FormalPara Key Summary Points

Despite the availability of more than 30 antiseizure medications, about one third of people with epilepsy do not achieve seizure freedom with existing pharmacological treatments.

Potentiation of GABAergic inhibition using novel approaches is among the strategies being pursued in the effort to develop better treatments for seizures and epilepsy.

Investigational treatments discussed in this article include novel therapeutic agents as well as drugs already in the market for non-epilepsy indications and being considered for repurposing in the treatment of seizure disorders.

Several drugs in clinical development are novel positive allosteric modulators of GABAA receptors, aimed at taking advantage of the structural and functional heterogeneity of these receptors. These agents include novel neurosteroids and subunit-selective allosteric GABAA receptor modulators.

Highly innovative treatments in early clinical development include an antisense oligonucleotide aimed at restoring the function of GABAergic interneurons in patients with Dravet syndrome, a cell therapy for mesial temporal lobe epilepsy based on stereotactically implanted GABAergic interneurons and a neuroprotective neuropeptide intended to upregulate GABA signalling. An adenoviral vector-based gene therapy is also scheduled for investigation in Dravet syndrome.

Overall, the current pipeline of-γ-aminobutyric acid targeting agents includes many promising treatments, but for most of these, clinical data are too limited to infer their actual value in the therapeutic setting.

1 Introduction

Because of the critical function of γ-aminobutyric acid (GABA) in modulating neuronal excitability [1] and the role played by a relative deficiency in GABA-mediated inhibition in the pathogenesis and expression of some seizure disorders [2,3,4], potentiation of GABAergic transmission has proved to be an attractive target for the development of new treatments for epilepsy for many years [5, 6]. Accordingly, the number of established antiseizure medications (ASMs) that act at least in part by enhancing GABAergic transmission has increased over time (Table 1) [5], with the two latest additions (cenobamate and ganaxolone) being discussed in detail in an accompanying article [6].

Table 1 Currently marketed antiseizure medications that exert their effects at least in part by enhancement of GABAergic transmissiona

Despite the current availability of more than 30 ASMs, the pharmacological management of epilepsy continues to face major unmet needs. In particular, the second-generation ASMs introduced in the last 30 years have not reduced substantially the proportion of patients (about one third) who do not achieve seizure freedom with available medical treatments [7, 8]. Drug resistance continues to be highly prevalent in the most severe forms of epilepsy, particularly developmental and epileptic encephalopathies [8,9,10]. There is also a need for newer medications with improved tolerability and safety profiles, as well as for medications that could modify favourably the course of epilepsy rather than simply exerting a symptomatic effect against seizures [7]. During the last several decades, knowledge of the molecular biology of the GABA system has advanced greatly, in parallel with the understanding of the clinical implications of pharmacological manipulation of this system [4, 6]. Progress has also been made towards innovative approaches such as gene and cell therapies that could target the underlying aetiology of some epilepsies [11]. Overall, these advances have created attractive opportunities to address at least some of the unmet needs in epilepsy management. The purpose of the present article is to review investigational GABA-targeting therapies that are currently in clinical development for the treatment of seizure disorders. As it will be apparent from the pipeline discussion below, these include not only therapies that are entirely novel, but also drugs acting on the GABA system that are already in clinical use for other indications and are currently being investigated as potential repurposed treatments for seizures and epilepsy.

2 Novel Therapies in Clinical Development

2.1 Darigabat

Darigabat (CVL-865, PF-06372865) is an imidazopyridazine derivative, structurally unrelated to benzodiazepines and neurosteroids, which emerged from a search for novel GABAA-positive allosteric modulators retaining the anxiolytic and antiseizure properties of benzodiazepines while minimising undesirable effects such as somnolence, cognitive dysfunction, tolerance to therapeutic effects and abuse potential [12]. There is evidence that amnestic, sedative, ataxic and dependence-inducing effects of benzodiazepines are largely mediated by enhanced GABA signalling at GABAA receptors containing α1 subunits [13,14,15]. Conversely, anxiolytic, antiseizure and pain-modulating properties are mediated to an important extent by actions at α2 and α3 subunits [13,14,15,16,17,18], even though actions at the α1 subunit appear to also contribute to antiseizure effects, at least in some rodent models [13]. Based on these findings, efforts aimed at minimising undesirable properties of benzodiazepines have focused on the identification of positive allosteric modulators acting selectively at α2 subunits and α3 subunits [14]. In vitro, darigabat binds with high affinity at GABAA receptors containing α1, α2, α3 or α5 subunits, and has very low affinity for GABAA receptors containing α4 or α6 subunits, which do not have a benzodiazepine binding site [12]. However, functionally, darigabat exerts a low level of allosteric modulation at α1 subunits, and it behaves pharmacologically as an α2/3/5 subtype-selective, GABAA-positive allosteric modulator [12, 19, 20]. Its functional selectivity for α2/3/5 subunits relative to the α1 subunit has been confirmed in vivo by assessment of pharmacodynamic biomarkers in healthy subjects [19] and may contribute to an improved tolerability profile.

