Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Jan 8.
Published in final edited form as: AAPS J. 2021 Jan 8;23(1):22. doi: 10.1208/s12248-020-00543-z

γ-Hydroxybutyric Acid: Pharmacokinetics, Pharmacodynamics, and Toxicology

Melanie A Felmlee 1, Bridget L Morse 2, Marilyn E Morris 3,4
PMCID: PMC8098080  NIHMSID: NIHMS1693767  PMID: 33417072

Abstract

Gamma-hydroxybutyrate (GHB) is a short-chain fatty acid present endogenously in the brain and used therapeutically for the treatment of narcolepsy, as sodium oxybate, and for alcohol abuse/withdrawal. GHB is better known however as a drug of abuse and is commonly referred to as the “date-rape drug”; current use in popular culture includes recreational “chemsex,” due to its properties of euphoria, loss of inhibition, amnesia, and drowsiness. Due to the steep concentration-effect curve for GHB, overdoses occur commonly and symptoms include sedation, respiratory depression, coma, and death. GHB binds to both GHB and GABAB receptors in the brain, with pharmacological/toxicological effects mainly due to GABAB agonist effects. The pharmacokinetics of GHB are complex and include nonlinear absorption, metabolism, tissue uptake, and renal elimination processes. GHB is a substrate for monocarboxylate transporters, including both sodium-dependent transporters (SMCT1, 2; SLC5A8; SLC5A12) and proton-dependent transporters (MCT1–4; SLC16A1, 7, 8, and 3), which represent significant determinants of absorption, renal reabsorption, and brain and tissue uptake. This review will provide current information of the pharmacology, therapeutic effects, and pharmacokinetics/pharmacodynamics of GHB, as well as therapeutic strategies for the treatment of overdoses.

Keywords: GABAB receptors addiction, monocarboxylate transporters, narcolepsy, overdose treatment strategies, pharmacology, toxicity

INTRODUCTION

γ-Hydroxybutyric acid (GHB) is an endogenous short-chain fatty acid (C4H8O3) present in the central nervous system (CNS) (Fig. 1) (1) and an analog of y-aminobutyric acid (GABA), a major inhibitory neurotransmitter in the CNS. GHB was first synthesized in 1874 but was not investigated for a potential clinical use until the 1960s when the French biochemist Henri Laborit tested GHB as an analog for the inhibitory CNS neurotransmitter GABA that was capable of crossing the blood-brain barrier (2). The US Food and Drug Administration (FDA) in 2002 approved GHB in its salt form as sodium oxybate (C4H7NaO3), for the treatment of narcolepsy associated with cataplexy in adults (1,3); in 2019, it was approved for use in children over 7 years of age. GHB has been approved in other countries for other therapeutic purposes. Currently, GHB is marketed in Austria and Italy for the treatment of alcohol withdrawal as Alcover® (4). A recent review indicates that sodium oxybate is an effective, well-tolerated, and safe treatment for withdrawal and relapse prevention treatment in alcohol-dependent patients (5). GHB is also approved for use as an anesthetic in Germany where it is marketed as Somsanit® (4). Additionally, GHB was marketed as a dietary supplement and used as a sleep aid and growth hormone enhancer. Despite these numerous clinical applications, the therapeutic utility of GHB has been overshadowed by its high prevalence of abuse. Over-the-counter sales of GHB were banned in 2000 due to reports of respiratory depression and deaths following its abuse, and it was classified through the US Controlled Substances Act as a Schedule I drug. GHB was also classified as a Schedule III controlled substance in the USA for its therapeutic use in narcolepsy in 2002.

Fig. 1.

Fig. 1.

Chemical structures of GHB, and its precursors GABA, GBL and 1,4-BD

GHB is exploited illicitly for many desirable effects including euphoria, decreased inhibition, and growth hormone release (1). The abuse of GHB (known as Fantasy, Liquid Ecstasy, G) carries the risk of several severe adverse effects such as sedation, respiratory depression, hypothermia, coma, and even death (1). Ingestion of GHB dietary supplements starting in the 1980s, by body builders and athletes as a steroid alternative, due to its growth hormone–stimulating effects, led to abuse and addiction. GHB became a popular rave drug often abused at clubs and dance parties, where it was frequently co-ingested with alcohol and other drugs of abuse. A survey of 131 GHB users reported that alcohol was co-ingested by 58% of individuals, and the risk of hospital treatment increased among GHB users following alcohol co-ingestion (6). Co-ingestion of ketamine and opiates has also been documented with GHB abuse (1,7). Ketamine (street names of Special K, Kit Kat) is a dissociative anesthetic that can have respiratory depressant effects similar to GHB and has been reported to be co-ingested with GHB by 30% of users (6). MDMA (3,4-methylenedioxymethamphetamine) is a member of the class of amphetamines and is a widely abused psychostimulant drug, very often co-abused with GHB (8). GHB is most commonly referred to as a “date-rape drug,” since it can be added to alcoholic drinks as it is colorless and nearly without taste, and ingestion results in sedation, euphoria, decreased inhibitions, enhanced sex drive, and anterograde amnesia.

GHB continues to be listed on NIDA’s “Commonly Abused Drugs,” and DAWN reports over the past decade indicate consistent abuse of GHB. However, it is also well known that problems exist with the documentation of GHB abuse, which is underreported. In view of concerns about the ongoing diversion of therapeutic GHB for illicit purposes and illicit trade of the GHB precursors γ-butyrolactone (GBL) and 1,4-butanediol (1,4-BD) (Fig. 1), the World Health Organization (WHO) Expert Committee on Drug Dependence critically reviewed GHB at its 35th meeting in June 2012 (9), and a 2018 theme issue in Current Drug Metabolism focused on GHB/GBL abuse (10). The reports highlight the use of “GHB kits” and recipes for synthesis available on the internet and in books. In the WHO report, a publication by Griffiths and Johnson (11) was cited, indicating that GHB was ranked second only to pentobarbital with respect to toxicity, when taking into account withdrawal severity, cognitive impairment, and, in particular, lethality after overdose. The WHO report states “The steep dose-response curve of GHB could also cause problems in terms of the user selecting the required dosage or taking subsequent doses in quick succession.” The report concluded that the overall data indicate that “the abuse of and dependence on GHB continues to be a public health problem.” As well, the European Drug Emergencies Network reported GHB as the fourth most commonly abused drug after heroin, cocaine, and cannabis (2013–2014). Consistent with these findings, recent publications that have surveyed the use of drugs of abuse have indicated continuous use of GHB, and deaths due to its overdose, and recent reports of overdoses from the UK in 2018 (12). A recent report investigated GHB-associated deaths in London over a 4-year period from January 2011 to December 2015 (13). There was a 119% increase in GHB-associated deaths in 2015 compared with those in 2014, which can be contrasted to only a 25% increase in cocaine-associated and 10% increase in MDMA-associated deaths in the same time (13). GHB was found to be in the top five drugs involved with emergency department visits by the European Drug Emergencies Network; GHB was associated with 711 visits, more than amphetamines (593 visits), over a 1 year period (2013–2014) (1). Accidental overdose of GHB also presents a real threat as the compound is used to facilitate sexual assault and due to its expanded clinical uses including as treatment of pediatric narcolepsy type 1 (1,14).

A 2019 report from the UK indicates that misuse of GHB or its prodrugs GBL and 1,4-BD has increased greatly since the early 1990s, particularly among lesbian, gay, bisexual, and transgender individuals in recreational and sexual settings for what is commonly referred to as chemsex or “party and play” (10). GHB use among gay and bisexual men has increased in recent years, as GHB is commonly cited as a sexual-enhancement drug. A prospective observational study of Australian gay and bisexual men found that 19.5% had a history of GHB use; overdose was reported by 14.7% of users and was more common among men who used GHB at least monthly (15). Recreational drugs commonly associated with chemsex along with GHB/GBL include crystal methamphetamine, mephedrone, and ketamine (16). Abuse of GHB and its resulting toxicity due to the steep concentration–effect relationship of GHB and its co-ingestion with alcohol and other drugs of abuse remain a societal problem.

GHB Pharmacology

GHB is both a precursor and metabolite of the neurotransmitter GABA and is present in the brain in μM concentrations. GHB binds to at least two distinct populations of low- and high-affinity binding sites in the brain. The physiological effects of GHB are mediated through binding to its own GHB receptor, identified as a subset of GABAA receptors characterized by the α4, δ, and β1 subunits (1719); [3H]GHB binding to the GHB receptor is specific, saturable, and pH-dependent with optimum binding at pH 5.5. GHB is thought to act as a neuromodulator in the brain at endogenous concentrations (20). Kinetic and pharmacological studies have focused on binding in the brain using [3H]-GHB as a ligand, identifying binding with Kd values for GHB in the nM and μM ranges, for both rats and humans (21), similar to endogenous GHB concentrations in brain tissue. GHB-induced epileptic generalized absence seizures were absent in mice and rats after knock-down of δ subunit of GABAA. The small-molecule ligand NCS-382 ((E)-2-(5-hydroxy-5,7,8,9-tetrahydro-6H-benzo[7]annulen-6-ylidene)acetic acid), an analog of γ-hydroxybutyric acid, can bind to the GHB receptor with higher affinity than GHB and can inhibit the uptake of GHB by GHB receptors. Other inhibitors are described by Bay et al. (17). GHB binding sites have also been reported in other tissues, including heart, pancreas, liver, and kidneys of rats, although the physiological functions of these peripheral GHB sites are unknown (21). Additionally, studies have demonstrated the ability of GHB to affect the release of neurotransmitters in the brain, including GABA, glutamate, and dopamine, which may contribute to its effect (2224). While some studies indicate the involvement of the GHB receptor in the pharmacological effects of GHB, many studies have demonstrated that the behavioral/pharmacological/toxicological effects of GHB are attributed to action at GABAB receptors.

Toxicological effects of GHB, and its prodrugs GBL and 1,4-BD, include sedation, hypothermia, respiratory depression, and fatality, and can be attributed to agonism at GABAB receptors (19,2527). The therapeutic use of GHB in reducing the symptoms of narcolepsy is due to the GHB-mediated stimulation of slow-wave “deep sleep” with no effect on REM sleep (28). The typical dose for this clinical indication is 4.5 g daily at bedtime in two divided doses 4 h apart. GHB is a partial weak agonist of the GABAB receptor, although with much lower affinity than for its own receptor, with Kd values estimated to be in the range of μM to mM (2932). The GABAB receptor is an obligate heterodimer, which functions as a G protein–coupled receptor (33). When activated, this receptor produces inhibitory responses at both presynaptic and postsynaptic sites. Presynaptically, GABAB activation inhibits calcium influx, thereby preventing the release of neurotransmitters (33). Postsynaptically, the activation of GABAB results in the activation of G protein–activated potassium (GIRK or Kir3) channels, which allow the efflux of potassium producing slow inhibitory postsynaptic currents (33). GHB binding to GABAB receptors increases brain concentrations of the endogenous neurosteroids 3a,5a-tetrahydroprogesterone (3a,5a-THP) and 3a,5a-tetrahydrodeoxycorticosterone (3a,5a-THDOC) in rats (17), which may play a role in its pharmacological effects. The administration of GABAB receptor antagonists, 2S)(+)-5,5-dimethyl-2-morpholineacetic acid (SCH50911) and (3-aminopropyl)(cyclohexylmethyl)phosphinic acid (SGS742, CGP46381), completely prevents the sedative/hypnotic and respiratory depressive effects and lethality of GHB in mice (19,2527). SCH50911 was also capable of completely abolishing the sedative/hypnotic effect of the GABAB receptor agonist, baclofen (19). Additional studies have demonstrated a lack of sedative/hypnotic and hypothermic effects following the administration of 1000-mg/kg GHB intraperitoneally to GABAB receptor-deficient mice (18,25). Studies from our laboratory have further shown that GHB produces a dose-dependent decline in breathing frequency and lethality in rats, which can be completely abolished by pretreatment with the GABAB receptor antagonists, SCH50911 and SGS742 (Fig. 2) (26). SCH50911 is a potent GABAB receptor antagonist that has an IC50 in rat brain of 1.1 μM (34,35). SCH50911 displays selectivity for the GABAB receptor and is able to penetrate the CNS, which makes it a useful compound for use as a GABAB receptor antagonist in vivo (34). SGS742 is another potent GABAB antagonist with an IC50 of 38 μM, and although it is less potent than SCH50911, it still has a higher affinity for GABAB receptors than GHB (36). SGS742 is currently in a phase II trial for the treatment of succinic semialdehyde dehydrogenase deficiency (SSADH), a rare neurological disorder characterized by lack of one of two enzymes involved in the breakdown of GABA in the brain, resulting in increased concentrations of both GABA and GHB. SGS742 has been shown to be effective in altering sedation and respiratory depression produced by both intravenously and orally administered GHB in vivo in rats. The compound reduces sleep time in rats, and ataxia and muscle relaxation in baboons, and precipitates withdrawal symptoms in chronic administration of the GBL (3739). These reports confirm the involvement of the GABAB receptors in the pharmacological and toxicological effects of GHB. However, it is of note that the effects of GHB differ from those of the GABAB agonist baclofen, indicating the presence of subsets of GABAB receptors or the influence of other neurotransmitter receptors including GHB receptors (40,41). Rats are able to distinguish between GHB and baclofen in drug discrimination studies, providing support for differing mechanisms (42). Additionally, chronic baclofen use is not associated with addiction, and it is not as effective as GHB in treating narcolepsy symptoms (41).

Fig. 2.

Fig. 2.

Impact of GABAB receptor inhibition on GHB inducedrespiratory depression. GHB (1500mg/kg) was administered intravenously,alone and after pretreatment with the GABAB receptor antagonist SCH50911(150mg/kg). Inhibitor was administered intravenously 5min beforeGHB. Data are presented as mean±S.D.; n=to 5.Figure and caption adapted from (26)

GHB Metabolism

GHB metabolism is complex and involves multiple reversible pathways. Endogenous GHB is formed as part of the GABA shunt pathway within the brain with approximately 1–2% of the pathways’ flux resulting in GHB production (43). Endogenous GHB concentrations range from 2 to 5 μM in all brain regions (22), but significantly higher concentrations are achieved following exogenous GHB consumption. Less than 2% of a therapeutic dose of GHB is excreted in the urine indicating that metabolism represents the major clearance pathway (44). Multiple pathways for GHB metabolism have been identified within the brain; however, there is little or no information on the contribution of additional tissues to GHB metabolism.

Figure 3 illustrates the known metabolic routes for GHB. GHB is rapidly formed following consumption of the GHB prodrugs 1,4-butanediol and GBL, which are also considered drugs of abuse (4547). GBL can be converted to GHB prior to ingestion with a strong base, or after ingestion via blood lactonases. 1,4-butanediol is converted to 4-hydroxybutyraldehyde via alcohol dehydrogenase (ADH), which is subsequently converted to GHB via aldehyde dehydrogenase (48). Metabolism of 1,4-butanediol is inhibited by fomepizole, a known ADH inhibitor leading to reduced systemic concentrations of GHB following pretreatment with fomepizole (48). GABA is metabolized to succinic semialdehyde (SSA) via GABA transaminase, which is then further reduced by SSA reductase to form endogenous GHB (49,50). GHB is metabolized within the body by distinct enzymes located in the cytosol and the mitochondria. Within the cytosol, GHB dehydrogenase converts GHB to SSA (51), which can either be taken up into the mitochondria or be converted within the cytosol to GABA (as part of the GABA shunt pathway) (49,50). GHB dehydrogenase has been identified as aldo-ketoreductase 1A1 (AKR1A1) in HepG2 cells; when AKR1A1 expression was knocked down with siRNA, GHB dehydrogenase activity was reduced by 82% (52). GHB is converted to SSA within the mitochondria via GHB transhydrogenase (also known as D-2-hydroxyglutarate transhydrogenase), which is coupled to the conversion of α-ketoglutarate to D-2-hydroxyglutaric acid (47,51). α-Ketoglutarate can be converted to glutamate, which can subsequently be converted to GABA, thereby completing the GABA shunt pathway (53). Within the mitochondria, SSA is metabolized to succinic acid by SSA dehydrogenase (ALDH5A1) (54); succinic acid subsequently enters the Kreb’s cycle and is excreted as carbon dioxide and water (55). GHB accumulation occurs in SSA dehydrogenase deficiency, suggesting that the conversion of GHB to SSA and entry into the Kreb’s cycle are the primary metabolic pathway for GHB (45,50,56). Cytosolic GHB dehydrogenase likely represents the main pathway for the conversion of GHB to SSA, as its inhibition by valproate and ethosuximide causes accumulation of GHB within the brain (57,58). GHB transhydrogenase, which converts GHB to SSA within the mitochondria, is not sensitive to inhibition by valproate and therefore is a minor pathway for GHB metabolism (57). SSA may be converted to 4,5-dihydroxyhexanoate within the mitochondria (59); however, the enzyme responsible for this reaction has not been determined. In addition, the β-oxidation spiral has been proposed as an excretion pathway for GHB resulting in the formation of dicarboxylic acids such as 3.4-dihydroxybutyric acid and glycolic acid (60,61).

Fig. 3.

Fig. 3.

Metabolism of GHB. Enzymes: 1, Alcohol dehydrogenase; 2, Aldehydedehydrogenase; 3, Blood lactonases; 4, GHB dehydrogenase (AKR1A1); 5, Succinicsemialdehyde reducatase; 6, GABA transaminase; 7, Glutamate decarboxylase; 8,Glutamate dehydrogenase; 9, GHB transhydrogenase; 10, β-oxidation spiral; 11,Succinic semialdehyde dehydrogenase (ALDH5A1)

Metabolism of GHB has primarily been evaluated in brain homogenates and crude synaptosomal membranes. AKR1A1 (GHB dehydrogenase) is ubiquitously expressed with high expression in the liver, suggesting that hepatic metabolism is likely the primary route of metabolism for exogenous GHB (62); however, quantitative analysis of the contribution of the liver to GHB metabolic clearance has not been conducted. Endogenous GHB concentrations are measurable in numerous peripheral tissues suggesting that GHB metabolism may not be restricted to the liver and brain. Mechanistic studies to elucidate GHB metabolism and metabolite kinetics in additional tissues including the liver, kidney, and intestine are necessary.

Monocarboxylate Transporters

Monocarboxylate transporters facilitate the transport of lactate and other monocarboxylates, and therefore play an important role in cellular metabolism and homeostasis.

Proton- and sodium-dependent monocarboxylate transporters (MCTs/SMCTs) are involved in the uptake and efflux of GHB in biologically important tissues and barriers, including the kidney, intestine, and blood-brain barrier (BBB) (63). Transporters are critically important for GHB pharmacokinetics due to the pKa of GHB (pKa = 4.7), which results in almost complete ionization of GHB at physiologic pH. MCT/SMCT expression governs the extent of GHB renal excretion, absorption, and brain and tissue distribution (6467).

Proton-dependent monocarboxylate transporters belong to the SLC16A family, with 14 members identified based on sequence homology (68). Only four members of this transporter family (MCT1–4) demonstrate proton-dependent transport and facilitate the transport of important endogenous monocarboxylates, including lactate, pyruvate, and ketone bodies (69,70). MCTs have a ubiquitous distribution in the body with variable expression depending on the specific isoform (70). Figure 4 is based on a recent review on MCTs by Felmlee et al. (70) that provides a detailed discussion of MCT tissue distribution. MCT1, the predominant isoform, is ubiquitous in its distribution in the body. Of importance in the distribution of GHB, MCT1 is expressed on the apical membrane of the liver and the basolateral membrane of the kidney and intestine (63,70,71). MCT2 demonstrates a more restricted tissue distribution but is present in the kidney, intestine, and other tissues including heart and skeletal muscle (70,72). MCT3 also exhibits a restricted tissue distribution, being present at the basolateral membrane of the retinal pigment epithelium and choroid plexus cells (70,71). MCT4 is present on the basolateral membrane of kidney and intestine, as well as in other tissues including skeletal muscle, heart, and lungs. MCT1 is the only isoform expressed at the BBB, with expression demonstrated at the apical and basolateral membranes in rats (73). MCT1 is also present on choroidal epithelial cells in humans and rats, present at both the apical and basolateral membranes in rats (71). Within the brain, MCT2 expression has been detected on neurons and astrocytes, although there are species differences, and MCT4 is mainly localized on astrocytes (74). Therefore, the proton-dependent MCTs 1–4 are responsible for the transport of GHB, as well as endogenous monocarboxylates such as L-lactate and other exogenous drugs including salicylate, valproic acid, and atorvastatin (8) in most tissues, including across the BBB and blood-CSF barrier, and in neurons and astrocytes within the brain.

Fig. 4.

Fig. 4.

