Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Apr 1.
Published in final edited form as: Epilepsia. 2024 Feb 12;65(4):833–845. doi: 10.1111/epi.17884

Current state of the epilepsy drug and device pipeline

Samuel W Terman 1, Laura Kirkpatrick 2,3, Lisa F Akiyama 4, Wadih Baajour 5, Deniz Atilgan 5, Maria Kristina C Dorotan 6, Hyoung Won Choi 7, Jacqueline A French 8
PMCID: PMC11018510  NIHMSID: NIHMS1959981  PMID: 38345387

Abstract

The field of epilepsy has undergone substantial advances as we develop novel drugs and devices. Yet, considerable challenges remain in developing broadly effective, well-tolerated treatments, but also precision treatments for rare epilepsies and seizure monitoring devices. We summarize major recent and ongoing innovations in diagnostic and therapeutic products presented at the seventeenth Epilepsy Therapies and Diagnostics Development conference, which occurred May 31st to June 2nd, 2023, in Aventura, Florida. Therapeutics under development are targeting genetics, ion channels and other neurotransmitters, and many other potentially first-in-class interventions such as stem cells, glycogen metabolism, cholesterol, the gut microbiome, and novel modalities for delivering electrical neuromodulation.

Keywords: Epilepsy, antiseizure medications, devices

Introduction

The need remains for innovation in epilepsy diagnostics and treatments. One-third of people with epilepsy may not have seizures controlled on antiseizure medications (ASMs).1 Thus, developing increasingly effective and tolerated therapeutics remains critical. Additionally, to study new therapeutics, rigorous modalities are needed for seizure tracking.

This manuscript describes promising, ongoing efforts in therapeutics and device development presented at the seventeenth Epilepsy Therapies and Diagnostics Development conference in May-June 2023. We describe many therapeutics in progress, such as targeting refractory seizures or else promoting precision medicine via potentially disease-modifying approaches. We review therapeutics pertaining to a wide variety of mechanisms of action (e.g., ion channels, neurotransmitters, genetics, and other) in addition to promising devices for seizure treatment and monitoring. These efforts seek to develop more effective, selective, better-tolerated ASMs, including novel routes of administration, targeting a wide array of disease states from rare epilepsies to status epilepticus to more generally drug-resistant epilepsy.

Therapeutics

Ion channels

Voltage-dependent potassium channels

Potassium channels have emerged as a promising target. The first potassium channel opener, ezogabine (retigabine), demonstrated efficacy2 leading to approval in 2011. However, warnings emerged in 2013 regarding skin and fundoscopic pigment changes3 and the drug was withdrawn in 2017 due to limited use. Three next-generation candidates targeting Kv7 are currently under development.

XEN1101 (Xenon Pharmaceuticals) is in the most advanced stage among next-generation potassium channel openers. It has demonstrated high potency at Kv7 channels in preclinical and in vitro/in vivo data, surpassing ezogabine. A recent double-blinded phase IIB study in focal epilepsy (N=325, median baseline 13–17 seizures/month, failed 6 ASMs) was encouraging.4 The high-dose arm of XEN1101 reduced monthly seizure counts by 53% compared to 18% in the placebo arm (p<0.001). Efficacy was seen as early as the first week at every dose, and further advantages include once-daily dosing with no titration. Adverse effects included dizziness (25%) and somnolence (16%) and rare urinary retention (N=2) and weight gain (N=7), but no tissue discoloration thus far (XEN1101 is chemically incapable of forming the phenazinium dimers implicated in ezogabine-related pigmentary abnormalities). Phase III studies enrolling adults taking 1–3 ASMs with focal (X-TOLE2 and X-TOLE3) or primary generalized (X-ACKT) epilepsy are underway. A phase II study in Major Depressive Disorder (X-NOVA) will complete before 2024.

Similarly, Biohaven is developing BHV-7000.5,6 Preclinical data has shown that BHV-7000 hyperpolarizes neurons and increases action potential thresholds. Furthermore, BHV-7000 has chemical modifications compared with ezogabine intended to avoid causing dimers causing tissue discoloration. The compound was also intended to be as selective as possible for the potassium channel, minimizing GABAergic adverse effects. The drug is currently in a phase I randomized double-blinded placebo-controlled SAD/MAD study. Among 61 healthy adults treated at several doses for 15 days, the most common adverse effect was headache (N=7), with no dose-limiting toxicity and no somnolence or dizziness. Phase I studies are nearing completion, with Phase II-III trials planned to start by the end of 2023.

ETX-123 (Eliem Therapeutics) is a potent Kv7.2/3 channel opener with high selectivity and efficacy in preclinical studies. ETX-123 exhibits high potency at Kv7.2/3 channels, per EC50 values that are orders of magnitude more effective than ezogabine (ezogabine: 2 μM: ETX-123: 0.0007–0.002 μM).7 In vivo testing using the maximal electroshock seizure (MES) rat model has also demonstrated remarkable efficacy, with 2 mg/kg doses providing 50% protection, and 8 and 14 mg/kg doses offering complete protection from seizures. Tolerability assessments, such as the Rotarod test, revealed a favorable safety profile for ETX-123. Some concern has existed regarding urinary retention (Kv-7.4 subunit related), though this has not borne out when tested in a human bladder contractility assay, with EC50 values for muscle relaxation 700-fold higher than typical anticonvulsant plasma concentrations, unlike ezogabine (only 4-fold higher). With such promising preclinical data, ETX-123 awaits further investigation through additional in vivo pharmacology studies before proceeding to human trials.

Voltage-dependent sodium channels

Sodium channel blockade is a canonical ASM mechanism, though innovations continue, expanding targets and populations.

One problem is that currently available sodium channel blockers are nonselective, inhibiting voltage-gated sodium channel isoforms in myocardial, skeletal, and cerebral tissue.8 NaV1.5 inhibition risks arrhythmogenesis given it produces the rising phase of the cardiac action potential,9 and Nav1.4 inhibition may alter skeletal muscle excitability.1012 Within the nervous system, NaV1.1 inhibition can aggravate certain syndromes (e.g. Dravet syndrome).13

Neurocrine Biosciences, Inc. is developing NBI-921352, a selective Nav1.6 sodium channel inhibitor. Study NBI-921352-FOS2021 is a phase II randomized, double-blind, placebo-controlled study that enrolled 101 patients aged 18–65, to investigate the safety, tolerability, pharmacokinetics, and efficacy of NBI-921352 as adjunctive therapy in focal seizures. NBI-921352-DEE2012 is a phase II randomized, double-blind, placebo-controlled study that will seek to enroll 52 patients aged 2–21, to investigate NBI-921352 as adjunctive therapy for SCN8A developmental and epileptic encephalopathy (DEE).

Lacosamide is another sodium channel blocker, which enhances slow inactivation of sodium channels. While it is already approved for patients at least one month old, data are sparse for neonates. SP0968 (UCB) is a phase II/III, multicenter, open-label, randomized study, comparing lacosamide with standard of care. Eligible neonates will be at least 34 weeks of gestational age who have any confirmed electrographic seizure ≥2 minutes or ≥3 electrographic seizures and have previously received either phenobarbital, levetiracetam, or midazolam, weigh at least 2.3 kg, and have not previously received other sodium channel blockers.

Neurotransmitters

Gamma-aminobutyric acid (GABA)

OV329 (Ovid Therapeutics) is a next-generation GABA aminotransferase inhibitor in development for treating rare epilepsies. Its potency is two hundred times greater than vigabatrin, which could allow use at lower doses to avoid visual field deficits seen with vigabatrin. OV329 has demonstrated antiseizure activity in six of eight mouse and rat models thus far. Combined with safety data in rats and dogs, this suggests that OV329 may offer an improved benefit risk profile compared with vigabatrin. For example, OV329 significantly reduced seizures in a corneal kindled mouse model in a dose-dependent manner, and hippocampal paroxysmal discharges were reduced by 46% after a single oral dose in a mesial temporal lobe epilepsy mouse model (unpublished, courtesy of Ovid Therapeutics). It is currently in a Phase I Single Ascending Dose/Multiple Ascending Dose (SAD/MAD) study with no significant adverse effects thus far. Further results are expected in 2024.

Inhaled alprazolam administered via the Staccato® breath-actuated device aims for rapid systemic exposure for seizure termination. A phase IIb randomized placebo-controlled study (N=116) recently tested its ability to abort long or repetitive seizures in an inpatient setting.14 Participants were randomized 1:1:1 to double-blinded treatment of a single seizure event with one dose of Staccato alprazolam 1 mg, 2 mg, or placebo. Both 1 and 2 mg doses demonstrated efficacy in rapidly terminating seizures (~66% responder rate in both drug arms versus 43% placebo responder rate; p=0.04). Both doses were also generally well-tolerated, the most common adverse effects being cough and somnolence (both 15%). While the initial responder rate was the same with 1 and 2 mg doses, the seizure recurrence rate was lower with 2 mg. A multicenter, randomized placebo-controlled study (N=250) remains underway to test the efficacy and safety in participants at least 12 years old with stereotypical generalized or focal prolonged seizures, beyond the inpatient setting.

The neuroactive steroid ganaxolone (Marinus Pharmaceuticals) acts as a positive allosteric modulator of GABAA receptors.15 It is FDA-approved for CDKL5 deficiency disorder in patients 2 years and older, based on a phase III study in 100 patients.16 That study found a 31% versus 7% median reduction in major motor seizures (p<0.01) with maintenance of response over a 2-year open-label phase. The odds ratio for improvement in the Clinical Global Impression scale was 2.6 and the 50% responder rate was 24% for ganaxolone versus 10% placebo (p=0.06). Most non-seizure neurodevelopmental outcomes were unchanged. An exploratory analysis on reduction in seizure intensity and severity favored ganaxolone [OR (95%CI) = 2.56 (1.20,5.45)]. Ongoing work includes testing ganaxolone in other DEEs (Lennox-Gastaut syndrome in phase I, TSC in phase III). Additionally, an intravenous formulation is being evaluated in two placebo-controlled trials in refractory status epilepticus.17

While temporal lobectomy is effective for appropriate candidates, many patients remain reluctant or are not candidates for resection,18 and resection and other invasive treatments cannot guarantee cure and may require indwelling hardware. NRTX-1001 (Neurona Therapeutics) is an investigational therapy for drug-resistant unilateral mesial temporal lobe epilepsy consisting of a single MRI-guided injection of GABAergic human stem cell-derived inhibitory interneurons into the hippocampus.19 Immunosuppression is required for one year. NRTX-1001 is currently undergoing a Phase I/II clinical trial, with a primary endpoint of 1-year adverse events. Secondary endpoints include 1-year reduction in seizure frequency and responder rates, seizure severity, quality of life, neuropsychological outcomes, imaging biomarkers, and ASM dose reduction. The first participant, with data available through 9 months post-transplant, experienced no serious adverse events and a 95% reduction in seizures with stable neurocognitive scores at 6 and 9 months. The second participant, with data available up to 6 months post-transplant, has experienced no serious adverse events and a similarly excellent 94% reduction in seizures. Neurocognitive scores were stable or improved at 6 months, excluding a one-point decline in the Rey Auditory Verbal Learning Test. Recruitment remains ongoing at 11 sites.

Synaptic vesicle glycoprotein 2A (SV2A)

Currently, there are few approved monotherapies for absence epilepsy.20 While levetiracetam is often used in generalized epilepsies, there may be advantages of brivaracetam due to its ten-fold higher affinity for the SV2A binding site. A 2007 subject-blinded placebo-controlled study (N=18) in patients with photosensitive epilepsy found that even a single dose of brivaracetam (10, 20, 40, or 80 mg) suppressed photoparoxysmal EEG responses.21 Therefore, EXPAND (UCB, N01269, started 7/2021, expected completion 2024) is a randomized, dose-finding and confirmatory, double-blinded, placebo-controlled, parallel-group, multicenter phase II/III trial evaluating the efficacy, safety, and tolerability of brivaracetam monotherapy in patients 2–25 years old with childhood or juvenile absence epilepsy. The trial consists of dose-selection and futility assessment stages, followed by an optimal-dose stage after interim analysis. This two-stage trial design will seek to investigate two potentially efficacious brivaracetam doses.22

Glutamate receptors

Radiprodil is a negative allosteric modulator that binds to the N-methyl-D-aspartate (NMDA) receptor NR2B (GluN2B) subunit.23 As GRIN genes encode GluN subunits of the NMDA receptor, radiprodil has the potential to become a precision therapy for GRIN-related disorders caused by gain-of-function mutations by reducing overactive NMDA receptors. In previously completed clinical studies, radiprodil has demonstrated favorable safety, tolerability, and pharmacokinetic profiles in animal models and adults. The 12 treatment-emergent adverse events (TEAE) reported by 8/10 healthy adults in a phase I study were all reported to be mild and temporary. The most common TEAEs were dizziness (40%), nausea (20%) and somnolence (20%).24 Additionally, spasms responded favorably in three infants with prednisolone- and vigabatrin-resistant infantile spasms; one patient achieved sustained seizure freedom and the other two experienced a reduction in seizure clusters.25 GRIN Therapeutics’ phase Ib radiprodil study is currently enrolling patients 6 months to 12 years old with gain-of-function GRIN variants in Europe to further assess safety, tolerability, pharmacokinetics, and efficacy as measured by impact on seizures, behavioral symptoms, and caregiver burden.

Basimglurant (NOE-101, Noema Pharma) is a negative allosteric modulator of mGluR5 to reduce glutamatergic function.26 In addition, basimglurant may have another mechanism of action – normalizing aberrant protein synthesis via activation of the MEK/ERK pathway. Attention has therefore recently focused on its potential in reducing mTOR-dependent protein synthesis in Tuberous Sclerosis Complex (TSC). Chronic treatment with an mGluR5 modulator in a TSC mouse model (n=6) decreased seizure frequency (p=0.046) and total seizure time (p=0.044) compared with placebo (n=5).26 Noema’s Galene global Phase IIb trial is enrolling pediatric and adult patients with TSC-associated seizures. Existing human trials in adults and adolescents with depression and Fragile X further support development for use in epilepsy.27 Nearly two-thirds (122 out of 183) of participants with Fragile X Syndrome in the FragXis phase II trial experienced at least one (mostly gastrointestinal or psychiatric) TEAE, eight of whom discontinued the drug and the remainder resolved during the study.27 This compound is also being developed for trigeminal neuralgia pain.

Serotonin

Fenfluramine’s (ZX008) antiseizure properties are proposed to be secondary to serotonin release, agonistic action at 5-HT1D, 5-HT2A and 5-HT2C receptors, and positive modulation of sigma-1 receptors.28 Currently, fenfluramine is approved for treating convulsive seizures in Dravet syndrome patients Recently, Devinsky et al29 demonstrated its efficacy and safety in six pediatric patients with cyclin-dependent kinase like-5 (CDKL5) deficiency disorder, particularly in those patients with generalized tonic-clonic seizures, with a median of 90% reduction in seizure frequency. The ongoing phase III GEMZ study is part of the FAiRE (Fenfluramine Assessment in Rare Epilepsy; UCB) program investigating efficacy and safety of fenfluramine in pediatric and adult patients with CDKL5 deficiency disorder-related refractory epilepsy across approximately 70 study sites globally.

LP352 (Longboard Pharmaceuticals) also acts at the serotonin receptor but is a selective 5-HT2C superagonist. The absence of action at 5H2A/B is felt to avoid side effects such as cardiac valve thickening. Like other serotonin agonists, LP352 is being developed for the treatment of DEEs. A Phase I open-label study (LP352 102; N=20) assessing the pharmacokinetics and pharmacodynamics of LP352 found that plasma and CSF concentrations increased in dose-dependent manners, confirming crossing of the blood brain barrier. Furthermore, LP352 affected quantitative EEG activity. The double-blinded Phase Ib/IIa PACIFIC study is currently evaluating LP352 for treatment of multiple DEEs, across 30 sites with three different doses, data expected early 2024.

Acetylcholine

Acetylcholine is not a typical target of existing ASMs. However, SPN-817 (formerly BIS-001ER) is a synthetic version of Huperazine A, which is an alkaloid acetylcholinesterase inhibitor from the Chinese clubmoss Huperzia serrata. It is proposed to reduce seizures by modulating the excitatory-inhibitory balance in neurons and reducing inflammation in glia via increased GABA release.30 The Supernus SPN-817 Phase I study completed in 2022 demonstrated safety and good tolerability with elimination of adverse effects with slower dose titration in healthy individuals. In the subsequent phase Ib/II pilot study enrolling adults with refractory focal seizures, two of three participants were seizure-free at 24 months, with mild/transient TEAEs including nausea, insomnia, unsteadiness, and nystagmus.31 Now, its phase IIa study with an optional open-label extension is investigating effects on inflammatory biomarkers, cognitive performance, and EEG.

Genetic

As genetic causes of epilepsies become increasingly understood, opportunities exist for increasing therapeutic precision.

STK-001 is an ASO engineered by Stoke Therapeutics, Inc. for Dravet syndrome. STK-001 is part of a technology called Targeted Augmentation of Nuclear Gene Output to treat haploinsufficiencies like Dravet syndrome. In Dravet syndrome caused by SCN1A loss-of-function mutations, an alternative exon to pre-mRNA leads to a non-productive transcript that is degraded by nonsense-mediated mRNA decay resulting in reduced Nav1.1 production.32 STK- 001 blocks the incorporation of this alternative exon to increase productive SCN1A mRNA expression. Initial data from 2022 suggested a 55% seizure frequency reduction in six patients at the 45 mg dose. Subsequent data in 2023 were less robust (N=16, only an 18% reduction), though the 70 mg dose has demonstrated median reductions up to 80–90%. Additionally, in vitro safety concerns surrounded off-target effects such as thrombocytopenia or complement activation,33 which have not yet been observed clinically. The compound is currently undergoing a phase I/IIa open-label study for continued efficacy and safety information.

AMT-260 (UniQure, Inc.) is an Adeno-Associated Virus (AAV) miRNA-based gene therapy targeting GRIK2 (glutamate receptor ionotropic kainate 2) for refractory temporal lobe epilepsy. The GRIK2 gene encodes a GluK2 subunit of the kainate receptor. Aberrant GluK2-containing kainate receptors play a key role in the pathogenesis of refractory temporal lobe epilepsy.34 In preclinical studies, AMT-260 has shown a dose-dependent decrease in GRIK2 gene expression in proportion to the expression of the miRNAs in the mouse hippocampus and achieved up to 75% seizure reduction. AMT-260 is injected into affected brain tissue; injection accuracy testing in primates has demonstrated post-administration transduction within the hippocampus. UniQure is planning to conduct a Phase I/II trial in adults with unilateral refractory mesial temporal lobe epilepsy in 2023. In this trial, AMT-260 will be administrated to the hippocampus under MRI-guidance following the same trajectory used for laser interstitial thermal therapy.

A third example of genetic innovation includes CAP-002 (Capsida Biotherapeutics), an AAV vector-based product for syntaxin-binding protein 1 (STXBP1) mutations. STXBP1 interacts with numerous synaptic proteins in the brain playing an essential role in synaptic vesicle fusion and neurotransmitter release.35 Genetic epilepsy caused by STXBP1 mutation has severe disease manifestations including early-onset refractory seizures, cognitive dysfunction, autistic behaviors, and gait disturbances. Drug development is vital, as seizures are typically refractory36 and there are no currently available disease-modifying treatments. Preclinical mouse data have demonstrated that intravenous administration of a capsid carrying STXBP1 cargo produced dose-dependent improvement in spike-wave discharges and cognitive (e.g., object recognition, fear response) and motor (e.g., limb clasping) deficits in adult mice, lasting at least 12 months post therapeutic intervention. Intravenous delivery of the compound resulted in widespread transduction in neocortex, caudate, putamen and thalamus, while avoiding uptake by the liver, in non-human primates. Thus, this drug remains a promising candidate in preclinical development.

Finally, NMT.001 represents another RNA-based therapy undergoing preclinical development (NEUmiRNA Therapeutics). It inhibits miR-134, which is a brain-specific miRNA implicated in controlling neuronal microstructure. miR-134 appears upregulated in experimental models of status epilepticus.37 Silencing miR-134 exerts prolonged seizure-suppression and neuroprotection in animal models.38 In a proof-of-concept screening test using intra-amygdala kainate mouse models of mesial temporal lobe epilepsy, treatment with NMT.001 dramatically reduced lorazepam-resistant status epilepticus seizures

Notably, diagnostic codes do not currently exist for many rare epilepsies. With increasing availability of such treatments, further efforts advocating for unique International Classification of Diseases codes to distinguish rare epilepsies could improve access and spur research.

Other/multiple targets

Intracerebroventricular (ICV) valproate

Another avenue for innovation includes using conventional ASMs in less conventional ways. ICV drug delivery with a device implanted under the scalp already has a track record in scenarios such as treating infections, pain, and neoplasms.39 It has several appealing features. In contrast to intrathecal delivery, ICV delivery avoids the need for repeated lumbar punctures. And in contrast to oral delivery, ICV delivery may reduce systemic toxicity. Valproate is an example with known systemic toxicity and teratogenicity. ICV administration of valproate could potentially avoid these pitfalls related to absorption into the bloodstream by producing a high drug concentration in the brain, but a low serum concentration. In a proof-of-concept study performed by Cerebral Therapeutics, five patients with refractory mesial temporal lobe epilepsy, who had previously not responded to oral valproate, received an implanted device delivering ICV valproate. Four subjects responded with >50% seizure reduction at the highest tested dose of 160 mg/day. All five subjects reported significant quality of life improvement.40 This observation could eventually open up a completely new path for drug-refractory patients.

Cholesterol inhibitor

Soticlestat (Takeda) is another first-in-class investigational compound. It inhibits cholesterol-24-hydroxylase, an enzyme primarily expressed in the brain that reduces 24-hydroxcholesterol (24HC).41 24HC is an NMDA modulator, thus implicated in hyperexcitability as well as inflammatory responses including induction of TNF-α. Phase I studies have confirmed drug-target binding and decreased 24HC via PET ligand studies and found a favorable drug-drug interaction profile. A 2022 proof of concept study (ELEKTRA) found robust reduction in convulsive seizures in Dravet syndrome (N=51; 46% median seizure reduction; p<0.001) and reduction in drop seizures in Lennox-Gastaut syndrome (N=83; 15% placebo-adjusted median seizure reduction; p=0.13),42 encouraging future phase III studies.

Polyglucosan

In epilepsy, the cellular machinery for glycogen metabolism may fail to synthesize homogeneous glycogen β-particles thus producing insoluble polyglucosans, which cannot be further metabolized. This process results in epileptogenesis due to detrimental neuronal effects.43 This may also impair the main function of astrocytic glycogenolysis (potassium uptake), which leads to sustained depolarization.43 VAL-1221 (FAB-GAA by Parasail LLC) is an antibody-enzyme fusion given every other week designed to block this pathway by clearing polyglucosan buildup. Parasail is currently fundraising to conduct expanded studies in Lafora disease, a disorder of glycogen metabolism which causes severe progressive myoclonic epilepsy and childhood dementia. Ten Lafora patients are currently being treated worldwide via Compassionate Use including three in the US under a prospective VAL-1221 Lafora Expanded Access Protocol filed as a commercial Investigational New Drug. Parasail is seeking NIH clinical grants and industry partnerships to execute additional formal studies against this rare but devastating condition without any other known disease-modifying treatment.

Cannabidiol (CBD)

Currently, CBD is available only as a solution that suffers from poor bioavailability, poor water solubility, pharmacokinetic and dosing variability, liver toxicity, and complex drug-drug interactions.44 London Research & Pharmaceuticals is developing LRP-01, a CBD derivative, for Dravet syndrome and LGS. LRP-01 enables CBD delivery via tablets and capsules for the first time. It has significantly better bioavailability (25%, rather than <6% for CBD) and predictable pharmacokinetics, with no first-pass metabolism. It has demonstrated up to a 25-fold increase in absorption and decreased alkaline phosphatase changes and cytochrome P450 effects (p<0.05, unpublished). These modifications may confer greater anti-seizure efficacy than typical CBD. The company projects rapid market access of this new drug through the 505 (b)(2) regulatory pathway for new drug applications.

Gut microbiome

BL-001 (BloomScience) is an orally administered live biotherapeutic product developed for Dravet syndrome and other DEEs. In a 6-Hz-induced seizure model of refractory epilepsy, the gut microbiome appeared to mediate the effect of ketogenic diet on seizures, by modulating hippocampal GABA/glutamate ratios.45 BL-001 contains two human gut microbes that have been shown in both cell-based assays and animal studies to reduce hyperexcitability, increase GABA in the hippocampus, and significantly reduce both seizure frequency and duration to replicate the antiseizure effect of ketogenic diet. Bloom Science, Inc. has completed enrollment of 32 healthy adult participants in a phase I clinical trial, across four dose cohorts and randomized in a 3 to 1 ratio of BL-001 to placebo. Following completion of the current Phase I trial, they intend to initiate a Phase II study in patients with Dravet syndrome.

Neurostimulation

Vagus Nerve Stimulation (VNS), Responsive Neurostimulation (RNS), and Deep Brain Stimulation (DBS) all present treatment palliative options for patients with drug-resistant epilepsy who are not candidates for or who have failed or decline resective/ablative surgery. These devices have responder rates of around 60% at 4 to 9 years post-implantation, with potentially increasing efficacy over time, and smaller subsets achieving prolonged seizure freedom.4648

Many unanswered questions surround candidate and stimulation parameter selection. For example, in a study among 10 patients with poor response to high-frequency stimulation, transition to low-frequency stimulation was associated with a 76% median seizure reduction compared to pre-implantation baseline,49 raising questions about how to tailor parameters. Moreover, future investigation should not just focus on optimizing stimulation parameters, but also how short-term biomarker changes might predict long-term clinical response, and how to tailor stimulation timing to an individual patient’s high-risk periods per RNS electrocorticography. To illustrate, in a retrospective analysis from 25 adults, stimulation showed differential impact on seizure frequency depending upon whether patients were in low versus high seizure risk states as determined by identifying seizure cycles.50

There remains a pipeline of novel neurostimulation devices. For drug-resistant neocortical focal epilepsy, these include Starstim and EASEE (Epicranial Application of Stimulation Electrodes for Epilepsy).

Starstim applies transcranial Direct Current Stimulation (tDCS) via a wearable device over the scalp above the seizure onset zone to suppress cortical excitability and reduce seizure frequency. An individualized montage is developed for each patient. Children and adults in an open-label pilot study experienced a median 44% seizure reduction with the device.51 About 1% of patients experienced skin injury.52 Starstim is currently recruiting for a Phase III trial in children and adults. This involves a 12-week baseline, followed by 20 minutes daily of cathodal stimulation in a laboratory setting for 10 consecutive weekdays, then follow up for 10 weeks. The primary endpoint is change in seizure frequency, with a secondary endpoint of responder rate. At-home tDCS also has improved refractory depression in adults,53 which also raises prospects for its use in a prevalent epilepsy comorbidity.

The EASEE device, in contrast, consists of epicranial electrodes implanted sub-scalp above the epileptogenic focus, connected via a cable through the neck under the skin to a pulse generator in the chest. The device acts via high-frequency stimulation every other second and low-frequency stimulation for twenty minutes per day. The parameters of the stimulation are programmable via a tablet by the treating physician according to patient needs. In an open-label trial of 33 patients in Europe, there was a 52% median seizure reduction from baseline to the 6th month with 4 patients becoming seizure-free and an overall responder rate of 53%. After receiving a European CE mark, the device became commercially available in several European countries. Next steps include an IDE submission to initiate clinical work in the US.

Seizure diaries and detection devices

Electronic seizure diaries for clinical trials

Patient-reported seizure counts are imperfect.54 Seizure diaries seek to reduce recall bias by tracking outcomes in real-time.55 With the explosion of apps, electronic seizure diaries have become common in trials. For example, electronic seizure diaries have already played central roles in approval of fenfluramine for Lennox-Gastaut and Dravet syndromes. Despite additional benefits of electronic data entry and storage, seizure diaries still pose challenges in their user acceptance, database structure, and ease of integration into clinical trial data collection and reporting activities.56 Data cleaning and assessments of data integrity present particular challenges, as guidelines do not yet exist for the maximum duration of data entry delay (retrospective data entry or lookback period) and what happens thereafter if data corrections are required, which oftentimes require complex auditing processes to be fit for regulatory purposes.

Two example electronic seizure diaries in development include those by Signant Health and WCG. Both were created in collaboration with The Epilepsy Study Consortium to overcome some of these issues. Goals include standardization of data with updated terminology and common data elements,57 complete audit trails, and improved ease of use to boost compliance. To address previously reported user challenges, both diaries allow for logging seizures with familiar wording, which are then matched to appropriate clinical classification based on ILAE terminology. Lookback periods (7 days for Signant Health and 1–7 days for WCG) allow users to add/edit entries, which can reduce or eliminate data corrections identified by the site or the sponsor. From the study team’s perspective, both electronic seizure diaries offer customizable data elements - Signant Health uses two standardized templates (“Daily Seizure Count” and “Daily Seizure Details”), and WCG’s eDiary offers pre-coded fields that can be added or removed, which allows for more efficient study startup. Data management is streamlined with visualization and periodic data reporting and alerts for both diaries.

Seizure detection devices

Seizure detection devices are available for clinical and research purposes. These devices seek to reduce morbidity and mortality associated with seizures and to address the limitations of patient reporting by improving objective documentation of seizure frequency.58,59 Various modalities, including heart rate monitoring, photoplethysmography, electrodermal activity, electromyography sensory and limited EEGs have been validated and are often used in combination. The ILAE and IFCN recommend the use of wearable seizure detection devices for patients with tonic-clonic seizures with significant safety concerns, while emphasizing limitations for use in non-motor seizures.60

Absence seizures represent one seizure type that is difficult to recognize clinically without video EEG. Due to their brief duration, without motor or autonomic changes, standard modalities for seizure detection have limited use for this seizure type. Epihunter, a wearable EEG device with an accompanying CE-marked smartphone-based application for behavioral testing, is designed for the detection of absence seizures, with reported 93% sensitivity and 0/hour median false alarm rates.61 This technology is being expanded to study non-convulsive seizures in rare epilepsies such as Dravet syndrome.

Diagnostic accuracy is higher with longer EEG monitoring.62 Though, prolonged ambulatory monitoring is limited with conventional EEG equipment. The REMI remote EEG monitoring system by Epitel uses four single patient, single use wireless sensors that transmit a single channel EEG to a mobile computing device and uses automated seizure detection using machine learning to tag events for clinician review. These hardware and machine learning algorithms are under FDA review, and a clinical trial comparing seizure count from three-day ambulatory EEG versus fourteen days of the REMI system is being planned.

EpiCare@Home from Byteflies, an end-to-end service which has received a CE mark in Europe, collects data using multi-modality (two-channel behind the ear EEG and accelerometer, heart rate, respiratory rate, and activity index) data. The two-channel EEG adhesives are hydrogel-based, biocompatible with regular replacement with intended use of up to four weeks. In a study of fifty patients, 84% had a change in clinical management using data gathered from the EpiCare@Home system.

Data from these devices could assist with changes in clinical management, seizure forecasting, seizure cycle assessment, and outcomes measurement in trials. Empatica Health Monitoring Platform, using the Embrace Plus (an FDA approved wearable seizure detection device using multiple modalities), offers continuous medical-grade data collection in real-world multi-site trials in addition to their already clinically approved available device.

National Institute of Neurological Disorders and Stroke (NINDS)

Dedicated funding mechanisms promote device development. For example, the NINDS Translational Neural Devices Program funds projects ranging from preclinical to feasibility phases. Blueprint Medtech is an NIH incubator which offers development research through its two Incubator Hubs (Center for Innovative NeuroTech Advancement & NeuroTech Harbor) and translational research through NIH funding opportunities for academic institutions (PAR-21–315) or small businesses (PAR-21–282). The Brain Research Through Advancing Innovative Neurotechnologies (BRAIN) Initiative offers several funding opportunities to develop and apply innovative neurotechnologies. Goals include better understanding of the human brain and developing new ways to treat and prevent brain disorders.

Conclusion

Despite the explosion in the number of available ASMs,63 we have yet to reduce the frequency of medically-refractory patients.1 This review highlights immense, ongoing innovation. Developing new mechanisms of action, expanding existing drugs into new populations or new routes of delivery, and refining our ability to track and detect seizures all hold promise for improving the lives of people with epilepsy.

Figure 1: Epicranial Application of Stimulation Electrodes for Epilepsy (EASEE) leads.

Figure 1:

These leads are implanted underneath the scalp above the epileptogenic focus and are connected via a cable under the skin to a pulse generator in the chest. This applies high-frequency direct stimulation every other minute and direct current stimulation for twenty minutes per day. Potential advantages of this device include being able to avoid intracranial surgery during implantation, and the device is easily removable and repositioned if needed.

Table 1:

Drugs

Target Drug Company Phase Population
Ion channels
 Volt-dept. K channels XEN1101 Xenon Pharmaceuticals III Focal and generalized epilepsy
 Volt-dept. K channels BHV-7000 Biohaven Pharmaceuticals I Drug-refractory focal and generalized epilepsy
 Volt-dept. K channels ETX-123 Eliem Therapeutics Preclinical Drug-refractory focal and generalized epilepsy
 Volt-dept. Na channels NBI-921352 Neurocrine Biosciences, Inc. II Focal onset seizures
 Volt-dept. Na channels Lacosamide UCB II/III Neonates
Neurotransmitters
 GABA OV329 Ovid Therapeutics I Rare adult and pediatric epilepsies
 GABA Alprazolam UCB III Long or clustering seizures
 GABA Ganaxolone Marinus Pharmaceuticals, Inc. IV CDKL5 deficiency disorder (approved), TSC, LGS, refractory SE
 Stem cell-derived GABAergic interneurons NRTX-1001 Neurona Therapeutics I/II Unilateral drug-refractory mesial temporal lobe epilepsy
 SV2A Brivaracetam UCB II/III Absence epilepsy
 NMDA subtype of glutamate receptors Radiprodil GRIN Therapeutics IIb Pediatric patients with gain-of-function GRIN variants: seizure cohort, behavioral cohort
 Glutamate Basimglurant Noema Pharma Pediatric and adult patients with epilepsy associated with TSC
 Serotonin LP352 Longboard Pharmaceuticals Ib/Iia DEE’s
 Serotonin Fenfluramine UCB III Pediatric and adult patients with CDKL5 deficiency disorder and refractory epilepsy
 Acetylcholine SPN-817 Supernus Pharmaceuticals Iia Adult patients with refractory focal impaired awareness epilepsy
Genetic
 TANGO ASO SKT-001 Stoke I/Iia SCN1A Dravet syndrome
 AAV-miRNA AMT-260 uniQure Preclinical Temporal lobe epilepsy
 Anti-miRNA NMT.001 NEUmiRNA Therapeutics Preclinical Drug refractory epilepsy
 DNA replacement CAP-002 Capsida Biotherapeutics Preclinical STXBP1 DEE
Other/multiple targets
 Valproate Intraventricular delivery Cerebral Therapeutics II Drug-refractory adults
 Cholesterol-24-hydroxylase Soticlestat Takeda Pharmaceuticals I Dravet syndrome, LGS
 Polyglucosan VAL-1221 Parasail LLC II Lafora disease
 Multiple Cannabidiol London Research and Pharmaceuticals Anticipated 505(b)(2) regulatory pathway
 Multiple BL-001/gut bacteria Bloom Science I Dravet syndrome

Abbreviations: AAV-miRNA (Adeno-associated Virus microRNA), ASO (antisense oligonucleotide), DEE (developmental and epileptic encephalopathy), LGS (Lennox-Gastaut syndrome), SE (status epilepticus), TANGO (Targeted Augmentation of Nuclear Gene Output), TSC (Tuberous Sclerosis Complex), Volt-dept. K channels (voltage-dependent potassium channels), Volt-dept. Na channels (voltage-dependent sodium channels).

Table 2:

Devices

Purpose Device Company Phase Population
Stimulation EASEE Precisis GmbH II Drug-refractory epilepsy
tDCS Neuroelectrics III Drug-refractory epilepsy
Electronic seizure diaries Signant Health All
WCG Clinical Endpoint Solutions All
Seizure detection devices EmbarcePlus Empatica, Inc. All
EpiCare@Home EpiCare@Home All
REMI Epitel, Inc. All
AWARE Epihunter All

Abbreviations: AWARE (Automated Awareness Testing in Absence Epilepsy), EASEE (Epicranial Application of Stimulation Electrodes for Epilepsy), REMI (Remote EEG Ambulatory Monitoring), tDCS (transcranial Direct Current Stimulation).

Key points box.

  • This review discusses the latest drugs and devices in the epilepsy diagnostics and therapeutics pipeline.

  • Many antiseizure medications plus other genetic and non-genetic therapeutics are currently under development, including numerous that are first-in-class.

  • Seizure tracking, detection, and stimulation devices all continue to make advances as devices enter the pipeline.

Acknowledgements

This manuscript summarizes content from the 17th Epilepsy Therapies and Diagnostics Development conference (formerly the Antiepileptic Drug and Device Symposium) that occurred from May 31st to June 2nd, 2023, in Aventura, Florida. The conference brought together leaders in academia, industry, and NIH to discuss the latest advances in epilepsy care.

We would like to acknowledge the following speakers whose lectures were summarized in this review: Daniel Abrams, MD, Dustin Armstrong, PhD, Mahnaz Asgharnejad, PharmD, Paolo Baroldi, MD, PhD, Andreas Borta, Manuela Bossi, PhD, Tim Buckinx, Manuela Cerina, PhD, Gail Farfel, PhD, Nicholas Flytzanis, PhD, Brooks Gross, PhD, Stine N. Hansen, PhD, Joseph Hulihan, MD, Randall Kaye, MD, Christopher Kenney, MD, Gregory Krauss, MD, Matteo Lai, Abdel-Rahman Lawendy, MD, PhD, Renata Lazarova, MD, Mark Lehmkuhle, PhD, Jason Lerner, MD, Angela Liedler, MD, Brian Moseley, MD, Cory Nicholas, PhD, Julie Nys, PhD, Welton O’Neal Jr., PharmD, Michael Panzara, MD, Alexander Rotenberg, MD, PhD, Svetlana Shore, PhD, Michael Sperling, MD, Shelly Steele, and Julia Tsai, PhD.

Funding

The Epilepsy Foundation, Epilepsy Study Consortium, and Pediatric Epilepsy Research Foundation pooled funds to provide travel grants to facilitate attendance at this conference (along with the May Liang and James Lintott Scholarship).

Dr Terman was supported by the Susan S Spencer Clinical Research Training Scholarship, the Michigan Institute for Clinical and Health Research J Award UL1TR002240, and the American Epilepsy Society/Epilepsy Study Consortium Mini-grant. He is now supported by the American Epilepsy Society Clinical Research and Training Fellowship for Clinicians.

Dr Kirkpatrick has received an honorarium from the Child Neurology Foundation and travel funding from the One8 Foundation. She is now supported by the Child Neurologist Career Development Program 5K12NS098482-07.

The other authors report no other relevant funding.

Disclosures of conflict of interest

No pharmaceutical or device company was involved in the initial drafting of this manuscript. Each speaker had the opportunity to provide comments on this manuscript for content feedback after the initial drafting phase.

Dr. Terman receives salary support from Xenon Pharmaceuticals as a site Primary Investigator for the ongoing Phase III trial of XEN1101 in focal epilepsy.

Dr. French receives salary support from the Epilepsy Foundation and for consulting work and/or attending Scientific Advisory Boards on behalf of the Epilepsy Study Consortium for Aeonian/Aeovian, Alterity Therapeutics Limited, Anavex, Arkin Holdings, Angelini Pharma S.p.A, Arvelle Therapeutics, Inc., Athenen Therapeutics/Carnot Pharma, Autifony Therapeutics Limited, Baergic Bio, Biogen, Biohaven Pharmaceuticals, BioMarin Pharmaceutical Inc., BioXcel Therapeutics, Bloom Science Inc., BridgeBio Pharma Inc., Camp4 Therapeutics Corporation, Cerebral Therapeutics, Cerevel, Clinical Education Alliance, Coda Biotherapeutics, Corlieve Therapeutics, Eisai, Eliem Therapeutics, Encoded Therapeutics, Encoded Therapeutics, Engage Therapeutics, Engrail, Epalex, Epihunter, Epiminder, Epitel Inc., Equilibre BioPharmaceuticals, Greenwich Biosciences, Grin Therapeutics, GW Pharma, Janssen Phamaceutica, Jazz Pharmaceuticals, Knopp Biosciences, Lipocine, LivaNova, Longboard Pharmaceuticals, Lundbeck, Marinus, Mend Neuroscience, Marck, NeuCyte Inc., Neumirna Therapeutics, Neurocrine, Neuroelectives USA Corporation, Neuronetics Inc., Neuropace, NxGen Medicine Inc., Ono Pharmaceutical Co., Otsuka Pharmaceutical Development, Ovid Therapeutics Inc., Paladin Labs, Passage Bio, Pfizer, Praxis, Pure Tech LTY Inc., Rafa Laboratories Ltd, SK Life Sciences, Sofinnova, Stoke, Supernus, Synergia Medical, Takeda, UCB Inc., Ventus Therapeutics, Xenon, Xeris, Zogenix, Zynerba. Dr. French also has received research support from the Epilepsy Study Consortium (Funded by Andrews Foundation, Eisai, Engage, Lundbeck, Pfizer, SK Life Science, Sunovion, UCB, Vogelstein Foundation), the Epilepsy Study Consortium/Epilepsy Foundation (Funded by UCB), GW/FACES, and NINDS. She is on the editorial board of Lancet Neurology and Neurology Today. She is Chief Medical/Innovation Officer of the Epilepsy Foundation. She has received travel reimbursement related to research, advisory meetings, or presentation of results at scientific meetings from the Epilepsy Study Consortium, the Epilepsy Foundation, Angelini Pharma S.p.A., Clinical Education Alliance, NeuCyte, Inc., Neurocrine, Praxis, and Xenon.

Footnotes

Ethical publication statement

We confirm that we have read the Journal’s position on issues involved in ethical publication and affirm that this report is consistent with those guidelines.

References

  • 1.Chen Z, Brodie MJ, Liew D, Kwan P. Treatment Outcomes in Patients With Newly Diagnosed Epilepsy Treated With Established and New Antiepileptic Drugs A 30-Year Longitudinal Cohort Study. JAMA Neurol. 2018; 75(3):279–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Porter RJ, Partiot A, Sachdeo R, Nohria V, Alves WM. Randomized, multicenter, dose-ranging trial of retigabine for partial-onset seizures. Neurology. 2007; 68(15):1197–204. [DOI] [PubMed] [Google Scholar]
  • 3.Clark S, Antell A, Kaufman K. New antiepileptic medication linked to blue discoloration of the skin and eyes. Ther Adv Drug Saf. 2015; 6(1):15–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.French JA, Porter RJ, Perucca E, Brodie MJ, Rogawski MA, Pimstone S, et al. Efficacy and Safety of XEN1101, a Novel Potassium Channel Opener, in Adults with Focal Epilepsy: A Phase 2b Randomized Clinical Trial. JAMA Neurol [Internet]. 2023; :E1–10. Available from: https://www.cochranelibrary.com/central/doi/10.1002/central/CN-02502531/full [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Picchione KE, Inglis AM, Resnick L, Mareska DA, Bozik ME, Dworetzky S. Discovery and Characterization of KB-3061: A Potent Kv7.2/7.3 Ion Channel Activator for the Treatment of KCNQ2-Neonatal Epileptic Encephalopathy. In: American Epilepsy Society Abstract number 3045 [Internet]. 2019. Available from: https://aesnet.org/abstractslisting/discovery-and-characterization-of-kb-3061--a-potent-kv7-2/7-3-ion-channel-activator-for-the-treatment-of-kcnq2-neonatal-epileptic-encephalopathy [Google Scholar]
  • 6.Biohaven slide deck (slides 48–50) [Internet]. Biohaven Pharmaceuticals. [cited 2023]. Available from: https://ir.biohaven.com/static-files/10426edf-1b55-4669-bbab-6b4a3e422c58 [Google Scholar]
  • 7.Gunthorpe MJ, Large CH, Sankar R. The mechanism of action of retigabine (ezogabine), a first-in-class K+ channel opener for the treatment of epilepsy. Epilepsia. 2012; 53(3):412–24. [DOI] [PubMed] [Google Scholar]
  • 8.England S, De Groot MJ. Subtype-selective targeting of voltage-gated sodium channels. Br J Pharmacol. 2009; 158(6):1413–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Erdemli G, Kim AM, Ju H, Springer C, Penland RC, Hoffmann PK. Cardiac safety implications of hNav1.5 blockade and a framework for pre-clinical evaluation. Front Pharmacol. 2012; 3 JAN(January):1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Loussouarn G, Sternberg D, Nicole S, Marionneau C, Le Bouffant F, Toumaniantz G, et al. Physiological and pathophysiological insights of Nav1.4 and Nav1.5 comparison. Front Pharmacol. 2016; 6(JAN). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Nicole S, Lory P. New Challenges Resulting From the Loss of Function of Nav1.4 in Neuromuscular Diseases. Front Pharmacol. 2021; 12(October):1–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Männikkö R, Wong L, Tester DJ, Thor MG, Sud R, Kullmann DM, et al. Dysfunction of NaV1.4, a skeletal muscle voltage-gated sodium channel, in sudden infant death syndrome: a case-control study. Lancet. 2018; 391(10129):1483–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Brunklaus A, Ellis R, Reavey E, Forbes GH, Zuberi SM. Prognostic, clinical and demographic features in SCN1A mutation-positive Dravet syndrome. Brain. 2012; 135(8):2329–36. [DOI] [PubMed] [Google Scholar]
  • 14.French J, Biton V, Dave H, Detyniecki K, Gelfand MA, Gong H, et al. A randomized phase 2b efficacy study in patients with seizure episodes with a predictable pattern using Staccato® alprazolam for rapid seizure termination. Epilepsia. 2023; 64(2):374–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Lattanzi S, Riva A, Striano P. Ganaxolone treatment for epilepsy patients: from pharmacology to place in therapy. Expert Rev Neurother [Internet]. 2021; 21(11):1317–32. Available from: 10.1080/14737175.2021.1904895 [DOI] [PubMed] [Google Scholar]
  • 16.Knight EMP, Amin S, Bahi-Buisson N, Benke TA, Cross JH, Demarest ST, et al. Safety and efficacy of ganaxolone in patients with CDKL5 deficiency disorder: results from the double-blind phase of a randomised, placebo-controlled, phase 3 trial. Lancet Neurol. 2022; 21(5):417–27. [DOI] [PubMed] [Google Scholar]
  • 17.Vaitkevicius H, Ramsay RE, Swisher CB, Husain AM, Aimetti A, Gasior M. Intravenous ganaxolone for the treatment of refractory status epilepticus: Results from an open-label, dose-finding, phase 2 trial. Epilepsia. 2022; 63(9):2381–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Samanta D, Ostendorf AP, Willis E, Singh R, Gedela S, Arya R, et al. Underutilization of epilepsy surgery: Part I: A scoping review of barriers. Epilepsy Behav [Internet]. 2021; 117:107837. Available from: 10.1016/j.yebeh.2021.107837 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bershteyn M, Broer S, Parekh M, Maury Y, Havlicek S, Kriks S, et al. Human pallial MGE-type GABAergic interneuron cell therapy for chronic focal epilepsy. Cell Stem Cell. 2023; 30(10):1331–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Brigo F, Igwe SC, Lattanzi S. Ethosuximide, sodium valproate or lamotrigine for absence seizures in children and adolescents. Cochrane Database Syst Rev. 2021; 2021(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kasteleijn-Nolst Trenité DGA, Genton P, Parain D, Masnou P, Steinhoff BJ, Jacobs T, et al. Evaluation of brivaracetam, a novel SV2A ligand, in the photosensitivity model. Neurology. 2007; 69(10):1027–34. [DOI] [PubMed] [Google Scholar]
  • 22.Bast T, Schulz AL, Floricel F, Morita D, Cleveland JM, Elshoff JP. Efficacy and tolerability of brivaracetam monotherapy in childhood and juvenile absence epilepsy: An innovative adaptive trial design. Epilepsia Open. 2022; 7(4):588–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Benke TA, Park K, Krey I, Camp CR, Song R, Ramsey AJ, et al. Clinical and therapeutic significance of genetic variation in the GRIN gene family encoding NMDARs. Neuropharmacology [Internet]. 2021; 199(July):108805. Available from: 10.1016/j.neuropharm.2021.108805 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sciberras D, Otoul C, Lurquin F, Smeraglia J, Lappert A, Bruyn S De, et al. A pharmacokinetic study of radiprodil oral suspension in healthy adults comparing conventional venous blood sampling with two microsampling techniques. Pharmacol Res Perspect. 2019; (e00459):1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Auvin S, Dozières-Puyravel B, Avbersek A, Sciberras D, Collier J, Leclercq K, et al. Radiprodil, a NR2B negative allosteric modulator, from bench to bedside in infantile spasm syndrome. Ann Clin Transl Neurol. 2020; 7(3):343–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kelly E, Schaeffer SM, Dhamne SC, Lipton JO, Lindemann L, Honer M, et al. mGluR5 Modulation of Behavioral and Epileptic Phenotypes in a Mouse Model of Tuberous Sclerosis Complex. Neuropsychopharmacology [Internet]. 2018; 43(6):1457–65. Available from: 10.1038/npp.2017.295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Youssef EA, Berry-Kravis E, Czech C, Hagerman RJ, Hessl D, Wong CY, et al. Effect of the mGluR5-NAM Basimglurant on Behavior in Adolescents and Adults with Fragile X Syndrome in a Randomized, Double-Blind, Placebo-Controlled Trial: FragXis Phase 2 Results. Neuropsychopharmacology [Internet]. 2018; 43(3):503–12. Available from: 10.1038/npp.2017.177 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bourson A, Wanner D, Wyler R, Petit N, Zwingelstein C, Rudler A, et al. Pharmacologic evaluation of the discriminative stimulus of metachlorophenylpiperazine. Pharmacol Biochem Behav. 1996; 53(1):107–14. [DOI] [PubMed] [Google Scholar]
  • 29.Devinsky O, King LT, Schwartz D, Conway E, Price D. Effect of fenfluramine on convulsive seizures in CDKL5 deficiency disorder. Epilepsia. 2021; 62(7):e98–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Gnatek Y, Zimmerman G, Goll Y, Najami N, Soreq H, Friedman A. Acetylcholinesterase loosens the brain’s cholinergic anti-inflammatory response and promotes epileptogenesis. Front Mol Neurosci. 2012; 5(MAY 2012):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Portelli J, Park L, Lujan B, Busse G, Rubin J, Nasser A. An Open-Label Pilot Trial Assessing the Safety and Efficacy of SPN-817 (Huperzine A Extended-Release) in Adults with Treatment-Resistant Focal Impaired Awareness Seizures (P12–8.003). 2022; May 3; 98(18 Supplement). Available from: https://n.neurology.org/content/98/18_Supplement/1388 [Google Scholar]
  • 32.Meng H, Xu HQ, Yu L, Lin GW, He N, Su T, et al. The SCN1A Mutation Database: Updating Information and Analysis of the Relationships among Genotype, Functional Alteration, and Phenotype. Hum Mutat. 2015; 36(6):573–80. [DOI] [PubMed] [Google Scholar]
  • 33.Chi X, Gatti P, Papoian T. Safety of antisense oligonucleotide and siRNA-based therapeutics. Drug Discov Today [Internet]. 2017; 22(5):823–33. Available from: 10.1016/j.drudis.2017.01.013 [DOI] [PubMed] [Google Scholar]
  • 34.Peret A, Christie LA, Ouedraogo DW, Gorlewicz A, Epsztein JÔ, Mulle C, et al. Contribution of Aberrant GluK2-Containing Kainate Receptors to Chronic Seizures in Temporal Lobe Epilepsy. Cell Rep. 2014; 8(2):347–54. [DOI] [PubMed] [Google Scholar]
  • 35.Abramov D, Guiberson NGL, Burré J. STXBP1 encephalopathies: Clinical spectrum, disease mechanisms, and therapeutic strategies. J Neurochem. 2021; 157(2):165–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Stamberger H, Nikanorova M, Accorsi P, Angriman M, Benkel-herrenbrueck I, Capovilla G, et al. STXBP1: A neurodevelopmental disorder including epilepsy. Neurology. 2016; 86(10):954–92. [DOI] [PubMed] [Google Scholar]
  • 37.Schratt GM, Tuebing F, Nigh EA, Kane CG, Sabatini ME, Kiebler M, et al. A brain-specific microRNA regulates dendritic spine development. Nature. 2006; 439(7074):283–9. [DOI] [PubMed] [Google Scholar]
  • 38.Jimenez-Mateos EM, Engel T, Merino-Serrais P, McKiernan RC, Tanaka K, Mouri G, et al. Silencing microRNA-134 produces neuroprotective and prolonged seizure-suppressive effects. Nat Med. 2012; 18(7):1087–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Cohen-Pfeffer JL, Gururangan S, Lester T, Lim DA, Shaywitz AJ, Westphal M, et al. Intracerebroventricular Delivery as a Safe, Long-Term Route of Drug Administration. Pediatr Neurol [Internet]. 2017; 67:23–35. Available from: 10.1016/j.pediatrneurol.2016.10.022 [DOI] [PubMed] [Google Scholar]
  • 40.Cook M, Murphy M, Bulluss K, D’Souza W, Plummer C, Priest E, et al. Anti-seizure therapy with a long-term, implanted intra-cerebroventricular delivery system for drug-resistant epilepsy: A first-in-man study. EClinicalMedicine [Internet]. 2020; 22:100326. Available from: 10.1016/j.eclinm.2020.100326 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Koike T, Yoshikawa M, Ando HK, Farnaby W, Nishi T, Watanabe E, et al. Discovery of Soticlestat, a Potent and Selective Inhibitor for Cholesterol 24-Hydroxylase (CH24H). J Med Chem. 2021; 64(16):12228–44. [DOI] [PubMed] [Google Scholar]
  • 42.Hahn CD, Jiang Y, Villanueva V, Zolnowska M, Arkilo D, Hsiao S, et al. A phase 2, randomized, double-blind, placebo-controlled study to evaluate the efficacy and safety of soticlestat as adjunctive therapy in pediatric patients with Dravet syndrome or Lennox–Gastaut syndrome (ELEKTRA). Epilepsia. 2022; 63(10):2671–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.DiNuzzo M, Mangia S, Maraviglia B, Giove F. Does abnormal glycogen structure contribute to increased susceptibility to seizures in epilepsy? Metab Brain Dis. 2015; 30(1):307–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Millar SA, Maguire RF, Yates AS, O’sullivan SE. Towards better delivery of cannabidiol (CBD). Pharmaceuticals. 2020; 13(9):1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Olson CA, Vuong HE, Yano JM, Liang QY, Nusbaum DJ, Hsiao EY. The Gut Microbiota Mediates the Anti-Seizure Effects of the Ketogenic Diet. Cell [Internet]. 2018; 173(7):1728–1741.e13. Available from: 10.1016/j.cell.2018.04.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Nair DR, Morrell MJ, Skarpaas TL, Murro AM, Park YD, Barkley GL, et al. Nine-year prospective efficacy and safety of brain-responsive neurostimulation for focal epilepsy. Neurology. 2020; 95(9):E1244–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Salanova V, Sperling MR, Gross RE, Irwin CP, Vollhaber JA, Giftakis JE, et al. The SANTÉ study at 10 years of follow-up: Effectiveness, safety, and sudden unexpected death in epilepsy. Epilepsia. 2021; 62(6):1306–17. [DOI] [PubMed] [Google Scholar]
  • 48.Englot DJ, Rolston JD, Wright CW, Hassnain KH, Chang EF. Rates and Predictors of Seizure Freedom with Vagus Nerve Stimulation for Intractable Epilepsy. Neurosurgery. 2016; 79(3):345–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Alcala-Zermeno JL, Starnes K, Gregg NM, Worrell G, Lundstrom BN. Responsive neurostimulation with low- frequency stimulation. Epilepsia. 2022; 64(2):e16–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chiang S, Khambhati AN, Wang ET, Vannucci M, Edward F, Rao VR, et al. Evidence of state-dependence in the effectiveness of responsive neurostimulation for seizure modulation. Brain Stimul. 2021; 14(2):366–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kaye HL, San-Juan D, Salvador R, Biagi MC, Dubreuil-Vall L, Damar U, et al. Personalized, Multisession, Multichannel Transcranial Direct Current Stimulation in Medication-Refractory Focal Epilepsy: An Open-Label Study. J Clin Neurophysiol. 2023; 40(1):53–62. [DOI] [PubMed] [Google Scholar]
  • 52.Auvichayapat N, Rotenberg A, Gersner R, Ngodklang S, Tiamkao S, Tassaneeyakul W, et al. Transcranial direct current stimulation for treatment of refractory childhood focal epilepsy. Brain Stimul [Internet]. 2013; 6(4):696–700. Available from: 10.1016/j.brs.2013.01.009 [DOI] [PubMed] [Google Scholar]
  • 53.Cappon D, den Boer T, Jordan C, Yu W, Lo A, LaGanke N, et al. Safety and Feasibility of Tele-Supervised Home-Based Transcranial Direct Current Stimulation for Major Depressive Disorder. Front Aging Neurosci. 2022; 13(February):1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Hoppe C, Poepel A, Elger CE. Accuracy of patient seizure counts. Arch Neurol. 2007; 64(11):1595–9. [DOI] [PubMed] [Google Scholar]
  • 55.Bidwell J, Khuwatsamrit T, Askew B, Ehrenberg JA, Helmers S. Seizure reporting technologies for epilepsy treatment: A review of clinical information needs and supporting technologies. Seizure [Internet]. 2015; 32:109–17. Available from: 10.1016/j.seizure.2015.09.006 [DOI] [PubMed] [Google Scholar]
  • 56.Gray TE, Hirsch LJ, Quraishi IH. Electronic seizure diaries for clinical care and research. Epileptic Disord. 2022; 24(5):803–11. [DOI] [PubMed] [Google Scholar]
  • 57.Goldenholz DM, Moss R, Jost DA, Crone NE, Krauss G, Picard R, et al. Common data elements for epilepsy mobile health systems. Epilepsia. 2018; 59(5):1020–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Bruno E, Böttcher S, Viana PF, Amengual-Gual M, Joseph B, Epitashvili N, et al. Wearable devices for seizure detection: Practical experiences and recommendations from the Wearables for Epilepsy And Research (WEAR) International Study Group. Epilepsia. 2021; 62(10):2307–21. [DOI] [PubMed] [Google Scholar]
  • 59.Shum J, Friedman D. Commercially available seizure detection devices: A systematic review. J Neurol Sci [Internet]. 2021; 428:117611. Available from: 10.1016/j.jns.2021.117611 [DOI] [PubMed] [Google Scholar]
  • 60.Beniczky S, Wiebe S, Jeppesen J, Tatum WO, Brazdil M, Wang Y, et al. Automated seizure detection using wearable devices: A clinical practice guideline of the International League Against Epilepsy and the International Federation of Clinical Neurophysiology. Clin Neurophysiol. 2021; 132(5):1173–84. [DOI] [PubMed] [Google Scholar]
  • 61.Japaridze G, Loeckx D, Buckinx T, Armand Larsen S, Proost R, Jansen K, et al. Automated detection of absence seizures using a wearable electroencephalographic device: a phase 3 validation study and feasibility of automated behavioral testing. Epilepsia. 2022; . [DOI] [PubMed] [Google Scholar]
  • 62.Hernandez-ronquillo L, Thorpe L, Feng C, Hunter G. Diagnostic Accuracy of Ambulatory EEG vs Routine EEG in Patients With First Single Unprovoked Seizure. Neurology. 2023; 13:3200160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Brodie MJ. Antiepileptic drug therapy the story so far. Seizure [Internet]. 2010; 19(10):650–5. Available from: 10.1016/j.seizure.2010.10.027 [DOI] [PubMed] [Google Scholar]

RESOURCES