Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci ; 36(49): 12312-12320, 2016 12 07.
Article in English | MEDLINE | ID: mdl-27927951

ABSTRACT

Alpha-5 gamma-aminobutyric acid type A receptors (α5-GABAARs) are located extrasynaptically, regulate neuronal excitability through tonic inhibition, and are fundamentally important for processes such as plasticity and learning. For example, pharmacological blockade of α5-GABAAR in mice with ischemic stroke improved recovery of function by normalizing exaggerated perilesional α5-GABAAR-dependent tonic inhibition. S44819 is a novel competitive selective antagonist of the α5-GABAAR at the GABA-binding site. Pharmacological modulation of α5-GABAAR-mediated tonic inhibition has never been investigated in the human brain. Here, we used transcranial magnetic stimulation (TMS) to test the effects of a single oral dose of 50 and 100 mg of S44819 on electromyographic (EMG) and electroencephalographic (EEG) measures of cortical excitability in 18 healthy young adults in a randomized, double-blinded, placebo-controlled, crossover phase I study. A dose of 100 mg, but not 50 mg, of S44819 decreased active motor threshold, the intensity needed to produce a motor evoked potential of 0.5 mV, and the amplitude of the N45, a GABAAergic component of the TMS-evoked EEG response. The peak serum concentration of 100 mg S44819 correlated directly with the decrease in N45 amplitude. Short-interval intracortical inhibition, a TMS-EMG measure of synaptic GABAAergic inhibition, and other components of the TMS-evoked EEG response remained unaffected. These findings provide first time evidence that the specific α5-GABAAR antagonist S44819 reached human cortex to impose an increase in cortical excitability. These data warrant further development of S44819 in a human clinical trial to test its efficacy in enhancing recovery of function after ischemic stroke. SIGNIFICANCE STATEMENT: The extrasynaptic α-5 gamma-aminobutyric acid type A receptor (α5-GABAAR) regulates neuronal excitability through tonic inhibition in the mammalian brain. Tonic inhibition is important for many fundamental processes such as plasticity and learning. Pharmacological modulation of α5-GABAAR-mediated tonic inhibition has never been investigated in the human brain. This study demonstrates that S44819, a selective α5-GABAAR antagonist, increases cortical excitability in healthy human subjects, as indicated by specific markers of transcranial magnetic stimulation-induced muscle and brain responses measured by electromyography and electroencephalography. Our findings imply that tonic inhibition in human cortex can be modified effectively and that this modification can be quantified with noninvasive brain stimulation methods. The actions of S44819 may be suitable to improve plasticity and learning.


Subject(s)
Brain/drug effects , GABA-A Receptor Antagonists/pharmacology , Receptors, GABA-A/drug effects , Transcranial Magnetic Stimulation/methods , Adult , Cross-Over Studies , Double-Blind Method , Electroencephalography/drug effects , Electromyography/drug effects , Evoked Potentials, Motor/drug effects , Healthy Volunteers , Humans , Male , Young Adult
2.
Hum Mol Genet ; 21(5): 1145-54, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22108178

ABSTRACT

Clomiphene citrate is the most used drug for the treatment of female infertility, a common condition in western societies and developing countries. Despite dose escalation, up to 30% of women do not respond. Since clomiphene shares structural similarities with tamoxifen, which is predominantly bioactivated by the polymorphic cytochrome P450 (CYP) 2D6, we systematically explored clomiphene metabolism and action in vitro and in vivo by pharmacogenetic, -kinetic and -dynamic investigations. Human liver microsomes were incubated with clomiphene citrate and nine metabolites were identified by mass spectrometry and tested at the oestrogen receptor for their antagonistic capacity. (E)-4-hydroxyclomiphene and (E)-4-hydroxy-N-desethylclomiphene showed strongest inhibition of the oestrogen receptor activity with 50% inhibitory concentrations of 2.5 and 1.4 nm, respectively. CYP2D6 has been identified as the major enzyme involved in their formation using recombinant CYP450 isozymes as confirmed by inhibition experiments with CYP monoclonal antibodies. We correlated the CYP2D6 genotype of 30 human liver donors with the microsomal formation rate of active metabolites and observed a strong gene-dose effect. A healthy female volunteer study confirmed our in vitro data that the CYP2D6 polymorphism substantially determines the formation of the active clomiphene metabolites. Comparison of the C(max) of (E)-4-hydroxyclomiphene and (E)-4-hydroxy-N-desethylclomiphene showed 8 and 12 times lower concentrations in subjects with non-functional CYP2D6 alleles. Our results highlight (E)-4-hydroxyclomiphene and (E)-4-hydroxy-N-desethylclomiphene as the active clomiphene metabolites, the formation of which strongly depends on the polymorphic CYP2D6 enzyme. Our data provide first evidence of a biological rationale for the variability in the response to clomiphene treatment.


Subject(s)
Clomiphene/metabolism , Clomiphene/pharmacology , Cytochrome P-450 CYP2D6/genetics , Estrogen Antagonists/metabolism , Estrogen Antagonists/pharmacology , Polymorphism, Genetic , Receptors, Estrogen/antagonists & inhibitors , Biotransformation , Clomiphene/analogs & derivatives , Clomiphene/chemistry , Cytochrome P-450 CYP2D6/metabolism , Female , Genotype , Humans , Microsomes, Liver/metabolism , Molecular Structure , Receptors, Estrogen/metabolism , Recombinant Proteins/metabolism
3.
J Clin Pharmacol ; 51(1): 93-101, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20308689

ABSTRACT

Tigecycline belongs to a new class of tetracyclines, the glycylcyclines, less than 20% of which is metabolized in the liver. Twenty-five patients with cirrhosis with varying degrees of functional hepatic reserve (Child-Pugh A, n = 10; B, n = 10; C, n = 5) and 23 healthy adults, matched by age, sex, weight, and smoking habits, received 100 mg of tigecycline infused intravenously over 60 minutes. Serum and urine samples were collected up to 120 hours after dosing. Pharmacokinetic data were derived using noncompartmental methods. The most common treatment-emergent adverse events in healthy volunteers were nausea (56.5%), vomiting (21.7%), and headache (21.7%) and in the patients with cirrhosis, albuminuria (12%). Mean (± 1 SD) tigecycline clearance values were 29.8 ± 11.3 L/h in healthy subjects and 31.2 ± 13.9 L/h (Child-Pugh A), 22.1 ± 9.3 L/h (Child-Pugh B), and 13.5 ± 2.7 L/h (Child-Pugh C) in the patients. A single intravenous dose of tigecycline 100 mg was safe and well-tolerated in patients with cirrhosis with varying degrees of hepatic functional reserve. No adjustment of tigecycline maintenance dosage is warranted in patients with compensated or moderately decompensated cirrhosis; doses should be reduced by 50%, to 25 mg, every 12 hours in patients with severely decompensated disease.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Liver Cirrhosis/complications , Minocycline/analogs & derivatives , Adult , Anti-Bacterial Agents/adverse effects , Case-Control Studies , Female , Humans , Infusions, Intravenous , Liver Cirrhosis/pathology , Male , Middle Aged , Minocycline/adverse effects , Minocycline/pharmacokinetics , Severity of Illness Index , Tigecycline
4.
J Clin Pharmacol ; 51(4): 594-602, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20484614

ABSTRACT

This nonrandomized, fixed-sequence, 2-period crossover study investigated potential pharmacokinetic interactions between the phosphodiesterase 4 inhibitor roflumilast, currently in clinical development for the treatment of chronic obstructive pulmonary disease, and the histamine 2 agonist cimetidine. Participants received roflumilast, 500 µg once daily, on days 1 and 13. Cimetidine, 400 mg twice daily, was administered from days 6 to 16. Pharmacokinetic analysis of roflumilast and its active metabolite roflumilast N-oxide was performed, and the ratio of geometric means for roflumilast alone and concomitantly with steady-state cimetidine was calculated. The effect of cimetidine on the total PDE4 inhibitory activity (tPDE4i; total exposure to roflumilast and roflumilast N-oxide) was also calculated. Coadministration of steady-state cimetidine increased mean tPDE4i of roflumilast and roflumilast N-oxide by about 47%. The maximum plasma concentration (C(max)) of roflumilast increased by about 46%, with no effect on C(max) of roflumilast N-oxide. The increase in tPDE4i of roflumilast and roflumilast N-oxide following coadministration with cimetidine was mainly due to the inhibitory effect of cimetidine on cytochrome P450 (CYP) isoenzymes CYP1A2, CYP3A, and CYP2C19. These moderate changes indicate that dose adjustment of roflumilast is not required when coadministered with a weak inhibitor of CYP1A2, CYP3A, and CYP2C19, such as cimetidine.


Subject(s)
Aminopyridines/administration & dosage , Benzamides/administration & dosage , Cimetidine/administration & dosage , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/administration & dosage , Phosphodiesterase 4 Inhibitors/administration & dosage , Adult , Aminopyridines/adverse effects , Aminopyridines/pharmacokinetics , Benzamides/adverse effects , Benzamides/pharmacokinetics , Cimetidine/adverse effects , Cimetidine/pharmacokinetics , Cross-Over Studies , Cyclopropanes/administration & dosage , Cyclopropanes/adverse effects , Cyclopropanes/pharmacokinetics , Drug Interactions , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Male , Phosphodiesterase 4 Inhibitors/adverse effects , Phosphodiesterase 4 Inhibitors/pharmacokinetics
5.
J Clin Pharmacol ; 49(4): 389-97, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19318692

ABSTRACT

This nonrandomized, fixed-sequence, 3-period study investigated potential pharmacokinetic interactions between the leukotriene receptor antagonist montelukast, approved for the treatment of asthma, and roflumilast, an oral, once-daily phosphodiesterase 4 inhibitor in clinical development for asthma and chronic obstructive pulmonary disease. Pharmacokinetic interactions are of interest because both drugs may be coadministered and share a common metabolic pathway via cytochrome P450 3A. Single-dose montelukast (10 mg, po) was administered alone in period 1, followed by repeated once-daily roflumilast alone (500 microg, po) for 12 days (period 2). In period 3, 500 microg qd roflumilast was coadministered with 10 mg qd montelukast for 8 days. Different pharmacokinetic parameters were evaluated for montelukast alone, for steady-state roflumilast and its pharmacologically active metabolite roflumilast N-oxide alone, for single-dose montelukast when coadministered with steady-state roflumilast, and for steady-state roflumilast and its N-oxide metabolite when coadministered with steady-state montelukast. The AUC and Cmax of montelukast were modestly increased by 9% and 8%, respectively, when single-dose montelukast was coadministered with steady-state roflumilast. The pharmacokinetics of roflumilast and roflumilast N-oxide in steady state remained unchanged when repeat-dose montelukast was coadministered at steady-state. Concomitant administration of both drugs was well tolerated. These findings suggest that no dose adjustment is warranted for either drug when roflumilast and montelukast are coadministered.


Subject(s)
Acetates/pharmacokinetics , Aminopyridines/pharmacokinetics , Anti-Asthmatic Agents/pharmacokinetics , Benzamides/pharmacokinetics , Leukotriene Antagonists/pharmacokinetics , Phosphodiesterase Inhibitors/pharmacokinetics , Quinolines/pharmacokinetics , Acetates/administration & dosage , Acetates/adverse effects , Administration, Oral , Adolescent , Adult , Aminopyridines/administration & dosage , Aminopyridines/adverse effects , Aminopyridines/blood , Aminopyridines/metabolism , Anti-Asthmatic Agents/administration & dosage , Anti-Asthmatic Agents/adverse effects , Benzamides/administration & dosage , Benzamides/adverse effects , Benzamides/blood , Benzamides/metabolism , Cyclopropanes/administration & dosage , Cyclopropanes/adverse effects , Cyclopropanes/blood , Cyclopropanes/metabolism , Cyclopropanes/pharmacokinetics , Drug Interactions , Drug Therapy, Combination , Humans , Leukotriene Antagonists/administration & dosage , Leukotriene Antagonists/adverse effects , Male , Middle Aged , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/adverse effects , Quinolines/administration & dosage , Quinolines/adverse effects , Sulfides
6.
Clin Pharmacokinet ; 47(5): 343-9, 2008.
Article in English | MEDLINE | ID: mdl-18399715

ABSTRACT

BACKGROUND AND OBJECTIVES: Cytochrome P450 (CYP) 3A4 isoenzyme has been identified in vitro as the key enzyme to metabolize desisobutyryl-ciclesonide (des-CIC), the pharmacologically active metabolite of the inhaled corticosteroid ciclesonide. This pharmacokinetic drug-drug interaction study was conducted to confirm this major metabolic pathway in vivo by using the strong CYP3A4 inhibitor ketoconazole, and to assess the effect of ketoconazole on the pharmacokinetics of ciclesonide and des-CIC. METHODS: Fourteen healthy adults participated in this open-label, nonrandomized, fixed sequence, two-period, repeated-dose pharmacokinetic study. During the first 7-day treatment period, the subjects orally inhaled ciclesonide 320 microg once daily. During the second 7-day treatment period, the subjects continued with the same dose of orally inhaled ciclesonide and concomitantly received oral ketoconazole 400 mg once daily. Pharmacokinetic profiles for ciclesonide and des-CIC were obtained on day 7 of each study period. RESULTS: For the parent compound, ciclesonide, no changes in the pharmacokinetic parameter estimates--the area under the serum concentration-time curve during the dosage interval (AUC(tau)), maximum serum concentration (C(max)) and time to reach the C(max)--were observed. In contrast, the AUC(tau) and C(max) of des-CIC increased approximately 3.5-fold and 2-fold under the influence of the CYP3A4 inhibitor ketoconazole. CONCLUSIONS: The CYP3A4 pathway is the major pathway for biotransformation of the active metabolite of ciclesonide in humans. While elimination of des-CIC was reduced by strong CYP3A4 inhibitor coadministration in vivo, activation of the parent compound ciclesonide to des-CIC was not affected. Dose adjustment is not necessary when ciclesonide needs to be coadministered with ketoconazole, because the potency of an inhaled corticosteroid is mediated by topical concentrations in the lung and because ciclesonide has a very low potential to produce systemic adverse effects.


Subject(s)
Anti-Allergic Agents/pharmacokinetics , Antifungal Agents/pharmacology , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Ketoconazole/pharmacology , Pregnenediones/pharmacokinetics , Adult , Anti-Allergic Agents/adverse effects , Antifungal Agents/adverse effects , Area Under Curve , Biotransformation , Enzyme Inhibitors/adverse effects , Female , Humans , Ketoconazole/adverse effects , Male , Middle Aged , Pregnenediones/adverse effects
7.
J Clin Pharmacol ; 47(1): 26-36, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17192499

ABSTRACT

The dose-proportional, intraindividual, single- and repeated-dose pharmacokinetics of roflumilast, an oral, once-daily phosphodiesterase 4 inhibitor under investigation for chronic obstructive pulmonary disease and asthma, was investigated in healthy subjects. In an open, randomized, 2-period, 2-sequence crossover study, 15 subjects received immediate-release tablets of roflumilast 250 or 500 microg as single (day 1) and as repeated, once-daily doses for 8 days (days 5-12). Dose-adjusted point estimates and 90% confidence intervals of test (500 microg)/reference (250 microg) ratios for AUC and Cmax of roflumilast and its pharmacologically active N-oxide metabolite after single and repeated dosing were all within the standard equivalence acceptance range (0.80, 1.25) indicating dose proportionality. The pharmacokinetic properties of both roflumilast dosage forms provide clinically relevant evidence of predictable, intraindividual total (AUC) and maximum (Cmax) exposure of roflumilast and roflumilast N-oxide. Repeated oral dosing with roflumilast 250 and 500 microg once daily was well tolerated.


Subject(s)
Aminopyridines/pharmacokinetics , Benzamides/pharmacokinetics , Phosphodiesterase Inhibitors/pharmacokinetics , Administration, Oral , Aminopyridines/administration & dosage , Aminopyridines/blood , Analysis of Variance , Area Under Curve , Benzamides/administration & dosage , Benzamides/blood , Chromatography, High Pressure Liquid , Cross-Over Studies , Cyclopropanes/administration & dosage , Cyclopropanes/blood , Cyclopropanes/pharmacokinetics , Dose-Response Relationship, Drug , Drug Tolerance , Female , Humans , Linear Models , Male , Mass Spectrometry , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/blood , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...