Elsevier

Biochemical Pharmacology

Volume 83, Issue 1, 1 January 2012, Pages 131-138
Biochemical Pharmacology

Stereoselective urinary MDMA (ecstasy) and metabolites excretion kinetics following controlled MDMA administration to humans

https://doi.org/10.1016/j.bcp.2011.09.023Get rights and content

Abstract

The R- and S-enantiomers of racemic 3,4-methylenedioxymethamphetamine (MDMA) exhibit different dose–concentration curves. In plasma, S-MDMA was eliminated at a higher rate, most likely due to stereoselective metabolism. Similar data were shown in various in vitro experiments. The aim of the present study was the in vivo investigation of stereoselective elimination of MDMA's phase I and phase II metabolites in human urine following controlled oral MDMA administration. Urine samples from 10 participants receiving 1.0 and 1.6 mg/kg MDMA separated by at least one week were analyzed blind by liquid chromatography–high resolution-mass spectrometry and gas chromatography–mass spectrometry after chiral derivatization with S-heptafluorobutyrylprolyl chloride. R/S ratios at Cmax were comparable after low and high doses with ratios >1 for MDMA, free DHMA, and HMMA sulfate, and with ratios <1 for MDA, free HMMA, DHMA sulfate and HMMA glucuronide. In the five days after the high MDMA dose, a median of 21% of all evaluated compounds were excreted as R-stereoisomers and 17% as S-stereoisomers. Significantly greater MDMA, DHMA, and HMMA sulfate R-enantiomers and HMMA and HMMA glucuronide S-stereoisomers were excreted. No significant differences were observed for MDA and DHMA sulfate stereoisomers. Changes in R/S ratios could be observed over time for all analytes, with steady increases in the first 48 h. R/S ratios could help to roughly estimate time of MDMA ingestion and therefore, improve interpretation of MDMA and metabolite urinary concentrations in clinical and forensic toxicology.

Introduction

3,4-Methylenedioxymethamphetamine (MDMA) or Ecstasy, is a recreational drug of abuse popular among young people. Recently, the Substance Abuse and Mental Health Services Administration reported increasing MDMA consumption in the United States [1]. MDMA is usually consumed as a 1:1 racemate of R- and S-enantiomers. The S-enantiomer is more potent than the R-enantiomer in producing euphoria, energy and a desire to socialize [2], [3]. MDMA also can induce severe acute toxic symptoms, such as tachycardia, hypertension, hyperthermia, and hepatotoxicity [3], severe and fatal intoxications also have been described [3]. Concerning chronic toxicity, animal data suggest that MDMA causes irreversible damage to serotonergic nerve terminals in the central nervous system [3], [4], [5], [6]. In humans, chronic MDMA toxicity is still controversial, as some recent publications suggest that animal doses may be too high compared to human pharmacokinetics [7], [8], while others indicate that humans are susceptible to brain serotonin neurotoxicity [9]. Admittedly, direct MDMA injection into rat brain failed to reproduce neurotoxic effects seen after systemic administration [10]. Furthermore, alteration of cytochrome P450 (CYP)-mediated MDMA metabolism influenced MDMA-induced neurotoxicity [10], [11]. Therefore, MDMA metabolism may be an important contributor to neurotoxicity [12], [13], [14], [15].

In vivo and in vitro MDMA studies revealed two main metabolic pathways. One includes multiple CYP enzyme-catalyzed O-demethylenation of MDMA to 3,4-dihydroxymethamphetamine (DHMA), followed by catechol-O-methyltransferase (COMT)-catalyzed O-methylation, primarily to 4-hydroxy-3-methoxymethamphetamine (HMMA). DHMA and HMMA also may be conjugated by uridine diphosphate glucuronyltransferases (UGT) to DHMA 3-glucuronide, DHMA 4-glucuronide, and HMMA glucuronide, or by sulfotransferases (SULT) to DHMA 3-sulfate, DHMA 4-sulfate, and HMMA sulfate. A minor pathway includes demethylation to 3,4-methylendioxy-amphetamine (MDA) followed by demethylenation to 3,4-dihydroxy-amphetamine (DHA), O-methylation to 4-hydroxy-3-methoxyamphetamine (HMA), and conjugation [5], [16], [17], [18], [19], [20], [21].

During the first 48 h after racemic MDMA ingestion, the S-enantiomer was eliminated from plasma at a faster rate than the R-enantiomer [2], [3], [22], [23], [24], [25]. Enantioselective metabolism is the most likely explanation for different MDMA enantiomer pharmacokinetics in humans. In vitro experiments mediated by CYP, COMT, SULT and UGT enzymes documented preferential metabolism of S-enantiomers [19], [26], [27], [28].

Enantioselective metabolism and elimination data in human blood or urine following controlled MDMA administration are available [2], [24], [25]; however, in all of these, DHMA, HMMA, and/or HMA data were obtained after conjugate cleavage either by acidic or enzymatic hydrolysis (Helix pomatia). There are no data available following direct measurement of intact glucuronides and sulfates that also might show different enantiomeric ratios. The aim of the present study was to investigate in vivo excretion of chiral MDMA phase I and phase II metabolites in human urine following controlled oral MDMA administration.

Section snippets

Chemicals and reagents

Racemic MDA, HMA, DHA, MDMA, HMMA, and DHMA hydrochlorides were obtained from Lipomed (Bad Saeckingen, Germany). Methanolic 1 mg/mL MDA-d5 and MDMA-d5 were purchased from LGC Promochem (Wesel, Germany), 4-hydroxymethamphetamine (pholedrine), 3,4-dihydroxybenzylamine (DHBA), and morphine 6-glucuronide (M6G), hexamethyldisilazane (HMDS) and sulfatases (EC no. 3.1.6.1) from Aerobacter aerogenes were from Sigma Aldrich (Steinheim, Germany). R/S-DHMA sulfates, R/S-HMMA sulfate, and single

Pharmacokinetic analysis

Table 1, Table 2 summarize Cmax, tmax, Cmax100, and tmax100 for each R- and S-enantiomer. Statistically significant differences were observed for the enantiomers of all compounds except HMMA sulfate after creatinine normalization. Higher median maximum concentrations were noted for the R-stereoisomers of MDMA, DHMA and HMMA sulfate with and without creatinine normalization. Conversely, median Cmax and Cmax100 were higher for the S-enantiomers of all other analytes. The S-enantiomer median tmax

Discussion

Few data on enantioselective urinary excretion kinetics of MDMA and its metabolites in human urine are available [32], [33], [34], [35]. None determined enantiomeric ratios of all major metabolites after controlled oral MDMA administration, and phase II metabolite data are rare. The present study is the first to monitor enantioselective excretion of phase I and II glucuronides and sulfates after controlled oral MDMA administration to humans. Direct analysis of intact conjugates was a

Acknowledgements

The authors would like to thank Carsten Schröder, Armin Weber, and Gabriele Ulrich from the Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University and Frank T. Peters from Schiller University Jena for their assistance and helpful discussions and Thermo Fisher for their support. Funding was provided by HOMFOR 2010, the research fund of the Medical Faculty, Saarland University, Homburg and the Intramural Research

References (36)

  • N. Easton et al.

    Ecstasy are animal data consistent between species and can they translate to humans?

    J Psychopharmacol

    (2006)
  • M.H. Baumann et al.

    Effects of dose and route of administration on pharmacokinetics of (+ or −)-3,4-methylenedioxymethamphetamine in the rat

    Drug Metab Dispos

    (2009)
  • M. Mueller et al.

    Direct comparison of (+/−) 3,4-methylenedioxymethamphetamine (ecstasy) disposition and metabolism in squirrel monkeys and humans

    Ther Drug Monit

    (2009)
  • U.D. McCann et al.

    Positron emission tomographic studies of brain dopamine and serotonin transporters in abstinent (+/−)3,4-methylenedioxymethamphetamine (ecstasy) users: relationship to cognitive performance

    Psychopharmacology (Berl)

    (2008)
  • B. Esteban et al.

    3,4-Methylenedioxymethamphetamine induces monoamine release, but not toxicity, when administered centrally at a concentration occurring following a peripherally injected neurotoxic dose

    Psychopharmacology (Berl)

    (2001)
  • R. Gollamudi et al.

    Influence of inducers and inhibitors on the metabolism in vitro and neurochemical effects in vivo of MDMA

    Neurotoxicology

    (1989)
  • F. Bai et al.

    Glutathione and N-acetylcysteine conjugates of alpha-methyldopamine produce serotonergic neurotoxicity: possible role in methylenedioxyamphetamine-mediated neurotoxicity

    Chem Res Toxicol

    (1999)
  • M. Hiramatsu et al.

    Metabolism of methylenedioxymethamphetamine: formation of dihydroxymethamphetamine and a quinone identified as its glutathione adduct

    J Pharmacol Exp Ther

    (1990)
  • Cited by (23)

    • Bioanalytical methods for determining ecstasy components in biological matrices: A review

      2018, TrAC - Trends in Analytical Chemistry
      Citation Excerpt :

      DHMA and HMMA also may be conjugated by uridine diphosphate glucuronyltransferases to DHMA 3-glucuronide, DHMA 4-glucuronide and HMMA glucuronide, or by sulfotransferases to DHMA 3-sulphate, DHMA 4-sulphate and HMMA sulphate. Other minor pathways include demethylation to MDA followed by demethylation to DHA, O-methylation to HMA and conjugation [15–22]. Fig. 3 shows the metabolism of MDMA.

    • Enantiomeric profiling of chiral illicit drugs in a pan-European study

      2018, Water Research
      Citation Excerpt :

      Due to the stereoselective metabolism of MDMA, HMMA and its glucuronide derivative are formed enriched with S-(+)-enantiomer. Interestingly, HMMA sulphate is formed via non-stereoselective route (Schwaninger et al., 2012). In this study, HMMA was enriched with the second eluting enantiomer.

    • Sensitive detection of anionic metabolites of drugs by positive ion mode HPLC-PIESI-MS

      2015, International Journal of Mass Spectrometry
      Citation Excerpt :

      The pharmacokinetics and pharmacodynamics of most drugs are known, but these are usually for highly controlled single dose experiments [3–7]. Further, it is well known that different classes of drugs and even different drugs within a class can have unique metabolic pathways and therefore very different elimination half-lives [7–11]. In the case of performance-enhancing drugs, several mitigating factors can affect individual drug-metabolite elimination/lifetimes/detection times.

    View all citing articles on Scopus
    View full text