Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters

Publication year range
1.
Anesthesiology ; 134(1): 35-51, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33064833

ABSTRACT

BACKGROUND: γ-Aminobutyric acid type A (GABAA) receptor agonists are known to cause involuntary muscle movements. The mechanism of these movements is not known, and its relationship to depth of anesthesia monitoring is unclear. We have explored the effect of involuntary muscle movement on the pharmacokinetic-pharmacodynamic model for the GABAA receptor agonist ABP-700 and its effects on the Bispectral Index (BIS) as well as the Modified Observer's Assessment of Alertness/Sedation (MOAA/S) scores. METHODS: Observations from 350 individuals (220 men, 130 women) were analyzed, comprising 6,312 ABP-700 concentrations, 5,658 ABP-700 metabolite (CPM-acid) concentrations, 25,745 filtered BIS values, and 6,249 MOAA/S scores, and a recirculatory model developed. Various subject covariates and pretreatment with an opioid or a benzodiazepine were explored as covariates. Relationships between BIS and MOAA/S models and involuntary muscle movements were examined. RESULTS: The final model shows that the pharmacokinetics of ABP-700 are characterized by small compartmental volumes and rapid clearance. The BIS model incorporates an effect-site for BIS suppression and a secondary excitatory/disinhibitory effect-site associated with a risk of involuntary muscle movements. The secondary effect-site has a threshold that decreases with age. The MOAA/S model did not show excitatory effects. CONCLUSIONS: The GABAA receptor agonist ABP-700 shows the expected suppressive effects for BIS and MOAA/S, but also disinhibitory effects for BIS associated with involuntary muscle movements and reduced by pretreatment. Our model provides information about involuntary muscle movements that may be useful to improve depth of anesthesia monitoring for GABAA receptor agonists.


Subject(s)
Anesthesia , Consciousness Monitors , Etomidate/analogs & derivatives , GABA-A Receptor Agonists/pharmacology , Imidazoles/pharmacology , Adult , Algorithms , Analgesics, Opioid , Benzodiazepines , Conscious Sedation , Etomidate/pharmacokinetics , Female , GABA-A Receptor Agonists/pharmacokinetics , Humans , Imidazoles/pharmacokinetics , Male , Monitoring, Intraoperative , Muscle, Smooth/drug effects , Preanesthetic Medication
2.
Acta Neuropsychiatr ; 29(5): 309-314, 2017 Oct.
Article in English | MEDLINE | ID: mdl-27938419

ABSTRACT

OBJECTIVE: Prenatal exposure to valproic acid (VPA) enhances the risk for later development of autism spectrum disorders (ASD). An altered gamma-aminobutyric acid (GABA) system may be a key factor in ASD. Here we investigated possible changes in the GABA system in rats exposed to a low dose of prenatal VPA. METHOD: We performed autoradiography with [3H]muscimol, (a GABAA receptor agonist), and [11C]Ro15-4513 (a partial agonist of the GABAA α1+5 receptor subtypes), in brain sections containing amygdala, thalamus and hippocampus of rats treated prenatally with 20 mg/kg VPA or saline from the 12th day of gestation. Result Prenatal VPA significantly increased [11C]Ro15-4513 binding in the left amygdala compared with controls (p<0.05). This difference was not observed in the hippocampus, thalamus or right amygdala. No differences were observed in [3H]muscimol binding. CONCLUSION: We observed an asymmetric increase in GABAA receptor binding. Disturbances in the GABAA receptor system have also been detected in human autism with [11C]Ro15-4513.


Subject(s)
Amygdala/drug effects , Amygdala/metabolism , Autism Spectrum Disorder/chemically induced , GABA Agents/administration & dosage , Prenatal Exposure Delayed Effects/chemically induced , Receptors, GABA-A/metabolism , Valproic Acid/administration & dosage , Animals , Autism Spectrum Disorder/metabolism , Autoradiography , Azides/pharmacokinetics , Benzodiazepines/pharmacokinetics , Carbon Radioisotopes , Disease Models, Animal , Female , GABA-A Receptor Agonists/pharmacokinetics , Male , Muscimol/pharmacokinetics , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats
3.
Bioorg Med Chem Lett ; 26(2): 445-453, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26706170

ABSTRACT

New derivatives of ethyl 8-oxo-5,6,7,8-tetrahydro-thiazolo[3,2-a][1,3]diazepin-3-carboxylate (HIE-124, 3), were synthesized as continuation to our previous patented efforts. Compounds 15 and 20 showed marginal hypnotic potency compared to 3. Compounds 15 (0.78mmol/kg) and 20 (0.39mmol/kg) showed remarkable 100% protection against PTZ induced convulsions with two and four fold increase in activity than sodium valproate (1.38mmol/kg), respectively. Molecular modeling studies showed hydrogen bonding interaction between 15 and Thr56 residues at the binding site of GABAA. Superposition, flexible alignment and surface mapping of 15, 20 and diazepam supports their biological resemblance where ADMET study suggested that those compounds could be used as oral anticonvulsants.


Subject(s)
Anticonvulsants/chemistry , Anticonvulsants/therapeutic use , Azepines/chemistry , Azepines/therapeutic use , Carboxylic Acids/chemistry , Carboxylic Acids/therapeutic use , Hypnotics and Sedatives/chemistry , Hypnotics and Sedatives/therapeutic use , Seizures/drug therapy , Thiazoles/chemistry , Thiazoles/therapeutic use , Animals , Anticonvulsants/pharmacokinetics , Anticonvulsants/pharmacology , Azepines/pharmacokinetics , Azepines/pharmacology , Carboxylic Acids/pharmacokinetics , Carboxylic Acids/pharmacology , GABA-A Receptor Agonists/chemistry , GABA-A Receptor Agonists/pharmacokinetics , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Agonists/therapeutic use , Hypnotics and Sedatives/pharmacokinetics , Hypnotics and Sedatives/pharmacology , Mice , Models, Molecular , Pentylenetetrazole , Receptors, GABA-A/metabolism , Seizures/chemically induced , Thiazoles/pharmacokinetics , Thiazoles/pharmacology
4.
BMC Neurosci ; 16: 67, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26480871

ABSTRACT

BACKGROUND: Synaptic dysfunction is a key event in pathogenesis of neurodegenerative diseases such as Alzheimer's disease (AD) where synapse loss pathologically correlates with cognitive decline and dementia. Although evidence suggests that aberrant protein production and aggregation are the causative factors in familial subsets of such diseases, drugs singularly targeting these hallmark proteins, such as amyloid-ß, have failed in late stage clinical trials. Therefore, to provide a successful disease-modifying compound and address synaptic dysfunction and memory loss in AD and mixed pathology dementia, we repurposed a clinically proven drug, CMZ, with neuroprotective and anti-inflammatory properties via addition of nitric oxide (NO) and cGMP signaling property. RESULTS: The novel compound, NMZ, was shown to retain the GABAA potentiating actions of CMZ in vitro and sedative activity in vivo. Importantly, NMZ restored LTP in hippocampal slices from AD transgenic mice, whereas CMZ was without effect. NMZ reversed amnestic blockade of acetylcholine receptors by scopolamine as well as NMDA receptor blockade by a benzodiazepine and a NO synthase inhibitor in the step-through passive avoidance (STPA) test of learning and working memory. A PK/PD relationship was developed based on STPA analysis coupled with pharmacokinetic measures of drug levels in the brain: at 1 nM concentration in brain and plasma, NMZ was able to restore memory consolidation in mice. CONCLUSION: Our findings show that NMZ embodies a promising pharmacological approach targeting synaptic dysfunction and opens new avenues for neuroprotective intervention strategies in mixed pathology AD, neurodegeneration, and dementia.


Subject(s)
Alzheimer Disease/drug therapy , Chlormethiazole/analogs & derivatives , Drug Repositioning/methods , GABA-A Receptor Agonists/pharmacology , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Neuroprotective Agents/pharmacology , Nootropic Agents/pharmacology , Animals , CREB-Binding Protein/metabolism , Cyclic GMP/metabolism , Disease Models, Animal , GABA-A Receptor Agonists/pharmacokinetics , Male , Mice , Mice, Transgenic , Neuroprotective Agents/pharmacokinetics , Nitric Oxide/metabolism , Nootropic Agents/pharmacokinetics , Signal Transduction/drug effects , Synapses/drug effects , Synapses/pathology , Xenopus laevis
5.
Eur J Clin Pharmacol ; 71(10): 1209-21, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26257250

ABSTRACT

PURPOSE: To quantify pharmacokinetic (PK) and pharmacodynamic (PD) relationships of various classes of GABAA agonists in healthy volunteers, in order to investigate the sensitivity of the biomarker responses due to differing GABAA-subtype selectivity and to explore the correlation between biomarker responses and side effects of these drugs. METHODS: A comprehensive search was conducted for published placebo-controlled clinical studies of non- and α1-selective GABAA drugs in healthy volunteers. PK/PD models were developed for concentrations and biomarker outcomes (saccadic eye movement (SEM), visual analogue scale (VAS), digit symbol substitution task (DSST), and critical flicker fusion test (CFFT)) extracted from included studies. Predicted responses and equivalent doses for biomarkers (based on predicted response) were used to compare drug effects. And the relationship between biomarkers and safety was explored by linear regression. RESULTS: A total of 2237 data from 163 articles were included. Based on PK and placebo effect modeling, linear biomarker-concentration relationships well fit the data. The α1-selective compounds had similar equivalent doses for VAS, DSST, and CFFT (4.7-6.7 mg), which were about three to seven times lower than that for SEM (14.4-35.5 mg), while such difference was less evident for non-selective drugs. DSST had the highest correlations with incidences of somnolence and dizziness. CONCLUSIONS: The integral PK/PD models of GABAA agonists were established in healthy volunteers. SEM was identified as the most sensitive biomarker in differentiating GABAA receptor α1 subtype selective compounds. The exploratory analysis implied that different relationships existed between the drug effects on biomarkers and the adverse event profiles in healthy volunteers.


Subject(s)
GABA-A Receptor Agonists/pharmacology , Biomarkers , Dose-Response Relationship, Drug , Double-Blind Method , GABA-A Receptor Agonists/pharmacokinetics , Healthy Volunteers , Humans , Protein Binding , Saccades , Visual Analog Scale
6.
BMC Gastroenterol ; 14: 188, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25407279

ABSTRACT

BACKGROUND: The γ-aminobutyric acid type B-receptor agonist lesogaberan (AZD3355) has been developed for use in patients with gastroesophageal reflux disease (GERD) symptoms despite proton pump inhibitor (PPI) therapy (partial responders). This study aimed to explore the dose-response effect of lesogaberan on reflux episodes in partial responders. METHODS: In this randomized, single-centre, double-blind, crossover, placebo-controlled study, partial responders taking optimised PPI therapy were given 30, 90, 120 and 240 mg doses of lesogaberan. Each dose was given twice (12 h apart) during a 24-h period, during which impedance-pH measurements were taken. RESULTS: Twenty-five patients were included in the efficacy analysis and 27 in the safety analysis. The effect of lesogaberan on the mean number of reflux episodes was dose-dependent, and all doses significantly reduced the mean number of reflux episodes relative to placebo. Lesogaberan also dose-dependently reduced the mean number of acid reflux episodes (except the 30 mg dose) and weakly acid reflux episodes (all doses) significantly, relative to placebo. Regardless of dose, lesogaberan had a similar effect on the percentage of time with esophageal pH < 4 [mean reduction: 68.5% (30 mg), 54.2% (90 mg), 65.9% (120 mg), 72.1% (240 mg); p < 0.05 except 90 mg dose]. No adverse events led to discontinuation and no serious adverse events occurred during active treatment. CONCLUSIONS: Lesogaberan inhibited reflux in a dose-dependent manner in partial responders taking optimised PPI therapy, and these effects were significant versus placebo. All lesogaberan doses were well tolerated and were not associated with clinically relevant adverse events. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT01043185.


Subject(s)
GABA-A Receptor Agonists/administration & dosage , Gastroesophageal Reflux/drug therapy , Gastrointestinal Agents/administration & dosage , Phosphinic Acids/administration & dosage , Propylamines/administration & dosage , Adult , Aged , Dose-Response Relationship, Drug , Double-Blind Method , Esophagus/physiopathology , Female , GABA-A Receptor Agonists/adverse effects , GABA-A Receptor Agonists/pharmacokinetics , Gastroesophageal Reflux/physiopathology , Headache/chemically induced , Humans , Hydrogen-Ion Concentration , Male , Middle Aged , Nausea/chemically induced , Phosphinic Acids/adverse effects , Phosphinic Acids/pharmacokinetics , Propylamines/adverse effects , Propylamines/pharmacokinetics , Young Adult
7.
Pharmacol Rev ; 63(1): 243-67, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21245208

ABSTRACT

GABA is the major inhibitory neurotransmitter in the central nervous system (CNS). The type A GABA receptor (GABA(A)R) system is the primary pharmacological target for many drugs used in clinical anesthesia. The α1, ß2, and γ2 subunit-containing GABA(A)Rs located in the various parts of CNS are thought to be involved in versatile effects caused by inhaled anesthetics and classic benzodiazepines (BZD), both of which are widely used in clinical anesthesiology. During the past decade, the emergence of tonic inhibitory conductance in extrasynaptic GABA(A)Rs has coincided with evidence showing that these receptors are highly sensitive to the sedatives and hypnotics used in anesthesia. Anesthetic enhancement of tonic GABAergic inhibition seems to be preferentially increased in regions shown to be important in controlling memory, awareness, and sleep. This review focuses on the physiology of the GABA(A)Rs and the pharmacological properties of clinically used BZDs. Although classic BZDs are widely used in anesthesiological practice, there is a constant need for new drugs with more favorable pharmacokinetic and pharmacodynamic effects and fewer side effects. New hypnotics are currently developed, and promising results for one of these, the GABA(A)R agonist remimazolam, have recently been published.


Subject(s)
Anesthetics, Intravenous/pharmacology , Benzodiazepines/pharmacology , Central Nervous System/drug effects , GABA-A Receptor Agonists/pharmacology , Neurons/drug effects , Receptors, GABA-A/metabolism , Anesthetics, Intravenous/chemistry , Anesthetics, Intravenous/pharmacokinetics , Anesthetics, Intravenous/therapeutic use , Animals , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/pharmacokinetics , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Benzodiazepines/chemistry , Benzodiazepines/pharmacokinetics , Benzodiazepines/therapeutic use , Central Nervous System/enzymology , Central Nervous System/metabolism , Drug Interactions , GABA-A Receptor Agonists/chemistry , GABA-A Receptor Agonists/pharmacokinetics , GABA-A Receptor Agonists/therapeutic use , Humans , Hypnotics and Sedatives/chemistry , Hypnotics and Sedatives/pharmacokinetics , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/therapeutic use , Neurons/enzymology , Neurons/metabolism , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , gamma-Aminobutyric Acid/metabolism
8.
CPT Pharmacometrics Syst Pharmacol ; 13(5): 823-836, 2024 05.
Article in English | MEDLINE | ID: mdl-38440939