Tissue distribution of MCTs and SMCTs in humans. (Data from references 7080)

SMCTs belong to the SLC5A family, with two members, SMCT1 (SLC5A8) and SMCT2 (SLC5A12), with neither having sequence homology with SLC16A members. SMCT1’s molecular structure differs from MCTs in that it has 7 transmembrane spanning domains with an extracellular amino terminus and an intracellular carboxyl terminus (75). SMCT1 protein expression has been identified in the kidney, intestine, brain, retina, and thyroid gland (7680). SMCT1 is expressed in the kidney cortex and outer medulla, with expression localized to the apical membrane of the S2 and S3 segments of the proximal tubules, while SMCT2 is localized to the apical membrane of the S1 segment (81). In the intestine, SMCT1 and SMCT2 are expressed on the brush border membrane. SMCTs are expressed in the brain in neurons (SMCT1) and astrocytes and glia (SMCT2) (82). Although there is no structural similarity between SMCTs and MCTs and the transport mechanisms differ, SMCTs share many substrates with proton-coupled MCTs, including D- and L-lactate, pyruvate, butyrate, and ketone bodies. A number of exogenous drugs are substrates for SMCT1, including GHB, benzoate, and salicylates; however, the pharmacological significance of SMCT2 is unknown (83). GHB has a higher affinity for SMCT1 than for MCT1, with a reported Km value of 0.68 mM (84).

Transport of GHB via Monocarboxylate Transporters

Transport of GHB is pH- and concentration-dependent in rat kidney membrane vesicles, human kidney HK-2 cells, Caco-2 cells, FRTL-5, and rat MCT1-transfected MDA-MB231 cells, and is inhibited by known MCT substrates/inhibitors such as lactate, pyruvate, and α-cyano-4-hydroxycinnamate (CHC) (72,8486). In rat kidney membrane vesicles, saturable uptake of GHB was demonstrated across the basolateral and brush border membranes and was inhibited by MCT/SMCT inhibitors (72). GHB transport was pH- and sodium-dependent, suggesting that MCTs and SMCTs are involved in active renal reabsorption of GHB (72); however, only MCT1 and MCT2 protein expressions were confirmed in the vesicles. Consistent with rat kidney membrane vesicles, transport of GHB in HK-2 cells was saturable and inhibited by MCT inhibitors (85), suggesting the role of MCT inhibitors in the renal reabsorption of GHB in both humans and rats. Following knock-down of MCT1 protein expression via siRNA in HK-2 cells, the uptake of L-lactate and GHB was significantly decreased (85), suggesting that MCT1 is the predominant transporter for GHB uptake in the kidney. GHB is a substrate for MCT2 and MCT4; however, the reductions in transport following knock-down were minimal compared to the reduction in transport following MCT1 knock-down (85). The presence of MCT1–4 protein expression was confirmed in Caco-2 cells, and GHB transport characteristics in these cells are consistent with proton-dependent MCT-mediated transport (86). In rat thyroid follicular (FRTL-5) cells, GHB transport was pH- and sodium-dependent with a Km for sodium-dependent transport of 0.68 mM, which is consistent with SMCT1-mediated transport (84). In rat MCT1-transfected MDA-MB231 cells, the Km for MCT1-mediated transport of GHB was found to be 4.6 mM (72). GHB affinity for MCT1 demonstrates pH dependence (87). In red blood cells, the Km for GHB transport shifted from 2.2 to 17.0 mM, when the extracellular pH was raised from pH 6.5 to 7.4 (87). This suggests that the Km for GHB transport will vary dependent on the tissue microenvironment.

GHB Transport at the BBB

Of the MCT/SMCT isoforms known to transport GHB, only MCT1 is expressed at the BBB in mice, rats, monkeys, and humans (71). GHB transport at the BBB was demonstrated to be saturable and carrier-mediated using in situ brain perfusion with a Km value of 11 mM (65). This is consistent with MCT1 transporter kinetics in RBCs in the absence of a pH gradient (87). Furthermore, GHB transport was inhibited by known MCT1 substrates and inhibitors, including lactate, pyruvate, and CHC (65). GHB transport kinetics were investigated in in vitro models of the rat and human BBB, RBE4, and hCMEC/D3 cells. These cell lines have been demonstrated to express MCT1 (88,89). GHB transport was concentration- and pH-dependent in RBE4 and hCMEC/D3 cells with Km values of 23.3 mM and 18.1 mM at pH 7.4 (90). MCT1-mediated uptake of GHB was inhibited in RBE4 cells by CHC and L-lactate (64,90) and in hCMEC/D3 cells by L-lactate (90) at concentrations obtained with in vivo GHB doses in rats.

GHB Pharmacokinetics

As a therapeutic agent, the pharmacokinetics of GHB have been well described in humans. GHB is marketed in the USA in the form of sodium oxybate under the brand name Xyrem®. In humans, GHB exhibits dose-dependent pharmacokinetics, even at therapeutic concentrations (9193). The nonlinear kinetics of GHB are due to saturable oral absorption, saturable metabolism, and saturable renal reabsorption (94,95). Dose-dependent pharmacokinetics have also been reported in rats and baboons (92,96). Compared to that in humans, pharmacokinetics in rats demonstrate similar nonlinearity attributed to saturable oral absorption, saturable metabolism, and saturable renal reabsorption as described below (94,96,97).

Absorption

MCTs/SMCTs are expressed in the intestine in multiple species, including rats, non-human primates, and humans, and are likely responsible for the observed saturable oral absorption of GHB (86). One clinical study reported dose-dependent oral absorption of GHB, with Tmax values increasing with increasing dose, suggesting saturable GHB absorption (92). In an evaluation of administration of GHB in doses of 200–1600 mg/kg orally in rats, the GHB Cmax increased less than proportionally with dose, and Tmax increased with dose, also suggesting saturable absorption of GHB in rats (96). This can be observed from similar subsequent experiments in rats shown in Fig. 5. In situ experiments using everted rat gut confirmed saturable intestinal transport of GHB at high mM concentrations (97). The oral administration of L-lactate with oral GHB in rats interestingly delayed the absorption of GHB, with no significant change in overall exposure, suggestive of the role of MCTs in GHB absorption and their high capacity in the intestine (98). The prolonged absorption of GHB after intragastric administration has also been observed in baboons, similar to rats, consistent with MCT-mediated absorption of GHB (97,100). Estimation of GHB bioavailability is confounded by its nonlinear pharmacokinetics. However, in rats, negligible GHB was detected in feces following oral administration, suggesting a high fraction absorbed and consistent with the high capacity of MCTs in the intestine (98).

Fig. 5.

Fig. 5.

Oral pharmacokinetics of GHB in rats and humans, with and withoutadministration of MCT inhibitor L-lactate (aand b) and of GHB following GBLadministration in rats, with and without administration of MCT inhibitorL-lactate (c). Rat figures adaptedfrom (98). Data in humans arefrom the study described in (99). Data are presentedas mean ± S.D.;n=7 to 10 in rat studies. Humanpharmacokinetic data are from 10 healthy volunteers

Distribution

Due to the ubiquitous expression of MCTs, it is likely that MCTs are involved in the distribution of GHB into tissues. The tissue distribution of GHB was assessed in rats (66), showing highest partitioning in kidney, with partition coefficients of < 1 in all other tissues. Tissue- and dose-dependent partitioning was observed, and L-lactate administration had tissue-specific effects on partitioning, consistent with the involvement of various MCT isoforms in different tissues and the bidirectional nature of MCTs. As MCT1 is highly expressed in RBCs, the dose-dependent partitioning of GHB into RBCs was separately assessed and was unexpectedly linear in rats across dose ranges that demonstrate nonlinear renal clearance (87). This is in part attributed to the higher Km value for MCT1 determined at blood pH (7.4), compared to other physiological sites, including the lumen of the proximal tubule (i.e., ~ 6.5).

As the site of GHB action and a known tissue with MCT expression, GHB partitioning into brain has been assessed in rats in various studies. Total partition coefficients in brain are < 1 and dependent on the brain region, being somewhat higher in hippocampus and frontal cortex compared to whole brain (66,101). Unbound partitioning in extracellular fluid (ECF) of the frontal cortex was also assessed in rats using microdialysis, similarly reporting low partitioning coefficients in ECF of < 0.1 across intravenous doses of 400–800 mg/kg (64,90). Intravenous administration of MCT inhibitors to rats has demonstrated a decrease in GHB brain partitioning, consistent with the role of MCTs at this barrier and may be due to either direct inhibition or potential trans-stimulation of MCT-mediated transport (64,74).

While such assessments on tissue distribution are not possible in humans, MCT expression is widespread in both humans and rats. Plasma protein binding has also been demonstrated to be similar between species (fraction unbound of ~ 1) (94,102).

Clearance

Metabolic Clearance.

Numerous reports indicate that the oral clearance of GHB decreases with increasing dose in humans, indicating saturable metabolism of GHB, even at therapeutic plasma concentrations (9193). Metabolism is the primary route of GHB elimination in humans, as very little is excreted unchanged in the urine (102). The proposed metabolic pathways for GHB are shown in Fig. 3 and described above. In rats, similarly to humans, the total and metabolic (non-renal) clearance decrease with increasing dose following IV and oral administration, as shown in Tables I and II, and the pharmacokinetics demonstrate Michaelis-Menten kinetics following IV administration (Fig. 6) (94). Through the use of pharmacokinetic modeling, the in vivo metabolic Km value for GHB in rats has been estimated to be in the range of 54–579 μg/ml or approximately 0.5–5 mM (96,103105).

Table I.

Dose-Dependent Oral Pharmacokinetics of GHB in Rats and Comparison of L-Lactate Effect on the Oral Pharmacokinetics of GHB in Rats and Humans. Adapted from (98) and (). Plasma GHB concentration profiles from these studies are shown in Fig. 5. Data are presented as mean (S.D.); n = 7 to 10 in rat studies. Human pharmacokinetic data are from 10 healthy volunteers

Rat Human
GHB alone
 Dose (mg/kg) 200 600 1500 63 mg/kg
 CL/F (ml/kg/min) 18.5 (6.3) 7.64 (2.8) 6.36 (1.1) 4.86 (1.8)
 CLR (ml/kg/min) 0.194 (0.098) 0.867 (0.51) 1.62 (0.63) 0.105 (0.053)
 % excreted 1.0 11.3 25.5 2.2
GHB with L-lactate
 CL/F (ml/kg/min) 20.9 (9.0) 15.7 (4.9)* 12.2 (1.4)* 4.86 (1.6)
 CLR (ml/kg/min) 0.48 (0.40)* 1.73 (0.92)* 2.48 (0.84) 0.146 (0.063)*
 % excreted 2.3 11.0 20.3 3.2*

CL/F total oral clearance, CLR renal clearance, CLNR non-renal (metabolic) clearance, % excreted, percent of dose excreted unchanged in urine

*

Significantly different from GHB alone (P < 0.05)

Table II.

Dose-Dependent Pharmacokinetics of GHB Following IV Administration and Effects of MCT Inhibitors (adapted from (26) and (95)). Data are presented as mean (S.D.); n = 7–10

GHB dose 200 mg/kg 600 mg/kg 1500 mg/kg 1500 mg/kg + L-lactate 1500 mg/kg + AR-C155858 1 mg/kg 1500 mg/kg + AR-C155858 5 mg/kg
Cl (ml/kg/min) 7.60 (0.29) 6.00 (0.74) 5.16 (0.70) 6.40* (0.62) 9.25* (0.75) 9.42* (0.98)
ClR (ml/kg/min) 0.444 (0.20) 1.68 (0.75) 3.18 (0.66) 4.22* (0.63) 7.09* (0.94) 7.44* (1.07)
% excreted 6% 27% 61% 66% 77% * 79% *

Cl total clearance, CLR renal clearance, % excreted, percent of dose excreted unchanged in urine

*

Significantly different from GHB 1500 mg/kg

Fig. 6.

Fig. 6.

Plasma concentrations of GHB after intravenous administration of GHB in rats. Datapresented as mean ± S.D., n=4–9.Figure and caption adapted from (26). Data are presentedas mean ± S.D.;n=4 to 6

Renal Clearance.

In humans, only 2–6% of GHB is excreted unchanged following an oral therapeutic dose (92). Since the fraction unbound in human plasma of GHB is ~ 1, it is assumed that GHB undergoes glomerular filtration, and the lack of elimination of GHB into urine can be attributed to almost complete renal reabsorption. In rats, renal clearance is similarly negligible at low doses and at plasma concentrations similar to those used therapeutically in humans (Table I); however as shown after both IV and oral administration in rats, it becomes the predominant route for its elimination at higher doses (Tables I and II) (94,106). In rats, the increased GHB renal clearance with increases in dose suggests that GHB undergoes saturable, carrier-mediated renal reabsorption (94). Administration of MCT inhibitors, including L-lactate, pyruvate, and dietary flavonoids, results in increased renal clearance of GHB in rats, further indicating saturable active renal reabsorption of GHB and that this process involves transport by MCTs (Tables I and II) (94,104,107). In humans, while renal clearance is nearly negligible at therapeutic oral doses, administration of L-lactate to humans significantly increased the renal clearance of GHB, consistent with a similar role of MCTs in the active renal reabsorption of GHB in humans, as in rats, shown in Table I (94).

Pharmacokinetics of GHB Precursors.

As a lactone, the GHB precursor GBL is rapidly converted to GHB via lactonases in the blood, and following GBL administration to rats and baboons, only GHB is detectable in plasma (108). As such, the disposition of GBL in vivo is likely irrelevant; however, the properties of GBL in the intestine, prior to systemic exposure, represent important differences with this agent compared to GHB itself. The everted rat gut studies mentioned above also evaluated the intestinal transport of GBL and interestingly demonstrated transport of GBL in these gut preparations to be much higher than GHB, lacking evident saturation of transport (97). Rat studies evaluating oral administration of equimolar doses of GHB and GBL demonstrate superior absorption of GBL compared to GHB with higher GHB Cmax values following GBL administration compared to GHB itself (Fig. 5) (109). Similar behavior of GHB following GBL absorption has been demonstrated in baboons (109). As GBL is undetectable in plasma following administration, it is unlikely that toxicodynamic effects of GBL are relevant in vivo. Additionally, intracerebroventricular administration of GBL in rats demonstrated no pharmacological effects of this agent, when avoiding systemic conversion to GHB (110). Therefore, toxicodynamic effects of concern following overdose of GBL are those of formed GHB.

Conversely, plasma concentrations of 1,4-butanediol (BD) are measurable following oral administration to rats, baboons, and humans along with those of GHB (111,112). Following an oral dose of 25 mg/kg BD to healthy volunteers, the oral clearance of BD showed high interindividual variability, as did the ratio of GHB:BD (113). A correlation was found in that subjects with lower clearance and lower GHB:BD carried variant alleles for the ALD gene. The mean GHB:BD plasma AUC ratio was 49 in this study, and mean plasma Cmax of GHB was greater than that of BD by over 10-fold, suggesting that the toxicological effects can likely be attributed to those of GHB. Additionally, in the same study with GBL, BD demonstrated no effect following intracerebroventricular administration in rats (110). The dose-dependent pharmacokinetics of BD were assessed in rats and were nonlinear, and pharmacokinetic modeling indicated that the bioconversion of BD to GHB was complete, via the two-step conversion through alcohol dehydrogenase, followed by aldehyde dehydrogenase. Similarly, the pharmacokinetics of BD were nonlinear following oral administration to baboons. However, unlike GHB, in both rats and baboons, the Tmax of BD occurred rapidly, at all doses (111,112).

GHB Overdose

Overdose of GHB can lead to serious adverse effects such as nausea, sedation, dizziness, seizure, respiratory depression, hypothermia, coma, and death (114). There are numerous reports in the clinic of GHB-related fatality among drug abusers. Currently, there is no antidote for the treatment of GHB overdose and treatment is limited to supportive care including mechanical ventilation to overcome respiratory depression observed in cases of overdose. Physostigmine, naloxone, and activated charcoal have been tried as antidotes but demonstrated little to no effect (98,115,116). Therefore, there is an urgent need to develop potential treatment strategies for the treatment of GHB overdose, alone and when it is co-ingested with other common club drugs.

Additionally, there is the potential for enhanced toxicity when GHB is administered with other drugs of abuse. In a recreational setting, GHB and GBL are typically not ingested alone and alcohol co-ingestion occurs in the majority of cases (115,117). In rats, GHB induced a decrease in respiratory frequency with a compensatory increase in tidal volume; co-administration of ethanol partially prevented the compensatory tidal volume increase and resulted in increased rate of fatality (39). Co-ingestion of ketamine and opiates has also been documented with GHB abuse (1,7). Ketamine can have respiratory depressant effects similar to GHB and has been reported to be co-ingested with GHB by 30% of users (6). Availability of ketamine may increase, since the therapeutic use of ketamine is increasing with its recent approval as an anti-depressant. Opioids including oxycodone and fentanyl are μ-opioid receptor agonists that can cause similar toxicities as GHB including respiratory depression, coma, and death (118120).

Although the relative contribution of MCT1 and SMCT1 to the renal reabsorption of GHB has not been determined in vivo, it has been demonstrated that inhibition of MCTs can increase GHB CLR, alter brain penetration, and reverse the effects of GHB on sleep time (measured by return of righting reflex), respiratory depression, and lethality (26,64,66,94,98). Animal studies with MCT inhibitors L-lactate and the flavonoid luteolin demonstrated significant increases in the renal and total clearances of GHB following its administration at high doses (104,107). Treatment with L-lactate also resulted in a significant decrease in the sedative/hypnotic effects of GHB and an improvement in GHB-induced respiratory depression in rats (26). A proof of concept study in humans utilized an infusion of the MCT1 inhibitor, L-lactate, and demonstrated a significant increase in GHB renal excretion (Table I) (99). Due to the minimal contribution of renal clearance at therapeutic doses assessed, the increase in renal clearance did not result in increased total clearance. This effect was similar to that observed at low oral doses in rats; however, the data in rats suggest that at higher doses, more relevant to overdose, the increase in renal clearance does translate into increased total clearance of GHB. The similarities between human and rat pharmacokinetics suggest that at high doses in humans, increased renal clearance with MCT inhibition should result in increased total clearance of GHB from the body. Specifically for L-lactate, the increase in renal clearance in humans and rats required a very high dose, which may limit the use of this MCT inhibitor for overdose treatment; however, the data support MCT inhibition as an overdose strategy in humans.

In addition to the inhibition of active renal reabsorption of GHB in the kidney, MCT inhibition may play an important role in blocking the entry of GHB into the brain, which is its site of action (64,65,74). Recent studies using in vivo microdialysis have demonstrated that L-lactate administration as a bolus followed by a continuous intravenous infusion to rats treated with GHB resulted in a decrease in plasma as well as frontal cortex ECF concentrations when compared to GHB alone (64). The reduction in plasma and frontal cortex ECF GHB concentrations was greater with a higher dose of lactate (64). This higher lactate dose also significantly reduced the unbound GHB brain ECF to plasma partition coefficient, whereas no such change was observed with lower lactate doses. These data suggest that L-lactate at higher doses can alter the BBB transport of GHB and represents a potential treatment strategy for GHB overdose (64). The Km value for GHB uptake has been shown to be higher at pH 7.4 when compared to pH 6.5 in red blood cells (87).

As the physiologically relevant pH at the BBB is 7.4, higher concentrations of L-lactate may be required to inhibit MCT-mediated transport of GHB across the BBB, compared with the intestine or kidneys where a lower physiological pH represents a driving force for absorption or reabsorption, respectively. Consistent with the reduction in plasma and brain ECF concentrations of GHB, L-lactate also significantly reduced GHB-induced sleep time measured as difference between return and loss of righting reflex (64). L-lactate was also able to inhibit GHB uptake into RBE4 cells in vitro at pH 7.4 at concentrations of 5 and 10 mM (64). The renal clearance of GHB was also increased by L-lactate administration due to inhibition of MCT-mediated active reabsorption in the proximal tubule of kidney, as demonstrated previously. These results together suggest that the transport of GHB across the BBB is mediated by MCTs. Since MCT1 is the predominant transporter expressed in the BBB, it is most likely responsible for the observed effects.

Subsequent pre-clinical work investigated more potent MCT1 inhibitors, AR-C15585 and AZD3965. Both compounds were identified by AstraZeneca, in an effort to synthesize potent and selective MCT1 inhibitors as immunosuppressants and as anti-cancer agents (121,122). AR-C155858 and AZD3965 are both highly potent MCT1 inhibitors with Ki values of 2.3 and 1.6 nM, respectively (123,124). AZD3965 exhibits 6-fold selectivity for MCT1 over MCT2 and does not have activity against MCT3 or MCT4 (122). Similarly, AR-C155858 is 4-fold more selective for MCT1 than MCT2 and does not inhibit MCT4 (123). In rats, treatment with AR-C155858 5 min after GHB administration prevented the respiratory depressant effects, reduced plasma exposure, and increased CLR of an intravenous dose of GHB (Table I and Fig. 7) (95). A similar effect was observed with AR-C155858 treatment following an oral dose of GHB. When AR-C155858 was administered 5 or 60 min after GHB, there was a decrease in exposure mediated by an increase in CLR (95). AZD3965 administration 60 min after IV or oral GHB administration results in decreased respiratory depression (125). Additionally, brain concentrations of GHB after AZD3965 administration were significantly decreased in rats, suggesting that a major effect of MCT1 inhibition is due to decreased uptake of GHB into the brain (125). The ability of AZD3965 to impact GHB TK even when the administration was delayed indicated that MCT1 inhibition with a potent inhibitor was a promising strategy for GHB overdose treatment.

Fig. 7.

Fig. 7.

Effect of increased clearance due to MCT inhibition by AR-C155858 onGHB-induced respiratory depression. Dose of GHB is 1500mg/kg IV AR-C155858was administered 5min after GHB. Figure adapted from (95). Extent of increaserenal and total clearance by AR-C155858 shown in Table I. Data arepresented as mean±S.D.; n=4 to 6

Addiction to GHB

Chronic treatment of drugs associated with addiction causes an increase in the neuronal activity of the mesocorticolimbic dopamine system. The actions of GHB on dopamine (DA) are mediated predominantly via the low-affinity binding of GHB to the GABAB receptors in the mesocorticolimbic DA system, and the addictive properties of GHB may represent the result of differential actions of GHB on DA and GABAB neurons. GHB is readily self-administered by rats and mice, and early drug discrimination studies conducted in rats indicated that GHB and its prodrugs GBL and BD are addictive GABAB agonists (126,127). Animals were clearly able to distinguish ethanol, baclofen, or diazepam from GHB and did not substitute them for GHB, suggesting differences in receptors involved in the reinforcing/addictive properties of these drugs (128,129). Rats that were chronically administered GHB showed tolerance after 3–6 days of daily administration and withdrawal symptoms (130,131). GHB and its prodrugs GBL and BD can also maintain self-administration in baboons, indicating the potential for physical dependence (37,132134). Administration of a GABAB antagonist results in withdrawal symptoms including tremors, vomiting, jerks, increased aggression, and increased duration to complete fine-motor tasks. Peak spontaneous withdrawal symptoms occur within 6–72 h following chronic administration of GHB or GBL. This is similar to that reported in humans following chronic GHB or GBL use (135).

Dependence liability of GHB was initially considered to be low for sodium oxybate (< 1%), and initial studies after clinical administration of GHB in patients with narcolepsy have not shown any cases of misuse or tolerance. However, further studies have indicated the potential for the drug to be mentally and physically addictive. Regular GHB use may result in dependence in weeks, and GHB withdrawal is known to cause autonomic dysfunction with severe CNS symptoms. Abrupt withdrawal can produce a range of neurological symptoms including tremor, anxiety attacks, confusion, seizures, and memory loss. These initial symptoms may progress to severe delirium with auditory and visual hallucinations and cardiovascular effects including tachycardia and hypertension (136). The withdrawal syndrome of GHB, GBL, or BD closely resembles that of other sedative-hypnotic agents (136). Brunt et al. (2014) (135) summarized the withdrawal symptoms from published studies; these consisted of tremor (67%), hallucinations (63%), tachycardia (63%), insomnia (58%), anxiety (46%), hypertension (44%), seizures (7%), and rhabdomyolysis (7%), plus one death. Additionally, little is known about treatment effectiveness for GHB-addicted subjects. Benzodiazepines, barbiturates, and antipsychotic medication have been used for treatment of withdrawal symptoms, and in such severe cases of GHB withdrawal, treatment with high doses of predominantly benzodiazepines is generally used. However, in many cases, these treatment regimens have proven ineffective. In the “Dutch GHB Monitor” study, 274 patients with GHB dependence were followed during treatment for GHB detoxification over the period of 2010 to 2012. After 3 months of follow-up, 65% of the patients in this study reported a relapse in GHB abuse (137). A newer treatment regimen in the Netherlands follows the recommended medication-assisted treatment for opioid addiction and treats GHB withdrawal with pharmaceutical GHB (Xyrem) with tapering of doses over time.

CONCLUSIONS

GHB is used therapeutically for the treatment of narcolepsy and for withdrawal symptoms in alcoholics. However, since the 1990s, there have been reports of GHB abuse in recreational settings and GHB has been referred to as a “date-rape drug.” One of the probable reasons for the abuse of GHB and, therefore, associated problems with toxicity and addiction is the relative ease to manufacture GHB and easy accessibility of its precursors. While physiological effects of GHB are due to binding to GHB receptors in the brain, pharmacological and toxicological effects of GHB are predominantly due to binding to GABAB receptors, effects that can be reversed by GABAB receptor antagonists. The pharmacokinetics of GHB are complex and involve capacity-limited absorption, metabolism, tissue distribution, and renal elimination. Monocarboxylate transporters represent major determinants of absorption, renal clearance, and tissue uptake, including uptake into the brain, its site of action. Toxicity and death after GHB overdoses in pre-clinical studies can be reversed by MCT1 inhibitors, indicating their potential use for the treatment of clinical GHB overdoses. Clinical studies, based on scale-up from physiologically based pharmacokinetics and pharmacodynamics models, are needed to implement MCT1 inhibition for the treatment of overdoses.

FUNDING

The studies from the Morris laboratory were supported in part by NIH grant R01DA023223. MAF is supported in part by NIH grant SC1 DA-052150.

Footnotes

Conflict of Interest The authors declare that they have no conflict of interest.

REFERENCES

  • 1.White CM. Pharmacologic, pharmacokinetic, and clinical assessment of illicitly used gamma-hydroxybutyrate. J Clin Pharmacol. 2017;57(1):33–9. [DOI] [PubMed] [Google Scholar]
  • 2.Laborit H. Sodium 4-hydroxybutyrate. Int J Neuropharmacol. 1964;3:433–51. [DOI] [PubMed] [Google Scholar]
  • 3.Zhang Y, Huo M, Zhou J, Xie S. PKSolver: an add-in program for pharmacokinetic and pharmacodynamic data analysis in Microsoft Excel. Comput Methods Prog Biomed. 2010;99(3):306–14. [DOI] [PubMed] [Google Scholar]
  • 4.Carter LP, Pardi D, Gorsline J, Griffiths RR. Illicit gamma-hydroxybutyrate (GHB) and pharmaceutical sodium oxybate (Xyrem): differences in characteristics and misuse. Drug Alcohol Depend. 2009;104(1–2):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.van den Brink W, Addolorato G, Aubin HJ, Benyamina A, Caputo F, Dematteis M, et al. Efficacy and safety of sodium oxybate in alcohol-dependent patients with a very high drinking risk level. Addict Biol. 2018;23(4):969–86. [DOI] [PubMed] [Google Scholar]
  • 6.Kim SY, Anderson IB, Dyer JE, Barker JC, Blanc PD. High-risk behaviors and hospitalizations among gamma hydroxybutyrate (GHB) users. Am J Drug Alcohol Abuse. 2007;33(3):429–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Liakoni E, Walther F, Nickel CH, Liechti ME. Presentations to an urban emergency department in Switzerland due to acute gamma-hydroxybutyrate toxicity. Scand J Trauma Resusc Emerg Med. 2016;24(1):107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Britt GC, McCance-Katz EF. A brief overview of the clinical pharmacology of “club drugs”. Subst Use Misuse. 2005;40(9–10):1189–201. [DOI] [PubMed] [Google Scholar]
  • 9.WHO. Gamma-hydroxybutyric acid (GHB) critical review report. Hammamet, Tunisia; 2012. [Google Scholar]
  • 10.Busardo FP, Gottardi M, Tini A, Minutillo A, Sirignano A, Marinelli E, et al. Replacing GHB with GBL in recreational settings: a new trend in chemsex. Curr Drug Metab. 2018;19(13):1080–5. [DOI] [PubMed] [Google Scholar]
  • 11.Griffiths RR, Johnson MW. Relative abuse liability of hypnotic drugs: a conceptual framework and algorithm for differentiating among compounds. J Clin Psychiatry. 2005;66(Suppl 9):31–41. [PubMed] [Google Scholar]
  • 12.Corkery JM, Loi B, Claridge H, Goodair C, Schifano F. Deaths in the lesbian, gay, bisexual and transgender United Kingdom communities associated with GHB and precursors. Curr Drug Metab. 2018;19(13):1086–99. [DOI] [PubMed] [Google Scholar]
  • 13.Hockenhull J, Murphy KG, Paterson S. An observed rise in gamma-hydroxybutyrate-associated deaths in London: evidence to suggest a possible link with concomitant rise in chemsex. Forensic Sci Int. 2017;270:93–7. [DOI] [PubMed] [Google Scholar]
  • 14.Moresco M, Pizza F, Antelmi E, Plazzi G. Sodium oxybate treatment in pediatric type 1 narcolepsy. Curr Drug Metab. 2018;19:1073–9. [DOI] [PubMed] [Google Scholar]
  • 15.Hammoud MA, Bourne A, Maher L, Jin F, Haire B, Lea T, et al. Intensive sex partying with gamma-hydroxybutyrate: factors associated with using gamma-hydroxybutyrate for chemsex among Australian gay and bisexual men-results from the Flux Study. Sex Health. 2018;15(2):123–34. [DOI] [PubMed] [Google Scholar]
  • 16.Schecke H, Lea T, Bohn A, Kohler T, Sander D, Scherbaum N, et al. Crystal methamphetamine use in sexual settings among German men who have sex with men. Front Psychiatry. 2019;10:886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bay T, Eghorn LF, Klein AB, Wellendorph P. GHB receptor targets in the CNS: focus on high-affinity binding sites. Biochem Pharmacol. 2014;87(2):220–8. [DOI] [PubMed] [Google Scholar]
  • 18.Absalom N, Eghorn LF, Villumsen IS, Karim N, Bay T, Olsen JV, et al. Alpha4betadelta GABA(A) receptors are high-affinity targets for gamma-hydroxybutyric acid (GHB). Proc Natl Acad Sci U S A. 2012;109(33):13404–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Carai MA, Colombo G, Brunetti G, Melis S, Serra S, Vacca G, et al. Role of GABA(B) receptors in the sedative/hypnotic effect of gamma-hydroxybutyric acid. Eur J Pharmacol. 2001;428(3):315–21. [DOI] [PubMed] [Google Scholar]
  • 20.Cash CD. Gamma-hydroxybutyrate: an overview of the pros and cons for it being a neurotransmitter and/or a useful therapeutic agent. Neurosci Biobehav Rev. 1994;18(2):291– 304. [DOI] [PubMed] [Google Scholar]
  • 21.Maitre M, Klein C, Mensah-Nyagan AG. Mechanisms for the specific properties of gamma-hydroxybutyrate in brain. Med Res Rev. 2016;36(3):363–88. [DOI] [PubMed] [Google Scholar]
  • 22.Gobaille S, Hechler V, Andriamampandry C, Kemmel V, Maitre M. Gamma-hydroxybutyrate modulates synthesis and extracellular concentration of gamma-aminobutyric acid in discrete rat brain regions in vivo. J Pharmacol Exp Ther. 1999;290(1):303–9. [PubMed] [Google Scholar]
  • 23.Castelli MP, Ferraro L, Mocci I, Carta F, Carai MA, Antonelli T, et al. Selective gamma-hydroxybutyric acid receptor ligands increase extracellular glutamate in the hippocampus, but fail to activate G protein and to produce the sedative/hypnotic effect of gamma-hydroxybutyric acid. J Neurochem. 2003;87(3):722–32. [DOI] [PubMed] [Google Scholar]
  • 24.Hechler V, Gobaille S, Bourguignon JJ, Maitre M. Extracellular events induced by gamma-hydroxybutyrate in striatum: a microdialysis study. J Neurochem. 1991;56(3):938–44. [DOI] [PubMed] [Google Scholar]
  • 25.Kaupmann K, Cryan JF, Wellendorph P, Mombereau C, Sansig G, Klebs K, et al. Specific gamma-hydroxybutyrate-binding sites but loss of pharmacological effects of gamma-hydroxybutyrate in GABA(B)(1)-deficient mice. Eur J Neurosci. 2003;18(10):2722–30. [DOI] [PubMed] [Google Scholar]
  • 26.Morse BL, Vijay N, Morris ME. Gamma-hydroxybutyrate (GHB)-induced respiratory depression: combined receptor-transporter inhibition therapy for treatment in GHB overdose. Mol Pharmacol. 2012;82(2):226–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Carai MA, Lobina C, Maccioni P, Cabras C, Colombo G, Gessa GL. Gamma-aminobutyric acidB (GABAB)-receptor mediation of different in vivo effects of gamma-butyrolactone. J Pharmacol Sci. 2008;106(2):199–207. [DOI] [PubMed] [Google Scholar]
  • 28.Dornbierer DA, Baur DM, Stucky B, Quednow BB, Kraemer T, Seifritz E, et al. Neurophysiological signature of gamma-hydroxybutyrate augmented sleep in male healthy volunteers may reflect biomimetic sleep enhancement: a randomized controlled trial. Neuropsychopharmacology. 2019;44(11):1985–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ito Y, Ishige K, Zaitsu E, Anzai K, Fukuda H. Gamma-hydroxybutyric acid increases intracellular Ca2+ concentration and nuclear cyclic AMP-responsive element- and activator protein 1 DNA-binding activities through GABAB receptor in cultured cerebellar granule cells. J Neurochem. 1995;65(1):75–83. [DOI] [PubMed] [Google Scholar]
  • 30.Mathivet P, Bernasconi R, De Barry J, Marescaux C, Bittiger H. Binding characteristics of gamma-hydroxybutyric acid as a weak but selective GABAB receptor agonist. Eur J Pharmacol. 1997;321(1):67–75. [DOI] [PubMed] [Google Scholar]
  • 31.Ishige K, Aizawa M, Ito Y, Fukuda H. Gamma-butyrolactone-induced absence-like seizures increase nuclear CRE- and AP-1 DNA-binding activities in mouse brain. Neuropharmacology. 1996;35(1):45–55. [DOI] [PubMed] [Google Scholar]
  • 32.Bernasconi R, Lauber J, Marescaux C, Vergnes M, Martin P, Rubio V, et al. Experimental absence seizures: potential role of gamma-hydroxybutyric acid and GABAB receptors. J Neural Transm Suppl. 1992;35:155–77. [DOI] [PubMed] [Google Scholar]
  • 33.Jacobson LH, Vlachou S, Slattery DA, Li X, Cryan JF. The gamma-aminobutyric acid B receptor in depression and reward. Biol Psychiatry. 2018;83(11):963–76. [DOI] [PubMed] [Google Scholar]
  • 34.Bolser DC, Blythin DJ, Chapman RW, Egan RW, Hey JA, Rizzo C, et al. The pharmacology of SCH 50911: a novel, orally-active GABA-beta receptor antagonist. J Pharmacol Exp Ther. 1995;274(3):1393–8. [PubMed] [Google Scholar]
  • 35.Lingenhoehl K, Brom R, Heid J, Beck P, Froestl W, Kaupmann K, et al. Gamma-hydroxybutyrate is a weak agonist at recombinant GABA(B) receptors. Neuropharmacology. 1999;38(11):1667–73. [DOI] [PubMed] [Google Scholar]
  • 36.Froestl W. Chemistry and pharmacology of GABAB receptor ligands. Adv Pharmacol. 2010;58:19–62. [DOI] [PubMed] [Google Scholar]
  • 37.Goodwin AK, Griffiths RR, Brown PR, Froestl W, Jakobs C, Gibson KM, et al. Chronic intragastric administration of gamma-butyrolactone produces physical dependence in baboons. Psychopharmacology. 2006;189(1):71–82. [DOI] [PubMed] [Google Scholar]
  • 38.Goodwin AK, Froestl W, Weerts EM. Involvement of gamma-hydroxybutyrate (GHB) and GABA-B receptors in the acute behavioral effects of GHB in baboons. Psychopharmacology. 2005;180(2):342–51. [DOI] [PubMed] [Google Scholar]
  • 39.Morse BL, Morris ME. Toxicokinetics/toxicodynamics of gamma-hydroxybutyrate-ethanol intoxication: evaluation of potential treatment strategies. J Pharmacol Exp Ther. 2013;346(3):504–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hodor A, Palchykova S, Gao B, Bassetti CL. Baclofen and gamma-hydroxybutyrate differentially altered behavior, EEG activity and sleep in rats. Neuroscience. 2015;284:18–28. [DOI] [PubMed] [Google Scholar]
  • 41.Koek W, France CP. Cataleptic effects of gamma-hydroxybutyrate (GHB) and baclofen in mice: mediation by GABA(B) receptors, but differential enhancement by N-methyl-d-aspartate (NMDA) receptor antagonists. Psychopharmacology 2008;199(2):191–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Koek W, Chen W, Mercer SL, Coop A, France CP. Discriminative stimulus effects of gamma-hydroxybutyrate: role of training dose. J Pharmacol Exp Ther. 2006;317(1):409–17. [DOI] [PubMed] [Google Scholar]
  • 43.Gold BI, Roth RH. Kinetics of in vivo conversion of gamma-[3H]aminobutyric acid to gamma-[3H]hydroxybutyric acid by rat brain. J Neurochem. 1977;28(5):1069–73. [DOI] [PubMed] [Google Scholar]
  • 44.Brenneisen R, Elsohly MA, Murphy TP, Passarelli J, Russmann S, Salamone SJ, et al. Pharmacokinetics and excretion of gamma-hydroxybutyrate (GHB) in healthy subjects. J Anal Toxicol 2004;28(8):625–30. [DOI] [PubMed] [Google Scholar]
  • 45.Roth RH, Giarman NJ. Gamma-butyrolactone and gamma-hydroxybutyric acid I: distribution and metabolism. Biochem Pharmacol. 1966;15:1333–48. [DOI] [PubMed] [Google Scholar]
  • 46.Wong CG, Gibson KM, Snead OC 3rd. From the street to the brain: neurobiology of the recreational drug gamma-hydroxybutyric acid. Trends Pharmacol Sci. 2004;25(1):29–34. [DOI] [PubMed] [Google Scholar]
  • 47.Struys EA, Verhoeven NM, Jansen EE, Ten Brink HJ, Gupta M, Burlingame TG, et al. Metabolism of gamma-hydroxybutyrate to d-2-hydroxyglutarate in mammals: further evidence for d-2-hydroxyglutarate transhydrogenase. Metabolism. 2006;55(3):353–8. [DOI] [PubMed] [Google Scholar]
  • 48.Liakoni E, Gugelmann H, Dempsey DA, Wiegand TJ, Havel C, Jacob P, et al. Butanediol conversion to gamma-hydroxybutyrate markedly reduced by the alcohol dehydrogenase blocker fomepizole. Clin Pharmacol Ther. 2019;105(5):1196–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Vayer P, Mandel P, Maitre M. Conversion of gamma-hydroxybutyrate to gamma-aminobutyrate in vitro. J Neurochem. 1985;45(3):810–4. [DOI] [PubMed] [Google Scholar]
  • 50.Snead OC 3rd, Furner R, Liu CC. In vivo conversion of gamma-aminobutyric acid and 1,4-butanediol to gamma-hydroxybutyric acid in rat brain. Studies using stable isotopes. Biochem Pharmacol. 1989;38(24):4375–80. [DOI] [PubMed] [Google Scholar]
  • 51.Kaufman EE, Nelson T. An overview of gamma-hydroxybutyrate catabolism: the role of the cytosolic NADP(+)-dependent oxidoreductase EC 1.1.1.19 and of a mitochondrial hydroxyacid-oxoacid transhydrogenase in the initial, rate-limiting step in this pathway. Neurochem Res. 1991;16(9):965–74. [DOI] [PubMed] [Google Scholar]
  • 52.Alzeer S, Ellis EM. Metabolism of gamma hydroxybutyrate in human hepatoma HepG2 cells by the aldo-keto reductase AKR1A1. Biochem Pharmacol. 2014;92(3):499–505. [DOI] [PubMed] [Google Scholar]
  • 53.Bhattacharya I, Boje KM. Feasibility of D-glucuronate to enhance gamma-hydroxybutyric acid metabolism during gamma-hydroxybutyric acid toxicity: pharmacokinetic and pharmacodynamic studies. Biopharm Drug Dispos. 2007;28(1):1–11. [DOI] [PubMed] [Google Scholar]
  • 54.Knerr I, Pearl PL, Bottiglieri T, Snead OC, Jakobs C, Gibson KM. Therapeutic concepts in succinate semialdehyde dehydrogenase (SSADH; ALDH5a1) deficiency (gamma-hydroxybutyric aciduria). Hypotheses evolved from 25 years of patient evaluation, studies in Aldh5a1−/− mice and characterization of gamma-hydroxybutyric acid pharmacology. J Inherit Metab Dis. 2007;30(3):279–94. [DOI] [PubMed] [Google Scholar]
  • 55.Gibson KM, Nyhan WL. Metabolism of [U-14C]-4-hydroxybutyric acid to intermediates of the tricarboxylic acid cycle in extracts of rat liver and kidney mitochondria. Eur J Drug Metab Pharmacokinet. 1989;14(1):61–70. [DOI] [PubMed] [Google Scholar]
  • 56.Pearl PL, Gibson KM, Cortez MA, Wu Y, Carter Snead O 3rd, Knerr I, et al. Succinic semialdehyde dehydrogenase deficiency: lessons from mice and men. J Inherit Metab Dis. 2009;32(3):343–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hechler V, Ratomponirina C, Maitre M. Gamma-hydroxybutyrate conversion into GABA induces displacement of GABAB binding that is blocked by valproate and ethosuximide. J Pharmacol Exp Ther. 1997;281(2):753–60. [PubMed] [Google Scholar]
  • 58.Snead OC 3rd, Bearden LJ, Pegram V. Effect of acute and chronic anticonvulsant administration on endogenous gamma-hydroxybutyrate in rat brain. Neuropharmacology. 1980;19(1):47–52. [DOI] [PubMed] [Google Scholar]
  • 59.Brown GK, Cromby CH, Manning NJ, Pollitt RJ. Urinary organic acids in succinic semialdehyde dehydrogenase deficiency: evidence of alpha-oxidation of 4-hydroxybutyric acid, interaction of succinic semialdehyde with pyruvate dehydrogenase and possible secondary inhibition of mitochondrial beta-oxidation. J Inherit Metab Dis. 1987;10(4):367–75. [DOI] [PubMed] [Google Scholar]
  • 60.Lee CR. Evidence for the beta-oxidation of orally administered 4-hydroxybutyrate in humans. Biochem Med. 1977;17(3):284–91. [DOI] [PubMed] [Google Scholar]
  • 61.Gibson KM, Goodman SI, Frerman FE, Glasgow AM. Succinic semialdehyde dehydrogenase deficiency associated with combined 4-hydroxybutyric and dicarboxylic acidurias: potential for clinical misdiagnosis based on urinary organic acid profiling. J Pediatr. 1989;114(4 Pt 1):607–10. [DOI] [PubMed] [Google Scholar]
  • 62.O’Connor T, Ireland LS, Harrison DJ, Hayes JD. Major differences exist in the function and tissue-specific expression of human aflatoxin B1 aldehyde reductase and the principal human aldo-keto reductase AKR1 family members. Biochem J. 1999;343(Pt 2):487–504. [PMC free article] [PubMed] [Google Scholar]
  • 63.Morris ME, Felmlee MA. Overview of the proton-coupled MCT (SLC16A) family of transporters: characterization, function and role in the transport of the drug of abuse gamma-hydroxybutyric acid. AAPS J. 2008;10(2):311–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Roiko SA, Vijay N, Felmlee MA, Morris ME. Brain extracellular gamma-hydroxybutyrate concentrations are decreased by L-lactate in rats: role in the treatment of overdoses. Pharm Res. 2013;30(5):1338–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Bhattacharya I, Boje KM. GHB (gamma-hydroxybutyrate) carrier-mediated transport across the blood-brain barrier. J Pharmacol Exp Ther. 2004;311(1):92–8. [DOI] [PubMed] [Google Scholar]
  • 66.Felmlee MA, Morse BL, Follman KE, Morris ME. The drug of abuse gamma-hydroxybutyric acid exhibits tissue-specific nonlinear distribution. AAPS J. 2017;20(1):21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Morse BL, Vijay N, Morris ME. Mechanistic modeling of monocarboxylate transporter-mediated toxicokinetic/toxicodynamic interactions between gamma-hydroxybutyrate and L-lactate. AAPS J. 2014;16(4):756–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wood DM, Greene SL, Dargan PI. Five-year trends in self-reported recreational drugs associated with presentation to a UK emergency department with suspected drug-related toxicity. Eur J Emerg Med : official journal of the European Society for Emergency Medicine. 2013;20(4):263–7. [DOI] [PubMed] [Google Scholar]
  • 69.Jackson VN, Halestrap AP. The kinetics, substrate, and inhibitor specificity of the monocarboxylate (lactate) transporter of rat liver cells determined using the fluorescent intracellular pH indicator, 2′,7′-bis(carboxyethyl)-5(6)-carboxyfluorescein. J Biol Chem. 1996;271(2):861–8. [DOI] [PubMed] [Google Scholar]
  • 70.Felmlee MA, Jones RS, Rodriguez-Cruz V, Follman KE, Morris ME. Monocarboxylate transporters (SLC16): function, regulation, and role in health and disease. Pharmacol Rev. 2020;72(2):466–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Morris ME, Rodriguez-Cruz V, Felmlee MA. SLC and ABC transporters: expression, localization, and species differences at the blood-brain and the blood-cerebrospinal fluid barriers. AAPS J. 2017;19:1317–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wang Q, Darling IM, Morris ME. Transport of gamma-hydroxybutyrate in rat kidney membrane vesicles: role of monocarboxylate transporters. J Pharmacol Exp Ther. 2006;318(2):751–61. [DOI] [PubMed] [Google Scholar]
  • 73.Gerhart DZ, Enerson BE, Zhdankina OY, Leino RL, Drewes LR. Expression of monocarboxylate transporter MCT1 by brain endothelium and glia in adult and suckling rats. Am J Phys. 1997;273(1 Pt 1):E207–13. [DOI] [PubMed] [Google Scholar]
  • 74.Vijay N, Morris ME. Role of monocarboxylate transporters in drug delivery to the brain. Curr Pharm Des. 2013;20:1487–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Otsuka Y, Furihata T, Nakagawa K, Ohno Y, Reien Y, Ouchi M, et al. Sodium-coupled monocarboxylate transporter 1 interacts with the RING finger- and PDZ domain-containing protein PDZRN3. J Physiol Sci. 2019;69(4):635–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Gopal E, Miyauchi S, Martin PM, Ananth S, Roon P, Smith SB, et al. Transport of nicotinate and structurally related compounds by human SMCT1 (SLC5A8) and its relevance to drug transport in the mammalian intestinal tract. Pharm Res. 2007;24(3):575–84. [DOI] [PubMed] [Google Scholar]
  • 77.Martin PM, Dun Y, Mysona B, Ananth S, Roon P, Smith SB, et al. Expression of the sodium-coupled monocarboxylate transporters SMCT1 (SLC5A8) and SMCT2 (SLC5A12) in retina. Invest Ophthalmol Vis Sci. 2007;48(7):3356–63. [DOI] [PubMed] [Google Scholar]
  • 78.Martin PM, Gopal E, Ananth S, Zhuang L, Itagaki S, Prasad BM, et al. Identity of SMCT1 (SLC5A8) as a neuron-specific Na+-coupled transporter for active uptake of L-lactate and ketone bodies in the brain. J Neurochem. 2006;98(1):279–88. [DOI] [PubMed] [Google Scholar]
  • 79.Gopal E, Fei YJ, Sugawara M, Miyauchi S, Zhuang L, Martin P, et al. Expression of slc5a8 in kidney and its role in Na(+)-coupled transport of lactate. J Biol Chem. 2004;279(43):44522–32. [DOI] [PubMed] [Google Scholar]
  • 80.Rodriguez AM, Perron B, Lacroix L, Caillou B, Leblanc G, Schlumberger M, et al. Identification and characterization of a putative human iodide transporter located at the apical membrane of thyrocytes. J Clin Endocrinol Metab. 2002;87(7):3500–3. [DOI] [PubMed] [Google Scholar]
  • 81.Iwanaga T, Kishimoto A. Cellular distributions of monocarboxylate transporters: a review. Biomed Res. 2015;36(5):279– 301. [DOI] [PubMed] [Google Scholar]
  • 82.Barac-Nieto M, Murer H, Kinne R. Lactate-sodium cotransport in rat renal brush border membranes. Am J Phys. 1980;239(5):F496–506. [DOI] [PubMed] [Google Scholar]
  • 83.Ganapathy V, Thangaraju M, Gopal E, Martin PM, Itagaki S, Miyauchi S, et al. Sodium-coupled monocarboxylate transporters in normal tissues and in cancer. AAPS J. 2008;10(1):193–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Cui D, Morris ME. The drug of abuse gamma-hydroxybutyrate is a substrate for sodium-coupled monocarboxylate transporter (SMCT) 1 (SLC5A8): characterization of SMCT-mediated uptake and inhibition. Drug Metab Dispos. 2009;37(7):1404–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wang Q, Lu Y, Morris ME. Monocarboxylate transporter (MCT) mediates the transport of gamma-hydroxybutyrate in human kidney HK-2 cells. Pharm Res. 2007;24(6):1067–78. [DOI] [PubMed] [Google Scholar]
  • 86.Lam WK, Felmlee MA, Morris ME. Monocarboxylate transporter-mediated transport of gamma-hydroxybutyric acid in human intestinal Caco-2 cells. Drug Metab Dispos. 2010;38(3):441–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Morse BL, Felmlee MA, Morris ME. Gamma-hydroxybutyrate blood/plasma partitioning: effect of physiologic pH on transport by monocarboxylate transporters. Drug Metab Dispos. 2012;40(1):64–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Smith JP, Drewes LR. Modulation of monocarboxylic acid transporter-1 kinetic function by the cAMP signaling pathway in rat brain endothelial cells. J Biol Chem. 2006;281(4):2053–60. [DOI] [PubMed] [Google Scholar]
  • 89.Carl SM, Lindley DJ, Das D, Couraud PO, Weksler BB, Romero I, et al. ABC and SLC transporter expression and proton oligopeptide transporter (POT) mediated permeation across the human blood–brain barrier cell line, hCMEC/D3 [corrected]. Mol Pharm. 2010;7(4):1057–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Roiko SA, Felmlee MA, Morris ME. Brain uptake of the drug of abuse gamma-hydroxybutyric acid in rats. Drug Metab Dispos. 2012;40(1):212–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Scharf MB, Lai AA, Branigan B, Stover R, Berkowitz DB. Pharmacokinetics of gamma-hydroxybutyrate (GHB) in narcoleptic patients. Sleep. 1998;21(5):507–14. [DOI] [PubMed] [Google Scholar]
  • 92.Palatini P, Tedeschi L, Frison G, Padrini R, Zordan R, Orlando R, et al. Dose-dependent absorption and elimination of gamma-hydroxybutyric acid in healthy volunteers. Eur J Clin Pharmacol. 1993;45(4):353–6. [DOI] [PubMed] [Google Scholar]
  • 93.Ferrara SD, Zotti S, Tedeschi L, Frison G, Castagna F, Gallimberti L, et al. Pharmacokinetics of gamma-hydroxybutyric acid in alcohol dependent patients after single and repeated oral doses. Br J Clin Pharmacol. 1992;34(3):231–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Morris ME, Hu K, Wang Q. Renal clearance of gamma-hydroxybutyric acid in rats: increasing renal elimination as a detoxification strategy. J Pharmacol Exp Ther. 2005;313(3):1194–202. [DOI] [PubMed] [Google Scholar]
  • 95.Vijay N, Morse BL, Morris ME. A novel monocarboxylate transporter inhibitor as a potential treatment strategy for gamma-hydroxybutyric acid overdose. Pharm Res. 2015;32(6):1894–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Lettieri JT, Fung HL. Dose-dependent pharmacokinetics and hypnotic effects of sodium gamma-hydroxybutyrate in the rat. J Pharmacol Exp Ther. 1979;208(1):7–11. [PubMed] [Google Scholar]
  • 97.Arena C, Fung HL. Absorption of sodium gamma-hydroxybutyrate and its prodrug gamma-butyrolactone: relationship between in vitro transport and in vivo absorption. J Pharm Sci. 1980;69(3):356–8. [DOI] [PubMed] [Google Scholar]
  • 98.Morse BL, Morris ME. Effects of monocarboxylate transporter inhibition on the oral toxicokinetics/toxicodynamics of gamma-hydroxybutyrate and gamma-butyrolactone. J Pharmacol Exp Ther. 2013;345(1):102–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Morris ME, Morse BL, Baciewicz GJ, Tessena MM, Acquisto NM, Hutchinson DJ, et al. Monocarboxylate transporter inhibition with osmotic diuresis increases gamma-hydroxybutyrate renal elimination in humans: a proof-of-concept study. J Clin Toxicol. 2011;1(2):1000105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Docherty JR, Green AR. The role of monoamines in the changes in body temperature induced by 3, 4 -methylenedioxymethamphetamine (MDMA, ecstasy) and its derivatives. Br J Pharmacol. 2010;160(5):1029–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Felmlee MA, Roiko SA, Morse BL, Morris ME. Concentration-effect relationships for the drug of abuse gamma-hydroxybutyric acid. J Pharmacol Exp Ther. 2010;333(3):764–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.FDA Xyrem Label 2020. [Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021196s033s034lbl.pdf. Accessed Sept 2020
  • 103.Felmlee MA, Wang Q, Cui D, Roiko SA, Morris ME. Mechanistic toxicokinetic model for gamma-hydroxybutyric acid: inhibition of active renal reabsorption as a potential therapeutic strategy. AAPS J. 2010;12(3):407–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Wang Q, Morris ME. Flavonoids modulate monocarboxylate transporter-1-mediated transport of gamma-hydroxybutyrate in vitro and in vivo. Drug Metab Dispos. 2007;35(2):201–8. [DOI] [PubMed] [Google Scholar]
  • 105.Raybon JJ, Boje KM. Pharmacokinetics and pharmacodynamics of gamma-hydroxybutyric acid during tolerance in rats: effects on extracellular dopamine. J Pharmacol Exp Ther. 2007;320(3):1252–60. [DOI] [PubMed] [Google Scholar]
  • 106.Lettieri J, Fung HL. Absorption and first-pass metabolism of 14C-gamma-hydroxybutyric acid. Res Commun Chem Pathol Pharmacol. 1976;13(3):425–37. [PubMed] [Google Scholar]
  • 107.Wang Q, Wang X, Morris ME. Effects of L-lactate and D-mannitol on gamma-hydroxybutyrate toxicokinetics and toxicodynamics in rats. Drug Metab Dispos.2008;36(11):2244–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Giarman NJ, Roth RH. Differential estimation of gamma-butyrolactone and gamma-hydroxybutyric acid in rat blood and brain. Science. 1964;145(3632):583–4. [DOI] [PubMed] [Google Scholar]
  • 109.Lettieri J, Fung HL. Improved pharmacological activity via pro-drug modification: comparative pharmacokinetics of sodium gamma-hydroxybutyrate and gamma-butyrolactone. Res Commun Chem Pathol Pharmacol. 1978;22(1):107–18. [PubMed] [Google Scholar]
  • 110.Carter LP, Koek W, France CP. Lack of effects of GHB precursors GBL and 1,4-BD following i.c.v. administration in rats. Eur J Neurosci. 2006;24(9):2595–600. [DOI] [PubMed] [Google Scholar]
  • 111.Fung HL, Tsou PS, Bulitta JB, Tran DC, Page NA, Soda D, et al. Pharmacokinetics of 1,4-butanediol in rats: bioactivation to gamma-hydroxybutyric acid, interaction with ethanol, and oral bioavailability. AAPS J. 2008;10(1):56–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Goodwin AK, Brown PR, Jansen EE, Jakobs C, Gibson KM, Weerts EM. Behavioral effects and pharmacokinetics of gamma-hydroxybutyrate (GHB) precursors gamma-butyrolactone (GBL) and 1,4-butanediol (1,4-BD) in baboons. Psychopharmacology. 2009;204(3):465–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Thai D, Dyer JE, Benowitz NL, Haller CA. Gamma-hydroxybutyrate and ethanol effects and interactions in humans. J Clin Psychopharmacol. 2006;26(5):524–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Okun MS, Boothby LA, Bartfield RB, Doering PL. GHB: an important pharmacologic and clinical update. J Pharm Pharm Sci : a publication of the Canadian Society for Pharmaceutical Sciences, Societe canadienne des sciences pharmaceutiques. 2001;4(2):167–75. [PubMed] [Google Scholar]
  • 115.Mason PE, Kerns WP 2nd. Gamma hydroxybutyric acid (GHB) intoxication. Acad Emerg Med Off J Soc Acad Emerg Med. 2002;9(7):730–9. [DOI] [PubMed] [Google Scholar]
  • 116.Bania TC, Chu J. Physostigmine does not effect arousal but produces toxicity in an animal model of severe gamma-hydroxybutyrate intoxication. Acad Emerg Med Off J Soc Acad Emerg Med. 2005;12(3):185–9. [DOI] [PubMed] [Google Scholar]
  • 117.Zvosec DL, Smith SW, Porrata T, Strobl AQ, Dyer JE. Case series of 226 gamma-hydroxybutyrate-associated deaths: lethal toxicity and trauma. Am J Emerg Med. 2011;29(3):319–32. [DOI] [PubMed] [Google Scholar]
  • 118.Berling I, Whyte IM, Isbister GK. Oxycodone overdose causes naloxone responsive coma and QT prolongation. QJM. 2013;106(1):35–41. [DOI] [PubMed] [Google Scholar]
  • 119.Fox LM, Hoffman RS, Vlahov D, Manini AF. Risk factors for severe respiratory depression from prescription opioid overdose. Addiction. 2018;113(1):59–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Marinetti LJ, Ehlers BJ. A series of forensic toxicology and drug seizure cases involving illicit fentanyl alone and in combination with heroin, cocaine or heroin and cocaine. J Anal Toxicol. 2014;38(8):592–8. [DOI] [PubMed] [Google Scholar]
  • 121.Pahlman C, Qi Z, Murray CM, Ferguson D, Bundick RV, Donald DK, et al. Immunosuppressive properties of a series of novel inhibitors of the monocarboxylate transporter MCT-1. Transpl Int. 2013;26(1):22–9. [DOI] [PubMed] [Google Scholar]
  • 122.Curtis NJ, Mooney L, Hopcroft L, Michopoulos F, Whalley N, Zhong H, et al. Pre-clinical pharmacology of AZD3965, a selective inhibitor of MCT1: DLBCL, NHL and Burkitt’s lymphoma anti-tumor activity. Oncotarget. 2017;8(41):69219– 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Ovens MJ, Davies AJ, Wilson MC, Murray CM, Halestrap AP. AR-C155858 is a potent inhibitor of monocarboxylate transporters MCT1 and MCT2 that binds to an intracellular site involving transmembrane helices 7–10. Biochem J. 2010;425(3):523–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Bola BM, Chadwick AL, Michopoulos F, Blount KG, Telfer BA, Williams KJ, et al. Inhibition of monocarboxylate transporter-1 (MCT1) by AZD3965 enhances radiosensitivity by reducing lactate transport. Mol Cancer Ther. 2014;13(12):2805–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Follman KE, Morris ME. Treatment of gamma-hydroxybutyric acid and gamma-butyrolactone overdose with two potent monocarboxylate transporter 1 inhibitors, AZD3965 and AR-C155858. J Pharmacol Exp Ther. 2019;370(1):84–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Lobina C, Agabio R, Reali R, Gessa GL, Colombo G. Contribution of GABA(A) and GABA(B) receptors to the discriminative stimulus produced by gamma-hydroxybutyric acid. Pharmacol Biochem Behav. 1999;64(2):363–5. [DOI] [PubMed] [Google Scholar]
  • 127.Baker LE, Van Tilburg TJ, Brandt AE, Poling A. Discriminative stimulus effects of gamma-hydroxybutyrate (GHB) and its metabolic precursor, gamma-butyrolactone (GBL) in rats. Psychopharmacology. 2005;181(3):458–66. [DOI] [PubMed] [Google Scholar]
  • 128.Baker LE, Searcy GD, Pynnonen DM, Poling A. Differentiating the discriminative stimulus effects of gamma-hydroxybutyrate and ethanol in a three-choice drug discrimination procedure in rats. Pharmacol Biochem Behav. 2008;89(4):598–607. [DOI] [PubMed] [Google Scholar]
  • 129.Lobina C, Colombo G, Gessa GL, Carai MA. Different sensitivity to the motor incoordinating effects of gamma-hydroxybutyric acid (GHB) and baclofen in GHB-sensitive and GHB-resistant rats. Brain Res. 2005;1033(1):109–12. [DOI] [PubMed] [Google Scholar]
  • 130.Carai MA, Quang LS, Atzeri S, Lobina C, Maccioni P, Orru A, et al. Withdrawal syndrome from gamma-hydroxybutyric acid (GHB) and 1,4-butanediol (1,4-BD) in Sardinian alcohol-preferring rats. Brain Res Brain Res Protoc. 2005;15(2):75–8. [DOI] [PubMed] [Google Scholar]
  • 131.Quang LS, Colombo G, Lobina C, Maccioni P, Orru A, Gessa GL, et al. Evaluation for the withdrawal syndrome from gamma-hydroxybutyric acid (GHB), gamma-butyrolactone (GBL), and 1,4-butanediol (1,4-BD) in different rat lines. Ann N Y Acad Sci. 2006;1074:545–58. [DOI] [PubMed] [Google Scholar]
  • 132.Goodwin AK, Gibson KM, Weerts EM. Physical dependence on gamma-hydroxybutrate (GHB) prodrug 1,4-butanediol (1,4-BD): time course and severity of withdrawal in baboons. Drug Alcohol Depend. 2013;132(3):427–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Goodwin AK, Kaminski BJ, Weerts EM. Self-administration of gamma-hydroxybutyric acid (GHB) precursors gamma-butyrolactone (GBL) and 1,4-butanediol (1,4-BD) in baboons. Psychopharmacology. 2013;225(3):637–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Weerts EM, Goodwin AK, Griffiths RR, Brown PR, Froestl W, Jakobs C, et al. Spontaneous and precipitated withdrawal after chronic intragastric administration of gamma-hydroxybutyrate (GHB) in baboons. Psychopharmacology. 2005;179(3):678–87. [DOI] [PubMed] [Google Scholar]
  • 135.Brunt TM, van Amsterdam JG, van den Brink W. GHB, GBL and 1,4-BD addiction. Curr Pharm Des. 2014;20(25):4076–85. [DOI] [PubMed] [Google Scholar]
  • 136.McDonough M, Kennedy N, Glasper A, Bearn J. Clinical features and management of gamma-hydroxybutyrate (GHB) withdrawal: a review. Drug Alcohol Depend. 2004;75(1):3–9. [DOI] [PubMed] [Google Scholar]
  • 137.van Noorden MS, Mol T, Wisselink J, Kuijpers W, Dijkstra BAG. Treatment consumption and treatment re-enrollment in GHB-dependent patients in The Netherlands. Drug Alcohol Depend. 2017;176:96–101. [DOI] [PubMed] [Google Scholar]

RESOURCES