Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
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.
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
3.
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
4.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...