Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Med Chem ; 64(6): 3427-3438, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33715378

ABSTRACT

Inhibition of the pituitary adenylate cyclase 1 receptor (PAC1R) is a novel mechanism that could be used for abortive treatment of acute migraine. Our research began with comparative analysis of known PAC1R ligand scaffolds, PACAP38 and Maxadilan, which resulted in the selection of des(24-42) Maxadilan, 6, as a starting point. C-terminal modifications of 6 improved the peptide metabolic stability in vitro and in vivo. SAR investigations identified synergistic combinations of amino acid replacements that significantly increased the in vitro PAC1R inhibitory activity of the analogs to the pM IC90 range. Our modifications further enabled deletion of up to six residues without impacting potency, thus improving peptide ligand binding efficiency. Analogs 17 and 18 exhibited robust in vivo efficacy in the rat Maxadilan-induced increase in blood flow (MIIBF) pharmacodynamic model at 0.3 mg/kg subcutaneous dosing. The first cocrystal structure of a PAC1R antagonist peptide (18) with PAC1R extracellular domain is reported.


Subject(s)
Blood Circulation/drug effects , Peptides/chemistry , Peptides/pharmacology , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/antagonists & inhibitors , Animals , Humans , Insect Proteins/pharmacology , Male , Mice , Migraine Disorders/drug therapy , Migraine Disorders/metabolism , Migraine Disorders/physiopathology , Molecular Docking Simulation , Peptides/pharmacokinetics , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Vasodilator Agents/pharmacology
2.
Pain ; 161(7): 1670-1681, 2020 07.
Article in English | MEDLINE | ID: mdl-32142016

ABSTRACT

Pituitary adenylate cyclase activating polypeptide-38 (PACAP38) may play an important role in primary headaches. Preclinical evidence suggests that PACAP38 modulates trigeminal nociceptive activity mainly through PAC1 receptors while clinical studies report that plasma concentrations of PACAP38 are elevated in spontaneous attacks of cluster headache and migraine and normalize after treatment with sumatriptan. Intravenous infusion of PACAP38 induces migraine-like attacks in migraineurs and cluster-like attacks in cluster headache patients. A rodent-specific PAC1 receptor antibody Ab181 was developed, and its effect on nociceptive neuronal activity in the trigeminocervical complex was investigated in vivo in an electrophysiological model relevant to primary headaches. Ab181 is potent and selective at the rat PAC1 receptor and provides near-maximum target coverage at 10 mg/kg for more than 48 hours. Without affecting spontaneous neuronal activity, Ab181 effectively inhibits stimulus-evoked activity in the trigeminocervical complex. Immunohistochemical analysis revealed its binding in the trigeminal ganglion and sphenopalatine ganglion but not within the central nervous system suggesting a peripheral site of action. The pharmacological approach using a specific PAC1 receptor antibody could provide a novel mechanism with a potential clinical efficacy in the treatment of primary headaches.


Subject(s)
Migraine Disorders , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Animals , Headache/chemically induced , Headache/drug therapy , Humans , Nociception , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Rats
3.
J Pharmacol Exp Ther ; 362(1): 146-160, 2017 07.
Article in English | MEDLINE | ID: mdl-28473457

ABSTRACT

Potent and selective antagonists of the voltage-gated sodium channel NaV1.7 represent a promising avenue for the development of new chronic pain therapies. We generated a small molecule atropisomer quinolone sulfonamide antagonist AMG8379 and a less active enantiomer AMG8380. Here we show that AMG8379 potently blocks human NaV1.7 channels with an IC50 of 8.5 nM and endogenous tetrodotoxin (TTX)-sensitive sodium channels in dorsal root ganglion (DRG) neurons with an IC50 of 3.1 nM in whole-cell patch clamp electrophysiology assays using a voltage protocol that interrogates channels in a partially inactivated state. AMG8379 was 100- to 1000-fold selective over other NaV family members, including NaV1.4 expressed in muscle and NaV1.5 expressed in the heart, as well as TTX-resistant NaV channels in DRG neurons. Using an ex vivo mouse skin-nerve preparation, AMG8379 blocked mechanically induced action potential firing in C-fibers in both a time-dependent and dose-dependent manner. AMG8379 similarly reduced the frequency of thermally induced C-fiber spiking, whereas AMG8380 affected neither mechanical nor thermal responses. In vivo target engagement of AMG8379 in mice was evaluated in multiple NaV1.7-dependent behavioral endpoints. AMG8379 dose-dependently inhibited intradermal histamine-induced scratching and intraplantar capsaicin-induced licking, and reversed UVB radiation skin burn-induced thermal hyperalgesia; notably, behavioral effects were not observed with AMG8380 at similar plasma exposure levels. AMG8379 is a potent and selective NaV1.7 inhibitor that blocks sodium current in heterologous cells as well as DRG neurons, inhibits action potential firing in peripheral nerve fibers, and exhibits pharmacodynamic effects in translatable models of both itch and pain.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/drug effects , Sodium Channel Blockers/pharmacology , Action Potentials/drug effects , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Humans , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Myocardium/metabolism , Neurons/drug effects , Pain/prevention & control , Pain/psychology , Patch-Clamp Techniques , Pruritus/prevention & control , Pruritus/psychology , Quinolones/pharmacology , Small Molecule Libraries , Stereoisomerism , Sulfonamides/pharmacology
4.
J Med Chem ; 60(14): 5990-6017, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28324649

ABSTRACT

Because of its strong genetic validation, NaV1.7 has attracted significant interest as a target for the treatment of pain. We have previously reported on a number of structurally distinct bicyclic heteroarylsulfonamides as NaV1.7 inhibitors that demonstrate high levels of selectivity over other NaV isoforms. Herein, we report the discovery and optimization of a series of atropisomeric quinolinone sulfonamide inhibitors [ Bicyclic sulfonamide compounds as sodium channel inhibitors and their preparation . WO 2014201206, 2014 ] of NaV1.7, which demonstrate nanomolar inhibition of NaV1.7 and exhibit high levels of selectivity over other sodium channel isoforms. After optimization of metabolic and pharmacokinetic properties, including PXR activation, CYP2C9 inhibition, and CYP3A4 TDI, several compounds were advanced into in vivo target engagement and efficacy models. When tested in mice, compound 39 (AM-0466) demonstrated robust pharmacodynamic activity in a NaV1.7-dependent model of histamine-induced pruritus (itch) and additionally in a capsaicin-induced nociception model of pain without any confounding effect in open-field activity.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/metabolism , Quinolones/chemistry , Sulfonamides/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Analgesics/chemistry , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Capsaicin , Cell Line , Dogs , Histamine , Mice, Inbred C57BL , Molecular Docking Simulation , Pain/chemically induced , Pain/prevention & control , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Pruritus/chemically induced , Pruritus/prevention & control , Quinolones/administration & dosage , Quinolones/chemical synthesis , Quinolones/pharmacokinetics , Quinolones/pharmacology , Rats , Structure-Activity Relationship , Sulfonamides/administration & dosage , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/pharmacology
5.
J Pharmacol Exp Ther ; 356(1): 223-31, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26559125

ABSTRACT

Therapeutic agents that block the calcitonin gene-related peptide (CGRP) signaling pathway are a highly anticipated and promising new drug class for migraine therapy, especially after reports that small-molecule CGRP-receptor antagonists are efficacious for both acute migraine treatment and migraine prevention. Using XenoMouse technology, we successfully generated AMG 334, a fully human monoclonal antibody against the CGRP receptor. Here we show that AMG 334 competes with [(125)I]-CGRP binding to the human CGRP receptor, with a Ki of 0.02 nM. AMG 334 fully inhibited CGRP-stimulated cAMP production with an IC50 of 2.3 nM in cell-based functional assays (human CGRP receptor) and was 5000-fold more selective for the CGRP receptor than other human calcitonin family receptors, including adrenomedullin, calcitonin, and amylin receptors. The potency of AMG 334 at the cynomolgus monkey (cyno) CGRP receptor was similar to that at the human receptor, with an IC50 of 5.7 nM, but its potency at dog, rabbit, and rat receptors was significantly reduced (>5000-fold). Therefore, in vivo target coverage of AMG 334 was assessed in cynos using the capsaicin-induced increase in dermal blood flow model. AMG 334 dose-dependently prevented capsaicin-induced increases in dermal blood flow on days 2 and 4 postdosing. These results indicate AMG 334 is a potent, selective, full antagonist of the CGRP receptor and show in vivo dose-dependent target coverage in cynos. AMG 334 is currently in clinical development for the prevention of migraine.


Subject(s)
Antibodies, Monoclonal/pharmacology , Calcitonin Gene-Related Peptide Receptor Antagonists , Animals , Antibodies, Monoclonal, Humanized , Binding, Competitive/drug effects , Calcitonin Gene-Related Peptide/metabolism , Capsaicin/pharmacology , Cyclic AMP/biosynthesis , Dogs , Dose-Response Relationship, Drug , Humans , Macaca fascicularis , Mice , Migraine Disorders/prevention & control , Rabbits , Rats , Receptors, Calcitonin/drug effects , Receptors, Calcitonin/metabolism , Regional Blood Flow/drug effects , Skin/blood supply
6.
Mol Pain ; 8: 36, 2012 May 09.
Article in English | MEDLINE | ID: mdl-22571355

ABSTRACT

BACKGROUND: Transient receptor potential cation channel subfamily M member 8 (TRPM8) is activated by cold temperature in vitro and has been demonstrated to act as a 'cold temperature sensor' in vivo. Although it is known that agonists of this 'cold temperature sensor', such as menthol and icilin, cause a transient increase in body temperature (Tb), it is not known if TRPM8 plays a role in Tb regulation. Since TRPM8 has been considered as a potential target for chronic pain therapeutics, we have investigated the role of TRPM8 in Tb regulation. RESULTS: We characterized five chemically distinct compounds (AMG0635, AMG2850, AMG8788, AMG9678, and Compound 496) as potent and selective antagonists of TRPM8 and tested their effects on Tb in rats and mice implanted with radiotelemetry probes. All five antagonists used in the study caused a transient decrease in Tb (maximum decrease of 0.98°C). Since thermoregulation is a homeostatic process that maintains Tb about 37°C, we further evaluated whether repeated administration of an antagonist attenuated the decrease in Tb. Indeed, repeated daily administration of AMG9678 for four consecutive days showed a reduction in the magnitude of the Tb decrease Day 2 onwards. CONCLUSIONS: The data reported here demonstrate that TRPM8 channels play a role in Tb regulation. Further, a reduction of magnitude in Tb decrease after repeated dosing of an antagonist suggests that TRPM8's role in Tb maintenance may not pose an issue for developing TRPM8 antagonists as therapeutics.


Subject(s)
Body Temperature Regulation , Naphthyridines/metabolism , TRPM Cation Channels/metabolism , Animals , Body Temperature Regulation/drug effects , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Inhibitory Concentration 50 , Ion Channel Gating/drug effects , Male , Membrane Transport Modulators/chemistry , Membrane Transport Modulators/pharmacology , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , TRPM Cation Channels/antagonists & inhibitors
7.
J Pharmacol Exp Ther ; 326(1): 218-29, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18420600

ABSTRACT

Antagonists of the vanilloid receptor TRPV1 (transient receptor potential vanilloid type 1) have been reported to produce antihyperalgesic effects in animal models of pain. These antagonists, however, also caused concomitant hyperthermia in rodents, dogs, monkeys, and humans. Antagonist-induced hyperthermia was not observed in TRPV1 knockout mice, suggesting that the hyperthermic effect is exclusively mediated through TRPV1. Since antagonist-induced hyperthermia is considered a hurdle for developing TRPV1 antagonists as therapeutics, we investigated the possibility of eliminating hyperthermia while maintaining antihyperalgesia. Here, we report four potent and selective TRPV1 modulators with unique in vitro pharmacology profiles (profiles A through D) and their respective effects on body temperature. We found that profile C modulator, (R,E)-N-(2-hydroxy-2,3-dihydro-1H-inden-4-yl)-3-(2-(piperidin-1-yl)-4-(trifluoromethyl)phenyl)acrylamide (AMG8562), blocks capsaicin activation of TRPV1, does not affect heat activation of TRPV1, potentiates pH 5 activation of TRPV1 in vitro, and does not cause hyperthermia in vivo in rats. We further profiled AMG8562 in an on-target (agonist) challenge model, rodent pain models, and tested for its side effects. We show that AMG8562 significantly blocks capsaicin-induced flinching behavior, produces statistically significant efficacy in complete Freund's adjuvant- and skin incision-induced thermal hyperalgesia, and acetic acid-induced writhing models, with no profound effects on locomotor activity. Based on the data shown here, we conclude that it is feasible to modulate TRPV1 in a manner that does not cause hyperthermia while maintaining efficacy in rodent pain models.


Subject(s)
Acrylamides/chemistry , Acrylamides/pharmacology , Analgesics/pharmacology , Fever , Hyperalgesia/drug therapy , Piperidines/pharmacology , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/physiology , Acrylamides/pharmacokinetics , Animals , Body Temperature/drug effects , Body Temperature/physiology , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Fever/chemically induced , Fever/physiopathology , Hyperalgesia/physiopathology , Male , Mice , Pain Measurement/drug effects , Pain Measurement/methods , Piperidines/chemistry , Piperidines/pharmacokinetics , Rats , Rats, Sprague-Dawley
8.
J Pharmacol Exp Ther ; 323(1): 128-37, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17652633

ABSTRACT

Capsaicin, the active ingredient in some pain-relieving creams, is an agonist of a nonselective cation channel known as the transient receptor potential vanilloid type 1 (TRPV1). The pain-relieving mechanism of capsaicin includes desensitization of the channel, suggesting that TRPV1 antagonism may be a viable pain therapy approach. In agreement with the above notion, several TRPV1 antagonists have been reported to act as antihyperalgesics. Here, we report the in vitro and in vivo characterization of a novel and selective TRPV1 antagonist, N-(4-[6-(4-trifluoromethyl-phenyl)-pyrimidin-4-yloxy]-benzothiazol-2-yl)-acetamide I (AMG 517), and compare its pharmacology with that of a closely related analog, tert-butyl-2-(6-([2-(acetylamino)-1,3-benzothiazol-4-yl]oxy)pyrimidin-4-yl)-5-(trifluoromethyl)phenylcarbamate (AMG8163). Both AMG 517 and AMG8163 potently and completely antagonized capsaicin, proton, and heat activation of TRPV1 in vitro and blocked capsaicin-induced flinch in rats in vivo. To support initial clinical investigations, AMG 517 was evaluated in a comprehensive panel of toxicology studies that included in vivo assessments in rodents, dogs, and monkeys. The toxicology studies indicated that AMG 517 was generally well tolerated; however, transient increases in body temperature (hyperthermia) were observed in all species after AMG 517 dosing. To further investigate this effect, we tested and showed that the antipyretic, acetaminophen, suppressed the hyperthermia caused by TRPV1 blockade. We also showed that repeated administration of TRPV1 antagonists attenuated the hyperthermia response, whereas the efficacy in capsaicin-induced flinch model was maintained. In conclusion, these studies suggest that the transient hyperthermia elicited by TRPV1 blockade may be manageable in the development of TRPV1 antagonists as therapeutic agents. However, the impact of TRPV1 antagonist-induced hyperthermia on their clinical utility is still unknown.


Subject(s)
Analgesics, Non-Narcotic/therapeutic use , Benzothiazoles/therapeutic use , Fever/drug therapy , Pain/drug therapy , Pyrimidines/therapeutic use , TRPV Cation Channels/antagonists & inhibitors , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/chemistry , Analgesics, Non-Narcotic/pharmacology , Animals , Benzothiazoles/administration & dosage , Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Body Temperature/drug effects , CHO Cells , Capsaicin/pharmacology , Cricetinae , Cricetulus , Disease Models, Animal , Drug Administration Schedule , Drug Design , Female , Fever/metabolism , Freund's Adjuvant/pharmacology , Macaca fascicularis , Male , Molecular Structure , Pain/metabolism , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Telemetry
9.
J Pharmacol Exp Ther ; 322(1): 282-7, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17431136

ABSTRACT

A considerable body of evidence implicates endogenous nerve growth factor (NGF) in conditions in which pain is a prominent feature, including neuropathic pain. However, previous studies of NGF antagonism in animal models of neuropathic pain have examined only the prevention of hyperalgesia and allodynia after injury, whereas the more relevant issue is whether treatment can provide relief of established pain, particularly without tolerance. In the current work, we studied the effects of potent, neutralizing anti-NGF antibodies on the reversal of tactile allodynia and thermal hyperalgesia in established models of neuropathic and inflammatory pain in rats and mice. In the complete Freund's adjuvant-induced hind-paw inflammation, spinal nerve ligation and streptozotocin-induced neuropathic pain models, a single intraperitoneal injection of a polyclonal anti-NGF antibody reversed established tactile allodynia from approximately day 3 to day 7 after treatment. Effects on thermal hyperalgesia were variable with a significant effect observed only in the spinal nerve ligation model. In the mouse chronic constriction injury (CCI) model, a mouse monoclonal anti-NGF antibody reversed tactile allodynia when administered 2 weeks after surgery. Repeated administration of this antibody to CCI mice for 3 weeks produced a sustained reversal (days 4 to 21) of tactile allodynia that returned 5 days after the end of dosing. In conclusion, NGF seems to play a critical role in models of established neuropathic and inflammatory pain in both rats and mice, with no development of tolerance to antagonism. Antagonists of NGF, such as fully human monoclonal anti-NGF antibodies, may have therapeutic utility in analogous human pain conditions.


Subject(s)
Antibodies/therapeutic use , Disease Models, Animal , Hyperalgesia/drug therapy , Nerve Growth Factor/antagonists & inhibitors , Animals , Brain-Derived Neurotrophic Factor/physiology , Drug Tolerance , Humans , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factor/physiology , Rats , Rats, Sprague-Dawley
10.
J Neurosci ; 27(13): 3366-74, 2007 Mar 28.
Article in English | MEDLINE | ID: mdl-17392452

ABSTRACT

The vanilloid receptor TRPV1 (transient receptor potential vanilloid 1) is a cation channel that serves as a polymodal detector of pain-producing stimuli such as capsaicin, protons (pH <5.7), and heat. TRPV1 antagonists block pain behaviors in rodent models of inflammatory, neuropathic, and cancer pain, suggesting their utility as analgesics. Here, we report that TRPV1 antagonists representing various chemotypes cause an increase in body temperature (hyperthermia), identifying a potential issue for their clinical development. Peripheral restriction of antagonists did not eliminate hyperthermia, suggesting that the site of action is predominantly outside of the blood-brain barrier. Antagonists that are ineffective against proton activation also caused hyperthermia, indicating that blocking capsaicin and heat activation of TRPV1 is sufficient to produce hyperthermia. All TRPV1 antagonists evaluated here caused hyperthermia, suggesting that TRPV1 is tonically activated in vivo and that TRPV1 antagonism and hyperthermia are not separable. TRPV1 antagonists caused hyperthermia in multiple species (rats, dogs, and monkeys), demonstrating that TRPV1 function in thermoregulation is conserved from rodents to primates. Together, these results indicate that tonic TRPV1 activation regulates body temperature.


Subject(s)
Acrylamides/pharmacology , Body Temperature Regulation/physiology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Sulfonamides/pharmacology , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/physiology , Thiourea/analogs & derivatives , Animals , Benzothiazoles/pharmacology , Blood-Brain Barrier/metabolism , CHO Cells , Capsaicin , Cells, Cultured , Conserved Sequence , Cricetinae , Cricetulus , Dogs , Female , Fever/chemically induced , Fever/physiopathology , Humans , Hypothermia/chemically induced , Hypothermia/physiopathology , Macaca fascicularis , Male , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Species Specificity , Thiourea/pharmacology
11.
J Med Chem ; 49(12): 3719-42, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16759115

ABSTRACT

The vanilloid receptor-1 (VR1 or TRPV1) is a membrane-bound, nonselective cation channel that is predominantly expressed by peripheral neurons sensing painful stimuli. TRPV1 antagonists produce antihyperalgesic effects in animal models of inflammatory and neuropathic pain. Herein, we describe the synthesis and the structure-activity relationships of a series of 2-(4-pyridin-2-ylpiperazin-1-yl)-1H-benzo[d]imidazoles as novel TRPV1 antagonists. Compound 46ad was among the most potent analogues in this series. This compound was orally bioavailable in rats and was efficacious in blocking capsaicin-induced flinch in rats in a dose-dependent manner. Compound 46ad also reversed thermal hyperalgesia in a model of inflammatory pain, which was induced by complete Freund's adjuvant (CFA).


Subject(s)
Analgesics/chemical synthesis , Benzimidazoles/chemical synthesis , Piperazines/chemical synthesis , TRPV Cation Channels/antagonists & inhibitors , Administration, Oral , Analgesics/chemistry , Analgesics/pharmacology , Animals , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Biological Availability , CHO Cells , Calcium/metabolism , Capsaicin/pharmacology , Cricetinae , Cricetulus , Freund's Adjuvant , Hot Temperature , Hydrogen-Ion Concentration , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Male , Pain Measurement , Piperazines/chemistry , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship
12.
J Pharmacol Exp Ther ; 313(1): 474-84, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15615864

ABSTRACT

The vanilloid receptor 1 (VR1 or TRPV1) is a membrane-bound, nonselective cation channel expressed by peripheral sensory neurons. TRPV1 antagonists produce antihyperalgesic effects in animal models of inflammatory and neuropathic pain. Here, we describe the in vitro and in vivo pharmacology of a novel TRPV1 antagonist, AMG 9810, (E)-3-(4-t-butylphenyl)-N-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)acrylamide. AMG 9810 is a competitive antagonist of capsaicin activation (IC50 value for human TRPV1, 24.5 +/- 15.7 nM; rat TRPV1, 85.6 +/- 39.4 nM) and blocks all known modes of TRPV1 activation, including protons (IC50 value for rat TRPV1, 294 +/- 192 nM; human TRPV1, 92.7 +/- 72.8 nM), heat (IC50 value for rat TRPV1, 21 +/- 17 nM; human TRPV1, 15.8 +/- 10.8 nM), and endogenous ligands, such as anandamide, N-arachidonyl dopamine, and oleoyldopamine. AMG 9810 blocks capsaicin-evoked depolarization and calcitonin gene-related peptide release in cultures of rat dorsal root ganglion primary neurons. Screening of AMG 9810 against a panel of G protein-coupled receptors and ion channels indicated selectivity toward TRPV1. In vivo, AMG 9810 is effective at preventing capsaicin-induced eye wiping in a dose-dependent manner, and it reverses thermal and mechanical hyperalgesia in a model of inflammatory pain induced by intraplantar injection of complete Freund's adjuvant. At effective doses, AMG 9810 did not show any significant effects on motor function, as measured by open field locomotor activity and motor coordination tests. AMG 9810 is the first cinnamide TRPV1 antagonist reported to block capsaicin-induced eye wiping behavior and reverse hyperalgesia in an animal model of inflammatory pain.


Subject(s)
Acrylamides/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Hyperalgesia/drug therapy , Receptors, Drug/antagonists & inhibitors , Animals , Behavior, Animal/drug effects , CHO Cells , Calcitonin Gene-Related Peptide/metabolism , Capsaicin/antagonists & inhibitors , Cells, Cultured , Cricetinae , Freund's Adjuvant , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hot Temperature , Humans , Hyperalgesia/chemically induced , Inflammation/complications , Inflammation/pathology , Motor Activity/drug effects , Neurons/drug effects , Neurons/metabolism , Pain Measurement/drug effects , Patch-Clamp Techniques , Protons , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...