ABSTRACT

Ciprofol (HSK3486) is a newly developed, highly selective γ-aminobutyric acid-A (GABAA) receptor potentiator that is recently approved for a new indication of sedation for patients in the intensive care unit (ICU) in China. This analysis aimed to characterize the population pharmacokinetics (PopPKs) of ciprofol and evaluate the relationship of exposure with hypotension in mechanically ventilated patients in the ICU. A total of 462 subjects with 3918 concentration measurements from two clinical trials of mechanically ventilated patients in the ICU, four clinical trials of elective surgical patients, and six clinical trials of healthy subjects were used in the PopPK analysis. Exposure-safety relationship for hypotension was evaluated based on the data gathered from 112 subjects in two clinical trials of mechanically ventilated patients in the ICU. Ciprofol pharmacokinetics (PKs) was adequately described by a three-compartment linear disposition model with first-order elimination. Body weight, age, sex, blood sampling site (vein vs. arterial), study design (long-term infusion vs. short-term infusion), and patient population (ICU vs. non-ICU) were identified as statistically significant covariates on the PKs of ciprofol. Within the exposure range of the mechanically ventilated ICU patient population, no meaningful association was observed between ciprofol exposure and the incidence of hypotension. These results support the dosing regimen currently used in mechanically ventilated patients in the ICU.


Subject(s)
Hypnotics and Sedatives , Intensive Care Units , Respiration, Artificial , Humans , Male , Middle Aged , Female , Adult , Aged , Hypnotics and Sedatives/pharmacokinetics , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/adverse effects , Hypotension/chemically induced , Young Adult , GABA-A Receptor Agonists/pharmacokinetics , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Agonists/adverse effects
9.
Brain Inj ; 27(11): 1320-9, 2013.
Article in English | MEDLINE | ID: mdl-23924270

ABSTRACT

INTRODUCTION: Zolpidem is a non-benzodiazepine drug used for the therapy of insomnia, which has selectivity for stimulating the effect of GABA-A receptors. Recently, a paradoxical arousing effect of zolpidem in patients with severe brain damage has been repeatedly reported. METHODS: A placebo-controlled magnetic resonance study was conducted to evaluate its effect on BOLD and metabolites spectral signals in a patient with severe brain injuries and an age-matched healthy volunteer. A multi-modal analysis was used to assess aspects in the pharmacologically-induced changes in the resting-state brain metabolism. RESULTS: A significantly increased BOLD signal was transiently localized in the left frontal cortices, bilateral anterior cingulated areas, left thalamus and right head of the caudate nucleus. The healthy subject showed a deactivation of the frontal, parietal and temporal cortices. BOLD signal changes were found to significantly correlate with concentrations of extravascular metabolites in the left frontal cortex. It is discussed that, when zolpidem attaches to modified GABA receptors of neurodormant brain cells, brain activation is induced. This might explain the significant correlations of BOLD signal changes and proton-MRS metabolites in this patient after zolpidem. CONCLUSION: It was concluded that proton-MRS and BOLD signal assessment could be used to study zolpidem-induced metabolic modulation in a resting state.


Subject(s)
GABA-A Receptor Agonists/therapeutic use , Oxygen/blood , Persistent Vegetative State/blood , Persistent Vegetative State/physiopathology , Pyridines/therapeutic use , Stroke/blood , Stroke/physiopathology , Wakefulness/drug effects , Adult , Female , GABA-A Receptor Agonists/pharmacokinetics , Hemodynamics , Humans , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Persistent Vegetative State/drug therapy , Pyridines/pharmacokinetics , Stroke/complications , Stroke/drug therapy , Treatment Outcome , Zolpidem
10.
Clin Exp Pharmacol Physiol ; 39(1): 9-12, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21985609

