Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters








Publication year range
1.
Br J Pharmacol ; 181(20): 3993-4011, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38922847

ABSTRACT

BACKGROUND AND PURPOSE: Inhibitors of voltage-gated sodium channels (NaVs) are important anti-epileptic drugs, but the contribution of specific channel isoforms is unknown since available inhibitors are non-selective. We aimed to create novel, isoform selective inhibitors of Nav channels as a means of informing the development of improved antiseizure drugs. EXPERIMENTAL APPROACH: We created a series of compounds with diverse selectivity profiles enabling block of NaV1.6 alone or together with NaV1.2. These novel NaV inhibitors were evaluated for their ability to inhibit electrically evoked seizures in mice with a heterozygous gain-of-function mutation (N1768D/+) in Scn8a (encoding NaV1.6) and in wild-type mice. KEY RESULTS: Pharmacologic inhibition of NaV1.6 in Scn8aN1768D/+ mice prevented seizures evoked by a 6-Hz shock. Inhibitors were also effective in a direct current maximal electroshock seizure assay in wild-type mice. NaV1.6 inhibition correlated with efficacy in both models, even without inhibition of other CNS NaV isoforms. CONCLUSIONS AND IMPLICATIONS: Our data suggest NaV1.6 inhibition is a driver of efficacy for NaV inhibitor anti-seizure medicines. Sparing the NaV1.1 channels of inhibitory interneurons did not compromise efficacy. Selective NaV1.6 inhibitors may provide targeted therapies for human Scn8a developmental and epileptic encephalopathies and improved treatments for idiopathic epilepsies.


Subject(s)
NAV1.6 Voltage-Gated Sodium Channel , Seizures , Voltage-Gated Sodium Channel Blockers , Animals , NAV1.6 Voltage-Gated Sodium Channel/genetics , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Seizures/drug therapy , Mice , Voltage-Gated Sodium Channel Blockers/pharmacology , Male , Gain of Function Mutation , Anticonvulsants/pharmacology , Mice, Inbred C57BL
2.
ACS Chem Neurosci ; 15(6): 1169-1184, 2024 03 20.
Article in English | MEDLINE | ID: mdl-38359277

ABSTRACT

Voltage-gated sodium channel (NaV) inhibitors are used to treat neurological disorders of hyperexcitability such as epilepsy. These drugs act by attenuating neuronal action potential firing to reduce excitability in the brain. However, all currently available NaV-targeting antiseizure medications nonselectively inhibit the brain channels NaV1.1, NaV1.2, and NaV1.6, which potentially limits the efficacy and therapeutic safety margins of these drugs. Here, we report on XPC-7724 and XPC-5462, which represent a new class of small molecule NaV-targeting compounds. These compounds specifically target inhibition of the NaV1.6 and NaV1.2 channels, which are abundantly expressed in excitatory pyramidal neurons. They have a > 100-fold molecular selectivity against NaV1.1 channels, which are predominantly expressed in inhibitory neurons. Sparing NaV1.1 preserves the inhibitory activity in the brain. These compounds bind to and stabilize the inactivated state of the channels thereby reducing the activity of excitatory neurons. They have higher potency, with longer residency times and slower off-rates, than the clinically used antiseizure medications carbamazepine and phenytoin. The neuronal selectivity of these compounds is demonstrated in brain slices by inhibition of firing in cortical excitatory pyramidal neurons, without impacting fast spiking inhibitory interneurons. XPC-5462 also suppresses epileptiform activity in an ex vivo brain slice seizure model, whereas XPC-7224 does not, suggesting a possible requirement of Nav1.2 inhibition in 0-Mg2+- or 4-AP-induced brain slice seizure models. The profiles of these compounds will facilitate pharmacological dissection of the physiological roles of NaV1.2 and NaV1.6 in neurons and help define the role of specific channels in disease states. This unique selectivity profile provides a new approach to potentially treat disorders of neuronal hyperexcitability by selectively downregulating excitatory circuits.


