U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Noebels JL, Avoli M, Rogawski MA, et al., editors. Jasper's Basic Mechanisms of the Epilepsies. 5th edition. New York: Oxford University Press; 2024. doi: 10.1093/med/9780197549469.003.0072

Cover of Jasper's Basic Mechanisms of the Epilepsies

Jasper's Basic Mechanisms of the Epilepsies. 5th edition.

Show details

Chapter 72Epileptogenic Channelopathies Guide Design of NBI-921352, a Highly Isoform-Selective Inhibitor of NaV1.6

, , , and .

Abstract

NBI-921352 is the first highly isoform selective inhibitor of NaV1.6 voltage-gated sodium channels (NaVs) to enter clinical development for epilepsy. Nonselective inhibitors of NaVs have long been a mainstay of epilepsy pharmacotherapy. Many such NaV inhibitors are available, but they are not mechanistically well differentiated. They all inhibit via the same highly conserved binding site, and they inhibit the distinct NaV isoforms equivalently. This broad NaV inhibition limits the utility of these drugs, as dose levels required to reduce seizures can cause tolerability issues in both the central nervous and cardiovascular systems. The rationale to develop NBI-921352 was to take a precision medicine approach to developing novel NaV inhibitors. This approach has been validated preclinically, both in scn8a gain-of-function mutant cell lines and in Scn8a gain-of-function mice. NBI-921352 should improve tolerability by stripping away effects mediated by inhibition of the NaVs of cardiac cells (NaV1.5), muscle cells (NaV1.4), and inhibitory interneurons (NaV1.1). Despite the selective nature of NBI-921352, the compound retains the ability to reduce neural excitability and increases seizure resistance in rodents. NBI-921352 is currently entering clinical trials to establish whether the improved properties observed in rodents translate to human epilepsy patients

Introduction

Drugs that inhibit voltage-gated sodium channels, such as phenytoin, carbamazepine, and lacosamide, are among the most widely used anticonvulsants. Many analogs of these compounds have been developed to progressively improve the pharmacodynamic properties and specificity for sodium channels. The mechanism of action of sodium channel inhibitors has been extensively reviewed, including an excellent summary in the fourth edition of this volume (Catterall, 2012). A limitation common to all of the currently used anticonvulsant sodium channel inhibitors is a relatively small therapeutic index. In general, the adverse effects seen with these compounds are a result of their lack of sodium channel selectivity. This is not unexpected because sodium channels are widely expressed in both excitatory and inhibitory neurons in the central nervous system (CNS) and play a key role in the activation of action potentials. The currently available sodium channel agents are non-isoform selective inhibitors and thus have similar potency for all sodium channel isoforms, including those in both excitatory and inhibitory circuits. All the agents produce state-dependent inhibition such that the inactivated states are stabilized, reducing the availability of channel opening during conditions that cause hyperexcitability. This profile favors inhibition of rapidly firing cells, as is encountered in epilepsy, but inhibitory interneurons can also fire rapidly. Ideally, one would strive to reduce the neuronal firing of the excitatory and not the inhibitory neurons. Moreover, the currently used sodium channel inhibitors have similar potency for both neuronal and muscle sodium channels. Inhibition of NaV1.4 and NaV1.5, the isoforms prevalent in skeletal and cardiac muscle, respectively, does not offer therapeutic benefit but does bring additional risk.

A strategy for enhancing the efficacy and therapeutic index of epileptic agents is to create sodium channel inhibitors that preferentially exist in excitatory neuronal circuits. Studies on channelopathies, especially from studies of severe myoclonic epilepsy of infancy, also known as SMEI or Dravet syndrome, add credence to this strategy. Three isoforms of voltage-gated sodium channels comprise the vast majority of those in the adult CNS: NaV1.1, NaV1.2, and NaV1.6. Electrophysiological and mouse genetic studies show that Dravet syndrome is caused by loss-of-function mutations in NaV1.1 and concomitant loss of excitability in inhibitory GABAergic neurons (Catterall, 2012). Thus, inhibition of NaV1.1 is likely counterproductive to antiseizure activity. A compound that selectively inhibits NaV1.6 and/or NaV1.2 and spares NaV1.1 and NaV1.5 could have superior efficacy and a better therapeutic index.

Another group of severe childhood epilepsy syndromes is attributed to de novo gain-of-function (GOF) mutations in SCN8A, the gene that codes for NaV1.6 (Veeramah et al., 2012; Meisler, 2019). As in Dravet syndrome, understanding of the channelopathy is derived from genetic studies in both humans and mice. The most severe of the SCN8A-related epilepsy syndromes (SCN8A-RES) is SCN8A developmental and epileptic encephalopathy (SCN8A-DEE) (Johannesen et al., 2019; Gardella and Moller, 2019). Since decreased NaV1.1 currents and increased NaV1.6 currents both cause seizure syndromes, it follows that a selective NaV1.6 inhibitor could rebalance the excitatory inhibitory balance and provide therapeutic benefit in epilepsy. We hypothesize that a selective inhibitor of NaV1.6 could provide a safer and more effective treatment for patients with SCN8A-RES and might also be efficacious in more common forms of epilepsy.

As shown in Figure 72–2, our earlier studies developing selective inhibitors of NaV1.7 served as a paradigm for targeting NaV1.6 and provided confidence that the appropriate selectivity among sodium channel isoforms might be achieved (Ahuja et al., 2015; Clairfeuille et al., 2020). The NaV inhibitors in use as antiseizure medicines bind to the pore domain of the channels, a site that is highly conserved among the isoforms. Time- and voltage-dependent inhibition can be also obtained by binding to an extracellular site on the domain IV voltage sensor (VSD4), and this site differs substantially among NaV1.6, NaV1.1, and NaV1.5. An extensive medicinal-chemistry effort focused on the VSD4 binding site produced NBI-921352 (formerly known as XEN901), a potent and isoform-selective inhibitor of NaV1.6 channels (Johnson et al., 2022).

Figure 72–2.. Targeting domain IV voltage sensor (VSD4) enables highly selective inhibitors.

Figure 72–2.

Targeting domain IV voltage sensor (VSD4) enables highly selective inhibitors. Therapeutically used NaV inhibitors bind in pore domain to a promiscuous, low-affinity binding site. Aryl sulfonamide inhibitors bind to a distinct extracellular binding site (more...)

We explored the properties of NBI-921352 in three preclinical rodent-seizure models. One model evaluated antiseizure activity in mice genetically engineered to carry a patient-identified heterozygous variant in the SCN8A gene coding for gain-of-function NaV1.6 channels (N1768D) that predispose the mice to seizures (Veeramah et al., 2012; Hammer et al., 2016; Wagnon et al., 2015). N1768D channels have impaired voltage-dependent inactivation gating that results in persistent sodium currents and enhanced resurgent currents (Veeramah et al., 2012). Because NaV1.6 channels are highly expressed in the neurons of the brain, and particularly so in excitatory pyramidal neurons, the increased Na+ flux leads to mice that are predisposed to seizures. Many, but not all, of the mice will develop spontaneous seizures between 6 weeks and 6 months after birth. In addition, the transgenic mice have a lower threshold for electrically induced seizures, and we evaluated the efficacy of NaV1.6 inhibitors to prevent seizures by stimuli that only provoke seizures in transgenic mice.

In addition to evaluating antiseizure activity in this genetically modified mouse, the selective NaV1.6 inhibitors were evaluated in the direct current maximal electroshock seizure (DC-MES) models in wild-type mice and rats, assays that have been used in the development of the currently used antiseizure medicine (ASM) NaV inhibitors. NBI-921352 is highly effective in these models and is a promising agent for treating both the childhood SCN8A-RES epilepsies as well as adult epilepsies with unknown etiologies (Johnson et al., 2022).

In this chapter, we will examine the rationale in support of creating NaV1.6 selective inhibitors for the treatment of epilepsy as well as preclinical data supporting this approach with our novel NaV1.6 selective inhibitor NBI-921352.

NaV Cellular and Subcellular Distribution

Central Nervous System

There are four NaV isoforms that predominate in the central nervous system, NaV1.1, NaV1.2, NaV1.3, and NaV1.6. Early in development, NaV1.2 and NaV1.3 are the most prevalent isoforms. NaV1.3 is largely embryonic, with high expression prenatally and perinatally. In the first year of life (or weeks in mice), NaV1.3 is largely replaced by NaV1.1. As shown in Figure 72–1, NaV1.1 is particularly important in inhibitory interneurons, where it supports action-potential firing and GABAergic signaling.

Figure 72–1.. NaV isoforms have diverse cellular and subcellular expression patterns.

Figure 72–1.

NaV isoforms have diverse cellular and subcellular expression patterns. Schematic showing NaV localization. NaV1.1 predominates in inhibitory interneurons. NaV1.2 and NaV1.6 are more prevalent in excitatory glutamatergic pyramidal neurons. NaV1.2 is (more...)

NaV1.2 is highly expressed throughout life but is particularly dominant at early stages of development. As shown in Figure 72–1, NaV1.6 expression is low at birth and increases in the first year of life, displacing NaV1.2 from the distal axon initial segments of many neurons as well as at the nodes of Ranvier (Sole and Tamkun, 2020). NaV1.2 remains the most abundant NaV in the dendrites and the proximal axon initial segment. This temporal and spatial differentiation of NaV isoform expression leads to diverse physiological roles as well as diverse roles in pathological conditions.

Peripheral Tissues

All of the NaV isoforms (NaV1.1–NaV1.9) are expressed in the periphery, but the relative contributions of NaV1.1, NaV1.2, and NaV1.6 are less well-understood in the periphery than in the CNS. As in the CNS, NaV1.3 is mostly expressed embryonically or perinatally in the periphery, though there is an extensive literature on re-expression of NaV1.3 after nerve injury, both peripherally and centrally (Samad et al., 2013; Hains et al., 2003; Yin et al., 2016). NaV1.4 is primarily expressed in skeletal muscle, where it is the most prevalent isoform. NaV1.5 is the NaV isoform most highly expressed in cardiac tissue. NaV1.7, NaV1.8, and NaV1.9 are largely restricted to peripheral neurons, particularly the nociceptor neurons of the dorsal root and trigeminal ganglia. For the purposes of epilepsy, there is no strong rationale to suggest that inhibition of peripheral sodium channels would be beneficial. Inhibition of NaVs in the skeletal or cardiac tissue creates obvious risks for motor and cardiovascular health.

Genetic Channelopathies Guide a Preferred Selectivity Profile for Antiseizure Medications

Peripheral NaVs

Many NaV associated ion channelopathies have been identified. The disorders linked to NaVs in the peripheral nervous system (PNS) are associated with pathologies consistent with their physiologic roles. NaV1.4 variants are associated with muscular dysfunction like periodic paralysis and myotonias (Sasaki et al., 1999; Jurkat-Rott et al., 2010; Cannon, 2018). NaV1.5 variants are associated with cardiac disfunction, such as long QT syndrome (gain of function) and Brugada syndrome (loss of function) (Roden et al., 1995; Chen et al., 1998; Campuzano et al., 2020). Mutations in NaV1.7, NaV1.8, or NaV1.9 have all been linked to pathogenic changes in pain sensitivity (Huang et al., 2017; Cox et al., 2006; Goldberg et al., 2007; Goodwin and McMahon, 2021). Both chronic pain syndromes, like inherited erythromelagia, paroxysmal extreme pain disorder, and impaired pain sensitivity, like congenital insensitivity to pain, can occur (Goldberg et al., 2012; Goodwin and McMahon, 2021; Alsaloum et al., 2020).

Because NaV1.4, NaV1.5, NaV 1.7, NaV1.8, and NaV1.9 are restricted to the periphery, inhibiting them is not expected to contribute to efficacy in epilepsy. Modulation of these channels can lead to adverse events, as suggested by the genetics and by drug-induced toxicity. Drugs that block NaV1.4, like nonselective NaV inhibitors, can cause motor control issues. Drugs that block NaV1.5, like class I antiarrhythmics, can cause cardiac arrhythmias, including drug-induced Brugada syndrome.

A recent FDA drug safety communication has applied a black-box warning to the non-isoform selective NaV inhibitor ASM Lamotrigine due to its inhibition of NaV1.5 and consequent risk of cardiac arrhythmias (FDA, 2021). Other ASM compounds in the class may also be required to perform postmarket studies to evaluate arrhythmia risks and are expected to carry similar liabilities. These compounds include Carbamazepine, Cenobamate, Eslicarbazepine, Fosphenytoin, Lacosamide, Oxcarbazepine, Phenytoin, Rufinamide, Topiramate, and Zonisamide. Other, ex-U.S. jurisdictions might also be expected to follow suit and increase scrutiny on the class of nonselective NaV inhibitors.

NaV inhibitors intended as local anesthetics for the treatment of trigeminal neuralgia and other pain syndromes, and class I cardiac antiarrhythmic drugs are often dose limited by CNS adverse events such as dizziness, sedation, and cognitive or motor impairment. These adverse events are likely caused by inhibition of CNS NaVs. Hence, the risk of non-isoform selective inhibitors extends to drugs targeting NaV isoforms in both peripheral and central compartments.

NaV1.1

Loss-of-function variants of NaV1.1 are known to impair inhibitory interneuron function and cause generalized epilepsy with seizures plus (GEFS+) and SCN1A-DEE (Dravet syndrome) (Claes et al., 2001; Gennaro et al., 2003; Escayg et al., 2000; Catterall et al., 2010). Missense variants that preserve some level of channel function tend to cause GEFS+. Null variants that disrupt production of the entire protein lead to the more extreme phenotype of Dravet syndrome. In some cases, missense mutations that severely disrupt gating, or do so in a dominant-negative fashion, can also cause Dravet syndrome. Nonselective NaV inhibitor ASMs are contraindicated for Dravet syndrome, since they have been found to exacerbate seizures. This is believed to be a consequence of inhibiting the already compromised inhibitory interneurons that depend on NaV1.1 to maintain efficient firing and balance excitatory inputs. Multiple high-quality reviews on the mechanisms and phenotypes of NaV1.1 variants are available (Catterall et al., 2010; Cheah et al., 2013; Mantegazza and Broccoli, 2019).

NaV1.6

NaV1.6 voltage-gated sodium channels are critical mediators of neural excitability (Royeck et al., 2008; Meisler, 2019). Mutations in the SCN8A gene result in disfunction of NaV1.6 sodium channels and have been linked to pathologic phenotypes associated with both loss- and gain-of-function variants. Mutations in mice that disrupt normal NaV1.6 function also result in abnormal phenotypes (Meisler, 2019; Veeramah et al., 2012; Burgess et al., 1995; Wagnon et al., 2015; Gardella and Moller, 2019; Johannesen et al., 2019). NaV1.6 channel variants can cause either gain or loss of function with respect to Na+ influx across neuronal membranes. Loss-of-function variants in humans usually lead to cognitive and developmental delays or autism, but most of these patients do not have seizures (Inglis et al., 2020; Liu et al., 2019). A few patients with loss-of-function variants have been found to have epilepsy, though those patients tend to develop seizures after the first year of life. In mice, loss-of-function variants of NaV1.6 cause motor impairment, but they actually increase resistance to seizure induction (Martin et al., 2007; Hawkins et al., 2011). Gain-of-function variants in human SCN8A cause early-onset SCN8A-related epilepsy syndromes (SCN8A-RES). The most severe of these epilepsy syndromes is SCN8A developmental and epileptic encephalopathy (SCN8A-DEE) (Gardella and Moller, 2019; Johannesen et al., 2019; Hammer et al., 2016). Most SCN8A-RES patients carry de novo heterozygous missense variants that lead to a gain of function of the NaV1.6 channel, but inherited and bi-allelic variants have been identified (Wengert et al., 2019; Gardella and Moller, 2019). SCN8A-DEE patients may be severely compromised in their cognitive and motor development and present with seizures early in life, usually in the first postnatal year. After seizure onset, patients begin to miss developmental milestones and to display additional comorbidities, including cognitive delay, motor dysfunction, hypotonia, and cortical blindness. SCN8A-DEE individuals are predisposed to early death, including sudden unexplained death in epilepsy (SUDEP). While SCN8A-RES patients often have treatment-resistant seizures, many can achieve seizure reduction, and sometimes seizure freedom, with ASMs that nonselectively inhibit voltage-gated sodium channels, like phenytoin and carbamazepine (Braakman et al., 2017; Boerma et al., 2016). SCN8A-RES patients may require doses that are higher than those prescribed for most epilepsy patients and, as a result, are more prone to drug-related adverse events (Boerma et al., 2016; Gardella and Moller, 2019). Even with high doses and multiple ASMs, many patients continue to have uncontrolled seizures and extensive comorbidities.

Designing a New Class of Isoform-Selective Sodium Channel Inhibitors

Based on the known risks of decreasing NaV currents through NaV1.1, NaV1.4, and NaV1.5, we set out to create a selective inhibitor of NaV1.6. We predicted that such a compound would be safer, and perhaps more efficacious, than nonselective ASMs, since the proconvulsant effect of inhibiting NaV1.1 would be avoided. This approach could provide better tolerated and more effective treatment for patients with SCN8A-RES and might also be efficacious in more common forms of epilepsy. An extensive medicinal-chemistry effort produced many compounds with diverse properties and selectivity profiles. NBI-921352 combines potent inhibition of NaV1.6, high selectivity against off-target NaVs, and favorable pharmaceutical properties. NBI-921352 is the first selective inhibitor of NaV1.6 channel currents to enter clinical development for epilepsy (Neurocrine., 2021; Johnson et al., 2022).

Most small-molecule inhibitors of sodium channels bind to the intracellular pore of the channel. Upon binding, they can impair Na+ flow through open channels directly, but they also stabilize nonconductive inactivated states of the channel. A fundamental challenge of this binding site is that it is well conserved across NaV isoforms and, hence, does not readily lend itself to isoform-selective inhibition.

Multiple drug discovery projects in the pharmaceutical industries have targeted selective NaV channels inhibition, usually NaV1.3, NaV1.7, or NaV1.8 for pain indications, and numerous molecules have been identified that bind with some selectivity. Some of these molecules have the desired antiepileptic selectivity profile for targeting NaV1.6 and NaV1.2, but sparing NaV1.1 and NaV1.5 (McCormack et al., 2013). These compounds also block NaV1.7, but that would not be expected to impact their performance for epilepsy. The selective profile of these NaV1.7 targeted molecules is made possible by the fact that they target a distinct binding site on the extracellular side of the channel voltage sensor, the domain IV voltage sensor (Ahuja et al., 2015). This site is more diverse across channel isoforms than the canonical binding site for sodium channel inhibitors. The biggest challenge for these NaV1.7 molecules as potential ASMs is that they were designed to be peripherally restricted (to improve tolerability) and, hence, are excluded from the brain. Thus, these molecules are trapped in the peripheral circulation where they cannot impact epilepsy.

We set out to target this domain IV voltage-sensor (VSD4) binding site to enable selectivity, but to do so with molecules that can cross the blood–brain barrier and target NaVs in the brain. After many iterations of medicinal chemistry, we created compounds that balance potency, isoform selectivity, and brain penetrance.

NBI-921352, the First Isoform-Selective Inhibitor of NaV1.6

As shown in Figure 72–3A, NBI-921352 potently inhibits human NaV1.6 channel currents with an inhibitory concentration 50% (IC50) of 0.0514 µM. Inhibition of other human NaV1.X isoforms requires >100-fold higher concentrations of NBI-921352. NBI-921352 was 756-fold selective for hNaV1.6 versus NaV1.1, 134-fold for NaV1.2, 276-fold for NaV1.7, and >583-fold for NaV1.3, NaV1.4, and NaV1.5.

Figure 72–3.. NBI-921352 is selective for NaV1.

Figure 72–3.

NBI-921352 is selective for NaV1.6 relative to other isoforms in both human (A) and mouse (B) channels. (C) NBI-921352 potently inhibits both resurgent and persistent currents in a patient-identified gain-of-function variant of NaV1.6, but it is a weak (more...)

We expressed point mutations in the putative binding site for our NaV1.6 inhibitors and defined the key residues for potency (see Fig 72–4; other data not shown). These studies suggested that the binding pocket for NBI-921352 in VSD IV is well conserved between the human and rodent orthologs and suggested that the potency would be similar among these species. We confirmed this by assessing the potency of NBI-921352 in the mouse NaV isoforms most highly expressed in the brain, NaV1.6, NaV1.1, and NaV1.2. As shown in Figure 72–3B, the potency and selectivity in mouse NaV channels closely paralleled that seen in the human orthologs with an IC50 of 0.058 µM for mNaV1.6. NBI-921352 was 709-fold selective for NaV1.6 versus NaV1.1, and 191-fold for NaV1.2. NBI-921352 potently inhibits both human and mouse NaV1.6 channels, and it does so at concentrations much lower than are needed to inhibit any of the other channel isoforms tested.

The species-independent potency and selectivity profile of NBI-921352 is in stark contrast to VSD IV binding compounds developed to target NaV1.7 and NaV1.3. Because of species-specific amino acid differences, compounds like PF-04856264 (hNaV1.7 selective) and ICA-121431 (hNaV1.3 and hNaV1.1 selective) exhibit strongly species-specific binding. This results in markedly different NaV isoform-selectivity profiles in humans versus rodents for those peripherally restricted compounds (McCormack et al., 2013). Species-selectivity issues can make preclinical mouse studies challenging. NBI-921352 is potent and NaV1.6 selective among both human and mouse isoforms and is, therefore, suited to both preclinical and human pharmacology.

Like Nonselective NaV Inhibitor ASMs That Bind the Pore Domain, NBI-921352 Is a State-Dependent Inhibitor

Most small-molecule inhibitors of NaV channels bind preferentially to open and or inactivated states (Bean et al., 1983; Courtney et al., 1978; Strichartz, 1976). State-dependent inhibition leads to higher affinity of the drug for the target channel under depolarized conditions, where the channel is more frequently in open or inactivated states—like when neurons are firing rapidly in a seizure. State dependence is a common feature of NaV inhibitor ASMs and has been suggested to improve the therapeutic margins for those compounds.

Supraphysiologic hyperpolarizations of the membrane potential (–120 mV) push most channels to reside in the resting (closed) state. Brief depolarizations to measure NaV1.6 current from –120 mV result in weak inhibition of the channels (IC50 of 36.3 µM). Holding the membrane potential more depolarized (–45 mV) promotes channels to transition to open and inactivated states. Brief hyperpolarizations allow rapid recovery from inactivation for channels that are not bound to drug followed by a short 20 ms test depolarization. NBI-921352 inhibits the reopening of inactivated channels much more efficiently than closed channels. As shown in Figure 72–3C, the IC50 of NBI-921352 for inactivated channels is 0.051 µM, demonstrating NBI-921352 is even more state dependent than currently available NaV ASMs, with a 750-fold preference for activated (open or inactivated) states relative to rested state closed channels.

NBI-921352 Inhibits Persistent and Resurgent Currents from Mutant NaV1.6 Channels

Elevated persistent and or resurgent currents are believed to underlie or contribute to the pathology of many sodium channel related pathologies, and NaV1.6 is a source of persistent and resurgent currents (Mason et al., 2019; Pan and Cummins, 2020; Potet et al., 2020; Tidball et al., 2020; Zaman et al., 2019). In most conditions, NaV channels inactivate rapidly and nearly completely after opening. The small persistent currents in healthy neurons have been proposed to be primarily carried by NaV1.6. Persistent currents can be larger in pathologic conditions (Lampl et al., 1998; Schwindt and Crill, 1980). The persistent currents exhibited by many GOF mutations in NaV1.6 that disrupt inactivation, including N1768D, are believed to be important contributors to their tendency to cause hyperexcitability and seizures in patients (Tidball et al., 2020; Wagnon et al., 2015). Thus, inhibition of persistent currents is a desirable feature. As shown in Figure 72–3C, NBI-921352 inhibits N1768D NaV1.6 persistent currents (measured as the non-inactivating current 10 ms after initiation of the depolarizing test pulse) with a similar potency as for open and inactivated wild-type NaV1.6 channels with an IC50 of 0.0593 µM.

Resurgent currents occur after repolarization following a strong depolarization as channels redistribute between closed, open, and inactivated states (Raman and Bean, 1997). These resurgent currents are enhanced in many SCN8A-RES variants and have also been suggested to drive pathologic hyperexcitability (Raman et al., 1997; Pan and Cummins, 2020). NBI-921352 also effectively inhibits resurgent currents from N1768D channels with an IC50 of 0.0373 µM.

NBI-921352 Inhibits Electrically Induced Seizures in Scn8a N1768D/+ Mice

A selective inhibitor of NaV1.6 should lend itself to the treatment of disease states caused by pathologic gain of function mutations of SCN8A. Hence, we examined the ability of NBI-921352 to inhibit electrically induced seizures in mice with a patient-identified gain-of-function variant in the Scn8a gene encoding NaV1.6. N1768D is a variant of NaV1.6 identified in the first-reported SCN8A-DEE patient (Veeramah et al., 2012). Subsequently, genetically modified mice bearing the same variant (Scn8aN1768D/+) were created and found to be seizure prone, producing a mouse model with a phenotype resembling that observed in SCN8A-DEE patients (Wagnon et al., 2015). To assess the ability of NBI-921352 to engage NaV1.6 channels in vivo, we designed a modified version of the 6Hz psychomotor seizure assay in Scn8aN1768D/+ mice (Barton et al., 2001, Focken et al., 2019). A low-level current stimulation evokes robust generalized tonic-clonic seizures (GTC) with hind-limb extension in Scn8aN1768D/+ mice, but not in their wild-type littermates.

As shown in Figure 72–5, oral administration of NBI-921352 prior to electrical stimulation prevented induction of GTC with hind-limb extension in Scn8aN1768D/+ mice in a concentration-dependent-manner.

Figure 72–5.. NBI-921352 increases seizure resistance in rodents.

Figure 72–5.

NBI-921352 increases seizure resistance in rodents. NBI-921352 reduces induced seizure incidence at concentrations more than 50-fold lower than those of three currently used NaV inhibitor ASMs (left). Similar brain concentrations of NBI-921352 are also (more...)

Figure 72–4.. Comparison of NBI-921352 potency on heterologously expressed human NaV1.

Figure 72–4.

Comparison of NBI-921352 potency on heterologously expressed human NaV1.6 and patient-identified variants of NaV1.6. Bars show the fold change in IC50 for the variant channel relative to WT (Variant IC50 / WT IC50). Values less than one mean that NBI-921352 (more...)

Concentration Dependence of NBI-921352 in Comparison to Common ASMs

Translating dose dependence between species is challenging, as there are many complications that make cross-species allometric predictions uncertain. Consequently, we use plasma concentration or, better yet, target tissue concentration as a means of comparing compound activity. To accomplish this, all animals are euthanized immediately after seizure assessment, and the concentration of the test compound is determined in the plasma and brain tissue from each animal tested. As shown in Figure 72–5, the average concentrations for each dose group were used to generate brain concentration versus efficacy relationships. The efficacy of NaV ASMs is driven by CNS exposure, and the tolerability of NaV inhibitor ASMs is generally limited by CNS signs. Therefore, we focus on brain concentrations. The brain EC50 (the concentration at which a half-maximal effect was observed) for preventing electrically induced seizures for NBI-921352 was 0.065 µM.

Currently available NaV inhibitor ASMs required higher brain concentrations to achieve efficacy in this model. The brain EC50s for carbamazepine, phenytoin, and lacosamide were 9.4 µM, 18 µM, and 3.3 µM, respectively. Lacosamide failed to provide full protection at the concentrations tested, and higher concentrations were not tolerated by the mice. Thus, NBI-921352 was 50-fold more potent than lacosamide, 144-fold more potent that carbamazepine, and 277-fold more potent than phenytoin. The improved potency of NBI-921352 in vivo is consistent with its greater potency for NaV1.6 channels in vitro.

NBI-921352 Inhibits Electrically Induced Seizures in Wild-Type Mice and Rats

NaV1.6 is an important mediator of neuronal excitability, even in animals without gain-of-function mutations, and selective inhibition of NaV1.6 has been suggested as a potential target for other epilepsies involving mutations, such as SCN8A-RES, the proline-rich transmembrane protein 2 related epilepsies (PRRT2-RES), and Dravet syndrome, as well as for more common idiopathic seizures (Fruscione et al., 2018). We wondered whether NBI-921352 might have broader application in epilepsy in other syndromes of neural hyperexcitability. To begin to explore this idea, we assessed NBI-921352 in a maximal electroshock assay induced by a direct-current electrical stimulus (DC-MES) in wild-type mice and rats. MES assays have been widely used to predict therapeutic efficacy of ASMs, including nonselective NaV inhibitor ASMs. Figure 72–5, right panel, shows that NBI-921352 protects wild-type rodents from DC-MES-induced seizures at brain concentrations that overlap those needed to protect mutant Scn8aN1768D/+ mice in the 6Hz assay. This suggests that NBI-921352 may be more broadly efficacious and may effectively prevent seizures, even in populations where no mutations have compromised NaV1.6 function (Johnson et al., 2022).

Efficacious Concentrations of NBI-921352 Are Well Separated from Concentrations That Provoke Behavioral Signs

The intent of creating a highly selective NaV1.6 antagonist was to make a better antiseizure medication. If a selective compound approach works, it should provide the efficacy of a classic, nonselective, sodium channel inhibitor drug, but prevent the adverse events caused by inhibiting other sodium channels and non-sodium channel targets. If sparing NaV1.1 and other off-target interactions does, in fact, reduce adverse events, then even higher receptor occupancy of NaV1.6 might be achievable, and this could further improve efficacy.

As shown in Figure 72–6, to evaluate our hypothesis, we compared the plasma concentrations required for seizure control (plasma EC50) with the minimal plasma concentration at which behavioral changes associated with the limits of tolerability were observed. We made this comparison both for NBI-921352 and for several widely used nonselective NaV inhibitor ASMs: carbamazepine, phenytoin, and lacosamide.

Figure 72–6.. Selective inhibition improves tolerability.

Figure 72–6.

Selective inhibition improves tolerability. Rat efficacy compared to acute tolerability for NBI-921352 is markedly higher than for other NaV inhibitor ASMs. Plasma concentration versus efficacy data is shown for the rat DC-MES assay for NBI-921352, carbamazepine, (more...)

NBI-921352 was well tolerated in these studies up to a plasma concentration of 71 µM. Dividing this concentration by the plasma EC50 of 0.15 µM in the rat DC-MES study results in a behavioral-signs concentration (BSC)/plasma EC50 ratio of 473-fold. The same calculation was repeated for the established ASMs. The minimal plasma concentrations provoking behavioral signs of intolerability for carbamazepine, phenytoin, and lacosamide were 110 µM, 11 µM, and 123 µM, respectively. Their plasma EC50s for seizure control were 30 µM, 4.5 µM, and 19.6 µM, respectively. Figure 75–7E shows BSC/plasma EC50 ratios for carbamazepine (3.7-fold), phenytoin (2.4-fold), and lacosamide (6.3-fold). These data indicate that increasing NaV1.6 selectivity can dramatically improve the tolerability of NaV inhibitors in mouse-seizure models.

In Vitro NaV1.6 Inhibition Predicts In Vivo Efficacy

Both NaV1.2 and NaV1.6 play critical roles in the firing of excitatory glutamatergic pyramidal neurons, despite differentiated profiles at the level of subcellular localization. NaV1.2 is more concentrated in the dendrites and proximal axon initial segment (AIS) (Spratt et al., 2019; Thompson et al., 2020). NaV1.6 is dominant in the distal AIS and nodes of Ranvier (Akin et al., 2015). NaV1.2 has been suggested to be an important mediator of backpropagating signals and integration of incoming signals, while NaV1.6 is more critical for action potentials propagating to downstream synapses (Ben-Shalom et al., 2017; Sanders et al., 2018).

De novo, it was unclear whether NaV1.2 selective compounds, NaV1.6 selective, or dual inhibitors would be the more fruitful approach. As shown in Figure 72–7, a comparison of efficacious brain concentrations in both Scn8aN1768D/+ mice and wild-type mice seizure assays to the in vitro NaV potency suggests that the potency on NaV1.6 channels is the best predictor of efficacy for these seizure models. NaV1.2 was loosely correlated with efficacy, and NaV1.1 potency appeared unrelated to efficacy.

Figure 72–7.. NaV1.

Figure 72–7.

NaV1.6 inhibition in vitro predicts efficacy in vivo. Each point represents a distinct compound tested in a mouse in vivo efficacy assay. Upper panels represent activity in the Scn8aN1768D/+ 6Hz assay. Lower panels display data from the mouse DC-MES (more...)

It is likely that the ideal selectivity profile may be impacted by the seizure model assessed or, in the clinic, the specific etiology of the seizure syndrome. For example, SCN2A-related epilepsy syndromes (SCN2A-RES) may benefit from NaV1.2 inhibition, since, in most cases, SCN2A-RES is associated with gain-of-function variants of NaV1.2. Nonetheless, our data suggest that inhibition of NaV1.6 in isolation is sufficient to impact neural excitability and to reduce the tendency for seizures in preclinical models.

Conclusions

The goal of ASMs is complete inhibition of seizures without adverse events. Dramatic reductions in seizure frequency are certainly welcome, but quality of life is greatly improved with complete suppression. Currently available NaV ASMs have dose-limiting side effects. Our studies indicate that NBI-921352 is very promising because it is highly isoform-selective for NaV1.6 and is well tolerated at brain concentrations well above those that suppress seizures.

A priori, it was unclear whether inhibition of multiple NaV isoforms was necessary to achieve seizure suppression. Our studies indicate that inhibition of NaV1.6 alone is sufficient, at least for the models that we have tested. The pharmacokinetic-pharmacodynamic (PK-PD) studies that we have conducted suggest that modest levels of inhibition of NaV1.6 are adequate to achieve seizure control and reestablish the balance between inhibitory and excitatory circuits in the CNS.

For SCN8A-RES patients, where disease is explicitly linked to excess NaV1.6 current, we anticipate that not only seizures but also other comorbidities that erode quality of life may be improved by a precision-medicine approach with NBI-92152. NBI-921352 is currently in development by Neurocrine Biosciences and is undergoing Phase II proof-of-concept trials for SCN8A-DEE (Neurocrine, 2019, 2021).

References

  1. Ahuja, S., Mukund, S., Deng, L., Khakh, K., Chang, E., Ho, H., Shriver, S., Young, C., Lin, S., Johnson, J. P., Jr., Wu, P., Li, J., Coons, M., Tam, C., Brillantes, B., Sampang, H., Mortara, K., Bowman, K. K., Clark, K. R., Estevez, A., Xie, Z., Verschoof, H., Grimwood, M., Dehnhardt, C., Andrez, J. C., Focken, T., Sutherlin, D. P., Safina, B. S., Starovasnik, M. A., Ortwine, D. F., Franke, Y., Cohen, C. J., Hackos, D. H., Koth, C. M. & Payandeh, J.  2015. Structural basis of Nav1.7 inhibition by an isoform-selective small-molecule antagonist.  Science, 350, aac5464. [PubMed: 26680203]
  2. Akin, E. J., Sole, L., Dib-Hajj, S. D., Waxman, S. G. & Tamkun, M. M.  2015. Preferential targeting of Nav1.6 voltage-gated Na+ channels to the axon initial segment during development.  PLoS One, 10, e0124397. [PMC free article: PMC4398423] [PubMed: 25874799]
  3. Alsaloum, M., Higerd, G. P., Effraim, P. R. & Waxman, S. G.  2020. Status of peripheral sodium channel blockers for non-addictive pain treatment.  Nat Rev Neurol, 16, 689–705. [PubMed: 33110213]
  4. Barton, M. E., Klein, B. D., Wolf, H. H. & White, H. S.  2001. Pharmacological characterization of the 6 Hz psychomotor seizure model of partial epilepsy.  Epilepsy Res, 47, 217–227. [PubMed: 11738929]
  5. Bean, B. P., Cohen, C. J. & Tsien, R. W.  1983. Lidocaine block of cardiac sodium channels.  J Gen Physiol, 81, 613–642. [PMC free article: PMC2216565] [PubMed: 6306139]
  6. Ben-Shalom, R., Keeshen, C. M., Berrios, K. N., An, J. Y., Sanders, S. J. & Bender, K. J.  2017. Opposing effects on NaV1.2 function underlie differences between SCN2A variants observed in individuals with autism spectrum disorder or infantile seizures.  Biol Psychiatry, 82, 224–232. [PMC free article: PMC5796785] [PubMed: 28256214]
  7. Boerma, R. S., Braun, K. P., Van De Broek, M. P., Van Berkestijn, F. M., Swinkels, M. E., Hagebeuk, E. O., Lindhout, D., Van Kempen, M., Boon, M., Nicolai, J., De Kovel, C. G., Brilstra, E. H. & Koeleman, B. P.  2016. Remarkable phenytoin sensitivity in four children with SCN8A-related epilepsy: A molecular neuropharmacological approach.  Neurotherapeutics, 13, 192–197. [PMC free article: PMC4720675] [PubMed: 26252990]
  8. Braakman, H. M., Verhoeven, J. S., Erasmus, C. E., Haaxma, C. A., Willemsen, M. H. & Schelhaas, H. J.  2017. Phenytoin as a last-resort treatment in SCN8A encephalopathy.  Epilepsia Open, 2, 343–344. [PMC free article: PMC5862112] [PubMed: 29588963]
  9. Burgess, D. L., Kohrman, D. C., Galt, J., Plummer, N. W., Jones, J. M., Spear, B. & Meisler, M. H.  1995. Mutation of a new sodium channel gene, Scn8a, in the mouse mutant “motor endplate disease.”  Nat Genet, 10, 461–465. [PubMed: 7670495]
  10. Campuzano, O., Sarquella-Brugada, G., Cesar, S., Arbelo, E., Brugada, J. & Brugada, R.  2020. Update on genetic basis of Brugada syndrome: monogenic, polygenic or oligogenic?  Int J Mol Sci, 21. [PMC free article: PMC7582739] [PubMed: 32998306]
  11. Cannon, S. C.  2018. Sodium channelopathies of skeletal muscle.  Handb Exp Pharmacol, 246, 309–330. [PMC free article: PMC5866235] [PubMed: 28939973]
  12. Catterall, W. A.  2012. Sodium Channel Mutations and Epilepsy. In: Th, Noebels, J. L., Avoli, M., Rogawski, M. A., Olsen, R. W. & Delgado-Escueta, A. V. (eds.) Jasper’s Basic Mechanisms of the Epilepsies. Bethesda (MD).
  13. Catterall, W. A., Kalume, F. & Oakley, J. C.  2010. NaV1.1 channels and epilepsy.  J Physiol, 588, 1849–1859. [PMC free article: PMC2901973] [PubMed: 20194124]
  14. Cheah, C. S., Westenbroek, R. E., Roden, W. H., Kalume, F., Oakley, J. C., Jansen, L. A. & Catterall, W. A.  2013. Correlations in timing of sodium channel expression, epilepsy, and sudden death in Dravet syndrome.  Channels (Austin), 7, 468–472. [PMC free article: PMC4042481] [PubMed: 23965409]
  15. Chen, Q., Kirsch, G. E., Zhang, D., Brugada, R., Brugada, J., Brugada, P., Potenza, D., Moya, A., Borggrefe, M., Breithardt, G., Ortiz-Lopez, R., Wang, Z., Antzelevitch, C., O’brien, R. E., Schulze-Bahr, E., Keating, M. T., Towbin, J. A. & Wang, Q.  1998. Genetic basis and molecular mechanism for idiopathic ventricular fibrillation.  Nature, 392, 293–296. [PubMed: 9521325]
  16. Claes, L., Del-Favero, J., Ceulemans, B., Lagae, L., Van Broeckhoven, C. & De Jonghe, P.  2001. De novo mutations in the sodium-channel gene SCN1A cause severe myoclonic epilepsy of infancy.  Am J Hum Genet, 68, 1327–1332. [PMC free article: PMC1226119] [PubMed: 11359211]
  17. Clairfeuille, T., Buchholz, K. R., Li, Q., Verschueren, E., Liu, P., Sangaraju, D., Park, S., Noland, C. L., Storek, K. M., Nickerson, N. N., Martin, L., Dela Vega, T., Miu, A., Reeder, J., Ruiz-Gonzalez, M., Swem, D., Han, G., Deponte, D. P., Hunter, M. S., Gati, C., Shahidi-Latham, S., Xu, M., Skelton, N., Sellers, B. D., Skippington, E., Sandoval, W., Hanan, E. J., Payandeh, J. & Rutherford, S. T.  2020. Structure of the essential inner membrane lipopolysaccharide-PbgA complex.  Nature, 584, 479–483. [PubMed: 32788728]
  18. Courtney, K. R., Kendig, J. J. & Cohen, E. N.  1978. The rates of interaction of local anesthetics with sodium channels in nerve.  J Pharmacol Exp Ther, 207, 594–604. [PubMed: 712641]
  19. Cox, J. J., Reimann, F., Nicholas, A. K., Thornton, G., Roberts, E., Springell, K., Karbani, G., Jafri, H., Mannan, J., Raashid, Y., Al-Gazali, L., Hamamy, H., Valente, E. M., Gorman, S., Williams, R., Mchale, D. P., Wood, J. N., Gribble, F. M. & Woods, C. G.  2006. An SCN9A channelopathy causes congenital inability to experience pain.  Nature, 444, 894–898. [PMC free article: PMC7212082] [PubMed: 17167479]
  20. Escayg, A., Macdonald, B. T., Meisler, M. H., Baulac, S., Huberfeld, G., An-Gourfinkel, I., Brice, A., Leguern, E., Moulard, B., Chaigne, D., Buresi, C. & Malafosse, A.  2000. Mutations of SCN1A, encoding a neuronal sodium channel, in two families with GEFS+2.  Nat Genet, 24, 343–345. [PubMed: 10742094]
  21. FDA. 2021. Drug Safety Communication: Studies show increased risk of heart rhythm problems with seizure and mental health medicine lamotrigine (Lamictal) in patients with heart disease.
  22. Focken, T., Burford, K., Grimwood, M. E., Zenova, A., Andrez, J. C., Gong, W., Wilson, M., Taron, M., Decker, S., Lofstrand, V., Chowdhury, S., Shuart, N., Lin, S., Goodchild, S. J., Young, C., Soriano, M., Tari, P. K., Waldbrook, M., Nelkenbrecher, K., Kwan, R., Lindgren, A., De Boer, G., Lee, S., Sojo, L., Devita, R. J., Cohen, C. J., Wesolowski, S. S., Johnson, J. P., Jr., Dehnhardt, C. M. & Empfield, J. R.  2019. Identification of CNS-Penetrant Aryl Sulfonamides as Isoform-Selective NaV1.6 Inhibitors with Efficacy in Mouse Models of Epilepsy.  J Med Chem, 62, 9618–9641. [PubMed: 31525968]
  23. Fruscione, F., Valente, P., Sterlini, B., Romei, A., Baldassari, S., Fadda, M., Prestigio, C., Giansante, G., Sartorelli, J., Rossi, P., Rubio, A., Gambardella, A., Nieus, T., Broccoli, V., Fassio, A., Baldelli, P., Corradi, A., Zara, F. & Benfenati, F.  2018. PRRT2 controls neuronal excitability by negatively modulating Na+ channel 1.2/1.6 activity.  Brain, 141, 1000–1016. [PMC free article: PMC5888929] [PubMed: 29554219]
  24. Gardella, E. & Moller, R. S.  2019. Phenotypic and genetic spectrum of SCN8A-related disorders, treatment options, and outcomes.  Epilepsia, 60 Suppl 3, S77–S85. [PubMed: 31904124]
  25. Gennaro, E., Veggiotti, P., Malacarne, M., Madia, F., Cecconi, M., Cardinali, S., Cassetti, A., Cecconi, I., Bertini, E., Bianchi, A., Gobbi, G. & Zara, F.  2003. Familial severe myoclonic epilepsy of infancy: truncation of Nav1.1 and genetic heterogeneity.  Epileptic Disord, 5, 21–25. [PubMed: 12773292]
  26. Goldberg, Y. P., Macfarlane, J., Macdonald, M. L., Thompson, J., Dube, M. P., Mattice, M., Fraser, R., Young, C., Hossain, S., Pape, T., Payne, B., Radomski, C., Donaldson, G., Ives, E., Cox, J., Younghusband, H. B., Green, R., Duff, A., Boltshauser, E., Grinspan, G. A., Dimon, J. H., Sibley, B. G., Andria, G., Toscano, E., Kerdraon, J., Bowsher, D., Pimstone, S. N., Samuels, M. E., Sherrington, R. & Hayden, M. R.  2007. Loss-of-function mutations in the Nav1.7 gene underlie congenital indifference to pain in multiple human populations.  Clin Genet, 71, 311–319. [PubMed: 17470132]
  27. Goldberg, Y. P., Pimstone, S. N., Namdari, R., Price, N., Cohen, C., Sherrington, R. P. & Hayden, M. R.  2012. Human Mendelian pain disorders: a key to discovery and validation of novel analgesics.  Clin Genet, 82, 367–373. [PubMed: 22845492]
  28. Goodwin, G. & Mcmahon, S. B.  2021. The physiological function of different voltage-gated sodium channels in pain.  Nat Rev Neurosci, 22, 263–274. [PubMed: 33782571]
  29. Hains, B. C., Klein, J. P., Saab, C. Y., Craner, M. J., Black, J. A. & Waxman, S. G.  2003. Upregulation of sodium channel Nav1.3 and functional involvement in neuronal hyperexcitability associated with central neuropathic pain after spinal cord injury.  J Neurosci, 23, 8881–8892. [PMC free article: PMC6740400] [PubMed: 14523090]
  30. Hammer, M. F., Wagnon, J. L., Mefford, H. C. & Meisler, M. H.  2016. SCN8A-Related Epilepsy with Encephalopathy.  In:  Adam, M. P., Ardinger, H. H., Pagon, R. A., Wallace, S. E., Bean, L. J. H., Stephens, K. & Amemiya, A. (eds.) GeneReviews((R)). Seattle (WA).
  31. Hawkins, N. A., Martin, M. S., Frankel, W. N., Kearney, J. A. & Escayg, A.  2011. Neuronal voltage-gated ion channels are genetic modifiers of generalized epilepsy with febrile seizures plus.  Neurobiol Dis, 41, 655–660. [PMC free article: PMC3035952] [PubMed: 21156207]
  32. Huang, J., Vanoye, C. G., Cutts, A., Goldberg, Y. P., Dib-Hajj, S. D., Cohen, C. J., Waxman, S. G. & George, A. L., Jr. 2017. Sodium channel NaV1.9 mutations associated with insensitivity to pain dampen neuronal excitability.  J Clin Invest, 127, 2805–2814. [PMC free article: PMC5490760] [PubMed: 28530638]
  33. Inglis, G. A. S., Wong, J. C., Butler, K. M., Thelin, J. T., Mistretta, O. C., Wu, X., Lin, X., English, A. W. & Escayg, A.  2020. Mutations in the Scn8a DIIS4 voltage sensor reveal new distinctions among hypomorphic and null Nav 1.6 sodium channels.  Genes Brain Behav, 19, e12612. [PubMed: 31605437]
  34. Johannesen, K. M., Gardella, E., Encinas, A. C., Lehesjoki, A. E., Linnankivi, T., Petersen, M. B., Lund, I. C. B., Blichfeldt, S., Miranda, M. J., Pal, D. K., Lascelles, K., Procopis, P., Orsini, A., Bonuccelli, A., Giacomini, T., Helbig, I., Fenger, C. D., Sisodiya, S. M., Hernandez-Hernandez, L., Krithika, S., Rumple, M., Masnada, S., Valente, M., Cereda, C., Giordano, L., Accorsi, P., Burki, S. E., Mancardi, M., Korff, C., Guerrini, R., Von Spiczak, S., Hoffman-Zacharska, D., Mazurczak, T., Coppola, A., Buono, S., Vecchi, M., Hammer, M. F., Varesio, C., Veggiotti, P., Lal, D., Brunger, T., Zara, F., Striano, P., Rubboli, G. & Moller, R. S.  2019. The spectrum of intermediate SCN8A-related epilepsy.  Epilepsia, 60, 830–844. [PubMed: 30968951]
  35. Johnson, J. P., Focken, T., Khakh, K., Tari, P. K., Dube, C., Goodchild, S. J., Andrez, J. C., Bankar, G., Bogucki, D., Burford, K., Chang, E., Chowdhury, S., Dean, R., De Boer, G., Decker, S., Dehnhardt, C., Feng, M., Gong, W., Grimwood, M., Hasan, A., Hussainkhel, A., Jia, Q., Lee, S., Li, J., Lin, S., Lindgren, A., Lofstrand, V., Mezeyova, J., Namdari, R., Nelkenbrecher, K., Shuart, N. G., Sojo, L., Sun, S., Taron, M., Waldbrook, M., Weeratunge, D., Wesolowski, S., Williams, A., Wilson, M., Xie, Z., Yoo, R., Young, C., Zenova, A., Zhang, W., Cutts, A. J., Sherrington, R. P., Pimstone, S. N., Winquist, R., Cohen, C. J. & Empfield, J. R.  2022. NBI-921352, a first-in-class, NaV1.6 selective, sodium channel inhibitor that prevents seizures in Scn8a gain-of-function mice, and wild-type mice and rats.  Elife, 11. [PMC free article: PMC8903829] [PubMed: 35234610]
  36. Jurkat-Rott, K., Holzherr, B., Fauler, M. & Lehmann-Horn, F.  2010. Sodium channelopathies of skeletal muscle result from gain or loss of function.  Pflugers Arch, 460, 239–248. [PMC free article: PMC2883924] [PubMed: 20237798]
  37. Lampl, I., Schwindt, P. & Crill, W.  1998. Reduction of cortical pyramidal neuron excitability by the action of phenytoin on persistent Na+ current.  J Pharmacol Exp Ther, 284, 228–237. [PubMed: 9435183]
  38. Liu, Y., Schubert, J., Sonnenberg, L., Helbig, K. L., Hoei-Hansen, C. E., Koko, M., Rannap, M., Lauxmann, S., Huq, M., Schneider, M. C., Johannesen, K. M., Kurlemann, G., Gardella, E., Becker, F., Weber, Y. G., Benda, J., Moller, R. S. & Lerche, H.  2019. Neuronal mechanisms of mutations in SCN8A causing epilepsy or intellectual disability.  Brain, 142, 376–390. [PubMed: 30615093]
  39. Mantegazza, M. & Broccoli, V.  2019. SCN1A/NaV 1.1 channelopathies: Mechanisms in expression systems, animal models, and human iPSC models.  Epilepsia, 60 Suppl 3, S25–S38. [PubMed: 31904127]
  40. Martin, M. S., Tang, B., Papale, L. A., Yu, F. H., Catterall, W. A. & Escayg, A.  2007. The voltage-gated sodium channel Scn8a is a genetic modifier of severe myoclonic epilepsy of infancy.  Hum Mol Genet, 16, 2892–2899. [PubMed: 17881658]
  41. Mason, E. R., Wu, F., Patel, R. R., Xiao, Y., Cannon, S. C. & Cummins, T. R.  2019. Resurgent and gating pore currents induced by de novo SCN2A epilepsy mutations.  eNeuro, 6. [PMC free article: PMC6795554] [PubMed: 31558572]
  42. Mccormack, K., Santos, S., Chapman, M. L., Krafte, D. S., Marron, B. E., West, C. W., Krambis, M. J., Antonio, B. M., Zellmer, S. G., Printzenhoff, D., Padilla, K. M., Lin, Z., Wagoner, P. K., Swain, N. A., Stupple, P. A., De Groot, M., Butt, R. P. & Castle, N. A.  2013. Voltage sensor interaction site for selective small molecule inhibitors of voltage-gated sodium channels.  Proc Natl Acad Sci U S A, 110, E2724–E2732. [PMC free article: PMC3718154] [PubMed: 23818614]
  43. Meisler, M. H.  2019. SCN8A encephalopathy: Mechanisms and models.  Epilepsia, 60 Suppl 3, S86–S91. [PMC free article: PMC6953611] [PubMed: 31904118]
  44. Neurocrine. 2019. Neurocrine biosciences and Xenon Pharmaceuticals announce agreement to develop first-in-class treatments for epilepsy.  Press Release.
  45. Neurocrine. 2021. Study to evaluate NBI-921352 as adjunctive therapy in Subjects With SCN8A Developmental and Epileptic Encephalopathy Syndrome (SCN8A-DEE).  ClinicalTrials.gov
  46. Pan, Y. & Cummins, T. R.  2020. Distinct functional alterations in SCN8A epilepsy mutant channels.  J Physiol, 598, 381–401. [PMC free article: PMC7216308] [PubMed: 31715021]
  47. Potet, F., Egecioglu, D. E., Burridge, P. W. & George, A. L., Jr. 2020. GS-967 and eleclazine block sodium channels in human induced pluripotent stem cell-derived cardiomyocytes.  Mol Pharmacol, 98, 540–547. [PubMed: 32938719]
  48. Raman, I. M. & Bean, B. P.  1997. Resurgent sodium current and action potential formation in dissociated cerebellar Purkinje neurons.  J Neurosci, 17, 4517–4526. [PMC free article: PMC6573347] [PubMed: 9169512]
  49. Raman, I. M., Sprunger, L. K., Meisler, M. H. & Bean, B. P.  1997. Altered subthreshold sodium currents and disrupted firing patterns in Purkinje neurons of Scn8a mutant mice.  Neuron, 19, 881–891. [PubMed: 9354334]
  50. Roden, D. M., George, A. L., Jr. & Bennett, P. B.  1995. Recent advances in understanding the molecular mechanisms of the long QT syndrome.  J Cardiovasc Electrophysiol, 6, 1023–1031. [PubMed: 8589871]
  51. Royeck, M., Horstmann, M. T., Remy, S., Reitze, M., Yaari, Y. & Beck, H.  2008. Role of axonal NaV1.6 sodium channels in action potential initiation of CA1 pyramidal neurons.  J Neurophysiol, 100, 2361–2380. [PubMed: 18650312]
  52. Samad, O. A., Tan, A. M., Cheng, X., Foster, E., Dib-Hajj, S. D. & Waxman, S. G.  2013. Virus-mediated shRNA knockdown of Na(v)1.3 in rat dorsal root ganglion attenuates nerve injury-induced neuropathic pain.  Mol Ther, 21, 49–56. [PMC free article: PMC3538302] [PubMed: 22910296]
  53. Sanders, S. J., Campbell, A. J., Cottrell, J. R., Moller, R. S., Wagner, F. F., Auldridge, A. L., Bernier, R. A., Catterall, W. A., Chung, W. K., Empfield, J. R., George, A. L., Jr., Hipp, J. F., Khwaja, O., Kiskinis, E., Lal, D., Malhotra, D., Millichap, J. J., Otis, T. S., Petrou, S., Pitt, G., Schust, L. F., Taylor, C. M., Tjernagel, J., Spiro, J. E. & Bender, K. J.  2018. Progress in understanding and treating SCN2A-mediated disorders.  Trends Neurosci, 41, 442–456. [PMC free article: PMC6015533] [PubMed: 29691040]
  54. Sasaki, R., Takano, H., Kamakura, K., Kaida, K., Hirata, A., Saito, M., Tanaka, H., Kuzuhara, S. & Tsuji, S.  1999. A novel mutation in the gene for the adult skeletal muscle sodium channel alpha-subunit (SCN4A) that causes paramyotonia congenita of von Eulenburg.  Arch Neurol, 56, 692–696. [PubMed: 10369308]
  55. Schwindt, P. & Crill, W.  1980. Role of a persistent inward current in motoneuron bursting during spinal seizures.  J Neurophysiol, 43, 1296–1318. [PubMed: 6246221]
  56. Sole, L. & Tamkun, M. M.  2020. Trafficking mechanisms underlying Nav channel subcellular localization in neurons.  Channels (Austin), 14, 1–17. [PMC free article: PMC7039628] [PubMed: 31841065]
  57. Spratt, P. W. E., Ben-Shalom, R., Keeshen, C. M., Burke, K. J., Jr., Clarkson, R. L., Sanders, S. J. & Bender, K. J.  2019. The autism-associated gene Scn2a contributes to dendritic excitability and synaptic function in the prefrontal cortex.  Neuron, 103, 673–685. [PMC free article: PMC7935470] [PubMed: 31230762]
  58. Strichartz, G.  1976. Molecular mechanisms of nerve block by local anesthetics.  Anesthesiology, 45, 421–441. [PubMed: 9844]
  59. Thompson, C. H., Ben-Shalom, R., Bender, K. J. & George, A. L.  2020. Alternative splicing potentiates dysfunction of early-onset epileptic encephalopathy SCN2A variants.  J Gen Physiol, 152. [PMC free article: PMC7054859] [PubMed: 31995133]
  60. Tidball, A. M., Lopez-Santiago, L. F., Yuan, Y., Glenn, T. W., Margolis, J. L., Clayton Walker, J., Kilbane, E. G., Miller, C. A., Martina Bebin, E., Scott Perry, M., Isom, L. L. & Parent, J. M.  2020. Variant-specific changes in persistent or resurgent sodium current in SCN8A-related epilepsy patient-derived neurons.  Brain, 143, 3025–3040. [PMC free article: PMC7780473] [PubMed: 32968789]
  61. Veeramah, K. R., O’brien, J. E., Meisler, M. H., Cheng, X., Dib-Hajj, S. D., Waxman, S. G., Talwar, D., Girirajan, S., Eichler, E. E., Restifo, L. L., Erickson, R. P. & Hammer, M. F.  2012. De novo pathogenic SCN8A mutation identified by whole-genome sequencing of a family quartet affected by infantile epileptic encephalopathy and SUDEP.  Am J Hum Genet, 90, 502–510. [PMC free article: PMC3309181] [PubMed: 22365152]
  62. Wagnon, J. L., Korn, M. J., Parent, R., Tarpey, T. A., Jones, J. M., Hammer, M. F., Murphy, G. G., Parent, J. M. & Meisler, M. H.  2015. Convulsive seizures and SUDEP in a mouse model of SCN8A epileptic encephalopathy.  Hum Mol Genet, 24, 506–515. [PMC free article: PMC4275076] [PubMed: 25227913]
  63. Wengert, E. R., Tronhjem, C. E., Wagnon, J. L., Johannesen, K. M., Petit, H., Krey, I., Saga, A. U., Panchal, P. S., Strohm, S. M., Lange, J., Kamphausen, S. B., Rubboli, G., Lemke, J. R., Gardella, E., Patel, M. K., Meisler, M. H. & Moller, R. S.  2019. Biallelic inherited SCN8A variants, a rare cause of SCN8A-related developmental and epileptic encephalopathy.  Epilepsia, 60, 2277–2285. [PMC free article: PMC6842408] [PubMed: 31625145]
  64. Yin, R., Liu, D., Chhoa, M., Li, C. M., Luo, Y., Zhang, M., Lehto, S. G., Immke, D. C. & Moyer, B. D.  2016. Voltage-gated sodium channel function and expression in injured and uninjured rat dorsal root ganglia neurons.  Int J Neurosci, 126, 182–192. [PubMed: 25562420]
  65. Zaman, T., Abou Tayoun, A. & Goldberg, E. M.  2019. A single-center SCN8A-related epilepsy cohort: clinical, genetic, and physiologic characterization.  Ann Clin Transl Neurol, 6, 1445–1455. [PMC free article: PMC6689675] [PubMed: 31402610]
Copyright Notice

This is an open access publication, available online and distributed under the terms of a Creative Commons Attribution-Non Commercial-No Derivatives 4.0 International licence (CC BY-NC-ND 4.0), a copy of which is available at https://creativecommons.org/licenses/by-nc-nd/4.0/. Subject to this license, all rights are reserved.

Bookshelf ID: NBK609897PMID: 39637216DOI: 10.1093/med/9780197549469.003.0072

Views

  • PubReader
  • Print View
  • Cite this Page
  • PDF version of this title (283M)

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...