ABSTRACT

1. Our objective was to evaluate a possible pharmacokinetic interaction between zolpidem and fluvoxamine in healthy volunteers. 2. The study consisted of two periods: Period 1 (reference), when each volunteer received a single dose of 5 mg zolpidem; and Period 2 (test), when each volunteer received a single dose of 5 mg zolpidem and 100 mg fluvoxamine. Between the two periods, the subjects were treated for 6 days with a single daily dose of 100 mg fluvoxamine. 3. Pharmacokinetic parameters of zolpidem given in each treatment period were calculated using non-compartmental analysis and the data from two periods were compared to determine statistically significant differences. 4. In the two periods of treatments, the mean peak plasma concentrations (C(max)) were 56.4 ± 25.6 ng/mL (zolpidem alone) and 67.3 ± 25.8 ng/mL (zolpidem after pretreatment with fluvoxamine). The t(max), times taken to reach C(max), were 0.83 ± 0.44 and 1.26 ± 0.74 h, respectively, and the total areas under the curve (AUC(0-∞)) were 200.9 ± 116.8 and 512.0 ± 354.6 ng h/mL, respectively. The half-life of zolpidem was 2.24 ± 0.81 h when given alone and 4.99 ± 2.92 h after pretreatment with fluvoxamine. 5. Fluvoxamine interacts with zolpidem in healthy volunteers and increases its exposure by approximately 150%. The experimental data showed the pharmacokinetic interaction between zolpidem and fluvoxamine, and suggest that the observed interaction might be clinically significant, but its relevance has to be confirmed.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Fluvoxamine/pharmacology , GABA-A Receptor Agonists/pharmacokinetics , Hypnotics and Sedatives/pharmacokinetics , Pyridines/pharmacokinetics , Selective Serotonin Reuptake Inhibitors/pharmacology , Adult , Antidepressive Agents, Second-Generation/adverse effects , Biological Availability , Cross-Over Studies , Drug Interactions , Fluvoxamine/adverse effects , GABA-A Receptor Agonists/adverse effects , GABA-A Receptor Agonists/blood , Half-Life , Humans , Hypnotics and Sedatives/adverse effects , Hypnotics and Sedatives/blood , Male , Metabolic Clearance Rate/drug effects , Pyridines/adverse effects , Pyridines/blood , Selective Serotonin Reuptake Inhibitors/adverse effects , Young Adult , Zolpidem
11.
Xenobiotica ; 42(4): 355-62, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22106962

ABSTRACT

Oxidative deamination of the GABA(A) partial agonist CP-409,092 and sumatriptan represents a major metabolic pathway and seems to play an important role for the clearance of these two compounds. Similar to sumatriptan, human mitochondrial incubations with deprenyl and clorgyline, probe inhibitors of monoamine oxidase B and monoamine oxidase A (MAO-B and MAO-A), respectively, showed that CP-409,092 was metabolized to a large extent by the enzyme MAO-A. The metabolism of CP-409,092 and sumatriptan was therefore studied in human liver mitochondria and in vitro intrinsic clearance (CL(int)) values were determined and compared to the corresponding in vivo oral clearance (CL(PO)) values. The overall objective was to determine whether an in vitro-in vivo correlation (IVIVC) could be described for compounds cleared by MAO-A. The intrinsic clearance, CL(int), of CP-409,092 was approximately 4-fold greater than that of sumatriptan (CL(int), values were calculated as 0.008 and 0.002 ml/mg/min for CP-409,092 and sumatriptan, respectively). A similar correlation was observed from the in vivo metabolic data where the unbound oral clearance, CL(u)(PO), values in humans were calculated as 724 and 178 ml/min/kg for CP-409,092 and sumatriptan, respectively. The present work demonstrates that it is possible to predict in vivo metabolic clearance from in vitro metabolic data for drugs metabolized by the enzyme monoamine oxidase.


Subject(s)
Anilides/pharmacokinetics , GABA-A Receptor Agonists/pharmacokinetics , Indoles/pharmacokinetics , Monoamine Oxidase/metabolism , Sumatriptan/pharmacokinetics , Clorgyline/pharmacology , Drug Interactions , Drug Partial Agonism , Humans , In Vitro Techniques , Kinetics , Metabolic Clearance Rate , Mitochondria, Liver/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Selegiline/pharmacology
12.
Gynecol Endocrinol ; 27(12): 1087-93, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21190418

ABSTRACT

Animal studies suggest regulatory effects on the hypothalamic-pituitary-gonad axis by allopregnanolone, an endogenous gamma-aminobutyric acid A (GABA(A)) receptor agonist. Elevated levels of allopregnanolone in women with hypothalamic amenorrhea have been seen. Isoallopregnanolone is an isomer to allopregnanolone, but without GABA(A) receptor effects. The purpose of this study was to investigate effects of allopregnanolone and isoallopregnanolone on gonadotropin levels in healthy women of fertile age. Ten women were given allopregnanolone and five women isoallopregnanolone intravenously in follicular phase. Repeated blood samples were drawn during the test day. Main outcomes were changes in serum levels of follicle-stimulating hormone (FSH), luteinising hormone (LH), oestradiol, and progesterone. Serum-FSH decreased between 5 and 105 min after the allopregnanolone injection (F(16,144)=2.18, p=0.008). Serum-LH was reduced between 5 and 35 min following the allopregnanolone injection (F(16,144)=2.63, p=0.001). Serum-oestradiol and -progesterone were not significantly changed after allopregnanolone injections. No effect on gonadotropin levels were seen after administration of isoallopregnanolone. Allopregnanolone reduces FSH and LH levels in women and the effect might be mediated via a specific GABA(A) receptor activation since isoallopregnanolone lacked this effect. Although the number of women was small, the results suggest a regulatory mechanism on the hypothalamic-pituitary-gonadal axis by allopregnanolon.


Subject(s)
Gonadotropins/blood , Pregnanolone/pharmacology , Adult , Down-Regulation/drug effects , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Follicular Phase/blood , Follicular Phase/drug effects , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Agonists/pharmacokinetics , GABA-A Receptor Agonists/pharmacology , Humans , Injections, Intravenous , Luteinizing Hormone/blood , Pilot Projects , Pregnanolone/administration & dosage , Pregnanolone/analogs & derivatives , Pregnanolone/pharmacokinetics , Progesterone/blood , Time Factors , Young Adult
13.
Clin Pharmacokinet ; 59(1): 111-120, 2020 01.
Article in English | MEDLINE | ID: mdl-31338688

ABSTRACT

BACKGROUND: SAGE-217, a novel γ-aminobutyric acid A (GABAA) receptor positive allosteric modulator, was evaluated in phase I, double-blind, placebo-controlled, single ascending dose (SAD) and multiple ascending dose (MAD) studies to assess the safety and pharmacokinetics (PK) of SAGE-217 following administration as an oral solution. METHODS: In the SAD study, subjects were randomized 6:2 to a single dose of SAGE-217 or placebo. Doses ranged from 0.25 to 66 mg across nine cohorts. In the MAD study, subjects were randomized 9:3 and received SAGE-217 (15, 30, or 35 mg) or placebo once daily for 7 days. In both studies, PK, maximum tolerated dose (MTD; against predetermined criteria), safety, and tolerability were assessed. RESULTS: A total of 108 healthy volunteers enrolled in the studies-72 subjects in the SAD study and 36 subjects in the MAD study. SAGE-217 was orally bioavailable, with a terminal-phase half-life of 16-23 h and a tmax of approximately 1 h. The MTDs for the oral solution of SAGE-217 in the SAD and MAD studies were determined to be 55 and 30 mg daily, respectively. In both studies, SAGE-217 was generally well tolerated, and no serious adverse events (SAEs) were reported. Most AEs were mild, dose-dependent, transient, occurred around the tmax, and related to drug pharmacology. CONCLUSIONS: SAGE-217 was generally well tolerated, and its PK profile was well characterized. Based on this profile, SAGE-217 has been advanced into multiple phase II clinical programs and pivotal studies of major depressive disorder and postpartum depression.


Subject(s)
Depressive Disorder, Major/drug therapy , GABA-A Receptor Agonists/pharmacokinetics , Pharmacology, Clinical/methods , Pregnanes/pharmacokinetics , Pyrazoles/pharmacokinetics , Administration, Oral , Adult , Allosteric Regulation , Depression, Postpartum/epidemiology , Depression, Postpartum/physiopathology , Depression, Postpartum/psychology , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Agonists/therapeutic use , Healthy Volunteers , Humans , Male , Middle Aged , Placebos/administration & dosage , Pregnanes/administration & dosage , Pregnanes/therapeutic use , Pyrazoles/administration & dosage , Pyrazoles/therapeutic use , Safety
14.
Nat Commun ; 11(1): 4929, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33004789

ABSTRACT

Non-invasive, molecularly-specific, focal modulation of brain circuits with low off-target effects can lead to breakthroughs in treatments of brain disorders. We systemically inject engineered ultrasound-controllable drug carriers and subsequently apply a novel two-component Aggregation and Uncaging Focused Ultrasound Sequence (AU-FUS) at the desired targets inside the brain. The first sequence aggregates drug carriers with millimeter-precision by orders of magnitude. The second sequence uncages the carrier's cargo locally to achieve high target specificity without compromising the blood-brain barrier (BBB). Upon release from the carriers, drugs locally cross the intact BBB. We show circuit-specific manipulation of sensory signaling in motor cortex in rats by locally concentrating and releasing a GABAA receptor agonist from ultrasound-controlled carriers. Our approach uses orders of magnitude (1300x) less drug than is otherwise required by systemic injection and requires very low ultrasound pressures (20-fold below FDA safety limits for diagnostic imaging). We show that the BBB remains intact using passive cavitation detection (PCD), MRI-contrast agents and, importantly, also by sensitive fluorescent dye extravasation and immunohistochemistry.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Diseases/drug therapy , Drug Carriers/radiation effects , GABA-A Receptor Agonists/administration & dosage , Ultrasonography, Interventional/methods , Animals , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/radiation effects , Dose-Response Relationship, Radiation , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Female , GABA-A Receptor Agonists/pharmacokinetics , Humans , Magnetic Resonance Imaging , Models, Animal , Muscimol/administration & dosage , Muscimol/pharmacokinetics , Rats , Stereotaxic Techniques , Ultrasonic Waves
16.
Drug Test Anal ; 11(7): 1076-1082, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30912258

ABSTRACT

BACKGROUND: Oral fluid zolpidem detection in the settings of drug-facilitated crime and roadside drug testing indicates recent zolpidem intake. Zolpidem pharmacokinetics in classical biological matrices such as blood and urine have been described; however, reports of such data based on oral fluids are limited. OBJECTIVE: The aim of this study is to describe the pharmacokinetics of zolpidem and its major metabolite zolpidem phenyl-4-carboxylic acid (ZPCA) in oral fluids after intake. METHODS: Ten milligrams of zolpidem tartrate tablets were orally administered to 14 volunteers, and oral fluid samples were collected at various times up to 72 hours and analyzed via liquid chromatography-tandem mass spectrometry (LC-MS/MS) with post-column reagent addition. RESULTS: Both zolpidem and ZPCA could be detected in oral fluid after 1 hour and were rapidly eliminated, with half-lives of 2.77 ± 0.71 hours and 5.11 ± 0.67 hours, respectively. Maximum zolpidem concentrations (36.73 ± 10.89 ng/mL) occurred at 2 ± 0.52 hours, and maximum ZPCA concentrations (0.28 ± 0.16 ng/mL) occurred at 2 ± 0.37 hours. Zolpidem/ZPCA ratios decreased after zolpidem intake, an observation that might be helpful for determining the time of drug use. CONCLUSION: The results showed that the measurement of zolpidem in oral fluid can be used for the non-invasive monitoring of zolpidem consumption and misuse in drug-facilitated crime and roadside drug testing settings.


Subject(s)
GABA-A Receptor Agonists/pharmacokinetics , Pyridines/pharmacokinetics , Saliva/metabolism , Sleep Aids, Pharmaceutical/pharmacokinetics , Zolpidem/pharmacokinetics , Administration, Oral , Adult , Chromatography, Liquid , Female , GABA-A Receptor Agonists/administration & dosage , Humans , Male , Pyridines/administration & dosage , Sleep Aids, Pharmaceutical/administration & dosage , Tandem Mass Spectrometry , Zolpidem/administration & dosage
17.
Neurology ; 92(15): e1786-e1795, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30877186

ABSTRACT

OBJECTIVE: The objective of this phase 2a study was to assess the activity of PF-06372865, a positive allosteric modulator (PAM) of α2/3/5 subunit-containing GABAA receptors with minimal activity at α1-containing receptors, which are believed to mediate many of the adverse events associated with benzodiazepines, in the epilepsy photosensitivity model as a proof-of-principle of efficacy. METHODS: Seven participants with a photoparoxysmal response to intermittent photic stimulation (IPS) at baseline were randomized in a double-blind, 4-period cross-over study examining single doses of 17.5 and 52.5 mg PF-06372865, 2 mg lorazepam (active control), and placebo. Standardized photosensitivity ranges (SPRs) to IPS were recorded at screening, predose, and 1, 2, 4, and 6 hours postdose. The primary endpoint was the average least squares mean change in the SPR in the participant's most sensitive eye condition, across all time points. RESULTS: Both doses of PF-06372865 produced a marked and statistically significant mean reduction in SPR compared to placebo, which was similar in degree to lorazepam. There was complete suppression of SPR in 6/7 participants following PF-06372865 or lorazepam administration. PF-06372865 was safe and well-tolerated. CONCLUSION: PF-06372865 demonstrated highly robust efficacy. This demonstrates anticonvulsant activity of a novel α2/3/5-subtype selective GABAA PAM in humans. Further study of the antiepileptic properties of PF-06372865 is warranted. CLINICALTRIALSGOV IDENTIFIER: NCT02564029. CLASSIFICATION OF EVIDENCE: This study provides Class II evidence that for people with a stable photoparoxysmal response to intermittent photic stimulation, PF-06372865 reduces the SPR.


Subject(s)
Anticonvulsants/therapeutic use , Epilepsy, Reflex/drug therapy , GABA-A Receptor Agonists/therapeutic use , Imidazoles/therapeutic use , Pyridazines/therapeutic use , Adolescent , Adult , Anticonvulsants/adverse effects , Anticonvulsants/pharmacokinetics , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , GABA Modulators/therapeutic use , GABA-A Receptor Agonists/adverse effects , GABA-A Receptor Agonists/pharmacokinetics , Humans , Imidazoles/adverse effects , Imidazoles/pharmacokinetics , Lorazepam/therapeutic use , Male , Middle Aged , Pyridazines/adverse effects , Pyridazines/pharmacokinetics , Treatment Outcome , Young Adult
18.
Neuropharmacology ; 137: 332-343, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29778948

ABSTRACT

HZ-166 has previously been characterized as an α2,3-selective GABAA receptor modulator with anticonvulsant, anxiolytic, and anti-nociceptive properties but reduced motor effects. We discovered a series of ester bioisosteres with reduced metabolic liabilities, leading to improved efficacy as anxiolytic-like compounds in rats. In the present study, we evaluated the anticonvulsant effects KRM-II-81 across several rodent models. In some models we also evaluated key structural analogs. KRM-II-81 suppressed hyper-excitation in a network of cultured cortical neurons without affecting the basal neuronal activity. KRM-II-81 was active against electroshock-induced convulsions in mice, pentylenetetrazole (PTZ)-induced convulsions in rats, elevations in PTZ-seizure thresholds, and amygdala-kindled seizures in rats with efficacies greater than that of diazepam. KRM-II-81 was also active in the 6 Hz seizure model in mice. Structural analogs of KRM-II-81 but not the ester, HZ-166, were active in all models in which they were evaluated. We further evaluated KRM-II-81 in human cortical epileptic tissue where it was found to significantly-attenuate picrotoxin- and AP-4-induced increases in firing rate across an electrode array. These molecules generally had a wider margin of separation in potencies to produce anticonvulsant effects vs. motor impairment on an inverted screen test than did diazepam. Ester bioisosters of HZ-166 are thus presented as novel agents for the potential treatment of epilepsy acting via selective positive allosteric amplification of GABAA signaling through α2/α3-containing GABA receptors. The in vivo data from the present study can serve as a guide to dosing parameters that predict engagement of central GABAA receptors.


Subject(s)
Anticonvulsants/pharmacology , GABA-A Receptor Agonists/pharmacology , Oxazoles/pharmacology , Seizures/drug therapy , Action Potentials/drug effects , Animals , Anticonvulsants/chemistry , Anticonvulsants/pharmacokinetics , Benzodiazepines/chemistry , Benzodiazepines/pharmacokinetics , Benzodiazepines/pharmacology , Biological Availability , Child , Diazepam/pharmacology , Disease Models, Animal , Drug Resistant Epilepsy/drug therapy , Drug Resistant Epilepsy/physiopathology , Female , GABA-A Receptor Agonists/chemistry , GABA-A Receptor Agonists/pharmacokinetics , Humans , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Male , Mice , Oxazoles/chemistry , Oxazoles/pharmacokinetics , Random Allocation , Rats, Sprague-Dawley , Seizures/physiopathology , Tissue Culture Techniques
19.
J Med Chem ; 60(18): 7810-7819, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28753313

ABSTRACT

Certain classes of neuroactive steroids (NASs) are positive allosteric modulators (PAM) of synaptic and extrasynaptic GABAA receptors. Herein, we report new SAR insights in a series of 5ß-nor-19-pregnan-20-one analogues bearing substituted pyrazoles and triazoles at C-21, culminating in the discovery of 3α-hydroxy-3ß-methyl-21-(4-cyano-1H-pyrazol-1'-yl)-19-nor-5ß-pregnan-20-one (SAGE-217, 3), a potent GABAA receptor modulator at both synaptic and extrasynaptic receptor subtypes, with excellent oral DMPK properties. Compound 3 has completed a phase 1 single ascending dose (SAD) and multiple ascending dose (MAD) clinical trial and is currently being studied in parallel phase 2 clinical trials for the treatment of postpartum depression (PPD), major depressive disorder (MDD), and essential tremor (ET).


Subject(s)
Allosteric Regulation/drug effects , GABA-A Receptor Agonists/chemistry , GABA-A Receptor Agonists/pharmacology , Pregnanolone/analogs & derivatives , Receptors, GABA-A/metabolism , Animals , Depression, Postpartum/drug therapy , Depressive Disorder, Major/drug therapy , Female , GABA-A Receptor Agonists/pharmacokinetics , Mice , Pregnanolone/chemistry , Pregnanolone/pharmacokinetics , Pregnanolone/pharmacology , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Rats
20.
Eur J Pharmacol ; 791: 433-443, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27639297

ABSTRACT

We have synthesized and characterized MP-III-022 ((R)-8-ethynyl-6-(2-fluorophenyl)-N,4-dimethyl-4H-benzo[f]imidazo[1,5-a][1,4]diazepine-3-carboxamide) in vitro and in vivo as a binding- and efficacy-selective positive allosteric modulator of GABAA receptors containing the α5 subunit (α5GABAARs). By approximation of the electrophysiological responses which the estimated free rat brain concentrations can induce, we demonstrated that convenient systemic administration of MP-III-022 in the dose range 1-10mg/kg may result in a selective potentiation, over a wide range from mild to moderate to strong, of α5ßγ2 GABAA receptors. For eliciting a comparable range of potentiation, the widely studied parent ligand SH-053-2'F-R-CH3 containing an ester moiety needs to be administered over a much wider dose range (10-200mg/kg), but at the price of activating non-α5 GABAARs as well as the desired α5GABAARs at the highest dose. At the dose of 10mg/kg, which elicits a strong positive modulation of α5GABAARs, MP-III-022 caused mild, but significant muscle relaxation, while at doses 1-10mg/kg was devoid of ataxia, sedation or an influence on the anxiety level, characteristic for non-selective benzodiazepines. As an amide compound with improved stability and kinetic properties, MP-III-022 may represent an optimized tool to study the influence of α5GABAARs on the neuronal pathways related to CNS disorders such as schizophrenia, Alzheimer's disease, Down syndrome or autism.


Subject(s)
Diazepam/analogs & derivatives , GABA-A Receptor Agonists/pharmacology , Imidazoles/pharmacology , Receptors, GABA-A/metabolism , Amphetamine/pharmacology , Animals , Diazepam/metabolism , Diazepam/pharmacokinetics , Diazepam/pharmacology , Drug Stability , Electrophysiological Phenomena/drug effects , GABA-A Receptor Agonists/metabolism , GABA-A Receptor Agonists/pharmacokinetics , HEK293 Cells , Hand Strength , Humans , Imidazoles/metabolism , Imidazoles/pharmacokinetics , Kinetics , Locomotion/drug effects , Male , Maze Learning/drug effects , Rats , Rats, Wistar , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL