Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
J Pharmacol Exp Ther ; 372(1): 1-10, 2020 01.
Article in English | MEDLINE | ID: mdl-31619465

ABSTRACT

Padsevonil is an antiepileptic drug (AED) candidate synthesized in a medicinal chemistry program initiated to rationally design compounds with high affinity for synaptic vesicle 2 (SV2) proteins and low-to-moderate affinity for the benzodiazepine binding site on GABAA receptors. The pharmacological profile of padsevonil was characterized in binding and electrophysiological experiments. At recombinant SV2 proteins, padsevonil's affinity for SV2A was greater than that of levetiracetam and brivaracetam (pKi 8.5, 5.2, and 6.6, respectively). Unlike the latter AEDs, both selective SV2A ligands, padsevonil also displayed high affinity for the SV2B and SV2C isoforms (pKi 7.9 and 8.5, respectively). Padsevonil's interaction with SV2A differed from that of levetiracetam and brivaracetam; it exhibited slower binding kinetics: dissociation t 1/2 30 minutes from the human protein at 37°C compared with <0.5 minute for levetiracetam and brivaracetam. In addition, its binding was not potentiated by the allosteric modulator UCB1244283. At recombinant GABAA receptors, padsevonil displayed low to moderate affinity (pIC50≤6.1) for the benzodiazepine site, and in electrophysiological studies, its relative efficacy compared with zolpidem (full-agonist reference drug) was 40%, indicating partial agonist properties. In in vivo (mice) receptor occupancy studies, padsevonil exhibited SV2A occupancy at low ED50 (0.2 mg/kg) and benzodiazepine site occupancy at higher doses (ED50 36 mg/kg), supporting in vitro results. Padsevonil's selectivity for its intended targets was confirmed in profiling studies, where it lacked significant effects on a wide variety of ion channels, receptors, transporters, and enzymes. Padsevonil is a first-in-class AED candidate with a unique target profile allowing for presynaptic and postsynaptic activity. SIGNIFICANCE STATEMENT: Padsevonil is an antiepileptic drug candidate developed as a single molecular entity interacting with both presynaptic and postsynaptic targets. Results of in vitro and in vivo radioligand binding assays confirmed this target profile: padsevonil displayed nanomolar affinity for the three synaptic vesicle 2 protein isoforms (SV2A, B, and C) and micromolar affinity for the benzodiazepine binding site on GABAA receptors. Furthermore, padsevonil showed greater affinity for and slower binding kinetics at SV2A than the selective SV2A ligands, levetiracetam, and brivaracetam.


Subject(s)
Anticonvulsants/pharmacokinetics , GABA Agonists/pharmacokinetics , Imidazoles/pharmacokinetics , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Pyrrolidinones/pharmacokinetics , Receptors, GABA-A/metabolism , Thiadiazoles/pharmacokinetics , Animals , Anticonvulsants/chemistry , COS Cells , Chlorocebus aethiops , GABA Agonists/chemistry , HEK293 Cells , Humans , Imidazoles/chemistry , Kinetics , Male , Mice , Mice, Inbred C57BL , Protein Binding , Pyrrolidinones/chemistry , Rats , Rats, Sprague-Dawley , Thiadiazoles/chemistry
2.
J Labelled Comp Radiopharm ; 61(5): 415-426, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29314165

ABSTRACT

Anxiolytic activity has been associated with GABAA α2 and α3 subunits. Several target compounds were identified and required in C-14 labeled form to enable a better understanding of their drug metabolism and pharmacokinetic properties. AZD7325 is a selective GABAA α2 and α3 receptor modulator intended for the treatment of anxiety through oral administration. A great number of AZD7325 metabolites were observed across species in vivo, whose identification was aided by [14 C]AZD7325. An interesting metabolic cyclization and aromatization pathway leading to the tricyclic core of M9 and the oxidative pathways to M10 and M42 are presented.


Subject(s)
GABA Agonists/chemistry , Heterocyclic Compounds, 2-Ring/chemistry , Inactivation, Metabolic , Animals , Carbon Radioisotopes/chemistry , GABA Agonists/pharmacokinetics , Hepatobiliary Elimination , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Rats
3.
J Neurosci ; 35(40): 13698-712, 2015 Oct 07.
Article in English | MEDLINE | ID: mdl-26446222

ABSTRACT

Interference between similar or overlapping memories formed at different times poses an important challenge on the hippocampal declarative memory system. Difficulties in managing interference are at the core of disabling cognitive deficits in neuropsychiatric disorders. Computational models have suggested that, in the normal brain, the sparse activation of the dentate gyrus granule cells maintained by tonic inhibitory control enables pattern separation, an orthogonalization process that allows distinct representations of memories despite interference. To test this mechanistic hypothesis, we generated mice with significantly reduced expression of the α5-containing GABAA (α5-GABAARs) receptors selectively in the granule cells of the dentate gyrus (α5DGKO mice). α5DGKO mice had reduced tonic inhibition of the granule cells without any change in fast phasic inhibition and showed increased activation in the dentate gyrus when presented with novel stimuli. α5DGKO mice showed impairments in cognitive tasks characterized by high interference, without any deficiencies in low-interference tasks, suggesting specific impairment of pattern separation. Reduction of fast phasic inhibition in the dentate gyrus through granule cell-selective knock-out of α2-GABAARs or the knock-out of the α5-GABAARs in the downstream CA3 area did not detract from pattern separation abilities, which confirms the anatomical and molecular specificity of the findings. In addition to lending empirical support to computational hypotheses, our findings have implications for the treatment of interference-related cognitive symptoms in neuropsychiatric disorders, particularly considering the availability of pharmacological agents selectively targeting α5-GABAARs. SIGNIFICANCE STATEMENT: Interference between similar memories poses a significant limitation on the hippocampal declarative memory system, and impaired interference management is a cognitive symptom in many disorders. Thus, understanding mechanisms of successful interference management or processes that can lead to interference-related memory problems has high theoretical and translational importance. This study provides empirical evidence that tonic inhibition in the dentate gyrus (DG), which maintains sparseness of neuronal activation in the DG, is essential for management of interference. The specificity of findings to tonic, but not faster, more transient types of neuronal inhibition and to the DG, but not the neighboring brain areas, is presented through control experiments. Thus, the findings link interference management to a specific mechanism, proposed previously by computational models.


Subject(s)
Dentate Gyrus/cytology , Memory/physiology , Neural Inhibition/genetics , Neurons/physiology , Receptors, GABA-A/metabolism , Animals , Discrimination, Psychological/physiology , Exploratory Behavior/physiology , GABA Agonists/pharmacokinetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Imidazoles/pharmacokinetics , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Receptors, GABA-A/genetics , Recognition, Psychology/physiology , Swimming/psychology
4.
J Pharmacol Exp Ther ; 356(1): 123-36, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26503377

ABSTRACT

Negative allosteric modulators (NAMs) of metabotropic glutamate receptor subtype 5 (mGlu5) have potential applications in the treatment of fragile X syndrome, levodopa-induced dyskinesia in Parkinson disease, Alzheimer disease, addiction, and anxiety; however, clinical and preclinical studies raise concerns that complete blockade of mGlu5 and inverse agonist activity of current mGlu5 NAMs contribute to adverse effects that limit the therapeutic use of these compounds. We report the discovery and characterization of a novel mGlu5 NAM, N,N-diethyl-5-((3-fluorophenyl)ethynyl)picolinamide (VU0477573) that binds to the same allosteric site as the prototypical mGlu5 NAM MPEP but displays weak negative cooperativity. Because of this weak cooperativity, VU0477573 acts as a "partial NAM" so that full occupancy of the MPEP site does not completely inhibit maximal effects of mGlu5 agonists on intracellular calcium mobilization, inositol phosphate (IP) accumulation, or inhibition of synaptic transmission at the hippocampal Schaffer collateral-CA1 synapse. Unlike previous mGlu5 NAMs, VU0477573 displays no inverse agonist activity assessed using measures of effects on basal [(3)H]inositol phosphate (IP) accumulation. VU0477573 acts as a full NAM when measuring effects on mGlu5-mediated extracellular signal-related kinases 1/2 phosphorylation, which may indicate functional bias. VU0477573 exhibits an excellent pharmacokinetic profile and good brain penetration in rodents and provides dose-dependent full mGlu5 occupancy in the central nervous system (CNS) with systemic administration. Interestingly, VU0477573 shows robust efficacy, comparable to the mGlu5 NAM MTEP, in models of anxiolytic activity at doses that provide full CNS occupancy of mGlu5 and demonstrate an excellent CNS occupancy-efficacy relationship. VU0477573 provides an exciting new tool to investigate the efficacy of partial NAMs in animal models.


Subject(s)
GABA Agonists/pharmacology , Picolinic Acids/pharmacology , Receptor, Metabotropic Glutamate 5/drug effects , Allosteric Regulation/drug effects , Animals , Anti-Anxiety Agents/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Behavior, Animal/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Drug Discovery , GABA Agonists/pharmacokinetics , HEK293 Cells , Humans , Inositol Phosphates/metabolism , MAP Kinase Signaling System/drug effects , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Picolinic Acids/pharmacokinetics , Pyridines/metabolism , Radioligand Assay , Rats , Receptor, Metabotropic Glutamate 5/metabolism , Synaptic Transmission/drug effects
5.
Br J Clin Pharmacol ; 81(5): 980-8, 2016 May.
Article in English | MEDLINE | ID: mdl-26659543

ABSTRACT

AIMS: γ-Hydroxybutyrate (GHB) is used as a treatment for narcolepsy and alcohol withdrawal and as a recreational substance. Nevertheless, there are limited data on the pharmacokinetics and pharmacokinetic-pharmacodynamic relationships of GHB in humans. We characterized the pharmacokinetic profile and exposure-psychotropic effect relationship of GHB in humans. METHODS: Two oral doses of GHB (25 and 35 mg kg(-1) ) were administered to 32 healthy male subjects (16 for each dose) using a randomized, placebo-controlled, cross-over design. RESULTS: Maximal concentrations of GHB were (geometric mean and 95% CI): 218 (176-270) nmol ml(-1) and 453 (374-549) nmol ml(-1) for the 25 and 35 mg kg(-1) GHB doses, respectively. The elimination half-lives (mean ± SD) were 36 ± 9 and 39 ± 7 min and the AUC∞ values (geometric mean and 95% CI) were 15 747 (12 854-19 290) and 40 113 (33 093-48 622) nmol∙min ml(-1) for the 20 and 35 mg kg(-1) GHB doses, respectively. Thus, plasma GHB exposure (AUC0-∞ ) rose disproportionally (+40%) with the higher dose. γ-Hydroxybutyrate produced mixed stimulant-sedative effects, with a dose-dependent increase in sedation and dizziness. It did not alter heart rate or blood pressure. A close relationship between plasma GHB exposure and its psychotropic effects was found, with higher GHB concentrations associated with higher subjective stimulation, sedation, and dizziness. No clockwise hysteresis was observed in the GHB concentration effect plot over time (i.e., no acute pharmacological tolerance). CONCLUSION: Evidence was found of a nonlinear dose-exposure relationship (i.e., no dose proportionality) at moderate doses of GHB. The effects of GHB on consciousness were closely linked to its plasma exposure and exhibited no acute tolerance.


Subject(s)
GABA Agonists/pharmacology , Hypnotics and Sedatives/pharmacology , Narcolepsy/drug therapy , Psychotropic Drugs/pharmacology , Sodium Oxybate/pharmacology , Substance Withdrawal Syndrome/drug therapy , Administration, Oral , Adult , Blood Pressure/drug effects , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Tolerance , GABA Agonists/administration & dosage , GABA Agonists/pharmacokinetics , Healthy Volunteers , Heart Rate/drug effects , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/pharmacokinetics , Male , Psychotropic Drugs/administration & dosage , Psychotropic Drugs/pharmacokinetics , Sodium Oxybate/administration & dosage , Sodium Oxybate/pharmacokinetics , Young Adult
6.
Alcohol Clin Exp Res ; 38(2): 316-21, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24033763

ABSTRACT

BACKGROUND: Baclofen is a GABA-B receptor agonist used in the treatment of spasticity. Recently, baclofen is used out of its label to decrease craving of alcoholic patients. Its optimal use in these patients requires further pharmacokinetic information. The objective of this study was to characterize the pharmacokinetics of baclofen in alcohol-dependent patients. Randomized clinical trials are ongoing to evaluate the efficacy for this new indication. METHODS: Thirty-seven outpatients (weight: 74.0 kg [42.0 to 104.0]; age: 49 years [31 to 68]) followed in the addictology unit were studied. Total mean dose of 77.9 mg (30 to 240) per day was administered by oral route. Therapeutic drug monitoring allowed the measurement of 139 plasma concentrations. The following covariates were evaluated: demographic data (age, body weight, height, sex), biological data (creatinine, urea, AST, ALT, albumin, PR, VGM, PAL, CDT, GGT), and tobacco consumption (number of cigarettes and Fagerstrom test). Pharmacokinetic analysis was performed by using a nonlinear mixed-effect population model (NONMEM 7.2 software). RESULTS: Data were modeled with a 1-compartment pharmacokinetic model. The population typical mean (95% confidence interval [95% CI]) values for clearance (CL), apparent volume of distribution (V), and rate constant of absorption (Ka) were 9.9 l/h (9.0 to 11.1), 80.7 l (63.6 to 96.9), and 4.6/h (1.5 to 19.9), respectively. The interindividual variability of CL (95% CI) and V (95% CI), and residual variability (95% CI) were 56.0% (47.9 to 60.7), 68.3% (48.7 to 80.1), and 0.096 mg/l (0.079 to 0.107), respectively. CONCLUSIONS: Baclofen exhibited a linear pharmacokinetics with a proportional relationship from 30 to 240 mg per day, the dose range currently used in alcoholic patients. A wide interpatient variability was observed which could not be explained by the covariates. This high variation of baclofen exposure may explain the lack of response observed for some patients.


Subject(s)
Alcoholism/metabolism , Baclofen/pharmacokinetics , GABA Agonists/pharmacokinetics , Adult , Aged , Algorithms , Area Under Curve , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Population , Regression Analysis
7.
Eksp Klin Farmakol ; 76(3): 38-40, 2013.
Article in Russian | MEDLINE | ID: mdl-23767103

ABSTRACT

Pharmacokinetic investigation of a new gamma-aminobutyric acid (GABA) derivative cirtocard showed that, upon the intravenous introduction, the drug is determined in high concentrations in organs of elimination--the liver and kidneys. The tissue accessibility amounts to 1.341 for the liver and 4.053 for the kidneys and the separation factor is 1.041 for the liver and 4.486 for the kidneys. The study of drug excretion showed that cirtocard is determined in the urine for 48 h, its nephritic clearance being 0.047 L/h and extra-nephritic clearance, 0.33 L/h. For the unchanged substance, a large significance ofhepatoduodenal circulation is low probable, since no more than 1 - 2% of the introduced dose was isolated with bile over entire experiment. It is established that the removal of the unchanged substance does not exceed 10% of the introduced dose. There is high probability of hepatoduodenal circulation and excretion of the preparation in the form of metabolites.


Subject(s)
Citric Acid/analogs & derivatives , Citric Acid/pharmacokinetics , Duodenum/metabolism , GABA Agonists/pharmacokinetics , Kidney/metabolism , Liver/metabolism , gamma-Aminobutyric Acid/analogs & derivatives , Animals , Citric Acid/pharmacology , GABA Agonists/pharmacology , Male , Rats , gamma-Aminobutyric Acid/pharmacokinetics , gamma-Aminobutyric Acid/pharmacology
8.
Int J Clin Pharmacol Ther ; 50(4): 307-14, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22456303

ABSTRACT

The novel Type B gamma-aminobutyric acid (GABAB)-receptor agonist lesogaberan (AZD3355) has been evaluated as an add-on to proton pump inhibitor treatment for gastroesophageal reflux disease, but the effect of food on the bioavailability of this compound has not been assessed. In this openlabel crossover study, healthy males received single 100 mg doses of lesogaberan (oral solution (A) or oral modified release (MR) capsules with a dissolution rate of 50% (B) or 100% (C) over 4 h) with and without food. Blood plasma concentrations of lesogaberan were assessed over 48 h. A log-transformed geometric mean Cmax and AUC ratio within the 90% confidence interval (CI) range (0.80 - 1.25) was defined as excluding a clinically relevant food effect. Overall, 57 subjects completed the study. Only the oral lesogaberan solution had a fed/fasting Cmax ratio outside the 90% CI range (Cmax ratio: 0.76). AUC ratios were within the 90% CI limits for all three lesogaberan formulations. The only substantial change in tmax associated with food intake was observed for the oral solution (1.0 h without food, 1.8 h with food). In conclusion, a clinically relevant food effect could be excluded for the lesogaberan MR formulations, but not for the oral lesogaberan solution.


Subject(s)
Food-Drug Interactions , GABA Agonists/administration & dosage , GABA Agonists/pharmacokinetics , Phosphinic Acids/administration & dosage , Phosphinic Acids/pharmacokinetics , Propylamines/administration & dosage , Propylamines/pharmacokinetics , Administration, Oral , Adolescent , Adult , Analysis of Variance , Area Under Curve , Biological Availability , Capsules , Cross-Over Studies , Delayed-Action Preparations , GABA Agonists/adverse effects , GABA Agonists/blood , Humans , Linear Models , Male , Middle Aged , Phosphinic Acids/adverse effects , Phosphinic Acids/blood , Propylamines/adverse effects , Propylamines/blood , Young Adult
9.
J Neurophysiol ; 106(4): 2057-64, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21795619

ABSTRACT

Extrasynaptic GABA(A) receptors (eGABARs) allow ambient GABA to tonically regulate neuronal excitability and are implicated as targets for ethanol and anesthetics. These receptors are thought to be heteropentameric proteins made up of two α subunits-either α4 or α6-two ß2 or ß3 subunits, and one δ subunit. The GABA analog 4,5,6,7-tetrahydroisoxazolo (5,4-c)pyridin-3(-ol) (THIP) has been proposed as a selective ligand for eGABARs. Behavioral and in vitro studies suggest that eGABARs have nanomolar affinity for THIP; however, all published studies on recombinant versions of eGABARs report micromolar affinities. Here, we examine THIP sensitivity of native eGABARs on cerebellar neurons and on reconstituted GABARs in heterologous systems. Concentration-response data for THIP, obtained from cerebellar granule cells and molecular layer interneurons in wild-type and δ subunit knockout slices, confirm that submicromolar THIP sensitivity requires δ subunits. In recombinant experiments, we find that δ subunit coexpression leads to receptors activated by nanomolar THIP concentrations (EC(50) of 30-50 nM for α4ß3δ and α6ß3δ), a sensitivity almost 1,000-fold higher than receptors formed by α4/6 and ß3 subunits. In contrast, γ2 subunit expression significantly reduces THIP sensitivity. Even when δ subunit cDNA or cRNA was supplied in excess, high- and low-sensitivity THIP responses were often apparent, indicative of variable mixtures of low-affinity αß and high-affinity αßδ receptors. We conclude that δ subunit incorporation into GABARs leads to a dramatic increase in THIP sensitivity, a defining feature that accounts for the unique behavioral and neurophysiological properties of THIP.


Subject(s)
GABA Agonists/pharmacology , GABAergic Neurons/drug effects , Isoxazoles/pharmacology , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Animals , Cerebellar Cortex/cytology , Dose-Response Relationship, Drug , GABA Agonists/administration & dosage , GABA Agonists/pharmacokinetics , GABAergic Neurons/physiology , HEK293 Cells , Humans , Isoxazoles/administration & dosage , Isoxazoles/pharmacokinetics , Mice , Mice, Knockout , Oocytes , Patch-Clamp Techniques , Protein Isoforms/physiology , Protein Multimerization , Protein Subunits , Receptors, GABA-A/chemistry , Receptors, GABA-A/deficiency , Receptors, GABA-A/genetics , Recombinant Fusion Proteins/physiology , Solubility , Xenopus laevis
10.
Bioorg Med Chem Lett ; 21(21): 6582-5, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21920749

ABSTRACT

Structure-activity studies have led to a discovery of 3-(4-pyridyl)methyl ether derivative 9d that has 25- to 50-fold greater functional potency than R-baclofen at human and rodent GABA(B) receptors in vitro. Mouse hypothermia studies confirm that this compound crosses the blood-brain barrier and is approximately 50-fold more potent after systemic administration.


Subject(s)
Baclofen/pharmacology , Drug Discovery , GABA Agonists/pharmacology , Receptors, GABA-B/drug effects , Animals , Baclofen/chemistry , Baclofen/pharmacokinetics , Blood-Brain Barrier , GABA Agonists/chemistry , GABA Agonists/pharmacokinetics , Humans , Mice
11.
J Pharmacol Exp Ther ; 330(3): 911-21, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19502531

ABSTRACT

Baclofen is a racemic GABA(B) receptor agonist that has a number of significant pharmacokinetic limitations, including a narrow window of absorption in the upper small intestine and rapid clearance from the blood. Arbaclofen placarbil is a novel transported prodrug of the pharmacologically active R-isomer of baclofen designed to be absorbed throughout the intestine by both passive and active mechanisms via the monocarboxylate type 1 transporter. Arbaclofen placarbil is rapidly converted to R-baclofen in human and animal tissues in vitro. This conversion seems to be primarily catalyzed in human tissues by human carboxylesterase-2, a major carboxylesterase expressed at high levels in various tissues including human intestinal cells. Arbaclofen placarbil was efficiently absorbed and rapidly converted to R-baclofen after oral dosing in rats, dogs, and monkeys. Exposure to R-baclofen was proportional to arbaclofen placarbil dose, whereas exposure to intact prodrug was low. Arbaclofen placarbil demonstrated enhanced colonic absorption, i.e., 5-fold higher R-baclofen exposure in rats and 12-fold higher in monkeys compared with intracolonic administration of R-baclofen. Sustained release formulations of arbaclofen placarbil demonstrated sustained R-baclofen exposure in dogs with bioavailability up to 68%. In clinical use, arbaclofen placarbil may improve the treatment of patients with gastroesophageal reflux disease, spasticity, and numerous other conditions by prolonging exposure and decreasing the fluctuations in plasma levels of R-baclofen.


Subject(s)
Baclofen/pharmacokinetics , GABA Agonists/pharmacokinetics , Prodrugs/pharmacokinetics , Animals , Binding, Competitive/drug effects , Butyrates/metabolism , Carboxylesterase/metabolism , Carboxylic Ester Hydrolases/metabolism , Cells, Cultured , Chemistry, Pharmaceutical , Cytochrome P-450 Enzyme System/metabolism , Humans , Hydrolysis , Intestinal Absorption , Isobutyrates , Isoenzymes/drug effects , LLC-PK1 Cells , Male , Membranes, Artificial , Oocytes/drug effects , Oocytes/metabolism , Rats , Rats, Sprague-Dawley , Tissue Distribution , Wine
12.
J Pharmacol Exp Ther ; 331(2): 470-84, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19704033

ABSTRACT

3-tert-Butyl-7-(5-methylisoxazol-3-yl)-2-(1-methyl-1H-1,2,4-triazol-5-ylmethoxy)-pyrazolo[1,5-d][1,2,4]triazine (MRK-016) is a pyrazolotriazine with an affinity of between 0.8 and 1.5 nM for the benzodiazepine binding site of native rat brain and recombinant human alpha1-, alpha2-, alpha3-, and alpha5-containing GABA(A) receptors. It has inverse agonist efficacy selective for the alpha5 subtype, and this alpha5 inverse agonism is greater than that of the prototypic alpha5-selective compound 3-(5-methylisoxazol-3-yl)-6-[(1-methyl-1,2,3-triazol-4-hdyl)methyloxy]-1,2,4-triazolo[3,4-a]phthalazine (alpha5IA). Consistent with its greater alpha5 inverse agonism, MRK-016 increased long-term potentiation in mouse hippocampal slices to a greater extent than alpha5IA. MRK-016 gave good receptor occupancy after oral dosing in rats, with the dose required to produce 50% occupancy being 0.39 mg/kg and a corresponding rat plasma EC(50) value of 15 ng/ml that was similar to the rhesus monkey plasma EC(50) value of 21 ng/ml obtained using [(11)C]flumazenil positron emission tomography. In normal rats, MRK-016 enhanced cognitive performance in the delayed matching-to-position version of the Morris water maze but was not anxiogenic, and in mice it was not proconvulsant and did not produce kindling. MRK-016 had a short half-life in rat, dog, and rhesus monkey (0.3-0.5 h) but had a much lower rate of turnover in human compared with rat, dog, or rhesus monkey hepatocytes. Accordingly, in human, MRK-016 had a longer half-life than in preclinical species ( approximately 3.5 h). Although it was well tolerated in young males, with a maximal tolerated single dose of 5 mg corresponding to an estimated occupancy in the region of 75%, MRK-016 was poorly tolerated in elderly subjects, even at a dose of 0.5 mg, which, along with its variable human pharmacokinetics, precluded its further development.


Subject(s)
GABA Agonists/pharmacology , GABA-A Receptor Agonists , Isoxazoles/pharmacology , Triazines/pharmacology , Animals , Anxiety/psychology , Behavior, Animal/drug effects , Convulsants/pharmacology , Dogs , Dose-Response Relationship, Drug , Electric Stimulation , Electrophysiology , Excitatory Postsynaptic Potentials/drug effects , Fibroblasts , Flumazenil/metabolism , GABA Agonists/metabolism , GABA Agonists/pharmacokinetics , GABA Modulators/metabolism , Hepatocytes/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Macaca mulatta , Male , Maze Learning/drug effects , Mice , Patch-Clamp Techniques , Postural Balance/drug effects , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Young Adult
13.
J Psychopharmacol ; 23(6): 625-32, 2009 Aug.
Article in English | MEDLINE | ID: mdl-18635696

ABSTRACT

Benzodiazepines are effective short-term treatments for anxiety disorders, but their use is limited by undesirable side effects related to Central Nervous System impairment and tolerance development. SL65.1498 is a new compound that acts in vitro as a full agonist at the gamma-aminobutyric acid(A) 2 and 3 receptor and as a partial agonist at the 1 and 5 receptor subtypes. It is thought that the compound could be anxiolytic by its activation at the alpha2 and alpha3 receptor subtypes, without causing unfavourable side effects, which are believed to be mediated by the alpha1 and alpha5 subtypes. This study was a double-blind, five-way cross-over study to investigate the effects of three doses of SL65.1498 in comparison with placebo and lorazepam 2 mg in healthy volunteers. The objective was to select a dose level (expected to be therapeutically active), free of any significant deleterious effect. Psychomotor and cognitive effects were measured using a validated battery of measurements, including eye movements, body sway, memory tests, reaction-time assessments, and visual analogue scales. The highest dose of SL65.1498 showed slight effects on saccadic peak velocity and smooth pursuit performance, although to a much lesser extent than lorazepam. In contrast to lorazepam, none of the SL65.1498 doses affected body sway, visual analogue scale alertness, attention, or memory tests. This study showed that the three doses of SL65.1498 were well tolerated and induced no impairments on memory, sedation, psychomotor, and cognitive functions.


Subject(s)
GABA Agonists/pharmacology , GABA Agonists/pharmacokinetics , GABA Modulators/pharmacology , GABA Modulators/pharmacokinetics , GABA-A Receptor Agonists , Indoles/pharmacology , Indoles/pharmacokinetics , Lorazepam/pharmacology , Lorazepam/pharmacokinetics , Pyrroles/pharmacology , Pyrroles/pharmacokinetics , Adult , Attention/drug effects , Cognition/drug effects , Cross-Over Studies , Double-Blind Method , Eye Movements/drug effects , Female , GABA Agonists/adverse effects , GABA Modulators/adverse effects , Humans , Indoles/adverse effects , Lorazepam/adverse effects , Male , Memory/drug effects , Neuropsychological Tests , Postural Balance/drug effects , Pursuit, Smooth/drug effects , Pyrroles/adverse effects , Receptors, GABA-A , Saccades/drug effects , Young Adult
14.
Biopharm Drug Dispos ; 30(1): 9-20, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19152227

ABSTRACT

Typically, the dose-occupancy curves for GABA-A receptor ligands are determined using in vivo binding of [3H]flumazenil. This study describes in vivo binding experiments without the use of tracer ligands. Bound and free fractions were measured directly using a highly sensitive LC/MS/MS detection method after in vivo administration of the GABA-A ligands zolpidem, (RS)-zopiclone, L-838417 and flumazenil, to demonstrate affinity and saturation of the filter-retained, membrane-bound fraction. The in vivo binding of flumazenil and L-838417 both saturated around 200 nM, at a similar level to the specific binding of (S)-zopiclone after doses of the racemic zopiclone, using (R)-zopiclone to estimate non-specific binding. This saturable component represented an estimate of benzodiazepine binding sites available on GABA-A receptors in vivo (200 nM). Dose-occupancy curves were constructed to estimate the dose required to achieve 50% occupancy and matched estimates obtained with tracer methods. In contrast to tracer methods, this method is uniquely suitable to the demonstration of stereoselective binding of the (S)-isomer in vivo after doses of racemic zopiclone. These results demonstrate that the LC/MS/MS measurements of total drug concentrations typically used in early drug development can be adapted to provide information about receptor occupancy in vivo.


Subject(s)
Receptors, GABA-A/metabolism , Algorithms , Animals , Azabicyclo Compounds/pharmacokinetics , Binding Sites , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Flumazenil/pharmacokinetics , Fluorobenzenes/pharmacokinetics , GABA Agonists/pharmacokinetics , GABA Modulators/pharmacokinetics , Hypnotics and Sedatives/pharmacokinetics , In Vitro Techniques , Indicators and Reagents , Ligands , Male , Mice , Models, Statistical , Piperazines/pharmacokinetics , Pyridines/pharmacokinetics , Stereoisomerism , Tandem Mass Spectrometry , Triazoles/pharmacokinetics , Zolpidem
15.
J Neurochem ; 104(1): 173-86, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17986225

ABSTRACT

Studies on animal models of epilepsy and cerebellar ataxia, e.g., stargazer mice (stg) have identified changes in the GABAergic properties of neurones associated with the affected brain loci. Whether these changes contribute to or constitute homeostatic adaptations to a state of altered neuronal excitability is as yet unknown. Using cultured cerebellar granule neurones from control [+/+; alpha-amino-3-hydroxyl-5-methyl-4-isoxazolepropionate receptor (AMPAR)-competent, Kainate receptor (KAR)-competent] and stg (AMPAR-incompetent, KAR-competent), we investigated whether non-NMDA receptor (NMDAR) activity regulates GABA(A) receptor (GABAR) expression. Neurones were maintained in 5 mmol/L KCl-containing basal media or depolarizing media containing either 25 mmol/L KCl or the non-NMDAR agonist kainic acid (KA) (100 micromol/L). KCl- and KA-mediated depolarization down-regulated GABAR alpha1, alpha6 and beta2, but up-regulated alpha4, beta3 and delta subunits in +/+ neurones. The KCl-evoked but not KA-evoked effects were reciprocated in stg neurones compatible with AMPAR-regulation of GABAR expression. Conversely, GABAR gamma2 expression was insensitive to KCl-mediated depolarization, but was down-regulated by KA-treatment in a 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX)-reversible manner in +/+ and stg neurones compatible with a KAR-mediated response. KA-mediated up-regulation of GABAR alpha4, beta3 and delta was inhibited by L-type voltage-gated calcium channel (L-VGCC) blockers and the Ca2+/calmodulin-dependent protein kinase inhibitor, 4-[(2S)-2-[(5-isoquinolinylsulfonyl)methylamino]-3-oxo-3-(4-phenyl-1-piperazinyl)propyl] phenyl isoquinoline sulfonic acid ester (KN-62). Up-regulation of GABAR alpha4 and beta3 was also prevented by calcineurin (CaN) inhibitors, FK506 and cyclosporin A. Down-regulation of GABAR alpha1, alpha6 and beta2 was independent of L-VGCC activity, but was prevented by inhibitors of CaN. Thus, we provide evidence that a KAR-mediated and at least three mutually exclusive AMPAR-mediated signalling mechanisms regulate neuronal GABAR expression.


Subject(s)
Cerebellum/cytology , Neurons/metabolism , Receptors, AMPA/physiology , Receptors, GABA-A/metabolism , Receptors, Kainic Acid/physiology , Animals , Animals, Newborn , Azides/pharmacokinetics , Benzodiazepines/pharmacokinetics , Cells, Cultured , Excitatory Amino Acids/pharmacology , GABA Agonists/pharmacokinetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Kainic Acid/pharmacology , Mice , Mice, Neurologic Mutants , Models, Biological , Muscimol/pharmacokinetics , Neurons/drug effects , Potassium Chloride/pharmacology , Protein Subunits/genetics , Protein Subunits/metabolism , Radioligand Assay/methods , Signal Transduction/drug effects , Signal Transduction/physiology , Tritium/pharmacokinetics
16.
Schizophr Res ; 90(1-3): 76-80, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17208412

ABSTRACT

Changes in GABA(A) receptors are observed in schizophrenia, with benzodiazepine-sensitive GABA(A) receptor subtypes being affected differently to other subtypes. However, long-term antipsychotic drug use in schizophrenia may underlie these changes. To test this, we examined the effects of administering a typical (haloperidol) and an atypical (olanzapine) antipsychotic drug on the GABA(A) receptor agonist (orthosteric) and benzodiazepine (allosteric) binding sites in rat prefrontal cortex. As antipsychotic drugs have delayed maximal therapeutic effects we also examined different drug treatment periods. Male SD rats received a sucrose solution containing either haloperidol (1.5 mg/kg), olanzapine (6.5 mg/kg) or no drug daily for either 7, 14 or 28 days. Sections of rat brain were then labelled with [(3)H]muscimol, which labels the total population of GABA(A) receptors, or the benzodiazepine site ligand [(3)H]flunitrazepam in separate saturation binding experiments using quantitative receptor autoradiography. [(3)H]Muscimol binding was enhanced in the prefrontal cortex after 7 days but no differences were observed after longer periods of drug administration. In contrast there was a delayed increase in density of benzodiazepine-sensitive GABA(A) receptors in the PFC, suggesting that antipsychotic drugs have different effects on different GABA(A) receptor subtypes. These changes in the properties of GABA(A) receptor binding following antipsychotic drug administration are not consistent with those observed in schizophrenia and suggest a 'reshuffling' in GABA(A) receptor subtypes over time.


Subject(s)
Antipsychotic Agents/pharmacology , Flunitrazepam/pharmacokinetics , GABA Agonists/pharmacokinetics , GABA Modulators/pharmacokinetics , Haloperidol/pharmacology , Muscimol/pharmacokinetics , Prefrontal Cortex/drug effects , Receptors, GABA-A/drug effects , Animals , Benzodiazepines/pharmacology , Binding, Competitive/drug effects , Humans , Long-Term Care , Male , Olanzapine , Prefrontal Cortex/pathology , Radioligand Assay , Rats , Rats, Sprague-Dawley , Schizophrenia/drug therapy , Schizophrenia/pathology
17.
Eur J Pharmacol ; 562(1-2): 47-52, 2007 May 07.
Article in English | MEDLINE | ID: mdl-17362924

ABSTRACT

Gaboxadol has been suggested to be a selective extrasynaptic GABA(A) receptor agonist. However, there is little information on Gaboxadol concentrations in the central nervous system (CNS) at therapeutically relevant doses. In order to investigate this, rats were injected subcutaneously with Gaboxadol and plasma and CNS concentrations were determined using the dynamic-no-net-flux and ultraslow microdialysis methods. Results using the 2 methods were similar and showed that Gaboxadol rapidly entered the brain and that peak CNS concentrations after 2.5, 5 and 10 mg/kg were in the range of 0.7 to 3 microM. Furthermore, a very short half-life (28 min) in both plasma and CNS was observed. It is concluded that concentrations of Gaboxadol in the CNS are in a range, which are likely to activate only extrasynaptic (nongamma subunit containing) GABA(A) receptors.


Subject(s)
Central Nervous System/metabolism , Isoxazoles/blood , Animals , Chromatography, Liquid , GABA Agonists/administration & dosage , GABA Agonists/blood , GABA Agonists/pharmacokinetics , Infusion Pumps , Injections, Subcutaneous , Isoxazoles/administration & dosage , Isoxazoles/pharmacokinetics , Male , Mass Spectrometry , Microdialysis/methods , Rats , Rats, Wistar , Time Factors
18.
J Control Release ; 262: 296-304, 2017 Sep 28.
Article in English | MEDLINE | ID: mdl-28774840

ABSTRACT

In this work, a fixed-dose combination of gabapentin and flurbiprofen formulated as multilayer tablets has been designed, developed and studied in vitro and in vivo. The aim was to construct a single dosage form of the two drugs, able to perform a therapeutic program involving three release kinetics and two delivery sites, i.e., immediate release of gabapentin, intra-gastric prolonged release of gabapentin and intestinal (delayed) release of flurbiprofen. An oblong three-layer tablet was manufactured having as top layer a floating hydrophilic polymeric matrix for gastric release of gabapentin, as middle layer a disintegrating formulation for immediate release of a gabapentin loading dose and as bottom layer, an uncoated hydrophilic polymeric matrix, swellable but insoluble in gastric fluids, for delayed and prolonged release of flurbiprofen in intestinal environment. The formulations were studied in vitro and in vivo in healthy volunteers. The in vitro release rate assessment confirmed the programmed delivery design. A significant higher bioavailability of gabapentin administered 30min after meal, compared to fasting conditions or to dose administration 10min before meal, argued in favor of the gastro-retention of gabapentin prolonged release layer. The two drugs were delivered at different anatomical sites, since the food presence prolonged the gastric absorption of gabapentin from the floating layer and delayed the flurbiprofen absorption. The attainment of a successful delayed release of flurbiprofen was realized by a matrix based on a polymers' combination. The combined use of three hydrophilic polymers with different pH sensitivity provided the dosage form layer containing flurbiprofen with gastro-resistant characteristics without the use of film coating.


Subject(s)
Amines/administration & dosage , Amines/pharmacokinetics , Cyclohexanecarboxylic Acids/administration & dosage , Cyclohexanecarboxylic Acids/pharmacokinetics , Flurbiprofen/administration & dosage , Flurbiprofen/pharmacokinetics , gamma-Aminobutyric Acid/administration & dosage , gamma-Aminobutyric Acid/pharmacokinetics , Administration, Oral , Adolescent , Adult , Analgesics/administration & dosage , Analgesics/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Cross-Over Studies , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Combinations , Drug Liberation , Female , Food-Drug Interactions , GABA Agonists/administration & dosage , GABA Agonists/pharmacokinetics , Gabapentin , Gastric Mucosa/metabolism , Humans , Intestinal Mucosa/metabolism , Male , Middle Aged , Polymers/administration & dosage , Polymers/pharmacokinetics , Tablets , Young Adult
19.
J Neurosci ; 25(39): 8978-87, 2005 Sep 28.
Article in English | MEDLINE | ID: mdl-16192388

ABSTRACT

In recent studies, inactivation of the dorsal hippocampus before the retrieval of extinguished fear memories disrupted the context-dependent expression of these memories. In the present experiments, we examined the role of the dorsal hippocampus in the acquisition of extinction. After pairing an auditory conditional stimulus (CS) with an aversive footshock [unconditional stimulus (US)], rats received an extinction session in which the CS was presented without the US. In experiment 1, infusion of muscimol, a GABAA receptor agonist, into the dorsal hippocampus before the extinction training session decreased the rate of extinction. Moreover, when later tested for fear to the extinguished CS, all rats that had received hippocampal inactivation before extinction training demonstrated renewed fear regardless of the context in which testing took place. This suggests a role for the dorsal hippocampus in both acquiring the extinction memory and encoding the CS-context relationship that yields the context dependence of extinction. In experiment 2, inactivation of the dorsal hippocampus before testing also disrupted the context dependence of fear to the extinguished CS. In experiment 3, quantitative autoradiography revealed the boundaries of muscimol diffusion after infusion into the dorsal hippocampus. Together, these results reveal that the dorsal hippocampus is involved in the acquisition, contextual encoding, and context-dependent retrieval of fear extinction. Learning and remembering when and where aversive events occur is essential for adaptive emotional regulation.


Subject(s)
Extinction, Psychological/physiology , Fear , Hippocampus/physiology , Acoustic Stimulation , Animals , Autoradiography , Behavior, Animal/drug effects , Conditioning, Psychological , Electroshock , Extinction, Psychological/drug effects , Fear/drug effects , GABA Agonists/administration & dosage , GABA Agonists/pharmacokinetics , GABA Agonists/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Infusion Pumps , Male , Muscimol/administration & dosage , Muscimol/pharmacokinetics , Muscimol/pharmacology , Rats , Rats, Long-Evans
20.
Drug Alcohol Depend ; 83(1): 25-32, 2006 Jun 09.
Article in English | MEDLINE | ID: mdl-16298087

ABSTRACT

BACKGROUND: Alcohol effects in humans involve gamma-amino butyric acid (GABA) neurotransmission. It has been proposed that GABAergic medications may be effective in the treatment of alcohol dependence. This study evaluated the acute effects of gabapentin, an anticonvulsant that increases extracellular GABA, on the subjective, physiological, and performance effects of alcohol in heavy (mean 34 drinks per week) alcohol drinkers. METHODS: Seventeen volunteers without alcohol dependence were tested using a double-blind design with three 3-day long inpatient phases, each separated by at least a 1-week wash-out period. Each phase, gabapentin (0, 1000, or 2000mg) was administered 4h before alcohol (0.75g/kg), which was given in four divided doses every 20min. RESULTS: Gabapentin impaired the ability to balance without producing changes in subjective, physiological or other performance measures. Pretreatment with gabapentin did not significantly alter subjective and performance effects of alcohol and did not alter alcohol craving. Gabapentin, dose-dependently enhanced alcohol-induced tachycardia. CONCLUSIONS: Acute gabapentin administration was well tolerated in combination with alcohol, but did not alter the effects of alcohol.


Subject(s)
Alcoholism/psychology , Amines/pharmacology , Anticonvulsants/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , Ethanol/adverse effects , GABA Agonists/pharmacology , gamma-Aminobutyric Acid/pharmacology , Adult , Alcoholic Intoxication/psychology , Alcoholism/blood , Amines/pharmacokinetics , Anticonvulsants/pharmacokinetics , Cyclohexanecarboxylic Acids/pharmacokinetics , Dose-Response Relationship, Drug , Double-Blind Method , Drug Interactions , Ethanol/blood , Female , GABA Agonists/pharmacokinetics , Gabapentin , Heart Rate/drug effects , Humans , Male , Mental Recall/drug effects , Motivation , Neuropsychological Tests , Postural Balance/drug effects , Premedication , Psychomotor Performance/drug effects , Verbal Learning/drug effects , gamma-Aminobutyric Acid/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL