ratio (0.05:1) in rodents. Following oral administration ABT-639 produced dose-dependent antinociception in a rat model of knee joint pain (ED50 = 2 mg/kg, p.o.). ABT-639 (10-100 mg/kg, p.o.) also increased tactile allodynia thresholds in multiple models of neuropathic pain (e.g. spinal nerve ligation, CCI, and vincristine-induced). [corrected]. ABT-639 did not attenuate hyperalgesia in inflammatory pain models induced by complete Freund's adjuvant or carrageenan. At higher doses (e.g. 100-300 mg/kg) ABT-639 did not significantly alter hemodynamic or psychomotor function. The antinociceptive profile of ABT-639 provides novel insights into the role of peripheral T-type (Ca(v)3.2) channels in chronic pain states.


Subject(s)
Calcium Channel Blockers/therapeutic use , Calcium Channels, T-Type/metabolism , Disease Models, Animal , Heterocyclic Compounds, 2-Ring/therapeutic use , Nerve Tissue Proteins/antagonists & inhibitors , Neuralgia/drug therapy , Nociceptive Pain/drug therapy , Peripheral Nerves/drug effects , Sulfonamides/therapeutic use , Animals , Behavior, Animal/drug effects , Biological Availability , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Calcium Channel Blockers/adverse effects , Calcium Channel Blockers/pharmacokinetics , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/genetics , Cells, Cultured , Chronic Pain/drug therapy , Chronic Pain/metabolism , Dose-Response Relationship, Drug , Heterocyclic Compounds, 2-Ring/adverse effects , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuralgia/metabolism , Nociceptive Pain/metabolism , Peripheral Nerves/cytology , Peripheral Nerves/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sulfonamides/adverse effects , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology
5.
J Pain ; 15(4): 387.e1-14, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24374196

ABSTRACT

UNLABELLED: Voltage-gated Ca(2+) channels play an important role in nociceptive transmission. There is significant evidence supporting a role for N-, T- and P/Q-type Ca(2+) channels in chronic pain. Here, we report that A-1264087, a structurally novel state-dependent blocker, inhibits each of these human Ca(2+) channels with similar potency (IC50 = 1-2 µM). A-1264087 was also shown to inhibit the release of the pronociceptive calcitonin gene-related peptide from rat dorsal root ganglion neurons. Oral administration of A-1264087 produces robust antinociceptive efficacy in monoiodoacetate-induced osteoarthritic, complete Freund adjuvant-induced inflammatory, and chronic constrictive injury of sciatic nerve-induced, neuropathic pain models with ED50 values of 3.0, 5.7, and 7.8 mg/kg (95% confidence interval = 2.2-3.5, 3.7-10, and 5.5-12.8 mg/kg), respectively. Further analysis revealed that A-1264087 also suppressed nociceptive-induced p38 and extracellular signal-regulated kinase 1/2 phosphorylation, which are biochemical markers of engagement of pain circuitry in chronic pain states. Additionally, A-1264087 inhibited both spontaneous and evoked neuronal activity in the spinal cord dorsal horn in complete Freund adjuvant-inflamed rats, providing a neurophysiological basis for the observed antihyperalgesia. A-1264087 produced no alteration of body temperature or motor coordination and no learning impairment at therapeutic plasma concentrations. PERSPECTIVE: The present results demonstrate that the neuronal Ca(2+) channel blocker A-1264087 exhibits broad-spectrum efficacy through engagement of nociceptive signaling pathways in preclinical pain models in the absence of effects on psychomotor and cognitive function.