At doses of 0.3–10 mg/kg orally, darigabat showed dose-dependent anticonvulsant activity in the subcutaneous pentylenetetrazole test in mice [12, 21]. Darigabat (1 and 10 mg/kg orally) also showed antiseizure activity in the amygdala-kindled rat model of focal seizures [12, 21] and, at doses of 3 and 10 mg/kg orally, it showed comparable activity to diazepam (2 mg/kg intraperitoneally) in reducing hippocampal paroxismal discharges in the intra-hippocampal kainate mouse model of mesial temporal lobe epilepsy [22]. Darigabat (0.3–10 mg/kg orally) was also effective in reducing dose dependently spike‐and‐wave discharges in the Genetic Absence Rat from Strasbourg (GAERS) model of absence epilepsy, with complete suppression of epileptic discharges 30 min after administration of a 10-mg/kg dose [23], an effect that coupled with results obtained in other seizure models suggests broad-spectrum antiseizure activity.

In phase I dose-escalating studies in healthy subjects, darigabat (oral suspension) was absorbed rapidly following single doses of 0.04–100 mg, with peak plasma drug concentrations (Cmax) occurring at a median of 1–4 h after dosing [19]. The half-life of darigabat across these doses ranged from 6.0 to 8.9 h. Area under the plasma drug concentration–time curve and Cmax values appeared to increase dose proportionally within the dose range investigated. In these studies, mean oral clearance of darigabat ranged from 17.4 to 26.9 L/h, and its mean apparent volume of distribution ranged from 194 to 260 L [19]. Multiple-dose pharmacokinetics was assessed in a phase Ib study in 18 healthy subjects who received darigabat orally for 21 days [24]. Treatment included a 7-day titration phase followed by a 14-day maintenance phase at a dose of 25 mg twice daily (Cohort 1) or 42.5 mg twice daily (Cohort 2), with the higher dose expected to achieve a > 80% receptor occupancy [24]. Darigabat Cmax occurred at 1–2 h after dosing, with Cmax and area under the plasma drug concentration–time curve values during a dosing interval at steady state being approximately proportional to dose. The half-life of sarigabat assessed after stopping multiple-dose administration was about 11 h.

A summary of tolerability data from several studies in 136 healthy subjects and a total of 153 patients with a variety of conditions (photosensitive epilepsy, generalised anxiety disorder and chronic low back pain) enrolled in phase II trials concluded that darigabat is generally well tolerated. Dizziness and somnolence were the most commonly reported treatment-emergent adverse events (TEAEs), and could be minimised by including a short titration period upon initiation of therapy [20].

In a proof-of-concept, double-blind, randomised, placebo-controlled trial in seven patients with photosensitive epilepsy, single doses of darigabat (17.5 and 52 mg, expected to produce GABAA receptor of occupancy ≤ 60% and about 80%, respectively) administered as a tablet formulation produced a statistically significant reduction in the photoparoxismal electroencephalographic (EEG) response compared with placebo [25]. The response did not differ between the two doses, and was similar to that achieved with a single 2-mg oral dose of lorazepam. The effect was already present at the first assessment (1 hour after dosing) and persisted throughout the 6-hour assessment period. Six of the seven patients showed complete suppression of the photoparoxismal response with darigabat.

A phase II, randomised, placebo-controlled, double-blind adjunctive-therapy trial (NCT04244175) to evaluate the safety and efficacy of darigabat doses of 7.5 mg and 25 mg twice daily in 150 patients with drug-resistant focal seizures is ongoing. Darigabat is also being developed as a potential treatment for panic disorder [26]. With respect to other potential indicatients, darigabat failed to meet the primary efficacy endpoint in phase II placebo-controlled exploratory trials in patients with generalised anxiety disorder [27] and chronic low back pain [28].

2.2 ENX-101

ENX-101 is a subtype-selective GABAA receptor-positive allosteric modulator acting at receptors containing α2, α3 or α5 subunits, but devoid of positive allosteric activity at α1 subunit-containing receptors [29, 30]. The compound has been designed with the aim of maximising antiseizure activity while minimising the potential for sedation and tolerance, which are considered to be mediated largely by α1 subunit activation [31,32,33]. In a placebo-controlled phase I study in healthy volunteers, ENX-101 given orally once daily in the morning for 10 days at doses that ranged from 5 to 50 mg/day without titration was well tolerated, with evidence of persistent target engagement based on changes in a number of pharmacodynamic biomarkers [29]. The most commonly reported TEAE was mild and transient somnolence. The half-life of ENX-101 was estimated to be about 20 h. A phase II, randomised, placebo-controlled, double-blind, adjunctive-therapy trial to assess the efficacy and tolerability of ENX (15 and 30 mg/day for 56 days) in comparison to placebo in 180 adults with focal epilepsy has been scheduled (NCT05481905).

2.3 ETX-101

ETX-101 can be described as “a non-replicating, recombinant adeno-associated viral vector serotype 9 (rAAV9) comprising a GABAergic regulatory element (reGABA) and an engineered transcription factor that increases transcription of the SCN1A gene (eTFSCN1A)” [34]. The product is intended as a one-time intracerebroventricular (i.c.v.) treatment for patients with Dravet syndrome.

ETX-101 has been designed with the aim to restore the function of inhibitory GABAergic interneurons of patients with Dravet syndrome by upregulating the expression of voltage-gated type I sodium channels (Nav1.1), thus correcting the functional defect underlying the disease. The regulatory element in ETX101 was engineered by interrogating human genetic sequences surrounding the GAD1 gene, expressed specifically in GABAergic interneurons, with the goal of limiting transgene expression to GABAergic inhibitory neurons and reducing off-target expression within excitatory cells [35].

In non-human primates, a one-time unilateral i.c.v. injection was found to be well tolerated and to be associated with widespread distribution of the vector and efficient transgene expression throughout the brain [35, 36]. In a genetic mouse model of Dravet syndrome, a one-time bilateral i.c.v. injection of ETX-101 resulted in increased SCN1A messenger RNA (mRNA) transcripts specifically within GABAergic inhibitory interneurons, and increased NaV1.1 protein levels in the brain. These changes were associated with reduction in spontaneous and hyperthermia-induced seizures, and prolonged survival. The protocol of the phase I/II trial of ETX-101 in infants and children with Dravet syndrome (NCT05419492) is available at ClinicalTrials.gov [34].

2.4 ETX-155

ETX-155 is a novel neuroactive steroid that shares with other neurosteroids an ability to act as a positive allosteric modulator of both synaptic and extrasynaptic GABAA receptors [37]. It is currently being developed for the treatment of focal epilepsy and major depressive disorder [38].

Preclinically, ETX-155 has been reported to be effective in various rodent seizure models, including seizures induced by pentylenetetrazole and maximal electroshock, as well as in animal models of anxiety and depression [37]. In a phase I, single ascending dose study in healthy subjects that evaluated oral doses between 5 and 300 mg, the maximal tolerated dose was found to be 135 mg, with somnolence and dizziness being dose-limiting TEAEs. In multiple-dose studies investigating doses of 60 mg once daily for 7 and 14 days, the most commonly reported TEAEs were somnolence, headache and fatigue [37]. Tolerability was improved when the dose was taken in the evening compared with dosing in the morning.

After single doses of 5–300 mg, ETX-155 is rapidly absorbed from the gastrointestinal tract with Cmax values occurring at 2–4 h after dosing [37]. Within the dose range tested, plasma ETX-155 concentrations increased dose proportionally. Co-administration with food had no clinically meaningful effect on the bioavailability of a 30-mg oral dose, suggesting that ETX-155 may be categorised as a Biopharmaceutics Classification System/Biopharmaceutical Drug Disposition and Classification System Class 1 drug. After multiple dosing, steady state is achieved after 8 days, consistent with a half-life of about 40 h [37]. A preliminary analysis of data from three patients included in a phase Ib proof-of-concept study on the effect of ETX-155 on the photoparoxismal EEG response in patients with photosensitive epilepsy showed inconclusive results, putatively because of plasma drug concentrations being significantly lower than expected [39].

2.5 NRP2945

NRP2945 is an engineered derivative of the neural regeneration peptide (NRP), which belongs to a family of gene-encoded proteins found in mammals and endowed with neuroregenerative and anti-inflammatory properties [40, 41]. NRP2945 is derived from the peptide encoded by the human NRP gene, also known as calcium-dependent activator of protein secretion 2 (CAPS2) and corresponds to the amino acid position 40–50 within the CAPS2 sequence [42, 43]. NRP2945 has been reported to interact with the chemokine receptor with cxc motif 4 and with another chemokine receptor subunit, leading to increased expression of GABAA receptor α and β subunits and upregulation of GABA signalling [41]. Although only a small proportion of circulating NRP2945 crosses the blood–brain barrier, the picomolar levels of the peptide that reach the brain have been deemed to be sufficient for pharmacological activity [43].

In rat studies, NRP2945 (20 μg/kg intraperitoneally) has been found to protect against seizures induced by pentylenetetrazole (50 μg/kg intraperitoneally) [42]. In the same model, NRP2945 inhibited the neuronal damage induced by pentylenetrazole and increased the expression of GABAA receptor α and β subunits and glutamic acid decarboxylase 65-kD isoform in the hippocampus and somatosensory cortex, suggesting that antiseizure and neuroprotective effects were related to increased GABAergic signalling. After a single subcutaneous dose of 20 μg/kg, NRP2945 inibited seizure activity in the GAERS model of absence epilepsy [43]. The reduction in seizure activity in the GAERS persisted for at least 10 days following a 28-day treatment with NRP2945; an effect that was was interpreted as suggestive of a disease-modifying effect [43]. An antiepileptogenic effect has also been reported in the pilocarpine rat model of temporal lobe epilepsy. In this model, treatment with NRP2945 (20 μg/kg, subcutaneously) for 7 days starting 24 h after cessation of pilocarpine-induced status epilepticus reduced the frequency of spontaneous recurrent seizures, but had no effect on seizure frequency when treatment was given after epilepsy had already developed [44]. NRP2945 has also been reported to have activity in models of other diseases, including motor neuron disease, peripheral neuropathy, multiple sclerosis and spinal cord injury, but details of these studies are not available [45].

Clinical studies of NRP2945 have not been published to date. Based on information listed in the sponsor’s website [45], multiple daily doses up to 25 μg/kg were well tolerated in healthy subjects over a 28-day treatment period (one subcutaneous bolus every 48 h). The half-life of NRP2945 was 22–25 min, with Cmax values of 1–1.5 ng/mL and no accumulation over time. An unspecified blood-based biomarker indicated target engagement in the brain. A phase IIa, placebo-controlled, adjunctive-therapy, cross-over trial of NRP2945 has been conducted in Melbourne, Australia, in patients with drug-resistant epilepsy and ‘absence epilepsy EEG patterns’ [45]. The trial apparently provided evidence of a favourable impact of the peptide on seizure activity, but the information provided in the sponsor’s website was insufficient to evaluate the data meaningfully. Further clinical trials are being planned in patients with Lennox–Gastaut syndrome and other types of epilepsy [45].

2.6 NRTX-1001

NRTX-1001 (previously known as NTX-001) is a cryopreserved product that comprises GABAergic post-mitotic interneurons of a specific pallial-type lineage derived from human pluripotent stem cells [46, 47]. The interneurons are designed for direct implantation into the epileptogenic tissue in order to permit release of GABA and suppress seizure activity at the site of the epileptic focus [48].

In the kainate mouse model of chronic mesiotemporal seizures, a single intrahippocampal administration of NRTX-1001 has been found to reduce or suppress electrographic seizures persistently without adverse behavioural effects [47,48,49]. Approximately 75% of epileptic mice treated with NRTX-1001 achieved stable seizure freedom compared with 8% of animals in the control group. Biodistribution studies showed that cell persistence was restricted to the hippocampus [48,49,50]. Metabolic abnormalities identified by magnetic resonance spectroscopy in the epileptic hippocampus partially normalised after administration of NRTX-1001 [50]. The antiseizure effect, which was consistent across different cell lots and studies, was associated with reduced hippocampal neurodegeneration and granule cell dispersion, without evidence of the appearance of ectopic tissues, tumours or teratomas at a histopathological examination 7.5 months after implantation [45, 47]. Preclinical safety studies included magnetic resonance imaging-guided intrahippocampal delivery in immunosuppressed non-human primates using a clinical-grade stereotactic delivery system, and found persistence of NRTX-1001 for at least 3 months post-implantation [50].

NRTX-1001 has recently entered clinical development in patients with drug-resistant unilateral mesial temporal lobe epilepsy. The first-in-human study (NCT05135091) includes an open-label dose escalation in up to ten patients, followed by a randomised trial with inclusion of a sham control group to evaluate safety and efficacy in up to 30 patients [47, 50]. The first two patients treated have shown encouraging results, with seizure counts reduced by > 90% at 6 months and 3 months after implantation, respectively [51].

2.7 STK-001

Dravet syndrome results from a pathogenic SCN1A gene variant that leads to impaired expression of functional voltage-gated type I sodium channels (Nav1.1) in inhibitory GABAergic interneurons [52]. Stimulation of expression of functional SCN1A gene protein in these interneurons to restore GABAergic inhibition provides a rational approach to counteract the manifestations of the disease. STK-001 is an antisense oligonucletide developed by applying the Targeted Augmentation of Nuclear Gene Output (TANGO) technology [53] to specifically activate the functional (wild-type) SCN1A allele in patients with Dravet syndrome, thereby increasing the levels of productive SCN1A mRNA transcript and boosting functional protein levels [20, 54]. Although STK-001 cannot be defined as a GABAergic agent per se, it is discussed here because ultimately its potential therapeutic action is dependent on its ability to improve the function of GABAergic interneurons.

After i.c.v. or intrathecal administration, STK-001 has been found to increase productive mRNA transcripts, and to upregulate brain Nav1.1 protein levels in mice, rats and non-human primates [20]. Intracerebroventricular STK-001 was also effective in increasing SCN1A mRNA levels and NaV1.1 protein expression in a mouse model of Dravet syndrome [55]. In the same model, these effects were associated with restored excitability of parvalbumin interneurons and reduced occurrence of electrographic seizures, together with a prolongation of survival [54, 55].