Subject(s)
Epilepsy , Voltage-Gated Sodium Channels , Humans , Neurons/metabolism , Voltage-Gated Sodium Channels/metabolism , Epilepsy/metabolism , Brain/metabolism , Seizures/drug therapy , Seizures/metabolism , Action Potentials/physiology
3.
Elife ; 112022 03 02.
Article in English | MEDLINE | ID: mdl-35234610

ABSTRACT

NBI-921352 (formerly XEN901) is a novel sodium channel inhibitor designed to specifically target NaV1.6 channels. Such a molecule provides a precision-medicine approach to target SCN8A-related epilepsy syndromes (SCN8A-RES), where gain-of-function (GoF) mutations lead to excess NaV1.6 sodium current, or other indications where NaV1.6 mediated hyper-excitability contributes to disease (Gardella and Møller, 2019; Johannesen et al., 2019; Veeramah et al., 2012). NBI-921352 is a potent inhibitor of NaV1.6 (IC500.051 µM), with exquisite selectivity over other sodium channel isoforms (selectivity ratios of 756 X for NaV1.1, 134 X for NaV1.2, 276 X for NaV1.7, and >583 Xfor NaV1.3, NaV1.4, and NaV1.5). NBI-921352is a state-dependent inhibitor, preferentially inhibiting inactivatedchannels. The state dependence leads to potent stabilization of inactivation, inhibiting NaV1.6 currents, including resurgent and persistent NaV1.6 currents, while sparing the closed/rested channels. The isoform-selective profile of NBI-921352 led to a robust inhibition of action-potential firing in glutamatergic excitatory pyramidal neurons, while sparing fast-spiking inhibitory interneurons, where NaV1.1 predominates. Oral administration of NBI-921352 prevented electrically induced seizures in a Scn8a GoF mouse,as well as in wild-type mouse and ratseizure models. NBI-921352 was effective in preventing seizures at lower brain and plasma concentrations than commonly prescribed sodium channel inhibitor anti-seizure medicines (ASMs) carbamazepine, phenytoin, and lacosamide. NBI-921352 waswell tolerated at higher multiples of the effective plasma and brain concentrations than those ASMs. NBI-921352 is entering phase II proof-of-concept trials for the treatment of SCN8A-developmental epileptic encephalopathy (SCN8A-DEE) and adult focal-onset seizures.


Subject(s)
Epilepsy , NAV1.6 Voltage-Gated Sodium Channel , Animals , Gain of Function Mutation , Mice , Mutation , NAV1.6 Voltage-Gated Sodium Channel/genetics , Neurons/physiology , Rats , Sodium , Sodium Channel Blockers/pharmacology
4.
J Med Chem ; 64(6): 2953-2966, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33682420

ABSTRACT

Nav1.7 is an extensively investigated target for pain with a strong genetic link in humans, yet in spite of this effort, it remains challenging to identify efficacious, selective, and safe inhibitors. Here, we disclose the discovery and preclinical profile of GDC-0276 (1) and GDC-0310 (2), selective Nav1.7 inhibitors that have completed Phase 1 trials. Our initial search focused on close-in analogues to early compound 3. This resulted in the discovery of GDC-0276 (1), which possessed improved metabolic stability and an acceptable overall pharmacokinetics profile. To further derisk the predicted human pharmacokinetics and enable QD dosing, additional optimization of the scaffold was conducted, resulting in the discovery of a novel series of N-benzyl piperidine Nav1.7 inhibitors. Improvement of the metabolic stability by blocking the labile benzylic position led to the discovery of GDC-0310 (2), which possesses improved Nav selectivity and pharmacokinetic profile over 1.


Subject(s)
Azetidines/pharmacology , Benzamides/pharmacology , Drug Discovery , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Sulfonamides/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Animals , Azetidines/chemistry , Azetidines/pharmacokinetics , Benzamides/chemistry , Benzamides/pharmacokinetics , Cells, Cultured , HEK293 Cells , Humans , Piperidines/chemistry , Piperidines/pharmacokinetics , Piperidines/pharmacology , Rats, Sprague-Dawley , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/pharmacokinetics
5.
J Med Chem ; 62(21): 9618-9641, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31525968

ABSTRACT

Nonselective antagonists of voltage-gated sodium (NaV) channels have been long used for the treatment of epilepsies. The efficacy of these drugs is thought to be due to the block of sodium channels on excitatory neurons, primarily NaV1.6 and NaV1.2. However, these currently marketed drugs require high drug exposure and suffer from narrow therapeutic indices. Selective inhibition of NaV1.6, while sparing NaV1.1, is anticipated to provide a more effective and better tolerated treatment for epilepsies. In addition, block of NaV1.2 may complement the anticonvulsant activity of NaV1.6 inhibition. We discovered a novel series of aryl sulfonamides as CNS-penetrant, isoform-selective NaV1.6 inhibitors, which also displayed potent block of NaV1.2. Optimization focused on increasing selectivity over NaV1.1, improving metabolic stability, reducing active efflux, and addressing a pregnane X-receptor liability. We obtained compounds 30-32, which produced potent anticonvulsant activity in mouse seizure models, including a direct current maximal electroshock seizure assay.


