Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jun 1.
Published in final edited form as: Pharmacol Ther. 2021 Nov 16;234:108035. doi: 10.1016/j.pharmthera.2021.108035

GABAkines – Advances in the Discovery, Development, and Commercialization of Positive Allosteric Modulators of GABAA Receptors1

Rok Cerne 1,2,3,4, Arnold Lippa 3, Michael M Poe 3, Jodi L Smith 1, Xiaoming Jin 4, Xingjie Ping 4, Lalit K Golani 5, James M Cook 3,5, Jeffrey M Witkin 1,3,5
PMCID: PMC9787737  NIHMSID: NIHMS1756985  PMID: 34793859

Abstract

Positive allosteric modulators of γ-aminobutyric acid-A (GABAA) receptors or GABAkines have been widely used medicines for over 70 years for anxiety, epilepsy, sleep, and other disorders. Traditional GABAkines like diazepam have safety and tolerability concerns that include sedation, motor-impairment, respiratory depression, tolerance and dependence. Multiple GABAkines have entered clinical development but the issue of side-effects has not been fully solved. The present review focuses on the new GABAkines in. The compounds that are presently being developed and commercialized include several neuroactive steroids (an allopregnanolone formulation (brexanolone), an allopregnanolone prodrug (LYT-300), Sage-324, zuranolone, and ganaxolone), the α2/3-preferring GABAkine, KRM-II-81, and the α2/3/5-preferring GABAkine PF-06372865 (darigabat). The neuroactive steroids are in clinical development for post-partum depression, intractable epilepsy, tremor, status epilepticus, and genetic epilepsy disorders. Darigabat is in development for epilepsy and anxiety. The imidazodiazepine, KRM-II-81 is efficacious in animal models for the treatment of epilepsy and post-traumatic epilepsy, acute and chronic pain, as well as anxiety and depression. The efficacy of KRM-II-81 in models of pharmacoresistant epilepsy, preventing the development of seizure sensitization, and in brain tissue of intractable epileptic patients bodes well for improved therapeutics. Medicinal chemistry efforts are also ongoing to identify novel and improved GABAkines. The data document gaps in our understanding of the molecular pharmacology of GABAkines that drive differential pharmacological profiles, but emphasize advancements in the ability to successfully utilize GABAA receptor potentiation for therapeutic gain in neurology and psychiatry.

Keywords: GABAkines, neuroactive steroids, KRM-II-81, darigabat, epilepsy, anxiety, depression, pain

I. Introduction.

Physiology and behavior are modulated by the nervous system through a tightly regulated balance of excitatory and inhibitory processes. Multiple neurotransmitters participate in this intricate control network where the primary excitatory neurotransmitter in the mammalian nervous system, glutamate, is offset by the principal inhibitory neurotransmitter, γ-aminobutyric acid or GABA (Fig. 1). These balancing processes of excitation and inhibition work in concert to finely tune physiological and behavioral function.

Figure 1.

Figure 1.

Structures of GABA and the direct-acting GABAA receptor agonists muscimol, gaboxadol (also known as THIP), and baclofen.

The importance of GABA receptor modulation for medical practice is highlighted by the large number of GABA receptor modulators that have been and continue to be highly valued therapeutic agents. The present review focuses on the positive allosteric modulators of GABA (known as GABAkines) that have recently been commercialized or are in development for the treatment of neurological and psychiatric disorders.

Upon its release, GABA activates two classes of GABA receptors, GABAA and GABAB that differ in structure, biophysical properties, and pharmacology. GABAA (ionotropic) receptors (GABAARs) are rapidly-responding ligand-gated ion channels belonging to the Cys-loop super-family (Hevers and Lüddens, 1998; Miller and Smart, 2010). This family also includes nicotinic acetylcholine, glycine and serotonin subtype 3 receptors and is characterized by a loop formed by 13 highly conserved amino acids between two cysteine (Cys) residues (Miller and Smart, 2010; Thompson et al., 2010), pentameric subunit complexes that form a central ion pore (Sine and Engel, 2006). The binding of GABA to the orthosteric binding site on the extracellular domain of GABAA receptors (GABAARs) opens permeability to chloride ions with a high degree of selectivity against other anions (Kaila and Voipio, 1987; Sigel and Steinmann, 2012). The direction of flow is, in most cases, from outside to inside the neuron, resulting in hyperpolarization and reduced neuronal excitability. In addition, the increased conductance can result in shunting inhibition and modulation of neuronal gain (Chance et al., 2002; Mitchell and Silver, 2003; Prescott and Koninck, 2003; Mody and Pearce, 2004). However, depolarizing GABAAR currents have been reported in immature neurons, and under pathological conditions (Coull et al., 2005) with a proposed role in development and in neuroplasticity (Ben-Ari et al., 2007; Deidda et al., 2015; Ohtawa et al., 2017). In addition to its synaptic localization, where they mediate phasic transmission, GABAARs are also present extrasynaptically where they are activated by low concentrations of ambient GABA and mediate tonic inhibition (Farrant and Nusser, 2005; Belelli et al., 2009).

Slow acting GABAB (metabotropic) receptors are members of the class C, G protein-coupled receptor family (Kaupmann et al., 1997). The receptors are mandatory heterodimers of GABAB1 and GABAB2 subunits with each subunit composed of an extracellular Venus Flytrap domain and a transmembrane domain of seven α-helices (Mao et al., 2020; Papasergi-Scott et al., 2020; Park et al., 2020; Shaye et al., 2020). Recent cryogenic electron microscopy studies have helped resolve multiple conformations of the receptor, providing insight into receptor function and pharmacology (Mao et al., 2020; Park et al., 2020; Shaye et al., 2020). GABAB receptors act through activation of pertussis toxin-sensitive G proteins (Knight and Bowery, 1996) resulting in a prolonged decrease in neuronal excitability through the inhibition of adenylyl cyclase and voltage-gated Ca2+ channels, as well as the opening of G protein-coupled inward rectifying K+ channels (Bettler et al., 2004; Gassmann and Bettler, 2012). GABAB receptors have been implicated in pathophysiological mechanisms of multiple neuronal processes and have been proposed as a drug target for a range of disorders including spasticity, pain, cough, bladder dysfunction, drug addiction, and epilepsy (Enna, 1997; Kumar et al., 2013). Substantial effort has been devoted to the discovery of compounds for GABABRs, but the compounds generally suffered from lack of efficacy, non-drug-like pharmacokinetic profiles, and side effects at sub-efficacious doses (Evenseth et al., 2020). Despite its poor brain penetration and rapid tolerance development, baclofen remains the only approved drug targeting GABABRs as an antispasmodic agent and muscle relaxant (Ertzgaard et al., 2017).

The primary excitatory neurotransmitter in the mammalian nervous system, glutamate, initiates excitatory changes in local neuronal circuits through binding to glutamate receptors. Glutamate receptors exist as both ionotropic receptors (iGluRs) and metabotropic receptors (mGluRs). IGluRs regulate synaptic neurotransmission through the gating of ions through an ion pore whereas mGluRs regulate slower transmission through coupling to G proteins (Schoepp, 1994; Nicoletti et al., 1996; Conn and Pin, 1997). Three types of ionotropic glutamate receptors have been identified that are differentiated by structure and pharmacology: α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), N-methyl-D-aspartate (NMDA), and kainate receptors (Collingridge and Lester, 1989). mGluRs are likewise classified based upon their distinct family structures and the compounds which regulate their function. mGluRs are classified into three families: type I receptors include mGluR1 and mGluR5 that stimulate the phosphoinositide biochemical pathway; type II receptors include the mGluR2 and mGluR3 receptors; and type III receptors include GluR4, mGluR6, mGluR7, and mGluR8 receptors. Activation of the type II and type III mGluRs inhibits adenylyl cyclase (Schoepp, 1994; Nicoletti et al., 1996; Conn and Pin, 1997).

The interplay of glutamatergic excitatory and GABAergic inhibitory neuronal processes has dominant oversight of neurological and psychological function. For example, in 2019, (S)-ketamine (Spravato) was approved for use as an adjunct treatment for treatment-refractory depression. Ketamine is the first compound in the new class of drugs known as rapid-acting antidepressants (Witkin et al., 2019b). In contrast to the selective serotonin reuptake inhibitor antidepressants that generally take weeks of daily dosing to produce a response (Katz et al., 1996, 2004), ketamine can transduce immediate relief from symptoms of major depressive disorder and can do so in patients that have been otherwise refractory to standard of care antidepressants (Zarate et al., 2006). In regions such as the hippocampus, glutamate release from pyramidal cells is kept in check through the activity of fast-spiking GABA inhibitory neurons. Ketamine, like its congener, phencyclidine (Homayoun and Moghaddam, 2007), acts by blocking the NMDA ion-channel and reducing the GABA-driven inhibitory tone on pyramidal cells, thus enabling the efflux of glutamate (Thelen et al., 2019). Free glutamate is then available to further the cascade of rapid-acting antidepressant response by binding to AMPA receptors (Alt et al., 2006) (Fig. 2). However, blockade of NMDA receptors by ketamine (Anis et al., 1983) is also an initiator of the characteristic dissociative and psychomimetic actions of ketamine (Nicholson and Balster, 2003). Understanding the inhibitory and excitatory balances involved in therapeutic actions and side-effects of compounds enables clues to creating rapid-acting antidepressants devoid of ketamine-like side effects (Witkin, 2020). In a similar fashion, such an understanding provides new directions for other therapeutic areas as well.

Figure 2.

Figure 2.

An example of GABA – glutamate dynamics. A simplified postulated mechanism of action of the rapid-acting antidepressant ketamine. Glutamate binding to AMPA receptors on postsynaptic neurons results in AMPA receptor amplification, initiating a biological cascade in a host of intermediary biology pathways that are associated with the antidepressant responses to ketamine. eEF2: eukaryotic elongation factor 2; ERK: extracellular signal–regulated kinase; mTOR: mammalian target of rapamycin. From Witkin et al. (2019b) with permission of the publisher.

II. GABAA Receptors

GABAARs have been mechanistically linked to a host of pathophysiological processes including sleep, pain, epilepsy, depression, and schizophrenia. GABAARs are a complex comprised of five subunits, which are expressed in humans as the following types: α1–6, β1–3, γ1–3, ρ1–3, δ, ε, π, and θ (Hevers and Lüddens, 1998; Olsen and Sieghart, 2009). Each functional GABAAR typically includes α-, β-, and γ or δ -subunits in a 2:2:1 ratio. The pentameric chloride ion channel is composed of a large extracellular domain containing an orthosteric binding pocket, a transmembrane domain composed of the pore-lining M2 helices and a small intracellular domain which is a site for modulation by associated proteins (Chen and Olsen, 2007; Han et al., 2020). The orthosteric binding pockets occupy the interfaces of the α and β subunits, typically two per pentamer (Phulera et al., 2018; Zhu et al., 2018; Laverty et al., 2019). The two binding pockets differ structurally (Masiulis et al., 2019), have different affinities for ligands, and act synergistically (Baumann et al., 2003). Binding of GABA, the endogenous agonist, induces local structural changes in the binding pocket, which result in a concerted rotation of the five extracellular domains (Masiulis et al., 2019). Rotation is translated into a conformational change of the transmembrane domain that affects channel activation and inactivation gates (Hevers and Lüddens, 1998; Masiulis et al., 2019).

Other agonists and antagonists can bind to the same orthosteric site but can produce different changes in both the binding pocket and in channel conformation (Masiulis et al., 2019). GABAARs also contain numerous binding sites for modulators including anesthetics, barbiturates, neurosteroids (Olsen, 2018; Solomon et al., 2019; Vega Alanis et al., 2020), and benzodiazepines. Like the orthosteric site, the benzodiazepine receptor is located in the extracellular domain, but on the interface of α and γ subunits (Laverty et al., 2019; Masiulis et al., 2019). Binding of benzodiazepine ligands does not produce marked changes in the conformation of the extracellular domain but rather facilitates the actions of orthosteric ligands. Different benzodiazepine receptor ligands have substantially different binding modes which likely impact ligand efficacy (Scott and Aricescu, 2019). For example, while full agonists of benzodiazepine receptors (diazepam, alprazolam) (Fig. 3) bind deep in the binding pocket (Masiulis et al., 2019) bretazenil, with partial agonism, and flumazenil with no activity, bind higher in the pocket (Miller et al., 2018; Scott and Aricescu, 2019).

Figure 3.

Figure 3.

Structures of first- and second-generation GABAkines used in the treatment of anxiety along with the currently widely prescribed drug alprazolam. Not shown here are barbiturates more prominently used as anxiolytics prior to the commercialization of the structures shown.

The complexity of the subunit composition of GABAARs in the CNS, together with distinct regional, cellular, and subcellular distributions, and diverse pharmacologic properties carries functional significance (Engin et al., 2018; Olsen and Sieghart, 2009; Siebert et al., 2018).

III. GABA and Therapeutics

Both positive and negative allosteric modulators of GABAARs exist (Olsen, 2018). The current review focuses on positive allosteric modulators (PAMs), or GABAkines. Consistent with the naming of AMPA receptor potentiators as AMPAkines, we are using the term GABAkines to denote positive allosteric modulators of GABAARs. The dominant role of GABA as an inhibitory neurotransmitter in the mammalian nervous system provides two a priori predictions regarding a role for GABA in therapeutics: 1) altering GABA neurotransmission should have pervasive actions on a host of neurological and psychiatric functions; and 2) alterations in GABA neurotransmission could be accompanied by GABA-related side effects. The data summarized below will illustrate both of these predictions.

The discovery, development, and clinical history of drugs that augment GABAergic neurotransmission is best illustrated by the work directed toward identifying non-sedating drugs to treat anxiety. This empirical adventure began long before a linkage drugs to GABA was uncovered. The first rationally-designed anxiolytic drug, meprobamate (Miltown) (Fig. 3) was discovered and championed by Frank Berger who, with Bernard Ludwig, modified the muscle relaxant mephenesin with the goal of reducing muscle-relaxing and sedative properties while augmenting anti-anxiety effects. Miltown was the first blockbuster drug and in the late 1950s was being used by so many people in the United States that its name was used in the popular language of the times (Tone, 2009). However, the popularity of Miltown led to its demise with people reporting dependence, a fate to later impact the medical prescriptions for diazepam (Valium) as well (Tone, 2009).

The carbamate, meprobamate, led to the next generation of anxiolytic drugs - the 1,4-substituted benzodiazepines. In search of a drug to compete with meprobamate, Hoffmann La-Roche synthesized many compounds without finding improvement over meprobamate and the project was terminated by management. Months later, these compounds were slated for destruction when a lab technician noted that Ro 5–0690 had not been tested (Winters, 2016). The head of medicinal chemistry, Leo Sternbach, directed animal testing (Randall et al., 1960) and took the compound himself, providing the first clinical data on chlordiazepoxide (Winters, 2016). With the FDA approval of chlordiazepoxide (Librium) (Fig. 3) in 1959 and its introduction into clinical practice, another generation of anxiolytic agents was born and, as with meprobamate, found widespread use for the treatment of anxiety.

Diazepam (Fig. 3) arose from the 1,4-substituted benzodiazepine chemical series and was approved for clinical use in 1965. By 1970, antianxiety drugs, mostly benzodiazepines, were used by 1 in 5 woman and 1 in 13 men in the United States (Parry et al., 1973). Diazepam was and still is a highly valuable drug used for the treatment of anxiety and other disorders (Woods et al., 1992; Szarmach et al., 2017; Weintraub, 2017; McTague et al., 2018; Jafarpour et al., 2019). Despite its bad press for being addictive (Hollister et al., 1963) it is still widely used. Diazepam is on the List of Essential Medicines of the World Health Organization. In the United States, diazepam as an anxiolytic has been largely supplanted by another benzodiazepine, alprazolam (Xanax) (Fig. 3) (Aden and Thein, 1980; Maletzky, 1980).

Benzodiazepines such as diazepam have some potential drawbacks, depending on their intended use. For example, when used as an anxiolytic, sedative effects at therapeutic doses can cause motor impairment. For sleep induction, however, sedation is desired. Memory impairment is another side-effect that is often unwanted. Like many CNS-acting drugs, benzodiazepines can produce tolerance to certain of their therapeutic effects, and induce physical dependence, and abuse (Woods et al., 1992). Tolerance to the anxiolytic effects of these drugs has been difficult to demonstrate (Margules and Stein, 1968; Laughren et al., 1982).

An overview of the pharmacology of meprobamate in comparison with the newer carbamates is available (Löscher et al., 2021). Machine-learning technologies were used to deduce that meprobamate would have antifungal properties (Udrescu et al., 2020). These findings were subsequently analyzed by molecular docking.

Combinations of clidinium with chlordiazepoxide were shown to be effective in patients with functional dyspepsia that were refractory to treatment by proton pump inhibitors (Puasripun et al., 2020). Librax® is a marketed formulation of this drug combination. The comparative clinical pharmacology of chlordiazepoxide and lorazepam in the treatment of alcohol withdrawal demonstrated that both were able to control delirium tremens, but it was suggested that chlordiazepoxide might be somewhat less efficacious in overall symptom control (March et al., 2019). Szarmach et al. (2017) provided an analysis of best practices for use of adjunctive benzodiazepine anxiolytics in combination with antipsychotic agents in the control of positive and negative symptoms in schizophrenic patients.

In preclinical studies using an inventive methodology, Leonard and Kangus (2020) evaluated the possible opioid-sparing (ability to reduce the amount of opioid needed to produce an anti-nociceptive effect) effects of diazepam. Diazepam had no antinociceptive actions of its own and had very limited ability to augment the effects of oxycodone. In addition, Pilipenko et al. (2019) reported on the potential favorable use of diazepam in a rat model of pre-dementia.

New delivery systems for diazepam have been studied including a nasal spray (Valtoco®), buccal, rectal gel (Diastat®) and intravenous forms (Cornett et al., 2021; Rogawski and Heller, 2019). Meta-analysis of randomized controlled trials indicated that i.v. lorazepam was better than i.v. diazepam for the cessation of status epilepticus in adults (Kobata et al., 2020).

A recent review of the pharmacology of alprazolam was published (George and Tripp, 2021). In addition to anxiety, prescribing practices for alprazolam have documented that it is also prescribed for dental procedures (Teoh et al., 2021) and major depressive disorder (Dold et al., 2020). Alprazolam also remains high on the list of medicines that are associated with withdrawal syndromes (Cosci and Chouinard, 2020).

IV. Direct-Acting Agonists

Inhibitory neurotransmission can be augmented through GABAAR ligands interacting with either orthosteric or allosteric domains on the GABAAR complex. Orthosteric agonists, like GABA itself, are also called direct-acting agonists. Muscimol and a structural analog, gaboxadol or THIP, are two common examples (Fig. 1). Gaboxadol is a potent agonist of GABA receptors that contain α4, α6, and δ, subunits, which have more restricted anatomic distribution in the thalamus, hippocampus, and cerebellum and are mainly extrasynaptic in location.

Attempts to capitalize on direct-acting GABAAR agonists for therapeutics in multiple areas of GABA insufficiency have generally failed due to a general lack of efficacy and GABA-associated side effects. A small trial with gaboxadol in patients with Huntington’s disease was conducted over a period of two weeks. Like muscimol, maximal doses of gaboxadol produced somnolence, unsteadiness of gait, and reduced attention to sensory stimulation. Despite these indicators of enhanced GABAergic impact, gaboxadol did not improve motor function or cognitive performances (Foster et al., 1983). Gaboxadol has also been considered as a medication to promote sleep (Wafford and Ebert, 2006). Ovid Therapeutics recently conducted a Phase-3 clinical trial of gaboxadol (OV-101) for treatment of Angelman Syndrome (Rakhit, 2020) where it failed to meet the primary endpoint and further development was suspended (Bryson, 2020). Heiss and colleagues (2019) have recently explored muscimol as a potential treatment for pharmacoresistant epilepsy but the data taken as a whole did not substantiate the efficacy of muscimol when administered intracerebrally.

V. GABAkines

A. History.

Although GABAkines have existed for years, it was not until the 1970s that GABA was implicated in their pharmacological actions. Traditional medicine utilized a host of hypnotic and sedative agents (e.g., ethanol, opioids, cannabinoids) with a transition to the use of paraldehyde, chloral hydrate, and bromides at the end of the 19th century. Barbiturates were initially synthesized by Adolf von Baeyer in 1864 and were introduced to the market as sedatives, hypnotics and antiepileptics in the early 20th century (López-Muñoz et al., 2005). Their use was rapidly expanded by the introduction of approximately 50 barbiturates, producing diverse clinical profiles but narrow therapeutic windows which created the potential for overdose, abuse, dependence and for exacerbation of seizures upon withdrawal (Isbell, 1950). The mechanism of action of barbiturates is not completely understood where modulation of multiple voltage-gated and ligand-gated ion channels has been proposed (Löscher and Rogawski, 2012). The main modulatory effect is through modulation of GABAARs with potentiation at low concentrations and direct activation at higher concentrations, pharmacological properties that decrease the safety of barbiturate GABAkines relative to the benzodiazepine GABAkines. Inter-subunit binding sites in the transmembrane domain of the human GABAAR were identified (Chiara et al., 2013) but precise binding domains and their structural impact upon binding remain to be fully characterized (Scott and Aricescu, 2019). The medical use of barbiturates declined in the 1950s after the introduction of meprobamate and the 1,4-benzodiazepines.

From the era of meprobamate and the benzodiazepines, there have been many new GABAkines discovered and some of these advanced into clinical development. The primary therapeutic areas of interest for these compounds were anxiety and epilepsy. However, other areas of interest were explored as well.

B. Anxiolytic Activity and Sedation.

One issue with the benzodiazepine anxiolytics that is key to understanding their therapeutic value as well as an aspect of their pharmacology that impedes therapeutic utility is dose-dependent sedation. While sedation is sometimes desired, as in sleep induction, it is a dose-limiting side-effect for the majority of therapeutic applications.

From the standpoint of both efficacy and side-effect liabilities, improvements in standard-of-care medicines for the symptomatic relief of anxiety are needed. Some antidepressant drugs are used for the treatment of anxiety (Bandelow, 2020). Although these compounds are generally not sedating, they bear other side effects such as treatment-mergent weight gain and sexual dysfunction (Atmaca, 2020; Gill et al., 2020). Moreover, their anxiolytic actions, like their effects on depression (Katz et al., 2004), require weeks of daily dosing (Cvjetkovic-Bosnjak et al., 2015). Due to such limitations, benzodiazepine anxiolytics continue to be used for the acute treatment of anxiety. Drugs like alprazolam are widely prescribed but come with the burden of sedation and motor-impairment, as well as abuse and dependence potential (Străulea and Chiriţă, 2009; Reissig et al., 2015; Duke et al., 2020). Moreover, drugs like alprazolam can facilitate the suppression of respiration that has led to emergency room visits and deaths (Jann et al., 2014; Hedegaard et al., 2018; Witkin et al., 2019a).

Rational drug discovery efforts directed at creating improved GABAkines came from basic pharmacological data along with the discovery of the benzodiazepine receptor (Möhler and Okada, 1977; Squires and Brastrup, 1977) and its role in potentiation of GABA currents by GABAkines (Choi et al., 1977) (see historical overviews by (Schallek et al., 1979; Tallman et al., 1980; Haefely, 1989)). This discovery enabled establishment of binding assays (Williamson et al., 1978), with promising ligands evaluated in animal models for efficacy and reduced unwanted side effects (reviewed in Skolnick, 2012).

Multiple compounds progressed into clinical trials due to their favorable preclinical profile including bretazenil, abecarnil, alpidem, and ocinaplon (Fig. 4). Alpidem was approved as an anxiolytic with relatively little sedation (Musch et al., 1988) but was later withdrawn due to a high occurrence of hepatitis (Baty et al., 1994). Ocinaplon demonstrated anxiolytic effects without sedation (Lippa et al., 2005; Czobor et al, 2010), but was later withdrawn from further development due to concern over incidents of liver enzyme elevation.

Figure 4.

Figure 4.

Structures of some compounds with anxio-selective profiles that entered into clinical investigation.

Initial pursuit of the ideal GABAkine had been directed toward the creation of a non-sedating anxiolytic (see Skolnick, 2012 for review). The potential for the discovery process to proceed on rational grounds was first given hope by Lippa and colleagues. Based upon the ability of some compounds to produce anxiolytic-like effects without sedation in animal models, it was hypothesized that multiple benzodiazepine receptors exist and that some mediate anxiolytic effects and others sedation (Klepner et al., 1979; Lippa et al., 1981; Lippa et al., 1982). The non-sedating anxiolytic-like compound, CL218,872 (Fig. 5) was the first to jump start this idea.

Figure 5.

Figure 5.

Structures of some early compounds developed for their reduced potentiation of GABAA receptors containing α1 protein subunits. The structure of NS11821 has not been publicly disclosed.

The advent of molecular biology enabled further refinement to the search for anxio-selective drugs. The concept of benzodiazepine type 1 and type 2 receptors (Lippa et al., 1978, 1981, 1982; Klepner et al., 1979) was integrated into the current understanding of the structure and function of the GABAAR. Basic and applied research in this area focused on the specific α-subunit comprising the GABAAR assembly since modifications of this subunit produced major changes in pharmacological activity. Genetic, pharmacological, and behavioral evidence was used to suggest that α1-subtype-containing GABAARs preferentially mediate the sedative, amnestic, ataxic effects, and dependence production of compounds (Rudolph et al., 1999; McKernan et al., 2000; Wafford, 2005; Licata et al., 2009; Ator et al., 2010; Tan et al., 2010), whereas α2- and α3-subtypes mediate anxiolytic effects (Löw et al., 2000; Rudolph and Möhler, 2014), anticonvulsant (Rivas et al., 2009), and pain therapeutics (Dias et al., 2005; Lewter, 2019). The α5-subtype has been implicated in memory function (Collinson et al., 2002; Dawson et al., 2006). Based primarily on the data associating α1-containing GABAARs receptors with sedation, discovery efforts were directed at the identification of GABAkines with preference for α2- and α3- relative to α1-containing GABAARs (Rudolph and Knoflach, 2011) (Fig. 5).

The α1 hypothesis has been challenged by both animal and human pharmacological data. For example, ocinaplon (Fig. 4), produced no sedation at the dose that produced significant anxiolytic effects, despite having higher efficacy at potentiating currents evoked in α1 GABAARs than α−2, −3 and −5-containing GABAARs (Lippa et al., 2005). And, as reviewed later, the α1-sparing, subtype-selective GABAkines showed more sedation than predicted from their pharmacological profiles in the preclinical studies. There continues to be a great deal unknown about the structural determinants of specific GABAARs that correspond to specific efficacy and side-effects (Sieghart and Savić, 2018; Skolnick, 2012). This uncertainty is exacerbated by the complexity of behavioral endpoints, specific GABAAR localization, and pharmacology (see also Maramai et al., 2020 for an overview).

One of the first such molecules investigated was L–838,417 (Fig. 5), a partial agonist at α2,3- and α5-containing GABAARs and a negative allosteric modulator at α1-containing receptors. L-838,417, produced anxiolytic-like effects in the elevated plus maze but did not impair motor activity (McKernan et al., 2000; Carling et al., 2005). Another three compounds progressed into clinical studies: two analogs of L–838,417 (TPA-023 and MRK-409) and a structurally unrelated compound, TPA-023B (Fig. 5) (Atack, 2011). All three compounds were partial agonists at α2/3 subtypes with no substantial efficacy at α1-containing GABAARs in vitro (Atack et al., 2006) and they were all efficacious in animal models of anxiety without observed sedation (Atack, 2009). Two other α1-sparing, subtype-selective GABAkines have also been brought forward: NS11821 from Neurosearch (structure not disclosed) and AZD7325 from Astra Zeneca (Fig. 5).

Despite preclinical hopes, clinical data on these compounds presented a more complex picture. All of these compounds produced more sedation, dizziness, drowsiness, and motor incoordination than was hoped (de Haas et al., 2007, 2008, 2012; Atack, 2009; Atack et al., 2010; Fujita et al., 1999). NS11821and AZD7325 also exhibited more dizziness, somnolence, and sedation in humans than hoped but were otherwise well-tolerated (Zuiker et al., 2016) (Chen et al., 2014; Jucaite et al., 2017). For example, in the study by (Zuiker et al., 2016), NS11821 is a partial GABAAR agonist with relatively dominant α2,3 and α5 subtype efficacy but negligible α1 agonism. The first-in-human study was performed in healthy male subjects using a single-dose, parallel, double blind, placebo-controlled, randomized, dose-escalation study design with six cohorts (N=48) enrolled. The eight subjects of each cohort received NS11821 (10 mg, 30 mg, 75 mg, 150 mg, 300 mg or 600 mg) or placebo in a 6:2 ratio. At low dose levels, NS11821 had relatively low exposure, probably due to poor solubility. Saccadic peak velocity decreased in a dose-related manner while limited impairments were seen on body sway and the visual analogue scale for alertness. The most common adverse events were somnolence and dizziness, which were more prominent with the higher doses. Although no positive control was used in this study, the results were compared post hoc with a dataset for lorazepam (2 mg). The maximum saccadic peak velocity effects seemed comparable to the typical effects of lorazepam, whereas the other central nervous system effects were smaller.

None of these compounds has yet progressed to become a medicine. There are many reasons for a drug not achieving the difficult milestone of becoming an approved therapeutic and the present review will not address this complex topic. For subtype selectivity discussions, the commentary by Sieghart and Savić (2018) and Skolnick (2012) is recommended.

Nonetheless, these compounds continue to be highly utilized research tools. For example, L838,417 was also utilized in modeling the pharmacophore for driving potency and efficacy of α1,2,3,5βγ2 GABAA receptor interactions where the α1-Gly2013-Glu225 appears to be pivotal (Söderhielm et al., 2018).

In an elegant series of experiments, Lorenzo et al. (2020) used L838,417 as a tool to help dissect the α2-GABAAR basis for regulation of neuropathic pain. The low abuse potential of this compound was shown by its lack of self-administration in Rhesus monkeys (Huskinson et al., 2019; Berro et al., 2021); triazolam and lorazepam were self-administered (Berro et al., 2021). As with chronic pain, itch continues to require improved therapies. TPA 023B was utilized as one of the tools to help define α2- and α3-containing GABAARs in the regulation of itch (Ralvenius et al., 2018). Zeilhofer and colleagues also reported that TPA-023B suppresses the affective component (tonic) of pain (Neumann et al., 2021). Taken together, these findings lead to increased mechanistic understanding of diseases and symptoms that are in search of better medicines.

Even though benzodiazepines, as a class, act at all γ subunit containing GABAARs (α1,2, 3, and 5), some compounds produce less sedation than others. One such compound is the 1,5 benzodiazepine clobazam (Wildin et al., 1990; Sankar, 2012), whose milder sedative liability could have contributed to its approval as an add-on therapy for Lennox-Gastaut syndrome (Ng et al., 2011). A small proof of concept clinical trial also reported reduction of capsaicin-induced hyperalgesia with clobazam (Besson et al., 2015). The activity of clobazam might be due, at least in part, to its active metabolite, N-desmethyl-clobazam. N-desmethyl-clobazam exhibits functional selectivity for α2/3/5-containing GABAARs (Ralvenius et al., 2016). N-desmethyl-clobazam produced significant analgesia in rodent models without sedation (Ralvenius et al., 2016). A patent for the clinical use of N-desmethyl-clobazam for chronic pain has been filed (Ralvenius et al., 2016). A Phase-1 clinical trial with N-desmethyl-clobazam showed no sedation, but there was also no significant efficacy in reversing hyperalgesia (Matthey et al., 2020). The compound has been registered for a Phase-2 clinical trial for treatment of peripheral neuropathic pain (Besson, 2020).

C. Epilepsy.

Many first-generation antiepileptic drugs have primary actions as GABAkines. These include valproic acid (Depakene), clorazepate dipotassium (Tranxene), clonazepam (Klonopin), diazepam (Valium), phenobarbital (Luminal), and primidone (Mysoline) (Gitto et al., 2010), some of which are still in use for the treatment of epilepsy (Gitto et al., 2010). For example, valproate is a GABAkine with use in generalized seizures and status-epilepticus (Rahman and Nguyen, 2020; Liampas et al., 2021). GABA-related side effects of dizziness and somnolence are induced and for which discontinuation of medication can occur (Rahman and Nguyen, 2020).

Despite the continued use of some older generation GABAkines, improvements are needed in both efficacy and safety (Witkin et al., 2021). It is estimated that in about 70% of patients, existing drugs are not fully efficacious in controlling seizures (Marson et al., 2007; Sinha and Siddiqui, 2011; Banerjee et al., 2014). Despite being maintained on multiple antiepileptic medicines, many patients continue to have seizures (Błaszczyk et al., 2018). For these reasons, some patients elect to have invasive therapeutic procedures such as surgical resection or disconnection (Adelson, 2001; Hwang and Kim, 2019). Pharmacoresistance to anticonvulsant therapy continues to be one of the key obstacles to the treatment of epilepsy (Franco et al., 2014). A recent review highlights the GABAkines that are being considered for focal epilepsy (Janković et al., 2021).

D. Other Therapeutic Indications.

Since GABA is a pervasive and primary neuro-inhibitor, GABA influences other biological functions in addition to anxiety and seizure activity. As such, GABAkines have been considered as potential therapeutic modulators for diverse disorders including schizophrenia, pain, depression, anxiety, cognition, and disorders of cardiovascular function. A recent review is highly informative (Maramai et al., 2020). This section will provide a very brief summary of the current focus of GABAARs that have α4, α5, or α6 subunit composition. The therapeutic potential of compounds acting at these receptors is less defined than those acting receptors containing α1-, α2, and α3 subunits.

Early work on GABAkines that specifically amplified GABA signaling through α5-containing GABAARs identified potential therapeutic value in the areas of schizophrenia and airway muscle relaxation (c.f., (Clayton et al., 2015)) (see reviews by Jacob, 2019; Mohamad and Has, 2019). Current work outlining a rationale for the potential therapeutic value of these selective GABAkines has recently been published in the diverse areas of schizophrenia, pain, depression, anxiety, cognition, and cardiovascular function (c.f., (Batinić et al., 2017; Donegan et al., 2019; Hernández-Reyes et al., 2019; Prevot et al., 2019; Bojić et al., 2021; Davenport et al., 2021; Fee et al., 2021; Franco-Enzástiga et al., 2021); however, see (Xue et al., 2017)).

α4-and α6-containing GABAARs are not sensitive to typical GABA modulators (diazepam-insensitive) and their focus is beyond the scope of this review. α4β3δ assemblies are considered extrasynaptic and as such have key relevance to some of the newer advancing neuroactive steroid GABAkines. α4-containing GABAARs also appear relevant for the control of breathing (Yocum et al., 2016).

The prospect of a therapeutic impact of α6-containing GABAARs has also not been neglected for multiple areas including pain and Tourette syndrome (c.f., (Huang et al., 1999; Chiou et al., 2018; Vasović et al., 2019; Tzeng et al., 2020; Cadeddu et al., 2021)).

VI. GABAkines in clinical development

The search for improved GABAkines has been directed toward achievement of efficacy at therapeutic levels that do not induce (unless specifically desired) sedation or sleep, but also that do not impair motor function or memory, and do not induce tolerance, dependence, or abuse. Another motive for the development of new GABAkines is the improvement of efficacy.

There are currently two classes of GABAkines in clinical development: several neuroactive steroids (Fig. 6) and two small molecules that have different degrees of selectivity for α2/3-containing GABAARs (Table 1).

Figure 6.

Figure 6.

Chemical structures of the neuroactive steroid GABAkines in clinical development including an intravenous formulation of the endogenous steroid allopregnanolone, brexanolone (Zulresso®), zuranolone, and ganaxolone. The structure of Sage-324 has not been disclosed. LYT-300 is an oral prodrug form of allopregnanolone whose structure has not been disclosed.

Table 1.

GABAkines in clinical development.

Compound Other Names Pharmacological Class Development Phase Current Clinical Use or Investigation Other Indications Investigated or Planned Company
Allopregnanol one Sage-547 Brexanolone Zulresso® Neuroactive steroid Marketed


Ph2

Ph1a/2b
Post-partem depression

FXTAS

Alzheimer’s disease
Refractory status epilepticus

Essential tremor

PTSD
Sage Therapeutics Inc
Sage-217 Zuranolone® Neuroactive steroid Ph2

Ph2
MDD

Parkinson’s disease
Bipolar disorder

Essential tremor

Insomnia model
Sage Therapeutics Inc
Sage-324 Neuroactive steroid Ph2 Essential tremor Sage Therapeutics Inc
Ganaxolone Ganaxolone® Neuroactive steroid Ph3

Ph2
CDLK5 epilepsy

PCDH19-related epilepsy

Refractory status epilepticus
Drug resistant partial onset seizures

Lennox-Gastaut Syndrome

Smoking cessation

PCDH19-related seizures

Post-partem depression

Fragile-X syndrome

PTSD

Infantile spasms

Treatment-resistant depression as adjunct
Marinus Pharmaceuticals
LYT-300 Neuroactive steroid Preclinical Not defined Pure Tech Health
PF-06372865 CVL-865 α2/3/5 GABAkine Ph2


Ph2

Ph2
Photosensiti ve epilepsy

Chronic back pain

GAD
Focal onset seizures

Panic disorder (CO2 model)
Cerevel Therapeutics
KRM-II-81 α2/3
GABAkine
IND enablement Not defined RespireRx Pharmaceuticals Inc

Other indications investigated were gleaned from the Clinical Trials Registry (https://clinicaltrials.gov/ct2/home).

FTXS: fragile-X associated tremor/ataxia disorder

GAD: generalized anxiety disorder

MDD: major depressive disorder

PTSD: post-traumatic stress disorder

A. Neuroactive steroids.

Three of the principal neuroactive steroids in clinical development are shown in Fig. 6. These GABAkines have potential for treating a host of neurological and psychiatric disorders (Gasior et al., 1999; Miziak et al., 2020), and are being developed primarily for epilepsy and depression. Indeed, a role for neuroactive steroids in epilepsy has been postulated since the publication of early preclinical data (Belelli et al., 2019; Gasior et al., 1999; Reddy and Rogawski, 2012; Miziak et al., 2020).

1. Allopregnanolone

a. Preclinical Pharmacology

Allopregnanolone is an endogenous GABAkine derived from progesterone. Preclinical studies have documented that allopregnanolone presents with the full phenotype of a GABA potentiator with anxiolytic-like, sedative-like, and anticonvulsant efficacy (Gasior et al., 1999; Reddy and Rogawski, 2010; Lévesque et al., 2017). Neuroactive steroids like allopregnanolone can prevent the development of seizure sensitization or kindling where diazepam is not effective (Gasior et al., 1998). Brexanolone is a special formulation of allopregnanolone approved for the treatment of post-partum depression, and is a mixture of allopregnanolone and sulfobutylether-β-cyclodextrin (a solubilizing agent) (Scott, 2019). In the following paragraphs, brexanolone refers to this specific formulation, while all other formulations are referred to as allopregnanolone.

Work on the potential adjunctive treatment of seizure disorders with neuroactive steroids and conventional anticonvulsants began twenty years ago (Gasior et al., 2000) and this work is ongoing (Chuang and Reddy, 2020; Dhir et al., 2020; Lumley et al., 2019).

Other formulations and delivery methods for allopregnanolone including intranasal (Zolkowska et al., 2021) and transdermal (Bhattaccharjee et al., 2020) are also being developed but as far as we are aware, these delivery systems have not yet been used in human studies.

b. Clinical Pharmacology.

The current clinical application of brexanolone is for post-partum depression (Zorumski et al., 2019). An intravenous formulation of allopregnanolone constitutes the approved form of the marketed drug Zulresso. The intravenous route bypasses the issue of allopregnanolone’s poor oral bioavailability. Clinical studies have documented the efficacy of brexanolone (Frieder et al., 2019; Gerbasi et al., 2020; Kanes et al., 2017; Meltzer-Brody et al., 2018), the results of which have been summarized and are under discussion (Zheng et al., 2019; Patatanian and Nguyen, 2020; Kleinman and Schatzberg, 2021; Payne, 2021; Shukla et al., 2021). Although direct head-to-head comparisons with other agents have not been reported, indirect analyses of the data suggest that brexanolone produced large changes in depression ratings as well as patient and clinician reports (Cooper et al., 2019). Recent post-hoc analyses of the three clinical trials for post-partem depression have indicated a rapid onset with intravenous treatment (about 60 h) and an antidepressant effect that endured to the end of the 30-day evaluation period (Gerbasi et al., 2020). Analyses of these data also demonstrated the value of brexanolone in improving health-related quality of life (Gerbasi et al., 2021).

Allopregnanolone has also been studied in patients with Fragile-X associated tremor/ataxia disorder. Wang et al. (2017) reported findings from six patients undergoing allopregnanolone infusions over 12 weeks. The patients showed improvements in executive functioning, episodic memory and learning; MRI data also suggested benefits in individual patients suggesting neuroprotective effects. Napoli et al. (2019) also reported beneficial effects of allopregnanolone treatment in patients with fragile X-associated tremor/ataxia syndrome when dosed for 12 weeks in an open-label study.

Another key area of clinical investigation has been in the area of status epilepticus and in particular super-refractory status epilepticus. Vaitkevicius et al. (2017) provided the first findings that allopregnanolone could successfully block seizures in two patients (120-h infusion). Another study confirmed the efficacy and tolerability of brexanolone infusion in 25 patients (Rosenthal et al., 2017).

The possible use of allopregnanolone in the treatment of Alzheimer’s disease has also been considered. A tolerability study of allopregnanolone (formulated in a similar manner as brexanolone) was reported in 24 early Alzheimer’s patients by Hernandez et al. (2020) where the safety and pharmacokinetic profiles suggested advancement into Phase-2 studies.

Brexanolone has also been in clinical investigation for other therapeutic indications (Table 1).

The safety and tolerability of brexanolone has been described now in multiple Phase-2 and 3 clinical reports (Powell et al., 2020). Dizziness and somnolence were the most prevalent events but generally did not exceed 20%. Loss of consciousness is one of the biggest concerns that must be monitored and medically managed. Euphoria, especially at higher concentrations has been reported. The dosing protocol for brexanolone takes into account the major tolerability issues by dose escalation and patient monitoring.

c. LYT-300.

An orally bioavailable prodrug form of allopregnanolone is being investigated. The compound avoids first-pass metabolism by the liver and was reported to achieve significant oral bioavailability of natural allopregnanolone in non-human primates. LYT-300 is targeted for a range of neurological and psychiatric disorders as indicated by the sponsoring company, Puretech Health (LYT-300 (puretechhealth.com).

2. Zuranolone

a. Preclinical Pharmacology.

The synthesis of Sage-217 (zuranolone) was reported (compound 3) by Martinez-Botella et al. (2017). The bioisosteric pyrazole substitution on the ester function of an allopregnanolone backbone was made with the potential of increasing oral bioavailability and selectivity for GABAARs over other protein targets was demonstrated.

Data from the preclinical characterization of zuranolone were disclosed (Martinez Botella et al., 2017; Althaus et al., 2020). In electrophysiological studies, zuranolone amplified GABAAR currents at nine unique human recombinant receptor subtypes; synaptic (γ subunit-containing) as well as extrasynaptic (δ subunit-containing) currents were potentiated. Diazepam enhanced current was also amplified. Cell surface GABAAR trafficking was suggested by data from electrophysiologic studies in brain slices where sustained increases in current were induced. Oral activity was demonstrated in pharmacokinetic and efficacy assay readouts. Intraperitoneal dosing produced greater plasma levels (Cmax) than oral dosing (bioavailability of 62%, p.o., compared to 89% with i.p. administration). Brain to plasma ratios were 1.4 to 1.6 for i.p., and p.o. dosing, respectively.

Tonic seizures induced by the GABAAR antagonist pentylenetetrazol (PTZ) were inhibited by zuranalone at a minimum effective dose of 1 mg/kg in mice. Oral administration augmented power in the β-frequency band of the EEG and induced sleep at the higher doses of 3 and 20 mg/kg. These data provided another indicator of on-target engagement with oral dosing and a potential translatable biomarker for further compound development (Althaus et al., 2020).

Studies with another neuroactive steroid, SGE-516, have shown activity in animal models that detect compounds with anticonvulsant and mood-enhancing efficacy (Althaus et al., 2017; Hammond et al., 2017; Hawkins et al., 2017; Melón et al., 2018). The potential for neuroactive steroids to differentiate from other GABAkines is intriguing. This was first demonstrated in vivo in a seizure kindling model where ganaxolone but not diazepam prevented the development of kindling (Gasior et al., 1998) (see also similar data with KRM-II-81 (Knutson et al., 2020). In a model of Dravet Syndrome (Hawkins et al., 2017) and a model of post-partum depression (Melón et al., 2018), SGE-516 showed overlapping as well as distinct efficacy compared to clobazam. For example, in the Scn1a +/− mice that mirror some symptoms of Dravet syndrome, SGE-516 increased long-term survival whereas clobazam was reported to be inactive against this biological endpoint (Hawkins et al., 2017).

b. Clinical Pharmacology

Single doses of zuranolone over 7 days were explored in healthy human volunteers as an oral solution (Hoffmann et al., 2020). Pharmacokinetics and adverse events were monitored (see below) where the data encouraged advancement of the compound into a number of Phase-2 studies as well as pivotal studies in major depressive disorder and post-partum depression. Zuranolone was orally bioavailable and dose-proportional, with a terminal-phase half-life of 16–23 h and a tmax of about 1 h in humans.

Zuranolone was studied for potential efficacy in major depressive disorder (MDD) (Frieder et al., 2019). In a study by Gunduz-Bruce (2019), zuranolone was administered to MDD patients in a double-blind, placebo-controlled manner with a single oral dose of 30 mg. Fourteen days of daily treatment resulted in a statistically significant reduction in depression rating scores at day 15 compared to baseline. The reduction in symptoms across several measurement instruments was not associated with improvements in insomnia or anxiety (Gunduz-Bruce et al., 2019). Analysis showed that there was a favorable benefit/risk ratio in the administration of zuranolone in MDD patients (Arnaud et al., 2021). Unfortunately, the efficacy observed in the study by Gunduz-Bruce was not systematically replicated (Pagliarulo, 2019).

Data from an open-label study of zuranolone (20–30 mg) was reported in patients with Parkinson’s disease where patients were given zuranolone for 7 days while they were concurrently taking their dopaminergic medications. Zuranolone was safe and significantly reduced tremor (Bullock et al., 2021).

In the dose-ranging study by Hoffmann et al. (2020), severe adverse events were observed after single oral dose at the level of 66 mg – a change in mental status in two people, two people were unresponsive to external stimuli, and one showed marked somnolence. No loss of consciousness was observed. Sedation was the prominent side-effect. In the study with MDD patients, the most common adverse events in the drug group (45 patients) were headache, dizziness, nausea, and somnolence.

3. Sage-324

a. Preclinical Pharmacology.

Sage-324 is reported to be a neuroactive steroid GABAkine with oral bioavailability and a delta-preferring GABAkine in contrast to the balanced pharmacological profile of Sage-217 (Rosenthal and Metzer-Brody, 2016). The comparative in vitro pharmacology of these two compounds was reported as – Sage-324: α1β2γ2 (EC50: 3000 nM); α4β3δ (EC50: 273.4 nM) and Sage-217 α1β2γ2 (EC50: 296.1 nM); α4β3δ(EC50: 274.6 nM). These electrophysiological data show Sage-324 to be a α4β3δ-preferring GABAkine (ratio = 10.97) whereas Sage-217 is equipotent (ratio = 1.08). Brexanolone (Sage-547) was reported at the same time to have potencies of α1β2γ2 (EC50: 184.6 nM); α4β3δ(EC50: 80 nM) with a α4β3δ-preferring ratio of 2.31.

b. Clinical Pharmacology

Results of a study with Sage-324 in patients with essential tremor were disclosed in April 2021 (Ulrich, 2021). Sage-324 was well tolerated and efficacious in reducing essential tremor (36% reduction vs 21% with placebo at day 29 of treatment). The data also suggest the possible utility of this compound in other Phase-2 exploratory studies. Higher doses were explored and dose reduction was necessary in 62% of patients and discontinuations occurred in 38% of patients. The side-effects reported were about twice as high as in the placebo group - somnolence 68%; dizziness 38%; balance disorder 15%; diplopia 12%; dysarthria 12%; and gait impairments 12%.

4. Ganaxolone

a. Preclinical Pharmacology

Synergistic studies with tiagabine or midazolam in rodent seizure models have recently been reported (Chuang and Reddy, 2020). Using two mouse models, Reddy et al. (2019) found better efficacy of ganaxolone in females that they suggested is related to a greater abundance of extrasynaptic GABAARs. A role for extrasynaptic GABAARs in the anticonvulsant actions of ganaxolone were further implicated with zinc blocking studies (Chuang and Reddy, 2019). Genotype differences in the efficacy of ganaxolone have also been reported in a mouse model of ethanol withdrawal-induced seizures (Nipper et al., 2019).

A host of newer preclinical studies are pointing the way for other potential therapeutic indications. Ganaxolone is under consideration for its potential beneficial efficacy in cases of nerve-agent-induced seizures (Reddy, 2019) and as a possible intervention in Angelman syndrome where epilepsy is one of the developmental clinical features. For example, Ciarlone et al. (2017) explored the effects of ganaxolone in the Ube3a-deficient mouse model of Angleman syndrome where anxiolytic, anticonvulsant and motoric improving efficacy were observed. Ganaxolone exhibited prolonged plasma exposures and prolonged efficacy compared to allopregnanolone against status epilepticus in rats (Saporito et al., 2019) with both compounds showing profound sedative effects at higher doses.

Ganaxolone has also been explored in preclinical studies evaluating several neuropsychiatric disorders. In combination with clonazepam, ganaxolone was suggested to be supra-additive in augmenting anti-anxiety-like effects in rats (Gunter et al., 2016). Ganaxolone has shown preclinical efficacy in ameliorating behavioral deficits induced by social isolation that in some cases are greater than that of fluoxetine (Locci et al., 2017). In a mouse model of autistic symptoms, ganaxolone showed enhancements in social behaviors (Kazdoba et al., 2016).

A role for ganaxolone in a host of psychiatric diseases has also gained recent preclinical scrutiny. Kazdoba et al. (2016) studied effects of ganaxolone in BTRB mice that model some aspects of autism spectrum disorder. They reported that ganaxolone improved some features of social approach and reciprocal social interactions in BTBR mice but did not significantly reduce repetitive self-grooming.

The abuse potential of ganaxolone was studied in primates where there were small differences in the drug-taking behavior of non-human primates compared to traditional benzodiazepine anxiolytics (Meng and Rowlett, 2016).

b. Clinical Pharmacology

Studies have demonstrated the efficacy of ganaxolone in multiple epilepsy patient populations (Pieribone et al., 2007; Sperling et al., 2017; Yawno et al., 2017; Bialer et al., 2018). Although a Phase-3 study failed to meet its primary endpoint, analysis of a patient subgroup showed superiority over placebo (Bialer et al., 2018). Adverse events with ganaxolone have been mild and reversible and include somnolence, fatigue, dizziness, and headache (Bialer et al., 2018). Phase-2 and Phase-3 studies are ongoing and planned where ganaxolone is being developed for post-partum depression, pharmacoresistant status epilepticus, and several rare, treatment-resistant genetic epilepsies (e.g. CDLK5 deficiency disorder). Ganaxolone has been targeted as a potential treatment for pharmacoresistant status epilepticus (Zolkowska et al., 2018). Marinus Pharmaceuticals recently reported that ganaxolone successfully treated refractory status epilepticus in a Phase-2 trial (Meglio, 2019) and has progressed to Phase-3 investigation (Meglio, 2021). Recent reviews of the antiepileptic drug studies with ganaxolone are available (Miziak et al., 2020; Lattanzi et al., 2021), as well as its effects for childhood epilepsy (Perry, 2020), neonatal epilepsy (Yawno et al., 2017), and neuroprotective properties (Thomas and Pang, 2020). Ganaxolone given at 1500 mg/day was reported to be an effective adjunct agent in the treatment of patients with partial-onset seizures (Sperling et al., 2017).

Studies are also ongoing with ganaxolone in the area of orphan diseases. A negative trial was reported by Ligsay et al. (2017) in Fragile-X syndrome, although subgroup analysis suggested some potential benefits. Some recent reviews have been published for Lennox-Gastaut Syndrome (Strzelczyk and Schubert-Bast, 2021). A Phase-3 study is ongoing to evaluate ganaxolone in PCDH19-related epilepsy (Samanta, 2020).

A review of treatments for post-partum depression placed ganaxolone on the list (Frieder et al., 2019). An open-label study with ganaxolone as an add-on for depression in post-menopausal women that did not respond to ongoing conventional antidepressants was conducted with b.i.d. doses increasing over 8 weeks (Dichtel et al., 2020). Antidepressant efficacy was observed but with somnolence, fatigue, and dizziness. There were no significant effects on quality of life or sexual function.

A commentary on the potential use of ganaxolone in patients with PTSD was published (Kawada, 2018) after the reported failed trial (Rasmusson et al., 2017).

B. PF‐06372865 (darigabat, formerly CVL-865)

1. Preclinical Pharmacology

The functional selectivity of PF-06372865 as determined in electrophysiological experiments on recombinant cell lines demonstrated it to be selective for α2/3/5 relative to α1-associated GABARs (Table 2) (Nickolls et al., 2018; Owen et al., 2019).

Table 2.

Potency and efficacy of KRM-II-81 and the structural analog, MP-III-080, compared to diazepam and PF-06372865 (CVL-865)1.

GABAA-α1 GABAA-α2 GABAA-α3 GABAA-α4 GABAA-α5 GABAA-α6
Compounds Potency (nM) Efficacy (%) Potency (nM) Efficacy (%) Potency (nM) Efficacy (%) Potency (nM) Efficacy (%) Potency (nM) Efficacy (%) Potency (nM) Efficacy (%) References
KRM-II-81 (9)2 1730.0 115.6 101.9 252.24 60.9 262.22 >30000 97.425 192.6 114.1 >30000 98.775 Lewter et al., 2017
KRM-III-80 (7)2 241.3 115.0 102.1 178.0 102.0 208.6 ND ND 61.3 173.6 ND ND Witkin et al., 2017
Diazepam 18.1 256.24 17.6 286.54 19.6 272.92 >30000 100.28 11.1 211.18 >30000 100.42 Poe et al., 2016, Witkin et al., 2017
PF-063728653 0.2 121.0 2.9 234.0 1.1 192.0 >19900 ND 18.0 191.0 >19900 ND Owen et al., 2019
1

Data are from different sources and sometimes from different methods as described in the references

2

Compound number in parentheses are compound designations in Poe et al. (2016) and in Witkin et al. (2017)

3

Potency data are affinity values

ND: no data exist for these diazepam insensitive GABAA receptor configurations

In rodent models, PF‐06372865 blocked pentylenetetrazol and amygdala kindled seizures (Buhl et al., 2017) and dampened seizure activity in the GAERS rat, a genetic model of absence seizures (Duveau et al., 2018). PF-06372865 also produced anti-hyperalgesia in rodent models of neuropathic pain (Owen et al., 2019).

2. Clinical Pharmacology

PF‐06372865 was studied in healthy human volunteers and in patients where it was well-tolerated (Gurrell et al., 2018; Nickolls et al., 2018; Simen et al., 2019). PF‐06372865 suppressed electrical activity in patients with photosensitive epilepsy (Gurrell et al., 2019) and has also demonstrated efficacy against multiple pain modalities in a Phase-1 clinical investigation (van Amerongen et al., 2019). When tested in a larger Phase-2 trial for lower back pain, it did not achieve its primary efficacy end point of reduction in pain intensity and produced benzodiazepine-like side effects including sedation and memory impairment (Gurrell et al., 2018). In a Phase-2 trial for anxiety, PF-06372865 failed for lack of efficacy and for induction of side effects (Simen et al., 2019). Lack of demonstrated efficacy might have been due to the relatively low estimated receptor occupancy (~50%) achieved with the maximal dose of 7.5 mg (Nickolls et al., 2018); however, the occurrence of somnolence, dizziness and memory impairment at this dose could preclude higher dose testing. Further development of PF-06372865 has been undertaken by Cerevel Therapeutics. The compound was renamed to darigabat (formerly CVL-865) and is in Phase-1 development as an anxiolytic (Zuiker et al., 2020) and Phase-2 as an antiepileptic (Dandurand, 2021).

In a small study of photosensitive epileptic patients, no severe side effects were reported with doses up to 52.5 mg, however sedation and dizziness observed in half of the photosensitive epilepsy patients (Gurrell et al., 2019). Larger clinical trials for anxiety and chronic pain however reported adverse events including dizziness, headache, somnolence, and cognitive impairment already at 7.5 mg dose (Gurrell et al., 2018; Simen et al., 2019). Another Phase-1 study that used twice daily dosing for 14 days reported no major side effects with the most prevalent minor side effect being dizziness, which was in most cases of transient nature (Eides, 2019). A 42.5 mg dose of PF-06372865 was able to achieve >80% receptor occupancy without significant somnolence (Eides, 2019), whereas benzodiazepines achieve 10% to 15% receptor occupancy with significant somnolence (Fujita et al., 1999).

PF-06372865 has also been studied in the clinic as a potential pain therapeutic. A pharmacodynamic biomarker study was reported by Nickolls et al. (2018). PF-06372865 suppressed evoked pain (electrical, pressure, heat, cold, inflammatory) in healthy human volunteers (van Amerongen et al., 2019). However, PF-06372865 failed to demonstrate efficacy in a Phase-2 trial for chronic low back pain where no sedation was also reported (Gurrell et al., 2018).

C. KRM-II-81

KRM-II-81 was first disclosed as a non-sedating anxiolytic-like compound in 2016 from the laboratory of James M. Cook (Poe et al., 2016) (Fig. 7). KRM-II-81 is an imidazodiazepine that preferentially activates α2/3-containing GABAARs (Table 2). Preclinical data support the proposition that KRM-II-81 demonstrates reduced sedation, motor-impairing effects, tolerance development, and abuse liability compared to 1,4-benzodiazepine GABAkines. KRM-II-81 has also shown efficacy in animal models that are used to detect compounds that are anxiolytic, antidepressant, as well as those used in chronic and neuropathic pain states. KRM-II-81 has most extensively been studied as an antiepileptic where it has shown greater potency and efficacy than diazepam in several animal models and greater efficacy in animal models of pharmaco-resistant epilepsy. In addition, KRM-II-81 has demonstrated in situ activity in human translational studies using brain tissue from treatment-resistant epileptic patients. No clinical data have been reported with KRM-II-81. It is currently under development by EndeavorRx, a business unit of RespireRx Pharmaceuticals Inc.

Figure 7.

Figure 7.

Structures of the imidazodiazepine KRM-II-81 and structural analogs are shown. Also pictured is the structurally-distinct compound PF-06372865. Both KRM-II-81 (α2/3-selective) and PF-06372865 (α2/3/5-selective), also known as CVL-865, are currently in development.

KRM-II-81 was first synthesized from HZ-166 (Poe et al., 2016). HZ-166 (Fig. 7) was identified as an α2/3-preferring GABAkine that displayed reduced propensity for sedation and motor impairment compared to diazepam (Cook et al., 2009), and was active in models that detect anxiolytic drugs (Fischer et al., 2010), anticonvulsant compounds (Rivas et al., 2009), and antinociceptive compounds (Di Lio et al., 2011). HZ-166 contains a metabolically-labile ester function that reduces its overall bioavailability (Poe et al., 2016); KRM-II-81 was rationally designed to increase oral bioavailability and retain selectivity for α2/3-containing GABAARs by replacing the ethyl ester with an oxazole bioisostere. KRM-II-81 displayed good bioavailability after i.p. and oral administration in rats with exposure of plasma and brain (Poe et al., 2016). Both oral and intraperitoneal dosing in rats produced detectable unbound plasma levels of KRM-II-81 from 0.25–12h post dosing (Witkin et al., 2019a).

Electrophysiological experiments have established that KRM-II-81 potentiates α2- and α3-containing GABAARs with little amplification of α1-containing GABAARs (Table 2). In contrast, diazepam also amplifies responses of GABA in recombinant cells containing α1- and α5-containing GABAARs (Table 2). Compared to PF-06372865 (α2/3/5-selective), KRM-II-81 is 9000 times less potent at α1 and 11 times less potent at α5. Thus, in addition to low efficacy at α1- and α 5-containing GABAARs, KRM-II-81 requires higher concentrations to potentiate these ‘off-target’ GABAARs. When studied for its activity against a host of other receptor proteins, KRM-II-81 did not display any off-target liabilities (Poe et al., 2016).

Preclinical data have been reported in a broad range of seizure models using rodents and human epileptic brain tissue. KRM-II-81 displayed wide-ranging anticonvulsant activity against acute chemoconvulsant challenges (Witkin et al., 2018; Knutson et al., 2020) and electrically-induced seizures (Witkin et al., 2018; Knutson et al., 2020). KRM-II-81 also blocked seizure sensitization (kindling) (Witkin et al., 2018; Knutson et al., 2020). Importantly, like the neuroactive steroids, but unlike diazepam, KRM-II-81 prevented the development of the seizure-kindling process (Knutson et al., 2020). That α2-containing GABAARs are sufficient for anticonvulsant activity is evidenced by the finding that blockade of α1-containing GABAARs by β-CCT did not nullify the anticonvulsant effects of diazepam at doses that were sufficient to block its motor-impairing effects (Witkin et al., 2018).

Traumatic brain injury engenders long-term negative health outcomes including posttraumatic epilepsy that is associated with affective, neurocognitive, and psychosocial disruption of life (Semple et al., 2019) and for which there are no effective treatments (Temkin, 2009; Wat et al., 2019). Effects of KRM-II-81 on inhibiting cortical network activity was studied using an in vivo two-photon imaging technique in which neuronal activity in cortical layer II/III pyramidal neurons was assessed in mice with traumatic brain injury. Dramatic increases in fluorescence, measuring the intensity of spiking activity of individual neurons and fraction of active neurons, was observed 3 months after brain injury. Both the mean integrated fluorescence and fraction of active neurons were markedly decreased after KRM-II-81 (Witkin et al., 2020). The possibility that KRM-II-81 might also positively reduce the probability of post-traumatic epilepsy is raised by these findings.

In animal models of pharmaco-resistant epilepsy, KRM-II-81 is also active and, in many cases, better than standard of care antiepileptics (Witkin et al., 2020). The mechanisms for pharmaco-resistance are likely multifaceted; nonetheless, tolerance development has been suggested to play a major role (Löscher and Schmidt, 2006). KRM-II-81 did not produce tolerance, over five days of dosing, to the suppression of convulsions in mice induced by pentylenetetrazol (Witkin et al., 2018). In a kainate-induced chronic epilepsy model, KRM-II-81 given daily for five days did not exhibit decreases in anticonvulsant efficacy (Witkin et al., 2020). Similar findings were reported by Rivas et al. (2009) with HZ-166. Longer term dosing studies are required to determine whether KRM-II-81 has a reduced propensity to produce tolerance as suggested by the data in pain models (described below) and as predicted from point mutation studies implicating α2-containing GABAARs as a target for the relief of tolerance development (Ralvenius et al., 2015).

To further establish translatability of the anticonvulsant effects of KRM-II-81 to patients, microelectrode recordings were made from freshly transected cortical tissue slices from the brains of juvenile epileptic patients that were refractory to antiepileptic medications. KRM-II-81 decreased cortical firing rates across the neural network (Witkin et al., 2018).

Pain pathways rely heavily on GABAAR-driven inhibitory neurotransmission (Hammond and Drower, 1984; Dirig and Yaksh, 1995; Enna and McCarson, 2006; Zeilhofer et al., 2015; Etlin et al., 2016). A nice illustration was presented over 25 years ago where inflammatory pain induced by formalin was reduced by intrathecal injection of the direct-acting agonist muscimol. That these effects of muscimol were due to stimulation of GABAARs was confirmed through the use of the GABAAR antagonist bicuculline (Dirig and Yaksh, 1995). More recent studies have shown that intrathecal muscimol can also reduce neuropathic pain endangered by spinal cord injury in rats (Hosseini et al., 2014).

Although GABA is known to be an integral biological mediator of pain, GABAkines are generally not used to control pain (Chou et al., 2017) although prescriptions for pain patients occurs (Wright, 2020). The sedative properties of benzodiazepines have been proposed to interfere with the attainment of the doses needed to produce analgesic benefit (McKernan et al., 2000; Knabl et al., 2009; Munro et al., 2009; Atack, 2010; Ralvenius et al., 2015; Rudolph et al., 1999). A potential role for α1-containing GABAARs in the mitigation of the antinociceptive effects of diazepam has been demonstrated in point mutation studies (Knabl et al., 2008, 2009; Ralvenius et al., 2015; Paul et al., 2014; Tudeau et al., 2020). That α2-containing GABAARs are responsible for this emergent anti-nociceptive activity was provided by the finding that α2-selective GABAkines, HZ-166 (di Lio et al., 2017), L-838417 (Knabl et al., 2008; Nickolls et al., 2011; Hofmann et al., 2012; Lorenzo et al., 2020), NS11294 (Nunro et al., 2008, 2009), NS16085 (de Lucas et al., 2015), and TPA023B (Nickolls et al., 2011) are analgesic in rodent models.

KRM-II-81 and structural analogs have been reported to be active in animal models of acute and inflammatory pain (Fisher et al., 2017; Lewter et al., 2017; Moerke et al., 2019; Witkin et al., 2019) and chronic neuropathic pain induced by L5/6 nerve ligation (Witkin et al., 2019) or by the chemotherapeutic agent paclitaxil (Biggerstaff et al., 2020). The potential for this mechanism to serve to reduce opioid burden (the amount of opioid needed to control pain) was reported by Rahman et al. (2021). When dosed together with morphine, the KRM-II-81 analog, MP-III-024, engendered synergistic pain reduction.

Tolerance to the antinociceptive effects of pain medications creates serious health risks both by placing the patient in undue pain and increased risk of medication dependence and lethality (Kalant et al., 1971; Huxtable et al., 2011). No tolerance was observed with KRM-II-81 against acute inflammatory pain after 11 days of dosing; midazolam showed tolerance development (Lewter, 2019). Similar findings were observed in a neuropathic pain preparation with chronic constrictive nerve injury (Lewter, 2019). Over 22 consecutive days of dosing, neither KRM-II-81 or MP-III-080 produced tolerance to their antinociceptive effect against chonic neuropathic pain in mice (Biggerstaff et al., 2020).

KRM-II-81 and analogs produced anxiolytic-like effects in rodents at doses that did not produce sedation or motor-impairment. This was reported in a marble-burying assay in mice (Poe et al., 2016; Knutson et al., 2020) and in a Vogel-conflict study in rats (Poe et al., 2016; Witkin et al., 2017). KRM-II-81 and two structural analogs also produced antidepressant-like effects in the mouse forced swim test although the efficacy was not as great as that of imipramine (Methuku et al., 2018).

On multiple measures of sedation and motor impairment, KRM-II-81 was less impacting than 1,4-benzodiazepines like diazepam (Poe et al., 2016; Lewter et al., 2017; Witkin et al., 2018). Given the reduced motoric impact of KRM-II-81, the structural basis of this reduced side-effect profile was interrogated with structural docking studies where alprazolam was shown to bind more strongly to the α1β3γ2L GABAAR compared to KRM-II-81 (Witkin et al., 2020). Respiratory depression is another potential medical liability of GABAkines when given with respiratory depressing drugs like opioids. In multiple measures of respiratory function, KRM-II-81 was either inactive or less potent than alprazolam (Witkin et al., 2019a). GABAkines can impair memory and cognitive function (Crowe and Stranks, 2018; Engin et al., 2018). Indeed, some benzodiazepines like midazolam are used as pre-anesthetic sedatives where they convey the useful side effect of surgical memory loss (Kim et al., 2015; Patel and Kurdi, 2015). In one study evaluating potential cognitive impact, midazolam reduced spontaneous alternation in a T-maze from 70 to 33% whereas KRM-II-81 did not significantly reduce this measure of cognition (61%) (Lewter, 2019).

1,4-Benzodiazepine anxiolytics can be abused (Griffiths and Wolf, 1990; Woods et al., 1992) and are legally scheduled as controlled substances for this reason. The U.S. FDA updated its boxed warning for this class of compounds in September 2020 over safety issues associated with their combined use with opioids (Hirschtritt et al., 2021). In two models of abuse liability, intracranial self-administration (Moerke et al., 2019) and drug discrimination (Lewter, 2019), diazepam or midazolam were detected but not KRM-II-81).

D. Early-Stage Compounds

Medicinal chemistry efforts to identify improved structures for GABAAR ligands is ongoing. This research enterprise has brought new compounds into use as tools to identify biological bases for disease and has led to increased understanding of the structural and molecular underpinnings of key drug-protein interactions. Importantly, these newer compounds stand as potential novel compounds from which the next breakthrough GABAkines will come. An excellent review of newly-patented compounds is available (Crocetti and Guerrini, 2020).

Reports have emerged on a new compound that is a selective GABAkine of α3-containing GABAARs. A study from Rowlett’s lab (Meng et al., 2020) showed that YT-III-31 (8-ethynyl-N-methyl-6-phenyl-4H-benzo[f]imidazo[1,5-a][1,4]diazepine-3-carboxamide) did not increase punished responding of Rhesus monkeys, a finding that is opposite to that of the anxiolytic-like effects found with traditional benzodiazepine anxiolytics and with the α2/3-GABAkine, KRM-II-81 outlined earlier in this review. Instead, YT-III-31 had unique sedative-like effects. These findings are consistent with the idea that α2 but not α3-containing GABAARs (for which YT-III-31 is preferring) are pivotal for driving anxiolytic-like effects. Another study from Rowlett’s lab showed that in Rhesus monkeys discriminating ethanol, only triazolam (non-selective), an α5-preferring GABAkine, and an α2/3/5-preferring GABAkine fully reproduced ethanol like discriminative stimulus effects. In monkeys self-administering ethanol orally, an α2/3-(HZ-166) and an α3-preferring (YT-III-31) GABAkine facilitated ethanol but not sucrose drinking (Berro et al., 2021). A study in rats from by the Savić lab showed that YT-III-31 was able to produce anxiolytic-like efficacy in the open field and elevated plus-maze assays at low, non-sedating doses (Batinić et al., 2018). They also did work to help identify the in vivo activity of this new compound. They used brain exposures to estimate in vivo potentiation of specific GABAAR configurations: YT-III-31 at doses that were anxiolytic-like in rats did not potentiate α1βγ2 GABAARs. Their estimation also revealed only a modest selectivity of YT-III-31 for α3γ2 over α2γ2 and α5γ2 sites.

Medicinal chemistry efforts have also brought forth novel GABAkines acting at GABABRs (Lobina et al., 2021). Novel compounds for the GABAAR have also been recently disclosed with both agonist and allosteric modulatory properties (Olander et al., 2018), α5-impacting GABAkines acting at extrasynaptic GABAARs (Etherington et al., 2017), a new chemical series of 1,2,3-triazolo-benzodiazepine anticonvulsant compounds (Shafie et al., 2020), new series of 11-dialkylaminomethyl-2,3,4,5-tetrahydrodiazepino[1,2-a]benzimidazoles with potent anxiolytic properties (Maltsev et al., 2021), new analogs of 4,6-diphenylpyrimidin-2-ol with in vivo activity (Khoramjouy et al., 2021), 5-(2-aryloxy-4-nitrophenyl)-4H-1,2,4-triazoles and 5-(2-aryloxy-3-pyridyl)-4H-1,2,4-triazoles, possessing C-3 thio or alkylthio substituents with potent anticonvulsant activity (Navidpour et al., 2021), a new series of anticonvulsants based upon the 3-{2-[1-acetyl-5-(substitutedphenyl)-4,5-dihydropyrazol-3-yl]hydrazinylidene}-1,3-dihydro-2H-indol-2-ones platform (Kerzare et al., 2021), and a novel series of 1,3-dihydro-2H-1,4-benzodiazepin-2-one azomethines and 1,3-dihydro-2H-1,4-benzodiazepin-2-one benzamides was disclosed for which the lead compound was more efficacious than diazepam under their assay conditions and with lead-like properties (Nilkanth et al., 2020).

The design and synthesis of three different series of 1,5-benzodiazepines substituted at the 2 and 4 position were reported with a lead compound having diazepam-like high potency and drug-like properties (Verma et al., 2020). A new series of 4-phenyl-6H-imidazo[1,5-a]thieno[3,2-f][1,4]diazepine-7-carboxylate esters were synthesized by Di Capua et al. (2020) and shown to have in vivo activity in a range of benzodiazepine-detecting assays systems; some compounds were active without notable ancillary side-effects. Pandey et al (2020) reported new analogs of HZ-166. These compounds used 2,4-disubstituted oxazoles and oxazolines as bioisosteric replacement of the ester function with the goal of improving oral bioavailability. Two new structural analogs, LKG-I-70 and KPP-III-5, were shown to be devoid of motor-impairing effects when given up to 100 mg/kg. MIDD0301 (Yocum et al., 2019) was reported by Forkuo et al. (2018) to selectively amplify α1–3,5β3γ2 GABAARs. The compound was orally active with a long half-life. Their data suggest that MIDD0301 represents a new candidate compound that relaxes airway smooth muscle, reduces lung inflammation and mitigates airway hyper-responsiveness in a mouse model of asthma.

Newer methods are also being introduced to help identify molecular substrates of compound binding to and functional activation of GABAARs as well as the identification of novel drug therapies. For example, in a recent study by Crocetti and colleagues (2021) 8-methoxypyrazolo[1,5-a]quinazolines were examined with proximity frequencies analyses (quantification of the frequencies that a compound intercepts two or more amino acids in the process of binding). Their work led to the elucidation of a combination of amino acids αVAL203- γTHR142 and αTYR 160- γTYR 58 that could predict GABAkine function. Sansolone et al. (2019) described new photochemical methods to interrogate individual GABAARs, a technology that should have important consequences for the design of improved GABAkines. Structural studies on a newer series of pyrazoloquinolinones gave some insights into the structural dynamics of the pharmacophore that might help guide new compound discovery (Iorio et al., 2020). A new potential drug target for diazepam-refractory status epilepticus was announced - proinflammatory cytokine high mobility group box-1 (HMGB1) (Zhao et al., 2020). Along with this, Burman et al. (2019) reported on other potential mechanisms of diazepam resistance. Novel compounds have been used to help identify new binding domains for neuroactive steroids (Yu et al., 2019; Jayakar et al., 2020). Another auxiliary protein associated with GABAARs was also recently disclosed; Shisa7 regulates GABAAR trafficking, function, and pharmacology relevant to the control of GABAkine activity (Han et al., 2020).

A recent review of the Translocator Protein 18 kDa (TSPO), long-postulated to be a binding site for benzodiazepine anxiolytics is now available (Barresi et al., 2021). Alpidem was used as a prototype to create novel structures interacting with the TSPO binding site (Gudasheva et al., 2020). The dipeptide, GD-102 (N-phenylpropionyl-l-tryptophanyl-l-leucine amide), was shown to produce potent anxiolytic-like effects in vivo that were blocked by a TSPO binding site antagonist.

Taken as a whole, it is clear that the search for improved GABAkine medications continue.

VI. Conclusions.

For well over a century, GABAkines have been widely used medicines with diazepam being on the list of Essential Medicines of the World Health Organization. Efforts to discover and develop improved compounds for therapeutic use has led to an array of pharmacological tools for research into the neurobiological substrates of disease and for disease therapeutics.

GABAkines can produce a host of undesirable side-effect and safety issues including sedation, memory-impairment, tolerance, dependence, and abuse. It remains to be seen whether the newer GABAkines reviewed here will, as hoped, display reduced liabilities.

Several new GABAkines are currently in development including neuroactive steroids, and an α2/3-preferring (KRM-II-81) and an α2/3/5-preferreing GABAkine (PF-06372865). The neuroactive steroids are in clinical development for depression and intractable epilepsy with clinical studies directed also at other indications. PF-06372865 (darigabat, formerly CRV-865) appears to be in development for epilepsy and anxiety. KRM-II-81, a non-benzodiazepine GABAkine, is currently in the late preclinical phase. KRM-II-81 has demonstrated efficacy in animal models of anxiety, depression, acute and chronic pain, epilepsy, and traumatic brain injury. Preclinical data also support the potential for reduced tolerance and abuse. The efficacy of KRM-II-81 in models of pharmacoresistant epilepsy, preventing the development of seizure sensitization, and in brain tissue of intractable epileptic patients bodes well for improved therapeutics. The data on these newer GABAkines highlight the possibility of developing improved medicines in areas of clinical need.

Acknowledgments

We would not have been able to provide as cogent a review of this topic had it not been for the diligence, time, and caring oversight of three expert reviewers and editors. Great thanks are also due to Maciej Gasior and Michael Rogawski for their help in understanding some of the developmental issues with the neuroactive steroids. Jun-Xu Li and Lakeisha Lewter helped in this review by allowing use of the data in Dr. Lewter’s doctoral dissertation and for their solid experimental support over many years. We are grateful to John and Nancy Peterson for their financial support of this research. We also thank The National Institutes of Health for support from [MH-096463] and [NS-076517] and The National Science Foundation, Division of Chemistry [CHE-1625735]. We also acknowledge UW-Milwaukee’s Shimadzu Laboratory for Advanced and Applied Analytical Chemistry and support from the Milwaukee Institute of Drug Discovery and the University of Wisconsin-Milwaukee Research Foundation.

Abbreviations

CNS

central nervous system

β-CCT

beta-carboline-3-carboxylate-t-butyl ester

CGS 9896

2-(4-Chlorophenyl)-1,2-dihydro-3h-pyrazolo[4,3-c]quinolin-3-one

DOV-51892

7-(2-chloropyridin-4-yl)pyrazolo-[1,5-a]-pyrimidin-3-yl](pyridin-2-yl)methanone

GABA

γ-aminobutyric acid

GABAAR

GABAA receptor

GABAkine

a positive allosteric modulator of GABAARs

iGlu

ionotropic glutamate

mGlu

metabotropic glutamate

NS11821

structure not publicly disclosed

PAM

positive allosteric modulator

THIP

4,5,6,7-tetrahydroisoxazolo(5,4-c)pyridin-3-ol (gaboxadol)

QH-II-66

7-Ethynyl-1-methyl-5-phenyl-1,3-dihydro-benzo[e][1,4]diazepin-2-one

XHe-II-053

ethyl 8-ethynyl-6-phenyl-4H-imidazo[1,5-a][1,4]benzodiazepine-3-carboxylate

Footnotes

1

This manuscript is dedicated to Dr. Michael A. Rogawski for his pioneering work on neuroactive steroids and other GABAkines and his continued devotion to bring improved medicines to patients in need.

Conflict of interest

James Cook and Michael Poe are named as inventors on patents describing KRM-II-81 and analogs, certain rights to which have been licensed to RespireRx Pharmaceuticals Inc. Rok Cerne, Michael Poe, James Cook, and Jeffrey Witkin are members of the research advisory group for RespireRx Pharmaceuticals Inc and Arnold Lippa serves as Executive Chairman and Chief Scientific Officer.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Adelson PD (2001) Temporal lobectomy in children with intractable seizures. Pediatr Neurosurg 34:268–277. [DOI] [PubMed] [Google Scholar]
  2. Aden GC, Thein SG (1980) Alprazolam compared to diazepam and placebo in the treatment of anxiety. J Clin Psychiatry 41:245–248. [PubMed] [Google Scholar]
  3. Afzalimoghaddam M, Khademi MF, Mirfazaelian H, Payandemehr P, Karimialavijeh E, Jalali A (2021) Comparing Diazepam Plus Fentanyl With Midazolam Plus Fentanyl in the Moderate Procedural Sedation of Anterior Shoulder Dislocations: A Randomized Clinical Trial. J Emerg Med 60:1–7. [DOI] [PubMed] [Google Scholar]
  4. Ahwazi HH, Abdijadid S (2021) Chlordiazepoxide. In: StatPearls. Treasure Island (FL): StatPearls Publishing. Available at: http://www.ncbi.nlm.nih.gov/books/NBK547659/ [Accessed April 20, 2021]. [Google Scholar]
  5. Alt A, Nisenbaum ES, Bleakman D, Witkin JM (2006) A role for AMPA receptors in mood disorders. Biochem Pharmacol 71:1273–1288. [DOI] [PubMed] [Google Scholar]
  6. Althaus AL, Ackley MA, Belfort GM, Gee SM, Dai J, Nguyen DP, Kazdoba TM, Modgil A, Davies PA, Moss SJ, Salituro FG, Hoffmann E, Hammond RS, Robichaud AJ, Quirk MC, Doherty JJ (2020) Preclinical characterization of zuranolone (SAGE-217), a selective neuroactive steroid GABAA receptor positive allosteric modulator. Neuropharmacology 181:108333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Althaus AL, McCarren HS, Alqazzaz A, Jackson C, McDonough JH, Smith CD, Hoffman E, Hammond RS, Robichaud AJ, Doherty JJ (2017) The synthetic neuroactive steroid SGE-516 reduces status epilepticus and neuronal cell death in a rat model of soman intoxication. Epilepsy Behav 68:22–30. [DOI] [PubMed] [Google Scholar]
  8. Amundarain MJ, Caffarena ER, Costabel MD (2021) How does α1Histidine102 affect the binding of modulators to α1β2γ2 GABAA receptors? molecular insights from in silico experiments. Phys Chem Chem Phys 23:3993–4006. [DOI] [PubMed] [Google Scholar]
  9. Andersson JA, Brekke M, Vallersnes OM (2020) Acute poisoning from substance abuse of benzodiazepines. Tidsskr Nor Laegeforen 140. [DOI] [PubMed] [Google Scholar]
  10. Anis NA, Berry SC, Burton NR, Lodge D (1983) The dissociative anaesthetics, ketamine and phencyclidine, selectively reduce excitation of central mammalian neurones by N-methyl-aspartate. Br J Pharmacol 79:565–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Arnaud A, Suthoff E, Stenson K, Werneburg B, Hodgkins P, Bonthapally V, Jonas J, Meyer K, O’Day K (2021) Number Needed to Treat and Number Needed to Harm analysis of the zuranolone phase 2 clinical trial results in major depressive disorder. J Affect Disord 285:112–119. [DOI] [PubMed] [Google Scholar]
  12. Artelsmair M, Gu C, Lewis RJ, Elmore CS (2018) Synthesis of C-14 labeled GABAA α2/α3 selective partial agonists and the investigation of late-occurring and long-circulating metabolites of GABAA receptor modulator AZD7325. J Labelled Comp Radiopharm 61:415–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Atack JR (2008) GABA(A) receptor subtype-selective efficacy: TPA023, an alpha2/alpha3 selective non-sedating anxiolytic and alpha5IA, an alpha5 selective cognition enhancer. CNS Neurosci Ther 14:25–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Atack JR (2009) Subtype-selective GABA(A) receptor modulation yields a novel pharmacological profile: the design and development of TPA023. Adv Pharmacol 57:137–185. [DOI] [PubMed] [Google Scholar]
  15. Atack JR et al. (2010) Benzodiazepine binding site occupancy by the novel GABAA receptor subtype-selective drug 7-(1,1-dimethylethyl)-6-(2-ethyl-2H-1,2,4-triazol-3-ylmethoxy)-3-(2-fluorophenyl)-1,2,4-triazolo[4,3-b]pyridazine (TPA023) in rats, primates, and humans. J Pharmacol Exp Ther 332:17–25. [DOI] [PubMed] [Google Scholar]
  16. Atack JR (2010) GABAA receptor alpha2/alpha3 subtype-selective modulators as potential nonsedating anxiolytics. Curr Top Behav Neurosci 2:331–360. [DOI] [PubMed] [Google Scholar]
  17. Atack JR (2011) GABAA receptor subtype-selective modulators. I. α2/α3-selective agonists as non-sedating anxiolytics. Curr Top Med Chem 11:1176–1202. [DOI] [PubMed] [Google Scholar]
  18. Atack JR, Wafford KA, Tye SJ, Cook SM, Sohal B, Pike A, Sur C, Melillo D, Bristow L, Bromidge F, Ragan I, Kerby J, Street L, Carling R, Castro JL, Whiting P, Dawson GR, McKernan RM (2006) TPA023 [7-(1,1-dimethylethyl)-6-(2-ethyl-2H-1,2,4-triazol-3-ylmethoxy)-3-(2-fluorophenyl)-1,2,4-triazolo[4,3-b]pyridazine], an agonist selective for alpha2- and alpha3-containing GABAA receptors, is a nonsedating anxiolytic in rodents and primates. J Pharmacol Exp Ther 316:410–422. [DOI] [PubMed] [Google Scholar]
  19. Atmaca M (2020) Selective Serotonin Reuptake Inhibitor-Induced Sexual Dysfunction: Current Management Perspectives. Neuropsychiatr Dis Treat 16:1043–1050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Ator NA, Atack JR, Hargreaves RJ, Burns HD, Dawson GR (2010) Reducing abuse liability of GABAA/benzodiazepine ligands via selective partial agonist efficacy at alpha1 and alpha2/3 subtypes. J Pharmacol Exp Ther 332:4–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Ator NA, Griffiths RR (2003) Principles of drug abuse liability assessment in laboratory animals. Drug Alcohol Depend 70:S55–72. [DOI] [PubMed] [Google Scholar]
  22. Ator NA, Weerts EM, Kaminski BJ, Kautz MA, Griffiths RR (2000) Zaleplon and triazolam physical dependence assessed across increasing doses under a once-daily dosing regimen in baboons. Drug Alcohol Depend 61:69–84. [DOI] [PubMed] [Google Scholar]
  23. Baandrup L, Ebdrup BH, Rasmussen JØ, Lindschou J, Gluud C, Glenthøj BY (2018) Pharmacological interventions for benzodiazepine discontinuation in chronic benzodiazepine users. Cochrane Database Syst Rev 3:CD011481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Baldwin DS, Anderson IM, Nutt DJ, Bandelow B, Bond A, Davidson JRT, den Boer JA, Fineberg NA, Knapp M, Scott J, Wittchen H-U, British Association for Psychopharmacology (2005) Evidence-based guidelines for the pharmacological treatment of anxiety disorders: recommendations from the British Association for Psychopharmacology. J Psychopharmacol 19:567–596. [DOI] [PubMed] [Google Scholar]
  25. Balster RL (1991) Drug abuse potential evaluation in animals. Br J Addict 86:1549–1558. [DOI] [PubMed] [Google Scholar]
  26. Bandelow B (2020) Current and Novel Psychopharmacological Drugs for Anxiety Disorders. Adv Exp Med Biol 1191:347–365. [DOI] [PubMed] [Google Scholar]
  27. Banerjee J, Chandra SP, Kurwale N, Tripathi M (2014) Epileptogenic networks and drug-resistant epilepsy: Present and future perspectives of epilepsy research-Utility for the epileptologist and the epilepsy surgeon. Ann Indian Acad Neurol 17:S134–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Barresi E, Robello M, Costa B, Da Pozzo E, Baglini E, Salerno S, Da Settimo F, Martini C, Taliani S (2021) An update into the medicinal chemistry of translocator protein (TSPO) ligands. Eur J Med Chem 209:112924. [DOI] [PubMed] [Google Scholar]
  29. Barton ME, Klein BD, Wolf HH, White HS (2001) Pharmacological characterization of the 6 Hz psychomotor seizure model of partial epilepsy. Epilepsy Res 47:217–227. [DOI] [PubMed] [Google Scholar]
  30. Batinić B, Santrač A, Jančić I, Li G, Vidojević A, Marković B, Cook JM, Savić MM (2017) Positive modulation of α5 GABAA receptors in preadolescence prevents reduced locomotor response to amphetamine in adult female but not male rats prenatally exposed to lipopolysaccharide. Int J Dev Neurosci 61:31–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Batinić B, Stanković T, Stephen MR, Kodali R, Tiruveedhula VV, Li G, Scholze P, Marković BD, Obradović AL, Ernst M, Cook JM, Savić MM (2018) Attaining in vivo selectivity of positive modulation of α3βγ2 GABAA receptors in rats: A hard task! Eur Neuropsychopharmacol 28:903–914. [DOI] [PubMed] [Google Scholar]
  32. Batista LA, Moreira FA (2019) Cannabinoid CB1 receptors mediate the anxiolytic effects induced by systemic alprazolam and intra-periaqueductal gray 5-HT1A receptor activation. Neurosci Lett 703:5–10. [DOI] [PubMed] [Google Scholar]
  33. Baty V, Denis B, Goudot C, Bas V, Renkes P, Bigard MA, Boissel P, Gaucher P (1994) [Hepatitis induced by alpidem (Ananxyl). Four cases, one of them fatal]. Gastroenterol Clin Biol 18:1129–1131. [PubMed] [Google Scholar]
  34. Baumann SW, Baur R, Sigel E (2003) Individual properties of the two functional agonist sites in GABA(A) receptors. J Neurosci 23:11158–11166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Belelli D, Harrison NL, Maguire J, Macdonald RL, Walker MC, Cope DW (2009) Extrasynaptic GABAA Receptors: Form, Pharmacology, and Function. J Neurosci 29:12757–12763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Ben-Ari Y, Gaiarsa J-L, Tyzio R, Khazipov R (2007) GABA: a pioneer transmitter that excites immature neurons and generates primitive oscillations. Physiol Rev 87:1215–1284. [DOI] [PubMed] [Google Scholar]
  37. Benasi G, Guidi J, Offidani E, Balon R, Rickels K, Fava GA (2018) Benzodiazepines as a Monotherapy in Depressive Disorders: A Systematic Review. Psychother Psychosom 87:65–74. [DOI] [PubMed] [Google Scholar]
  38. Berro LF, Overton JS, Reeves-Darby JA, Rowlett JK (2021) Alprazolam-induced EEG spectral power changes in rhesus monkeys: a translational model for the evaluation of the behavioral effects of benzodiazepines. Psychopharmacology (Berl). [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Besson M (2020) A Randomized Double-blind Ascending-dose Placebo-controlled Study of N-desmethylclobazam in Patients With Peripheral Neuropathic Pain. clinicaltrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT04480164 [Accessed July 19, 2021].
  40. Besson M, Matthey A, Daali Y, Poncet A, Vuilleumier P, Vuillemier P, Curatolo M, Zeilhofer HU, Desmeules J (2015) GABAergic modulation in central sensitization in humans: a randomized placebo-controlled pharmacokinetic-pharmacodynamic study comparing clobazam with clonazepam in healthy volunteers. Pain 156:397–404. [DOI] [PubMed] [Google Scholar]
  41. Bettler B, Kaupmann K, Mosbacher J, Gassmann M (2004) Molecular structure and physiological functions of GABA(B) receptors. Physiol Rev 84:835–867. [DOI] [PubMed] [Google Scholar]
  42. Bhattaccharjee SA, Murnane KS, Banga AK (2020) Transdermal delivery of breakthrough therapeutics for the management of treatment-resistant and post-partum depression. Int J Pharm 591:120007. [DOI] [PubMed] [Google Scholar]
  43. Bialer M, Johannessen SI, Koepp MJ, Levy RH, Perucca E, Tomson T, White HS (2018) Progress report on new antiepileptic drugs: A summary of the Fourteenth Eilat Conference on New Antiepileptic Drugs and Devices (EILAT XIV). II. Drugs in more advanced clinical development. Epilepsia 59:1842–1866. [DOI] [PubMed] [Google Scholar]
  44. Biggerstaff A, Kivell B, Smith JL, Mian MY, Golani LK, Rashid F, Sharmin D, Knutson DE, Cerne R, Cook JM, Witkin JM (2020) The α2,3-selective potentiators of GABAA receptors, KRM-II-81 and MP-III-80, produce anxiolytic-like effects and block chemotherapy-induced hyperalgesia in mice without tolerance development. Pharmacol Biochem Behav 196:172996. [DOI] [PubMed] [Google Scholar]
  45. Blanco-Hinojo L, Pujol J, Macià D, Martínez-Vilavella G, Martín-Santos R, Pérez-Sola V, Deus J (2021) Mapping the Synchronization Effect of Gamma-Aminobutyric Acid Inhibition on the Cerebral Cortex Using Magnetic Resonance Imaging. Brain Connect. [DOI] [PubMed] [Google Scholar]
  46. Błaszczyk B, Miziak B, Czuczwar P, Wierzchowska-Cioch E, Pluta R, Czuczwar SJ (2018) A viewpoint on rational and irrational fixed-drug combinations. Expert Rev Clin Pharmacol 11:761–771. [DOI] [PubMed] [Google Scholar]
  47. Bojić MG, Todorović L, Santrač A, Mian MY, Sharmin D, Cook JM, Savić MM (2021) Vasodilatory effects of a variety of positive allosteric modulators of GABAA receptors on rat thoracic aorta. Eur J Pharmacol 899:174023. [DOI] [PubMed] [Google Scholar]
  48. Bolkvadze T, Pitkänen A (2012) Development of post-traumatic epilepsy after controlled cortical impact and lateral fluid-percussion-induced brain injury in the mouse. J Neurotrauma 29:789–812. [DOI] [PubMed] [Google Scholar]
  49. Bouilleret V, Ridoux V, Depaulis A, Marescaux C, Nehlig A, Le Gal La Salle G (1999) Recurrent seizures and hippocampal sclerosis following intrahippocampal kainate injection in adult mice: electroencephalography, histopathology and synaptic reorganization similar to mesial temporal lobe epilepsy. Neuroscience 89:717–729. [DOI] [PubMed] [Google Scholar]
  50. Bryson S (2020) Ovid’s OV101 for Angelman Fails to Meet NEPTUNE Trial Goals. Available at: https://angelmansyndromenews.com/news-posts/2020/12/08/ov101-for-angelman-failed-to-meet-neptune-primary-endpoint-ovid-therapeutics-will-suspend-development/ [Accessed July 21, 2021].
  51. Buhl DL, DaSilva JS, Tyszkiewicz C (2017) https://www.aesnet.org/meetings_events/annual_meeting_abstracts/view/345068 American Epilepsy Society Available at: https://www.aesnet.org/meetings_events/annual_meeting_abstracts/view/345068 [Accessed July 11, 2019].
  52. Bullock A, Kaul I, Li S, Silber C, Doherty J, Kanes SJ (2021) Zuranolone as an oral adjunct to treatment of Parkinsonian tremor: A phase 2, open-label study. J Neurol Sci 421:117277. [DOI] [PubMed] [Google Scholar]
  53. Burman RJ, Selfe JS, Lee JH, van den Berg M, Calin A, Codadu NK, Wright R, Newey SE, Parrish RR, Katz AA, Wilmshurst JM, Akerman CJ, Trevelyan AJ, Raimondo JV (2019) Excitatory GABAergic signalling is associated with benzodiazepine resistance in status epilepticus. Brain 142:3482–3501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Cadeddu R, Knutson DE, Mosher LJ, Loizou S, Odeh K, Fisher JL, Cook JM, Bortolato M (2021) The α6 GABAA Receptor Positive Allosteric Modulator DK-I-56–1 Reduces Tic-Related Behaviors in Mouse Models of Tourette Syndrome. Biomolecules 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Carling RW et al. (2005) 7-(1,1-Dimethylethyl)-6-(2-ethyl-2H-1,2,4-triazol-3-ylmethoxy)-3-(2-fluorophenyl)-1,2,4-triazolo[4,3-b]pyridazine: a functionally selective gamma-aminobutyric acid(A) (GABA(A)) alpha2/alpha3-subtype selective agonist that exhibits potent anxiolytic activity but is not sedating in animal models. J Med Chem 48:7089–7092. [DOI] [PubMed] [Google Scholar]
  56. Chance FS, Abbott LF, Reyes AD (2002) Gain modulation from background synaptic input. Neuron 35:773–782. [DOI] [PubMed] [Google Scholar]
  57. Chen X, Jacobs G, de Kam M, Jaeger J, Lappalainen J, Maruff P, Smith MA, Cross AJ, Cohen A, van Gerven J (2014) The central nervous system effects of the partial GABA-Aα2,3-selective receptor modulator AZD7325 in comparison with lorazepam in healthy males. British Journal of Clinical Pharmacology 78:1298–1314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Chen Z-W, Olsen RW (2007) GABAA receptor associated proteins: a key factor regulating GABAA receptor function. J Neurochem 100:279–294. [DOI] [PubMed] [Google Scholar]
  59. Chiara DC, Jayakar SS, Zhou X, Zhang X, Savechenkov PY, Bruzik KS, Miller KW, Cohen JB (2013) Specificity of intersubunit general anesthetic binding sites in the transmembrane domain of the human α1β3γ2 GABAA receptor. J Biol Chem:jbc.M113.479725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Chiou L-C, Lee H-J, Ernst M, Huang W-J, Chou J-F, Chen H-L, Mouri A, Chen L-C, Treven M, Mamiya T, Fan P-C, Knutson DE, Witzigmann C, Cook J, Sieghart W, Nabeshima T (2018) Cerebellar α6 -subunit-containing GABAA receptors: a novel therapeutic target for disrupted prepulse inhibition in neuropsychiatric disorders. Br J Pharmacol 175:2414–2427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Choi DW, Farb DH, Fischbach GD (1977) Chlordiazepoxide selectively augments GABA action in spinal cord cell cultures. Nature 269:342–344. [DOI] [PubMed] [Google Scholar]
  62. Chou R, Deyo R, Friedly J, Skelly A, Weimer M, Fu R, Dana T, Kraegel P, Griffin J, Grusing S (2017) Systemic Pharmacologic Therapies for Low Back Pain: A Systematic Review for an American College of Physicians Clinical Practice Guideline. Ann Intern Med 166:480. [DOI] [PubMed] [Google Scholar]
  63. Chuang S-H, Reddy DS (2019) Zinc reduces antiseizure activity of neurosteroids by selective blockade of extrasynaptic GABA-A receptor-mediated tonic inhibition in the hippocampus. Neuropharmacology 148:244–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Chuang S-H, Reddy DS (2020) Isobolographic Analysis of Antiseizure Activity of the GABA Type A Receptor-Modulating Synthetic Neurosteroids Brexanolone and Ganaxolone with Tiagabine and Midazolam. J Pharmacol Exp Ther 372:285–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Ciarlone SL, Wang X, Rogawski MA, Weeber EJ (2017) Effects of the synthetic neurosteroid ganaxolone on seizure activity and behavioral deficits in an Angelman syndrome mouse model. Neuropharmacology 116:142–150. [DOI] [PubMed] [Google Scholar]
  66. Clayton T, Poe MM, Rallapalli S, Biawat P, Savić MM, Rowlett JK, Gallos G, Emala CW, Kaczorowski CC, Stafford DC, Arnold LA, Cook JM (2015) A Review of the Updated Pharmacophore for the Alpha 5 GABA(A) Benzodiazepine Receptor Model. Int J Med Chem 2015:430248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Clyburn C, Howe CA, Arnold AC, Lang CH, Travagli RA, Browning KN (2019) Perinatal high-fat diet alters development of GABAA receptor subunits in dorsal motor nucleus of vagus. Am J Physiol Gastrointest Liver Physiol 317:G40–G50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Collingridge GL, Lester RA (1989) Excitatory amino acid receptors in the vertebrate central nervous system. Pharmacol Rev 41:143–210. [PubMed] [Google Scholar]
  69. Collinson N, Kuenzi FM, Jarolimek W, Maubach KA, Cothliff R, Sur C, Smith A, Otu FM, Howell O, Atack JR, McKernan RM, Seabrook GR, Dawson GR, Whiting PJ, Rosahl TW (2002) Enhanced learning and memory and altered GABAergic synaptic transmission in mice lacking the alpha 5 subunit of the GABAA receptor. J Neurosci 22:5572–5580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Conn PJ, Pin JP (1997) Pharmacology and functions of metabotropic glutamate receptors. Annu Rev Pharmacol Toxicol 37:205–237. [DOI] [PubMed] [Google Scholar]
  71. Cook JM, Zhou H, Huang S, Sarma PVVS, Zhang C (2009) Stereospecific anxiolytic and anticonvulsant agents with reduced muscle-relaxant, sedative-hypnotic and ataxic effects. Available at: https://patents.google.com/patent/US7618958B2/en [Accessed July 11, 2019].
  72. Cooper MC, Kilvert HS, Hodgkins P, Roskell NS, Eldar-Lissai A (2019) Using Matching-Adjusted Indirect Comparisons and Network Meta-analyses to Compare Efficacy of Brexanolone Injection with Selective Serotonin Reuptake Inhibitors for Treating Postpartum Depression. CNS Drugs 33:1039–1052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Cornett EM, Amarasinghe SN, Angelette A, Abubakar T, Kaye AM, Kaye AD, Neuchat EE, Urits I, Viswanath O (2021) VALTOCO® (Diazepam Nasal Spray) for the Acute Treatment of Intermittent Stereotypic Episodes of Frequent Seizure Activity. Neurol Int 13:64–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Cosci F, Chouinard G (2020) Acute and Persistent Withdrawal Syndromes Following Discontinuation of Psychotropic Medications. Psychother Psychosom 89:283–306. [DOI] [PubMed] [Google Scholar]
  75. Costa A, D’Angelo A, Ramusino MC, Perini G, Bosone D, Derosa G, Fogari R (2021) Effects of Oral Administration of Alprazolam and Lorazepam as Hypnotics on Cardiovascular Parameters in Hypertensive Patients. J Clin Psychopharmacol 41:191–195. [DOI] [PubMed] [Google Scholar]
  76. Coull JAM, Beggs S, Boudreau D, Boivin D, Tsuda M, Inoue K, Gravel C, Salter MW, De Koninck Y (2005) BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain. Nature 438:1017–1021. [DOI] [PubMed] [Google Scholar]
  77. Crocetti L, Guerrini G (2020) GABAA receptor subtype modulators in medicinal chemistry: an updated patent review (2014-present). Expert Opin Ther Pat 30:409–432. [DOI] [PubMed] [Google Scholar]
  78. Crocetti L, Guerrini G, Cantini N, Vergelli C, Melani F, Mascia MP, Giovannoni MP (2021) “Proximity frequencies” a new parameter to evaluate the profile of GABAAR modulators. Bioorg Med Chem Lett 34:127755. [DOI] [PubMed] [Google Scholar]
  79. Crowe SF, Stranks EK (2018) The Residual Medium and Long-term Cognitive Effects of Benzodiazepine Use: An Updated Meta-analysis. Arch Clin Neuropsychol 33:901–911. [DOI] [PubMed] [Google Scholar]
  80. Cvjetkovic-Bosnjak M, Soldatovic-Stajic B, Babovic SS, Boskovic K, Jovicevic M (2015) Pregabalin versus sertraline in generalized anxiety disorder. An open label study. Eur Rev Med Pharmacol Sci 19:2120–2124. [PubMed] [Google Scholar]
  81. Dandurand A (2021) A Randomized, Double-blind, Placebo-controlled, Parallel Group, Multicenter Trial of CVL-865 as Adjunctive Therapy in Adults With Drug-Resistant Focal Onset Seizures (REALIZE Trial). clinicaltrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT04244175 [Accessed July 19, 2021].
  82. Davenport CM, Rajappa R, Katchan L, Taylor CR, Tsai M-C, Smith CM, de Jong JW, Arnold DB, Lammel S, Kramer RH (2021) Relocation of an Extrasynaptic GABAA Receptor to Inhibitory Synapses Freezes Excitatory Synaptic Strength and Preserves Memory. Neuron 109:123–134.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Dawson GR, Maubach KA, Collinson N, Cobain M, Everitt BJ, MacLeod AM, Choudhury HI, McDonald LM, Pillai G, Rycroft W, Smith AJ, Sternfeld F, Tattersall FD, Wafford KA, Reynolds DS, Seabrook GR, Atack JR (2006) An inverse agonist selective for alpha5 subunit-containing GABAA receptors enhances cognition. J Pharmacol Exp Ther 316:1335–1345. [DOI] [PubMed] [Google Scholar]
  84. de Haas SL, de Visser SJ, van der Post JP, de Smet M, Schoemaker RC, Rijnbeek B, Cohen AF, Vega JM, Agrawal NGB, Goel TV, Simpson RC, Pearson LK, Li S, Hesney M, Murphy MG, van Gerven JMA (2007) Pharmacodynamic and pharmacokinetic effects of TPA023, a GABA(A) alpha(2,3) subtype-selective agonist, compared to lorazepam and placebo in healthy volunteers. J Psychopharmacol (Oxford) 21:374–383. [DOI] [PubMed] [Google Scholar]
  85. de Haas SL, Zoethout RWM, Van Dyck K, De Smet M, Rosen LB, Murphy MG, Gottesdiener KM, Schoemaker RC, Cohen AF, van Gerven JMA (2012) The effects of TPA023, a GABAAα2,3 subtype-selective partial agonist, on essential tremor in comparison to alcohol. J Psychopharmacol (Oxford) 26:282–291. [DOI] [PubMed] [Google Scholar]
  86. De la Luz-Cuellar YE, Rodríguez-Palma EJ, Franco-Enzástiga Ú, Salinas-Abarca AB, Delgado-Lezama R, Granados-Soto V (2019) Blockade of spinal α5-GABAA receptors differentially reduces reserpine-induced fibromyalgia-type pain in female rats. Eur J Pharmacol 858:172443. [DOI] [PubMed] [Google Scholar]
  87. de Lucas AG, Ahring PK, Larsen JS, Rivera-Arconada I, Lopez-Garcia JA, Mirza NR, Munro G (2015) GABAA α5 subunit-containing receptors do not contribute to reversal of inflammatory-induced spinal sensitization as indicated by the unique selectivity profile of the GABAA receptor allosteric modulator NS16085. Biochem Pharmacol 93:370–379. [DOI] [PubMed] [Google Scholar]
  88. Dean L (2017) Carisoprodol Therapy and CYP2C19 Genotype. In: Medical Genetics Summaries (Pratt VM, Scott SA, Pirmohamed M, Esquivel B, Kane MS, Kattman BL, Malheiro AJ, eds). Bethesda (MD): National Center for Biotechnology Information (US). Available at: http://www.ncbi.nlm.nih.gov/books/NBK425390/ [Accessed April 20, 2021]. [Google Scholar]
  89. Deidda G, Allegra M, Cerri C, Naskar S, Bony G, Zunino G, Bozzi Y, Caleo M, Cancedda L (2015) Early depolarizing GABA controls critical-period plasticity in the rat visual cortex. Nat Neurosci 18:87–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Dhaliwal JS, Rosani A, Saadabadi A (2021) Diazepam. In: StatPearls. Treasure Island (FL): StatPearls Publishing. Available at: http://www.ncbi.nlm.nih.gov/books/NBK537022/ [Accessed April 20, 2021]. [Google Scholar]
  91. Dhir A, Bruun DA, Guignet M, Tsai Y-H, González E, Calsbeek J, Vu J, Saito N, Tancredi DJ, Harvey DJ, Lein PJ, Rogawski MA (2020) Allopregnanolone and perampanel as adjuncts to midazolam for treating diisopropylfluorophosphate-induced status epilepticus in rats. Ann N Y Acad Sci 1480:183–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Di Capua A, Reale A, Paolino M, Chemi G, Brogi S, Cappelli A, Giorgi G, Grande F, Di Cesare Mannelli L, Ghelardini C, Matucci R, Garofalo A, Anzini M (2020) Design, synthesis and biological evaluation of 7-substituted 4-phenyl-6H-imidazo[1,5-a]thieno[3,2-f] [1,4]diazepines as safe anxiolytic agents. Eur J Med Chem 200:112405. [DOI] [PubMed] [Google Scholar]
  93. Di Lio A, Benke D, Besson M, Desmeules J, Daali Y, Wang Z, Edwankar R, Cook JM, Zeilhofer HU (2011) HZ166, a Novel GABAA Receptor Subtype-Selective Benzodiazepine Site Ligand, Is Antihyperalgesic in Mouse Models of Inflammatory and Neuropathic Pain. Neuropharmacology 60:626–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Dias R et al. (2005) Evidence for a Significant Role of α3-Containing GABAA Receptors in Mediating the Anxiolytic Effects of Benzodiazepines. J Neurosci 25:10682– 10688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Dichtel LE, Nyer M, Dording C, Fisher LB, Cusin C, Shapero BG, Pedrelli P, Kimball AS, Rao EM, Mischoulon D, Fava M, Miller KK (2020) Effects of Open-Label, Adjunctive Ganaxolone on Persistent Depression Despite Adequate Antidepressant Treatment in Postmenopausal Women: A Pilot Study. J Clin Psychiatry 81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Dirig DM, Yaksh TL (1995) Intrathecal baclofen and muscimol, but not midazolam, are antinociceptive using the rat-formalin model. J Pharmacol Exp Ther 275:219–227. [PubMed] [Google Scholar]
  97. Dold M, Bartova L, Fugger G, Mitschek MMM, Kautzky A, Frey R, Montgomery S, Zohar J, Mendlewicz J, Souery D, Fabbri C, Serretti A, Kasper S (2020) Add-on benzodiazepine treatment in patients with major depressive disorder - results from a European cross-sectional multicenter study. Eur Neuropsychopharmacol 41:70–80. [DOI] [PubMed] [Google Scholar]
  98. Donegan JJ, Boley AM, Yamaguchi J, Toney GM, Lodge DJ (2019) Modulation of extrasynaptic GABAA alpha 5 receptors in the ventral hippocampus normalizes physiological and behavioral deficits in a circuit specific manner. Nat Commun 10:2819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Duke AN, Platt DM, Rowlett JK (2020) Tolerance and dependence following chronic alprazolam treatment: quantitative observation studies in female rhesus monkeys. Psychopharmacology (Berl) 237:1183–1194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Duveau V, Buhl DL, Evrard A, Ruggiero C, Mandé‐Niedergang B, Roucard C, Gurrell R (2018) Pronounced antiepileptic activity of the subtype‐selective GABAA-positive allosteric modulator PF‐06372865 in the GAERS absence epilepsy model. CNS Neurosci Ther 25:255–260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Eides R (2019) Cerevel Therapeutics Announces Phase 1 Results for CVL-865, a Novel α2/3/5-subtype GABAA Positive Allosteric Modulator in Development to Treat Epilepsy. BioSpace Available at: https://www.biospace.com/article/cerevel-therapeutics-announces-phase-1-results-for-cvl-865-a-novel-α2-3-5-subtype-gabaa-positive-allosteric-modulator-in-development-to-treat-epilepsy/ [Accessed July 20, 2021]. [Google Scholar]
  102. El Zahaf NA, Elhwuegi AS (2014) The effect of GABAmimetics on the duration of immobility in the forced swim test in albino mice. Libyan J Med 9:23480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Engin E, Benham RS, Rudolph U (2018) An Emerging Circuit Pharmacology of GABAA Receptors. Trends Pharmacol Sci 39:710–732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Enna SJ (1997) GABAB receptor agonists and antagonists: pharmacological properties and therapeutic possibilities. Expert Opin Investig Drugs 6:1319–1325. [DOI] [PubMed] [Google Scholar]
  105. Enna SJ, McCarson KE (2006) The role of GABA in the mediation and perception of pain. Adv Pharmacol 54:1–27. [DOI] [PubMed] [Google Scholar]
  106. Ertzgaard P, Campo C, Calabrese A (2017) Efficacy and safety of oral baclofen in the management of spasticity: A rationale for intrathecal baclofen. J Rehabil Med 49:193–203. [DOI] [PubMed] [Google Scholar]
  107. Etlin A, Bráz JM, Kuhn JA, Wang X, Hamel KA, Llewellyn-Smith IJ, Basbaum AI (2016) Functional Synaptic Integration of Forebrain GABAergic Precursors into the Adult Spinal Cord. J Neurosci 36:11634–11645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Evenseth LSM, Gabrielsen M, Sylte I (2020) The GABAB Receptor-Structure, Ligand Binding and Drug Development. Molecules 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Farrant M, Nusser Z (2005) Variations on an inhibitory theme: phasic and tonic activation of GABA(A) receptors. Nat Rev Neurosci 6:215–229. [DOI] [PubMed] [Google Scholar]
  110. Fava M, Asnis GM, Shrivastava RK, Lydiard B, Bastani B, Sheehan DV, Roth T (2011a) Improved insomnia symptoms and sleep-related next-day functioning in patients with comorbid major depressive disorder and insomnia following concomitant zolpidem extended-release 12.5 mg and escitalopram treatment: a randomized controlled trial. J Clin Psychiatry 72:914–928. [DOI] [PubMed] [Google Scholar]
  111. Fava M, McCall WV, Krystal A, Wessel T, Rubens R, Caron J, Amato D, Roth T (2006) Eszopiclone co-administered with fluoxetine in patients with insomnia coexisting with major depressive disorder. Biol Psychiatry 59:1052–1060. [DOI] [PubMed] [Google Scholar]
  112. Fava M, Schaefer K, Huang H, Wilson A, Iosifescu DV, Mischoulon D, Wessel TC (2011b) A post hoc analysis of the effect of nightly administration of eszopiclone and a selective serotonin reuptake inhibitor in patients with insomnia and anxious depression. J Clin Psychiatry 72:473–479. [DOI] [PubMed] [Google Scholar]
  113. Fee C, Prevot TD, Misquitta K, Knutson DE, Li G, Mondal P, Cook JM, Banasr M, Sibille E (2021) Behavioral deficits induced by somatostatin-positive GABA neuron silencing are rescued by alpha 5 GABA-A receptor potentiation. Int J Neuropsychopharmacol. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Fischer BD, Licata SC, Edwankar RV, Wang Z-J, Huang S, He X, Yu J, Zhou H, Johnson EM, Cook JM, Furtmüller R, Ramerstorfer J, Sieghart W, Roth BL, Majumder S, Rowlett JK (2010) Anxiolytic-like effects of 8-acetylene imidazobenzodiazepines in a rhesus monkey conflict procedure. Neuropharmacology 59:612–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Flugy A, Gagliano M, Cannizzaro C, Novara V, Cannizzaro G (1992) Antidepressant and anxiolytic effects of alprazolam versus the conventional antidepressant desipramine and the anxiolytic diazepam in the forced swim test in rats. Eur J Pharmacol 214:233–238. [DOI] [PubMed] [Google Scholar]
  116. Forkuo GS, Nieman AN, Kodali R, Zahn NM, Li G, Rashid Roni MS, Stephen MR, Harris TW, Jahan R, Guthrie ML, Yu OB, Fisher JL, Yocum GT, Emala CW, Steeber DA, Stafford DC, Cook JM, Arnold LA (2018) A Novel Orally Available Asthma Drug Candidate That Reduces Smooth Muscle Constriction and Inflammation by Targeting GABAA Receptors in the Lung. Mol Pharm 15:1766–1777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Foster NL, Chase TN, Denaro A, Hare TA, Tamminga CA (1983) THIP treatment of Huntington’s disease. Neurology 33:637–639. [DOI] [PubMed] [Google Scholar]
  118. Franco V, Canevini MP, Capovilla G, De Sarro G, Galimberti CA, Gatti G, Guerrini R, La Neve A, Rosati E, Specchio LM, Striano S, Tinuper P, Perucca E (2014) Off-label prescribing of antiepileptic drugs in pharmacoresistant epilepsy: a cross-sectional drug utilization study of tertiary care centers in Italy. CNS Drugs 28:939–949. [DOI] [PubMed] [Google Scholar]
  119. Franco-Enzástiga Ú, García G, Murbartián J, González-Barrios R, Salinas-Abarca AB, Sánchez-Hernández B, Tavares-Ferreira D, Herrera LA, Barragán-Iglesias P, Delgado-Lezama R, Price TJ, Granados-Soto V (2021) Sex-dependent pronociceptive role of spinal α5 -GABAA receptor and its epigenetic regulation in neuropathic rodents. J Neurochem 156:897–916. [DOI] [PubMed] [Google Scholar]
  120. French JA, Wechsler R, Gelfand MA, Pollard JR, Vazquez B, Friedman D, Gong LH, Kamemoto E, Isojarvi J, Cassella JV (2019) Inhaled alprazolam rapidly suppresses epileptic activity in photosensitive participants. Epilepsia 60:1602–1609. [DOI] [PubMed] [Google Scholar]
  121. Frias AT, Fernandes GG, Zangrossi H (2019) GABAA/benzodiazepine receptors in the dorsal periaqueductal gray mediate the panicolytic but not the anxiolytic effect of alprazolam in rats. Behav Brain Res 364:99–105. [DOI] [PubMed] [Google Scholar]
  122. Frieder A, Fersh M, Hainline R, Deligiannidis KM (2019) Pharmacotherapy of Postpartum Depression: Current Approaches and Novel Drug Development. CNS Drugs 33:265–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Fujita M, Woods SW, Verhoeff NP, Abi-Dargham A, Baldwin RM, Zoghbi SS, Soares JC, Jatlow PA, Krystal JH, Rajeevan N, Charney DS, Seibyl JP, Innis RB (1999) Changes of benzodiazepine receptors during chronic benzodiazepine administration in humans. Eur J Pharmacol 368:161–172. [DOI] [PubMed] [Google Scholar]
  124. Gasior M, Carter RB, Witkin JM (1999) Neuroactive steroids: potential therapeutic use in neurological and psychiatric disorders. Trends Pharmacol Sci 20:107–112. [DOI] [PubMed] [Google Scholar]
  125. Gasior M, Beekman M, Carter RB, Goldberg SR and Witkin JM Antiepileptogenic effects of the novel synthetic neuroactive steroid, ganaxolone, against pentylentetrazol-induced kindled seizures: Comparison with diazepam and valproate. Drug Development Research 44: 21–33, 1998. [Google Scholar]
  126. Gasior M, Ungard JT, Beekman M, Carter RB, Witkin JM (2000) Acute and chronic effects of the synthetic neuroactive steroid, ganaxolone, against the convulsive and lethal effects of pentylenetetrazol in seizure-kindled mice: comparison with diazepam and valproate. Neuropharmacology 39:1184–1196. [DOI] [PubMed] [Google Scholar]
  127. Gassmann M, Bettler B (2012) Regulation of neuronal GABA(B) receptor functions by subunit composition. Nat Rev Neurosci 13:380–394. [DOI] [PubMed] [Google Scholar]
  128. Gee KW, Yamamura HI (1982) A novel pyrazoloquinoline that interacts with brain benzodiazepine receptors: characterization of some in vitro and in vivo properties of CGS 9896. Life Sci 30:2245–2252. [DOI] [PubMed] [Google Scholar]
  129. George TT, Tripp J (2021) Alprazolam. In: StatPearls. Treasure Island (FL): StatPearls Publishing. Available at: http://www.ncbi.nlm.nih.gov/books/NBK538165/ [Accessed April 20, 2021]. [Google Scholar]
  130. Gerbasi ME, Kosinski M, Meltzer-Brody S, Acaster S, Fridman M, Huang M-Y, Bonthapally V, Hodgkins P, Kanes SJ, Eldar-Lissai A (2021) Achieving clinical response in postpartum depression leads to improvement in health-related quality of life. Curr Med Res Opin:1–11. [DOI] [PubMed] [Google Scholar]
  131. Gerbasi ME, Meltzer-Brody S, Acaster S, Fridman M, Bonthapally V, Hodgkins P, Kanes SJ, Eldar-Lissai A (2020) Brexanolone in Postpartum Depression: Post Hoc Analyses to Help Inform Clinical Decision-Making. J Womens Health (Larchmt). [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Gilbert M, Dinh La A, Romulo Delapaz N, Kenneth Hor W, Fan P, Qi X, Guo X, Ying J, Wang L (2020) An Emulation of Randomized Trials of Administrating Benzodiazepines in PTSD Patients for Outcomes of Suicide-Related Events. J Clin Med 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Gill H, Gill B, El-Halabi S, Chen-Li D, Lipsitz O, Rosenblat JD, Van Rheenen TE, Rodrigues NB, Mansur RB, Majeed A, Lui LMW, Nasri F, Lee Y, Mcintyre RS (2020) Antidepressant Medications and Weight Change: A Narrative Review. Obesity (Silver Spring) 28:2064–2072. [DOI] [PubMed] [Google Scholar]
  134. Gitto R, Luca LD, Sarro GD (2010) Anticonvulsants. In: Burger’s Medicinal Chemistry and Drug Discovery, pp 121–160. American Cancer Society. Available at: 10.1002/0471266949.bmc099.pub2 [Accessed July 20, 2020]. [DOI] [Google Scholar]
  135. Goddard GV, McIntyre DC, Leech CK (1969) A permanent change in brain function resulting from daily electrical stimulation. Exp Neurol 25:295–330. [DOI] [PubMed] [Google Scholar]
  136. Greenblatt DJ, Harmatz JS, Shader RI (2020) Diazepam in the Elderly: Looking Back, Ahead, and at the Evidence. J Clin Psychopharmacol 40:215–219. [DOI] [PubMed] [Google Scholar]
  137. Griebel G, Perrault G, Letang V, Granger P, Avenet P, Schoemaker H, Sanger DJ (1999) New evidence that the pharmacological effects of benzodiazepine receptor ligands can be associated with activities at different BZ (omega) receptor subtypes. Psychopharmacology (Berl) 146:205–213. [DOI] [PubMed] [Google Scholar]
  138. Griffiths RR, Wolf B (1990) Relative abuse liability of different benzodiazepines in drug abusers. J Clin Psychopharmacol 10:237–243. [PubMed] [Google Scholar]
  139. Gu C, Artelsmair M, Elmore CS, Lewis RJ, Davis P, Hall JE, Dembofsky BT, Christoph G, Smith MA, Chapdelaine M, Sunzel M (2018) Late-occurring and Long-circulating Metabolites of GABAA α 2,3 Receptor Modulator AZD7325 Involving Metabolic Cyclization and Aromatization: Relevance to MIST Analysis and Application for Patient Compliance. Drug Metab Dispos 46:303–315. [DOI] [PubMed] [Google Scholar]
  140. Gudasheva TA, Deeva OA, Pantileev AS, Mokrov GV, Rybina IV, Yarkova MA, Seredenin SB (2020) The New Dipeptide TSPO Ligands: Design, Synthesis and Structure-Anxiolytic Activity Relationship. Molecules 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Gunduz-Bruce H, Silber C, Kaul I, Rothschild AJ, Riesenberg R, Sankoh AJ, Li H, Lasser R, Zorumski CF, Rubinow DR, Paul SM, Jonas J, Doherty JJ, Kanes SJ (2019) Trial of SAGE-217 in Patients with Major Depressive Disorder. N Engl J Med 381:903–911. [DOI] [PubMed] [Google Scholar]
  142. Gunter BW, Jones SA, Paul IA, Platt DM, Rowlett JK (2016) Benzodiazepine and neuroactive steroid combinations in rats: anxiolytic-like and discriminative stimulus effects. Psychopharmacology (Berl) 233:3237–3247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Gurrell R, Dua P, Feng G, Sudworth M, Whitlock M, Reynolds DS, Butt RP (2018) A randomised, placebo-controlled clinical trial with the α2/3/5 subunit selective GABAA positive allosteric modulator PF-06372865 in patients with chronic low back pain. Pain 159:1742–1751. [DOI] [PubMed] [Google Scholar]
  144. Gurrell R, Gorman D, Whitlock M, Ogden A, Reynolds DS, DiVentura B, Abou-Khalil B, Gelfand M, Pollard J, Hogan RE, Krauss G, Sperling M, Vazquez B, Wechsler RT, Friedman D, Butt RP, French J (2019) Photosensitive epilepsy: Robust clinical efficacy of a selective GABA potentiator. Neurology 92:e1786–e1795. [DOI] [PubMed] [Google Scholar]
  145. Haefely WE (1989) Pharmacology of the benzodiazepine receptor. Eur Arch Psychiatr Neurol Sci 238:294–301. [DOI] [PubMed] [Google Scholar]
  146. Hammond DL, Drower EJ (1984) Effects of intrathecally administered THIP, baclofen and muscimol on nociceptive threshold. Eur J Pharmacol 103:121–125. [DOI] [PubMed] [Google Scholar]
  147. Hammond RS, Althaus AL, Ackley MA, Maciag C, Martinez Botella G, Salituro FG, Robichaud AJ, Doherty JJ (2017) Anticonvulsant profile of the neuroactive steroid, SGE-516, in animal models. Epilepsy Res 134:16–25. [DOI] [PubMed] [Google Scholar]
  148. Han W, Shepard RD, Lu W (2020) Regulation of GABAARs by Transmembrane Accessory Proteins. Trends Neurosci. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Hawkins NA, Lewis M, Hammond RS, Doherty JJ, Kearney JA (2017) The synthetic neuroactive steroid SGE-516 reduces seizure burden and improves survival in a Dravet syndrome mouse model. Sci Rep 7:15327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Hedegaard H, Bastian BA, Trinidad JP, Spencer M, Warner M (2018) Drugs Most Frequently Involved in Drug Overdose Deaths: United States, 2011–2016. Natl Vital Stat Rep 67:1–14. [PubMed] [Google Scholar]
  151. Hedegaard H, Bastian BA, Trinidad JP, Spencer MR, Warner M (2019) Regional Differences in the Drugs Most Frequently Involved in Drug Overdose Deaths: United States, 2017. Natl Vital Stat Rep 68:1–16. [PubMed] [Google Scholar]
  152. Heiss JD, Argersinger DP, Theodore WH, Butman JA, Sato S, Khan OI (2019) Convection-Enhanced Delivery of Muscimol in Patients with Drug-Resistant Epilepsy. Neurosurgery 85:E4–E15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Hernandez GD, Solinsky CM, Mack WJ, Kono N, Rodgers KE, Wu C-Y, Mollo AR, Lopez CM, Pawluczyk S, Bauer G, Matthews D, Shi Y, Law M, Rogawski MA, Schneider LS, Brinton RD (2020) Safety, tolerability, and pharmacokinetics of allopregnanolone as a regenerative therapeutic for Alzheimer’s disease: A single and multiple ascending dose phase 1b/2a clinical trial. Alzheimers Dement (N Y) 6:e12107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Hernández-Reyes JE, Salinas-Abarca AB, Vidal-Cantú GC, Raya-Tafolla G, Elias-Viñas D, Granados-Soto V, Delgado-Lezama R (2019) α5GABAA receptors play a pronociceptive role and avoid the rate-dependent depression of the Hoffmann reflex in diabetic neuropathic pain and reduce primary afferent excitability. Pain 160:1448–1458. [DOI] [PubMed] [Google Scholar]
  155. Hevers W, Lüddens H (1998) The diversity of GABAA receptors. Pharmacological and electrophysiological properties of GABAA channel subtypes. Mol Neurobiol 18:35–86. [DOI] [PubMed] [Google Scholar]
  156. Hines RM, Maric HM, Hines DJ, Modgil A, Panzanelli P, Nakamura Y, Nathanson AJ, Cross A, Deeb T, Brandon NJ, Davies P, Fritschy J-M, Schindelin H, Moss SJ (2018) Developmental seizures and mortality result from reducing GABAA receptor α2-subunit interaction with collybistin. Nat Commun 9:3130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Hirschtritt ME, Olfson M, Kroenke K (2021) Balancing the Risks and Benefits of Benzodiazepines. JAMA. [DOI] [PubMed] [Google Scholar]
  158. Hoffmann E, Nomikos GG, Kaul I, Raines S, Wald J, Bullock A, Sankoh AJ, Doherty J, Kanes SJ, Colquhoun H (2020) SAGE-217, A Novel GABAA Receptor Positive Allosteric Modulator: Clinical Pharmacology and Tolerability in Randomized Phase I Dose-Finding Studies. Clin Pharmacokinet 59:111–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Hofmann M, Kordás KS, Gravius A, Bölcskei K, Parsons CG, Dekundy A, Danysz W, Dézsi L, Wittko-Schneider IM, Sághy K, Gyertyán I, Horváth C (2012) Assessment of the effects of NS11394 and L-838417, α2/3 subunit-selective GABA(A) [corrected] receptor-positive allosteric modulators, in tests for pain, anxiety, memory and motor function. Behavioural pharmacology. [DOI] [PubMed] [Google Scholar]
  160. Hollister LE, Bennett JL, Kimbell I, Savage C, Overall JE (1963) DIAZEPAM IN NEWLY ADMITTED SCHIZOPHRENICS. Dis Nerv Syst 24:746–750. [PubMed] [Google Scholar]
  161. Holmes MK, Erickson K, Luckenbaugh DA, Drevets WC, Bain EE, Cannon DM, Snow J, Sahakian BJ, Manji HK, Zarate CA (2008) A comparison of cognitive functioning in medicated and unmedicated subjects with bipolar depression. Bipolar Disord 10:806–815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Homayoun H, Moghaddam B (2007) NMDA receptor hypofunction produces opposite effects on prefrontal cortex interneurons and pyramidal neurons. J Neurosci 27:11496–11500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Hosseini M, Karami Z, Janzadenh A, Jameie SB, Haji Mashhadi Z, Yousefifard M, Nasirinezhad F (2014) The Effect of Intrathecal Administration of Muscimol on Modulation of Neuropathic Pain Symptoms Resulting from Spinal Cord Injury; an Experimental Study. Emerg (Tehran) 2:151–157. [PMC free article] [PubMed] [Google Scholar]
  164. Huang Q, Cox ED, Gan T, Ma C, Bennett DW, McKernan RM, Cook JM (1999) Studies of molecular pharmacophore/receptor models for GABAA/benzodiazepine receptor subtypes: binding affinities of substituted beta-carbolines at recombinant alpha x beta 3 gamma 2 subtypes and quantitative structure-activity relationship studies via a comparative molecular field analysis. Drug Des Discov 16:55–76. [PubMed] [Google Scholar]
  165. Huang Q, He X, Ma C, Liu R, Yu S, Dayer CA, Wenger GR, McKernan R, Cook JM (2000) Pharmacophore/Receptor Models for GABAA/BzR Subtypes (α1β3γ2, α5β3γ2, and α6β3γ2) via a Comprehensive Ligand-Mapping Approach. J Med Chem 43:71–95. [DOI] [PubMed] [Google Scholar]
  166. Huskinson SL, Freeman KB, Rowlett JK (2019) Self-administration of benzodiazepine and cocaine combinations by male and female rhesus monkeys in a choice procedure: role of α1 subunit-containing GABAA receptors. Psychopharmacology (Berl) 236:3271–3279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Huxtable CA, Roberts LJ, Somogyi AA, MacIntyre PE (2011) Acute pain management in opioid-tolerant patients: a growing challenge. Anaesth Intensive Care 39:804–823. [DOI] [PubMed] [Google Scholar]
  168. Hwang JK, Kim D-S (2019) From Resection to Disconnection for Seizure Control in Pediatric Epilepsy Children. J Korean Neurosurg Soc 62:336–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Iorio MT, Vogel FD, Koniuszewski F, Scholze P, Rehman S, Simeone X, Schnürch M, Mihovilovic MD, Ernst M (2020) GABAA Receptor Ligands Often Interact with Binding Sites in the Transmembrane Domain and in the Extracellular Domain-Can the Promiscuity Code Be Cracked? Int J Mol Sci 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Isbell H (1950) Addiction to barbiturates and the barbiturate abstinence syndrome. Ann Intern Med 33:108–121. [DOI] [PubMed] [Google Scholar]
  171. Jacob TC (2019) Neurobiology and Therapeutic Potential of α5-GABA Type A Receptors. Front Mol Neurosci 12:179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Jafarpour S, Hirsch LJ, Gaínza-Lein M, Kellinghaus C, Detyniecki K (2019) Seizure cluster: Definition, prevalence, consequences, and management. Seizure 68:9–15. [DOI] [PubMed] [Google Scholar]
  173. Janković SM, Dješević M, Janković SV (2021) Experimental GABA A Receptor Agonists and Allosteric Modulators for the Treatment of Focal Epilepsy. J Exp Pharmacol 13:235–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Jann M, Kennedy WK, Lopez G (2014) Benzodiazepines: a major component in unintentional prescription drug overdoses with opioid analgesics. J Pharm Pract 27:5–16. [DOI] [PubMed] [Google Scholar]
  175. Jayakar SS, Chiara DC, Zhou X, Wu B, Bruzik KS, Miller KW, Cohen JB (2020) Photoaffinity labeling identifies an intersubunit steroid-binding site in heteromeric GABA type A (GABAA) receptors. J Biol Chem 295:11495–11512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Jucaite A, Cselényi Z, Lappalainen J, McCarthy DJ, Lee C-M, Nyberg S, Varnäs K, Stenkrona P, Halldin C, Cross A, Farde L (2017) GABAA receptor occupancy by subtype selective GABAAα2,3 modulators: PET studies in humans. Psychopharmacology (Berl) 234:707–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Kaila K, Voipio J (1987) Postsynaptic fall in intracellular pH induced by GABA-activated bicarbonate conductance. Nature 330:163–165. [DOI] [PubMed] [Google Scholar]
  178. Kasteleijn-Nolst Trenité DGA, Groenwold RHH, Schmidt B, Löscher W (2016) Single dose efficacy evaluation of two partial benzodiazepine receptor agonists in photosensitive epilepsy patients: A placebo-controlled pilot study. Epilepsy Res 122:30–36. [DOI] [PubMed] [Google Scholar]
  179. Katz MM, Koslow SH, Frazer A (1996) Onset of antidepressant activity: reexamining the structure of depression and multiple actions of drugs. Depress Anxiety 4:257–267. [DOI] [PubMed] [Google Scholar]
  180. Katz MM, Tekell JL, Bowden CL, Brannan S, Houston JP, Berman N, Frazer A (2004) Onset and early behavioral effects of pharmacologically different antidepressants and placebo in depression. Neuropsychopharmacology 29:566–579. [DOI] [PubMed] [Google Scholar]
  181. Kaupmann K, Huggel K, Heid J, Flor PJ, Bischoff S, Mickel SJ, McMaster G, Angst C, Bittiger H, Froestl W, Bettler B (1997) Expression cloning of GABA B receptors uncovers similarity to metabotropic glutamate receptors. Nature 386:239–246. [DOI] [PubMed] [Google Scholar]
  182. Kawada T (2018) Effect of ganaxolone in patients with posttraumatic stress disorder. Psychopharmacology (Berl) 235:1619. [DOI] [PubMed] [Google Scholar]
  183. Kazdoba TM, Hagerman RJ, Zolkowska D, Rogawski MA, Crawley JN (2016) Evaluation of the neuroactive steroid ganaxolone on social and repetitive behaviors in the BTBR mouse model of autism. Psychopharmacology (Berl) 233:309–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Kerzare DR, Menghani SS, Rarokar NR, Khedekar PB (2021) Development of novel indole-linked pyrazoles as anticonvulsant agents: A molecular hybridization approach. Arch Pharm (Weinheim) 354:e2000100. [DOI] [PubMed] [Google Scholar]
  185. Khisti RT, Chopde CT, Jain SP (2000) Antidepressant-like effect of the neurosteroid 3alpha-hydroxy-5alpha-pregnan-20-one in mice forced swim test. Pharmacol Biochem Behav 67:137–143. [DOI] [PubMed] [Google Scholar]
  186. Khoramjouy M, Rezaee E, Khoshnevis A, Nazari M, Nematpour M, Shahhosseini S, Tabatabai SA, Faizi M (2021) Synthesis of 4,6-diphenylpyrimidin-2-ol derivatives as new benzodiazepine receptor ligands. Bioorg Chem 109:104737. [DOI] [PubMed] [Google Scholar]
  187. Kim D, Lee S, Pyeon T, Jeong S (2015) Use of triazolam and alprazolam as premedication for general anesthesia. Korean J Anesthesiol 68:346–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Kingston L, Gu C, Guo J, Swallow S, Elmore CS (2021) The impact of radiochemistry in drug projects: The use of C-14 label in the AZD8529, AZD7325, and AZD6280 projects. J Labelled Comp Radiopharm 64:65–72. [DOI] [PubMed] [Google Scholar]
  189. Kleinman RA, Schatzberg AF (2021) Understanding the Clinical Effects and Mechanisms of Action of Neurosteroids. Am J Psychiatry 178:221–223. [DOI] [PubMed] [Google Scholar]
  190. Klepner CA, Lippa AS, Benson DI, Sano MC, Beer B (1979) Resolution of two biochemically and pharmacologically distinct benzodiazepine receptors. Pharmacol Biochem Behav 11:457–462. [DOI] [PubMed] [Google Scholar]
  191. Knabl J, Witschi R, Hösl K, Reinold H, Zeilhofer UB, Ahmadi S, Brockhaus J, Sergejeva M, Hess A, Brune K, Fritschy J-M, Rudolph U, Möhler H, Zeilhofer HU (2008) Reversal of pathological pain through specific spinal GABAA receptor subtypes. Nature 451:330–334. [DOI] [PubMed] [Google Scholar]
  192. Knabl J, Zeilhofer UB, Crestani F, Rudolph U, Zeilhofer HU (2009) Genuine antihyperalgesia by systemic diazepam revealed by experiments in GABAA receptor point-mutated mice. Pain 141:233–238. [DOI] [PubMed] [Google Scholar]
  193. Knight AR, Bowery NG (1996) The pharmacology of adenylyl cyclase modulation by GABAB receptors in rat brain slices. Neuropharmacology 35:703–712. [DOI] [PubMed] [Google Scholar]
  194. Knutson DE, Smith JL, Ping X, Jin X, Golani LK, Li G, Tiruveedhula VVNPB, Rashid F, Mian MY, Jahan R, Sharmin D, Cerne R, Cook JM, Witkin JM (2020) Imidazodiazepine Anticonvulsant, KRM-II-81, Produces Novel, Non-diazepam-like Antiseizure Effects. ACS Chem Neurosci 11:2624–2637. [DOI] [PubMed] [Google Scholar]
  195. Kobata H, Hifumi T, Hoshiyama E, Yamakawa K, Nakamura K, Soh M, Kondo Y, Yokobori S, Japan Resuscitation Council (JRC) Neuroresuscitation Task Force and the Guidelines Editorial Committee (2020) Comparison of diazepam and lorazepam for the emergency treatment of adult status epilepticus: a systemic review and meta-analysis. Acute Med Surg 7:e582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Kohut SJ, Ator NA (2008) Novel discriminative stimulus effects of TPA023B, subtype-selective gamma-aminobutyric-acid(A)/benzodiazepine modulator: comparisons with zolpidem, lorazepam, and TPA023. Pharmacol Biochem Behav 90:65–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Kostowski W, Malatyńska E, Płaźnik A, Dyr W, Danysz W (1986) Comparative studies on antidepressant action of alprazolam in different animal models. Pol J Pharmacol Pharm 38:471–481. [PubMed] [Google Scholar]
  198. Kowski AB, Weissinger F, Gaus V, Fidzinski P, Losch F, Holtkamp M (2016) Specific adverse effects of antiepileptic drugs--A true-to-life monotherapy study. Epilepsy Behav 54:150–157. [DOI] [PubMed] [Google Scholar]
  199. Krall J, Balle T, Krogsgaard-Larsen N, Sørensen TE, Krogsgaard-Larsen P, Kristiansen U, Frølund B (2015) GABAA receptor partial agonists and antagonists: structure, binding mode, and pharmacology. Adv Pharmacol 72:201–227. [DOI] [PubMed] [Google Scholar]
  200. Krystal A, Fava M, Rubens R, Wessel T, Caron J, Wilson P, Roth T, McCall WV (2007) Evaluation of eszopiclone discontinuation after cotherapy with fluoxetine for insomnia with coexisting depression. J Clin Sleep Med 3:48–55. [PubMed] [Google Scholar]
  201. Kumar K, Sharma S, Kumar P, Deshmukh R (2013) Therapeutic potential of GABA(B) receptor ligands in drug addiction, anxiety, depression and other CNS disorders. Pharmacol Biochem Behav 110:174–184. [DOI] [PubMed] [Google Scholar]
  202. Kumar M, Dillon GH (2016) Assessment of direct gating and allosteric modulatory effects of meprobamate in recombinant GABA(A) receptors. Eur J Pharmacol 775:149–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Lattanzi S, Riva A, Striano P (2021) Ganaxolone treatment for epilepsy patients: from pharmacology to place in therapy. Expert Rev Neurother:1–16. [DOI] [PubMed] [Google Scholar]
  204. Laughren TP, Battey YW, Greenblatt DJ (1982) Chronic diazepam treatment in psychiatric outpatients. J Clin Psychiatry 43:461–462. [PubMed] [Google Scholar]
  205. Laverty D, Desai R, Uchański T, Masiulis S, Stec WJ, Malinauskas T, Zivanov J, Pardon E, Steyaert J, Miller KW, Aricescu AR (2019) Cryo-EM structure of the human α1β3γ2 GABAA receptor in a lipid bilayer. Nature 565:516–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Leclercq K, Matagne A, Kaminski RM (2014) Low potency and limited efficacy of antiepileptic drugs in the mouse 6Hz corneal kindling model. Epilepsy Research 108:675–683. [DOI] [PubMed] [Google Scholar]
  207. Leonard MZ, Kangas BD (2020) Effects of oxycodone and diazepam alone and in combination on operant nociception. Behav Pharmacol 31:168–173. [DOI] [PubMed] [Google Scholar]
  208. Lévesque M, Herrington R, Leclerc L, Rogawski MA, Avoli M (2017) Allopregnanolone decreases interictal spiking and fast ripples in an animal model of mesial temporal lobe epilepsy. Neuropharmacology 121:12–19. [DOI] [PubMed] [Google Scholar]
  209. Lewter L, Cook J, Li J-X (2018) Discriminative stimulus effects of α2/α3 subtype-selective GABAA receptor positive allosteric modulators in rats. [Google Scholar]
  210. Lewter LA (2019) Characterization of novel α2/α3 subtype-selective GABAA receptor positive allosteric modulators. Buffalo, NY: A dissertation submitted to the faculty of the Graduate School of the University at Buffalo, The State University of New York. [Google Scholar]
  211. Lewter LA, Fisher JL, Siemian JN, Methuku KR, Poe MM, Cook JM, Li J-X (2017) Antinociceptive effects of a novel α2/α3 subtype selective GABAA receptor positive allosteric modulator. ACS Chem Neurosci 8:1305–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Li G, Golani LK, Jahan R, Rashid F, Cook JM (2018) Improved Synthesis of Anxiolytic, Anticonvulsant, and Antinociceptive α2/α3-GABA(A)-ergic Receptor Subtype Selective Ligands as Promising Agents to Treat Anxiety, Epilepsy, and Neuropathic Pain. Synthesis 50:4124–4132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Li Y-F (2020) A hypothesis of monoamine (5-HT) - Glutamate/GABA long neural circuit: Aiming for fast-onset antidepressant discovery. Pharmacol Ther 208:107494. [DOI] [PubMed] [Google Scholar]
  214. Liampas I, Siokas V, Brotis A, Zintzaras E, Stefanidis I, Dardiotis E (2021) Intravenous sodium valproate in status epilepticus: review and Meta-analysis. Int J Neurosci 131:70–84. [DOI] [PubMed] [Google Scholar]
  215. Licata SC, Platt DM, Cook JM, Van Linn ML, Rowlett JK (2009) Contribution of alpha1 subunit-containing gamma-aminobutyric acidA (GABAA) receptors to motor-impairing effects of benzodiazepines in squirrel monkeys. Psychopharmacology (Berl) 203:539–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Ligsay A, Van Dijck A, Nguyen DV, Lozano R, Chen Y, Bickel ES, Hessl D, Schneider A, Angkustsiri K, Tassone F, Ceulemans B, Kooy RF, Hagerman RJ (2017) A randomized double-blind, placebo-controlled trial of ganaxolone in children and adolescents with fragile X syndrome. J Neurodev Disord 9:26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Lippa A, Czobor P, Stark J, Beer B, Kostakis E, Gravielle M, Bandyopadhyay S, Russek SJ, Gibbs TT, Farb DH, Skolnick P (2005) Selective anxiolysis produced by ocinaplon, a GABA(A) receptor modulator. Proc Natl Acad Sci USA 102:7380–7385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Lippa AS, Beer B, Sano MC, Vogel RA, Meyerson LR (1981) Differential ontogeny of type 1 and type 2 benzodiazepine receptors. Life Sci 28:2343–2347. [DOI] [PubMed] [Google Scholar]
  219. Lippa AS, Klepner CA, Yunger L, Sano MC, Smith WV, Beer B (1978) Relationship between benzodiazepine receptors and experimental anxiety in rats. Pharmacol Biochem Behav 9:853–856. [DOI] [PubMed] [Google Scholar]
  220. Lippa AS, Meyerson LR, Beer B (1982) Molecular substrates of anxiety: clues from the heterogeneity of benzodiazepine receptors. Life Sci 31:1409–1417. [DOI] [PubMed] [Google Scholar]
  221. Liu J, Wang L-N (2020) Efficacy and safety of valproic acid in dementia: A systematic review with meta-analysis. Arch Gerontol Geriatr 89:104091. [DOI] [PubMed] [Google Scholar]
  222. Lobina C, Maccioni P, Lorrai I, Zaru A, Collu M, Carai MAM, Brizzi A, Mugnaini C, Gessa GL, Corelli F, Colombo G (2021) Suppressing effect of the novel positive allosteric modulator of the GABAB receptor, COR659, on locomotor hyperactivity induced by different drugs of abuse. Behav Brain Res 400:113045. [DOI] [PubMed] [Google Scholar]
  223. Locci A, Geoffroy P, Miesch M, Mensah-Nyagan A-G, Pinna G (2017) Social Isolation in Early versus Late Adolescent Mice Is Associated with Persistent Behavioral Deficits That Can Be Improved by Neurosteroid-Based Treatment. Front Cell Neurosci 11:208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Lopez E, Jeanne G, Lefort L-H, Autissier C, Picot M-C, Peyrière H, Donnadieu-Rigole H (2021) Characterization of benzodiazepine misuse and comorbidities in patients with alcohol use disorder. Fundam Clin Pharmacol. [DOI] [PubMed] [Google Scholar]
  225. López-Muñoz F, Ucha-Udabe R, Alamo C (2005) The history of barbiturates a century after their clinical introduction. Neuropsychiatr Dis Treat 1:329–343. [PMC free article] [PubMed] [Google Scholar]
  226. Lorenzo L-E, Godin AG, Ferrini F, Bachand K, Plasencia-Fernandez I, Labrecque S, Girard AA, Boudreau D, Kianicka I, Gagnon M, Doyon N, Ribeiro-da-Silva A, De Koninck Y (2020) Enhancing neuronal chloride extrusion rescues α2/α3 GABAA-mediated analgesia in neuropathic pain. Nat Commun 11:869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Löscher W, Rogawski MA (2012) How theories evolved concerning the mechanism of action of barbiturates. Epilepsia 53 Suppl 8:12–25. [DOI] [PubMed] [Google Scholar]
  228. Löscher W, Schmidt D (2006) Experimental and clinical evidence for loss of effect (tolerance) during prolonged treatment with antiepileptic drugs. Epilepsia 47:1253–1284. [DOI] [PubMed] [Google Scholar]
  229. Löscher W, Sills GJ, White HS (2021) The ups and downs of alkyl-carbamates in epilepsy therapy: How does cenobamate differ? Epilepsia 62:596–614. [DOI] [PubMed] [Google Scholar]
  230. Löw K, Crestani F, Keist R, Benke D, Brünig I, Benson JA, Fritschy JM, Rülicke T, Bluethmann H, Möhler H, Rudolph U (2000) Molecular and neuronal substrate for the selective attenuation of anxiety. Science 290:131–134. [DOI] [PubMed] [Google Scholar]
  231. Lumley L, Miller D, Muse WT, Marrero-Rosado B, de Araujo Furtado M, Stone M, McGuire J, Whalley C (2019) Neurosteroid and benzodiazepine combination therapy reduces status epilepticus and long-term effects of whole-body sarin exposure in rats. Epilepsia Open 4:382–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Maletzky BM (1980) Anxiolytic efficacy of alprazolam compared to diazepam and placebo. J Int Med Res 8:139–143. [DOI] [PubMed] [Google Scholar]
  233. Maltsev DV, Spasov AA, Yakovlev DS, Vassiliev PM, Skripka MO, Miroshnikov MV, Sultanova KT, Kochetkov AN, Divaeva LN, Kuzmenko TA, Morkovnik AS (2021) Searching for new anxiolytic agents among derivatives of 11-dialkylaminoethyl-2,3,4,5-tetrahydrodiazepino[1,2-a]benzimidazole. Eur J Pharm Sci 161:105792. [DOI] [PubMed] [Google Scholar]
  234. Mao C, Shen C, Li C, Shen D-D, Xu C, Zhang S, Zhou R, Shen Q, Chen L-N, Jiang Z, Liu J, Zhang Y (2020) Cryo-EM structures of inactive and active GABAB receptor. Cell Res 30:564–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Maramai S, Benchekroun M, Ward SE, Atack JR (2020) Subtype Selective γ-Aminobutyric Acid Type A Receptor (GABAAR) Modulators Acting at the Benzodiazepine Binding Site: An Update. J Med Chem 63:3425–3446. [DOI] [PubMed] [Google Scholar]
  236. March KL, Twilla JD, Reaves AB, Self TH, Slayton MM, Bergeron JB, Sakaan SA (2019) Lorazepam versus chlordiazepoxide for the treatment of alcohol withdrawal syndrome and prevention of delirium tremens in general medicine ward patients. Alcohol 81:56–60. [DOI] [PubMed] [Google Scholar]
  237. Margules DL, Stein L (1968) Increase of “antianxiety” activity and tolerance of behavioral depression during chronic administration of oxazepam. Psychopharmacologia 13:74–80. [DOI] [PubMed] [Google Scholar]
  238. Marson AG et al. (2007) The SANAD study of effectiveness of carbamazepine, gabapentin, lamotrigine, oxcarbazepine, or topiramate for treatment of partial epilepsy: an unblinded randomised controlled trial. The Lancet 369:1000–1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Martinez Botella G, Salituro FG, Harrison BL, Beresis RT, Bai Z, Blanco M-J, Belfort GM, Dai J, Loya CM, Ackley MA, Althaus AL, Grossman SJ, Hoffmann E, Doherty JJ, Robichaud AJ (2017) Neuroactive Steroids. 2. 3α-Hydroxy-3β-methyl-21-(4-cyano-1H-pyrazol-1′-yl)-19-nor-5β-pregnan-20-one (SAGE-217): A Clinical Next Generation Neuroactive Steroid Positive Allosteric Modulator of the (γ-Aminobutyric Acid)A Receptor. J Med Chem 60:7810–7819. [DOI] [PubMed] [Google Scholar]
  240. Masiulis S, Desai R, Uchański T, Martin IS, Laverty D, Karia D, Malinauskas T, Zivanov J, Pardon E, Kotecha A, Steyaert J, Miller KW, Aricescu AR (2019) GABA A receptor signalling mechanisms revealed by structural pharmacology. Nature 565:454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Matthey A, Daali Y, Curtin F, Poncet A, Desmeules J, Besson M (2020) GABAergic modulation of secondary hyperalgesia: A randomized controlled 4-way crossover trial with the α2-subunit preferring GABA positive allosteric modulator, N-desmethyl-clobazam in healthy volunteers. Eur J Pain 24:1094–1106. [DOI] [PubMed] [Google Scholar]
  242. McAlonan DG (2019) Modulation of the Brain Excitatory/Inhibitory (E/I) Balance Through Neuronal Systems in Autism Spectrum Disorder (ASD). clinicaltrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT03678129 [Accessed July 19, 2021].
  243. McKernan RM et al. (2000) Sedative but not anxiolytic properties of benzodiazepines are mediated by the GABA(A) receptor alpha1 subtype. Nat Neurosci 3:587–592. [DOI] [PubMed] [Google Scholar]
  244. McTague A, Martland T, Appleton R (2018) Drug management for acute tonic-clonic convulsions including convulsive status epilepticus in children. Cochrane Database Syst Rev 1:CD001905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Meglio M (2019) Status Epilepticus Drug Ganaxolone Shows Benefit in Refractory Seizures. Neurology live Available at: https://www.neurologylive.com/view/status-epilepticus-drug-ganaxolone-shows-benefit-in-refractory-seizures- [Accessed July 22, 2021]. [Google Scholar]
  246. Meglio M (2021) First Patient Enrolled in Ganaxolone Phase 3 Refractory Status Epilepticus Trial. Neurology live Available at: https://www.neurologylive.com/view/first-patient-enrolled-in-ganaxolone-phase-3-refractory-status-epilepticus-trial [Accessed July 22, 2021]. [Google Scholar]
  247. Melón L, Hammond R, Lewis M, Maguire J (2018) A Novel, Synthetic, Neuroactive Steroid Is Effective at Decreasing Depression-Like Behaviors and Improving Maternal Care in Preclinical Models of Postpartum Depression. Front Endocrinol (Lausanne) 9:703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Meng Z, Rowlett JK (2016) Self-administration of progesterone and synthetic neuroactive steroids by male rhesus monkeys. Drug Alcohol Depend 165:265–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Methuku KR, Li X, Cerne R, Gleason SD, Schkeryantz JM, Tiruveedhula VVNPB, Golani LK, Li G, Poe MM, Rahman MdT, Cook JM, Fisher JL, Witkin JM (2018) An antidepressant-related pharmacological signature for positive allosteric modulators of α2/3-containing GABAA receptors. Pharmacology Biochemistry and Behavior 170:9–13. [DOI] [PubMed] [Google Scholar]
  250. Miller PS et al. (2018) Heteromeric GABA A receptor structures in positively-modulated active states. Biophysics. Available at: 10.1101/338343 [Accessed December 31, 2020]. [DOI] [Google Scholar]
  251. Miller PS, Smart TG (2010) Binding, activation and modulation of Cys-loop receptors. Trends in Pharmacological Sciences 31:161–174. [DOI] [PubMed] [Google Scholar]
  252. Mitchell SJ, Silver RA (2003) Shunting Inhibition Modulates Neuronal Gain during Synaptic Excitation. Neuron 38:433–445. [DOI] [PubMed] [Google Scholar]
  253. Miziak B, Chrościńska-Krawczyk M, Czuczwar SJ (2020) Neurosteroids and Seizure Activity. Front Endocrinol (Lausanne) 11:541802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  254. Mody I, Pearce RA (2004) Diversity of inhibitory neurotransmission through GABA(A) receptors. Trends Neurosci 27:569–575. [DOI] [PubMed] [Google Scholar]
  255. Moerke MJ, Li G, Golani LK, Cook J, Negus SS (2019) Effects of the α2/α3-subtype-selective GABAA receptor positive allosteric modulator KRM-II-81 on pain-depressed behavior in rats: comparison with ketorolac and diazepam. Behav Pharmacol 30:452–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Mohamad FH, Has ATC (2019) The α5-Containing GABAA Receptors-a Brief Summary. J Mol Neurosci 67:343–351. [DOI] [PubMed] [Google Scholar]
  257. Möhler H, Okada T (1977) Benzodiazepine receptor: demonstration in the central nervous system. Science 198:849–851. [DOI] [PubMed] [Google Scholar]
  258. Munro G, Ahring PK, Mirza NR (2009) Developing analgesics by enhancing spinal inhibition after injury: GABAA receptor subtypes as novel targets. Trends in Pharmacological Sciences 30:453–459. [DOI] [PubMed] [Google Scholar]
  259. Munro G, Lopez-Garcia JA, Rivera-Arconada I, Erichsen HK, Nielsen EØ, Larsen JS, Ahring PK, Mirza NR (2008) Comparison of the novel subtype-selective GABAA receptor-positive allosteric modulator NS11394 [3’-[5-(1-hydroxy-1-methyl-ethyl)-benzoimidazol-1-yl]-biphenyl-2-carbonitrile] with diazepam, zolpidem, bretazenil, and gaboxadol in rat models of inflammatory and neuropathic pain. J Pharmacol Exp Ther 327:969–981. [DOI] [PubMed] [Google Scholar]
  260. Musch B, Morselli PL, Priore P (1988) Clinical studies with the new anxiolytic alpidem in anxious patients: an overview of the European experiences. Pharmacol Biochem Behav 29:803–806. [DOI] [PubMed] [Google Scholar]
  261. Nagatani T, Yamamoto T, Sugihara T, Ueki S (1987) The effect of agonists at the GABA-benzodiazepine receptor complex on the duration of immobility of mice in the forced swimming test. Eur J Pharmacol 142:17–22. [DOI] [PubMed] [Google Scholar]
  262. Napoli E, Schneider A, Wang JY, Trivedi A, Carrillo NR, Tassone F, Rogawski M, Hagerman RJ, Giulivi C (2019) Allopregnanolone Treatment Improves Plasma Metabolomic Profile Associated with GABA Metabolism in Fragile X-Associated Tremor/Ataxia Syndrome: a Pilot Study. Mol Neurobiol 56:3702–3713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. Navidpour L, Shabani S, Heidari A, Bashiri M, Ebrahim-Habibi A, Shahhosseini S, Shafaroodi H, Abbas Tabatabai S, Toolabi M (2021) 5-[Aryloxypyridyl (or nitrophenyl)]-4H-1,2,4-triazoles as novel flexible benzodiazepine analogues: Synthesis, receptor binding affinity and lipophilicity-dependent anti-seizure onset of action. Bioorg Chem 106:104504. [DOI] [PubMed] [Google Scholar]
  264. Negus SS, Miller LL (2014) Intracranial self-stimulation to evaluate abuse potential of drugs. Pharmacol Rev 66:869–917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  265. Nelson TS, Holstein SE, Baird J-P, Pittman DW (2019) Selective stimulation of central GABAAα2,3,5 receptors increases intake and motivation to consume sucrose solution in rats. Neuroscience 409:111–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  266. Neugebauer NM, Miyauchi M, Sato T, Tadano J, Akal H, Ardehali H, Meltzer HY (2018) Hippocampal GABAA antagonism reverses the novel object recognition deficit in sub-chronic phencyclidine-treated rats. Behav Brain Res 342:11–18. [DOI] [PubMed] [Google Scholar]
  267. Neumann E, Küpfer L, Zeilhofer HU (2021) The α2/α3GABAA receptor modulator TPA023B alleviates not only the sensory but also the tonic affective component of chronic pain in mice. Pain 162:421–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  268. Ng YT, Conry JA, Drummond R, Stolle J, Weinberg MA, OV-1012 Study Investigators (2011) Randomized, phase III study results of clobazam in Lennox-Gastaut syndrome. Neurology 77:1473–1481. [DOI] [PubMed] [Google Scholar]
  269. Nicholson KL, Balster RL (2003) Evaluation of the phencyclidine-like discriminative stimulus effects of novel NMDA channel blockers in rats. Psychopharmacology (Berl) 170:215–224. [DOI] [PubMed] [Google Scholar]
  270. Nickolls SA et al. (2018) Pharmacology in translation: the preclinical and early clinical profile of the novel α2/3 functionally selective GABAA receptor positive allosteric modulator PF-06372865. Br J Pharmacol 175:708–725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  271. Nickolls SA, Mace H, Fish R, Edye M, Gurrell R, Ivarsson M, Pitcher T, Tanimoto-Mori S, Richardson D, Sweatman C, Nicholson J, Ward C, Jinks J, Bell C, Young K, Rees H, Moss A, Kinloch R, McMurray G (2011) A Comparison of the α2/3/5 Selective Positive Allosteric Modulators L-838,417 and TPA023 in Preclinical Models of Inflammatory and Neuropathic Pain. Adv Pharmacol Sci 2011:608912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  272. Nicoletti F, Bruno V, Copani A, Casabona G, Knöpfel T (1996) Metabotropic glutamate receptors: a new target for the therapy of neurodegenerative disorders? Trends Neurosci 19:267–271. [DOI] [PubMed] [Google Scholar]
  273. Nilkanth PR, Ghorai SK, Sathiyanarayanan A, Dhawale K, Ahamad T, Gawande MB, Shelke SN (2020) Synthesis and Evaluation of Anticonvulsant Activity of Some Schiff Bases of 7-Amino-1,3-dihydro-2H-1,4-benzodiazepin-2-one. Chem Biodivers 17:e2000342. [DOI] [PubMed] [Google Scholar]
  274. Nipper MA, Jensen JP, Helms ML, Ford MM, Crabbe JC, Rossi DJ, Finn DA (2019) Genotype Differences in Sensitivity to the Anticonvulsant Effect of the Synthetic Neurosteroid Ganaxolone during Chronic Ethanol Withdrawal. Neuroscience 397:127–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  275. Nomura T, Hawkins NA, Kearney JA, George AL, Contractor A (2019) Potentiating α2 subunit containing perisomatic GABAA receptors protects against seizures in a mouse model of Dravet syndrome. J Physiol 597:4293–4307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  276. Nors JW, Gupta S, Goldschen-Ohm MP (2021) A critical residue in the α1M2-M3 linker regulating mammalian GABAA receptor pore gating by diazepam. Elife 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  277. Ogawa Y, Takeshima N, Hayasaka Y, Tajika A, Watanabe N, Streiner D, Furukawa TA (2019) Antidepressants plus benzodiazepines for adults with major depression. Cochrane Database Syst Rev 6:CD001026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  278. Ohtawa M, Krambis MJ, Cerne R, Schkeryantz JM, Witkin JM, Shenvi RA (2017) Synthesis of (−)-11-O-Debenzoyltashironin: Neurotrophic Sesquiterpenes Cause Hyperexcitation. J Am Chem Soc 139:9637–9644. [DOI] [PubMed] [Google Scholar]
  279. Olander ER, Madjroh N, Bunch L, Söderhielm PC, Jensen AA (2018) Delineation of the functional properties and the mechanism of action of AA29504, an allosteric agonist and positive allosteric modulator of GABAA receptors. Biochem Pharmacol 150:305–319. [DOI] [PubMed] [Google Scholar]
  280. Olsen RW (2018) GABAA receptor: Positive and negative allosteric modulators. Neuropharmacology 136:10–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  281. Olsen RW, Sieghart W (2009) GABA A receptors: subtypes provide diversity of function and pharmacology. Neuropharmacology 56:141–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Owen RM et al. (2019) Design and Identification of a Novel, Functionally Subtype Selective GABAA Positive Allosteric Modulator (PF-06372865). J Med Chem 62:5773–5796. [DOI] [PubMed] [Google Scholar]
  283. Pagliarulo N (2019) Trial setback strikes blow to Sage antidepressant ambitions. BioPharma Dive Available at: https://www.biopharmadive.com/news/sage-trial-setback-mountain-antidepressant/568513/ [Accessed April 22, 2021]. [Google Scholar]
  284. Paine TA, Chang S, Poyle R (2020) Contribution of GABAA receptor subunits to attention and social behavior. Behav Brain Res 378:112261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  285. Pandey KP, Khan ZA, Golani LK, Mondal P, Mian Y, Rashid F, Tiruveedhula VVNPB, Knutson DE, Sharmin D, Ahmed T, Rezvanian S, Zahn NM, Arnold LA, Witkin JM, Cook JM (2020) Design, synthesis and characterization of novel gamma‑aminobutyric acid type A receptor ligands. ARKIVOC 2020:242–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Papasergi-Scott MM, Robertson MJ, Seven AB, Panova O, Mathiesen JM, Skiniotis G (2020) Structures of metabotropic GABAB receptor. Nature 584:310–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  287. Park J et al. (2020) Structure of human GABAB receptor in an inactive state. Nature 584:304–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Parry HJ, Balter MB, Mellinger GD, Cisin IH, Manheimer DI (1973) National Patterns of Psychotherapeutic Drug Use. Arch Gen Psychiatry 28:769–783. [PubMed] [Google Scholar]
  289. Patatanian E, Nguyen DR (2020) Brexanolone: A Novel Drug for the Treatment of Postpartum Depression. J Pharm Pract:897190020979627. [DOI] [PubMed] [Google Scholar]
  290. Patel JB, Martin C, Malick JB (1982) Differential antagonism of the anticonflict effects of typical and atypical anxiolytics. Eur J Pharmacol 86:295–298. [DOI] [PubMed] [Google Scholar]
  291. Patel T, Kurdi MS (2015) A comparative study between oral melatonin and oral midazolam on preoperative anxiety, cognitive, and psychomotor functions. J Anaesthesiol Clin Pharmacol 31:37–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  292. Paul J, Yévenes GE, Benke D, Lio AD, Ralvenius WT, Witschi R, Scheurer L, Cook JM, Rudolph U, Fritschy J-M, Zeilhofer HU (2014) Antihyperalgesia by α2-GABAA Receptors Occurs Via a Genuine Spinal Action and Does Not Involve Supraspinal Sites. Neuropsychopharmacology 39:477–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  293. Payne JL (2021) Evaluating brexanolone for the treatment of postpartum depression. Expert Opin Pharmacother:1–6. [DOI] [PubMed] [Google Scholar]
  294. Pedapati E (2021) An Initial Double-Blind, Placebo-Controlled Two-Dose Crossover Study of AZD7325 in Adults With Fragile X Syndrome. clinicaltrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT03140813 [Accessed July 19, 2021].
  295. Perry MS (2020) New and Emerging Medications for Treatment of Pediatric Epilepsy. Pediatr Neurol 107:24–27. [DOI] [PubMed] [Google Scholar]
  296. Petrache AL, Rajulawalla A, Shi A, Wetzel A, Saito T, Saido TC, Harvey K, Ali AB (2019) Aberrant Excitatory-Inhibitory Synaptic Mechanisms in Entorhinal Cortex Microcircuits During the Pathogenesis of Alzheimer’s Disease. Cereb Cortex 29:1834–1850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Phulera S, Zhu H, Yu J, Claxton DP, Yoder N, Yoshioka C, Gouaux E (2018) Cryo-EM structure of the benzodiazepine-sensitive α1β1γ2S tri-heteromeric GABAA receptor in complex with GABA. Elife 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  298. Pieribone VA, Tsai J, Soufflet C, Rey E, Shaw K, Giller E, Dulac O (2007) Clinical Evaluation of Ganaxolone in Pediatric and Adolescent Patients with Refractory Epilepsy. Epilepsia 48:1870–1874. [DOI] [PubMed] [Google Scholar]
  299. Pilipenko V, Narbute K, Pupure J, Rumaks J, Jansone B, Klusa V (2019) Neuroprotective action of diazepam at very low and moderate doses in Alzheimer’s disease model rats. Neuropharmacology 144:319–326. [DOI] [PubMed] [Google Scholar]
  300. Ping X, Jin X (2016) Transition from Initial Hypoactivity to Hyperactivity in Cortical Layer V Pyramidal Neurons after Traumatic Brain Injury In Vivo. J Neurotrauma 33:354–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  301. Platt DM, Duggan A, Spealman RD, Cook JM, Li X, Yin W, Rowlett JK (2005) Contribution of alpha 1GABAA and alpha 5GABAA receptor subtypes to the discriminative stimulus effects of ethanol in squirrel monkeys. J Pharmacol Exp Ther 313:658–667. [DOI] [PubMed] [Google Scholar]
  302. Poe MM, Methuku KR, Li G, Verma AR, Teske KA, Stafford DC, Arnold LA, Cramer JW, Jones TM, Cerne R, Krambis MJ, Witkin JM, Jambrina E, Rehman S, Ernst M, Cook JM, Schkeryantz JM (2016) Synthesis and characterization of a novel gamma-aminobutyric acid type A (GABAA) receptor ligand that combines outstanding metabolic stability, pharmacokinetics, and anxiolytic efficacy. J Med Chem 59:10800–10806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  303. Powell JG, Garland S, Preston K, Piszczatoski C (2020) Brexanolone (Zulresso): Finally, an FDA-Approved Treatment for Postpartum Depression. Ann Pharmacother 54:157–163. [DOI] [PubMed] [Google Scholar]
  304. Prescott SA, Koninck YD (2003) Gain control of firing rate by shunting inhibition: Roles of synaptic noise and dendritic saturation. PNAS 100:2076–2081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  305. Prevot TD, Li G, Vidojevic A, Misquitta KA, Fee C, Santrac A, Knutson DE, Stephen MR, Kodali R, Zahn NM, Arnold LA, Scholze P, Fisher JL, Marković BD, Banasr M, Cook JM, Savic M, Sibille E (2019) Novel Benzodiazepine-Like Ligands with Various Anxiolytic, Antidepressant, or Pro-Cognitive Profiles. Mol Neuropsychiatry 5:84–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  306. Puasripun S, Thinrungroj N, Pinyopornpanish K, Kijdamrongthum P, Leerapun A, Chitapanarux T, Thongsawat S, Praisontarangkul O-A (2020) Efficacy and Safety of Clidinium/Chlordiazepoxide as an Add-on Therapy in Functional Dyspepsia: A Randomized, Controlled, Trial. J Neurogastroenterol Motil 26:259–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  307. Puretech Health (n.d.) LYT-300. Puretech Health Available at: https://puretechhealth.com/programs/details/lyt-300 [Accessed April 22, 2021].
  308. Qiu Z-K, Li M-S, He J-L, Liu X, Zhang G-H, Lai S, Ma J-C, Zeng J, Li Y, Wu H-W, Chen Y, Shen Y-G, Chen J-S (2015) Translocator protein mediates the anxiolytic and antidepressant effects of midazolam. Pharmacol Biochem Behav 139:77–83. [DOI] [PubMed] [Google Scholar]
  309. Rahman M, Nguyen H (2020) Valproic Acid. In: StatPearls. Treasure Island (FL): StatPearls Publishing. Available at: http://www.ncbi.nlm.nih.gov/books/NBK559112/ [Accessed January 24, 2021]. [Google Scholar]
  310. Rahman MA, Keck TM, Poe MM, Sharmin D, Cook JM, Fischer BD (2021) Synergistic antihyperalgesic and antinociceptive effects of morphine and methyl 8-ethynyl-6-(pyridin-2-yl)-4H-benzo[f]imidazo[1,5-a][1,4]diazepine-3-carboxylate (MP-III-024): a positive allosteric modulator at α2GABAA and α3GABAA receptors. Psychopharmacology (Berl) 238:1585–1592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  311. Rajagopal L, Huang M, Michael E, Kwon S, Meltzer HY (2018) TPA-023 attenuates subchronic phencyclidine-induced declarative and reversal learning deficits via GABAA receptor agonist mechanism: possible therapeutic target for cognitive deficit in schizophrenia. Neuropsychopharmacology 43:2468–2477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  312. Rakhit A (2020) A Phase 2, Randomized, Double-Blind, Parallel-Group Study Evaluating the Safety, Tolerability, and Efficacy of OV101 in Fragile X Syndrome. clinicaltrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT03697161 [Accessed July 20, 2021].
  313. Ralvenius WT, Acuña MA, Benke D, Matthey A, Daali Y, Rudolph U, Desmeules J, Zeilhofer HU, Besson M (2016) The clobazam metabolite N-desmethyl clobazam is an α2 preferring benzodiazepine with an improved therapeutic window for antihyperalgesia. Neuropharmacology 109:366–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  314. Ralvenius WT, Benke D, Acuña MA, Rudolph U, Zeilhofer HU (2015) Analgesia and unwanted benzodiazepine effects in point-mutated mice expressing only one benzodiazepine-sensitive GABAA receptor subtype. Nat Commun 6:6803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  315. RALVENIUS WT, DESMEULES J, BESSON M, Zeilhofer HU, DAALI Y, MATTHEY A (2016) Use of n-desmethylclobazam in the treatment of chronic pain disorders and related methods. Available at: https://patents.google.com/patent/EP3064208A1/en [Accessed October 8, 2019].
  316. Ralvenius WT, Neumann E, Pagani M, Acuña MA, Wildner H, Benke D, Fischer N, Rostaher A, Schwager S, Detmar M, Frauenknecht K, Aguzzi A, Hubbs JL, Rudolph U, Favrot C, Zeilhofer HU (2018) Itch suppression in mice and dogs by modulation of spinal α2 and α3GABAA receptors. Nat Commun 9:3230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  317. Randall LO, Schallek W, Heise GA, Keith EF, Bagdon RE (1960) The Psychosedative Properties of Methaminodiazepoxide. J Pharmacol Exp Ther 129:163–171. [PubMed] [Google Scholar]
  318. Rasmusson AM, Marx CE, Jain S, Farfel GM, Tsai J, Sun X, Geracioti TD, Hamner MB, Lohr J, Rosse R, Summerall L, Naylor JC, Cusin C, Lang AJ, Raman R, Stein MB (2017) A randomized controlled trial of ganaxolone in posttraumatic stress disorder. Psychopharmacology (Berl) 234:2245–2257. [DOI] [PubMed] [Google Scholar]
  319. Reddy DS (2019) Mechanism-based novel antidotes for organophosphate neurotoxicity. Curr Opin Toxicol 14:35–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  320. Reddy DS, Carver CM, Clossen B, Wu X (2019) Extrasynaptic γ-aminobutyric acid type A receptor-mediated sex differences in the antiseizure activity of neurosteroids in status epilepticus and complex partial seizures. Epilepsia 60:730–743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  321. Reddy DS, Rogawski MA (2010) Neurosteroids as endogenous regulators of seizure susceptibility and use in the treatment of epilepsy. Epilepsia 51:84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  322. Reddy DS, Rogawski MA (2012) Neurosteroids — Endogenous Regulators of Seizure Susceptibility and Role in the Treatment of Epilepsy. In: Jasper’s Basic [Google Scholar]
  323. Mechanisms of the Epilepsies, 4th ed. (Noebels JL, Avoli M, Rogawski MA, Olsen RW, Delgado-Escueta AV, eds). Bethesda (MD): National Center for Biotechnology Information (US). Available at: http://www.ncbi.nlm.nih.gov/books/NBK98218/ [Accessed January 25, 2021]. [PubMed] [Google Scholar]
  324. Reissig CJ, Harrison JA, Carter LP, Griffiths RR (2015) Inhaled vs. oral alprazolam: subjective, behavioral and cognitive effects, and modestly increased abuse potential. Psychopharmacology (Berl) 232:871–883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  325. Riban V, Bouilleret V, Pham-Lê BT, Fritschy J-M, Marescaux C, Depaulis A (2002) Evolution of hippocampal epileptic activity during the development of hippocampal sclerosis in a mouse model of temporal lobe epilepsy. Neuroscience 112:101–111. [DOI] [PubMed] [Google Scholar]
  326. Rivas FM, Stables JP, Murphree L, Edwankar RV, Edwankar CR, Huang S, Jain HD, Zhou H, Majumder S, Sankar S, Roth BL, Ramerstorfer J, Furtmüller R, Sieghart W, Cook JM (2009) Antiseizure activity of novel gamma-aminobutyric acid (A) receptor subtype-selective benzodiazepine analogues in mice and rat models. J Med Chem 52:1795–1798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  327. Rogawski MA, Heller AH (2019) Diazepam buccal film for the treatment of acute seizures. Epilepsy Behav 101:106537. [DOI] [PubMed] [Google Scholar]
  328. Rosenthal E, Metzer-Brody S (2016) Sage Therapeutics R&D Day, 2016. Available at: https://investor.sagerx.com/static-files/487e787c-b1a6-433c-a345-9e0d80b5359b [Accessed July 19, 2021].
  329. Rosenthal ES, Claassen J, Wainwright MS, Husain AM, Vaitkevicius H, Raines S, Hoffmann E, Colquhoun H, Doherty JJ, Kanes SJ (2017) Brexanolone as adjunctive therapy in super‐refractory status epilepticus. Ann Neurol 82:342–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  330. Rudolph U, Crestani F, Benke D, Brünig I, Benson JA, Fritschy JM, Martin JR, Bluethmann H, Möhler H (1999) Benzodiazepine actions mediated by specific gamma-aminobutyric acid(A) receptor subtypes. Nature 401:796–800. [DOI] [PubMed] [Google Scholar]
  331. Rudolph U, Knoflach F (2011) Beyond classical benzodiazepines: novel therapeutic potential of GABA A receptor subtypes. Nat Rev Drug Discov 10:685–697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  332. Rudolph U, Möhler H (2014) GABAA receptor subtypes: Therapeutic potential in Down syndrome, affective disorders, schizophrenia, and autism. Annu Rev Pharmacol Toxicol 54:483–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  333. Saberi A, Nemati S, Lili EK, Esmaeilpour H, Panahi R (2021) Investigating the efficacy of fluoxetine vs. fluoxetine plus alprazolam (single therapy vs. combination therapy) in treatment of chronic tinnitus: A placebo-controlled study. Am J Otolaryngol 42:102898. [DOI] [PubMed] [Google Scholar]
  334. Samanta D (2020) PCDH19-Related Epilepsy Syndrome: A Comprehensive Clinical Review. Pediatr Neurol 105:3–9. [DOI] [PubMed] [Google Scholar]
  335. Sankar R (2012) GABA(A) receptor physiology and its relationship to the mechanism of action of the 1,5-benzodiazepine clobazam. CNS Drugs 26:229–244. [DOI] [PubMed] [Google Scholar]
  336. Sansalone L, Bratsch-Prince J, Tang S, Captain B, Mott DD, Raymo FM (2019) Photopotentiation of the GABAA receptor with caged diazepam. Proc Natl Acad Sci U S A 116:21176–21184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  337. Saporito MS, Gruner JA, DiCamillo A, Hinchliffe R, Barker-Haliski M, White HS (2019) Intravenously Administered Ganaxolone Blocks Diazepam-Resistant Lithium-Pilocarpine-Induced Status Epilepticus in Rats: Comparison with Allopregnanolone. J Pharmacol Exp Ther 368:326–337. [DOI] [PubMed] [Google Scholar]
  338. Schallek W, Horst WD, Schlosser W (1979) Mechanisms of action of benzodiazepines. Adv Pharmacol Chemother 16:45–87. [DOI] [PubMed] [Google Scholar]
  339. Schoepp DD (1994) Novel functions for subtypes of metabotropic glutamate receptors. Neurochem Int 24:439–449. [DOI] [PubMed] [Google Scholar]
  340. Scott LJ (2019) Brexanolone: First Global Approval. Drugs 79:779–783. [DOI] [PubMed] [Google Scholar]
  341. Scott S, Aricescu AR (2019) A structural perspective on GABAA receptor pharmacology. Current Opinion in Structural Biology 54:189–197. [DOI] [PubMed] [Google Scholar]
  342. Semple BD, Zamani A, Rayner G, Shultz SR, Jones NC (2019) Affective, neurocognitive and psychosocial disorders associated with traumatic brain injury and post-traumatic epilepsy. Neurobiol Dis 123:27–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  343. Shafie A, Mohammadi-Khanaposhtani M, Asadi M, Rahimi N, Ranjbar PR, Ghasemi JB, Larijani B, Mahdavi M, Shafaroodi H, Dehpour AR (2020) Novel fused 1,2,3-triazolo-benzodiazepine derivatives as potent anticonvulsant agents: design, synthesis, in vivo, and in silico evaluations. Mol Divers 24:179–189. [DOI] [PubMed] [Google Scholar]
  344. Shaye H, Ishchenko A, Lam JH, Han GW, Xue L, Rondard P, Pin J-P, Katritch V, Gati C, Cherezov V (2020) Structural basis of the activation of a metabotropic GABA receptor. Nature 584:298–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  345. Shukla AK, Jhaj R, Sadasivam B (2021) Brexanolone: panacea for postpartum depression? Reply to: “Intravenous brexanolone for postpartum depression: what it is, how well does it work, and will it be used?” Ther Adv Psychopharmacol 11:2045125321997293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  346. Sigel E, Steinmann ME (2012) Structure, Function, and Modulation of GABAA Receptors. J Biol Chem 287:40224–40231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  347. Simen A, Whitlock M, Qiu R, Miceli J, Zumpano L, Du Metz M, Dua P, Binneman B (2019) An 8-Week, Randomized, Phase 2, Double-Blind, Sequential Parallel-Group Comparison Study of Two Dose Levels of the GABAA Positive Allosteric Modulator PF-06372865 Compared With Placebo as an Adjunctive Treatment in Outpatients With Inadequate Response to Standard of Care for Generalized Anxiety Disorder. J Clin Psychopharmacol 39:20–27. [DOI] [PubMed] [Google Scholar]
  348. Sine SM, Engel AG (2006) Recent advances in Cys-loop receptor structure and function. Nature 440:448–455. [DOI] [PubMed] [Google Scholar]
  349. Sinha S, Siddiqui KA (2011) Definition of intractable epilepsy. Neurosciences (Riyadh) 16:3–9. [PubMed] [Google Scholar]
  350. Skolnick P (2012) Anxioselective anxiolytics: on a quest for the Holy Grail. Trends Pharmacol Sci 33:611–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  351. Söderhielm PC, Balle T, Bak-Nyhus S, Zhang M, Hansen KM, Ahring PK, Jensen AA (2018) Probing the molecular basis for affinity/potency- and efficacy-based subtype-selectivity exhibited by benzodiazepine-site modulators at GABAA receptors. Biochem Pharmacol 158:339–358. [DOI] [PubMed] [Google Scholar]
  352. Solomon VR, Tallapragada VJ, Chebib M, Johnston G a. R, Hanrahan JR (2019) GABA allosteric modulators: An overview of recent developments in non-benzodiazepine modulators. Eur J Med Chem 171:434–461. [DOI] [PubMed] [Google Scholar]
  353. Sperling MR, Klein P, Tsai J (2017) Randomized, double-blind, placebo-controlled phase 2 study of ganaxolone as add-on therapy in adults with uncontrolled partial-onset seizures. Epilepsia 58:558–564. [DOI] [PubMed] [Google Scholar]
  354. Speth RC, Wastek GJ, Reisine TD, Yamamura HI (1979) Benzodiazepine receptors: effect of tissue preincubation at 37 degrees C. Neurosci Lett 13:243–247. [DOI] [PubMed] [Google Scholar]
  355. Squires RF, Brastrup C (1977) Benzodiazepine receptors in rat brain. Nature 266:732–734. [DOI] [PubMed] [Google Scholar]
  356. Srivastava AK, Alex AB, Wilcox KS, White HS (2013) Rapid loss of efficacy to the antiseizure drugs lamotrigine and carbamazepine: a novel experimental model of pharmacoresistant epilepsy. Epilepsia 54:1186–1194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  357. Stone RH, Abousaud M, Abousaud A, Kobak W (2020) A Systematic Review of Intravaginal Diazepam for the Treatment of Pelvic Floor Hypertonic Disorder. J Clin Pharmacol 60 Suppl 2:S110–S120. [DOI] [PubMed] [Google Scholar]
  358. Străulea AO, Chiriţă V (2009) [The withdrawal syndrome in benzodiazepine dependence and its management]. Rev Med Chir Soc Med Nat Iasi 113:879–884. [PubMed] [Google Scholar]
  359. Strzelczyk A, Schubert-Bast S (2021) Expanding the Treatment Landscape for Lennox-Gastaut Syndrome: Current and Future Strategies. CNS Drugs 35:61–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  360. Szarmach J, Włodarczyk A, Cubała WJ, Wiglusz MS (2017) Benzodiazepines as adjunctive therapy in treatment refractory symptoms of schizophrenia. Psychiatr Danub 29:349–352. [PubMed] [Google Scholar]
  361. Tallman JF, Paul SM, Skolnick P, Gallager DW (1980) Receptors for the age of anxiety: pharmacology of the benzodiazepines. Science 207:274–281. [DOI] [PubMed] [Google Scholar]
  362. Tan KR, Brown M, Labouèbe G, Yvon C, Creton C, Fritschy J-M, Rudolph U, Lüscher C (2010) Neural bases for addictive properties of benzodiazepines. Nature 463:769–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  363. Te Beek ET, Chen X, Jacobs GE, Nahon KJ, de Kam ML, Lappalainen J, Cross AJ, van Gerven JMA, Hay JL (2015) The effects of the nonselective benzodiazepine lorazepam and the α2 /α3 subunit-selective GABAA receptor modulators AZD7325 and AZD6280 on plasma prolactin levels. Clin Pharmacol Drug Dev 4:149–154. [DOI] [PubMed] [Google Scholar]
  364. Temkin NR (2009) Preventing and treating posttraumatic seizures: the human experience. Epilepsia 50 Suppl 2:10–13. [DOI] [PubMed] [Google Scholar]
  365. Teoh L, Thompson W, Hubbard CC, Gellad W, Finn K, Suda KJ (2021) Comparison of Dental Benzodiazepine Prescriptions From the U.S., England, and Australia From 2013 to 2018. Am J Prev Med. [DOI] [PMC free article] [PubMed] [Google Scholar]
  366. Thelen C, Flaherty E, Saurine J, Sens J, Mohamed S, Pitychoutis PM (2019) Sex Differences in the Temporal Neuromolecular and Synaptogenic Effects of the Rapid-acting Antidepressant Drug Ketamine in the Mouse Brain. Neuroscience 398:182–192. [DOI] [PubMed] [Google Scholar]
  367. Thomas P, Pang Y (2020) Anti-apoptotic Actions of Allopregnanolone and Ganaxolone Mediated Through Membrane Progesterone Receptors (PAQRs) in Neuronal Cells. Front Endocrinol (Lausanne) 11:417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  368. Thompson AJ, Lester HA, Lummis SCR (2010) The structural basis of function in Cys-loop receptors. Q Rev Biophys 43:449–499. [DOI] [PubMed] [Google Scholar]
  369. Tian J, Dang H, Karashchuk N, Xu I, Kaufman DL (2019) A Clinically Applicable Positive Allosteric Modulator of GABA Receptors Promotes Human β-Cell Replication and Survival as well as GABA’s Ability to Inhibit Inflammatory T Cells. J Diabetes Res 2019:5783545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  370. Tikhonova TA, Rassokhina IV, Kondrakhin EA, Fedosov MA, Bukanova JV, Rossokhin AV, Sharonova IN, Kovalev GI, Zavarzin IV, Volkova YA (2020) Development of 1,3-thiazole analogues of imidazopyridines as potent positive allosteric modulators of GABAA receptors. Bioorg Chem 94:103334. [DOI] [PubMed] [Google Scholar]
  371. Tinker SC, Reefhuis J, Bitsko RH, Gilboa SM, Mitchell AA, Tran EL, Werler MM, Broussard CS, National Birth Defects Prevention Study (2019) Use of benzodiazepine medications during pregnancy and potential risk for birth defects, National Birth Defects Prevention Study, 1997–2011. Birth Defects Res 111:613–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  372. Tone A (2009) The age of anxiety: A history of America’s turbulent affair with tranquilizers. New York, NY, US: Basic Books. [Google Scholar]
  373. Tudeau L, Acuña MA, Albisetti GW, Neumann E, Ralvenius WT, Scheurer L, Poe M, Cook JM, Johannssen HC, Zeilhofer HU (2020) Mice lacking spinal α2GABAA receptors: Altered GABAergic neurotransmission, diminished GABAergic antihyperalgesia, and potential compensatory mechanisms preventing a hyperalgesic phenotype. Brain Res 1741:146889. [DOI] [PubMed] [Google Scholar]
  374. Tzeng H-R, Lee MT, Fan P-C, Knutson DE, Lai T-H, Sieghart W, Cook J, Chiou L-C (2020) α6GABAA Receptor Positive Modulators Alleviate Migraine-like Grimaces in Mice via Compensating GABAergic Deficits in Trigeminal Ganglia. Neurotherapeutics. [DOI] [PMC free article] [PubMed] [Google Scholar]
  375. Udrescu L, Bogdan P, Chiş A, Sîrbu IO, Topîrceanu A, Văruţ R-M, Udrescu M (2020) Uncovering New Drug Properties in Target-Based Drug-Drug Similarity Networks. Pharmaceutics 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  376. Ulrich M (2021) The Biologics News and Reports Portal. pipelinereview Available at: https://pipelinereview.com [Accessed July 20, 2021]. [Google Scholar]
  377. Vaitkevicius H, Husain AM, Rosenthal ES, Rosand J, Bobb W, Reddy K, Rogawski MA, Cole AJ (2017) First-in-man allopregnanolone use in super-refractory status epilepticus. Ann Clin Transl Neurol 4:411–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  378. van Amerongen G, Siebenga PS, Gurrell R, Dua P, Whitlock M, Gorman D, Okkerse P, Hay JL, Butt RP, Groeneveld GJ (2019) Analgesic potential of PF-06372865, an α2/α3/α5 subtype-selective GABAA partial agonist, in humans. Br J Anaesth 123:e194–e203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  379. van Marwijk H, Allick G, Wegman F, Bax A, Riphagen II (2012) Alprazolam for depression. Cochrane Database Syst Rev:CD007139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  380. Vasović D, Divović B, Treven M, Knutson DE, Steudle F, Scholze P, Obradović A, Fabjan J, Brković B, Sieghart W, Ernst M, Cook JM, Savić MM (2019) Trigeminal neuropathic pain development and maintenance in rats are suppressed by a positive modulator of α6 GABAA receptors. Eur J Pain 23:973–984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  381. Vega Alanis BA, Iorio MT, Silva LL, Bampali K, Ernst M, Schnürch M, Mihovilovic MD (2020) Allosteric GABAA Receptor Modulators—A Review on the Most Recent Heterocyclic Chemotypes and Their Synthetic Accessibility. Molecules 25 Available at: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7070463/ [Accessed December 31, 2020]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  382. Verma R, Bhatia R, Singh G, Kumar B, Mehan S, Monga V (2020) Design, synthesis and neuropharmacological evaluation of new 2,4-disubstituted-1,5-benzodiazepines as CNS active agents. Bioorg Chem 101:104010. [DOI] [PubMed] [Google Scholar]
  383. Vinkers CH, Olivier B, Hanania T, Min W, Schreiber R, Hopkins SC, Campbell U, Paterson N (2011) Discriminative stimulus properties of GABAA receptor positive allosteric modulators TPA023, ocinaplon and NG2–73 in rats trained to discriminate chlordiazepoxide or zolpidem. Eur J Pharmacol 668:190–193. [DOI] [PubMed] [Google Scholar]
  384. Vuilleumier PH, Besson M, Desmeules J, Arendt-Nielsen L, Curatolo M (2013) Evaluation of Anti-Hyperalgesic and Analgesic Effects of Two Benzodiazepines in Human Experimental Pain: A Randomized Placebo-Controlled Study. PLoS One 8 Available at: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3598812/ [Accessed April 17, 2020]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  385. Wafford KA (2005) GABAA receptor subtypes: any clues to the mechanism of benzodiazepine dependence? Current Opinion in Pharmacology 5:47–52. [DOI] [PubMed] [Google Scholar]
  386. Wafford KA, Ebert B (2006) Gaboxadol--a new awakening in sleep. Curr Opin Pharmacol 6:30–36. [DOI] [PubMed] [Google Scholar]
  387. Wafford KA, Whiting PJ, Kemp JA (1993) Differences in affinity and efficacy of benzodiazepine receptor ligands at recombinant gamma-aminobutyric acidA receptor subtypes. Mol Pharmacol 43:240–244. [PubMed] [Google Scholar]
  388. Wang B, Lv K, Liu H, Su Y, Wang H, Wang S, Bao S, Zhou W-H, Lian Q-Q (2018) Contribution of the α5 GABAA receptor to the discriminative stimulus effects of propofol in rat. Neuroreport 29:347–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  389. Wang JY, Trivedi AM, Carrillo NR, Yang J, Schneider A, Giulivi C, Adams P, Tassone F, Kim K, Rivera SM, Lubarr N, Wu C-Y, Irwin RW, Brinton RD, Olichney JM, Rogawski MA, Hagerman RJ (2017) Open-Label Allopregnanolone Treatment of Men with Fragile X-Associated Tremor/Ataxia Syndrome. Neurotherapeutics 14:1073–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  390. Wat R, Mammi M, Paredes J, Haines J, Alasmari M, Liew A, Lu VM, Arnaout O, Smith TR, Gormley WB, Aglio LS, Mekary RA, Zaidi H (2019) The Effectiveness of Antiepileptic Medications as Prophylaxis of Early Seizure in Patients with Traumatic Brain Injury Compared with Placebo or No Treatment: A Systematic Review and Meta-Analysis. World Neurosurg 122:433–440. [DOI] [PubMed] [Google Scholar]
  391. Weintraub SJ (2017) Diazepam in the Treatment of Moderate to Severe Alcohol Withdrawal. CNS Drugs 31:87–95. [DOI] [PubMed] [Google Scholar]
  392. West PJ, Saunders GW, Billingsley P, Smith MD, White HS, Metcalf CS, Wilcox KS (2018) Recurrent epileptiform discharges in the medial entorhinal cortex of kainate-treated rats are differentially sensitive to antiseizure drugs. Epilepsia 59:2035–2048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  393. Wilcox KS, Dixon-Salazar T, Sills GJ, Ben-Menachem E, White HS, Porter RJ, Dichter MA, Moshé SL, Noebels JL, Privitera MD, Rogawski MA (2013) Issues related to development of new antiseizure treatments. Epilepsia 54 Suppl 4:24–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  394. Wildin JD, Pleuvry BJ, Mawer GE, Onon T, Millington L (1990) Respiratory and sedative effects of clobazam and clonazepam in volunteers. Br J Clin Pharmacol 29:169–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  395. Wilkinson ST, Sanacora G (2019) A new generation of antidepressants: an update on the pharmaceutical pipeline for novel and rapid-acting therapeutics in mood disorders based on glutamate/GABA neurotransmitter systems. Drug Discov Today 24:606–615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  396. Williamson MJ, Paul SM, Skolnick P (1978) Labelling of benzodiazepine receptors in vivo. Nature 275:551–553. [DOI] [PubMed] [Google Scholar]
  397. Winters RW (2016) Accidental Medical Discoveries: How Tenacity and Pure Dumb Luck Changed the World, 1 edition. Skyhorse Publishing. [Google Scholar]
  398. Witkin JM et al. (2018) Bioisosteres of ethyl 8-ethynyl-6-(pyridin-2-yl)-4H-benzo[f]imidazo [1,5-a][1,4]diazepine-3-carboxylate (HZ-166) as novel alpha 2,3 selective potentiators of GABAA receptors: Improved bioavailability enhances anticonvulsant efficacy. Neuropharmacology 137:332–343. [DOI] [PubMed] [Google Scholar]
  399. Witkin JM et al. (2019a) The α2,3-selective potentiator of GABAA receptors, KRM-II-81, reduces nociceptive-associated behaviors induced by formalin and spinal nerve ligation in rats. Pharmacol Biochem Behav 180:22–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  400. Witkin JM (2020) mGlu2/3 receptor antagonism: A mechanism to induce rapid antidepressant effects without ketamine-associated side-effects. Pharmacol Biochem Behav 190:172854. [DOI] [PubMed] [Google Scholar]
  401. Witkin JM, Cerne R, Wakulchik M, S J, Gleason SD, Jones TM, Li G, Arnold LA, Li J-X, Schkeryantz JM, Methuku KR, Cook JM, Poe MM (2017) Further evaluation of the potential anxiolytic activity of imidazo[1,5-a][1,4]diazepin agents selective for α2/3-containing GABAA receptors. Pharmacology Biochemistry and Behavior 157:35–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  402. Witkin JM, Golani L, Smith JL (2021) New and emerging antiepileptic drugs. In: Burger’s Medicinal Chemistry, Drug Discovery and Development, Eighth Edition. Hoboken, NJ: John Wiley and Sons, Inc. [Google Scholar]
  403. Witkin JM, Li G, Golani LK, Xiong W, Smith JL, Ping X, Rashid F, Jahan R, Cerne R, Cook JM, Jin X (2020) The Positive Allosteric Modulator of α2/3-Containing GABAA Receptors, KRM-II-81, Is Active in Pharmaco-Resistant Models of Epilepsy and Reduces Hyperexcitability after Traumatic Brain Injury. J Pharmacol Exp Ther 372:83–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  404. Witkin JM, Martin AE, Golani LK, Xu NZ, Smith JL (2019b) Rapid-acting antidepressants. Adv Pharmacol 86:47–96. [DOI] [PubMed] [Google Scholar]
  405. Woods JH, Katz JL, Winger G (1992) Benzodiazepines: use, abuse, and consequences. Pharmacol Rev 44:151–347. [PubMed] [Google Scholar]
  406. Wright SL (2020) Limited Utility for Benzodiazepines in Chronic Pain Management: A Narrative Review. Adv Ther 37:2604–2619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  407. Xu L, Chockalingam A, Stewart S, Shea K, Matta MK, Narayanasamy S, Pilli NR, Volpe DA, Weaver J, Zhu H, Davis MC, Rouse R (2020) Developing an animal model to detect drug-drug interactions impacting drug-induced respiratory depression. Toxicol Rep 7:188–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  408. Xue M, Liu JP, Yang YH, Suo ZW, Yang X, Hu XD (2017) Inhibition of α5 subunit-containing GABAA receptors facilitated spinal nociceptive transmission and plasticity. Eur J Pain 21:1061–1071. [DOI] [PubMed] [Google Scholar]
  409. Yawno T, Miller SL, Bennet L, Wong F, Hirst JJ, Fahey M, Walker DW (2017) Ganaxolone: A New Treatment for Neonatal Seizures. Front Cell Neurosci 11 Available at: 10.3389/fncel.2017.00246/full [Accessed January 24, 2021]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  410. Yeh C-B, Tsai M-C, Teng Y-H, Ku M-S, Huang J-Y, Wang B-Y, Tai C-L, Kornelius E, Ho S-W (2020) Association of alprazolam with major cardiovascular events in patients with hypertension. J Eval Clin Pract 26:983–991. [DOI] [PubMed] [Google Scholar]
  411. Yocum GT, Gallos G, Zhang Y, Jahan R, Stephen MR, Varagic Z, Puthenkalam R, Ernst M, Cook JM, Emala CW (2016) Targeting the γ-Aminobutyric Acid A Receptor α4 Subunit in Airway Smooth Muscle to Alleviate Bronchoconstriction. Am J Respir Cell Mol Biol 54:546–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  412. Yocum GT, Perez-Zoghbi JF, Danielsson J, Kuforiji AS, Zhang Y, Li G, Rashid Roni MS, Kodali R, Stafford DC, Arnold LA, Cook JM, Emala CW (2019) A novel GABAA receptor ligand MIDD0301 with limited blood-brain barrier penetration relaxes airway smooth muscle ex vivo and in vivo. Am J Physiol Lung Cell Mol Physiol 316:L385–L390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  413. Yu Z, Chiara DC, Savechenkov PY, Bruzik KS, Cohen JB (2019) A photoreactive analog of allopregnanolone enables identification of steroid-binding sites in a nicotinic acetylcholine receptor. J Biol Chem 294:7892–7903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  414. Zarate CA, Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, Charney DS, Manji HK (2006) A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry 63:856–864. [DOI] [PubMed] [Google Scholar]
  415. Zeilhofer HU, Ralvenius WT, Acuña MA (2015) Restoring the spinal pain gate: GABA(A) receptors as targets for novel analgesics. Adv Pharmacol 73:71–96. [DOI] [PubMed] [Google Scholar]
  416. Zhao J, Zheng Y, Liu K, Chen J, Lai N, Fei F, Shi J, Xu C, Wang S, Nishibori M, Wang Y, Chen Z (2020) HMGB1 Is a Therapeutic Target and Biomarker in Diazepam-Refractory Status Epilepticus with Wide Time Window. Neurotherapeutics 17:710–721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  417. Zheng W, Cai D-B, Zheng W, Sim K, Ungvari GS, Peng X-J, Ning Y-P, Wang G, Xiang Y-T (2019) Brexanolone for postpartum depression: A meta-analysis of randomized controlled studies. Psychiatry Res 279:83–89. [DOI] [PubMed] [Google Scholar]
  418. Zhu S, Noviello CM, Teng J, Walsh RM, Kim JJ, Hibbs RE (2018) Structure of a human synaptic GABAA receptor. Nature 559:67–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  419. Zolkowska D, Wu C-Y, Rogawski MA (2018) Intramuscular allopregnanolone and ganaxolone in a mouse model of treatment-resistant status epilepticus. Epilepsia 59 Suppl 2:220–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  420. Zolkowska D, Wu C-Y, Rogawski MA (2021) Intranasal Allopregnanolone Confers Rapid Seizure Protection: Evidence for Direct Nose-to-Brain Delivery. Neurotherapeutics. [DOI] [PMC free article] [PubMed] [Google Scholar]
  421. Zorumski CF, Paul SM, Covey DF, Mennerick S (2019) Neurosteroids as novel antidepressants and anxiolytics: GABA-A receptors and beyond. Neurobiol Stress 11:100196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  422. Zuiker R, Pour KS, Jacobs GE (2020) A Randomized, Double-blind, Placebo- and Active Comparator-controlled, Crossover Trial to Examine the Effect of Multiple Doses of CVL-865 on Panic Symptoms Induced by Carbon Dioxide Inhalation in Healthy Subject. clinicaltrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT04592536 [Accessed July 19, 2021].
  423. Zuiker RGJA, Chen X, Østerberg O, Mirza NR, Muglia P, de Kam M, Klaassen ES, van Gerven JMA (2016) NS11821, a partial subtype-selective GABAA agonist, elicits selective effects on the central nervous system in randomized controlled trial with healthy subjects. J Psychopharmacol (Oxford) 30:253–262. [DOI] [PubMed] [Google Scholar]

RESOURCES