You are on page 1of 34

pubs.acs.

org/jmc Perspective

Advances in Epilepsy: Mechanisms, Clinical Trials, and Drug


Therapies
Tingting Hu, Jifa Zhang, Jiaxing Wang, Leihao Sha, Yilin Xia, Tyler C. Ortyl, Xiaohe Tian, and Lei Chen*

Cite This: J. Med. Chem. 2023, 66, 4434−4467 Read Online

ACCESS Metrics & More Article Recommendations


See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

ABSTRACT: Epilepsy is a common disease of the nervous system


characterized by transient brain dysfunction caused by an abnormal
electrical discharge from the brain neurons. The pathogenesis of epilepsy
Downloaded via ZHEJIANG UNIV on February 27, 2024 at 14:02:48 (UTC).

is complex and remains elusive. Nowadays, drug therapy is the mainstay


method for the treatment of epilepsy. More than 30 antiseizure drugs
(ASDs) were approved for clinical use. Unfortunately, about 30% of
patients still display pharmacoresistance against ASDs. The long-term use
of ASDs may cause adverse effects, raise tolerability concerns, bring
unexpected drug interactions, generate withdrawal symptoms, and
increase the economic burden. Thus, the research uncovering more
effective ASDs that are safe is still a difficult and urgent task. In this
Perspective, we describe the pathogenesis, clinical trials, and drug therapy
progress of epilepsy, focusing on summarizing the current situation of
small-molecule drug candidates progressing in epilepsy therapy, which provides future directions for the development of more
promising ASDs.

1. INTRODUCTION specific drug-resistant focal epilepsy by removing the area of


Epilepsy is a chronic neurological disease caused by an the seizure focus. However, it is hurdled by the risks and
abnormal discharge of brain neurons. Spontaneous recurrent postoperative complications.7 Nowadays, drug therapy is the
seizures (SRS) are the hallmark of epilepsy.1 The main clinical mainstay method for treating epilepsy, which can effectively
symptoms of epilepsy include convulsions, loss of conscious- control seizures.8 More than 30 antiseizure drugs (ASDs, also
ness, myoclonus, hypotonia, and prolonged duration of muscle referred to as anticonvulsants and antiepileptic medications)
contraction.2 Based on the 2017 International League Against have been approved for clinical use. New ASDs such as
Epilepsy (ILAE) latest definition, epilepsy is mainly divided brivaracetam, cannabidiol, cenobamate, everolimus, and
into focal epilepsy, generalized epilepsy, combined generalized fenfluramine have high efficacy, few adverse reactions, and
and focal epilepsy, and unknown epilepsy.2 Epilepsy, with a
low drug resistance. However, long-term efficacy and safety
worldwide prevalence of 0.5−1.0%, can occur in people of any
age, region, and race, but it is more common in children and need to be further verified.9 Unfortunately, about 30% of
adolescents.3 Etiology of epilepsy can be due to genetics, patients respond poorly to existing ASDs that may develop into
acquired brain injury, infection, and metabolic or immune treatment-resistant epilepsy.10 Therefore, more effective ASDs,
disorders.2 The pathogenesis of epilepsy has not been particularly with reduced side effects and novel mechanisms of
completely elucidated and is considered closely related to action, are still in high demand for patients with DRE.
ion channels, neurotransmitter imbalance, inflammation, and Herein, we summarize the pathogenesis, clinical trials, and
immune abnormalities.4,5 Therefore, strengthening the explo- drug therapy progress of epilepsy, focusing on small-molecule
ration of the pathogenesis of epilepsy will help provide the compounds with potential antiepileptic effects developed in
necessary theoretical basis for its diagnosis, prevention, and the past five years.
treatment.
Drug therapy, surgery, and ketogenic diets are the classic
methods for treating of epilepsy. The ketogenic diets refer to Received: December 2, 2022
high-fat, low-carbohydrate, and moderate-protein diets, which Published: April 4, 2023
have been used for decades to treat drug-resistant epilepsy
(DRE). Although the antiepileptic mechanism is still unclear,
its efficacy and safety have reached a consensus.6 Epilepsy
surgery can achieve long-term seizure freedom in patients with

© 2023 American Chemical Society https://doi.org/10.1021/acs.jmedchem.2c01975


4434 J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 1. Diagram for the pathogenesis of epilepsy, including abnormal ion channel function, imbalance of neurotransmitter secretion, irregular
glial system, neuroinflammation and oxidative stress, and abnormal neural circuits.

2. THE PATHOGENESIS OF EPILEPSY including SCN1A, SCN2A, SCN3A, SCN8A, and SCN1B
Although the pathogenesis of epilepsy is complex, researchers are associated with a spectrum of epilepsy phenotypes. SCN1A
are gradually unraveling its mystery. Seizures are due to an is the most common epilepsy gene identified to date, NaV1.1
imbalance of excitatory and inhibitory neuronal firing, which is encoded by SCN1A is highly expressed in inhibitory neurons.4
closely related to abnormal ion channel function, imbalance of SCN1A-related seizure disorders encompass simple febrile
neurotransmitter secretion, irregular glial system, neuro- seizures (FS), generalized epilepsy with febrile seizures plus
inflammation and oxidative stress, and abnormal neural circuits (GEFS+), Dravet syndrome (DS), and early infantile epileptic
(Figure 1). A more precise understanding of resistance encephalopathy (EIEE). NaV1.2 (SCN2A) and NaV1.6
mechanisms may lead to the development of more effective (SCN8A) show the highest expression in excitatory neurons.13
and tolerated drugs. SCN2A has emerged as one of the most prominent epilepsy
2.1. Abnormal Ion Channel Function. Ion channels, genes broadly associated with seizure and neurodevelopmental
including voltage-gated sodium channels (VGSCs), voltage-
phenotypes.14 Mutations in the SCN8A cause catastrophic
gated potassium channels (VGPCs), and voltage-gated calcium
EIEE13 and SCN8A developmental and epileptic encephalop-
channels (VGCCs), are the structural basis of neuronal
signaling. Mutations in genes encoding these ion channels athy (SCN8A-DEE), which are also associated with intractable
will lead to changes in ion channel functions, resulting in epilepsy.15 A few pathogenic SCN3A variants have been
genetic epilepsy.11,12 reported in familial focal epilepsy with variable foci, as well as
2.1.1. Voltage-Gated Sodium Channels. VGSCs play a in patients with early onset DEEs.16 SCN1B encodes sodium
crucial role in generating and transmitting cellular action channel β subunit (NaVβ1), which is enriched in the neuronal
potentials. Pathogenic variants in sodium channel genes axon initiation segment and node of Ranvier.17 SCN1B
4435 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

variants are associated with severe DEEs resembling DS and transmission has been shown to inhibit or attenuate seizures.37
GEFS+.18 GABA is mainly synthesized at axonal terminals, which is
2.1.2. Voltage-Gated Potassium Channels. VGPCs are formed by decarboxylate of glutamate precursor under the
necessary in the maintenance of the cell resting membrane action of glutamic acid decarboxylase (GAD). Synthetic GABA
potential, regulating action potential discharge, and releasing is released from vesicles into the synaptic cleft via vesicular
neurotransmitters. A variety of seizure-related potassium GABA transporters after neuron depolarization. Then GABA
channel mutations have been found, such as KCNQ2, can bind to receptors (GABA-A, GABA-B, and GABA-C
KCNQ3, KCNA1, KCNA2, KCNB1, KCNC1, KCNMA1, receptors) on the postsynaptic membrane to mediate
KCNT1, and KCTD7.11 KV7 (KCNQ channel) is the seventh excitatory transmission. Also, GABA can be retaken to
subfamily of VGPCs, which consists of five members (KV7.1− presynaptic neurons by GABA transporters (GATs) or
7.5) that each have a different tissue distribution and transported to glial cells, reducing its concentration in the
physiological function.19 KCNQ1 is restricted to cardiac synaptic cleft, thus terminating the inhibitory synaptic
tissues,20 and KCNQ2−5 are mainly expressed in the central transmission of GABAergic. GABA is ultimately catabolic to
and peripheral nervous systems. Therefore, they are often succinic acid semialdehyde mediated by GABA amino-
referred to as neuronal KCNQs. The KCNQ2/3 hetero- transferase (GABA-AT). Clinical studies have demonstrated
tetramer assembled by KCNQ2 and KCNQ3 subunits is the that drugs enhancing GABA-mediated inhibitory synaptic
main molecular entity of M current, which can stabilize the transmission are effective in treating epilepsy.38,39 GABA
resting membrane potential and limit the repetitive firing of receptors (GABAR), GABA-AT, and GATs have become
neurons.21 It has been confirmed that mutation-induced important targets for the development of ASDs recently.40−42
reduction of KV7.2 subunit stability or changes in M current 2.2.2. Glutamate. Glutamate, the most abundant amino
gating may lead to the development of neonatal epilepsy.22 acid in the brain, is transmitted between astrocytes and glial
Therefore, neuronal KCNQ2/3 channels are important targets cells to achieve rapid signaling transduction. It plays an
for treating diseases such as epilepsy and chronic pain.23 important role in neuronal excitability.43 Four glutamate
KCNT1 encodes the sodium-activated potassium channel receptors have been identified: ionotropic glutamate receptors
KNa1.1 (Slack, Slo2.2), which is widely expressed in the CNS.24 (iGluR), including mammalian α-amino-3-hydroxy-5-methyl-
Gain-of-function (GOF) mutations in the KCNT1 gene are 4-isoxazole propionic acid receptors (AMPAR), kainate
associated with DREs, including nocturnal frontal lobe epilepsy receptors (KAR) and N-methyl- D -aspartate receptors
and epilepsy of infancy with migrating focal seizures.25 (NMDAR), and metabotropic glutamate receptor
2.1.3. Voltage-Gated Calcium Channels. VGCCs that (mGluR).44 Studies have shown that overstimulation of
involve in the release of presynaptic transmitters, the glutamate receptors will lead to glutamate accumulation,
synchronous firing of neurons, and paroxysmal depolarization promoting the occurrence and development of epilepsy.45
drift, are widely distributed in various tissues and cells.26 The GluA2 subunit of AMPAR is selectively permeable to Na+
VGCCs can be classified as high-voltage activation (L, P/Q, and prevents Ca2+ influx. AMPAR blockers can significantly
and N-type) or low-voltage activation (T-type) based on the reduce or inhibit the occurrence of epilepsy. 46 The
range of membrane potentials at which the channel becomes heteromeric GluK2/GluK5 of KARs have been shown to
active.27 L-Type channels consist of the α1 subunit from the significantly contribute to epileptic relapses in chronic
CaV1 family, while P/Q and N-type channels consist of CaV2.1 epilepsy.47 NMDAR is primarily permeable to Ca2+ and
and CaV2.2 α1 subunits, respectively.28 Low-voltage activated important for seizure activity.48 Missense mutations in genes
T-type calcium channel (TTCC), mainly including CAC- encoding NMDAR have been reported to cause early onset
NA1G (Ca3.1), CACNA1H (Ca3.2), and CACNA1I (Ca3.3), epilepsy and intellectual disability.49
has been studied the most.29 The mice overexpressing CaV3.1 2.3. Irregular Glial System. The glial cells in CNS mainly
showed frequent spike discharges (a sign of absence seizure), include astrocytes, microglia, and oligodendrocytes.50 In recent
while knockout mice lacking CaV3.1 were protected from years, it has been found that abnormal glial cells can change the
absence seizure.30 Mutations in CACNA1H (CaV3.2) have microenvironment by altering the neurotransmitters, regulating
been identified in patients with idiopathic generalized epilepsy the switch of ion channels, promoting the release of
(IGE).31 CACNA1A (CaV2.1) is a susceptible gene for the inflammatory factors, and changing the nerve myelin sheath,
absence of epilepsy. Abnormal CaV2.1 channel will affect contributing to epilepsy progression. Astrocytes are abundantly
membrane excitability, calcium influx-dependent neurotrans- distributed in the surrounding neurons, which play a
mitter release, and abnormal oscillation rhythm of the loop, supporting and nutritional role. Astrocyte dysfunction destroys
eventually leading to the absence of seizure. 32 The the function of membrane protein (EAAT2), which is
pharmacological studies in GAERS or WAG/Rij genetic responsible for glutamate uptake. A large amount of glutamate
models have shown that T-type calcium channel blockers accumulates outside the cell, resulting in increased neuronal
significantly reduced epileptic activity.33,34 excitability, which induces epilepsy.45 Astrocytes are also major
2.2. Imbalance of Neurotransmitter Secretion. Neuro- regulators of K+ and aquaporins.51 Astrocyte dysfunction
transmitters are endogenous chemicals that are typically stored impairs the uptake of potassium (Kir4.1) and changes the
in synaptic vesicles and axon terminals and released into the intracellular and extracellular osmotic pressure (AQP4).52
synaptic cleft during signal transduction. They modulate Seizures activate many astrocytes, and those astrocytes release
excitatory/inhibitory neuronal function by binding to their inflammatory factors that further aggravate seizures.53 Micro-
receptors.35 The imbalance of γ-aminobutyric acid (GABA) glia are macrophages in CNS and play an important role in
and glutamate secretion in neurons is closely related to immune functions. Under stress or pathological conditions,
seizures.36 excessive activation of microglia can produce oxygen free
2.2.1. GABA. GABA is a major inhibitory neurotransmitter radicals and cytokines, resulting in neuronal apoptosis and
in the mammalian CNS. Enhancement of GABAergic neuro- recruitment of immune cells to attack the blood−brain barrier
4436 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 2. Diagram for the pathogenesis of DRE, including the transporter hypothesis, target hypothesis, gene variation hypothesis, intrinsic severity
hypothesis, neural network hypothesis, and pharmacokinetic hypothesis.

(BBB).54 Oligodendrocytes are the only cells that produce the prostaglandin-E2 (PG-E2) axis, and activated intracellular
nerve myelin sheath that allows neuronal cells to perform their signaling pathways.58 Inflammatory mediators can promote the
conduction function. Studies have found that changes in nerve transcriptional activation of autocrine and/or paracrine
myelin can also cause epilepsy.55 inflammatory genes. They can also directly activate homolo-
2.4. Neuroinflammation and Oxidative Stress. The gous receptors expressed on neurons of epileptogenic tissues,
destruction of the BBB emerges when inflammatory responses which can alter the transcriptional activity of glutamate, GABA
in the peripheral and central nervous systems become receptors, and other ion channels, thereby increasing neuronal
activated, ultimately caused by infections, trauma, or tumors excitability.59 After seizures or during recurrent seizures,
within the brain. The damage of BBB can cause the endogenous IL-1β and HMGB1 release are increased, which
extravasation of serum lymphocytes, immunoglobulins, and can bind to IL-1β and TLR4 receptors to activate downstream
albumin into the CNS. Subsequently, this can cause the intracellular signaling molecules and finally induce the
activation and proliferation of glial cells, which enhances the transcription of inflammatory genes regulated by nuclear
excitability of astrocyte nerves and eventually leads to factor-κ B (NF-κB).60 In epilepsy patients and animal models,
seizures.56 Studies have shown that the ABC efflux transporters the COX-2 level in brain is up-regulated, which can induce the
(P-gp, BCRP, MRP) in vascular endothelial cells can effectively conversion of arachidonic acid to PGs. Therefore, inhibition of
inhibit therapeutic drugs through BBB, leading to the COX-2 or blockade of the PG receptor can protect neurons
occurrence of refractory epilepsy and DRE.57 and inhibit seizure.61 Recent studies have shown that albumin
Neuroinflammation is associated with inflammatory media- generated by BBB damage can penetrate the intercellular space
tors released from neurons, glial cells, and BBB endothelial in the brain, then activate TGF-β signaling pathway changes in
cells, including interleukin-1β(L-1β), high-mobility group box astrocytes and destroy the potassium buffering capacity and
1(HMGB1), tumor necrosis factor α (TNF-α), transforming glutamate metabolism, causing an increase in nerve excitability
growth factor β (TNF-β), cyclooxygenases-2 (COX-2)− and lowers the seizure threshold.62 Studies have suggested that
4437 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

a profound interaction between inflammation and epilepsy,63 hypothesis are currently controversial, and further studies are
which indicates blocking unwanted inflammatory signals in the needed to determine the clinical relevance of efflux transporter
brain may provide new strategies for treating epilepsy. overexpression at the BBB.72 The target hypothesis postulates
Oxidative stress (OS) is oxidative damage caused by the that acquired alterations of ASD targets at molecular levels,
production of reactive oxygen species (ROS) and reactive such as changes in voltage-gated ion channels, neuro-
nitrogen species (RNS) due to mitochondrial dysfunction, transmitter receptors, transporters, or metabolic enzymes,
increased activities of NADPH oxidase (NOX), xanthine lead to decreased drug sensitivity to treatment.78 Remy et al.
oxidase, and inducible nitric oxide synthase (iNOS). Free showed that loss of use-dependent blockade of voltage-gated
radicals produced by seizures can lead to OS damage and Na+ channels by carbamazepine was observed in patients with
increased the ratio of OS markers, such as iNOS oxidized and carbamazepine-resistant TLE.79 In the pilocarpine model of
reduced glutathione can be detected in patients with TLE.64 epilepsy, the changes in GABAA receptor makes patients
Endoplasmic reticulum stress (ERS) is a condition that resistant to ASDs acting through this receptor.80 Changes in
various physical and chemical factors (such as oxidative stress, the GABAA receptor reducing drug sensitivity in patients with
mutant protein expression, and abnormal Ca+ concentration) refractory TLE have also been reported.81 The evidence of this
disrupt ER function and lead to unfolded or misfolded proteins hypothesis is limited and needs to be further studied and
accumulating in the ER lumen.65 Increasing studies have verified. One of the most faithful hypotheses, and possibly the
suggested that abnormal ER stress may participate in various most basic cause of resistance, is the genetic variation
pathological processes associated with epilepsy.66 hypothesis, which states that variations in genes, encoding
2.5. Abnormal Neural Circuits. The pathogenesis of ion channels, and neurotransmitter receptors, or enzymes that
epilepsy is related to the enhancement of neural network metabolize ASDs, associated with ASD pharmacokinetics, and
excitability caused by abnormal synaptic connections between pharmacodynamics cause inherent pharmacoresistance.82 The
brain neurons and the establishment of pathological neural polymorphism of the SCN1A gene may affect the con-
circuits. Recent studies have found that factors containing formation of the encoded protein or stability of post-
long-term potentiation (LTP), mossy fiber sprouting (MFS), transcriptional miRNAs and results in the reduced response
neuropeptide Y (NPY), neural cell adhesion molecule of ASDs to sodium channel blockade, ultimately leading to
(NCAM), and mGluRs are involved in the occurrence and drug resistance.83 Some studies have found a strong association
development of epilepsy and the plasticity of the neuronal between CYP2C9 polymorphism and phenytoin dose require-
synapse. Seizures can induce LTP production at glutamatergic ment.83,84 However, this hypothesis is limited by incon-
synapses in epileptic regions, which leads to persistent sistencies and poor reproducibility of study findings, more
remodeling between synapses.67 A variety of axon-guiding powerful and repeatable pharmacogenomics research is needed
molecules, such as semaphorins and Eprins, affect the normal to prove it. The intrinsic severity hypothesis states that drug
growth of axons and ultimately lead to abnormal neuronal resistance is related to the severity of epilepsy.85 The frequency
networks in seizures.68 MFS is the main pathological change of of seizures is considered to be an important indicator of the
temporal lobe epilepsy. It has been found that MFS projects to disease severity.86 Data supporting this hypothesis suggest that
glutamatergic interneurons of the dentate gyrus, leading to patients with more than 10 seizures before initiation of therapy
increased excitability of the dentate gyrus and transmission to were more than twice as likely to develop refractory epilepsy.87
the hippocampus in epileptic animal models.69 In addition, However, Musicco et al. reported that patients treated after the
NPYs are released in the hippocampus to bind the receptors in first seizure and those who received treatment after seizure
granulosa cells, which can promote mossy fiber budding and recurrence had the same probability of achieving 1 and 2
synaptic reconstruction in seizures.70 seizure-free years.88 A randomized controlled trial found that
2.6. Drug-Resistant Epilepsy. The mechanism of DRE immediate ASD treatment can reduce seizure occurrence in
likely involves multifactors, including environment, genetics, as the next 1−2 years, but there was no difference in the long
well as factors related to disease and drugs.71 Several term remission in individuals with single or infrequent
hypotheses have been proposed, including the transporter seizures.89 There is little evidence to prove a direct connection
hypothesis, target hypothesis, gene variation hypothesis, between the severity of epilepsy and ASD response. Therefore,
intrinsic severity hypothesis, neural network hypothesis, and drug resistance in epilepsy cannot be fully explained by this
pharmacokinetic hypothesis (Figure 2).72 The transporter hypothesis.90 The neural network hypothesis suggests that the
hypothesis and target hypothesis are the most cited theories of endogenous antiepileptic system is inhibited by degeneration
ASD resistance, but neither can fully explain the neuro- and reconstruction of the neural network (including neuro-
biological basis of drug resistance. 73 The transporter genesis, gliosis, neurodegeneration, axonal sprouting, and
hypothesis suggests that overexpression of multidrug efflux synaptic reorganization), which inhibits ASDs from accessing
transporters at the BBB barrier in epilepsy decreases ASD brain to neuronal targets.91 Molecular evidence shows that
uptake, thus causing ASD resistance.72,74 The best-understood cytoskeletal genes and cytoskeletal effectors in the growth
efflux transporters in epilepsy are members of the ATP-binding cone at the tip of each axon can affect growth cone steering,
cassette (ABC) superfamily, specifically P-glycoprotein (P-gp, were abnormally expressed in the brains of DRE patients.91,92
MDR1), the multidrug resistance-associated proteins (MRP1, Before this hypothesis, it had been suggested that hippocampal
ABCC1; MRP2, ABCC2), and breast cancer resistance protein sclerosis plays an important role in the mechanisms of drug
(BCRP, ABCG2).75 Overexpression of efflux transporters in resistance in ASD.93 This view has been confirmed in drug-
brain capillary endothelium, astrocytes, and/or dysplastic resistant patients with the resection of the affected temporal
neurons with refractory epilepsy has been documented in lobe, as resection of affected tissue usually reverses drug
numerous studies.76 It has been suggested that inhibiting P-gp resistance.94 The pharmacokinetic hypothesis states that
in epileptic rats with proven drug resistance can counteract overexpression of exogenous transporters in peripheral organs
resistance.77 However, some aspects of the transporter can reduce the plasma level of ASD in patients with intractable
4438 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 3. History of ASDs development can be divided into three generations. The year of introduction refers to the first clinical use for epilepsy in
Europe, the United States, or Japan is depicted. The figure does not include drugs that are rarely used today.

epilepsy, thereby reducing the number of ASD reaching In recent years, some novel ASDs, including cannabidiol,
intracerebral epileptic lesions.95 P-gp may be a barrier to acetazolamide, cenobamate, and ganaxolone, were introduced
prevent drugs from the intestinal lumen into the bloodstream, into the market. However, their clinical application time is
thus reducing their oral bioavailability.96 It was found that relatively short, and long-term efficacy and safety need to be
there was no difference in plasma concentration of drugs further verified.11
between ASDs responders and nonresponders in animal ASDs mechanisms can be divided into five categories (Table
epilepsy models.97 However, this hypothesis postulated by 1): (1) Modulation of voltage-gated ion channels, including
Lazarowski et al. based on only two case studies, and it is not VGSCs, VGPCs, and VGCCs; (2) enhancement of GABA-
clear if their observation is limited to these cases or a mediated inhibitory neurotransmission, through the effects on
widespread phenomenon, which needs to be further verified.95 GABAR, GATs, and GABA-ATs; (3) attenuation of glutamate-
Nevertheless, based on current evidence, the mechanisms of mediated excitatory neurotransmission via acting on AMPAR
drug resistance epilepsy are still not fully understood and and NMDAR; (4) modulation of neurotransmitters release
remain challenging in treating epilepsy patients. through a presynaptic action; (5) mechanism-targeted agents,
such as carbonic anhydrase (CA) inhibitors and mechanistic
3. CURRENT CLINICAL DEVELOPMENT STATUS target of rapamycin (mTOR) inhibitors. Some ASDs may
prevent seizures by acting on multiple targets, and the
Drug therapy is the mainstay treatment for epilepsy, which can mechanisms of action of several ASDs remain elusive.101
reduce the occurrence of seizures but cannot essentially cure Table 2 summarizes some drugs in clinical development as
the disease because of the diversity of epilepsy causes, genetic potential treatments for epilepsy. CVL-865 (PF-06372865) is
heterogeneity, and different pathogenesis.98 Nowadays, over 30 the first α2/3/5 subtype selective GABA-positive allosteric
ASDs have been approved for clinical use, which is different in modulator (PAM) to undergo phase 2 clinical trials in patients
pharmacokinetics, efficacy, and adverse reactions.99 The with drug-resistant focal epilepsy (NCT04244175).38 JNJ-
history of ASDs development can be divided into three 40411813 is a positive allosteric regulator of mGlu2. A phase 2
generations (Figure 3).8,100 The first generation is traditional clinical trial is currently underway to evaluate the effect of JNJ-
ASDs (before 1958), including potassium bromide, pheno- 40411813 combined with Levetiracetam or Brivaracetam in
barbital, and barbiturate analogues (phenytoin and ethosux- treating epilepsy (NCT04836559).128 Soticlestat (TAK 935/
imide). The second-generation ASDs were introduced between OV935), a specific inhibitor of cholesterol 24-hydroxylase, was
1960 and 1975, including carbamazepine, valproate, and the developed as a potential ASD with novel mechanisms of
benzodiazepines. Phenytoin and valproate are still the first-line action.129 Soticlestat has completed a phase 2 clinical study
drugs for epilepsy treatment, but other first- and second- (NCT03650452) evaluating its efficacy, safety, and tolerability
generation ASDs are rarely used because of obvious adverse as an adjunctive treatment for developmental and/or epileptic
effects, unfavorable pharmacokinetics, and drug interaction encephalopathy in children. XEN1101 is a novel positive
profiles. The era of the third-generation AEDs started in the allosteric regulator of KV7.2−7.5 (KCNQ2−5) potassium
1980s, such as lamotrigine, levetiracetam, and oxcarbazepine. channels in neurons. The phase 3 clinical trials are designed to
Due to new targets and mechanisms, third-generation drugs evaluate XEN1101 as adjunctive therapy in focal-onset seizures
further reduce adverse drug reactions, improve pharmacoki- (FOS) (NCT05614063) and Primary Generalized Tonic-
netics and drug resistance, and have fewer drug interactions.100 Clonic Seizures (PGTCS) (NCT05667142). In phase 2b
4439 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Table 1. Molecular Targets of Clinically Used ASDs

4440 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Table 1. continued

4441 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Table 1. continued

4442 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Table 1. continued

Table 2. Progress in Clinical Research of ASDs


recruitment
drugs targets sponsors phase NUT identifier status condition and disease
CVL-865 (PF- GABA-A receptor Cerevel Therapeutics, 2 NCT04244175 recruiting seizures
06372865) LLC
JNJ-40411813 mGlu2 Janssen Research and 2 NCT04836559 recruiting focal onset seizures
Development, LLC
Soticlestat (TAK- cholesterol Takeda 2 NCT03650452 completed epilepsy; Dravet syndrome; Lennox−Gastaut
935/OV935) 24-hydroxylase syndrome

XEN1101 KV7.2/7.3 Xenon Pharmaceuticals 3 NCT05614063; recruiting FOS and PGTCS


(IOP-2198) Inc.
NCT05667142 not yet
recruiting

NBI-921352 NaV1.6 Neurocrine Biosciences 2 NCT05159908 recruiting focal onset seizures


(XEN901)
NCT04873869 SCN8A developmental and epileptic
encephalopathy syndrome (SCN8A-DEE)

retigabine KV7.2/7.3 GlaxoSmithKline 4 NCT01721317 terminated epilepsy


(XEN496)
Xenon Pharmaceuticals 3 NCT04639310 recruiting KCNQ2 developmental and epileptic
Inc. encephalopathy (KCNQ2-DEE)

Padsevonil SV2/GABA-A receptor UCB Biopharma SPRL 2 NCT03373383 completed DRE;


focal-onset seizures

CX-8998 CaV3 Jazz Pharmaceuticals 2 NCT03406702 completed idiopathic generalized epilepsy

Ataluren antisense oligonucleotide NYU Langone Health 2 NCT02758626 completed Dravet syndrome
(PTC124)
CDKL5 deficiency

ACT-709478 T-type calcium channel Idorsia Pharmaceuticals 2 NCT03239691 completed photosensitive epilepsy
Ltd.
EXP-100 5-HT Epygenix 2 NCT04462770 recruiting Dravet syndrome

4443 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 4. (A) The optimization process in developing compounds 3, 4, and 5. (B) Chemical structures of other reported NaV1.6 or NaV1.2
inhibitors. Compound with a docking score higher than a threshold value (−9.0 kcal/mol) was defined as an inactive compound in the virtual
screening of the database.

clinical trial, XEN1101 demonstrated a statistically significant premature and abnormal stop codons, bypassing nonsense
and dose-dependent reduction from baseline in monthly focal mutations. The phase 2 clinical study (NCT02758626)
seizure frequency compared to placebo (monotonic dose characterizes the safety profile of ataluren in patients with
response; p < 0.001). NBI-921352 (formerly XEN901), a CDKL5 or DS resulting from a nonsense mutation. ACT-
heterocyclic sulfonamide compound, is a novel small-molecule 709478, a T-type calcium channel agonist, has completed a
selective NaV1.6 sodium channel inhibitor. The phase 2 clinical phase 2 clinical trial (NCT03239691), evaluating its effect in
study assesses the efficacy, safety, and pharmacokinetics of photosensitive epilepsy patients. EXP-100, a clemizole
NBI-921352 as adjunctive therapy for seizures in subjects with analogue that binds to the 5-HT receptor, is undergoing
SCN8A-DEE. The most common treatment-emergent adverse phase 2 studies in DS (NCT04462770). These clinical drugs
events of NBI-921352 include headache, dizziness, and nausea. have relatively mild or moderate side effects, such as headache,
Retigabine is an ASD developed for the treatment of adults dizziness, and somnolence.131
with partial-onset seizures, which is restricted in 2013
(NCT01721317) because of its ophthalmological, dermato- 4. SMALL-MOLECULE COMPOUNDS FOR THE
logical pigmentation, and discoloration side effects. It has been TREATMENT OF EPILEPSY
now withdrawn from the market in 2017.130 XEN496, a new
oral formulation with RTG active ingredient, is currently in Although the discovery of ASDs has rapidly improved in recent
phase 3 clinical trial (NCT04639310) for the treatment of years, a challenge remains to prevent and cure epilepsy or to
KCNQ2-DEE. Padsevonil (UCB0942) was designed to act on alleviate the plight of drug-resistance in epilepsy. The screening
both synaptic vesicle protein 2 (SV2) and GABAA receptors. of compounds with antiepileptic activity is mainly based on
The phase 2 clinical study has been completed to test the three different strategies: (1) Optimizing existing ASDs,
efficacy and safety of adjunctive treatment of focal-onset finding safer and more effective analogues or derivatives
seizures in adults with DRE (NCT03373383). CX-8998 is an through in vivo epilepsy models; 2) unveiling the pathogenesis
ASD that selectively blocks the CaV3 receptor. Currently, its of epilepsy, finding new targets to develop new synthetic
phase 2 clinical trial (NCT03406702) has been completed to compounds. For example, carbonic anhydrase (CA) inhibitors
investigate the effect of IGE in adolescents and adults. Ataluren and mTOR inhibitors have been favored by scientists as
is a daily oral drug that selectively induces readthrough antiepileptic candidates with novel mechanisms.132,133 (3)
4444 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Screening for drugs that include ASDs or drugs for other et al. identified indoline scaffold derivative 1 that has moderate
diseases that have been approved by the U.0.S Food and Drug NaV1.6 efficacy and low topological polar surface area (TPSA),
Administration (FDA) though epilepsy animal models.134 but its metabolic stability and MDR1-mediated efflux are
Preclinical screening of ASDs primarily depends on poor.141 Ring opening of the indoline provided benzylami-
establishing epileptic models in vivo. Common animal models noarylsulfonamide compound 2 with over 17-fold improved
are divided into three categories according to different NaV1.6 potency compared to 1. After more systematic and
induction methods: acute animal epilepsy model, chronic extensive SAR exploration of this new scaffold, compounds 3, 4
animal epilepsy model, and genetic absence animal epilepsy (XPC-6444), and 5 (XPC-6379) were determined as potent
model.97 The maximal electroshock (MES) and subcutaneous NaV1.6 inhibitors, which also display potent blockage of
pentylenetetrazol (scPTZ) acute epilepsy models, which serve NaV1.2 (Figure 4A). These three compounds show excellent
as the gold standard for preliminary screening of ASDs, are the anticonvulsant activity and reduce seizure activity in the
most widely used. However, these two epilepsy models cannot SCN8AN1768D/+ model. They also have good tolerability,
detect drugs for DRE and chronic spontaneous epilepsy.97 The metabolic stability, and low MDR1-mediated efflux. However,
chronic epilepsy models can be divided into kindling epilepsy these compounds faced challenges in preclinical toxicology
model, persistent epilepsy model, and spontaneous epilepsy studies.141 Other reported NaV1.6 or NaV1.2 inhibitors are
model based on the intensity of stimulation and the severity of shown in Figure 4B. Compound 6 (NBI-921352), a novel
the disease. The 6 Hz corneal kindling mouse model is a selective NaV1.6 channel inhibitor (IC50 = 0.051 μM), can
medium or high throughput model of secondary generalized effectively prevent electrically induced seizures in
epilepsy, usually for studying drug resistance. The amygdala Scn8aN1768D/+ mouse, wild-type mouse, and rat-seizure models.
kindling model, because of its high sensitivity and the Compound 6 was tolerated at high multiples of effective
observation of brain pathological changes similar to human plasma and brain concentrations, which is entering the phase II
temporal lobe epilepsy (TLE), is also widely used in studying clinical trial (NCT04873869) for the treatment of SCN8A-
epilepsy drug resistance.135 Genetic epilepsy models are often DEE and adult focal-onset seizures. This compound has
generalized seizures, which can simulate the absence of seizures significant promise to be translated into a therapeutic for
in human genetic epilepsy diseases. The main animal strains individuals with treatment-resistant epilepsy.142 Compounds 7
are GAERS, WAG/Rij rats, DAB/2J mice, and human (Valsartan), 8 (Ciprofloxacin), and 9 (N.N′-diphenethylsulfa-
mutation-based mouse models such as SCN1AR1407K/+, mide), which can interact with the open conformation of the
SCN2AA263 V/+, SCNQ2A306T/+, and SCN8AN1768D/+.136 Nav1.2 pore but not with P-gp efflux transporter and were
Here, we summarize the small-molecule compounds discovered by a sequential virtual screening campaign, are
discovered in recent years, which are potential therapeutic potentially effective compounds for the treatment of P-gp
candidates against epilepsy with different mechanism of action. mediated resistant epilepsy. These three compounds exhibited
4.1. Modulation of Voltage-Gated Ion Channels. inhibitory effects on the NaV1.2 channel activity and showed
Voltage-gated ion channels play an important role in regulating anticonvulsant activity in vivo.143 Compound 10, a mexiletine
neuronal excitability and are closely related to epilepsy.137 We analogue, is a highly selective NaV1.2 inhibitor that can inhibit
summarize the small molecule compounds developed recently seizures in a kindled mouse model of refractory epilepsy (6HZ
to modulate voltage-gated ion channels, which may become 44 mA).144
potential ASDs, including 10 types of sodium channel 4.1.2. Potassium Channel Agonists. The activation of
inhibitors, 15 types of potassium channel agonists, and four VGPC-KV7, which includes five members (KV7.1−5), can
types of calcium channel blockers. reduce the excitability of neurons, which has been proven to be
4.1.1. Sodium Channel Inhibitors. Nine isoforms of VGSCs an effective method for treating epilepsy.145 Retigabine (RTG)
are known in mammals (NaV1.1 to NaV1.9). The four isoforms is the first approved neuronal potassium channel agonist for
(NaV1.1, NaV1.2, NaV1.3, and NaV1.6), which are mainly treating epilepsy. RTG can cause permanent retinal abnormal-
expressed in the CNS, are closely related to epilepsy.138 NaV1.2 ities, peripheral skin discoloration, bladder dysfunction, and
and NaV1.6 isoforms are highly expressed in excitatory other side effects. It was withdrawn from markets in 2017.146
neurons, while NaV1.1 is mainly expressed in inhibitory Flupirtine, an atypical analgesic drug, was identified as a
neurons.139 NaV inhibitors, such as phenytoin, carbamazepine, KV7.2/3 channel opener.147 It has been proved that Flupirtine
lamotrigine, and oxcarbazepine, have been used for decades as is an extremely effective treatment for neonatal seizures in
anticonvulsants for the treatment of epilepsies.140 However, rats.148 However, because of rare but severe adverse drug
these existing NaV inhibitors have no selectivity in the nine reactions, namely drug-induced liver injury (DILI), the
sodium channel isoforms, require a large number of drug European Medical Agency (EMA) recommended the with-
contacts, and have narrow therapeutic indicators. Moreover, drawal of flupirtine from the market in 2018.149 Studies have
inhibition of these nonselective drugs on the off-target found that retigabine and flupirine, both containing a
pathways will damage skeletal muscle (NaV1.4), cardiac tissue triaminoaryl scaffold, can be oxidized to reactive quinone
(NaV1.5), and peripheral neurons (NaV1.7, NaV1.8, NaV1.9). It diimide or azoquinone diimide metabolites in vivo.150
has been found that selective inhibition of NaV1.6 and NaV1.2 Flupirtine is an active metabolite, such as azoquinone diimide,
while retaining high selectivity to NaV1.1 is more effective than that can directly cause cellular damage or form haptens in the
currently available ASDs and has fewer side effects on liver, which can trigger toxic autoimmune reaction and
CNS.139,141 Therefore, isoform-selective or dual-acting eventually lead to rare but serious liver toxicity.151 Retigabine
NaV1.6 and NaV1.2 inhibitors may be beneficial to epilepsy active metabolite, such as quinone diimide, can dimerize to
patients.142 phenazinium structures, thus causing blue discoloration of
In recent years, NaV inhibitors for epilepsy have focused on various tissue.152
the development of subtype-selective inhibitors, mainly To avoid the formation of quinone or azoquinone diimine
including NaV1.6 and NaV1.2 inhibitors (Figure 4). Focken metabolites, improve the drugs photosensitivity, improve
4445 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 5. (A) Chemical structures of novel Kv7 inhibitors. (B) Chemical structures of KNa1.1 inhibitors.

brain/plasma (B/P) concentration ratio, reduce the undesir- propargyl group at the N position of the RTG linker. The B/P
able side effects, and increase potency at specific Kv7 channel concentration ratio of 13 increased to 2.3 (RTG = 0.16). The
subtypes, novel candidate drugs have been developed (Figure antiepileptic activity of 13 was significantly enhanced to a value
5A). Compound 11 was obtained by replacing the carbamate 2.5 times greater than that of RTG.153 Compound 14 (HN37)
group of retigabine and flupirtine with an inverted amide and was obtained by deleting the ortho-liable −NH2 group and
introducing an essential methyl group. Compound 11 showed installing two adjacent methyl groups to the carbamate motif
six times more potent compared to flupirtine and is devoid of of compound 13. Compound 14, with its improved chemical
the potential for azaquinone diimine formation.150 Compound stability, strong efficacy, and better safety threshold, has strong
12 is a potent and photochemically stable neuronal Kv7 activation potency toward neuronal KV7 channels and high
channel activator, which displayed a higher B/P distribution efficacy in vivo. Compound 14 has entered the clinical trial in
ratio, no photoinduced dimer formation, a longer plasma half- China for epilepsy treatment.154 Compounds 15 and 16 are
life in vivo and higher potent and effective anticonvulsant two RTG conformational restriction analogues obtained by
effects in an acute seizure model in mice.19 Compound 13 (P- introducing the N5-alkyl amine indole moiety, which showed
RTG) is an RTG analogue developed by incorporating a higher potency in activating heteromeric KV7.2/KV7.3 channels
4446 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 6. (A) The optimization process of compound 31. (B) The optimization process of compound 34. (C) The optimization process of
compound 36. (D) Chemical structure of compound 37.

and improved chemical stability.155 Compound 17 selectively Treatment options for KCNT1-related diseases are limited,
activates the KV7.2/7.3 channel in a dose-dependent manner in and conventional drugs are often ineffective. Quinidine,
HEK293 cells. The EC50 value of 17 is 100 times stronger, and clofilium, and bepridil have been proposed as treatment
its antiepileptic effect is 10 times that of RTG.156 options for seizures and comorbidities.157,158 However, novel
4447 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 7. (A) Chemical structures of GABAAR modulators. (B) Chemical structures of GAT inhibitors.

therapies are urgently required because of their nonspecific, potencies, which are more potent than quinidine. Except for
low potency or side effects.158,159 Several compounds that compound 21 and 24, the other four compounds are effective
specifically inhibit KNa1.1 have been investigated (Figure 5B). in hERG inhibition and cytotoxicity assays. These may provide
Compound 18 (VU0606170) is a selective KNa1.1 inhibitor a starting point for developing new pharmacophores, and could
with low micromolar potency screened by a high-throughput become key compounds for further research on this
thallium flux assay. Compound 18 can significantly reduce the channel.158 Compound 25, a novel oxadiazole KNa1.1 inhibitor,
firing rate in overexcited and spontaneously firing cortical can reduce seizures and interictal spikes in a KCNT1-GoF
neuron cultures.160 Compounds 19−24, obtained by virtual mouse model.161
screening based on chicken KNa1.1 cryo-EM structure, can 4.1.3. Calcium Channels Blockers. TTCCs (CaV3.1,
inhibit the KNa1.1 channel with low- and submicromolar CaV3.2, and CaV3.3 channels) play an important role in IGE
4448 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

in humans and animals.162 Ethosuximide as a small-molecule is active against electric-shock-induced convulsions in mice,
blocker of TTCCs is the earliest ASD used to treat absence PTZ-induced convulsions in rats and amygdala-kindled
epilepsy.163 However, TTCCs are widely distributed in various seizures in rats. It shows a wide therapeutic range for exercise
tissues and have close homology with other voltage-gated side effects.172 Compound 39 (PF-06372865/CVL-865) is an
channels. Therefore, it is challenging to design selective T-type α2/3/5-selective GABAAR modulator and can effectively
calcium channel blockers. It is important to break through this inhibit the peak-wave discharge of rats in a GAERS genetic
limitation and find more effective compounds.164 absence epilepsy rat model, which has entered a phase 2
Recent research focuses on discoverying and developing clinical trial (NCT04244175).173 Compound 40 is a β2/3
potent, selective, and brain-penetrant TTCC blockers (Figure subunit-selective GABAAR modulator screened by pharmaco-
6). Bezençon et al.165 identified compound 26 as a moderately phore model, which has the strongest regulatory ability on
effective TTCC blocker that has certain selectivity over GABA-induced chloride current and stronger protective effect
CaV1.2. Optimization of the phenylacetic acid part and the on PTZ-induced seizures. It can promote the phased and
N-benzyl substituent on the pyrazole ring of compound 26 led persistent GABAergic inhibition of hippocampal neurons.174
to compound 27 as a triple point TTCC blocker that showed Compound 41 is an α-asaronol esters analogue that exhibits
excellent efficiency in the WAG/Rij rat epilepsy model. To good modulatory activity on human α1β2γ2-GABAAR and can
search for a good balance between solubility, metabolic significantly attenuate neuropathic pain in the mice formalin
stability, and active efflux, compounds 28 and 29 were model. Compound 41 was a promising candidate for treating
obtained by introducing indolyl and oxalkyl into the para- refractory epilepsy such as Dravet syndrome. 175 The
position of phenylacetic acid. To find the amino-pyrazole benzodiazepine site (BZ-S) on the GABAA receptor is an
metabolite that tested negative in the Ames test, moiety 30 was important active site of ASDs. Benzenediazepines (BZDs)
introduced into the compound. After the final optimization, enhance the role of the neurotransmitter GABA mainly
compound 31 (ACT-709478) was obtained, which is in phase through agonists binding to the BZD pocket, which play an
2 clinical trial (NCT03239691) (Figure 6A). Remen et al.166 important role in treating epilepsy.176 However, the non-
identified dihydropyrazole 32 as a compound with a promising selective combination of these drugs leads to serious side
potency on CaV3.2 and good selectivity for CaV1.2 by high- effects, including dependence, unnecessary sedation, and
throughput FLIPR experiment. Compound 33 was found to amnesia.177 Different effective antiepileptic compounds 42−
have higher potency toward CaV3.2 and was optimized by 47 (Figure 7A) with a BZD scaffold have been proposed to
introducing a para-fluoro substituent to the urea moiety of 32. target GABAARs through computer-aided drug design.40,178
A 4-pyridinyl group was introduced to position 3 to obtain
4.2.2. GAT inhibitors. GATs belong to the solute carrier 6
compound 34 with acceptable solubility. Compound 34
gene family A (SCL-6). There are four different subtypes in
advanced to in vivo studies and demonstrated efficacy in the
mice: mGAT1, mGAT2, mGAT3, and mGAT4, which
WAG/Rij rat model of generalized nonconvulsive, absence-like
correspond to hGAT-1, hBGT-1, hGAT-2, and hGAT-3 in
epilepsy (Figure 6B). Siegris et al.167 identified that compound
humans, respectively.179 mGAT1 is the most prevalent GAT,
35 has good activity for CaV3.1, CaV3.2, and CaV3.3 channels
which is almost exclusively located in the CNS. It was
and a good selectivity for CaV1.2 channels but poor
physicochemical and in vitro DMPK properties. Optimization identified as a major pharmacological target.180 Tiagabine was
work led to preparing pyridodiazepine 36 with improved the first highly effective and selective GAT1 inhibitor that was
physicochemical properties, solubility, and metabolic stability, approved by the U.S. FDA for treating partial epileptic seizures.
which showed promising efficacy in the WAG/Rij-rat model However, the half-life of tiagabine is very short and additionally
(Figure 6C). Compound 37 (Z944), a potent TTCC may cause dizziness, weakness, trembling, tension, diarrhea,
antagonist, can significantly inhibit absence seizures (85− depression, and other side effects.181
90%) in the GAERS model and the kindling process in the Figure 7B shows GAT inhibitors identified in recent years.
amygdala kindling model of TLE (Figure 6D).34,168 Compound 48 (DDPM-2571), a tetrahydronicotinic acid
4.2. Enhancement of GABA-Mediated Inhibitory derivative, is a novel, highly potent and in vivo active GAT1
Neurotransmission. GABA is a major inhibitory transmitter inhibitor with anticonvulsant, anxiolytic, antidepressant, and
that controls synaptic transmission and neuronal excitability. It antinociceptive properties.182 Compounds 49−51 with high
was found that enhancing GABA inhibition can reduce or antiseizure activity, low neurotoxicity, and good safety profiles
inhibit seizures.169 Here, we summarize the small-molecule that have been evaluated by epilepsy animal models, whose
compounds that can enhance GABAergic inhibition reported potential target was speculated as GAT1 by molecular
in recent years, including 10 types of GABAAR modulators, six docking.183 Compound 52 (a butylamine derivative) is a
types of GAT inhibitors, and six types of GABA-AT inhibitors. nonselective GAT inhibitor with a slight preference toward
4.2.1. GABAAR Modulators. GABAARs are composed of 19 mGAT4 (pIC50: 5.02 ± 0.11) and showed predominant
members of the GABA protein family (α1-6, β1-3, γ1-3, δ, ε, θ, anticonvulsant activity and antinociception in the formalin
π, and ρ1-3), which are important targets for CNS drugs, model of tonic pain.41 In recent years, studies have found that
especially epilepsy.170 Although several ASDs that have been BGT1 inhibitors also have antiseizure effects.184 Compound
approved for marketing can act on focal epilepsy through 53 (SBV2-114), derived from tiagabine, is a BGT1 inhibitor
GABAAR to enhance the inhibitory effects of GABA, it is still with >13-fold selectivity for BGT1 over the other GAT
necessary to develop more potent and more specific GABA subtypes.185 SBV2-114 was found to have an elusive biphasic
ASDs that are effective in drug-resistant patients with focal inhibition profile with two IC50 values (4.7 and 556 μM)
epilepsy.171 determined by a cell-based [3H] GABA uptake assay. SBV2-
Figure 7A shows the novel representative of selective 114 shows antiseizure effects in an audiogenic-induced seizure
GABAAR modulators developed in recent years. Compound model. This compound may ultimately contribute to our
38 (KRM-II-81) is an α2/3-selective GABAAR modulator and understanding of GAT transport mechanisms and become a
4449 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 8. (A) Chemical structures of GABA-AT inhibitors. (B) Chemical structures of AMPAR and NMDAR antagonists.

lead compound for designing of more selective BGT1 the pentylenetetrazole seizure threshold test and in amygdala-
inhibitors.186 kindled rats.192 It has been reported that OV329 is well
4.2.3. GABA-AT Inhibitors. GABA-AT is a pyridoxal-5- tolerated for assessed ocular end points.193 Compounds 56 and
phosphate (PLP) dependent enzyme that can degrade GABA 57, two 5,6-dihydropyrimidine-2(1H)-thione derivatives, were
into succinic acid semialdehyde.187 GABA-AT is a recognized found active in two epilepsy models (MES and scPTZ). They
target for ASDs, and GABA-AT selective inhibition plays an showed in vitro GABA-AT inhibitory potency of 18.42 μM and
antiseizure role by increasing the concentration of GABA in 19.23 μM, respectively.194 Compound 58 is a pyrimidine-
the brain.188 Vigabatrin, a known GABA-AT inhibitor, was carbothioamide derivative that can effectively suppress seizure
approved by FDA in 2009 as an adjunctive treatment for in different animal models with median doses of 15.6 mg/kg
refractory partial seizures.189 However, long-term use of (MES ED50), 278.4 mg/kg (scPTZ ED50), and 534.4 mg/kg
vigabatrin may cause permanent visual damage.190 (TD50), with no sign of neurotoxicity. In vitro, the GABA-AT
To increase the potency and eliminate retinal toxicity, enzyme activity assay of 58 showed inhibitory potency (IC50)
several vigabatrin analogues have been designed (Figure 8A). of 12.23 μM.195 Compounds 59 is a parabanic acid derivative,
Compound 54 (CPP-115) is a novel GABA-AT inhibitor that exhibiting the most potent activity against electric stimuli-
is 187 times more potent in inactivating GABA-AT than induced seizures and a protective index greater than 36.5.
vigabatrin, markedly reduced seizures with no evidence of Additionally, a molecular docking study of this compound
retinal dysfunction in a patient with refractory infantile indicated good interaction on the active site of GABA-AT.196
spasms.191 Compound 55 (OV329) is modified from 54 and 4.3. Attenuation of Glutamate-Mediated Excitatory
appears superior in both anticonvulsant potency and efficacy to Neurotransmission. Glutamate-mediated neuronal over-
vigabatrin, which shows pronounced anticonvulsant effects in excitation plays a critical role in inducing seizures. NMDAR
4450 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 9. Chemical structures of CA inhibitors.

or AMPAR antagonists have been shown to inhibit seizures in derivative, with high efficacy in vitro, low drug clearance, and
animal models, indicating their potential role in the develop- AMPAR/TARP γ-8 selectivity, which displayed potent
ment of ASDs. antiseizure efficacy in rats against PTZ-induced convulsions
4.3.1. AMPAR Antagonists. AMPAR is a transmembrane without motor side effects.201 Compound 61 (JNJ-55511118)
complex composed of four subunits (GluA1−GluA4), which is is highly potent and selective for AMPA receptors containing
a therapeutic target for epilepsy treatment.197 Perampanel is TARP γ-8. It had good pharmacokinetic characteristics and
the only clinically approved noncompetitive AMPAR antago- high receptor occupancy after oral administration. Although
nist. It acts as an adjunctive treatment for partial-onset and hepatotoxicity in rats prevented developing compound 61, this
generalized tonic-clonic seizures over 12 years of age.115 compound showed a strong dose-dependent inhibition of
However, CNS-related side effects such as dizziness, sedation, neurotransmission in the hippocampus and a strong anti-
and falls have been reported at treatment doses.198 Although
convulsant effect without causing sedation or motor impair-
AMPARs are widely distributed throughout the brain, their
ment.202 Replacement of the benzimidazolone core in 61 with
activity can be regulated by transmembrane AMPAR
regulatory proteins (TARPs). TARPs are classified into type an oxindole scaffold and introduction of a methyl group in the
I (γ-2, γ-3, γ-4, γ-8) and type II (γ-5, γ-7) by sequence 7-position leads to the identification of compound 62, which
homology. Among them, TARP γ-2 is highly expressed within demonstrated robust target engagement in the hippocampus
the cerebellum, which is involved in motor coordination, while (ED50 = 0.6 mg/kg, plasma EC50 = 9 ng/mL).This compound
TARP γ-8 is highly expressed in the hippocampus.199 Recent exhibited higher potency and lower efficacious plasma
studies have found that selective TARP γ-8-dependent concentration, and can reduce the formation of a reactive
AMPAR antagonist is expected to become novel effective metabolite.203 Compound 63 (JNJ- 61432059) is a TARP γ-8
ASDs without motor impairment.200 selective AMPAR modulator, which showed time- and dose-
Representative TARP γ-8-dependent AMPAR antagonists dependent receptor occupation in mouse hippocampus after
are shown in Figure 8B. Compound 60 is an aminothiazole oral administration and strong seizure protection in corneal
4451 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 10. (A) Chemical structures of mTOR inhibitors. (B) Co-crystal structure of p110a and compound 80 complex. Compound 80 was best
accommodated with the methyl group in one of the two hydrophobic interfaces formed by side chains of Tyr836, Ile848, Ile932 or Trp780, Ile800,
and Val850 (PDB code 6OAC). (C) Chemical structures of a COX-2 inhibitor, H3R antagonists, and an sEH inhibitor.

kindling and PTZ anticonvulsant models without damaging antagonists can control epilepsy, there is no sufficient evidence
motor function.46 to prove their efficacy and safety.116,206
4.3.2. NMDAR Antagonist. NMDARs are related to the There is little research on NMDAR antagonists in recent
pathogenesis of epilepsy and are widely expressed in the years. Compound 64 (DDBM) (Figure 8B) is a novel indolyl
cerebral cortex and hippocampus.204 NMDAR antagonists, derivative that has robust anticonvulsive activity in a rat
electroconvulsive shock (ECS) model of epilepsy. It may
including topiramate (TPM) and zolesamide, can inhibit
antagonize the binding sites of the NMDA receptor as
glutamate release. Ketamine, another NMDAR antagonist, has indicated by computational docking.207
no stable effect when used alone in the status epilepticus 4.4. New Target Compounds. With an in-depth under-
animal model, but it has synergistic effect when used in standing of the pathogenesis of epilepsy, researchers have
combination with other drugs.205 Although these NMDAR found new targets to develop novel antiepileptic compounds,
4452 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

such as mTOR inhibitors, CA inhibitors, cannabinol, sitol 3-kinase (PI3K) related kinase protein family.219 The
adenosine inhibitors, and anti-inflammatory drugs. earliest mTOR inhibitors are mainly rapamycin and its
4.4.1. CA Inhibitors. Carbonic anhydrase (CA) is a derivatives, whose mechanism of action is mainly to inhibit
ubiquitous zinc metalloproteinase, which has 15 different mTOR activity by binding with other intracellular receptors
subtypes in human (hCA I−hCA XV).208 CAs can catalyze the FKBP12. Tuberous sclerosis complex (TSC) is a genetic
reversible hydration of carbon dioxide into bicarbonate epilepsy that involves mutations in the TSC1 and/or TSC2
(HCO3−) and proton (H+). The CO2/HCO3 buffer system genes, which lead to reduced inhibition of mTOR or abnormal
mainly maintains the balance of pH in the brain and plays a activation of mTOR.220 In a TSC animal model, rapamycin
key role in the initiation and progression of epilepsy.209 CAs can significantly prevent the occurrence and development of
can influence the function of transmembrane proteins epilepsy and improve its survival rate. Everolimus is a
implicated in neuronal signaling, such as NMDARs and rapamycin analogue that can effectively reduce the frequency
GABAARs, which may affect epileptic activity.210 Some of seizures in TSC patients and the size of TSC-related solid
sulfonamide CA inhibitors, including acetazolamide (AAZ), tumors. It was approved for market in 2018.221 However, the
topiramate (TPM), metazolamide (MZA), and zonisamide inhibition of rapamycin and its analogues on TORC1 is not
(ZNS), have good anticonvulsant activity and are still used in effective for all TSC patients because their drug resistance and
the clinic for treating various forms of seizures.211 However, it adverse side effects will lead to treatment termination and dose
is clinically demonstrated that some CA inhibitors are not reduction.222 Therefore, finding mTOR inhibitors with optimal
effective when used alone or in long-term epilepsy therapy and brain permeability is of paramount importance for CNS
have serious side effects which are related to the inhibition of disease treatment.
the off-target CA isomers.212 Therefore, developing new CA Beaufils et al.223 reported a pan-PI3K/mTOR inhibitor 76
inhibitors with long-lasting efficacy and minor side effects is (PQR309) (Figure 10A), which has good brain penetrability
required. and oral bioavailability. It has entered clinical phase II for
In the past years, one strategy to develop compounds treating solid tumors and lymphoma. Rageot et al.224
displaying effective anticonvulsant activity is to target CA II optimized PQR309 and developed compound 77 (PQR620),
and CA VII isoforms in the brain.213 The benzenesulfonamide which is an effective, selective, and a brain-penetrable
(ZBG) scaffold (Ar-SO2NH2) has emerged as a pharmacody- mTORC1/2 kinase inhibitor. It has a strong selective
namic head for designing effective and isomer-selective CA inhibitory effect on mTOR kinase (Ki mTOR: 0.33 nM). It
inhibitors.214 The pharmacophore frame of CA inhibitors reduces seizures in a TSC mouse model and has antitumor
consists of a head region (ZBG), a linker/spacer such as
effects in vitro and in vivo. Ethylene-bridged morpholine in
carbamide, urea, thiourea, hydrazide, and amide, and a tail
compound 77 has been identified as the main metabolic site of
region with alkyl/aryl/heteroaryl/sugar scaffolds (Figure 9).132
human liver microsome cells.225 Therefore, Borsari et al.
For compounds 65−67, the head ZBG is connected with a
optimized 77 to obtain compound 78 (PQR626) with higher
piperazine tail through ethyl urea/acetamide. They have the
metabolic stability. Compared with 77, the stability of 78 in
strongest inhibitory effect on hCA II and hCA VII and display
human hepatocytes has been improved by 50%. Compound 78
effective seizure protection against MES and scPTZ-induced
seizures. They showed minor toxicity in subacute toxicity has excellent brain penetration ability and shows effectiveness
studies.215 For compounds 68−70, the BZG head is connected in the TSC mouse model.133 Rageot et al.226 explored the
to the piperazine tail through the benzylidene hydrazine/ morpholine part of PQR309 for compound 79, which contains
benzylidene hydrazine carbonyl. Compounds 68 and 69 have a a 1(S)-3-methylmorpholine (M2) group, and they found the
good inhibitory effect on hCA II and hCA VII. Compound 70 strongest effect on PI3K and mTOR. Its affinity increased by
only has an inhibitory effect on hCA VII. These three potent two times compared with PQR309. After optimizing the
inhibitors significantly inhibited seizures induced by MES and trifluoromethyl group, they obtained compound 80
scPTZ in mice. Compound 70 has the strongest anticonvulsant (PQR530), which is a drug-like inhibitor of PI3K/mTOR
effect, without obvious neurotoxicity and hepatorenal toxicity, kinase that has subnanomolar Kd values of 0.84 and 0.33 nM
which can effectively eliminate epilepsy in a rat model.216 For for PI3K and mTOR kinase, respectively. Compound 80 can
compounds 71−73, the BZG head is connected with the quickly enter the brain, reaching a brain-to-plasma ratio of
piperazine tail through N-methylacetamide and N-methylpro- ∼1.6. In addition, it significantly reduced the phosphorylation
pionamide, which are the most effective inhibitors of hCA II level of S6 ribosomal protein in the hippocampus of normal
and hCA VII. The three derivatives show effective anti- and epileptic mice, showing an effective inhibition of mTOR.
convulsant activity against epilepsy induced by MES and Compound 80 significantly increased the seizure threshold at a
scPTZ. Among them, compound 71 has the strongest tolerable dose (Figure 10A,B).
anticonvulsant effect and low therapeutic dosage (30 mg/ 4.4.3. COX-2 Inhibitors. Cyclooxygenase-2 (COX-2) is an
kg). After oral administration, it can eliminate the MES enzyme that synthesizes proinflammatory prostaglandins, is a
stimulation of male Wistar rats without showing neuro- potential therapeutic target for epilepsy.227 Currently, the
toxicity.217 Compounds 74 and 75 were synthesized by research of COX-2 inhibitors as ASDs is still in the preclinical
substituting acetamide for propionamide linker and piperazine stage. Some drugs, such as aspirin and rofecoxib, have been
for aniline/benzothiazole tail based on 73. These two evaluated to have antiepileptic effects in acute epilepsy and
compounds are the most effective inhibitors of hCA II and chemical convulsions or electrical models of epilepsy.228
hCA VII, which have long-term effects on seizures caused by However, these compounds have low selectivity to COX-2,
MES. After 6 h of administration, they show 75% and 25% with poor pharmacokinetics and severe hepatotoxicity after
protective effects respectively, without neurotoxicity.218 long-time use.229 Therefore, COX-2 inhibitors must be
4.4.2. mTOR Inhibitors. mTOR is a highly conserved researched to have small toxicity, higher potency, and
serine/threonine kinase that belongs to the phosphatidylino- increased selectivity.
4453 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Figure 11. (A) Design strategy and general structure of hybrid compounds. (B) Chemical structure of other multitargeted compounds.

Compound 81 (MLT-1) (Figure 10C) is a methyl sulfonyl glutamate, and aminobutyric acid in different regions of the
phenyl derivative, showed good protection against scPTZ- brain.232 Therefore, H3R is considered a pharmacological
induced acute seizures and 67% long-term epilepsy protection target for developing central nervous system diseases, including
after administration, and successfully improved the cognitive epilepsy and cognitive impairment.233 Currently, a series of
impairment of PTZ kindled rats. The molecular simulation nonimidazole H3R antagonists with anticonvulsant activity
showed that MTL has good interaction with the COX-2 active have been developed, including compounds 82−85 (Figure
site. Therefore, MLT-1 was determined an effective COX-2 10C) that show dose-dependent anticonvulsant and cognitive
inhibitor, which also shows excellent anticonvulsant and promoting properties in MES-induced seizure model.234
cognitive enhancement potential without any significant 4.4.5. sEH Inhibitors. In recent years, the roles of the
toxicity.230 arachidonic acid (AA) metabolic pathway in inflammation
4.4.4. H3R Antagonists. Histamine is an important neuro- have been widely studied. In the brain parenchyma, free AA is
transmitter that exerts biological activity through four different metabolized into different types of epoxyeicosatrienoid acids
G-protein coupled receptors (H1−H4).231 H3 receptors (EETs) by cytochrome P450 (CYP450) epoxygenases, which
(H3Rs) are abundantly expressed in the central nervous are considered antineuroinflammatory or antiapoptotic mole-
system and can also act as foreign receptors to regulate the cules.235 However, EETs are easily hydrolyzed by soluble
release of other neurotransmitters, such as acetylcholine, epoxide hydrolase (sEH) to form the less active form of
4454 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

dihydroxyeicosatrienoic acids.236 It has been suggested that 5. CONCLUSIONS AND PROSPECTS


inhibition of sEH can increase the level of EETs and improve Epilepsy is a common neurological disease that affects about
the neuronal damage caused by cerebral ischemia.237 In a 65 million people worldwide. Nowadays, ASDs are the
murine model of acute tetramethylenedisulfotetramine mainstay treatment for epilepsy. More than 30 ASDs have
(TETS) intoxication, postexposure administration of diazepam been approved for clinical use. Unfortunately, about 20−40%
combined with an sEH inhibitor stops seizures and modulates of patients do not respond to these ASDs, resulting in DRE. As
neuroinflammation.238 Therefore, sEH is a potential target for the mechanism of epilepsy pathogenesis and drug-resistance
the treatment of epilepsy. 86 (TPPU), a type of sEH inhibitor continue to be investigated, ASDs with novel mechanisms of
that can cross the blood−brain barrier, could alleviate action, higher efficacy, and reduced side effects are in high
spontaneous recurrent seizures (SRS) and epilepsy-associated demand, especially for patients with treatment-resistant
depressive behaviors in the lithium chloride (LiCl)-pilocar- epilepsy or treatment-refractory seizures. We elaborated on
pine-induced poststatus epilepticus (SE) rat model.239 the pathogenesis and clinical research progress of epilepsy and
4.5. Multitargeted Compounds. Multifunctional (or summarized 79 kinds of new antiepileptic small molecules.
multitargeted) ASDs, such as valproic acid (VPA), are the These molecules include 27 ion channel modulators, 22 GABA
most commonly used and effective compounds for treating inhibitors, five glutamate-mediated neuronal excitation modu-
different types of epilepsy.240 The combination of different lators, 11 CA inhibitors, three mTOR signal pathway blockers,
molecular mechanisms is often more beneficial for DRE.241 one COX-2 inhibitor, four H3R antagonists, one sEH inhibitor,
Multitargeted ASDs (involving synergistic or additive mecha- and 15 small antiepileptic molecules with multiple targets.
nisms) may provide a wider activity spectrum and a better These compounds offer great opportunities for the develop-
therapeutic window (Figure 11). ment of more promising ASDs.
A series of hybrid molecules with a pyrrolidine-2,5-dione Nowadays, the marketed ASDs are based on ionic
core was developed by combining the active fragments of mechanisms, which directly or indirectly act on sodium,
ethosuximide, levetiracetam, and lacosamide with the frame- potassium, and calcium ion channels or GABAAR, AMPAR,
work combination approach. Compounds 87 and 88 showed and NMDAR. The specificity of these ASDs is not strong, and
high anticonvulsant activity in MES, scPTZ, and 6 Hz seizure many are effective against multiple diseases. More targeted and
models. Compound 87 had higher protection and safety, and specific drugs for epilepsy need to be developed. In recent
compound 88 could effectively reduce pain responses in years, small-molecule compounds related to ion channels with
formalin-induced tonic pain, capsaicin-induced neurogenic potential antiepileptic effects have also made great progress.
pain, and oxaliplatin-induced neuropathic pain in mice.242 A VGSCs are mainly focused on developing selective inhibitors.
new hybrid molecule 89 (KA-104) was obtained by integrating Compared with traditional nonselective inhibitors, new
active moieties of compound 87, BCTC (a TRPV1 compounds can reduce the adverse reactions caused by off-
antagonist), and lacosamide, which showed the most potent target effects, thus improving safety.142 RTG is the only ASD
anticonvulsant activity and efficacy in the formalin-induced that acts on VGPCs and was withdrawn from the market due
tonic pain.243 To improve the water solubility of the above to serious side effects. Therefore, in recent years, researchers
compounds, compound 90 was obtained by introducing an optimized and modified the chemical structure of RTG to
amino group into the pyrroline-2,5-dione ring. Compound 90 improve the chemical structure, stability, and potency. Among
had good anticonvulsant and antineurogenic pain activity. them, XEN1101 (1OP-2198) has been used in clinical phase 2
Additionally, compound 90 has good in vitro ADME Tox for treating focal epilepsy (NCT03468725).249 HN37 has
characteristics (high metabolic stability, weak influence on completed a clinical phase 1a study (CTR20201676) in China,
CYPs, no neurotoxicity) and in vivo pharmacokinetics with good safety and metabolic characteristics.250 Small-
characteristics.244 Compounds 91 and 92 were obtained by molecule compounds targeting VGCCs have made a great
combining the structural fragment of TRPV1 antagonist with breakthrough in treating absence epilepsy, such as ACT-
89 (KA-104), which showed robust in vivo antiseizure activity 709478, which has entered phase 2 clinical trials
in the MES test and 6 Hz seizure model (using both 32 and 44 (NCT03239691).251 Small-molecule compounds targeting
mA current intensities) and were effective in the ivPTZ seizure GABAAR have also turned to developing selective inhibitors
threshold.245 The mechanism of compounds 89−92 may be to reduce their side effects, which has made good progress in
due to the inhibition of peripheral and central sodium calcium treating refractory epilepsy, such as DS and absence epilepsy.
currents and the antagonism of the TRPV1 receptor observed PF-06372865/CVL-865 has entered the clinical phase 2 trial
in vitro. In addition, compounds 93−98 with the pyrrolidine- (NCT04244175).38 Selective TARP γ-8-dependent AMPAR
2,5-dione core also have broad-spectrum antiepileptic activity antagonist (TDAA) has become the research direction in this
developed by combining different drug fragments. In vitro field, and many small molecules have been developed.
binding studies showed that the most reasonable mechanism of Compared with traditional TARP-independent AMPAR
these compounds was to affect the voltage-sensitive sodium antagonists, these new TDAA compounds are believed to
channel and L-type calcium channel of neurons.246 Com- provide an increased dose range and better efficacy in patients.
pounds 99 and 100, two isatin arylhydrazone analogues, have With the in-depth study of the pathogenesis of epilepsy,
strong anticonvulsant activity and show 100% protective effect nonionic targets have also attracted wide attention, such as CA,
at a dose of 5 mg/kg. Molecular docking studies show that the mTOR, inflammation-related protein (COX-2/IL-1β/
binding of VGSCs and GABA-A receptors is the most likely HMGB1), 5-HT, and adenosine kinase. Nowadays, only a
mechanism of these two compounds.247 Compound 101 few drugs targeting these targets, such as everolimus,
(SKA-19), a compound that can activate KCa2 channel and cannabinol, fenfluramine, and anabolin, have achieved clinical
block NaV channel, has oral bioavailability and shows activity transformation. There are still a large number of nonionic
in a wide range of rodent seizure models.248 small antiepileptic compounds in the preclinical development
4455 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

stage, which may be a future direction for developing new Leihao Sha − Department of Neurology, Joint Research
ASDs. Researchers have spent a lot of effort designing a variety Institution of Altitude Health, State Key Laboratory of
of effective and isomer-selective CA inhibitors with benzene- Biotherapy and Cancer Center, National Clinical Research
sulfonamide as the head, which may represent a new promising Center for Geriatrics, West China Hospital, Sichuan
treatment strategy. mTOR inhibitor everolimus has been University, Chengdu 610041 Sichuan, China
marketed to treat patients with TSC epilepsy, but its long-term Yilin Xia − Department of Neurology, Joint Research
use is limited by immunosuppression, poor brain penetration, Institution of Altitude Health, State Key Laboratory of
and feedback circulation induction. Therefore, three novel dual Biotherapy and Cancer Center, National Clinical Research
P13K/mTOR inhibitors with good brain permeability have Center for Geriatrics, West China Hospital, Sichuan
been developed in recent years, which may overcome the University, Chengdu 610041 Sichuan, China
shortcomings of rap therapy for epilepsy and mTOR diseases Tyler C. Ortyl − Department of Pharmaceutical Sciences,
directly related to mTOR signal cascade mutations. Inflam- College of Pharmacy, University of Tennessee Health Science
mation is closely related to epilepsy, and a variety of pro- Center, Memphis 38163 Tennessee, United States
inflammatory factors have also become targets for the Xiaohe Tian − Department of Neurology, Joint Research
development of ASDs. There are still many nonionic Institution of Altitude Health, State Key Laboratory of
antiepileptic mechanisms that have not been clinically Biotherapy and Cancer Center, National Clinical Research
transformed, and it is believed that they will become a new Center for Geriatrics, West China Hospital, Sichuan
direction of clinical research and development in the future. University, Chengdu 610041 Sichuan, China; orcid.org/
In recent years, the development of ASDs has made great 0000-0002-2294-3945
progress, but epilepsy is a very complex disease. Present ASDs Complete contact information is available at:
mainly control the symptoms of seizures but do not aim at the https://pubs.acs.org/10.1021/acs.jmedchem.2c01975
cause of treatment. In the future, drug research on epilepsy
treatment is anticipated to be more in-depth from the Author Contributions
pathogenesis of epilepsy, revealing molecular targets for
epilepsy for personalized precise treatment, including repur- T.H., J.Z., and J.W. contributed equally to this work.
posed drugs,252 new small molecules, and other treatments Notes
based on innovative technologies, such as antisense oligonu- The authors declare no competing financial interest.
cleotides and gene therapy.9,253 More and more researchers Biographies
also treat the occurrence of epilepsy and other manifestations
of the disease from the perspective of epilepsy prevention. Tingting Hu received her Ph.D. degree from Sichuan University in
These studies are expected to break through the current 2019 and is currently a postdoctoral fellow at the State Key
dilemma of epilepsy treatment so that drug treatment is no Laboratory of Biotherapy and Cancer Center, West China Hospital,
longer just to controlling seizure symptoms but also to address and Collaborative Innovation Center of Biotherapy, Sichuan
the pathogenesis of epilepsy and fundamentally solve the University. Her research interest focuses on drug design and discovery
outcome of epilepsy treatment. based on structure biology and studying the antineurotic diseases
mechanism of small-molecule compounds.
■ AUTHOR INFORMATION
Corresponding Author
Jifa Zhang received his Ph.D. degree from Southwest Jiaotong
University in 2018 and is currently a postdoctoral fellow at the State
Lei Chen − Department of Neurology, Joint Research Key Laboratory of Biotherapy and Cancer Center, West China
Institution of Altitude Health, State Key Laboratory of Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan
Biotherapy and Cancer Center, National Clinical Research University. His research interests focus on the discovery, optimization
Center for Geriatrics, West China Hospital, Sichuan and studying the molecular mechanism of candidate small-molecule
University, Chengdu 610041 Sichuan, China; compounds with antitumor activity.
Email: leilei_25@126.com Jiaxing Wang received his B.S. degree and M.S. degrees from Peking
University Health Science Center (Beijing, China). He then joined
Authors
Dr. Wei Li’s lab at the University of Tennessee Health Science Center
Tingting Hu − Department of Neurology, Joint Research
(UTHSC; Memphis, TN, United States) in 2019. His current
Institution of Altitude Health, State Key Laboratory of
Biotherapy and Cancer Center, National Clinical Research research focuses on biological evaluation of TRPC3 antagonists
Center for Geriatrics, West China Hospital, Sichuan developed by the lab.
University, Chengdu 610041 Sichuan, China; orcid.org/ Leihao Sha is currently a M.D. candidate at the Department of
0000-0002-5323-0702 Neurology, West China Hospital, Sichuan University. His research
Jifa Zhang − Department of Neurology, Joint Research interests focus on safer small-molecule drug therapy in neuro-
Institution of Altitude Health, State Key Laboratory of degenerative diseases for women of childbearing age, especially
Biotherapy and Cancer Center, National Clinical Research epilepsy.
Center for Geriatrics, West China Hospital and Precision
Yilin Xia is currently a M.D. candidate at the Department of
Medicine Key Laboratory of Sichuan Province & Precision
Neurology, West China Hospital, Sichuan University. Her research
Medicine Research Center, West China Hospital, Sichuan
interests focus on genetic mechanism and small-molecule therapy in
University, Chengdu 610041 Sichuan, China; orcid.org/
epilepsy and comorbidities including polycystic ovary syndrome.
0000-0001-8618-9200
Jiaxing Wang − Department of Pharmaceutical Sciences, Tyler C. Ortyl received his B.S. degree from The University of
College of Pharmacy, University of Tennessee Health Science Toledo (Toledo, OH, United States). He then joined Dr. Francesca-
Center, Memphis 38163 Tennessee, United States Fang Liao’s lab as a research assistant at the University of Tennessee

4456 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

Health Science Center (UTHSC; Memphis, TN, United States) in necrosis factor α; TNF-β, transforming growth factor β; TLE,
2021. temporal lobe epilepsy; TARPs, transmembrane AMPAR
Xiaohe Tian obtained his M.S. degree under cosupervision of Prof. regulatory proteins; TPM, topiramate; TSC, tuberous sclerosis
Jim A. Thomas and Prof. Giuseppe Battaglia from the department of complex; TETS, tetramethylenedisulfotetramine; VGSCs,
Engineering Materials. He continued a Ph.D. degree with Prof. G. voltage-gated sodium channels; VGPCs, voltage-gated potas-
Battaglia at University of Sheffield at the department of Biomedical sium channels; VGCCs, voltage-gated calcium channels; ZNS,
Science until 2013. This was followed by two years postdoctoral zonisamide; ZBG, benzene sulfonamide
program at University College London, department of Chemistry
(2013−2015). He now working in Sichuan University, West China
Hospital as a full professor. His research has concentrated on
■ REFERENCES
(1) Fisher, R. S.; van Emde Boas, W.; Blume, W.; Elger, C.; Genton,
functional nanosystem for treating central nervous system disease, and P.; Lee, P.; Engel, J., Jr. Epileptic seizures and epilepsy: definitions
functional bioimaging including super-resolution imaging and electron proposed by the International League Against Epilepsy (ILAE) and
microscopic imaging. the International Bureau for Epilepsy (IBE). Epilepsia 2005, 46, 470−
472.
Lei Chen received her Ph.D. degree from Sichuan University and is (2) Scheffer, I. E.; Berkovic, S.; Capovilla, G.; Connolly, M. B.;
now a neurologist and professor in the Neurology department of West French, J.; Guilhoto, L.; Hirsch, E.; Jain, S.; Mathern, G. W.; Moshe,
China Hospital, Sichuan University. Her research interests focus on S. L.; Nordli, D. R.; Perucca, E.; Tomson, T.; Wiebe, S.; Zhang, Y. H.;
the etiology, pathogenesis, diagnosis, and small molecule drug therapy Zuberi, S. M. ILAE classification of the epilepsies: Position paper of
of neurodegenerative diseases, especially epilepsy. Her current the ILAE commission for classification and terminology. Epilepsia.
research focuses on the molecular mechanism and optimization of 2017, 58, 512−521.
small-molecule compounds as a therapy to epilepsy, role of noncoding (3) Fiest, K. M.; Sauro, K. M.; Wiebe, S.; Patten, S. B.; Kwon, C. S.;
RNA in refractory epilepsy, brain imaging studies of epilepsy and Dykeman, J.; Pringsheim, T.; Lorenzetti, D. L.; Jetté, N. Prevalence
metabolic mechanisms of epilepsy. and incidence of epilepsy: A systematic review and meta-analysis of
international studies. Neurology. 2017, 88, 296−303.

■ ACKNOWLEDGMENTS
This work was supported by grants from National Natural
(4) Symonds, J. D.; Zuberi, S. M.; Stewart, K.; McLellan, A.;
O’Regan, M.; MacLeod, S.; Jollands, A.; Joss, S.; Kirkpatrick, M.;
Brunklaus, A.; Pilz, D. T.; Shetty, J.; Dorris, L.; Abu-Arafeh, I.;
Science Foundation of China (82271500, 81903502), Key Andrew, J.; Brink, P.; Callaghan, M.; Cruden, J.; Diver, L. A.; Findlay,
R&D Plan of Sichuan Province (23ZDYF2200), and Natural C.; Gardiner, S.; Grattan, R.; Lang, B.; MacDonnell, J.; McKnight, J.;
Science Foundation of Sichuan Province (2022NSFSC1365). Morrison, C. A.; Nairn, L.; Slean, M. M.; Stephen, E.; Webb, A.;
Vincent, A.; Wilson, M. Incidence and phenotypes of childhood-onset

■ ABBREVIATIONS USED
ASDs, antiseizure drugs; AMPAR, α-amino-3-hydroxy-5-
genetic epilepsies: a prospective population-based national cohort.
Brain. 2019, 142, 2303−2318.
(5) Brooks-Kayal, A. R.; Shumate, M. D.; Jin, H.; Rikhter, T. Y.;
methyl-4-isoxazole propionic acid receptor; AAZ, acetazola- Coulter, D. A. Selective changes in single cell GABA(A) receptor
mide; AA, arachidonic acid; BBB, blood−brain barrier; brain/ subunit expression and function in temporal lobe epilepsy. Nat. Med.
plasma, B/P; COX2, cyclooxygenases-2; CA, carbonic 1998, 4, 1166−1172. Choi, J.; Nordli, D. R., Jr.; Alden, T. D.; DiPatri,
anhydrase; CYP450, cytochrome P450; DRE, drug-resistant A., Jr.; Laux, L.; Kelley, K.; Rosenow, J.; Schuele, S. U.; Rajaram, V.;
epilepsy; DS, Dravet syndrome; DEE, developmental and Koh, S. Cellular injury and neuroinflammation in children with
chronic intractable epilepsy. J. Neuroinflammation 2009, 6, 38. Ong,
epileptic encephalopathy; EIEE, early infantile epileptic M. S.; Kohane, I. S.; Cai, T.; Gorman, M. P.; Mandl, K. D.
encephalopathy; ERS, endoplasmic reticulum stress; EET, Population-level evidence for an autoimmune etiology of epilepsy.
epoxyeicosatrienoid acid; FS, febrile seizures; FDA, Food and JAMA Neurol. 2014, 71, 569−574.
Drug Administration; FAAH, fatty acid amide hydrolase; (6) Cervenka, M. C.; Henry, B. J.; Felton, E. A.; Patton, K.; Kossoff,
GEFS+, generalized epilepsy with febrile seizures plus; GOF, E. H. Establishing an adult epilepsy diet center: Experience, efficacy
gain-of-function; GABA, γ-aminobutyric acid; GAD, glutamic and challenges. Epilepsy Behav. 2016, 58, 61−68.
acid decarboxylase; GATs, GABA transporters; GABA-AT, (7) Burneo, J. G.; Shariff, S. Z.; Liu, K.; Leonard, S.; Saposnik, G.;
GABA aminotransferase; GABAR, GABA receptors; HMGB1, Garg, A. X. Disparities in surgery among patients with intractable
high-mobility group box 1; H3Rs, H3 receptors; ILAE, epilepsy in a universal health system. Neurology. 2016, 86, 72−78.
International League Against Epilepsy; IGE, idiopathic (8) Rho, J. M.; White, H. S. Brief history of anti-seizure drug
development. Epilepsia Open. 2018, 3, 114−119.
generalized epilepsy; iGluR, ionotropic glutamate receptors; (9) Perucca, E. The pharmacological treatment of epilepsy: recent
IL-1β, interleukin-1β; iNOS, inducible nitric oxide synthase; advances and future perspectives. Acta Epileptologica 2021, 3, 22.
KAR, kainate receptor; LTP, long-term potentiation; LiCl, (10) Brodie, M. J.; Barry, S. J.; Bamagous, G. A.; Norrie, J. D.; Kwan,
lithium chloride; mGluR, metabotropic glutamate receptor; P. Patterns of treatment response in newly diagnosed epilepsy.
MFS, mossy fiber sprouting; mTOR, mechanistic target of Neurology. 2012, 78, 1548−1554.
rapamycin; MES, maximal electroshock; NPY, neuropeptide Y; (11) Oyrer, J.; Maljevic, S.; Scheffer, I. E.; Berkovic, S. F.; Petrou, S.;
MZA, methazolamide; NCAM, neural cell adhesion molecule; Reid, C. A. Ion channels in genetic epilepsy: From genes and
NMDAR, N-methyl-D-aspartate receptor; NOX, NADPH mechanisms to disease-targeted therapies. Pharmacol Rev. 2018, 70,
oxidase; NF-κB, nuclear factor-Κ B; OS, oxidative stress; 142−173.
PGE2, prostaglandin-E2; PAMs, positive allosteric modulators; (12) Mastrangelo, M.; Sartori, S.; Simonati, A.; Brinciotti, M.; Moro,
F.; Nosadini, M.; Pezzini, F.; Doccini, S.; Santorelli, F. M.; Leuzzi, V.
PLP, pyridoxal-5-phosphate; PI3K, phosphatidylinositol 3- Progressive myoclonus epilepsy and ceroidolipofuscinosis 14: The
kinase; ROS, reactive oxygen species; RNS, reactive nitrogen multifaceted phenotypic spectrum of KCTD7-related disorders. Eur. J.
species; RTG, retigabine; SRS, spontaneous recurrent seizures; Med. Genet. 2019, 62, 103591.
scPTZ, subcutaneous pentylenetetrazol; SCL-6, solute carrier 6 (13) Gasser, A.; Ho, T. S.; Cheng, X.; Chang, K. J.; Waxman, S. G.;
gene family; sEH, soluble epoxide hydrolase; SE, status Rasband, M. N.; Dib-Hajj, S. D. An ankyrinG-binding motif is
epilepticus; TTCC, T-type calcium channel; TNF-α, tumor necessary and sufficient for targeting Nav1.6 sodium channels to axon

4457 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

initial segments and nodes of Ranvier. J. Neurosci. 2012, 32, 7232− a KCNQ2 C terminus mutation causing benign familial neonatal
7243. Hu, W.; Tian, C.; Li, T.; Yang, M.; Hou, H.; Shu, Y. Distinct convulsions. J. Biol. Chem. 2006, 281, 418−428.
contributions of Na(v)1.6 and Na(v)1.2 in action potential initiation (23) Main, M. J.; Cryan, J. E.; Dupere, J. R.; Cox, B.; Clare, J. J.;
and backpropagation. Nat. Neurosci. 2009, 12, 996−1002. Burbidge, S. A. Modulation of KCNQ2/3 potassium channels by the
(14) Wolff, M.; Johannesen, K. M.; Hedrich, U. B. S.; Masnada, S.; novel anticonvulsant retigabine. Mol. Pharmacol. 2000, 58, 253−262.
Rubboli, G.; Gardella, E.; Lesca, G.; Ville, D.; Milh, M.; Villard, L.; (24) Kaczmarek, L. K. Slack, slick and sodium-activated potassium
Afenjar, A.; Chantot-Bastaraud, S.; Mignot, C.; Lardennois, C.; Nava, channels. ISRN Neurosci. 2013, 2013, 354262.
C.; Schwarz, N.; Gérard, M.; Perrin, L.; Doummar, D.; Auvin, S.; (25) Alsaleem, M.; Carrion, V.; Weinstock, A.; Chandrasekharan, P.
Miranda, M. J.; Hempel, M.; Brilstra, E.; Knoers, N.; Verbeek, N.; van Infantile refractory seizures due to de novo KCNT 1 mutation. BMJ.
Kempen, M.; Braun, K. P.; Mancini, G.; Biskup, S.; Hörtnagel, K.; Case Rep. 2019, 12, e231178.
Döcker, M.; Bast, T.; Loddenkemper, T.; Wong-Kisiel, L.; Baumeister, (26) Alexander, S. P.; Catterall, W. A.; Kelly, E.; Marrion, N.; Peters,
F. M.; Fazeli, W.; Striano, P.; Dilena, R.; Fontana, E.; Zara, F.; J. A.; Benson, H. E.; Faccenda, E.; Pawson, A. J.; Sharman, J. L.;
Kurlemann, G.; Klepper, J.; Thoene, J. G.; Arndt, D. H.; Deconinck, Southan, C.; Davies, J. A. The concise guide to PHARMACOLOGY
N.; Schmitt-Mechelke, T.; Maier, O.; Muhle, H.; Wical, B.; Finetti, C.; 2015/16: Voltage-gated ion channels. Br. J. Pharmacol. 2015, 172,
Brückner, R.; Pietz, J.; Golla, G.; Jillella, D.; Linnet, K. M.; Charles, P.; 5904−5941.
Moog, U.; Õ iglane-Shlik, E.; Mantovani, J. F.; Park, K.; Deprez, M.; (27) Ryglewski, S.; Lance, K.; Levine, R. B.; Duch, C. Ca(v)2
Lederer, D.; Mary, S.; Scalais, E.; Selim, L.; Van Coster, R.; Lagae, L.; channels mediate low and high voltage-activated calcium currents in
Nikanorova, M.; Hjalgrim, H.; Korenke, G. C.; Trivisano, M.; Drosophila motoneurons. J. Physiol. 2012, 590, 809−825.
Specchio, N.; Ceulemans, B.; Dorn, T.; Helbig, K. L.; Hardies, K.; (28) Ehlinger, D. G.; Commons, K. G. Cav1.2 L-type calcium
Stamberger, H.; de Jonghe, P.; Weckhuysen, S.; Lemke, J. R.; channels regulate stress coping behavior via serotonin neurons.
Krägeloh-Mann, I.; Helbig, I.; Kluger, G.; Lerche, H.; Møller, R. S. Neuropharmacology. 2019, 144, 282−290. Davydova, D.; Marini, C.;
Genetic and phenotypic heterogeneity suggest therapeutic implica- King, C.; Klueva, J.; Bischof, F.; Romorini, S.; Montenegro-Venegas,
tions in SCN2A-related disorders. Brain. 2017, 140, 1316−1336. C.; Heine, M.; Schneider, R.; Schröder, M. S.; Altrock, W. D.;
(15) Veeramah, K. R.; O’Brien, J. E.; Meisler, M. H.; Cheng, X.; Dib- Henneberger, C.; Rusakov, D. A.; Gundelfinger, E. D.; Fejtova, A.
Hajj, S. D.; Waxman, S. G.; Talwar, D.; Girirajan, S.; Eichler, E. E.; Bassoon specifically controls presynaptic P/Q-type Ca(2+) channels
Restifo, L. L.; Erickson, R. P.; Hammer, M. F. De novo pathogenic via RIM-binding protein. Neuron. 2014, 82, 181−194.
SCN8A mutation identified by whole-genome sequencing of a family (29) Mirlohi, S.; Bladen, C.; Santiago, M.; Connor, M. Modulation
quartet affected by infantile epileptic encephalopathy and SUDEP. of recombinant human T-type calcium channels by Δ(9)-tetrahy-
Am. J. Hum. Genet. 2012, 90, 502−510. drocannabinolic acid in vitro. Cannabis Cannabinoid Res. 2022, 7, 34−
(16) Ma, H.; Guo, Y.; Chen, Z.; Wang, L.; Tang, Z.; Zhang, J.; Miao, 45.
Q.; Zhai, Q. Mutations in the sodium channel genes SCN1A, SCN3A, (30) Ernst, W. L.; Zhang, Y.; Yoo, J. W.; Ernst, S. J.; Noebels, J. L.
and SCN9A in children with epilepsy with febrile seizures plus(EFS Genetic enhancement of thalamocortical network activity by elevating
+). Seizure. 2021, 88, 146−152. Vanoye, C. G.; Gurnett, C. A.; alpha 1g-mediated low-voltage-activated calcium current induces pure
Holland, K. D.; George, A. L., Jr.; Kearney, J. A. Novel SCN3A absence epilepsy. J. Neurosci. 2009, 29, 1615−1625. Kim, D.; Song, I.;
variants associated with focal epilepsy in children. Neurobiol Dis. 2014,
Keum, S.; Lee, T.; Jeong, M. J.; Kim, S. S.; McEnery, M. W.; Shin, H.
62, 313−322.
S. Lack of the burst firing of thalamocortical relay neurons and
(17) Wimmer, V. C.; Harty, R. C.; Richards, K. L.; Phillips, A. M.;
resistance to absence seizures in mice lacking alpha(1G) T-type
Miyazaki, H.; Nukina, N.; Petrou, S. Sodium channel β1 subunit
Ca(2+) channels. Neuron. 2001, 31, 35−45.
localizes to axon initial segments of excitatory and inhibitory neurons
(31) Eckle, V. S.; Shcheglovitov, A.; Vitko, I.; Dey, D.; Yap, C. C.;
and shows regional heterogeneity in mouse brain. J. Comp Neurol.
Winckler, B.; Perez-Reyes, E. Mechanisms by which a CACNA1H
2015, 523, 814−830.
(18) Scheffer, I. E.; Harkin, L. A.; Grinton, B. E.; Dibbens, L. M.; mutation in epilepsy patients increases seizure susceptibility. J. Physiol.
Turner, S. J.; Zielinski, M. A.; Xu, R.; Jackson, G.; Adams, J.; 2014, 592, 795−809.
Connellan, M.; Petrou, S.; Wellard, R. M.; Briellmann, R. S.; Wallace, (32) Rajakulendran, S.; Graves, T. D.; Labrum, R. W.;
R. H.; Mulley, J. C.; Berkovic, S. F. Temporal lobe epilepsy and GEFS Kotzadimitriou, D.; Eunson, L.; Davis, M. B.; Davies, R.; Wood, N.
+ phenotypes associated with SCN1B mutations. Brain. 2006, 130, W.; Kullmann, D. M.; Hanna, M. G.; Schorge, S. Genetic and
100−109. functional characterisation of the P/Q calcium channel in episodic
(19) Musella, S.; Carotenuto, L.; Iraci, N.; Baroli, G.; Ciaglia, T.; ataxia with epilepsy. J. Physiol. 2010, 588, 1905−1913.
Nappi, P.; Basilicata, M. G.; Salviati, E.; Barrese, V.; Vestuto, V.; (33) Yang, Z. Q.; Barrow, J. C.; Shipe, W. D.; Schlegel, K. A.; Shu,
Pignataro, G.; Pepe, G.; Sommella, E.; Di Sarno, V.; Manfra, M.; Y.; Yang, F. V.; Lindsley, C. W.; Rittle, K. E.; Bock, M. G.; Hartman,
Campiglia, P.; Gomez-Monterrey, I.; Bertamino, A.; Taglialatela, M.; G. D.; Uebele, V. N.; Nuss, C. E.; Fox, S. V.; Kraus, R. L.; Doran, S.
Ostacolo, C.; Miceli, F. Beyond retigabine: design, synthesis, and M.; Connolly, T. M.; Tang, C.; Ballard, J. E.; Kuo, Y.; Adarayan, E. D.;
pharmacological characterization of a potent and chemically stable Prueksaritanont, T.; Zrada, M. M.; Marino, M. J.; Graufelds, V. K.;
neuronal Kv7 channel activator with anticonvulsant activity. J. Med. DiLella, A. G.; Reynolds, I. J.; Vargas, H. M.; Bunting, P. B.;
Chem. 2022, 65, 11340−11364. Woltmann, R. F.; Magee, M. M.; Koblan, K. S.; Renger, J. J. Discovery
(20) Barhanin, J.; Lesage, F.; Guillemare, E.; Fink, M.; Lazdunski, of 1,4-substituted piperidines as potent and selective inhibitors of T-
M.; Romey, G. K(V)LQT1 and lsK (minK) proteins associate to form type calcium channels. J. Med. Chem. 2008, 51, 6471−6477.
the I(Ks) cardiac potassium current. Nature. 1996, 384, 78−80. (34) Tringham, E.; Powell, K. L.; Cain, S. M.; Kuplast, K.;
(21) Wang, H. S.; Pan, Z.; Shi, W.; Brown, B. S.; Wymore, R. S.; Mezeyova, J.; Weerapura, M.; Eduljee, C.; Jiang, X.; Smith, P.;
Cohen, I. S.; Dixon, J. E.; McKinnon, D. KCNQ2 and KCNQ3 Morrison, J. L.; Jones, N. C.; Braine, E.; Rind, G.; Fee-Maki, M.;
potassium channel subunits: molecular correlates of the M-channel. Parker, D.; Pajouhesh, H.; Parmar, M.; O’Brien, T. J.; Snutch, T. P. T-
Science. 1998, 282, 1890−1893. type calcium channel blockers that attenuate thalamic burst firing and
(22) Castaldo, P.; del Giudice, E. M.; Coppola, G.; Pascotto, A.; suppress absence seizures. Sci. Transl Med. 2012, 4, 121ra19.
Annunziato, L.; Taglialatela, M. Benign familial neonatal convulsions (35) Zhao, T.; Chen, Y.; Sun, Z.; Shi, Z.; Qin, J.; Lu, J.; Li, C.; Ma,
caused by altered gating of KCNQ2/KCNQ3 potassium channels. J. D.; Zhou, L.; Song, X. Prenatal sevoflurane exposure causes neuronal
Neurosci. 2002, 22, RC199. Soldovieri, M. V.; Castaldo, P.; Iodice, L.; excitatory/inhibitory imbalance in the prefrontal cortex and neuro-
Miceli, F.; Barrese, V.; Bellini, G.; del Giudice, E. M.; Pascotto, A.; functional abnormality in rats. Neurobiol Dis. 2020, 146, 105121.
Bonatti, S.; Annunziato, L.; Taglialatela, M. Decreased subunit Mahmoud, M. A.; Zaitone, S. A.; Ammar, A. M. Binary and ternary
stability as a novel mechanism for potassium current impairment by Cu(II) complexes of pregabalin with excitatory and inhibitory

4458 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

neurotransmitters and their antiepileptic effect. Mater. Sci. Eng. C Miyake, N.; Tanaka, F.; Kato, M.; Ogata, K.; Saitsu, H.; Matsumoto,
Mater. Biol. Appl. 2020, 110, 110650. N. GRIN1 mutations cause encephalopathy with infantile-onset
(36) Wang, H. T.; Zhu, Z. A.; Li, Y. Y.; Lou, S. S.; Yang, G.; Feng, epilepsy, and hyperkinetic and stereotyped movement disorders.
X.; Xu, W.; Huang, Z. L.; Cheng, X.; Xiong, Z. Q. CDKL5 deficiency Epilepsia. 2015, 56, 841−848.
in forebrain glutamatergic neurons results in recurrent spontaneous (50) Patel, D. C.; Tewari, B. P.; Chaunsali, L.; Sontheimer, H.
seizures. Epilepsia. 2021, 62, 517−528. Neuron-glia interactions in the pathophysiology of epilepsy. Nat. Rev.
(37) George, S.; James, S.; De Blas, A. L. Selective overexpression of Neurosci. 2019, 20, 282−297.
collybistin in mouse hippocampal pyramidal cells enhances (51) Walch, E.; Murphy, T. R.; Cuvelier, N.; Aldoghmi, M.;
GABAergic neurotransmission and protects against PTZ-induced Morozova, C.; Donohue, J.; Young, G.; Samant, A.; Garcia, S.;
seizures. eNeuro 2021, 8, ENEURO.0561-20. Zhang, T.; Yu, F.; Xu, Alvarez, C.; Bilas, A.; Davila, D.; Binder, D. K.; Fiacco, T. A.
H.; Chen, M.; Chen, X.; Guo, L.; Zhou, C.; Xu, Y.; Wang, F.; Yu, J.; Astrocyte-selective volume increase in elevated extracellular potassium
Wu, B. Dysregulation of REV-ERBα impairs GABAergic function and conditions is mediated by the Na(+)/K(+) ATPase and occurs
promotes epileptic seizures in preclinical models. Nat. Commun. 2021, independently of aquaporin 4. ASN Neuro. 2020, 12,
12, 1216. 1759091420967152.
(38) Gurrell, R.; Whitlock, M.; Wei, H.; Shen, Z.; Ogden, A. Safety, (52) David, Y.; Cacheaux, L. P.; Ivens, S.; Lapilover, E.; Heinemann,
tolerability, and pharmacokinetics of multiple repeated oral doses of U.; Kaufer, D.; Friedman, A. Astrocytic dysfunction in epilepto-
the α2/3/5-subtype selective GABA(A) -positive allosteric modulator genesis: consequence of altered potassium and glutamate homeo-
PF-06372865 in healthy volunteers. Clin Pharmacol Drug Dev. 2021, stasis? J. Neurosci. 2009, 29, 10588−10599.
10, 756−764. (53) Espinosa-Garcia, C.; Zeleke, H.; Rojas, A. Impact of stress on
(39) Pong, A. W.; Ross, J.; Tyrlikova, I.; Giermek, A. J.; Kohli, M. P.; epilepsy: focus on neuroinflammation-A mini review. Int. J. Mol. Sci.
Khan, Y. A.; Salgado, R. D.; Klein, P. Epilepsy: expert opinion on 2021, 22, 4061.
emerging drugs in phase 2/3 clinical trials. Expert Opin Emerg Drugs. (54) Vandresen-Filho, S.; Martins, W. C.; Bertoldo, D. B.; Mancini,
2022, 27, 75−90. G.; De Bem, A. F.; Tasca, C. I. Cerebral cortex, hippocampus,
(40) Wang, S.; Liu, H.; Wang, X.; Lei, K.; Li, G.; Li, J.; Liu, R.; striatum and cerebellum show differential susceptibility to quinolinic
Quan, Z. Synthesis of 1,3,4-oxadiazole derivatives with anticonvulsant acid-induced oxidative stress. Neurol Sci. 2015, 36, 1449−1456.
activity and their binding to the GABA(A) receptor. Eur. J. Med. (55) Hoffmann, K.; Lindner, M.; Gröticke, I.; Stangel, M.; Löscher,
Chem. 2020, 206, 112672. W. Epileptic seizures and hippocampal damage after cuprizone-
(41) Zaręba, P.; Sałat, K.; Höfner, G. C.; Ła̧tka, K.; Bajda, M.; induced demyelination in C57BL/6 mice. Exp. Neurol. 2008, 210,
Latacz, G.; Kotniewicz, K.; Rapacz, A.; Podkowa, A.; Maj, M.; 308−321.
Jóźwiak, K.; Filipek, B.; Wanner, K. T.; Malawska, B.; Kulig, K. (56) Liu, X. X.; Yang, L.; Shao, L. X.; He, Y.; Wu, G.; Bao, Y. H.; Lu,
Development of tricyclic N-benzyl-4-hydroxybutanamide derivatives N. N.; Gong, D. M.; Lu, Y. P.; Cui, T. T.; Sun, N. H.; Chen, D. Y.;
as inhibitors of GABA transporters mGAT1−4 with anticonvulsant, Shi, W. X.; Fukunaga, K.; Chen, H. S.; Chen, Z.; Han, F.; Lu, Y. M.
antinociceptive, and antidepressant activity. Eur. J. Med. Chem. 2021, Endothelial Cdk5 deficit leads to the development of spontaneous
221, 113512. epilepsy through CXCL1/CXCR2-mediated reactive astrogliosis. J.
(42) Jamal Gilani, S.; Zaheen Hassan, M.; Sarim Imam, S.; Kala, C.; Exp Med. 2020, 217, e20180992.
Prakash Dixit, S. Novel benzothiazole hydrazine carboxamide hybrid (57) Weidner, L. D.; Kannan, P.; Mitsios, N.; Kang, S. J.; Hall, M.
scaffolds as potential in vitro GABA AT enzyme inhibitors: Synthesis, D.; Theodore, W. H.; Innis, R. B.; Mulder, J. The expression of
molecular docking and antiepileptic evaluation. Bioorg. Med. Chem. inflammatory markers and their potential influence on efflux
Lett. 2019, 29, 1825−1830. transporters in drug-resistant mesial temporal lobe epilepsy tissue.
(43) Hamdan, S. K.; Mohd Zain, A. In vivo electrochemical Epilepsia. 2018, 59, 1507−1517.
biosensor for brain glutamate detection: a mini review. Malays J. Med. (58) van Vliet, E. A.; Aronica, E.; Vezzani, A.; Ravizza, T.
Sci. 2014, 21, 12−26. Neuroinflammatory pathways as treatment targets and biomarker
(44) Mihály, A. The reactive plasticity of hippocampal ionotropic candidates in epilepsy: emerging evidence from preclinical and clinical
glutamate receptors in animal epilepsies. Int. J. Mol. Sci. 2019, 20, studies. Neuropathol Appl. Neurobiol. 2018, 44, 91−111. Klement, W.;
1030. Blaquiere, M.; Zub, E.; deBock, F.; Boux, F.; Barbier, E.; Audinat, E.;
(45) Zaitsev, A. V.; Kim, K.; Vasilev, D. S.; Lukomskaya, N. Y.; Lerner-Natoli, M.; Marchi, N. A pericyte-glia scarring develops at the
Lavrentyeva, V. V.; Tumanova, N. L.; Zhuravin, I. A.; Magazanik, L. leaky capillaries in the hippocampus during seizure activity. Epilepsia.
G. N-methyl-D-aspartate receptor channel blockers prevent pentyle- 2019, 60, 1399−1411.
netetrazole-induced convulsions and morphological changes in rat (59) Vezzani, A.; Viviani, B. Neuromodulatory properties of
brain neurons. J. Neurosci Res. 2015, 93, 454−465. inflammatory cytokines and their impact on neuronal excitability.
(46) Savall, B. M.; Wu, D.; Swanson, D. M.; Seierstad, M.; Wu, N.; Neuropharmacology. 2015, 96, 70−82.
Vives Martinez, J.; García Olmos, B.; Lord, B.; Coe, K.; Koudriakova, (60) Roseti, C.; van Vliet, E. A.; Cifelli, P.; Ruffolo, G.; Baayen, J. C.;
T.; Lovenberg, T. W.; Carruthers, N. I.; Maher, M. P.; Ameriks, M. K. Di Castro, M. A.; Bertollini, C.; Limatola, C.; Aronica, E.; Vezzani, A.;
Discovery of imidazo[1,2-a]pyrazines and pyrazolo[1,5-c]pyrimidines Palma, E. GABAA currents are decreased by IL-1β in epileptogenic
as TARP γ-8 selective AMPAR negative modulators. ACS Med. Chem. tissue of patients with temporal lobe epilepsy: implications for
Lett. 2019, 10, 267−272. ictogenesis. Neurobiol Dis. 2015, 82, 311−320.
(47) Taniguchi, S.; Stolz, J. R.; Swanson, G. T. The antiseizure drug (61) Jiang, J.; Yang, M. S.; Quan, Y.; Gueorguieva, P.; Ganesh, T.;
perampanel is a subunit-selective negative allosteric modulator of Dingledine, R. Therapeutic window for cyclooxygenase-2 related anti-
kainate receptors. J. Neurosci. 2022, 42, 5499−5509. inflammatory therapy after status epilepticus. Neurobiol Dis. 2015, 76,
(48) Bertocchi, I.; Eltokhi, A.; Rozov, A.; Chi, V. N.; Jensen, V.; Bus, 126−136.
T.; Pawlak, V.; Serafino, M.; Sonntag, H.; Yang, B.; Burnashev, N.; Li, (62) Ivens, S.; Kaufer, D.; Flores, L. P.; Bechmann, I.; Zumsteg, D.;
S. B.; Obenhaus, H. A.; Both, M.; Niewoehner, B.; Single, F. N.; Tomkins, O.; Seiffert, E.; Heinemann, U.; Friedman, A. TGF-beta
Briese, M.; Boerner, T.; Gass, P.; Rawlins, J. N. P.; Köhr, G.; receptor-mediated albumin uptake into astrocytes is involved in
Bannerman, D. M.; Sprengel, R. Voltage-independent GluN2A-type neocortical epileptogenesis. Brain. 2007, 130, 535−547.
NMDA receptor Ca(2+) signaling promotes audiogenic seizures, (63) Dong, X.; Fan, J.; Lin, D.; Wang, X.; Kuang, H.; Gong, L.;
attentional and cognitive deficits in mice. Commun. Biol. 2021, 4, 59. Chen, C.; Jiang, J.; Xia, N.; He, D.; Shen, W.; Jiang, P.; Kuang, R.;
(49) Ohba, C.; Shiina, M.; Tohyama, J.; Haginoya, K.; Lerman- Zeng, L.; Xie, Y. Captopril alleviates epilepsy and cognitive
Sagie, T.; Okamoto, N.; Blumkin, L.; Lev, D.; Mukaida, S.; Nozaki, F.; impairment by attenuation of C3-mediated inflammation and synaptic
Uematsu, M.; Onuma, A.; Kodera, H.; Nakashima, M.; Tsurusaki, Y.; phagocytosis. J. Neuroinflammation. 2022, 19, 226.

4459 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

(64) Castañeda-Cabral, J. L.; Ureña-Guerrero, M. E.; Beas-Zárate, proteins in patients with focal cortical dysplasia. Seizure. 2007, 16,
C.; Colunga-Durán, A.; Nuñez-Lumbreras, M. L. A.; Orozco-Suárez, 493−503.
S.; Alonso-Vanegas, M.; Guevara-Guzmán, R.; Deli, M. A.; Rocha, L. (77) Brandt, C.; Bethmann, K.; Gastens, A. M.; Löscher, W. The
Increased expression of proinflammatory cytokines and iNOS in the multidrug transporter hypothesis of drug resistance in epilepsy: Proof-
neocortical microvasculature of patients with temporal lobe epilepsy. of-principle in a rat model of temporal lobe epilepsy. Neurobiol Dis.
Immunol Res. 2020, 68, 169−176. 2006, 24, 202−211.
(65) Rashid, H. O.; Yadav, R. K.; Kim, H. R.; Chae, H. J. ER stress: (78) Remy, S.; Beck, H. Molecular and cellular mechanisms of
autophagy induction, inhibition and selection. Autophagy. 2015, 11, pharmacoresistance in epilepsy. Brain. 2006, 129, 18−35.
1956−1977. (79) Remy, S.; Gabriel, S.; Urban, B. W.; Dietrich, D.; Lehmann, T.
(66) Fu, J.; Peng, L.; Wang, W.; He, H.; Zeng, S.; Chen, T. C.; N.; Elger, C. E.; Heinemann, U.; Beck, H. A novel mechanism
Chen, Y. Sodium valproate reduces neuronal apoptosis in acute underlying drug resistance in chronic epilepsy. Ann. Neurol. 2003, 53,
pentylenetetrzole-induced seizures via inhibiting ER stress. Neuro- 469−479.
chem. Res. 2019, 44, 2517−2526. (80) Naylor, D. E.; Liu, H.; Wasterlain, C. G. Trafficking of
(67) Debanne, D.; Thompson, S. M.; Gähwiler, B. H. A brief period GABA(A) receptors, loss of inhibition, and a mechanism for
of epileptiform activity strengthens excitatory synapses in the rat pharmacoresistance in status epilepticus. J. Neurosci. 2005, 25,
hippocampus in vitro. Epilepsia. 2006, 47, 247−256. Nateri, A. S.; 7724−7733.
Raivich, G.; Gebhardt, C.; Da Costa, C.; Naumann, H.; Vreugdenhil, (81) Loup, F.; Wieser, H. G.; Yonekawa, Y.; Aguzzi, A.; Fritschy, J.
M.; Makwana, M.; Brandner, S.; Adams, R. H.; Jefferys, J. G.; Kann, M. Selective alterations in GABAA receptor subtypes in human
O.; Behrens, A. ERK activation causes epilepsy by stimulating NMDA temporal lobe epilepsy. J. Neurosci. 2000, 20, 5401−5419.
receptor activity. Embo j. 2007, 26, 4891−4901. (82) Depondt, C. The potential of pharmacogenetics in the
(68) Acker, D. W. M.; Wong, I.; Kang, M.; Paradis, S. Semaphorin treatment of epilepsy. Eur. J. Paediatr Neurol. 2006, 10, 57−65.
4D promotes inhibitory synapse formation and suppresses seizures in Löscher, W.; Schmidt, D. Modern antiepileptic drug development has
vivo. Epilepsia. 2018, 59, 1257−1268. Liu, T. T.; Li, Y.; Shu, Y.; Xiao, failed to deliver: ways out of the current dilemma. Epilepsia. 2011, 52,
B.; Feng, L. Ephrin-b3 modulates hippocampal neurogenesis and the 657−678.
reelin signaling pathway in a pilocarpine-induced model of epilepsy. (83) Tate, S. K.; Depondt, C.; Sisodiya, S. M.; Cavalleri, G. L.;
Int. J. Mol. Med. 2018, 41, 3457−3467. Schorge, S.; Soranzo, N.; Thom, M.; Sen, A.; Shorvon, S. D.; Sander,
(69) Botterill, J. J.; Lu, Y. L.; LaFrancois, J. J.; Bernstein, H. L.; J. W.; Wood, N. W.; Goldstein, D. B. Genetic predictors of the
Alcantara-Gonzalez, D.; Jain, S.; Leary, P.; Scharfman, H. E. An maximum doses patients receive during clinical use of the anti-
excitatory and epileptogenic effect of dentate gyrus mossy cells in a epileptic drugs carbamazepine and phenytoin. Proc. Natl. Acad. Sci. U.
mouse model of epilepsy. Cell Rep. 2019, 29, 2875. S. A. 2005, 102, 5507−5512.
(70) Richichi, C.; Lin, E. J.; Stefanin, D.; Colella, D.; Ravizza, T.; (84) Van der Weide, J.; Steijns, L. S.; van Weelden, M. J.; de Haan,
Grignaschi, G.; Veglianese, P.; Sperk, G.; During, M. J.; Vezzani, A.
K. The effect of genetic polymorphism of cytochrome P450 CYP2C9
Anticonvulsant and antiepileptogenic effects mediated by adeno-
on phenytoin dose requirement. Pharmacogenetics. 2001, 11, 287−
associated virus vector neuropeptide Y expression in the rat
291.
hippocampus. J. Neurosci. 2004, 24, 3051−3059.
(85) Vega-García, A.; Orozco-Suárez, S.; Villa, A.; Rocha, L.; Feria-
(71) Kalilani, L.; Sun, X.; Pelgrims, B.; Noack-Rink, M.; Villanueva,
Romero, I.; Alonso Vanegas, M. A.; Guevara-Guzmán, R. Cortical
V. The epidemiology of drug-resistant epilepsy: A systematic review
expression of IL1-β, Bcl-2, Caspase-3 and 9, SEMA-3a, NT-3 and P-
and meta-analysis. Epilepsia. 2018, 59, 2179−2193.
(72) Tang, F.; Hartz, A. M. S.; Bauer, B. Drug-Resistant Epilepsy: glycoprotein as biological markers of intrinsic severity in drug-
Multiple Hypotheses, Few Answers. Front Neurol. 2017, 8, 301. resistant temporal lobe epilepsy. Brain Res. 2021, 1758, 147303.
(73) Beleza, P. Refractory epilepsy: a clinically oriented review. Eur. (86) Rogawski, M. A.; Johnson, M. R. Intrinsic severity as a
Neurol. 2009, 62, 65−71. determinant of antiepileptic drug refractoriness. Epilepsy Curr. 2008,
(74) Löscher, W.; Potschka, H. Drug resistance in brain diseases and 8, 127−130.
the role of drug efflux transporters. Nat. Rev. Neurosci. 2005, 6, 591− (87) Hitiris, N.; Mohanraj, R.; Norrie, J.; Sills, G. J.; Brodie, M. J.
602. Tishler, D. M.; Weinberg, K. I.; Hinton, D. R.; Barbaro, N.; Predictors of pharmacoresistant epilepsy. Epilepsy Res. 2007, 75, 192−
Annett, G. M.; Raffel, C. MDR1 gene expression in brain of patients 196.
with medically intractable epilepsy. Epilepsia. 1995, 36, 1−6. (88) Musicco, M.; Beghi, E.; Solari, A.; Viani, F. Treatment of first
(75) Xue, T.; Lu, Z. N. Association between the polymorphisms in tonic-clonic seizure does not improve the prognosis of epilepsy. First
the ATP-binding cassette genes ABCB1 and ABCC2 and the risk of Seizure Trial Group (FIRST Group). Neurology. 1997, 49, 991−998.
drug-resistant epilepsy in a Chinese Han population. Genet Mol. Res. (89) Marson, A.; Jacoby, A.; Johnson, A.; Kim, L.; Gamble, C.;
2016, 15, gmr15048752. Song, C.; Li, X.; Zhang, H.; Bao, J.; Lu, W.; Chadwick, D. Immediate versus deferred antiepileptic drug treatment
Peng, Q.; Zhang, Y. Impact of MDR1 and UGT gene polymorphisms for early epilepsy and single seizures: a randomised controlled trial.
on sodium valproate plasma concentration in patients with epilepsy. Lancet. 2005, 365, 2007−2013.
Clin Lab. 2022, 68, 220101. Mousavi, S. F.; Hasanpour, K.; (90) Schmidt, D.; Löscher, W. New developments in antiepileptic
Nazarzadeh, M.; Adli, A.; Bazghandi, M. S.; Asadi, A.; Rad, A.; drug resistance: an integrative view. Epilepsy Curr. 2009, 9, 47−52.
Gholami, O. ABCG2, SCN1A and CYP3A5 genes polymorphism and (91) Fang, M.; Xi, Z. Q.; Wu, Y.; Wang, X. F. A new hypothesis of
drug-resistant epilepsy in children: A case-control study. Seizure. drug refractory epilepsy: neural network hypothesis. Med. Hypotheses.
2022, 97, 58−62. 2011, 76, 871−876.
(76) Sun, Y.; Luo, X.; Yang, K.; Sun, X.; Li, X.; Zhang, C.; Ma, S.; (92) Yuan, J.; Wang, L. Y.; Li, J. M.; Cao, N. J.; Wang, L.; Feng, G.
Liu, Y.; Yin, J. Neural overexpression of multidrug resistance- B.; Xue, T.; Lu, Y.; Wang, X. F. Altered expression of the small
associated protein 1 and refractory epilepsy: a meta-analysis of nine guanosine triphosphatase RhoA in human temporal lobe epilepsy. J.
studies. Int. J. Neurosci. 2016, 126, 308−317. Aronica, E.; Gorter, J. A.; Mol. Neurosci. 2010, 42, 53−58. Xi, Z. Q.; Xiao, F.; Yuan, J.; Wang, X.
Ramkema, M.; Redeker, S.; Ozbas-Gerçeker, F.; van Vliet, E. A.; F.; Wang, L.; Quan, F. Y.; Liu, G. W. Gene expression analysis on
Scheffer, G. L.; Scheper, R. J.; van der Valk, P.; Baayen, J. C.; Troost, anterior temporal neocortex of patients with intractable epilepsy.
D. Expression and cellular distribution of multidrug resistance-related Synapse. 2009, 63, 1017−1028.
proteins in the hippocampus of patients with mesial temporal lobe (93) Chakir, A.; Fabene, P. F.; Ouazzani, R.; Bentivoglio, M. Drug
epilepsy. Epilepsia. 2004, 45, 441−451. Ak, H.; Ay, B.; Tanriverdi, T.; resistance and hippocampal damage after delayed treatment of
Sanus, G. Z.; Is, M.; Sar, M.; Oz, B.; Ozkara, C.; Ozyurt, E.; Uzan, M. pilocarpine-induced epilepsy in the rat. Brain Res. Bull. 2006, 71,
Expression and cellular distribution of multidrug resistance-related 127−138.

4460 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

(94) Engel, J., Jr; McDermott, M. P.; Wiebe, S.; Langfitt, J. T.; Stern, (112) Zolkowska, D.; Wu, C. Y.; Rogawski, M. A. Intramuscular
J. M.; Dewar, S.; Sperling, M. R.; Gardiner, I.; Erba, G.; Fried, I.; allopregnanolone and ganaxolone in a mouse model of treatment-
Jacobs, M.; Vinters, H. V.; Mintzer, S.; Kieburtz, K. Early surgical resistant status epilepticus. Epilepsia 2018, 59, 220−227.
therapy for drug-resistant temporal lobe epilepsy: a randomized trial. (113) Schmidt, D.; Gram, L.; Brodie, M.; Krämer, G.; Perucca, E.;
Jama. 2012, 307, 922−930. Kälviäinen, R.; Elger, C. E. Tiagabine in the treatment of epilepsy–a
(95) Lazarowski, A.; Czornyj, L.; Lubienieki, F.; Girardi, E.; clinical review with a guide for the prescribing physician. Epilepsy Res.
Vazquez, S.; D’Giano, C. ABC transporters during epilepsy and 2000, 41, 245−251.
mechanisms underlying multidrug resistance in refractory epilepsy. (114) Bröer, S.; Backofen-Wehrhahn, B.; Bankstahl, M.; Gey, L.;
Epilepsia. 2007, 48, 140−149. Gernert, M.; Löscher, W. Vigabatrin for focal drug delivery in
(96) Hartz, A. M.; Pekcec, A.; Soldner, E. L.; Zhong, Y.; Schlichtiger, epilepsy: bilateral microinfusion into the subthalamic nucleus is more
J.; Bauer, B. P-gp protein expression and transport activity in rodent effective than intranigral or systemic administration in a rat seizure
seizure models and human epilepsy. Mol. Pharmaceutics 2017, 14, model. Neurobiol Dis. 2012, 46, 362−376.
999−1011. (115) de Grazia, U.; DʼUrso, A.; Ranzato, F.; De Riva, V.;
(97) Löscher, W. Animal models of seizures and epilepsy: past, Contarato, G.; Billo, G.; Perini, F.; Galloni, E. A liquid
chromatography-mass spectrometry assay for determination of
present, and future role for the discovery of antiseizure drugs.
perampanel and concomitant antiepileptic drugs in the plasma of
Neurochem. Res. 2017, 42, 1873−1888.
patients with epilepsy compared with a fluorescent HPLC assay. Ther
(98) Xu, C.; Wang, Y.; Zhang, S.; Nao, J.; Liu, Y.; Wang, Y.; Ding,
Drug Monit. 2018, 40, 477−485.
F.; Zhong, K.; Chen, L.; Ying, X.; Wang, S.; Zhou, Y.; Duan, S.; Chen,
(116) Prüss, H.; Holtkamp, M. Ketamine successfully terminates
Z. Subicular pyramidal neurons gate drug resistance in temporal lobe malignant status epilepticus. Epilepsy Res. 2008, 82, 219−222.
epilepsy. Ann. Neurol. 2019, 86, 626−640. (117) Lynch, B. A.; Lambeng, N.; Nocka, K.; Kensel-Hammes, P.;
(99) Moshé, S. L.; Perucca, E.; Ryvlin, P.; Tomson, T. Epilepsy: new Bajjalieh, S. M.; Matagne, A.; Fuks, B. The synaptic vesicle protein
advances. Lancet. 2015, 385, 884−898. SV2A is the binding site for the antiepileptic drug levetiracetam. Proc.
(100) Perucca, E.; Brodie, M. J.; Kwan, P.; Tomson, T. 30 years of Natl. Acad. Sci. U. S. A. 2004, 101, 9861−9866.
second-generation antiseizure medications: impact and future (118) Niespodziany, I.; Rigo, J. M.; Moonen, G.; Matagne, A.;
perspectives. Lancet Neurol. 2020, 19, 544−556. Klitgaard, H.; Wolff, C. Brivaracetam does not modulate ionotropic
(101) Sills, G. J.; Rogawski, M. A. Mechanisms of action of currently channels activated by glutamate, γ-aminobutyric acid, and glycine in
used antiseizure drugs. Neuropharmacology. 2020, 168, 107966. hippocampal neurons. Epilepsia. 2017, 58, e157−e161.
Rogawski, M. A.; Löscher, W.; Rho, J. M. Mechanisms of action of (119) Elger, C. E.; Brodie, M. J.; Anhut, H.; Lee, C. M.; Barrett, J. A.
antiseizure drugs and the ketogenic diet. Cold Spring Harb Perspect Pregabalin add-on treatment in patients with partial seizures: a novel
Med. 2016, 6, a022780. evaluation of flexible-dose and fixed-dose treatment in a double-blind,
(102) Lu, Y.; Xiao, Z.; Yu, W.; Xiao, F.; Xiao, Z.; Hu, Y.; Chen, Y.; placebo-controlled study. Epilepsia. 2005, 46, 1926−1936.
Wang, X. Efficacy and safety of adjunctive zonisamide in adult (120) Hamidi, S.; Avoli, M. Carbonic anhydrase inhibition by
patients with refractory partial-onset epilepsy: a randomized, double- acetazolamide reduces in vitro epileptiform synchronization. Neuro-
blind, placebo-controlled trial. Clin Drug Investig. 2011, 31, 221−229. pharmacology. 2015, 95, 377−387.
(103) Panebianco, M.; Prabhakar, H.; Marson, A. G. Rufinamide (121) MacKeigan, J. P.; Krueger, D. A. Differentiating the mTOR
add-on therapy for drug-resistant epilepsy. Cochrane Database Syst. inhibitors everolimus and sirolimus in the treatment of tuberous
Rev. 2020, 11, CD011772. sclerosis complex. Neuro Oncol. 2015, 17, 1550−1559.
(104) Flor-Hirsch, H.; Heyman, E.; Livneh, A.; Reish, O.; (122) Lawthom, C. Valproate and epilepsy: for women as well as
Watemberg, N.; Litmanovits, I.; Ben Sason Lilli, A.; Lev, D.; men. Pract Neurol. 2018, 18, 222−223.
Lerman Sagie, T.; Bassan, H. Lacosamide for SCN2A-related (123) Abelaira, H. M.; Réus, G. Z.; Ribeiro, K. F.; Zappellini, G.;
intractable neonatal and infantile seizures. Epileptic Disord. 2018, Cipriano, A. L.; Scaini, G.; Streck, E. L.; Quevedo, J. Lamotrigine
20, 440−446. treatment reverses depressive-like behavior and alters BDNF levels in
(105) Chang, X. C.; Yuan, H.; Wang, Y.; Xu, H. Q.; Hong, W. K.; the brains of maternally deprived adult rats. Pharmacol., Biochem.
Zheng, R. Y. Eslicarbazepine acetate add-on therapy for drug-resistant Behav. 2012, 101, 348−353. Dibué-Adjei, M.; Kamp, M. A.;
focal epilepsy. Cochrane Database Syst. Rev. 2021, 6, CD008907. Alpdogan, S.; Tevoufouet, E. E.; Neiss, W. F.; Hescheler, J.;
(106) Nakamura, M.; Cho, J. H.; Shin, H.; Jang, I. S. Effects of Schneider, T. Cav2.3 (R-Type) calcium channels are critical for
cenobamate (YKP3089), a newly developed anti-epileptic drug, on mediating anticonvulsive and neuroprotective properties of lamo-
trigine in vivo. Cell Physiol Biochem. 2017, 44, 935−947.
voltage-gated sodium channels in rat hippocampal CA3 neurons. Eur.
(124) Lee, H.; Kim, D. W. Usefulness of extended-release topiramate
J. Pharmacol. 2019, 855, 175−182.
in patients with epilepsy: a two-year retention study. J. Clin Pharm.
(107) Yang, N. D.; Kanyo, R.; Zhao, L.; Li, J.; Kang, P. W.; Dou, A.
Ther. 2021, 46, 1412−1417.
K.; White, K. M.; Shi, J.; Nerbonne, J. M.; Kurata, H. T.; Cui, J.
(125) Marson, A.; Burnside, G.; Appleton, R.; Smith, D.; Leach, J.
Electro-mechanical coupling of KCNQ channels is a target of P.; Sills, G.; Tudur-Smith, C.; Plumpton, C.; Hughes, D. A;
epilepsy-associated mutations and retigabine. Sci. Adv. 2022, 8, Williamson, P.; Baker, G. A; Balabanova, S.; Taylor, C.; Brown, R.;
eabo3625. Hindley, D.; Howell, S.; Maguire, M.; Mohanraj, R.; Smith, P. E;
(108) Gomora, J. C.; Daud, A. N.; Weiergräber, M.; Perez-Reyes, E. Lanyon, K.; Manford, M.; Chitre, M.; Parker, A.; Swiderska, N.;
Block of cloned human T-type calcium channels by succinimide Appleton, R.; Pauling, J.; Hughes, A.; Gupta, R.; Hanif, S.; Awadh, M.;
antiepileptic drugs. Mol. Pharmacol. 2001, 60, 1121−1132. Ragunathan, S.; Cable, N.; Cooper, P.; Hindley, D.; Rakshi, K.;
(109) Zhu, S.; Noviello, C. M.; Teng, J.; Walsh, R. M., Jr.; Kim, J. J.; Molloy, S.; Reuber, M.; Ayonrinde, K.; Wilson, M.; Saladi, S.; Gibb, J.;
Hibbs, R. E. Structure of a human synaptic GABA(A) receptor. Funston, L.-A.; Cassidy, D.; Boyd, J.; Ratnayaka, M.; Faza, H.; Sadler,
Nature. 2018, 559, 67−72. M.; Al-Moasseb, H.; Galtrey, C.; Wren, D.; Olabi, A.; Fuller, G.;
(110) Quilichini, P. P.; Chiron, C.; Ben-Ari, Y.; Gozlan, H. Khan, M.; Kallappa, C.; Chinthapalli, R.; Aji, B.; Davies, R.; Foster,
Stiripentol, a putative antiepileptic drug, enhances the duration of K.; Hitiris, N.; Maguire, M.; Hussain, N.; Dowson, S.; Ellison, J.;
opening of GABA-A receptor channels. Epilepsia. 2006, 47, 704−716. Sharrack, B.; Gandhi, V.; Powell, R.; Tittensor, P.; Summers, B.;
(111) Sharma, R.; Nakamura, M.; Neupane, C.; Jeon, B. H.; Shin, Shashikiran, S.; Dison, P. J; Samarasekera, S.; McCorry, D.; White, K.;
H.; Melnick, S. M.; Glenn, K. J.; Jang, I. S.; Park, J. B. Positive Nithi, K.; Richardson, M.; Brown, R.; Page, R.; Deekollu, D.; Slaght,
allosteric modulation of GABA(A) receptors by a novel antiepileptic S.; Warriner, S.; Ahmed, M.; Chaudhuri, A.; Chow, G.; Artal, J.;
drug cenobamate. Eur. J. Pharmacol. 2020, 879, 173117. Kucinskiene, D.; Sreenivasa, H.; Velmurugan, S.; Zipitis, C. S;

4461 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

McLean, B.; Lal, V.; Gregoriou, A.; Maddison, P.; Pickersgill, T.; F.; Hamburger, M.; Hebeisen, P.; Loscher, W.; Fabbro, D.; Hillmann,
Anderson, J.; Lawthom, C.; Howell, S.; Whitlingum, G.; Rakowicz, P.; Wymann, M. P. 4-(Difluoromethyl)-5-(4-((3R,5S)-3,5-dimethyl-
W.; Kinton, L.; McLellan, A.; Zuberi, S.; Kelso, A.; Hughes, I.; morpholino)-6-((R)-3-methylmorpholino)-1,3,5-triazin-2-yl)pyridin-
Martland, J.; Emsley, H.; de Goede, C.; Singh, R.; Moor, C.-C.; Aram, 2-amine (PQR626), a potent, orally available, and brain-penetrant
J.; Mohanraj, R.; Sakthivel, K.; Nelapatla, S.; Rittey, C.; Pinto, A.; mTOR inhibitor for the treatment of neurological disorders. J. Med.
Leach, J. P.; Cock, H.; Richardson, A.; Houston, E.; Cooper, C.; Chem. 2020, 63, 13595−13617.
Lawson, G.; Massarano, A.; Burness, C.; Marson, A.; Smith, D.; (134) Campos, G.; Fortuna, A.; Falcão, A.; Alves, G. In vitro and in
Wieshmann, U.; Dey, I.; Sivakumar, P.; Yeung, L.-K.; Smith, P.; vivo experimental models employed in the discovery and development
Bentur, H.; Heafield, T.; Mathew, A.; Smith, D.; Jauhari, P. The of antiepileptic drugs for pharmacoresistant epilepsy. Epilepsy Res.
SANAD II study of the effectiveness and cost-effectiveness of 2018, 146, 63−86.
levetiracetam, zonisamide, or lamotrigine for newly diagnosed focal (135) Goddard, G. V.; McIntyre, D. C.; Leech, C. K. A permanent
epilepsy: an open-label, non-inferiority, multicentre, phase 4, change in brain function resulting from daily electrical stimulation.
randomised controlled trial. Lancet. 2021, 397, 1363−1374. Exp. Neurol. 1969, 25, 295−330. Sato, M.; Racine, R. J.; McIntyre, D.
(126) Mazurkiewicz-Bełdzińs ka, M.; Zawadzka, M. Use of C. Kindling: basic mechanisms and clinical validity. Electroencephalogr
cannabidiol in the treatment of epilepsy. Neurol Neurochir Polym. Clin Neurophysiol. 1990, 76, 459−472.
2022, 56, 14−20. (136) Vergnes, M.; Marescaux, C.; Micheletti, G.; Reis, J.; Depaulis,
(127) Rodríguez-Muñoz, M.; Sánchez-Blázquez, P.; Garzón, J. A.; Rumbach, L.; Warter, J. M. Spontaneous paroxysmal electro-
Fenfluramine diminishes NMDA receptor-mediated seizures via its clinical patterns in rat: a model of generalized non-convulsive
mixed activity at serotonin 5HT2A and type 1 sigma receptors. epilepsy. Neurosci. Lett. 1982, 33, 97−101. van Luijtelaar, G.;
Oncotarget. 2018, 9, 23373−23389. Zobeiri, M. Progress and outlooks in a genetic absence epilepsy
(128) Salih, H.; Anghelescu, I.; Kezic, I.; Sinha, V.; Hoeben, E.; Van model (WAG/Rij). Curr. Med. Chem. 2014, 21, 704−721. Lopez-
Nueten, L.; De Smedt, H.; De Boer, P. Pharmacokinetic and Santiago, L. F.; Yuan, Y.; Wagnon, J. L.; Hull, J. M.; Frasier, C. R.;
pharmacodynamic characterisation of JNJ-40411813, a positive O’Malley, H. A.; Meisler, M. H.; Isom, L. L. Neuronal hyper-
allosteric modulator of mGluR2, in two randomised, double-blind excitability in a mouse model of SCN8A epileptic encephalopathy.
phase-I studies. J. Psychopharmacol. 2015, 29, 414−425. Proc. Natl. Acad. Sci. U. S. A. 2017, 114, 2383−2388. Singh, N. A.;
(129) Paul, S. M.; Doherty, J. J.; Robichaud, A. J.; Belfort, G. M.; Otto, J. F.; Dahle, E. J.; Pappas, C.; Leslie, J. D.; Vilaythong, A.;
Chow, B. Y.; Hammond, R. S.; Crawford, D. C.; Linsenbardt, A. J.; Noebels, J. L.; White, H. S.; Wilcox, K. S.; Leppert, M. F. Mouse
Shu, H. J.; Izumi, Y.; Mennerick, S. J.; Zorumski, C. F. The major models of human KCNQ2 and KCNQ3 mutations for benign familial
brain cholesterol metabolite 24(S)-hydroxycholesterol is a potent neonatal convulsions show seizures and neuronal plasticity without
allosteric modulator of N-methyl-D-aspartate receptors. J. Neurosci.
synaptic reorganization. J. Physiol. 2008, 586, 3405−3423. Schattling,
2013, 33, 17290−17300.
B.; Fazeli, W.; Engeland, B.; Liu, Y.; Lerche, H.; Isbrandt, D.; Friese,
(130) Brickel, N.; Hewett, K.; Rayner, K.; McDonald, S.; De’Ath, J.;
M. A. Activity of Na(V)1.2 promotes neurodegeneration in an animal
Daniluk, J.; Joshi, K.; Boll, M. C.; Tiamkao, S.; Vorobyeva, O.;
model of multiple sclerosis. JCI Insight 2016, 1, e89810. Yu, F. H.;
Cooper, J. Safety of retigabine in adults with partial-onset seizures
Mantegazza, M.; Westenbroek, R. E.; Robbins, C. A.; Kalume, F.;
after long-term exposure: focus on unexpected ophthalmological and
dermatological events. Epilepsy Behav. 2020, 102, 106580. Burton, K. A.; Spain, W. J.; McKnight, G. S.; Scheuer, T.; Catterall, W.
(131) Bialer, M.; Johannessen, S. I.; Koepp, M. J.; Levy, R. H.; A. Reduced sodium current in GABAergic interneurons in a mouse
Perucca, E.; Perucca, P.; Tomson, T.; White, H. S. Progress report on model of severe myoclonic epilepsy in infancy. Nat. Neurosci. 2006, 9,
new antiepileptic drugs: A summary of the Fifteenth Eilat Conference 1142−1149.
conference on new antiepileptic drugs and devices (EILAT XV). I. (137) Musto, E.; Gardella, E.; Møller, R. S. Recent advances in
Drugs in preclinical and early clinical development. Epilepsia 2020, 61, treatment of epilepsy-related sodium channelopathies. Eur. J. Paediatr
2340−2364. Bialer, M.; Johannessen, S. I.; Koepp, M. J.; Levy, R. H.; Neurol. 2020, 24, 123−128.
Perucca, E.; Perucca, P.; Tomson, T.; White, H. S. Progress report on (138) Catterall, W. A.; Goldin, A. L.; Waxman, S. G. International
new antiepileptic drugs: A summary of the Fifteenth Eilat Conference Union of Pharmacology. XLVII. Nomenclature and structure-function
on new antiepileptic Drugs and devices (EILAT XV). II. Drugs in relationships of voltage-gated sodium channels. Pharmacol Rev. 2005,
more advanced clinical development. Epilepsia 2020, 61, 2365−2385. 57, 397−409.
Bialer, M.; Johannessen, S. I.; Koepp, M. J.; Levy, R. H.; Perucca, E.; (139) Catterall, W. A. Sodium channels, inherited epilepsy, and
Tomson, T.; White, H. S. Progress report on new antiepileptic drugs: antiepileptic drugs. Annu. Rev. Pharmacol. Toxicol. 2014, 54, 317−338.
A summary of the Fourteenth Eilat Conference on new antiepileptic (140) Brodie, M. J. Sodium channel blockers in the treatment of
drugs and devices (EILAT XIV). I. Drugs in preclinical and early epilepsy. CNS Drugs. 2017, 31, 527−534.
clinical development. Epilepsia. 2018, 59, 1811−1841. Bialer, M.; (141) Focken, T.; Burford, K.; Grimwood, M. E.; Zenova, A.;
Johannessen, S. I.; Koepp, M. J.; Levy, R. H.; Perucca, E.; Tomson, T.; Andrez, J. C.; Gong, W.; Wilson, M.; Taron, M.; Decker, S.;
White, H. S. Progress report on new antiepileptic drugs: A summary Lofstrand, V.; Chowdhury, S.; Shuart, N.; Lin, S.; Goodchild, S. J.;
of the Fourteenth Eilat Conference on new antiepileptic drugs and Young, C.; Soriano, M.; Tari, P. K.; Waldbrook, M.; Nelkenbrecher,
devices (EILAT XIV). II. Drugs in more advanced clinical K.; Kwan, R.; Lindgren, A.; de Boer, G.; Lee, S.; Sojo, L.; DeVita, R.
development. Epilepsia. 2018, 59, 1842−1866. Bialer, M.; J.; Cohen, C. J.; Wesolowski, S. S.; Johnson, J. P.; Dehnhardt, C. M.
Johannessen, S. I.; Koepp, M. J.; Levy, R. H.; Perucca, E.; Perucca, Identification of CNS-penetrant aryl sulfonamides as isoform-selective
P.; Tomson, T.; White, H. S. Progress report on new antiepileptic Na(V)1.6 inhibitors with efficacy in mouse models of epilepsy. J. Med.
drugs: A summary of the Sixteenth Eilat Conference on new Chem. 2019, 62, 9618−9641.
antiepileptic drugs and devices (EILAT XVI): II. Drugs in more (142) Johnson, J. P.; Focken, T.; Khakh, K.; Tari, P. K.; Dube, C.;
advanced clinical development. Epilepsia. 2022, 63, 2883−2910. Goodchild, S. J.; Andrez, J. C.; Bankar, G.; Bogucki, D.; Burford, K.;
Steriade, C.; French, J.; Devinsky, O. Epilepsy: key experimental Chang, E.; Chowdhury, S.; Dean, R.; de Boer, G.; Decker, S.;
therapeutics in early clinical development. Expert Opin Investig Drugs. Dehnhardt, C.; Feng, M.; Gong, W.; Grimwood, M.; Hasan, A.;
2020, 29, 373−383. Hussainkhel, A.; Jia, Q.; Lee, S.; Li, J.; Lin, S.; Lindgren, A.; Lofstrand,
(132) Ciccone, L.; Cerri, C.; Nencetti, S.; Orlandini, E. Carbonic V.; Mezeyova, J.; Namdari, R.; Nelkenbrecher, K.; Shuart, N. G.; Sojo,
anhydrase inhibitors and epilepsy: State of the art and future L.; Sun, S.; Taron, M.; Waldbrook, M.; Weeratunge, D.; Wesolowski,
Perspectives. Molecules 2021, 26, 6380. S.; Williams, A.; Wilson, M.; Xie, Z.; Yoo, R.; Young, C.; Zenova, A.;
(133) Borsari, C.; Keles, E.; Rageot, D.; Treyer, A.; Bohnacker, T.; Zhang, W.; Cutts, A. J. NBI-921352, a first-in-class, Na(V)1.6
Bissegger, L.; De Pascale, M.; Melone, A.; Sriramaratnam, R.; Beaufils, selective, sodium channel inhibitor that prevents seizures in Scn8a

4462 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

gain-of-function mice, and wild-type mice and rats. eLife 2022, 11, (158) Cole, B. A.; Johnson, R. M.; Dejakaisaya, H.; Pilati, N.;
e72468. Fishwick, C. W. G.; Muench, S. P.; Lippiat, J. D. Structure-based
(143) Palestro, P. H.; Enrique, N.; Goicoechea, S.; Villalba, M. L.; identification and characterization of inhibitors of the epilepsy-
Sabatier, L. L.; Martin, P.; Milesi, V.; Bruno Blanch, L. E.; Gavernet, associated K(Na)1.1 (KCNT1) potassium channel. iScience. 2020, 23,
L. Searching for New Leads To Treat Epilepsy: Target-based virtual 101100.
screening for the discovery of anticonvulsant agents. J. Chem. Inf (159) Mullen, S. A.; Carney, P. W.; Roten, A.; Ching, M.; Lightfoot,
Model. 2018, 58, 1331−1342. P. A.; Churilov, L.; Nair, U.; Li, M.; Berkovic, S. F.; Petrou, S.;
(144) van der Peet, P. L.; Sandanayake, S.; Jarrott, B. Discovery of Scheffer, I. E. Precision therapy for epilepsy due to KCNT1
N-aryloxypropylbenzylamines as voltage-gated sodium channel Na(V) mutations: a randomized trial of oral quinidine. Neurology. 2018,
1.2-subtype-selective inhibitors. ChemMedChem 2019, 14, 570−582. 90, e67−e72.
(145) Robbins, J. KCNQ potassium channels: physiology, (160) Spitznagel, B. D.; Mishra, N. M.; Qunies, A. M.; Prael, F. J.;
pathophysiology, and pharmacology. Pharmacol Ther. 2001, 90, 1−19. Du, Y.; Kozek, K. A.; Lazarenko, R. M.; Denton, J. S.; Emmitte, K. A.
(146) Rode, F.; Svalø, J.; Sheykhzade, M.; Rønn, L. C. Functional VU0606170, a selective slack channels inhibitor, decreases calcium
effects of the KCNQ modulators retigabine and XE991 in the rat oscillations in cultured cortical neurons. ACS Chem. Neurosci. 2020,
urinary bladder. Eur. J. Pharmacol. 2010, 638, 121−127. Streng, T.; 11, 3658−3671.
Christoph, T.; Andersson, K. E. Urodynamic effects of the K+ channel (161) Griffin, A. M.; Kahlig, K. M.; Hatch, R. J.; Hughes, Z. A.;
(KCNQ) opener retigabine in freely moving, conscious rats. J. Urol. Chapman, M. L.; Antonio, B.; Marron, B. E.; Wittmann, M.;
2004, 172, 2054−2058. Faulkner, M. A.; Burke, R. A. Safety profile of Martinez-Botella, G. Discovery of the first orally available, selective
two novel antiepileptic agents approved for the treatment of refractory K(Na)1.1 inhibitor: In vitro and in vivo activity of an oxadiazole
partial seizures: ezogabine (retigabine) and perampanel. Expert Opin series. ACS Med. Chem. Lett. 2021, 12, 593−602.
Drug Saf. 2013, 12, 847−855. Brickel, N.; Gandhi, P.; (162) Powell, K. L.; Cain, S. M.; Snutch, T. P.; O’Brien, T. J. Low
VanLandingham, K.; Hammond, J.; DeRossett, S. The urinary safety threshold T-type calcium channels as targets for novel epilepsy
profile and secondary renal effects of retigabine (ezogabine): a first-in- treatments. Br. J. Clin. Pharmacol. 2014, 77, 729−739.
class antiepileptic drug that targets KCNQ (K(v)7) potassium (163) Huguenard, J. R. Block of T -type Ca(2+) channels is an
channels. Epilepsia. 2012, 53, 606−612. important action of succinimide antiabsence drugs. Epilepsy Curr.
(147) Kornhuber, J.; Maler, M.; Wiltfang, J.; Bleich, S.; Degner, D.; 2002, 2, 49−52.
Rüther, E. [Neuronal potassium channel opening with flupirtine]. (164) Weiss, N.; Zamponi, G. W. T-type channel druggability at a
Fortschr Neurol Psychiatr. 1999, 67, 466−475. crossroads. ACS Chem. Neurosci. 2019, 10, 1124−1126.
(148) Sampath, D.; Valdez, R.; White, A. M.; Raol, Y. H. (165) Bezençon, O.; Heidmann, B.; Siegrist, R.; Stamm, S.; Richard,
Anticonvulsant effect of flupirtine in an animal model of neonatal S.; Pozzi, D.; Corminboeuf, O.; Roch, C.; Kessler, M.; Ertel, E. A.;
hypoxic-ischemic encephalopathy. Neuropharmacology. 2017, 123, Reymond, I.; Pfeifer, T.; de Kanter, R.; Toeroek-Schafroth, M.;
126−135. Moccia, L. G.; Mawet, J.; Moon, R.; Rey, M.; Capeleto, B.; Fournier,
(149) Withdrawal of Pain Medicine Flupirtine Endorsed; European E. Discovery of a potent, selective T-type calcium channel blocker as a
Medicines Agency, 2018. drug candidate for the treatment of generalized epilepsies. J. Med.
(150) Wurm, K. W.; Bartz, F. M.; Schulig, L.; Bodtke, A.; Bednarski, Chem. 2017, 60, 9769−9789.
P. J.; Link, A. Modifications of the triaminoaryl metabophore of (166) Remen, L.; Bezençon, O.; Simons, L.; Gaston, R.; Downing,
flupirtine and retigabine aimed at avoiding quinone diimine D.; Gatfield, J.; Roch, C.; Kessler, M.; Mosbacher, J.; Pfeifer, T.;
formation. ACS Omega. 2022, 7, 7989−8012. Grisostomi, C.; Rey, M.; Ertel, E. A.; Moon, R. Preparation,
(151) Cho, T.; Uetrecht, J. How reactive metabolites induce an antiepileptic activity, and cardiovascular safety of dihydropyrazoles
immune response that sometimes leads to an idiosyncratic drug as brain-penetrant T-type calcium channel blockers. J. Med. Chem.
reaction. Chem. Res. Toxicol. 2017, 30, 295−314. 2016, 59, 8398−8411.
(152) Groseclose, M. R.; Castellino, S. An investigation into (167) Siegrist, R.; Pozzi, D.; Jacob, G.; Torrisi, C.; Colas, K.;
Retigabine (Ezogabine) associated dyspigmentation in rat eyes by Braibant, B.; Mawet, J.; Pfeifer, T.; de Kanter, R.; Roch, C.; Kessler,
MALDI imaging mass spectrometry. Chem. Res. Toxicol. 2019, 32, M.; Moon, R.; Corminboeuf, O.; Bezençon, O. Correction to
294−303. structure-activity relationship, drug metabolism and pharmacokinetics
(153) Zhou, P.; Zhang, Y.; Xu, H.; Chen, F.; Chen, X.; Li, X.; Pi, X.; properties optimization, and in vvivo studies of new brain penetrant
Wang, L.; Zhan, L.; Nan, F.; Gao, Z. P-retigabine: an N-propargyled triple T-type calcium channel blockers. J. Med. Chem. 2017, 60, 2163.
retigabine with improved brain distribution and enhanced anti- (168) Casillas-Espinosa, P. M.; Hicks, A.; Jeffreys, A.; Snutch, T. P.;
epileptic activity. Mol. Pharmacol. 2015, 87, 31−38. O’Brien, T. J.; Powell, K. L. Z944, a novel selective T-type calcium
(154) Zhang, Y. M.; Xu, H. Y.; Hu, H. N.; Tian, F. Y.; Chen, F.; Liu, channel antagonist delays the progression of seizures in the amygdala
H. N.; Zhan, L.; Pi, X. P.; Liu, J.; Gao, Z. B.; Nan, F. J. Discovery of kindling model. PLoS One. 2015, 10, e0130012.
HN37 as a potent and chemically stable antiepileptic drug candidate. (169) Treiman, D. M. GABAergic mechanisms in epilepsy. Epilepsia
J. Med. Chem. 2021, 64, 5816−5837. 2001, 42, 8−12.
(155) Ostacolo, C.; Miceli, F.; Di Sarno, V.; Nappi, P.; Iraci, N.; (170) Olsen, R. W.; Sieghart, W. International Union of
Soldovieri, M. V.; Ciaglia, T.; Ambrosino, P.; Vestuto, V.; Lauritano, Pharmacology. LXX. Subtypes of gamma-aminobutyric acid(A)
A.; Musella, S.; Pepe, G.; Basilicata, M. G.; Manfra, M.; Perinelli, D. receptors: classification on the basis of subunit composition,
R.; Novellino, E.; Bertamino, A.; Gomez-Monterrey, I. M.; Campiglia, pharmacology, and function. Update. Pharmacol Rev. 2008, 60,
P.; Taglialatela, M. Synthesis and pharmacological characterization of 243−260. Hernandez, C. C.; Macdonald, R. L. A structural look at
conformationally restricted retigabine analogues as novel neuronal GABA(A) receptor mutations linked to epilepsy syndromes. Brain
Kv7 channel activators. J. Med. Chem. 2020, 63, 163−185. Res. 2019, 1714, 234−247.
(156) Zhang, F.; Liu, Y.; Tang, F.; Liang, B.; Chen, H.; Zhang, H.; (171) Janković, S. M.; Dješević, M.; Janković, S. V. Experimental
Wang, K. Electrophysiological and pharmacological characterization GABA A receptor agonists and allosteric modulators for the treatment
of a novel and potent neuronal Kv7 channel opener SCR2682 for of focal epilepsy. J. Exp Pharmacol. 2021, 13, 235−244.
antiepilepsy. Faseb j. 2019, 33, 9154−9166. (172) Witkin, J. M.; Smith, J. L.; Ping, X.; Gleason, S. D.; Poe, M.
(157) Klaerke, D. A.; Tejada, M.; Stolpe; Meinild. Clofilium inhibits M.; Li, G.; Jin, X.; Hobbs, J.; Schkeryantz, J. M.; McDermott, J. S.;
Slick and Slack potassium channels. Biologics 2012, 6, 465−470. Alatorre, A. I.; Siemian, J. N.; Cramer, J. W.; Airey, D. C.; Methuku,
Bearden, D.; Strong, A.; Ehnot, J.; DiGiovine, M.; Dlugos, D.; K. R.; Tiruveedhula, V.; Jones, T. M.; Crawford, J.; Krambis, M. J.;
Goldberg, E. M. Targeted treatment of migrating partial seizures of Fisher, J. L.; Cook, J. M.; Cerne, R. Bioisosteres of ethyl 8-ethynyl-6-
infancy with quinidine. Ann. Neurol. 2014, 76, 457−461. (pyridin-2-yl)-4H-benzo[f]imidazo [1,5-a][1,4]diazepine-3-carboxy-

4463 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

late (HZ-166) as novel alpha 2,3 selective potentiators of GABA(A) Excavating precursors from the traditional Chinese herb Polygala
receptors: Improved bioavailability enhances anticonvulsant efficacy. tenuifolia and Gastrodia elata: Synthesis, anticonvulsant activity
Neuropharmacology. 2018, 137, 332−343. evaluation of 3,4,5-trimethoxycinnamic acid (TMCA) ester deriva-
(173) Owen, R. M.; Blakemore, D. C; Cao, L.; Flanagan, N.; Fish, tives. Bioorg Chem. 2019, 88, 102832. Singh, R. B.; Das, N.; Singh, G.
R.; Gibson, K. R; Gurrell, R.; Huh, C. W.; Kammonen, J.; Mortimer- K.; Singh, S. K.; Zaman, K. Synthesis and pharmacological evaluation
Cassen, E.; Nickolls, S.; Omoto, K.; Owen, D. R; Pike, A.; Pryde, D. of 3-[5-(aryl-[1,3,4]oxadiazole-2-yl]-piperidine derivatives as anti-
C.; Reynolds, D.; Roeloffs, R.; Rose, C. R.; Stead, C.; Takeuchi, M.; convulsant and antidepressant agents. Arabian Journal of Chemistry.
Warmus, J. S; Watson, C. Design and identification of a novel, 2020, 13, 5299−5311.
functionally subtype selective GABA(A) positive allosteric modulator (184) Vogensen, S. B.; Jørgensen, L.; Madsen, K. K.; Borkar, N.;
(PF-06372865). J. Med. Chem. 2019, 62, 5773−5796. Wellendorph, P.; Skovgaard-Petersen, J.; Schousboe, A.; White, H. S.;
(174) Stadler, M.; Monticelli, S.; Seidel, T. Design, synthesis, and Krogsgaard-Larsen, P.; Clausen, R. P. Selective mGAT2 (BGT-1)
pharmacological evaluation of novel β2/3 subunit-selective γ-amino- GABA uptake inhibitors: design, synthesis, and pharmacological
butyric acid type A (GABA(A)) receptor modulators. J. Med. Chem. characterization. J. Med. Chem. 2013, 56, 2160−2164.
2019, 62, 317−341. (185) Vogensen, S. B.; Jørgensen, L.; Madsen, K. K.; Jurik, A.;
(175) Bai, Y.; He, X.; Bai, Y.; Sun, Y.; Zhao, Z.; Chen, X.; Li, B.; Xie, Borkar, N.; Rosatelli, E.; Nielsen, B.; Ecker, G. F.; Schousboe, A.;
J.; Li, Y.; Jia, P.; Meng, X.; Zhao, Y.; Ding, Y.; Xiao, C.; Wang, S.; Yu, Clausen, R. P. Structure activity relationship of selective GABA uptake
J.; Liao, S.; Zhang, Y.; Zhu, Z.; Zhang, Q.; Zhao, Y.; Qin, F.; Zhang, inhibitors. Bioorg. Med. Chem. 2015, 23, 2480−2488.
Y.; Wei, X.; Zeng, M.; Liang, J.; Cuan, Y.; Shan, G.; Fan, T. P.; Wu, B.; (186) Lie, M. E. K.; Kickinger, S.; Skovgaard-Petersen, J.; Ecker, G.
Zheng, X. Polygala tenuifolia-Acori tatarinowii herbal pair as an F.; Clausen, R. P.; Schousboe, A.; White, H. S.; Wellendorph, P.
inspiration for substituted cinnamic α-asaronol esters: Design, Pharmacological characterization of a betaine/GABA transporter 1
synthesis, anticonvulsant activity, and inhibition of lactate dehydro- (BGT1) inhibitor displaying an unusual biphasic inhibition profile
genase study. Eur. J. Med. Chem. 2019, 183, 111650. and anti-seizure effects. Neurochem. Res. 2020, 45, 1551−1565.
(176) Costa, E.; Guidotti, A.; Mao, C. C.; Suria, A. New concepts on (187) Storici, P.; Capitani, G.; De Biase, D.; Moser, M.; John, R. A.;
the mechanism of action of benzodiazepines. Life Sci. 1975, 17, 167− Jansonius, J. N.; Schirmer, T. Crystal structure of GABA-amino-
185. transferase, a target for antiepileptic drug therapy. Biochemistry. 1999,
(177) Atack, J. R. The benzodiazepine binding site of GABA(A) 38, 8628−8634.
receptors as a target for the development of novel anxiolytics. Expert (188) Abdulfatai, U.; Uzairu, A.; Uba, S. Quantitative structure-
Opin Investig Drugs. 2005, 14, 601−618. activity relationship and molecular docking studies of a series of
(178) Mohammadi-Khanaposhtani, M.; Shabani, M.; Faizi, M.; quinazolinonyl analogues as inhibitors of gamma amino butyric acid
Aghaei, I.; Jahani, R.; Sharafi, Z.; Shamsaei Zafarghandi, N.; Mahdavi, aminotransferase. J. Adv. Res. 2017, 8, 33−43.
M.; Akbarzadeh, T.; Emami, S.; Shafiee, A.; Foroumadi, A. Design, (189) Krauss, G.; Faught, E.; Foroozan, R.; Pellock, J. M.; Sergott, R.
synthesis, pharmacological evaluation, and docking study of new C.; Shields, W. D.; Ziemann, A.; Dribinsky, Y.; Lee, D.; Torri, S.;
acridone-based 1,2,4-oxadiazoles as potential anticonvulsant agents. Othman, F.; Isojarvi, J. Sabril® registry 5-year results: Characteristics
Eur. J. Med. Chem. 2016, 112, 91−98. Zhang, H. J.; Shen, Q. K.; Jin, of adult patients treated with vigabatrin. Epilepsy Behav. 2016, 56, 15−
C. M.; Quan, Z. S. Synthesis and pharmacological evaluation of new 19.
3,4-dihydroisoquinolin derivatives containing heterocycle as potential (190) Wild, J. M.; Smith, P. E. M.; Knupp, C. Objective derivation of
anticonvulsant agents. Molecules. 2016, 21, 1635. Kothayer, H.; the morphology and staging of visual field loss associated with long-
Ibrahim, S. M.; Soltan, M. K.; Rezq, S.; Mahmoud, S. S. Synthesis, in term vigabatrin therapy. CNS Drugs. 2019, 33, 817−829.
vivo and in silico evaluation of novel 2,3-dihydroquinazolin-4(1H)- (191) Pan, Y.; Qiu, J.; Silverman, R. B. Design, synthesis, and
one derivatives as potential anticonvulsant agents. Drug Dev Res. 2019, biological activity of a difluoro-substituted, conformationally rigid
80, 343−352. Huang, L.; Ding, J.; Li, M.; Hou, Z.; Geng, Y.; Li, X.; vigabatrin analogue as a potent gamma-aminobutyric acid amino-
Yu, H. Discovery of [1,2,4]-triazolo [1,5-a]pyrimidine-7(4H)-one transferase inhibitor. J. Med. Chem. 2003, 46, 5292−5293. Doumlele,
derivatives as positive modulators of GABA(A1) receptor with potent K.; Conway, E.; Hedlund, J.; Tolete, P.; Devinsky, O. A case report on
anticonvulsant activity and low toxicity. Eur. J. Med. Chem. 2020, 185, the efficacy of vigabatrin analogue (1S, 3S)-3-amino-4-difluorome-
111824. thylenyl-1-cyclopentanoic acid (CPP-115) in a patient with infantile
(179) Bröer, S.; Gether, U. The solute carrier 6 family of spasms. Epilepsy Behav Case Rep. 2016, 6, 67−69.
transporters. Br. J. Pharmacol. 2012, 167, 256−278. Madsen, K. K.; (192) Feja, M.; Meller, S.; Deking, L. S.; Kaczmarek, E.; During, M.
Clausen, R. P.; Larsson, O. M.; Krogsgaard-Larsen, P.; Schousboe, A.; J.; Silverman, R. B.; Gernert, M. OV329, a novel highly potent γ-
White, H. S. Synaptic and extrasynaptic GABA transporters as targets aminobutyric acid aminotransferase inactivator, induces pronounced
for anti-epileptic drugs. J. Neurochem. 2009, 109, 139−144. anticonvulsant effects in the pentylenetetrazole seizure threshold test
(180) Lehre, A. C.; Rowley, N. M.; Zhou, Y.; Holmseth, S.; Guo, C.; and in amygdala-kindled rats. Epilepsia. 2021, 62, 3091−3104.
Holen, T.; Hua, R.; Laake, P.; Olofsson, A. M.; Poblete-Naredo, I.; Juncosa, J. I.; Takaya, K.; Le, H. V.; Moschitto, M. J.; Weerawarna,
Rusakov, D. A.; Madsen, K. K.; Clausen, R. P.; Schousboe, A.; White, P. M.; Mascarenhas, R.; Liu, D.; Dewey, S. L.; Silverman, R. B. Design
H. S.; Danbolt, N. C. Deletion of the betaine-GABA transporter and Mechanism of (S)-3-Amino-4-(difluoromethylenyl)cyclopent-1-
(BGT1; slc6a12) gene does not affect seizure thresholds of adult ene-1-carboxylic Acid, a Highly Potent γ-Aminobutyric Acid Amino-
mice. Epilepsy Res. 2011, 95, 70−81. transferase Inactivator for the Treatment of Addiction. J. Am. Chem.
(181) Kälviäinen, R. Long-term safety of tiagabine. Epilepsia 2001, Soc. 2018, 140, 2151−2164.
42, 46−48. (193) Bialer, M.; Johannessen, S. I.; Koepp, M. J.; Levy, R. H.;
(182) Sałat, K.; Podkowa, A.; Malikowska, N.; Kern, F.; Pabel, J.; Perucca, E.; Perucca, P.; Tomson, T.; White, H. S. Progress report on
Wojcieszak, E.; Kulig, K.; Wanner, K. T.; Strach, B.; Wyska, E. Novel, new antiepileptic drugs: A summary of the Sixteenth Eilat Conference
highly potent and in vivo active inhibitor of GABA transporter on New Antiepileptic Drugs and Devices (EILAT XVI): I. Drugs in
subtype 1 with anticonvulsant, anxiolytic, antidepressant and preclinical and early clinical development. Epilepsia. 2022, 63, 2865−
antinociceptive properties. Neuropharmacology. 2017, 113, 331−342. 2882.
(183) Singh, R. B.; Singh, G. K.; Chaturvedi, K.; Kumar, D.; Singh, (194) Sahu, M.; Siddiqui, N.; Iqbal, R.; Sharma, V.; Wakode, S.
S. K.; Zaman, M. K. Design, synthesis, characterization, and molecular Design, synthesis and evaluation of newer 5,6-dihydropyrimidine-
modeling studies of novel oxadiazole derivatives of nipecotic acid as 2(1H)-thiones as GABA-AT inhibitors for anticonvulsant potential.
potential anticonvulsant and antidepressant agents. Medicinal Bioorg Chem. 2017, 74, 166−178.
Chemistry Research. 2018, 27, 137−152. Zhao, Z.; Bai, Y.; Xie, J.; (195) Sahu, M.; Siddiqui, N.; Sharma, V.; Wakode, S. 5,6-
Chen, X.; He, X.; Sun, Y.; Bai, Y.; Zhang, Y.; Wu, S.; Zheng, X. Dihydropyrimidine-1(2H)-carbothioamides: Synthesis, in vitro

4464 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

GABA-AT screening, anticonvulsant activity and molecular modelling (207) Rothan, H. A.; Amini, E.; Faraj, F. L.; Golpich, M.; Teoh, T.
study. Bioorg Chem. 2018, 77, 56−67. C.; Gholami, K.; Yusof, R. NMDA receptor antagonism with novel
(196) Aboutabl, M. E.; Hassan, R. M.; El-Azzouny, A. A.; Aboul- indolyl, 2-(1,1-Dimethyl-1,3-dihydro-benzo[e]indol-2-ylidene)-malo-
Enein, M. N.; Abd-Allah, W. H. Design and synthesis of novel naldehyde, reduces seizures duration in a rat model of epilepsy. Sci.
parabanic acid derivatives as anticonvulsants. Bioorg Chem. 2020, 94, Rep. 2017, 7, 45540.
103473. (208) Supuran, C. T. Carbonic anhydrases: novel therapeutic
(197) Rogawski, M. A. Revisiting AMPA receptors as an applications for inhibitors and activators. Nat. Rev. Drug Discovery
antiepileptic drug target. Epilepsy Curr. 2011, 11, 56−63. Lee, K.; 2008, 7, 168−181. Mishra, C. B.; Tiwari, M.; Supuran, C. T. Progress
Goodman, L.; Fourie, C.; Schenk, S.; Leitch, B.; Montgomery, J. M. in the development of human carbonic anhydrase inhibitors and their
AMPA receptors as therapeutic targets for neurological disorders. Adv. pharmacological applications: Where are we today? Med. Res. Rev.
Protein Chem. Struct Biol. 2016, 103, 203−261. 2020, 40, 2485−2565. Alterio, V.; Di Fiore, A.; D’Ambrosio, K.;
(198) French, J. A.; Krauss, G. L.; Biton, V.; Squillacote, D.; Yang, Supuran, C. T.; De Simone, G. Multiple binding modes of inhibitors
H.; Laurenza, A.; Kumar, D.; Rogawski, M. A. Adjunctive perampanel to carbonic anhydrases: how to design specific drugs targeting 15
for refractory partial-onset seizures: randomized phase III study 304. different isoforms? Chem. Rev. 2012, 112, 4421−4468.
Neurology. 2012, 79, 589−596. (209) Ruusuvuori, E.; Kaila, K. Carbonic anhydrases and brain pH in
(199) Jackson, A. C.; Nicoll, R. A. The expanding social network of the control of neuronal excitability. Subcell Biochem. 2014, 75, 271−
ionotropic glutamate receptors: TARPs and other transmembrane 290.
auxiliary subunits. Neuron. 2011, 70, 178−199. Schwenk, J.; Harmel, (210) Tang, C. M.; Dichter, M.; Morad, M. Modulation of the N-
N.; Brechet, A.; Zolles, G.; Berkefeld, H.; Müller, C. S.; Bildl, W.; methyl-D-aspartate channel by extracellular H+. Proc. Natl. Acad. Sci.
Baehrens, D.; Hüber, B.; Kulik, A.; Klöcker, N.; Schulte, U.; Fakler, B. U. S. A. 1990, 87, 6445−6449. Ruusuvuori, E.; Huebner, A. K;
High-resolution proteomics unravel architecture and molecular Kirilkin, I.; Yukin, A. Y; Blaesse, P.; Helmy, M.; Jung Kang, H.; El
diversity of native AMPA receptor complexes. Neuron. 2012, 74, Muayed, M.; Christopher Hennings, J; Voipio, J.; Sestan, N.; Hubner,
621−633. C. A; Kaila, K. Neuronal carbonic anhydrase VII provides GABAergic
(200) Kato, A. S.; Burris, K. D.; Gardinier, K. M.; Gernert, D. L.; excitatory drive to exacerbate febrile seizures. EMBO J. 2013, 32,
Porter, W. J.; Reel, J.; Ding, C.; Tu, Y.; Schober, D. A.; Lee, M. R.; 2275−2286.
Heinz, B. A.; Fitch, T. E.; Gleason, S. D.; Catlow, J. T.; Yu, H.; (211) Ilies, M. A.; Masereel, B.; Rolin, S.; Scozzafava, A.; Câmpeanu,
Fitzjohn, S. M.; Pasqui, F.; Wang, H.; Qian, Y.; Sher, E.; Zwart, R.; G.; Cîmpeanu, V.; Supuran, C. T. Carbonic anhydrase inhibitors:
Wafford, K. A.; Rasmussen, K.; Ornstein, P. L.; Isaac, J. T.; aromatic and heterocyclic sulfonamides incorporating adamantyl
Nisenbaum, E. S.; Bredt, D. S.; Witkin, J. M. Forebrain-selective moieties with strong anticonvulsant activity. Bioorg. Med. Chem.
AMPA-receptor antagonism guided by TARP γ-8 as an antiepileptic 2004, 12, 2717−2726. Herrero, A. I.; Del Olmo, N.; González-
mechanism. Nat. Med. 2016, 22, 1496−1501. Escalada, J. R.; Solís, J. M. Two new actions of topiramate: inhibition
(201) Gardinier, K. M.; Gernert, D. L.; Porter, W. J.; Reel, J. K.; of depolarizing GABA(A)-mediated responses and activation of a
Ornstein, P. L.; Spinazze, P.; Stevens, F. C.; Hahn, P.; Hollinshead, S. potassium conductance. Neuropharmacology 2002, 42, 210−220.
Katayama, F.; Miura, H.; Takanashi, S. Long-term effectiveness and
P.; Mayhugh, D.; Schkeryantz, J.; Khilevich, A.; De Frutos, O.;
side effects of acetazolamide as an adjunct to other anticonvulsants in
Gleason, S. D.; Kato, A. S.; Luffer-Atlas, D.; Desai, P. V.; Swanson, S.;
the treatment of refractory epilepsies. Brain Dev. 2002, 24, 150−154.
Burris, K. D.; Ding, C.; Heinz, B. A.; Need, A. B.; Barth, V. N.;
(212) Anderson, R. E.; Chiu, P.; Woodbury, D. M. Mechanisms of
Stephenson, G. A.; Diseroad, B. A.; Woods, T. A.; Yu, H.; Bredt, D.;
tolerance to the anticonvulsant effects of acetazolamide in mice:
Witkin, J. M. Discovery of the first α-Amino-3-hydroxy-5-methyl-4-
relation to the activity and amount of carbonic anhydrase in brain.
isoxazolepropionic acid (AMPA) receptor antagonist dependent upon Epilepsia. 1989, 30, 208−216. White, J. R.; Walczak, T. S.; Marino, S.
transmembrane AMPA receptor regulatory protein (TARP) γ-8. J. E.; Beniak, T. E.; Leppik, I. E.; Birnbaum, A. K. Zonisamide
Med. Chem. 2016, 59, 4753−4768. discontinuation due to psychiatric and cognitive adverse events: a
(202) Maher, M. P.; Wu, N.; Ravula, S.; Ameriks, M. K.; Savall, B. case-control study. Neurology. 2010, 75, 513−518.
M.; Liu, C.; Lord, B.; Wyatt, R. M.; Matta, J. A.; Dugovic, C.; Yun, S.; (213) Lakkis, M. M.; O’Shea, K. S.; Tashian, R. E. Differential
Ver Donck, L.; Steckler, T.; Wickenden, A. D.; Carruthers, N. I.; expression of the carbonic anhydrase genes for CA VII (Car7) and
Lovenberg, T. W. Discovery and characterization of AMPA receptor CA-RP VIII (Car8) in mouse brain. J. Histochem Cytochem. 1997, 45,
modulators selective for TARP-γ8. J. Pharmacol Exp Ther. 2016, 357, 657−662. Nocentini, A.; Supuran, C. T. Advances in the structural
394−414. annotation of human carbonic anhydrases and impact on future drug
(203) Ravula, S.; Savall, B. M.; Wu, N.; Lord, B.; Coe, K.; Wang, K.; discovery. Expert Opin Drug Discovery 2019, 14, 1175−1197.
Seierstad, M.; Swanson, D. M.; Ziff, J.; Nguyen, M.; Leung, P.; (214) Fisher, S. Z.; Govindasamy, L.; Boyle, N.; Agbandje-McKenna,
Rynberg, R.; La, D.; Pippel, D. J.; Koudriakova, T.; Lovenberg, T. W.; M.; Silverman, D. N.; Blackburn, G. M.; McKenna, R. X-ray
Carruthers, N. I.; Maher, M. P.; Ameriks, M. K. Lead optimization of crystallographic studies reveal that the incorporation of spacer groups
5-aryl benzimidazolone- and oxindole-based AMPA receptor modu- in carbonic anhydrase inhibitors causes alternate binding modes. Acta
lators selective for TARP γ-8. ACS Med. Chem. Lett. 2018, 9, 821− Crystallogr. Sect F Struct Biol. Cryst. Commun. 2006, 62, 618−622.
826. (215) Mishra, C. B.; Kumari, S.; Angeli, A.; Monti, S. M.; Buonanno,
(204) Zhu, X.; Dong, J.; Shen, K.; Bai, Y.; Zhang, Y.; Lv, X.; Chao, J.; M.; Tiwari, M. Discovery of benzenesulfonamides with potent human
Yao, H. NMDA receptor NR2B subunits contribute to PTZ-kindling- carbonic anhydrase inhibitory and effective anticonvulsant action:
induced hippocampal astrocytosis and oxidative stress. Brain Res. Bull. Design, synthesis, and pharmacological assessment. J. Med. Chem.
2015, 114, 70−78. Di Maio, R.; Mastroberardino, P. G.; Hu, X.; 2017, 60, 2456−2469.
Montero, L.; Greenamyre, J. T. Pilocapine alters NMDA receptor (216) Mishra, C. B.; Kumari, S.; Angeli, A.; Bua, S.; Buonanno, M.;
expression and function in hippocampal neurons: NADPH oxidase Monti, S. M.; Tiwari, M.; Supuran, C. T. Discovery of potent anti-
and ERK1/2 mechanisms. Neurobiol Dis. 2011, 42, 482−495. convulsant carbonic anhydrase inhibitors: Design, synthesis, in vitro
(205) Zanos, P.; Moaddel, R.; Morris, P. J.; Riggs, L. M.; Highland, and in vivo appraisal. Eur. J. Med. Chem. 2018, 156, 430−443.
J. N.; Georgiou, P.; Pereira, E. F. R.; Albuquerque, E. X.; Thomas, C. (217) Mishra, C. B.; Kumari, S.; Angeli, A.; Bua, S.; Tiwari, M.
J.; Zarate, C. A., Jr.; Gould, T. D. Ketamine and ketamine metabolite Discovery of benzenesulfonamide derivatives as carbonic anhydrase
pharmacology: Insights into therapeutic mechanisms. Pharmacol Rev. inhibitors with effective anticonvulsant action: Design, synthesis, and
2018, 70, 621−660. pharmacological evaluation. J. Med. Chem. 2018, 61, 3151−3165.
(206) Czapiński, P.; Blaszczyk, B.; Czuczwar, S. J. Mechanisms of (218) Mishra, C. B.; Kumari, S.; Angeli, A.; Bua, S.; Mongre, R. K.;
action of antiepileptic drugs. Curr. Top Med. Chem. 2005, 5, 3−14. Tiwari, M.; Supuran, C. T. Discovery of potent carbonic anhydrase

4465 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

inhibitors as effective anticonvulsant agents: Drug design, synthesis, (232) Walter, M.; Stark, H. Histamine receptor subtypes: a century
and in vitro and in vivo investigations. J. Med. Chem. 2021, 64, 3100− of rational drug design. Front Biosci (Schol Ed). 2012, 4, 461−488.
3114. (233) Berlin, M.; Boyce, C. W.; de Lera Ruiz, M. Histamine H3
(219) Yang, H.; Rudge, D. G.; Koos, J. D.; Vaidialingam, B.; Yang, receptor as a drug discovery target. J. Med. Chem. 2011, 54, 26−53.
H. J.; Pavletich, N. P. mTOR kinase structure, mechanism and (234) Sadek, B.; Saad, A.; Subramanian, D.; Shafiullah, M.;
regulation. Nature. 2013, 497, 217−223. Łażewska, D.; Kieć-Kononowiczc, K. Anticonvulsant and procognitive
(220) Salussolia, C. L.; Klonowska, K.; Kwiatkowski, D. J.; Sahin, M. properties of the non-imidazole histamine H3 receptor antagonist
Genetic etiologies, diagnosis, and treatment of tuberous sclerosis DL77 in male adult rats. Neuropharmacology. 2016, 106, 46−55.
complex. Annu. Rev. Genomics Hum Genet. 2019, 20, 217−240. Alachkar, A.; Łażewska, D.; Latacz, G.; Frank, A.; Siwek, A.; Lubelska,
(221) Lechuga, L.; Franz, D. N. Everolimus as adjunctive therapy for A.; Honkisz-Orzechowska, E.; Handzlik, J.; Stark, H.; Kieć-
tuberous sclerosis complex-associated partial-onset seizures. Expert Kononowicz, K.; Sadek, B. Studies on anticonvulsant effects of
Rev. Neurother. 2019, 19, 913−925. novel histamine H3R antagonists in electrically and chemically
(222) Krueger, D. A.; Wilfong, A. A.; Mays, M.; Talley, C. M.; induced seizures in rats. Int. J. Mol. Sci. 2018, 19, 3386. Szczepańska,
Agricola, K.; Tudor, C.; Capal, J.; Holland-Bouley, K.; Franz, D. N. K.; Karcz, T.; Mogilski, S.; Siwek, A.; Kuder, K. J.; Latacz, G.;
Long-term treatment of epilepsy with everolimus in tuberous Kubacka, M.; Hagenow, S.; Lubelska, A.; Olejarz, A.; Kotańska, M.;
sclerosis. Neurology. 2016, 87, 2408−2415. Sadek, B.; Stark, H.; Kieć-Kononowicz, K. Synthesis and biological
(223) Beaufils, F.; Cmiljanovic, N.; Cmiljanovic, V.; Bohnacker, T.; activity of novel tert-butyl and tert-pentylphenoxyalkyl piperazine
Melone, A.; Marone, R.; Jackson, E.; Zhang, X.; Sele, A.; Borsari, C. 5- derivatives as histamine H(3)R ligands. Eur. J. Med. Chem. 2018, 152,
(4,6-Dimorpholino-1,3,5-triazin-2-yl)-4-(trifluoromethyl)pyridin-2- 223−234. Bastaki, S. M.; Abdulrazzaq, Y. M.; Shafiullah, M.; Więcek,
amine (PQR309), a potent, brain-penetrant, orally bioavailable, pan- M.; Kieć-Kononowicz, K.; Sadek, B. Anticonvulsant and reproductive
class I PI3K/mTOR inhibitor as clinical candidate in oncology. J. toxicological studies of the imidazole-based histamine H3R antagonist
Med. Chem. 2017, 60, 7524−7538. 2−18 in mice. Drug Des Devel Ther. 2018, 12, 179−194. Song, M.;
(224) Rageot, D.; Bohnacker, T.; Melone, A.; Langlois, J. B.; Borsari, Yan, R.; Zhang, Y.; Guo, D.; Zhou, N.; Deng, X. Design, synthesis,
C. Discovery and Preclinical Characterization of 5-[4,6-Bis({3-oxa-8- and anticonvulsant effects evaluation of nonimidazole histamine H(3)
azabicyclo[3.2.1]octan-8-yl})-1,3,5-triazin-2-yl]-4-(difluoromethyl)- receptor antagonists/inverse agonists containing triazole moiety. J.
pyridin-2-amine (PQR620), a highly potent and selective mTORC1/ Enzyme Inhib Med. Chem. 2020, 35, 1310−1321.
2 inhibitor for cancer and neurological disorders. J. Med. Chem. 2018, (235) Iliff, J. J.; Close, L. N.; Selden, N. R.; Alkayed, N. J. A novel
61, 10084−10105. role for P450 eicosanoids in the neurogenic control of cerebral blood
(225) Venkatesan, A. M.; Chen, Z.; dos Santos, O.; Dehnhardt, C.; flow in the rat. Exp Physiol. 2007, 92, 653−658. Spiecker, M.; Liao, J.
de los Santos, E. D.; Ayral-Kaloustian, S.; Mallon, R.; Hollander, I.; K. Vascular protective effects of cytochrome p450 epoxygenase-
Feldberg, L.; Lucas, J.; Yu, K.; Chaudhary, I.; Mansour, T. S. PKI-179: derived eicosanoids. Arch. Biochem. Biophys. 2005, 433, 413−420.
an orally efficacious dual phosphatidylinositol-3-kinase (PI3K)/ (236) Morisseau, C.; Hammock, B. D. Impact of soluble epoxide
mammalian target of rapamycin (mTOR) inhibitor. Bioorg. Med. hydrolase and epoxyeicosanoids on human health. Annu. Rev.
Chem. Lett. 2010, 20, 5869−5873. Pharmacol. Toxicol. 2013, 53, 37−58.
(226) Rageot, D.; Bohnacker, T.; Keles, E.; McPhail, J. A.; (237) Inceoglu, B.; Jinks, S. L.; Schmelzer, K. R.; Waite, T.; Kim, I.
Hoffmann, R. M.; Melone, A.; Borsari, C. (S)-4-(Difluoromethyl)-5- H.; Hammock, B. D. Inhibition of soluble epoxide hydrolase reduces
(4-(3-methylmorpholino)-6-morpholino-1,3,5-triazin-2-yl)pyridin-2- LPS-induced thermal hyperalgesia and mechanical allodynia in a rat
amine (PQR530), a potent, orally bioavailable, and brain-penetrable model of inflammatory pain. Life Sci. 2006, 79, 2311−2319.
dual inhibitor of class I PI3K and mTOR kinase. J. Med. Chem. 2019, (238) Vito, S. T.; Austin, A. T.; Banks, C. N.; Inceoglu, B.; Bruun, D.
62, 6241−6261. Brandt, C.; Hillmann, P.; Noack, A.; Römermann, K.; A.; Zolkowska, D.; Tancredi, D. J.; Rogawski, M. A.; Hammock, B. D.;
Ö hler, L. A.; Rageot, D.; Beaufils, F.; Melone, A.; Sele, A. M.; Lein, P. J. Post-exposure administration of diazepam combined with
Wymann, M. P.; Fabbro, D.; Löscher, W. The novel, catalytic soluble epoxide hydrolase inhibition stops seizures and modulates
mTORC1/2 inhibitor PQR620 and the PI3K/mTORC1/2 inhibitor neuroinflammation in a murine model of acute TETS intoxication.
PQR530 effectively cross the blood-brain barrier and increase seizure Toxicol. Appl. Pharmacol. 2014, 281, 185−194.
threshold in a mouse model of chronic epilepsy. Neuropharmacology. (239) Shen, Y.; Peng, W.; Chen, Q.; Hammock, B. D.; Liu, J.; Li, D.;
2018, 140, 107−120. Yang, J.; Ding, J.; Wang, X. Anti-inflammatory treatment with a
(227) Ricciotti, E.; FitzGerald, G. A. Prostaglandins and soluble epoxide hydrolase inhibitor attenuates seizures and epilepsy-
inflammation. Arterioscler Thromb Vasc Biol. 2011, 31, 986−1000. associated depression in the LiCl-pilocarpine post-status epilepticus
Dey, A.; Kang, X.; Qiu, J.; Du, Y.; Jiang, J. Anti-inflammatory small rat model. Brain Behav Immun. 2019, 81, 535−544.
molecules to treat seizures and epilepsy: From bench to bedside. (240) Margineanu, D. G. Systems biology, complexity, and the
Trends Pharmacol. Sci. 2016, 37, 463−484. impact on antiepileptic drug discovery. Epilepsy Behav. 2014, 38,
(228) Akula, K. K.; Dhir, A.; Kulkarni, S. K. Rofecoxib, a selective 131−142.
cyclooxygenase-2 (COX-2) inhibitor increases pentylenetetrazol (241) Brodie, M. J.; Covanis, A.; Gil-Nagel, A.; Lerche, H.; Perucca,
seizure threshold in mice: possible involvement of adenosinergic E.; Sills, G. J.; White, H. S. Antiepileptic drug therapy: does
mechanism. Epilepsy Res. 2008, 78, 60−70. Ma, L.; Cui, X. L.; Wang, mechanism of action matter? Epilepsy Behav. 2011, 21, 331−341.
Y.; Li, X. W.; Yang, F.; Wei, D.; Jiang, W. Aspirin attenuates (242) Abram, M.; Zagaja, M.; Mogilski, S.; Andres-Mach, M.;
spontaneous recurrent seizures and inhibits hippocampal neuronal Latacz, G.; Baś, S.; Łuszczki, J. J.; Kieć-Kononowicz, K.; Kamiński, K.
loss, mossy fiber sprouting and aberrant neurogenesis following Multifunctional hybrid compounds derived from 2-(2,5-dioxopyrro-
pilocarpine-induced status epilepticus in rats. Brain Res. 2012, 1469, lidin-1-yl)-3-methoxypropanamides with anticonvulsant and antino-
103−113. ciceptive properties. J. Med. Chem. 2017, 60, 8565−8579.
(229) DeBisschop, M. What are the risks of long-term NSAIDs and (243) Abram, M.; Rapacz, A.; Mogilski, S.; Latacz, G.; Lubelska, A.;
COX-2 inhibitors? J. Fam. Pract. 2003, 52, 199−200. Kamiński, R. M.; Kamiński, K. Multitargeted compounds derived
(230) Mishra, C. B.; Kumari, S.; Prakash, A.; Yadav, R.; Tiwari, A. from (2,5-dioxopyrrolidin-1-yl)(phenyl)-acetamides as candidates for
K.; Pandey, P.; Tiwari, M. Discovery of novel Methylsulfonyl phenyl effective anticonvulsant and antinociceptive agents. ACS Chem.
derivatives as potent human Cyclooxygenase-2 inhibitors with Neurosci. 2020, 11, 1996−2008.
effective anticonvulsant action: Design, synthesis, in-silico, in-vitro (244) Abram, M.; Jakubiec, M.; Rapacz, A. Identification of new
and in-vivo evaluation. Eur. J. Med. Chem. 2018, 151, 520−532. compounds with anticonvulsant and antinociceptive properties in a
(231) Parsons, M. E.; Ganellin, C. R. Histamine and its receptors. Br. group of 3-substituted (2,5-dioxo-pyrrolidin-1-yl)(phenyl)-acet-
J. Pharmacol. 2006, 147, S127. amides. Int. J. Mol. Sci. 2021, 22, 13092.

4466 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467
Journal of Medicinal Chemistry pubs.acs.org/jmc Perspective

(245) Jakubiec, M.; Abram, M.; Zagaja, M.; Andres-Mach, M.;


Szewczyk, A.; Latacz, G.; Szulczyk, B.; Socała, K.; Nieoczym, D.; Wlaź,
P.; Metcalf, C. S.; Wilcox, K.; Kamiński, R. M.; Kamiński, K. New
phenylglycinamide derivatives with hybrid structure as candidates for
new broad-spectrum anticonvulsants. Cells. 2022, 11, 1862.
(246) Kamiński, K.; Zagaja, M.; Rapacz, A.; Łuszczki, J. J.; Andres-
Mach, M.; Abram, M.; Obniska, J. New hybrid molecules with
anticonvulsant and antinociceptive activity derived from 3-methyl- or
3,3-dimethyl-1-[1-oxo-1-(4-phenylpiperazin-1-yl)propan-2-yl]-
pyrrolidine-2,5-diones. Bioorg. Med. Chem. 2016, 24, 606−618. Góra,
M.; Czopek, A. Synthesis, anticonvulsant and antinociceptive activity
of new hybrid compounds: Derivatives of 3-(3-Methylthiophen-2-yl)-
pyrrolidine-2,5-dione. Int. J. Mol. Sci. 2020, 21, 5750. Rapacz, A.;
Obniska, J.; Wiklik-Poudel, B.; Rybka, S.; Sałat, K.; Filipek, B.
Anticonvulsant and antinociceptive activity of new amides derived
from 3-phenyl-2,5-dioxo-pyrrolidine-1-yl-acetic acid in mice. Eur. J.
Pharmacol. 2016, 781, 239−249. Góra, M.; Czopek, A. Design,
synthesis and biological activity of new amides derived from 3-
benzhydryl and 3-sec-butyl-2,5-dioxo-pyrrolidin-1-yl-acetic acid.
ChemMedChem 2021, 16, 1619−1630. Góra, M.; Czopek, A.
Synthesis, anticonvulsant, and antinociceptive activity of new 3-(2-
chlorophenyl)- and 3-(3-chlorophenyl)-2,5-dioxo-pyrrolidin-1-yl-acet-
amides. Molecules 2021, 26, 1664.
(247) Emami, S.; Valipour, M.; Kazemi Komishani, F.; Sadati-
Ashrafi, F.; Rasoulian, M.; Ghasemian, M.; Tajbakhsh, M.;
Honarchian Masihi, P.; Shakiba, A.; Irannejad, H.; Ahangar, N.
Synthesis, in silico, in vitro and in vivo evaluations of isatin
aroylhydrazones as highly potent anticonvulsant agents. Bioorg
Chem. 2021, 112, 104943.
(248) Coleman, N.; Nguyen, H. M.; Cao, Z.; Brown, B. M.; Jenkins,
D. P.; Zolkowska, D.; Chen, Y. J.; Tanaka, B. S.; Goldin, A. L.;
Rogawski, M. A.; Pessah, I. N.; Wulff, H. The riluzole derivative 2-
amino-6-trifluoromethylthio-benzothiazole (SKA-19), a mixed KCa2
activator and NaV blocker, is a potent novel anticonvulsant.
Neurotherapeutics. 2015, 12, 234−249.
(249) Premoli, I.; Rossini, P. G.; Goldberg, P. Y.; Posadas, K.;
Green, L.; Yogo, N.; Pimstone, S.; Abela, E.; Beatch, G. N.; Recommended by ACS
Richardson, M. P. TMS as a pharmacodynamic indicator of cortical
activity of a novel anti-epileptic drug, XEN1101. Ann. Clin Transl
Neurol. 2019, 6, 2164−2174. Classics in Chemical Neuroscience: Selegiline, Isocarboxazid,
(250) Tian, F.; Cao, B.; Xu, H.; Zhan, L.; Nan, F.; Li, N.; Phenelzine, and Tranylcypromine
Taglialatela, M.; Gao, Z. Epilepsy phenotype and response to KCNQ Gavin R. Hoffman, Maureen E. Stabio, et al.
openers in mice harboring the Kcnq2 R207W voltage-sensor NOVEMBER 15, 2023
mutation. Neurobiol Dis. 2022, 174, 105860. ACS CHEMICAL NEUROSCIENCE READ
(251) Richard, M.; Kaufmann, P.; Ort, M.; Kornberger, R.;
Dingemanse, J. Multiple-ascending dose study in healthy subjects to Antidepressant-like Effect of 1-(2-(4-(4-Ethylphenyl)-1H-
assess the pharmacokinetics, tolerability, and CYP3A4 interaction 1,2,3-triazol-1-yl)phenyl)ethan-1-one in Mice: Evidence of the
potential of the T-type calcium channel blocker ACT-709478, a
Contribution of the Serotonergic System
potential new antiepileptic drug. CNS Drugs. 2020, 34, 311−323.
(252) Specchio, N.; Pietrafusa, N.; Perucca, E.; Cross, J. H. New Marcelo Heinemann Presa, César Augusto Brüning, et al.
paradigms for the treatment of pediatric monogenic epilepsies: JUNE 09, 2023
Progressing toward precision medicine. Epilepsy Behav. 2022, 131, ACS CHEMICAL NEUROSCIENCE READ
107961.
(253) Higurashi, N.; Broccoli, V.; Hirose, S. Genetics and gene Palmitoylation Regulates Human Serotonin Transporter
therapy in Dravet syndrome. Epilepsy Behav. 2022, 131, 108043. Activity, Trafficking, and Expression and Is Modulated by
Escitalopram
Christopher R. Brown and James D. Foster
AUGUST 29, 2023
ACS CHEMICAL NEUROSCIENCE READ

Novel Psilocin Prodrugs with Altered Pharmacological


Properties as Candidate Therapies for Treatment-Resistant
Anxiety Disorders
Sheetal A. Raithatha, Peter J. Facchini, et al.
NOVEMBER 20, 2023
JOURNAL OF MEDICINAL CHEMISTRY READ
Get More Suggestions >

4467 https://doi.org/10.1021/acs.jmedchem.2c01975
J. Med. Chem. 2023, 66, 4434−4467

You might also like