Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
CPT Pharmacometrics Syst Pharmacol ; 7(10): 687-697, 2018 10.
Article in English | MEDLINE | ID: mdl-30091858

ABSTRACT

Low-dose oral S-ketamine is increasingly used in chronic pain therapy, but extensive cytochrome P450 (CYP) mediated metabolism makes it prone to pharmacokinetic drug-drug interactions (DDIs). In our study, concentration-time data from five studies were used to develop a semimechanistic model that describes the ticlopidine-mediated inhibition of S-ketamine biotransformation. A mechanistic model was implemented to account for reversible and time-dependent hepatic CYP2B6 inactivation by ticlopidine, which causes elevated S-ketamine exposure in vivo. A pharmacokinetic model was developed with gut wall and hepatic clearances for S-ketamine, its primary metabolite norketamine, and ticlopidine. Nonlinear mixed effects modeling approach was used (NONMEM version 7.3.0), and the final model was evaluated with visual predictive checks and the sampling-importance-resampling procedure. Our final model produces biologically plausible output and demonstrates that ticlopidine is a strong inhibitor of CYP2B6 mediated S-ketamine metabolism. Simulations from our model may be used to evaluate chronic pain therapy with S-ketamine.


Subject(s)
Analgesics/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , Ketamine/pharmacokinetics , Models, Biological , Ticlopidine/pharmacokinetics , Analgesics/pharmacology , Cross-Over Studies , Drug Interactions , Enzyme Inhibitors/pharmacology , Healthy Volunteers , Humans , Ketamine/pharmacology , Pain Management/methods , Placebos , Randomized Controlled Trials as Topic , Ticlopidine/pharmacology
2.
Clin Pharmacokinet ; 55(9): 1059-77, 2016 09.
Article in English | MEDLINE | ID: mdl-27028535

ABSTRACT

Ketamine is a phencyclidine derivative, which functions primarily as an antagonist of the N-methyl-D-aspartate receptor. It has no affinity for gamma-aminobutyric acid receptors in the central nervous system. Ketamine shows a chiral structure consisting of two optical isomers. It undergoes oxidative metabolism, mainly to norketamine by cytochrome P450 (CYP) 3A and CYP2B6 enzymes. The use of S-ketamine is increasing worldwide, since the S(+)-enantiomer has been postulated to be a four times more potent anesthetic and analgesic than the R(-)-enantiomer and approximately two times more effective than the racemic mixture of ketamine. Because of extensive first-pass metabolism, oral bioavailability is poor and ketamine is vulnerable to pharmacokinetic drug interactions. Sublingual and nasal formulations of ketamine are being developed, and especially nasal administration produces rapid maximum plasma ketamine concentrations with relatively high bioavailability. Ketamine produces hemodynamically stable anesthesia via central sympathetic stimulation without affecting respiratory function. Animal studies have shown that ketamine has neuroprotective properties, and there is no evidence of elevated intracranial pressure after ketamine dosing in humans. Low-dose perioperative ketamine may reduce opioid consumption and chronic postsurgical pain after specific surgical procedures. However, long-term analgesic effects of ketamine in chronic pain patients have not been demonstrated. Besides analgesic properties, ketamine has rapid-acting antidepressant effects, which may be useful in treating therapy-resistant depressive patients. Well-known psychotomimetic and cognitive adverse effects restrict the clinical usefulness of ketamine, even though fewer psychomimetic adverse effects have been reported with S-ketamine in comparison with the racemate. Safety issues in long-term use are yet to be resolved.


Subject(s)
Analgesics/pharmacokinetics , Anesthesia/methods , Ketamine/analogs & derivatives , Ketamine/pharmacokinetics , Pain/drug therapy , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Analgesics/administration & dosage , Analgesics/adverse effects , Analgesics/blood , Animals , Biological Availability , Central Nervous System/drug effects , Child , Cytochrome P-450 CYP2B6/metabolism , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Drug Administration Routes , Humans , Ketamine/administration & dosage , Ketamine/adverse effects , Ketamine/blood , Pain/prevention & control , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism
3.
Basic Clin Pharmacol Toxicol ; 111(5): 325-32, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22676424

ABSTRACT

Low-dose ketamine is currently used in several acute and chronic pain conditions as an analgesic. Ketamine undergoes extensive metabolism and is thus susceptible to drug-drug interactions. We examined the effect rifampicin, a well-known inducer of many cytochrome P450 (CYP) enzymes and transporters, on the pharmacokinetics of intravenous and oral S-ketamine in healthy volunteers. Eleven healthy volunteers were administered in randomized order 600 mg rifampicin or placebo orally for 6 days in a four-session paired cross-over study. On day 6, S-ketamine was administered intravenously (0.1 mg/kg) in the first part of the study and orally (0.3 mg/kg) in the second part. Plasma concentrations of ketamine and norketamine were measured up to 24 hr and behavioural and analgesic effects up to 12 hr. Rifampicin treatment decreased the mean area under the plasma ketamine concentration-time curve extrapolated to infinity (AUC (0-∞)) of intravenous and oral S-ketamine by 14% (p = 0.005) and 86% (p < 0.001), respectively. Rifampicin decreased greatly the peak plasma concentration of oral S-ketamine by 81% (p < 0.001), but shortened only moderately the elimination half-life of intravenous and oral S-ketamine. Rifampicin decreased the ratio of norketamine AUC (0-∞) to ketamine AUC (0-∞) after intravenous S-ketamine by 66%, (p < 0.001) but increased the ratio by 147% (p < 0.001) after the oral administration of S-ketamine. Rifampicin profoundly reduces the plasma concentrations of ketamine and norketamine after oral administration of S-ketamine, by inducing mainly its first-pass metabolism.


Subject(s)
Analgesics/pharmacokinetics , Enzyme Inhibitors/pharmacology , Ketamine/pharmacokinetics , Rifampin/pharmacology , Administration, Oral , Analgesics/administration & dosage , Analgesics/blood , Biological Availability , Biotransformation/drug effects , Cross-Over Studies , Drug Interactions , Enzyme Induction/drug effects , Female , Half-Life , Humans , Injections, Intravenous , Ketamine/administration & dosage , Ketamine/analogs & derivatives , Ketamine/blood , Male , Metabolic Clearance Rate , Psychomotor Performance/drug effects , Sleep Stages/drug effects
4.
Eur J Clin Pharmacol ; 68(6): 979-86, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22286159

ABSTRACT

PURPOSE: We examined the effect of grapefruit juice on the pharmacokinetics and pharmacodynamics of oral S-ketamine. METHODS: A randomized crossover open-label study design with two phases at an interval of 4 weeks was conducted in 12 healthy volunteers. Grapefruit juice or water was ingested 200 ml t.i.d. for 5 days. An oral dose of 0.2 mg/kg of S-ketamine was ingested on day 5 with 150 ml grapefruit juice or water. Plasma concentrations of ketamine and norketamine were determined for 24 h, and pharmacodynamic variables were recorded for 12 h. Noncompartmental methods were used to calculate pharmacokinetic parameters. RESULTS: Grapefruit juice increased the geometric mean value of the area under the plasma ketamine concentration-time curve(AUC0-∞) by 3.0-fold (range 2.4- to 3.6-fold; P<0.001), the peak plasma concentration (Cmax) by 2.1-fold (range 1.8- to 2.6-fold; P<0.001), and the elimination half-life by 24% (P<0.05) as compared to the water phase. The ratio of main metabolite norketamine to ketamine (AUCm/AUCp) was decreased by 57% (P<0.001) during the grapefruit phase.Self-rated relaxation was decreased (P<0.05) and the performance in the digit symbol substitution test was increased (P<0.05) after grapefruit juice, but other behavioral or analgesic effects were not affected. CONCLUSIONS: Grapefruit juice significantly increased the plasma concentrations of oral ketamine in healthy volunteers.Dose reductions of ketamine should be considered when using oral ketamine concomitantly with grapefruit juice.