One phase I/II trial of single and multiple ascending doses of intrathecally administered STK-001 in patients with Dravet syndrome aged 2–18 years (NCT04442295) is ongoing in the USA, together with an associated extension study (NCT04740476). A multiple-dose study is also ongoing in the UK. [56]. The choice of the initial doses was based on a pharmacokinetic model developed with data from non-human primates and published data for animal-human scaling [57]. As of 21 February, 2022, 29 patients had received at least one dose at the 10-mg, 20-mg or 30-mg dose level, and no safety concerns related to the antisense nucleotide had emerged [58]. Dose-dependent increases in plasma STK-001 exposure were observed, and the antisense oligonucleotide was detected in cerebrospinal fluid samples up to 6 months after a single dose [58]. Although preliminary findings suggest an improvement in seizure frequency following STK-001 treatment [56], a full assessment of efficacy data has not been reported to date.

3 Drugs Under Clinical Investigation for Potential Repurposing in Epilepsy

3.1 Alprazolam

Alprazolam is a 1,4-benzodiazepine approved in the USA and other countries for the acute treatment of generalised anxiety disorder in adults, and for the treatment of panic disorder with or without agoraphobia in adults. Its actions are mediated by positive allosteric modulation of GABAA receptors. Like other benzodiazepines, alprazolam is active in animal models of genetic epilepsy and against seizures induced by electrical stimulation and various chemical convulsants [59,60,61,62,63]. It has not generally been used as an ASM, but its potential value in the treatment of seizure disorders has recently been considered as part of a broader effort to explore novel routes of administration for the suppression of prolonged or repetitive seizures.

Currently available rescue seizure treatments using the rectal, buccal or intranasal route utilise compounds such as midazolam and diazepam, which are established ASMs [64]. Alprazolam is being repurposed as an ASM for use by the inhalation route. The Staccato® device delivers alprazolam as an aerosol that, via normal breathing, reaches the lung for rapid uptake into the systemic circulation [65]. In a placebo-controlled, proof-of-concept study in five individuals with photosensitive idiopathic generalised epilepsy, Staccato® alprazolam (S-ALP) at doses of 0.5, 1.0 and 2.0 mg was effective in suppressing the photoparoxismal EEG response, with an onset of activity within 2 min after dosing and persistence of effect through 4 h at the lowest doses and at least 6 h at higher doses [65]. Treatment-emergent adverse events were mild to moderate, appeared to be dose dependent, and consisted in cough, dysgeusia, oral dysesthesia, sedation and somnolence. Mean (± standard deviation) plasma concentrations of alprazolam at 2 min after the 0.5-mg dose were 5.1 ± 2.4 ng/mL, and increased dose proportionally at higher doses. Visual inspection of plasma alprazolam concentration profiles suggested a biphasic decline, with a terminal phase consistent with the drug’s known half-life of 9–16 h [66].

A phase IIb, randomised, double-blind, placebo-controlled, parallel-group, in-patient trial of S-ALP (1.0 and 2.0 mg) has been completed in 163 adults with focal or generalised epilepsy and a predictable pattern of prolonged or repetitive seizures [67]. The primary endpoint was the proportion of responders (response rate), defined as those patients in whom seizure activity ceased within 2 min after administration of the study medication with no recurrence of seizure activity within 2 h. The response rate was identical (65.8%) at S-ALP doses of 1.0 and 2.0 mg, compared with 42.5% for placebo (p < 0.04 for each S-ALP dose vs placebo). Treatment-emergent adverse events were mostly mild to moderate and consisted mainly of cough and somnolence (both 14.5%) and dysgeusia (13.2%), with no clear dose dependency. Plasma alprazolam concentration–time profiles were similar across different seizure types. Overall, these data demonstrate that S-ALP is effective in terminating seizure activity rapidly. Of note, none of the patients had generalised tonic-clonic seizures or focal-to-bilateral-tonic-clonic seizures during the study, and therefore efficacy in terminating this seizure type cannot be inferred. The hypothesis that S-ALP may have a faster onset of action compared with other benzodiazepines given by alternative non-parenteral routes will also require formal head-to-head testing in an ad hoc study [67]. A phase III trial of S-ALP in patients 12 years of age and older with stereotypical prolonged seizures is ongoing (NCT05077904).

3.2 Bumetanide

The rationale for investigating bumetamide as a potential antiseizure drug stems from evidence that certain seizure disorders, most notably neonatal seizures, may be caused by an increased intracellular level of chloride ions (Cl) in cerebral neurons. Under these conditions, GABA-induced opening of chloride channels leads from an outflow of Cl from the cytoplasm and a consequent paradoxical depolarising (excitatory) response [68, 69]. The increase in the intracellular Cl level can result from upregulation of the sodium (Na+)-potassium (K+)-Cl- transporter isoform 1 (NKCC1), which is involved in Cl inflow, and/or downregulation of the K+-Cl-transporter isoform 2 (KCC2), which extrudes Cl from the cytoplasm [68, 69]. While other mechanisms can play a role in intracellular Cl homeosthasis [70], inhibitors of NKCC1 such as bumetanide have been considered as potential treatments to reduce intracellular Cl levels and correct the dysfunction in GABA signalling [71]. Of note, bumetanide inhibits not only NKKC1, but also Na+-K+-Cl-transporter isoform 2 (NKCC2), which is expressed in the kidney and other organs and plays a key role in the diuretic effects of bumetanide [72]. Several off target effects, some of which might contribute to antiseizure effects, have also been described, including inhibition of carbonic anhydrases, modulation of G protein-coupled receptor 35, inhibition of organic anion transporter 3 and other organic anion transporter transporters, and an interaction of unclear significance with the α3 subunit of the GABAA receptor [72].

Although bumetanide does show antiseizure activity in vitro and in vivo [71, 73], in most seizure models, including those intended to mimic neonatal seizures, its main effect consists of potentiating the antiseizure activity of phenobarbital [20, 68, 74, 75]. Preclinical evidence of seizure protection, however, has not been consistent [76, 77], possibly owing to its poor brain penetration [77]. Concerns have also been expressed about the implications of some of the off-target effects of bumetanide, as well as the consequences of NKCC1 inhibition at non-neuronal sites such as glial cells, the choroid plexus, endocrine cells and immune cells [20]. As a result, the potential value of bumetanide as an ASM, particularly for the treatment of neonatal seizures, has been a subject of intense debate [71, 72, 77, 78].

The results of two recent clinical studies conducted to date have contributed to the controversy. The first of these studies was an open-label, dose-finding, feasibility trial designed to assess the pharmacokinetics, efficacy and safety of bumetanide (0.05–0.3 mg/kg) added on to phenobarbital in full-term neonates with hypoxic ischaemic encephalopathy and electrographic seizures [79]. The trial was stopped prematurely after the enrolment of 14 participants because of the limited evidence of seizure reduction, and the occurrence of hearing impairment in three of 11 surviving infants. However, two of the three infants with hearing impairments had also received aminoglycosides, the ototoxic potential of which can be increased by bumetanide [71]. The second study used a randomised, double-blind, placebo-controlled, dose-escalation design in neonates with an electrographic seizure occurring at least 30 min after a loading dose of phenobarbital (≥ 20 and < 40 mg/kg) [80]. Bumetanide 0.1–0.3 mg/kg (treated group, n = 27) or placebo (control group, n = 16) was administered in conjunction with phenobarbital 5–10 mg/kg, with continuous EEG monitoring from ≥ 2 h before (baseline) to ≥ 48 h after study drug administration (post-treatment). The primary endpoints of the study were the pharmacokinetics and safety of bumetanide, and electrographic seizure burden was included as an exploratory endpoint. Bumetanide was found to have a median half-life of 16.0 h and a median clearance of 0.10 mL/min/kg, with large individual variability. Some of the neonates were also treated with hypothermia, which was associated with lower bumetanide clearance values compared with neonates not exposed to hypothermia. Diuresis as an adverse event was recorded more often in the treated group than in the control group (48% vs 13%, p = 0.02). Four neonates died, one in the treated group and three in the control group. Among survivors, hearing impairment was recorded in 2 of 26 treated neonates, both of whom also received aminoglycosides, versus 0 of 13 controls. After adjustment for seizure burden (which differed markedly between groups), the decrease from baseline in seizure burden from 0 to 4 h and from 2 to 4 h post-treatment was significantly greater among the treated neonates than among controls. Overall, the findings were considered to provide reassuring safety data, and a strong signal of antiseizure efficacy [71]. As mentioned above, the rationale for continuing the clinical development of bumetanide in neonatal seizures is currently being debated [71, 72, 77, 78].

Of note, a bumetanide analogue (NPT 2042), ultimately intended for use as ‘adjunct antiseizure treatment for patients with medically intractable epilepsy’, has recently entered clinical development (NCT05503511), but no information on its structure, pharmacological properties and rationale for development appears to be available [81]. Another compound, IAMA-6, a putatively selective NKCC1 inhibitor targeting epilepsy and other central nervous system disorders as potential indications, is currently in preclinical development [82].

3.3 Ivermectin

Ivermectin is an anthelmintic that exerts its action against parasites by activating glutamate-gated Cl channels, which are absent in vertebrates [83]. Invermectin, however, also acts as an activator of GABAA receptors by binding to at least two different binding sites distinct from the benzodiazepine binding site. At low (nanomolar) concentrations in vitro, ivermectin enhances GABAA receptor-mediated responses to GABA, and also increases benzodiazepine binding to GABAA receptors, while at higher concentrations it causes a direct activation of the GABAA receptor [84, 85]. As recently reviewed by Löscher [85], ivermectin has many additional actions, including activation of glycine receptors, α7 nicotinic cholinergic receptors and G-protein-gated inwardly rectifying K+ channels. It also acts as a positive allosteric modulator of purinergic adenosine triphosphate-gated P2X4 receptors, as a ligand of farnesoid X receptors [85] and, possibly, as an activator of ATP-sensitive potassium channels and a modulator of the opioidergic system and nitric oxide pathway [86, 87]. Finally, ivermectin has been reported to exert anti-inflammatory actions through a variety of mechanisms [88, 89] and, in particular, to inhibit neuroinflammation in a murine model of experimental autoimmune encephalomyelitis through regulation of T-cell proliferation and function [89]. Some of these properties, particularly those related to inhibition of inflammation, may have implications for seizure control, even though based on available information, the antiseizure effects of ivermectin can be mainly ascribed to actions on the GABAergic system [85, 90, 91].

Interest in evaluating potential effects of ivermectin on seizure susceptibility was stimulated initially by the discovery of its interactions with the GABA system [92]. Antiseizure effects of the drug were reported as early as 1986 in a genetic avian model of photosensitive epilepsy [92]. Since then, protective activity against electrically and chemically induced seizures has been reported in various rodent models [86, 87, 91, 93, 94], including the 6-Hz model in mice and hippocampal kindling in rats [85]. In these models, however, antiseizure effects are generally seen at doses that produce toxicity, which together with other considerations led Löscher [85] to advise against clinical development of the drug for the treatment of epilepsy. Although there have been reports of reduced seizure frequency after ivermectin treatment in patients with onchocerciasis-associated epilepsy, the improvement in these patients may have simply reflected an indirect effect secondary to reduced microfilarial load in the central nervous system [95,96,97]. There has also been a report of improved seizure control in a small group of patients with drug-resistant mostly focal epilepsy treated with ivermectin (10 mg/day once daily three or seven times a week) added on to pre-existing ASMs [98]. Because of the lack of a control group and other methodological limitations, however, the findings of the latter study are difficult to interpret.

After oral administration, peak plasma ivermectin concentrations are usually attained at about 4 h after dosing. The drug is cleared primarily by cytochrome P450 3A-mediated metabolism, with a mean half-life of about 18 h [99]. Ivermectin shows poor penetration across the blood–brain barrier because it is a substrate of the efflux transport P-glycoprotein, which limits access of many molecules that are P-glycoprotein substrates to the brain [85]. Although ivermectin is generally well tolerated at the low doses used in the treatment of onchocerciasis (approximately 150 µg/kg body weight as a single oral dose) [99], concerns have been expressed about potential neurotoxic effects (especially in patients with a dysfunctional blood–brain barrier) at the higher doses (up to 60 mg/day) and prolonged duration of treatment that are currently being tested in patients with epilepsy [85].

A capsule formulation of ivermectin (EQU-001) is currently in development for the treatment of various neurological conditions, including drug-resistant epilepsy [85, 100]. A 12-week, phase II, randomised, double-blind, adjunctive-therapy, placebo-controlled, safety, tolerability and exploratory efficacy study (NCT05063877) has been completed recently. The trial evaluated EQU-001 doses of 10, 20, 40 and 60 mg, administered once daily, in a small group of patients (n = 43) with uncontrolled, mostly focal seizures. According to a press release from the sponsor [100], EQU-001 was found to be safe well tolerated, and no treatment-related serious adverse events were reported. Neurological and psychological adverse events occurred in less than 10% of participants and were similar in the EQU-001 and placebo arms. There was a trend for seizure frequency to improve with increasing EQU-001 doses (after excluding the 40-mg dose), but the study was not powered for efficacy. A reduction in inflammatory biomarkers in the circulation was identified in the 60-mg dose group [100]. A 16-week, randomised, double-blind, placebo-controlled trial (NCT05473442) to assess the antiseizure efficacy of EQU-001 at maintenance doses of 20 and 60 mg/day in adults with uncontrolled focal seizures is currently ongoing.

4 Summary and Conclusions

Although many of the currently marketed ASMs act on the GABA system (Table 1), only three of those, namely vigabatrin, tiagabine and ganaxolone, were rationally designed to exert a GABAergic effect. A fourth rationally designed compound, progabide, was developed in the 1980s as a GABA prodrug but never became established because of disappointing clinical efficacy results and the propensity to cause liver toxicity [101,102,103,104]. Gabapentin was developed as a GABA derivative but it soon became clear that, although it can allosterically modulate GABAA receptors, its primary mode of action is not via GABAergic mechanisms but through interference with voltage-gated calcium channels [105]. All other ASMs listed in Table 1 were discovered through random screening or serendipitous observations, and their interactions with the GABA system became apparent only when their development was already advanced [5].

The last decade saw a renewed interest in targeting the GABA system for the treatment of epilepsy. As discussed in the previous sections of this article, several treatments are now in development that are specifically intended to target GABAergic transmission. A review of their modes of action and pursued indications reveals a remarkable heterogeneity in objectives and approaches (Table 2), which may allow the broad categorisation of these treatments into three groups.

Table 2 Mechanisms of action and proposed indications of GABA-targeting investigational treatments in clinical development for seizure disorders

The first group consists of positive allosteric modulators of GABAA receptors. One of these, S-ALP, is a benzodiazepine not used in the past as an ASM, and being now repurposed as a rescue medication for the treatment of prolonged and repetitive seizures. Utilisation of a benzodiazepine for this indication is not a novelty, but the mode of administration of S-ALP by inhalation does represent a first in epilepsy treatment. Whether S-ALP represents an advance over other benzodiazepines currently used for the same indication remains to be determined. It is also unclear whether the inhalation route is applicable to individuals with severe seizure types such as generalised or focal-to-bilateral tonic-clonic seizures. Another set of compounds in this category consists of subunit-selective allosteric modulators. The rationale for developing these drugs, which include the α2/3/5 subtype-selective agents darigabat and ENX-101, builds on evidence that subunit selectivity and, in particular, avoidance of α1 subunit activation, may retain some favourable effects of benzodiazepines, including their antiseizure action, while avoiding or minimising undesirable effects such as cognitive dysfunction, somnolence, tolerance and abuse liability [14, 19, 106,107,108,109,110,111]. Other subunit-selective GABAA receptor allosteric modulators that target seizures as their primary indication have been described, including KRM-II-81 [109, 110] and SAN2219 [112], but to our knowledge they are not yet in clinical development. Among subtype-selective agents, darigabat is the most advanced in development, and has shown promising antiseizure activity in a proof-of-concept trial in photosensitive epilepsy [25]. Larger scale studies, however, are needed to determine whether the promise of subunit-selective GABAA receptor allosteric modulators being superior to benzodiazepines for the maintenance treatment of epilepsy will be fulfilled in the clinical setting. A different approach to GABAA receptor allosteric modulation is the targeting of the neurosteroid modulatory site. ETX-155 is described as a ‘next-generation’ orally active neurosteroid that enhances GABAergic responses at synaptic and extrasynaptic GABAA receptors [38]. Publicly accessible information on this compound is very limited, and it is unclear how it differs from other neurosteroids such as ganaxolone [6]. This question is especially relevant because ETX-155 is apparently being targeted for the treatment of focal epilepsy [38], an indication that was also pursued with ganaxolone with disappointing results [6, 113]. Similar considerations apply to LPCN 2101, another orally administered neurosteroid scheduled for a proof-of-concept study in patients with photosensitive epilepsy, although the indication to be pursued for this compound are as yet unclear [114, 115].

A second group of investigational compounds consists of two marketed non-central nervous system drugs, namely bumetanide and ivermectin, that have been considered for repurposing in the treatment of seizure disorders. These agents differ in pharmacological properties as well as indications being pursued. Bumetanide is a NKCC1 inhibitor investigated for the treatment of phenobarbital-resistant neonatal seizures based on evidence that increased intracellular levels of chloride in the immature brain can result in GABA having a paradoxical depolarising effect. Ivermectin, in contrast, has a very complex pharmacology. It has many other mechanisms of action in addition to those involving the GABA system and is being investigated in epilepsy mainly based on the results of preclinical studies in experimental seizure models. One common feature of bumetanide and ivermectin is that the rationale for pursuing their clinical development in seizure disorders has come recently under criticism [71, 72, 77, 85]. Apparently, no additional clinical studies are currently scheduled for bumetanide in neonatal seizures, whereas a randomised controlled trial of ivermectin in patients with uncontrolled focal seizures is ongoing (NCT05473442).

The third and most innovative group of treatments includes cell and gene therapies, as well as treatments to regulate gene expression. Intracerebral implantation of GABAergic interneurons (NRTX-001) is being evaluated in patients with mesial temporal lobe epilepsy, whereas an antisense oligonucleotide (STK-001) to restore the function of GABAergic interneurons by upregulating NaV1.1 sodium channels is under investigation in patients with Dravet syndrome. A clinical trial of gene therapy using an adenoviral vector (ETX-101) is scheduled for initiation. Some of these treatments could theoretically prove to be game changers, but it would be premature to speculate on their value prior to completion of safety and efficacy studies. The same applies to NRP2945, a subcutaneously administered neuroactive peptide that reportedly upregulates the expression of GABAA receptor α and β subunits.

Overall, the current pipeline of investigational treatments demonstrates that targeting the GABA system remains an attractive approach in epilepsy drug development. Although this article focused on investigational treatments for which clinical data are available, or likely to be available in the short term, preclinical research in this area is also very active. Drug candidates being investigated preclinically include, among others, novel GABAA receptor agonists and allosteric modulators [14, 33, 111], compounds targeting GABAB receptors [116, 117], a highly potent inhibitor of GABA transaminase [118], a new class of selective NKCC1 inhibitors [82] and novel GABA-targeting viral vectors [119]. At the other extreme of the development spectrum, clinical studies are ongoing to assess novel applications of already marketed GABA-targeting ASMs. Notable examples include the investigation of vigabatrin as an antiepileptogenic and disease-modifying agent in infants with tuberous sclerosis complex [120], and the evaluation of low-dose valproic acid delivered by continuous i.c.v. infusion as a long-term treatment for patients with drug-resistant epilepsy [121, 122].