Subject(s)
Amides/chemistry , Central Nervous System/metabolism , Epilepsy/drug therapy , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Sodium Channel Blockers/metabolism , Sodium Channel Blockers/pharmacology , Animals , Dogs , Hep G2 Cells , Humans , Madin Darby Canine Kidney Cells , Mice , Models, Molecular , NAV1.6 Voltage-Gated Sodium Channel/chemistry , Protein Domains , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/therapeutic use , Structure-Activity Relationship
6.
J Med Chem ; 62(2): 908-927, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30499663

ABSTRACT

Herein, we report the discovery and optimization of a series of orally bioavailable acyl sulfonamide NaV1.7 inhibitors that are selective for NaV1.7 over NaV1.5 and highly efficacious in in vivo models of pain and hNaV1.7 target engagement. An analysis of the physicochemical properties of literature NaV1.7 inhibitors suggested that acyl sulfonamides with high fsp3 could overcome some of the pharmacokinetic (PK) and efficacy challenges seen with existing series. Parallel library syntheses lead to the identification of analogue 7, which exhibited moderate potency against NaV1.7 and an acceptable PK profile in rodents, but relatively poor stability in human liver microsomes. Further, design strategy then focused on the optimization of potency against hNaV1.7 and improvement of human metabolic stability, utilizing induced fit docking in our previously disclosed X-ray cocrystal of the NaV1.7 voltage sensing domain. These investigations culminated in the discovery of tool compound 33, one of the most potent and efficacious NaV1.7 inhibitors reported to date.


Subject(s)
Analgesics/chemistry , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Sulfonamides/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Analgesics/metabolism , Analgesics/therapeutic use , Animals , Binding Sites , Drug Design , Half-Life , Humans , Male , Mice , Mice, Transgenic , Microsomes, Liver/metabolism , Molecular Docking Simulation , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pain/chemically induced , Pain/drug therapy , Pain/pathology , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Sulfonamides/metabolism , Sulfonamides/therapeutic use , Voltage-Gated Sodium Channel Blockers/metabolism , Voltage-Gated Sodium Channel Blockers/therapeutic use
7.
J Med Chem ; 61(11): 4810-4831, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29737846

ABSTRACT

The sodium channel NaV1.7 has emerged as a promising target for the treatment of pain based on strong genetic validation of its role in nociception. In recent years, a number of aryl and acyl sulfonamides have been reported as potent inhibitors of NaV1.7, with high selectivity over the cardiac isoform NaV1.5. Herein, we report on the discovery of a novel series of N-([1,2,4]triazolo[4,3- a]pyridin-3-yl)methanesulfonamides as selective NaV1.7 inhibitors. Starting with the crystal structure of an acyl sulfonamide, we rationalized that cyclization to form a fused heterocycle would improve physicochemical properties, in particular lipophilicity. Our design strategy focused on optimization of potency for block of NaV1.7 and human metabolic stability. Lead compounds 10, 13 (GNE-131), and 25 showed excellent potency, good in vitro metabolic stability, and low in vivo clearance in mouse, rat, and dog. Compound 13 also displayed excellent efficacy in a transgenic mouse model of induced pain.


Subject(s)
Drug Design , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pain/drug therapy , Sulfonamides/chemistry , Sulfonamides/pharmacology , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/pharmacology , Amino Acid Sequence , Animals , Dogs , Drug Stability , Humans , Kinetics , Mice , Molecular Conformation , Pain/metabolism , Rats , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/therapeutic use
8.
ACS Med Chem Lett ; 7(3): 277-82, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26985315

ABSTRACT

We report on a novel series of aryl sulfonamides that act as nanomolar potent, isoform-selective inhibitors of the human sodium channel hNaV1.7. The optimization of these inhibitors is described. We aimed to improve potency against hNaV1.7 while minimizing off-target safety concerns and generated compound 3. This agent displayed significant analgesic effects in rodent models of acute and inflammatory pain and demonstrated that binding to the voltage sensor domain 4 site of NaV1.7 leads to an analgesic effect in vivo. Our findings corroborate the importance of hNaV1.7 as a drug target for the treatment of pain.