Subject(s)
Analgesics/pharmacology , Azabicyclo Compounds/pharmacology , Calcium Channel Blockers/pharmacology , Leucine/analogs & derivatives , Neurons/metabolism , Nociception/drug effects , Spinal Cord/drug effects , Animals , Disease Models, Animal , Immunohistochemistry , Leucine/pharmacology , Male , Neurons/drug effects , Pain/metabolism , Patch-Clamp Techniques , Rats, Sprague-Dawley , Spinal Cord/metabolism
6.
Neuroimage ; 64: 341-55, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22982372

ABSTRACT

The earliest stages of osteoarthritis are characterized by peripheral pathology; however, during disease progression chronic pain emerges-a major symptom of osteoarthritis linked to neuroplasticity. Recent clinical imaging studies involving chronic pain patients, including osteoarthritis patients, have demonstrated that functional properties of the brain are altered, and these functional changes are correlated with subjective behavioral pain measures. Currently, preclinical osteoarthritis studies have not assessed if functional properties of supraspinal pain circuitry are altered, and if these functional properties can be modulated by pharmacological therapy either by direct or indirect action on brain systems. In the current study, functional connectivity was first assessed in order to characterize the functional neuroplasticity occurring in the rodent medial meniscus tear (MMT) model of osteoarthritis-a surgical model of osteoarthritis possessing peripheral joint trauma and a hypersensitive pain state. In addition to knee joint trauma at week 3 post-MMT surgery, we observed that supraspinal networks have increased functional connectivity relative to sham animals. Importantly, we observed that early and sustained treatment with a novel, peripherally acting broad-spectrum matrix metalloproteinase (MMP) inhibitor (MMPi) significantly attenuates knee joint trauma (cartilage degradation) as well as supraspinal functional connectivity increases in MMT animals. At week 5 post-MMT surgery, the acute pharmacodynamic effects of celecoxib (selective cyclooxygenase-2 inhibitor) on brain function were evaluated using pharmacological magnetic resonance imaging (phMRI) and functional connectivity analysis. Celecoxib was chosen as a comparator, given its clinical efficacy for alleviating pain in osteoarthritis patients and its peripheral and central pharmacological action. Relative to the vehicle condition, acute celecoxib treatment in MMT animals yielded decreased phMRI infusion responses and decreased functional connectivity, the latter observation being similar to what was detected following chronic MMPi treatment. These findings demonstrate that an assessment of brain function may provide an objective means by which to further evaluate the pathology of an osteoarthritis state as well as measure the pharmacodynamic effects of therapies with peripheral or peripheral and central pharmacological action.


Subject(s)
Action Potentials/drug effects , Brain/physiopathology , Disease Models, Animal , Nerve Net/physiopathology , Osteoarthritis/physiopathology , Pain/physiopathology , Pyrazoles/administration & dosage , Sulfonamides/administration & dosage , Animals , Brain/drug effects , Celecoxib , Humans , Male , Nerve Net/drug effects , Osteoarthritis/complications , Osteoarthritis/drug therapy , Pain/etiology , Pain/prevention & control , Pain Measurement/drug effects , Rats , Rats, Inbred Lew
7.
J Peripher Nerv Syst ; 17(3): 324-30, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22971094

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN) is a major toxicity of chemotherapy treatment for which no therapy is approved. Poly(ADP-ribose) polymerase (PARP)1/2 are nuclear enzymes activated upon DNA damage, and PARP1/2 inhibition provides resistance against DNA damage. A role for PARP inhibition in sensory neurotransmission has also been established. PARP inhibitors attenuate pain-like behaviors and neuropathy-associated decreased peripheral nerve function in diabetic models. The hypothesis tested was that PARP inhibition protects against painful neuropathy. The objective of this study was to investigate whether the novel, selective PARP1/2 inhibitors (ABT-888 and related analogues) would attenuate development of mechanical allodynia in vincristine-treated rats. PARP inhibitors were dosed for 2 days, and then co-administered with vincristine for 12 days. Mechanical allodynia was observed in rats treated with vincristine. PARP1/2 inhibition significantly attenuated development of mechanical allodynia and reduced poly ADP-ribose (PAR) activation in rat skin. The data presented here show that PARP inhibition attenuates vincristine-induced mechanical allodynia in rats, and supports that PARP inhibition may represent a novel therapeutic approach for CIPN.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Benzimidazoles/therapeutic use , Neuralgia/chemically induced , Neuralgia/prevention & control , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Benzimidazoles/pharmacology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Male , Neuralgia/enzymology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Sprague-Dawley
8.
J Pain ; 13(6): 519-23, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22543045

ABSTRACT

UNLABELLED: The use of von Frey filaments, originally developed by Maximilian von Frey, has become the cornerstone for assaying mechanical sensitivity in animal models and is widely used for human assessment. While there are certain limitations associated with their use that make comparisons between studies not straightforward at times, such as stimulus duration and testing frequency, von Frey filaments provide a good measurement of mechanosensation. Here we describe the application of von Frey filaments to testing in animal models, specifically with respect to determining changes in sensory thresholds in a pain state using the Dixon up-down method. In a literature survey, we found that up to 75% of reports using this method analyze the data with parametric statistical analysis and of those that used nonparametric analysis, none took into account that mechanical sensation is perceived on a logarithmic scale (Weber's Law) when calculating efficacy. Here we outline a more rigorous analysis for calculating efficacy and ED(50)'s from von Frey data that incorporates Weber's Law. We show that this analysis makes statistical and biological sense and provide a specific example of how this change affects data analysis that brings results from animal models more in line with clinical observations. PERSPECTIVE: This focus article argues that analyzing von Frey paw withdrawal threshold data obtained by using the Dixon up-down method without considering Weber's Law is inappropriate. An analysis method that incorporates how mechanical sensation is perceived and how its application brings results from animal models more in line with clinical data is presented.


Subject(s)
Models, Statistical , Pain Measurement/methods , Physical Stimulation/methods , Sensation/physiology , Animals , Pain Threshold/physiology , Research Design
9.
J Pharmacol Exp Ther ; 342(2): 416-28, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22570364

ABSTRACT

The transient receptor potential vanilloid-1 (TRPV1) channel is involved in the development and maintenance of pain and participates in the regulation of temperature. The channel is activated by diverse agents, including capsaicin, noxious heat (≥ 43°C), acidic pH (< 6), and endogenous lipids including N-arachidonoyl dopamine (NADA). Antagonists that block all modes of TRPV1 activation elicit hyperthermia. To identify efficacious TRPV1 antagonists that do not affect temperature antagonists representing multiple TRPV1 pharmacophores were evaluated at recombinant rat and human TRPV1 channels with Ca(2+) flux assays, and two classes of antagonists were identified based on their differential ability to inhibit acid activation. Although both classes of antagonists completely blocked capsaicin- and NADA-induced activation of TRPV1, select compounds only partially inhibited activation of the channel by protons. Electrophysiology and calcitonin gene-related peptide release studies confirmed the differential pharmacology of these antagonists at native TRPV1 channels in the rat. Comparison of the in vitro pharmacological properties of these TRPV1 antagonists with their in vivo effects on core body temperature confirms and expands earlier observations that acid-sparing TRPV1 antagonists do not significantly increase core body temperature. Although both classes of compounds elicit equivalent analgesia in a rat model of knee joint pain, the acid-sparing antagonist tested is not effective in a mouse model of bone cancer pain.


Subject(s)
Body Temperature/drug effects , TRPV Cation Channels/antagonists & inhibitors , Analgesics/pharmacology , Animals , Calcitonin Gene-Related Peptide/metabolism , Calcium/metabolism , Capsaicin/pharmacology , Cell Line, Transformed , Fever/drug therapy , Fever/physiopathology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C3H , Neurons/drug effects , Neurons/metabolism , Pain/drug therapy , Pain/metabolism , Pain/physiopathology , Protons , Rats , Rats, Sprague-Dawley , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , TRPV Cation Channels/metabolism
10.
Pain ; 152(5): 1165-1172, 2011 May.
Article in English | MEDLINE | ID: mdl-21402443

ABSTRACT

Despite the increasing interest in TRPA1 channel as a pain target, its role in cold sensation and body temperature regulation is not clear; the efficacy and particularly side effects resulting from channel blockade remain poorly understood. Here we use a potent, selective, and bioavailable antagonist to address these issues. A-967079 potently blocks human (IC(50): 51 nmol/L, electrophysiology, 67 nmol/L, Ca(2+) assay) and rat TRPA1 (IC(50): 101 nmol/L, electrophysiology, 289 nmol/L, Ca(2+) assay). It is >1000-fold selective over other TRP channels, and is >150-fold selective over 75 other ion channels, enzymes, and G-protein-coupled receptors. Oral dosing of A-967079 produces robust drug exposure in rodents, and exhibits analgesic efficacy in allyl isothiocyanate-induced nocifensive response and osteoarthritic pain in rats (ED(50): 23.2 mg/kg, p.o.). A-967079 attenuates cold allodynia produced by nerve injury but does not alter noxious cold sensation in naive animals, suggesting distinct roles of TRPA1 in physiological and pathological states. Unlike TRPV1 antagonists, A-967079 does not alter body temperature. It also does not produce locomotor or cardiovascular side effects. Collectively, these data provide novel insights into TRPA1 function and suggest that the selective TRPA1 blockade may present a viable strategy for alleviating pain without untoward side effects.


Subject(s)
Body Temperature Regulation/drug effects , Calcium Channels/metabolism , Cold Temperature/adverse effects , Hyperalgesia/drug therapy , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Pain/physiopathology , Sensation/physiology , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Body Temperature/drug effects , Body Temperature/physiology , Body Temperature Regulation/genetics , Body Temperature Regulation/physiology , Calcitonin Gene-Related Peptide/metabolism , Calcium/metabolism , Calcium Channels/genetics , Cells, Cultured , Disease Models, Animal , Drug Interactions , Ganglia, Spinal/pathology , Heart Rate/drug effects , Heart Rate/physiology , Humans , Hyperalgesia/physiopathology , Inhibitory Concentration 50 , Isothiocyanates/pharmacology , Magnetic Resonance Imaging/methods , Male , Mice , Nerve Tissue Proteins/genetics , Neurons/drug effects , Oximes/pharmacology , Oximes/therapeutic use , Pain/drug therapy , Pain/genetics , Pain/metabolism , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Sensation/drug effects , Sensory Thresholds/drug effects , TRPA1 Cation Channel , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/genetics , Tritium
11.
Bioorg Med Chem Lett ; 21(5): 1338-41, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21315587

ABSTRACT

Novel chroman and tetrahydroquinoline ureas were synthesized and evaluated for their activity as TRPV1 antagonists. It was found that aryl substituents on the 7- or 8-position of both bicyclic scaffolds imparted the best in vitro potency at TRPV1. The most potent chroman ureas were assessed in chronic and acute pain models, and compounds with the ability to cross the blood-brain barrier were shown to be highly efficacious. The tetrahydroquinoline ureas were found to be potent CYP3A4 inhibitors, but replacement of bulky substituents at the nitrogen atom of the tetrahydroisoquinoline moiety with small groups such as methyl can minimize the inhibition.


Subject(s)
Chromans , Quinolines , TRPV Cation Channels/antagonists & inhibitors , Urea/pharmacology , Chromans/chemical synthesis , Chromans/chemistry , Chromans/pharmacology , Humans , Inhibitory Concentration 50 , Molecular Structure , Quinolines/chemistry , Urea/chemical synthesis , Urea/chemistry
12.
Brain Res ; 1369: 158-66, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21047496

ABSTRACT

The TRPV1 receptor functions as a molecular integrator, and blockade of this receptor modulates enhanced somatosensitivity across several animal models of pathological pain, including models of osteoarthritic (OA) pain. In order to further characterize the contributions of TRPV1 to OA-related pain, we investigated the systemic effects of a selective TRPV1 receptor antagonist, A-889425, on grip force behavior, and on the evoked and spontaneous firing of spinal wide dynamic range (WDR) and nociceptive specific (NS) neurons in the monoiodoacetate (MIA) model of OA. Administration of A-889425 (10-300 µmol/kg, p.o.) alleviated grip force impairment in OA rats 3 weeks after the MIA injection. Also at 3 weeks post-MIA injection, the responses of WDR and NS neurons to 300 g von Frey hair stimulation of the knee joint were significantly reduced by A-889425 administration (10 and 30 µmol/kg, i.v.) in OA, but not sham-OA rats. Spontaneous firing of WDR neurons was elevated in the OA rats compared to sham-OA rats and may reflect ongoing discomfort in the OA animal. In addition to an effect on mechanotransmission, systemic administration of A-889425 reduced the elevated spontaneous firing of WDR neurons in OA rats but did not alter spontaneous firing in sham rats. The present data demonstrate that blockade of TRPV1 receptors modulates the firing of two important classes of spinal nociceptive neurons in a rat model of OA. The effect of A-889425 on neuronal responses to intense mechanical stimulation of the knee and on the spontaneous firing of WDR neurons adds to the growing appreciation for the role of TRPV1 receptors in pathological mechanotransmission and possibly non-evoked discomfort, respectively.


Subject(s)
Nociceptors/metabolism , Pain/metabolism , Spinal Cord/metabolism , TRPV Cation Channels/metabolism , Animals , Disease Models, Animal , Electrophysiology , Male , Nociceptors/drug effects , Osteoarthritis/complications , Osteoarthritis/metabolism , Osteoarthritis/physiopathology , Pain/physiopathology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , TRPV Cation Channels/drug effects
13.
Bioorg Med Chem ; 18(22): 7816-25, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20965738

ABSTRACT

Na(v)1.8 (also known as PN3) is a tetrodotoxin-resistant (TTx-r) voltage-gated sodium channel (VGSC) that is highly expressed on small diameter sensory neurons. It has been implicated in the pathophysiology of inflammatory and neuropathic pain, and we envisioned that selective blockade of Na(v)1.8 would be analgesic, while reducing adverse events typically associated with non-selective VGSC blocking therapeutic agents. Herein, we describe the preparation and characterization of a series of 6-aryl-2-pyrazinecarboxamides, which are potent blockers of the human Na(v)1.8 channel and also block TTx-r sodium currents in rat dorsal root ganglia (DRG) neurons. Selected derivatives display selectivity versus human Na(v)1.2. We further demonstrate that an example from this series is orally bioavailable and produces antinociceptive activity in vivo in a rodent model of neuropathic pain following oral administration.


Subject(s)
Neuralgia/drug therapy , Pyrazines/chemistry , Sodium Channel Blockers/chemistry , Sodium Channels/chemistry , Administration, Oral , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Ganglia, Spinal/cytology , Humans , Microsomes/metabolism , NAV1.8 Voltage-Gated Sodium Channel , Neurons/metabolism , Pyrazines/pharmacokinetics , Pyrazines/therapeutic use , Rats , Sodium Channel Blockers/pharmacokinetics , Sodium Channel Blockers/therapeutic use , Sodium Channels/metabolism , Structure-Activity Relationship
14.
Bioorg Med Chem Lett ; 20(22): 6812-5, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20855211

ABSTRACT

A series of aryl-substituted nicotinamide derivatives with selective inhibitory activity against the Na(v)1.8 sodium channel is reported. Replacement of the furan nucleus and homologation of the anilide linker in subtype-selective blocker A-803467 (1) provided potent, selective derivatives with improved aqueous solubility and oral bioavailability. Representative compounds from this series displayed efficacy in rat models of inflammatory and neuropathic pain.


Subject(s)
Niacinamide/pharmacology , Sodium Channel Blockers/pharmacology , Administration, Oral , Animals , Biological Availability , Niacinamide/chemistry , Niacinamide/pharmacokinetics , Rats , Sodium Channel Blockers/administration & dosage , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacokinetics , Structure-Activity Relationship
15.
Pain ; 150(2): 319-326, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20621685

ABSTRACT

The TRPV1 antagonist A-995662 demonstrates analgesic efficacy in monoiodoacetate-induced osteoarthritic (OA) pain in rat, and repeated dosing results in increased in vivo potency and a prolonged duration of action. To identify possible mechanism(s) underlying these observations, release of neuropeptides and the neurotransmitter glutamate from isolated spinal cord was measured. In OA rats, basal release of glutamate, bradykinin and calcitonin gene-related peptide (CGRP) was significantly elevated compared to naïve levels, whereas substance P (SP) levels were not changed. In vitro studies showed that capsaicin-evoked TRPV1-dependent CGRP release was 54.7+/-7.7% higher in OA, relative to levels measured for naïve rats, suggesting that TRPV1 activity was higher under OA conditions. The efficacy of A-995662 in OA corresponded with its ability to inhibit glutamate and CGRP release from the spinal cord. A single, fully efficacious dose of A-995662, 100 micromol/kg, reduced spinal glutamate and CGRP release, while a single sub-efficacious dose of A-995662 (25 micromol/kg) was ineffective. Multiple dosing with A-995662 increased the potency and duration of efficacy in OA rats. Changes in efficacy did not correlate with plasma concentrations of A-995662, but were accompanied with reductions in spinal glutamate release. These findings suggest that repeated dosing of TRPV1 antagonists enhances therapeutic potency and duration of action against OA pain, at least in part, by the sustained reduction in release of glutamate and CGRP from the spinal cord.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Glutamic Acid/metabolism , Osteoarthritis, Knee/metabolism , Pain/metabolism , Spinal Cord/drug effects , TRPV Cation Channels/antagonists & inhibitors , Tetrahydronaphthalenes/pharmacology , Analysis of Variance , Animals , Bradykinin/metabolism , Osteoarthritis, Knee/chemically induced , Pain/chemically induced , Pain Measurement , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Substance P/metabolism
16.
Bioorg Med Chem ; 18(13): 4821-9, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20570528

ABSTRACT

The synthesis and structure-activity relationships of a series of 5-monosubstituted and 4,5-disubstituted 2-arylaminooxazoles as novel antagonists of the transient receptor potential vanilloid 1 (TRPV1) receptor are described. The 7-hydroxy group of the tetrahydronaphthyl moiety on the 2-amino substituent of the oxazole ring was important for obtaining excellent in vitro potency at the human TRPV1 receptor, while a variety of alkyl and phenyl substituents at the 4- and 5-positions of the oxazole ring were well tolerated and yielded potent TRPV1 antagonists. Despite excellent in vitro potency, the 5-monosubstituted compounds suffered from poor pharmacokinetics. It was found that 4,5-disubstitution on the oxazole ring was critical to the improvement of the overall pharmacokinetic profile of these analogues, which led to the discovery of compound (R)-27, a novel TRPV1 antagonist with good oral activity in preclinical animal models of pain.


Subject(s)
Naphthols/chemical synthesis , Oxazoles/chemistry , TRPV Cation Channels/antagonists & inhibitors , Cell Line , Crystallography, X-Ray , Humans , Molecular Conformation , Naphthols/chemistry , Naphthols/pharmacokinetics , Oxazoles/chemical synthesis , Oxazoles/pharmacokinetics , TRPV Cation Channels/metabolism
17.
Neuropharmacology ; 59(3): 201-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20566409

ABSTRACT

Activation of sodium channels is essential to action potential generation and propagation. Recent genetic and pharmacological evidence indicates that activation of Na(v)1.8 channels contributes to chronic pain. Herein, we describe the identification of a novel series of structurally related pyridine derivatives as potent Na(v)1.8 channel blockers. A-887826 exemplifies this series and potently (IC(50)=11nM) blocked recombinant human Na(v)1.8 channels. A-887826 was approximately 3 fold less potent to block Na(v)1.2, approximately 10 fold less potent to block tetrodotoxin-sensitive sodium (TTX-S Na(+)) currents and was >30 fold less potent to block Na(V)1.5 channels. A-887826 potently blocked tetrodotoxin-resistant sodium (TTX-R Na(+)) currents (IC(50)=8nM) from small diameter rat dorsal root ganglion (DRG) neurons in a voltage-dependent fashion. A-887826 effectively suppressed evoked action potential firing when DRG neurons were held at depolarized potentials and reversibly suppressed spontaneous firing in small diameter DRG neurons from complete Freund's adjuvant inflamed rats. Following oral administration, A-887826 significantly attenuated tactile allodynia in a rat neuropathic pain model. Further characterization of TTX-R current block in rat DRG neurons demonstrated that A-887826 (100nM) shifted the mid-point of voltage-dependent inactivation of TTX-R currents by approximately 4mV without affecting voltage-dependent activation and did not exhibit frequency-dependent inhibition. The present data demonstrate that A-887826 is a structurally novel and potent Na(v)1.8 blocker that inhibits rat DRG TTX-R currents in a voltage-, but not frequency-dependent fashion. The ability of this structurally novel Na(v)1.8 blocker to effectively reduce tactile allodynia in neuropathic rats further supports the role of Na(v)1.8 sodium channels in pathological pain states.


Subject(s)
Hyperalgesia/drug therapy , Pain Threshold/drug effects , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use , Sodium Channels/metabolism , Animals , Biophysics , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Stimulation/methods , Ganglia, Spinal/cytology , Humans , Hyperalgesia/etiology , Male , Membrane Potentials/drug effects , Morpholines/chemistry , Morpholines/pharmacology , Morpholines/therapeutic use , NAV1.8 Voltage-Gated Sodium Channel , Neuralgia/complications , Neuralgia/etiology , Niacinamide/analogs & derivatives , Niacinamide/chemistry , Niacinamide/pharmacology , Niacinamide/therapeutic use , Patch-Clamp Techniques/methods , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Sodium Channel Blockers/chemistry , Sodium Channels/drug effects , Spinal Cord Injuries/complications , Tetrodotoxin/pharmacology , Transfection/methods
18.
Pain ; 142(1-2): 27-35, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19135797

ABSTRACT

Transient receptor potential vanilloid type 1 (TRPV1) is a ligand-gated ion channel that functions as an integrator of multiple pain stimuli including heat, acid, capsaicin and a variety of putative endogenous lipid ligands. TRPV1 antagonists have been shown to decrease inflammatory pain in animal models and to produce limited hyperthermia at analgesic doses. Here, we report that ABT-102, which is a potent and selective TRPV1 antagonist, is effective in blocking nociception in rodent models of inflammatory, post-operative, osteoarthritic, and bone cancer pain. ABT-102 decreased both spontaneous pain behaviors and those evoked by thermal and mechanical stimuli in these models. Moreover, we have found that repeated administration of ABT-102 for 5-12 days increased its analgesic activity in models of post-operative, osteoarthritic, and bone cancer pain without an associated accumulation of ABT-102 concentration in plasma or brain. Similar effects were also observed with a structurally distinct TRPV1 antagonist, A-993610. Although a single dose of ABT-102 produced a self-limiting increase in core body temperature that remained in the normal range, the hyperthermic effects of ABT-102 effectively tolerated following twice-daily dosing for 2 days. Therefore, the present data demonstrate that, following repeated administration, the analgesic activity of TRPV1 receptor antagonists is enhanced, while the associated hyperthermic effects are attenuated. The analgesic efficacy of ABT-102 supports its advancement into clinical studies.


Subject(s)
Analgesics/administration & dosage , Fever/drug therapy , Indazoles/administration & dosage , Pain Threshold/drug effects , Pain/drug therapy , TRPV Cation Channels/metabolism , Urea/analogs & derivatives , Animals , Body Temperature/drug effects , Bone Neoplasms/complications , Calcium/metabolism , Disease Models, Animal , Drug Interactions , Fever/chemically induced , Inflammation/complications , Male , Mice , Mice, Inbred C3H , Motor Activity/drug effects , Osteoarthritis/complications , Pain/etiology , Pain Measurement , Rats , Rats, Sprague-Dawley , TRPV Cation Channels/antagonists & inhibitors , Urea/administration & dosage
19.
J Neurophysiol ; 100(6): 3158-66, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18829846

ABSTRACT

TRPV1 receptors are activated and/or modulated by noxious heat, capsaicin, protons and other endogenous agents released following tissue injury. There is a growing appreciation that this molecular integrator may also have a role in mechanosensation. To further understand this role, we investigated the systemic and site-specific effects of a selective TRPV1 receptor antagonist, A-889425, on low-intensity mechanical stimulation in inflamed rats. Systemic administration of A-889425 (30 and 100 micromol/kg po) reduced mechanical allodynia in complete Freund's adjuvant (CFA)-inflamed rats. Systemic A-889425 (3 and 10 micromol/kg iv) also decreased the responses of spinal wide dynamic range (WDR) neurons to low-intensity mechanical stimulation in CFA-inflamed but not uninjured rats. This effect of A-889425 was likely mediated via multiple sites since local injection of A-889425 into the spinal cord (1-3 nmol), ipsilateral hindpaw (200 nmol), and cerebral ventricles (30-300 nmol) all attenuated WDR responses to low-intensity mechanical stimulation. In addition to an effect on mechanotransmission, systemic administration of A-889425 reduced the spontaneous firing of WDR neurons in inflamed but not uninjured rats. Spontaneous firing is elevated after injury and may reflect ongoing pain in the animal. Local injection experiments indicated that this effect of A-889425 on spontaneous firing was mainly mediated via TRPV1 receptors in the spinal cord. Thus the current data demonstrate that TRPV1 receptors have an enhanced role after an inflammatory injury, impacting both low-intensity mechanotransmission and possibly spontaneous pain. Furthermore this study delineates the differential contribution of central and peripheral TRPV1 receptors to affect spontaneous or mechanically evoked firing of WDR neurons.


Subject(s)
Action Potentials/physiology , Inflammation/pathology , Neurons/physiology , Spinal Cord/pathology , TRPV Cation Channels/physiology , Action Potentials/drug effects , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Evoked Potentials, Somatosensory/drug effects , Freund's Adjuvant/pharmacology , Inflammation/chemically induced , Male , Neurons/drug effects , Pain Threshold/drug effects , Physical Stimulation/methods , Pyridines/chemistry , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , TRPV Cation Channels/antagonists & inhibitors
20.
Bioorg Med Chem ; 16(12): 6379-86, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18501613

ABSTRACT

The synthesis and pharmacological characterization of a novel furan-based class of voltage-gated sodium channel blockers is reported. Compounds were evaluated for their ability to block the tetrodotoxin-resistant sodium channel Na(v)1.8 (PN3) as well as the Na(v)1.2 and Na(v)1.5 subtypes. Benchmark compounds from this series possessed enhanced potency, oral bioavailability, and robust efficacy in a rodent model of neuropathic pain, together with improved CNS and cardiovascular safety profiles compared to the clinically used sodium channel blockers mexiletine and lamotrigine.


Subject(s)
Analgesics, Non-Narcotic/chemistry , Analgesics, Non-Narcotic/pharmacology , Furans/chemistry , Furans/pharmacology , Neuralgia/drug therapy , Piperazines/chemistry , Piperazines/pharmacology , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Analgesics, Non-Narcotic/chemical synthesis , Animals , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Furans/chemical synthesis , Humans , Male , Mice , Piperazines/chemical synthesis , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/chemical synthesis , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...