Subject(s)
Beverages , Citrus paradisi , Food-Drug Interactions , Ketamine/pharmacokinetics , Adult , Aged , Aryl Hydrocarbon Hydroxylases/metabolism , Cross-Over Studies , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP3A/metabolism , Female , Humans , Ketamine/administration & dosage , Ketamine/analogs & derivatives , Ketamine/blood , Male , Middle Aged , Oxidoreductases, N-Demethylating/metabolism , Young Adult
5.
Fundam Clin Pharmacol ; 26(6): 743-50, 2012 Dec.
Article in English | MEDLINE | ID: mdl-21635359

ABSTRACT

Ketamine is an intravenous anaesthetic and analgesic agent but it can also be used orally as an adjuvant in the treatment of chronic pain. This study investigated the effect of the herbal antidepressant St John's wort, an inducer of cytochrome P450 3A4 (CYP3A4), on the pharmacokinetics and pharmacodynamics of oral S-ketamine. In a randomized cross-over study with two phases, 12 healthy subjects were pretreated with oral St John's wort or placebo for 14 days. On day 14, they were given an oral dose of 0.3 mg/kg of S-ketamine. Plasma concentrations of ketamine and norketamine were measured for 24 h and pharmacodynamic variables for 12 h. St John's wort decreased the mean area under the plasma concentration-time curve (AUC(0-∞)) of ketamine by 58% (P < 0.001) and decreased the peak plasma concentration (C(max)) of ketamine by 66% (P < 0.001) when compared with placebo. Mean C(max) of norketamine (the major metabolite of ketamine) was decreased by 23% (P = 0.002) and mean AUC(0-∞) of norketamine by 18% (P < 0.001) by St John's wort. There was a statistically significant linear correlation between the self-reported drug effect and C(max) of ketamine (r = 0.55; P < 0.01). St John's wort greatly decreased the exposure to oral S-ketamine in healthy volunteers. Although this decrease was not associated with significant changes in the analgesic or behavioural effects of ketamine in the present study, usual doses of S-ketamine may become ineffective if used concomitantly with St John's wort.


Subject(s)
Analgesics/blood , Antidepressive Agents/pharmacology , Hypericum/chemistry , Ketamine/analogs & derivatives , Plant Preparations/pharmacology , Adult , Analgesics/administration & dosage , Analgesics/adverse effects , Analgesics/pharmacology , Antidepressive Agents/administration & dosage , Antidepressive Agents/adverse effects , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Interactions , Female , Half-Life , Humans , Ketamine/administration & dosage , Ketamine/adverse effects , Ketamine/blood , Ketamine/pharmacology , Male , Neuropsychological Tests , Plant Preparations/administration & dosage , Plant Preparations/adverse effects , Psychomotor Performance/drug effects , Regression Analysis , Young Adult
6.
Eur J Pain ; 14(6): 625-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19897389

ABSTRACT

BACKGROUND: Oral ketamine is used as an adjuvant in the treatment of refractory neuropathic and cancer-related pain. Drug interactions may alter the analgesic or other effects of ketamine. AIM AND METHODS: The aim of the study was to investigate the effect of cytochrome P450 3A enzyme inhibition with clarithromycin on the pharmacokinetics and pharmacodynamics of oral S-ketamine in a randomized controlled cross-over study with two phases. Ten healthy subjects were pre-treated with oral clarithromycin or placebo for 4 days. On day 4, they ingested an oral dose of 0.2mg/kg of S-ketamine syrup. Plasma concentrations of ketamine and norketamine were measured for 24h. Analgesic effects were evaluated in a cold pressor test and psychomotor effects were followed for 12h. RESULTS: Clarithromycin increased the mean C(max) of ketamine by 3.6-fold (p<0.001) and the mean AUC(0-infinity) of ketamine by 2.6-fold (p=0.001). The relative amount of the CYP3A dependent metabolite norketamine was decreased by 54% by clarithromycin (p=0.004). Self-rated drug effect of S-ketamine was enhanced by clarithromycin (p<0.05) but other behavioral effects or cold pain scores were not affected. CONCLUSIONS: Clarithromycin strongly increases plasma concentrations of oral S-ketamine probably by inhibiting its CYP3A-mediated N-demethylation. This increase is reflected as modest changes in behavioral effects of oral S-ketamine.


Subject(s)
Clarithromycin/administration & dosage , Ketamine/administration & dosage , Ketamine/pharmacokinetics , Pain Measurement/drug effects , Adult , Analgesics/administration & dosage , Analgesics/blood , Analgesics/pharmacokinetics , Area Under Curve , Clarithromycin/blood , Cross-Over Studies , Drug Interactions , Female , Humans , Ketamine/blood , Male , Protein Synthesis Inhibitors/administration & dosage , Protein Synthesis Inhibitors/blood
SELECTION OF CITATIONS
SEARCH DETAIL