Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 7(10): e47599, 2012.
Article in English | MEDLINE | ID: mdl-23112822

ABSTRACT

UNLABELLED: The synthetic psychostimulant MDMA (± 3,4-methylenedioxymethamphetamine, ecstasy) acts as an indirect serotonin, dopamine, and norepinephrine agonist and as a mechanism-based inhibitor of the cytochrome P-450 2D6 (CYP2D6). It has been suggested that women are more sensitive to MDMA effects than men but no clinical experimental studies have satisfactorily evaluated the factors contributing to such observations. There are no studies evaluating the influence of genetic polymorphism on the pharmacokinetics (CYP2D6; catechol-O-methyltransferase, COMT) and pharmacological effects of MDMA (serotonin transporter, 5-HTT; COMT). This clinical study was designed to evaluate the pharmacokinetics and physiological and subjective effects of MDMA considering gender and the genetic polymorphisms of CYP2D6, COMT, and 5-HTT. A total of 27 (12 women) healthy, recreational users of ecstasy were included (all extensive metabolizers for CYP2D6). A single oral weight-adjusted dose of MDMA was administered (1.4 mg/kg, range 75-100 mg) which was similar to recreational doses. None of the women were taking oral contraceptives and the experimental session was performed during the early follicular phase of their menstrual cycle. Principal findings show that subjects reached similar MDMA plasma concentrations, and experienced similar positive effects, irrespective of gender or CYP2D6 (not taking into consideration poor or ultra-rapid metabolizers) or COMT genotypes. However, HMMA plasma concentrations were linked to CYP2D6 genotype (higher with two functional alleles). Female subjects displayed more intense physiological (heart rate, and oral temperature) and negative effects (dizziness, sedation, depression, and psychotic symptoms). Genotypes of COMT val158met or 5-HTTLPR with high functionality (val/val or l/*) determined greater cardiovascular effects, and with low functionality (met/* or s/s) negative subjective effects (dizziness, anxiety, sedation). In conclusion, the contribution of MDMA pharmacokinetics following 1.4 mg/kg MDMA to the gender differences observed in drug effects appears to be negligible or even null. In contrast, 5-HTTLPR and COMT val158met genotypes play a major role. TRIAL REGISTRATION: ClinicalTrials.gov NCT01447472.


Subject(s)
Adrenergic Uptake Inhibitors/blood , Adrenergic Uptake Inhibitors/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/blood , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Serotonin Agents/blood , Serotonin Agents/pharmacology , Adrenergic Uptake Inhibitors/adverse effects , Adult , Body Temperature/drug effects , Catechol O-Methyltransferase/genetics , Cytochrome P-450 CYP2D6/genetics , Female , Heart Rate/drug effects , Humans , Male , N-Methyl-3,4-methylenedioxyamphetamine/adverse effects , Polymorphism, Genetic , Serotonin Agents/adverse effects , Serotonin Plasma Membrane Transport Proteins/genetics , Sex Factors , Young Adult
2.
Drug Metab Dispos ; 37(7): 1448-55, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19349378

ABSTRACT

3,4-Methylenedioxymethamphetamine (MDMA, Ecstasy) is a widely misused synthetic amphetamine derivative and a serotonergic neurotoxicant in animal models and possibly humans. The underlying mechanism of neurotoxicity involves the formation of reactive oxygen species although their source remains unclear. It has been postulated that MDMA-induced neurotoxicity is mediated via the formation of bioreactive metabolites. In particular, the primary catechol metabolites, 3,4-dihydroxymethamphetamine (HHMA) and 3,4-dihydroxyamphetamine (HHA), subsequently cause the formation of glutathione and N-acetylcysteine conjugates, which retain the ability to redox cycle and are serotonergic neurotoxicants in rats. Although the presence of such metabolites has been recently demonstrated in rat brain microdialysate, their formation in humans has not been reported. The present study describes the detection of 5-(N-acetylcystein-S-yl)-3,4-dihydroxymethamphetamine (N-Ac-5-Cys-HHMA) and 5-(N-acetylcystein-S-yl)-3,4-dihydroxyamphetamine (N-Ac-5-Cys-HHA) in human urine of 15 recreational users of MDMA (1.5 mg/kg) in a controlled setting. The results reveal that in the first 4 h after MDMA ingestion approximately 0.002% of the administered dose was recovered as thioether adducts. Genetic polymorphisms in CYP2D6 and catechol-O-methyltransferase expression, the combination of which are major determinants of steady-state levels of HHMA and 4-hydroxy-3-methoxyamphetamine, probably explain the interindividual variability seen in the recovery of N-Ac-5-Cys-HHMA and N-Ac-5-Cys-HHA. In summary, the formation of neurotoxic thioether adducts of MDMA has been demonstrated for the first time in humans. The findings lend weight to the hypothesis that the bioactivation of MDMA to neurotoxic metabolites is a relevant pathway to neurotoxicity in humans.


Subject(s)
N-Methyl-3,4-methylenedioxyamphetamine/isolation & purification , N-Methyl-3,4-methylenedioxyamphetamine/metabolism , Sulfides/chemistry , Urine/chemistry , Adult , Eating , Female , Humans , Male , Mass Spectrometry , Metabolic Clearance Rate , Young Adult
3.
J Clin Psychopharmacol ; 28(5): 523-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18794647

ABSTRACT

3,4-Methylenedioxymethamphetamine (MDMA, ecstasy) is a widely abused substituted amphetamine. MDMA is predominantly O-demethylenated in humans by cytochrome P450 isoforms 2D6 and 1A2 (CYP2D6 and CYP CYP1A2, respectively). MDMA is also a mechanism-based inhibitor of CYP2D6. A controlled clinical trial was conducted in 15 healthy male subjects whereby a probe drug, dextromethorphan (DEX), was administered after an oral dose of 1.5 mg/kg MDMA. The pharmacokinetics of DEX and its metabolites were used to evaluate changes in CYP2D6 activity. The urinary metabolic ratio of DEX and dextrorphan was used to calculate a recovery half-life of CYP2D6. After MDMA, DEX Cmax and area under the curve increased approximately 10-fold with corresponding decreases in dextrorphan pharmacokinetic parameters. The metabolic ratio increased almost 100-fold from 0.0061 +/- 0.0056 to 0.4322 +/- 0.2848 after MDMA administration, with 67% of the subjects having a value greater than the antimode of 0.3 for assigning the poor metabolizer phenotype. CYP2D6 activity recovered after 10 days with a recovery half-life of 46.6 hours. In addition to the possible long-term serotonergic effects of MDMA, users must be warned of the consequences of such an inhibition.


Subject(s)
Cytochrome P-450 CYP2D6/drug effects , Enzyme Inhibitors/pharmacology , Hallucinogens/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Adult , Area Under Curve , Cytochrome P-450 CYP2D6/metabolism , Dextromethorphan/pharmacokinetics , Dextrorphan/pharmacokinetics , Half-Life , Humans , Male , Phenotype , Pilot Projects , Time Factors
4.
Psychopharmacology (Berl) ; 197(2): 263-78, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18074122

ABSTRACT

RATIONALE: A close relationship appears to exist between 3,4-methylenedioxymethamphetamine (MDMA)-induced changes in core body temperature and long-term serotonin (5-HT) loss. OBJECTIVE: We investigated whether changes in core body temperature affect MDMA metabolism. MATERIALS AND METHODS: Male Wistar rats were treated with MDMA at ambient temperatures of 15, 21.5, or 30 degrees C to prevent or exacerbate MDMA-induced hyperthermia. Plasma concentrations of MDMA and its main metabolites were determined for 6 h. Seven days later, animals were killed and brain indole content was measured. RESULTS: The administration of MDMA at 15 degrees C blocked the hyperthermic response and long-term 5-HT depletion found in rats treated at 21.5 degrees C. At 15 degrees C, plasma concentrations of MDMA were significantly increased, whereas those of three of its main metabolites were reduced when compared to rats treated at 21.5 degrees C. By contrast, hyperthermia and indole deficits were exacerbated in rats treated at 30 degrees C. Noteworthy, plasma concentrations of MDMA metabolites were greatly enhanced in these animals. Instrastriatal perfusion of MDMA (100 microM for 5 h at 21 degrees C) did not potentiate the long-term depletion of 5-HT after systemic MDMA. Furthermore, interfering in MDMA metabolism using the catechol-O-methyltransferase inhibitor entacapone potentiated the neurotoxicity of MDMA, indicating that metabolites that are substrates for this enzyme may contribute to neurotoxicity. CONCLUSIONS: This is the first report showing a direct relationship between core body temperature and MDMA metabolism. This finding has implications on both the temperature dependence of the mechanism of MDMA neurotoxicity and human use, as hyperthermia is often associated with MDMA use in humans.


Subject(s)
Body Temperature/physiology , Hallucinogens/metabolism , Hallucinogens/toxicity , N-Methyl-3,4-methylenedioxyamphetamine/metabolism , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Neurotoxicity Syndromes/psychology , Animals , Body Temperature/drug effects , Brain Chemistry/drug effects , Catechol O-Methyltransferase/metabolism , Catechol O-Methyltransferase Inhibitors , Catechols/pharmacology , Data Interpretation, Statistical , Drug Synergism , Enzyme Inhibitors/pharmacology , Fever/chemically induced , Hydroxyindoleacetic Acid/metabolism , Male , Microdialysis , Nitriles/pharmacology , Rats , Rats, Wistar , Serotonin/metabolism , Tyrosine/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...