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










Publication year range
1.
Br J Pharmacol ; 2024 Jun 24.
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.

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(24): 17777-17794, 2021 12 23.
Article in English | MEDLINE | ID: mdl-34871500

ABSTRACT

In our efforts to identify novel small molecule inhibitors for the treatment of adrenoleukodystrophy (ALD), we conducted a high-throughput radiometric screen for inhibitors of elongation of very long chain fatty acid 1 (ELOVL1) enzyme. We developed a series of highly potent, central nervous system (CNS)-penetrant pyrimidine ether-based compounds with favorable pharmacokinetics culminating in compound 22. Compound 22 is a selective inhibitor of ELOVL1, reducing C26:0 VLCFA synthesis in ALD patient fibroblasts and lymphocytes in vitro. Compound 22 reduced C26:0 lysophosphatidyl choline (LPC), a subtype of VLCFA, in the blood of ATP binding cassette transporter D1 (ABCD1) KO mice, a murine model of ALD to near wild-type levels. Compound 22 is a low-molecular-weight, potent ELOVL1 inhibitor that may serve as a useful tool for exploring therapeutic approaches to the treatment of ALD.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Fatty Acid Elongases/antagonists & inhibitors , Pyrimidines/pharmacology , Administration, Oral , Adrenoleukodystrophy/drug therapy , Animals , Biological Availability , Dogs , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Ethers/chemistry , HEK293 Cells , Humans , Macaca fascicularis , Mice , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Rats
5.
Bioorg Med Chem Lett ; 45: 128133, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34044121

ABSTRACT

We describe the synthesis and biological evaluation of a series of novel aryl sulfonamides that exhibit potent inhibition of NaV1.5. Unlike local anesthetics that are currently used for treatment of Long QT Syndrome 3 (LQT-3), the most potent compound (-)-6 in this series shows high selectivity over hERG and other cardiac ion channels and has a low brain to plasma ratio to minimize CNS side effects. Compound (-)-6 is also effective inshortening prolonged action potential durations (APDs) in a pharmacological model of LQT-3 syndrome in pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). Unlike most aryl sulfonamide NaV inhibitors that bind to the channel voltage sensors, these NaV1.5 inhibitors bind to the local anesthetic binding site in the central pore of the channel.


Subject(s)
NAV1.5 Voltage-Gated Sodium Channel/metabolism , Sulfonamides/pharmacology , Dose-Response Relationship, Drug , Humans , Molecular Structure , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
6.
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
7.
Trends Neurosci ; 41(7): 442-456, 2018 07.
Article in English | MEDLINE | ID: mdl-29691040

ABSTRACT

Advances in gene discovery for neurodevelopmental disorders have identified SCN2A dysfunction as a leading cause of infantile seizures, autism spectrum disorder, and intellectual disability. SCN2A encodes the neuronal sodium channel NaV1.2. Functional assays demonstrate strong correlation between genotype and phenotype. This insight can help guide therapeutic decisions and raises the possibility that ligands that selectively enhance or diminish channel function may improve symptoms. The well-defined function of sodium channels makes SCN2A an important test case for investigating the neurobiology of neurodevelopmental disorders more generally. Here, we discuss the progress made, through the concerted efforts of a diverse group of academic and industry scientists as well as policy advocates, in understanding and treating SCN2A-related disorders.


Subject(s)
NAV1.2 Voltage-Gated Sodium Channel/genetics , NAV1.2 Voltage-Gated Sodium Channel/metabolism , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/metabolism , Animals , Humans , Neurodevelopmental Disorders/drug therapy
8.
IDrugs ; 13(12): 869-73, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21154145

ABSTRACT

Rising expenditure in pharmaceutical R&D has not been matched by increased productivity. There is an urgent need to solve the current high levels of pipeline attrition. Changing the current failed model of drug discovery and development, in which high numbers of candidate drugs are produced and high attrition is accepted, is essential. A different model is needed, in which the focus shifts to identifying better-quality candidate drugs that allow scientifically robust testing of disease and targets in humans. Lowering the risks of compound-based attrition in small-molecule drug discovery and development (ie, addressing toxicity, specificity, potency, duration and exposure) is achievable by improved control of physical properties and by setting more demanding candidate criteria. Separating the key scientific experiment--proof-of-concept clinical trials in humans--from commercial development imperatives is a necessary step for the industry.


Subject(s)
Drug Discovery/methods , Drug Industry , Animals , Drug Discovery/economics , Drug Evaluation, Preclinical/economics , Drug Evaluation, Preclinical/methods , Drug Industry/economics , Drug Industry/methods , Efficiency, Organizational , Humans , Models, Organizational , Pharmacokinetics , Research Design
9.
Bioorg Med Chem Lett ; 20(24): 7312-6, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21055935

ABSTRACT

We determined the experimental solubility of CNS marketed drugs. Of the 98 drugs measured, greater than 90% had solubility >10 µM in pH 7.4 buffer. Only seven drugs had solubility <10 µM. Using these data, we established a solubility criterion to support CNS discovery. The implication of poor solubility with potential safety concerns and undesirable side effects are discussed.


Subject(s)
Central Nervous System Agents/chemistry , Pharmaceutical Preparations/chemistry , Drug Evaluation, Preclinical , Hydrogen-Ion Concentration , Solubility
10.
J Med Chem ; 50(13): 3113-31, 2007 Jun 28.
Article in English | MEDLINE | ID: mdl-17542571

ABSTRACT

A series of 7-chloro-2,3-dihydro-2-[1-(pyridinyl)alkyl]-pyridazino[4,5-b]quinoline-1,4,10(5H)-triones were synthesized and found to have potent activity at the glycine site of the NMDA receptor. In some cases, these compounds possessed poor aqueous solubility that may have contributed to poor rat oral bioavailability. Subsequently, compounds have been identified with improved aqueous solubility and oral bioavailability. Several of these compounds were examined in a rat chronic constrictive injury (CCI) model of neuropathic pain and found to have potent activity when dosed orally.


Subject(s)
Analgesics/chemical synthesis , Pain/drug therapy , Peripheral Nervous System Diseases/drug therapy , Pyridazines/chemical synthesis , Quinolines/chemical synthesis , Receptors, Glycine/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Administration, Oral , Alkynes/chemical synthesis , Alkynes/chemistry , Alkynes/pharmacology , Analgesics/chemistry , Analgesics/pharmacology , Animals , Brain/metabolism , Chronic Disease , Constriction, Pathologic/complications , Male , Pain/etiology , Peripheral Nervous System Diseases/etiology , Pyridazines/chemistry , Pyridazines/pharmacology , Pyridines/chemical synthesis , Pyridines/chemistry , Pyridines/pharmacology , Quinolines/chemistry , Quinolines/pharmacology , Radioligand Assay , Rats , Rats, Sprague-Dawley , Sciatic Nerve/pathology , Structure-Activity Relationship
11.
Appl Spectrosc ; 58(1): 41-6, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14727719

ABSTRACT

Integration of a Raman probe into a commercially available microwave synthesizer has demonstrated unprecedented utility in understanding chemical processes within the rapidly emerging field of microwave-assisted organic synthesis. The real-time spectral feedback afforded by this system has facilitated analysis of reaction mechanisms, reactive intermediates, and reaction kinetics via optical sampling through the sidewall of the sealed reaction vial within the microwave chamber. The feasibility, attributes, and limitations of the system are illustrated using amine coupling and Knoevenagel coupling example reactions. In addition to the reported analyses, this system provided the safety of remote sensing, adequate sensitivity, ease of alignment, and optimized "depth of field" for analysis of solutions with solids content.


Subject(s)
Fiber Optic Technology/instrumentation , Microchemistry/methods , Microwaves , Online Systems , Organic Chemicals/chemistry , Organic Chemicals/radiation effects , Spectrum Analysis, Raman/instrumentation , Spectrum Analysis, Raman/methods , Electromagnetic Fields , Equipment Design , Equipment Failure Analysis , Feasibility Studies , Organic Chemicals/analysis , Organic Chemicals/chemical synthesis , Temperature
12.
Bioorg Med Chem Lett ; 13(20): 3553-6, 2003 Oct 20.
Article in English | MEDLINE | ID: mdl-14505669

ABSTRACT

Several members of the 7-chloro-2,3-dihydro-2-[1-(pyridinyl)alkyl]-pyridazino[4,5-b]quinoline-1,4,10(5H)-triones (2) have been identified as being potent and selective NMDA glycine-site antagonists. Increasing size of the alkyl substituent on the alpha-carbon led to a progressive decrease in binding affinity. Some of these analogues possess improved drug-like properties such as cellular permeability, solubility and oral absorption.


Subject(s)
Glycine/chemistry , Pyridazines/chemical synthesis , Quinolines/chemical synthesis , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Pyridazines/chemistry , Pyridazines/pharmacology , Quinolines/chemistry , Quinolines/pharmacology , Receptors, N-Methyl-D-Aspartate/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...