9.
Science ; 350(6267): aac5464, 2015 Dec 18.
Article in English | MEDLINE | ID: mdl-26680203

ABSTRACT

Voltage-gated sodium (Nav) channels propagate action potentials in excitable cells. Accordingly, Nav channels are therapeutic targets for many cardiovascular and neurological disorders. Selective inhibitors have been challenging to design because the nine mammalian Nav channel isoforms share high sequence identity and remain recalcitrant to high-resolution structural studies. Targeting the human Nav1.7 channel involved in pain perception, we present a protein-engineering strategy that has allowed us to determine crystal structures of a novel receptor site in complex with isoform-selective antagonists. GX-936 and related inhibitors bind to the activated state of voltage-sensor domain IV (VSD4), where their anionic aryl sulfonamide warhead engages the fourth arginine gating charge on the S4 helix. By opposing VSD4 deactivation, these compounds inhibit Nav1.7 through a voltage-sensor trapping mechanism, likely by stabilizing inactivated states of the channel. Residues from the S2 and S3 helices are key determinants of isoform selectivity, and bound phospholipids implicate the membrane as a modulator of channel function and pharmacology. Our results help to elucidate the molecular basis of voltage sensing and establish structural blueprints to design selective Nav channel antagonists.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/chemistry , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Thiadiazoles/chemistry , Thiadiazoles/pharmacology , Amino Acid Sequence , Cell Membrane/chemistry , Crystallization/methods , Crystallography, X-Ray , DNA Mutational Analysis , Humans , Models, Molecular , Molecular Sequence Data , NAV1.7 Voltage-Gated Sodium Channel/genetics , Pain Perception/drug effects , Protein Engineering , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary
10.
Beilstein J Org Chem ; 10: 1848-77, 2014.
Article in English | MEDLINE | ID: mdl-25246946

ABSTRACT

A review of the synthesis of natural products and bioactive compounds adopting phosphonamide anion technology is presented highlighting the utility of phosphonamide reagents in stereocontrolled bond-forming reactions. Methodologies utilizing phosphonamide anions in asymmetric alkylations, Michael additions, olefinations, and cyclopropanations will be summarized, as well as an overview of the synthesis of the employed phosphonamide reagents.

11.
J Org Chem ; 75(16): 5601-18, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20704433

ABSTRACT

An enantioselective synthesis of the antifungal natural product (+)-ambruticin S has been accomplished starting with the readily available methyl alpha-d-glucopyranoside, (R)-Roche ester, and (S)-glycidol as chirons, which encompassed seven of the 10 stereogenic centers of the target molecule. The remaining three centers were set by a highly diastereoselective, asymmetric cyclopropanation employing a chiral, nonracemic phosphonamide reagent. Our strategy for the construction of the dihydropyran subunit involved a highly syn-selective Lewis acid catalyzed 6-endo-trig cyclization. Other key steps in the synthesis featured an epoxide opening with a dithiane anion, two efficient phosphonamide-anion based olefinations, and a late-stage C-glycosylation.


Subject(s)
Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Biological Factors/chemical synthesis , Biological Factors/chemistry , Cyclization , Glycosylation , Molecular Conformation , Pyrans/chemical synthesis , Pyrans/chemistry , Stereoisomerism
12.
Org Lett ; 12(14): 3172-5, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20565089

ABSTRACT

The first total synthesis of the antifungal polyketide jerangolid A has been accomplished. Starting with the readily available (R)-Roche ester and (S)-glycidol as chirons, the synthesis involved a highly syn-selective Lewis acid catalyzed 6-endo-trig cyclization for the construction of the dihydropyran subunit. The lactone segment was built through a tandem NaOMe conjugate addition-lactonization reaction, and further functionalized through a sequence consisting of iodination, I-Mg exchange, and hydroxymethylation. Other key steps in the synthesis featured a novel application of a phosphonamide-anion based olefination and a Julia-Kocienski reaction.


Subject(s)
Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Biological Products/chemical synthesis , Biological Products/chemistry , Catalysis , Pyrans/chemical synthesis , Pyrans/chemistry , Stereoisomerism
13.
Org Lett ; 8(14): 2985-8, 2006 Jul 06.
Article in English | MEDLINE | ID: mdl-16805533

ABSTRACT

[reaction: see text] The stereoselective synthesis of pyridinones was accomplished by the nucleophilic addition of Grignard reagents to a chiral pyridinium salt derived from 4-methoxypyridine. This methodology was applied to an expedient synthesis of (-)-barrenazine A and B. After N-functionalization and 1,4-reduction of the pyridinone system, the corresponding alpha-amino piperidinones readily undergo dimerization to give the hexahydrodipyridinopyrazine skeleton of the barrenazine alkaloids.


Subject(s)
Alkaloids/chemical synthesis , Heterocyclic Compounds, 3-Ring/chemical synthesis , Pyridones/chemical synthesis , Alkaloids/chemistry , Animals , Heterocyclic Compounds, 3-Ring/chemistry , Molecular Structure , Pyridinium Compounds/chemistry , Stereoisomerism , Urochordata/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL