Duloxetine inhibits effects of MDMA ("ecstasy") in vitro and in humans in a randomized placebo-controlled laboratory study

Cédric M Hysek, Linda D Simmler, Valentina G Nicola, Nerina Vischer, Massimiliano Donzelli, Stephan Krähenbühl, Eric Grouzmann, Jörg Huwyler, Marius C Hoener, Matthias E Liechti, Cédric M Hysek, Linda D Simmler, Valentina G Nicola, Nerina Vischer, Massimiliano Donzelli, Stephan Krähenbühl, Eric Grouzmann, Jörg Huwyler, Marius C Hoener, Matthias E Liechti

Abstract

This study assessed the effects of the serotonin (5-HT) and norepinephrine (NE) transporter inhibitor duloxetine on the effects of 3,4-methylenedioxy-methamphetamine (MDMA, ecstasy) in vitro and in 16 healthy subjects. The clinical study used a double-blind, randomized, placebo-controlled, four-session, crossover design. In vitro, duloxetine blocked the release of both 5-HT and NE by MDMA or by its metabolite 3,4-methylenedioxyamphetamine from transmitter-loaded human cells expressing the 5-HT or NE transporter. In humans, duloxetine inhibited the effects of MDMA including elevations in circulating NE, increases in blood pressure and heart rate, and the subjective drug effects. Duloxetine inhibited the pharmacodynamic response to MDMA despite an increase in duloxetine-associated elevations in plasma MDMA levels. The findings confirm the important role of MDMA-induced 5-HT and NE release in the psychotropic effects of MDMA. Duloxetine may be useful in the treatment of psychostimulant dependence.

Trial registration: Clinicaltrials.gov NCT00990067.

Conflict of interest statement

Competing Interests: One of the authors (MCH) is employed by a commercial company (Hoffmann-La Roche Ltd.). This does not alter the authors’ adherence to all the PLoS ONE policies on sharing data and materials.

Figures

Figure 1. CONSORT flowchart.
Figure 1. CONSORT flowchart.
Figure 2. Duloxetine inhibited the psychotropic effects…
Figure 2. Duloxetine inhibited the psychotropic effects of MDMA.
MDMA produced stimulant-like (b–d) and “entactogenic" (e, f) effects compared with placebo (p<0.001 for all scales). Duloxetine significantly inhibited MDMA-induced elevations in all of these subjective effects (a–f) (p<0.001 for all scales). Values are expressed as mean+SEM (n  = 16).
Figure 3. Duloxetine prevented the acute emotional…
Figure 3. Duloxetine prevented the acute emotional effects of MDMA in the Adjective Mood Rating Scale.
MDMA produced a state of well-being (a), emotional excitation (b), increased introversion at drug onset at 1.25 h (d), increased extroversion at 2 h (c), increased dreaminess (e), and decreased performance-oriented activity (f) (*p<0.05, **p<0.01, ***p<0.001, vs. placebo-placebo). Duloxetine prevented MDMA-induced elevations in well-being, emotional excitation, and extroversion (a-c) (###p<0.001, placebo-MDMA vs. duloxetine-MDMA). Values are expressed as mean+SEM (n  = 16).
Figure 4. Duloxetine prevented the acute effects…
Figure 4. Duloxetine prevented the acute effects of MDMA in the Altered States of Consciousness (ASC) scale.
MDMA significantly increased the ASC sum score, Oceanic Boundlessness (OB), Anxious Ego Dissolution (AED), and Visionary Restructuralization (VR) dimensions, and most of the subscales (*p#p<0.05, ##p<0.01, ###p<0.001, placebo-MDMA vs. duloxetine-MDMA). Values are expressed as mean+SEM (n  = 16).
Figure 5. Duloxetine reduced the cardiostimulant response…
Figure 5. Duloxetine reduced the cardiostimulant response to MDMA.
Duloxetine reduced the elevations in mean arterial blood pressure (a) and heart rate (b) in response to MDMA. Duloxetine also nonsignificantly lowered the MDMA-induced increase in body temperature (c). Values are expressed as mean+SEM of 16 subjects.
Figure 6. Duloxetine increased MDMA exposure.
Figure 6. Duloxetine increased MDMA exposure.
Pharmacokinetics of MDMA, MDA, HMMA, and duloxetine (a–d). Duloxetine was administered 16 h and 4 h before MDMA, which was administered at the 0 h time point. Duloxetine increased the Cmax and AUC0–6 of MDMA (a), had no significant effect on MDA exposure (b), and decreased the Cmax and AUC0–6 of HMMA (c). Plasma duloxetine concentrations were similar in the duloxetine-placebo and duloxetine-MDMA groups before MDMA administration (at –4 h and 0 h). Duloxetine concentrations increased 1 h after MDMA administration in the duloxetine-MDMA vs. duloxetine-placebo group (d). Values are expressed as mean±SEM of 16 subjects. MDMA, 3,4-methylenedioxymethamphetamine; MDA, 3,4-methylenedioxyamphetamine; HMMA, 4-hydroxy-3-methoxymethamphetamine.
Figure 7. Pharmacokinetic-pharmacodynamic (PK-PD) relationship.
Figure 7. Pharmacokinetic-pharmacodynamic (PK-PD) relationship.
MDMA effects are plotted against simultaneous MDMA plasma concentrations (a, b). The time of sampling is noted next to each point in minutes or hours after MDMA administration. The clockwise hysteresis indicates acute tolerance to the effects of MDMA. Duloxetine pretreatment markedly reduced physical and subjective responses to MDMA in the hysteresis loops (a, b).
Figure 8. Duloxetine blocked MDMA- and MDA-induced…
Figure 8. Duloxetine blocked MDMA- and MDA-induced 5-HT and NE efflux.
Duloxetine inhibited SERT-mediated 5-HT release by MDMA (a) and MDA (b). Duloxetine also inhibited NET-mediated NE release by MDMA (c) and MDA (d). Values are expressed as mean ± SEM (n  = 3–6) of retained radiolabeled substrate following incubation with various concentrations of MDMA and MDA.
Figure 9. Pharmacokinetic-pharmacodynamic modeling.
Figure 9. Pharmacokinetic-pharmacodynamic modeling.
Duloxetine lowered Emax in the MDMA concentration-effect curve (a) with little effect on EC50, similar to the effect of MDMA on monoamine release in vitro. Diamonds and circles represent concentration-effect data pairs for ascending concentrations for placebo-MDMA and duloxetine-MDMA, respectively (a). The solid lines show the fit of a sigmoid Emax PD model to the observed PK data (a). Dashed lines indicate the 95% confidence interval (CI) of the estimation error (a). NE release predicted the observed subjective effect of MDMA in vivo (b). Predicted effects are shown as curves (mean ±95% CI) that represent the fit of the in vitro concentration-effect data to the 16 individual plasma concentration-time curves (b). Observed values are expressed as mean±SEM of 16 subjects (b). MDMA, 3,4-methylenedioxymethamphetamine; NE, norepinephrine; 5-HT, serotonin.

References

    1. Rudnick G, Wall SC. The molecular mechanism of “ecstasy" [3,4-methylenedioxy-methamphetamine (MDMA)]: serotonin transporters are targets for MDMA-induced serotonin release. Proc Natl Acad Sci U S A. 1992;89:1817–1821.
    1. Rothman RB, Baumann MH, Dersch CM, Romero DV, Rice KC, et al. Amphetamine-type central nervous system stimulants release norepinephrine more potently than they release dopamine and serotonin. Synapse. 2001;39:32–41.
    1. Verrico CD, Miller GM, Madras BK. MDMA (ecstasy) and human dopamine, norepinephrine, and serotonin transporters: implications for MDMA-induced neurotoxicity and treatment. Psychopharmacology. 2007;189:489–503.
    1. Wall SC, Gu H, Rudnick G. Biogenic amine flux mediated by cloned transporters stably expressed in cultured cell lines: amphetamine specificity for inhibition and efflux. Mol Pharmacol. 1995;47:544–550.
    1. Rothman RB, Partilla JS, Baumann MH, Dersch CM, Carroll FI, et al. Neurochemical neutralization of methamphetamine with high-affinity nonselective inhibitors of biogenic amine transporters: a pharmacological strategy for treating stimulant abuse. Synapse. 2000;35:222–227.
    1. Rothman RB, Baumann MH, Blough BE, Jacobson AE, Rice KC, et al. Evidence for noncompetitive modulation of substrate-induced serotonin release. Synapse. 2010;64:862–869.
    1. Liechti ME, Baumann C, Gamma A, Vollenweider FX. Acute psychological effects of 3,4-methylenedioxymethamphetamine (MDMA, “ecstasy") are attenuated by the serotonin uptake inhibitor citalopram. Neuropsychopharmacology. 2000;22:513–521.
    1. Tancer M, Johanson CE. The effects of fluoxetine on the subjective and physiological effects of 3,4-methylenedioxymethamphetamine (MDMA) in humans. Psychopharmacology. 2007;189:565–573.
    1. Farre M, Abanades S, Roset PN, Peiro AM, Torrens M, et al. Pharmacological interaction between 3,4-methylenedioxymethamphetamine (ecstasy) and paroxetine: pharmacological effects and pharmacokinetics. J Pharmacol Exp Ther. 2007;323:954–962.
    1. Hysek CM, Simmler LD, Ineichen M, Grouzmann E, Hoener MC, et al. The norepinephrine transporter inhibitor reboxetine reduces stimulant effects of MDMA (“ecstasy") in humans. Clin Pharmacol Ther. 2011;90:246–255.
    1. Sofuoglu M, Poling J, Hill K, Kosten T. Atomoxetine attenuates dextroamphetamine effects in humans. Am J Drug Alcohol Abuse. 2009;35:412–416.
    1. Hysek CM, Brugger R, Simmler LD, Bruggisser M, Donzelli M, et al. Effects of the α2-adrenergic agonist clonidine on the pharmacodynamics and pharmacokinetics of methylenedioxymethamphetamine in healthy volunteers. J Pharmacol Exp Ther. 2012;340:286–294.
    1. Bymaster FP, Dreshfield-Ahmad LJ, Threlkeld PG, Shaw JL, Thompson L, et al. Comparative affinity of duloxetine and venlafaxine for serotonin and norepinephrine transporters in vitro and in vivo, human serotonin receptor subtypes, and other neuronal receptors. Neuropsychopharmacology. 2001;25:871–880.
    1. Wong DT. Duloxetine (LY 248686): an inhibitor of serotonin and noradrenaline uptake and an antidepressant drug candidate. Expert Opin Investig Drugs. 1998;7:1691–1699.
    1. Segura M, Farre M, Pichini S, Peiro AM, Roset PN, et al. Contribution of cytochrome P450 2D6 to 3,4-methylenedioxymethamphetamine disposition in humans: use of paroxetine as a metabolic inhibitor probe. Clin Pharmacokinet. 2005;44:649–660.
    1. Knadler MP, Lobo E, Chappell J, Bergstrom R. Duloxetine: clinical pharmacokinetics and drug interactions. Clin Pharmacokinet. 2011;50:281–294.
    1. Battaglia G, Brooks BP, Kulsakdinun C, De Souza EB. Pharmacologic profile of MDMA (3,4-methylenedioxymethamphetamine) at various brain recognition sites. Eur J Pharmacol. 1988;149:159–163.
    1. Wittchen HU, Wunderlich U, Gruschwitz S, Zaudig M. Göttingen: Hogrefe-Verlag; 1997. SKID-I: Strukturiertes Klinisches Interview für DSM-IV.
    1. Derogatis LR, Rickels K, Rock AF. The SCL-90 and the MMPI: a step in the validation of a new self-report scale. Br J Psychiatry. 1976;128:280–289.
    1. Schmitz N, Hartkamp N, Kiuse J, Franke GH, Reister G, et al. The Symptom Check-List-90-R (SCL-90-R): a German validation study. Qual Life Res. 2000;9:185–193.
    1. Janke W, Debus G. Göttingen, Germany: Hogrefe; 1978. Die Eigenschaftswörterliste.
    1. Dittrich A. The standardized psychometric assessment of altered states of consciousness (ASCs) in humans. Pharmacopsychiatry 31. 1998;S2:80–84.
    1. Studerus E, Gamma A, Vollenweider FX. Psychometric evaluation of the altered states of consciousness rating scale (OAV). PLoS One. 2010;5:e12412.
    1. Cami J, Farre M, Mas M, Roset PN, Poudevida S, et al. Human pharmacology of 3,4-methylenedioxymethamphetamine (“ecstasy"): psychomotor performance and subjective effects. J Clin Psychopharmacol. 2000;20:455–466.
    1. Kolbrich EA, Goodwin RS, Gorelick DA, Hayes RJ, Stein EA, et al. Physiological and subjective responses to controlled oral 3,4-methylenedioxymethamphetamine administration. J Clin Psychopharmacol. 2008;28:432–440.
    1. Zerssen DV. Münchener Informationssystem. München: Psychis; 1976. Die Beschwerden-Liste.
    1. Simmler LD, Hysek CM, Liechti ME. Sex differences in the effects of MDMA (ecstasy) on plasma copeptin in healthy subjects. J Clin Endocrinol Metab. 2011;96:2844–2850.
    1. Tatsumi M, Groshan K, Blakely RD, Richelson E. Pharmacological profile of antidepressants and related compounds at human monoamine transporters. Eur J Pharmacol. 1997;340:249–258.
    1. Cheng Y, Prusoff WH. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem Pharmacol. 1973;22:3099–3108.
    1. Torok M, Huwyler J, Drewe J, Gutmann H, Fricker G. Transport of the β-lactam antibiotic benzylpenicillin and the dipeptide glycylsarcosine by brain capillary endothelial cells in vitro. Drug Metab Dispos. 1998;26:1144–1148.
    1. Hermle L, Spitzer M, Borchardt D, Kovar KA, Gouzoulis E. Psychological effects of MDE in normal subjects: are entactogens a new class of psychoactive agents? Neuropsychopharmacology. 1993;8:171–176.
    1. Bedi G, Hyman D, de Wit H. Is ecstasy an “empathogen"? Effects of±3,4-methylenedioxymethamphetamine on prosocial feelings and identification of emotional states in others. Biol Psychiatry. 2010;68:1134–1140.
    1. Van Dyke C, Jatlow P, Ungerer J, Barash PG, Byck R. Oral cocaine: plasma concentrations and central effects. Science. 1978;200:211–213.
    1. Pifl C, Nagy G, Berenyi S, Kattinger A, Reither H, et al. Pharmacological characterization of ecstasy synthesis byproducts with recombinant human monoamine transporters. J Pharmacol Exp Ther. 2005;314:346–354.
    1. Han DD, Gu HH. Comparison of the monoamine transporters from human and mouse in their sensitivities to psychostimulant drugs. BMC Pharmacol. 2006;6:6.
    1. Rothman RB, Baumann MH. Monoamine transporters and psychostimulant drugs. Eur J Pharmacol. 2003;479:23–40.
    1. Kahlig KM, Binda F, Khoshbouei H, Blakely RD, McMahon DG, et al. Amphetamine induces dopamine efflux through a dopamine transporter channel. Proc Natl Acad Sci U S A. 2005;102:3495–3500.
    1. Saunders C, Ferrer JV, Shi L, Chen J, Merrill G, et al. Amphetamine-induced loss of human dopamine transporter activity: an internalization-dependent and cocaine-sensitive mechanism. Proc Natl Acad Sci U S A. 2000;97:6850–6855.
    1. Robertson SD, Matthies HJ, Galli A. A closer look at amphetamine-induced reverse transport and trafficking of the dopamine and norepinephrine transporters. Mol Neurobiol. 2009;39:73–80.
    1. Matthies HJ, Moore JL, Saunders C, Matthies DS, Lapierre LA, et al. Rab11 supports amphetamine-stimulated norepinephrine transporter trafficking. J Neurosci. 2010;30:7863–7877.
    1. Field JR, Henry LK, Blakely RD. Transmembrane domain 6 of the human serotonin transporter contributes to an aqueously accessible binding pocket for serotonin and the psychostimulant 3,4-methylenedioxymethamphetamine. J Biol Chem. 2010;285:11270–11280.
    1. Thompson BJ, Jessen T, Henry LK, Field JR, Gamble KL, et al. Transgenic elimination of high-affinity antidepressant and cocaine sensitivity in the presynaptic serotonin transporter. Proc Natl Acad Sci U S A. 2011;108:3785–3790.
    1. Baumann MH, Clark RD, Rothman RB. Locomotor stimulation produced by 3,4-methylenedioxymethamphetamine (MDMA) is correlated with dialysate levels of serotonin and dopamine in rat brain. Pharmacol Biochem Behav. 2008;90:208–217.
    1. Verrico CD, Lynch L, Fahey MA, Fryer AK, Miller GM, et al. MDMA-induced impairment in primates: antagonism by a selective norepinephrine or serotonin, but not by a dopamine/norepinephrine transport inhibitor. J Psychopharmacol. 2008;22:187–202.
    1. Liechti ME, Vollenweider FX. Acute psychological and physiological effects of MDMA (“Ecstasy") after haloperidol pretreatment in healthy humans. Eur Neuropsychopharmacol. 2000;10:289–295.
    1. Zhao RK, Cheng G, Tang J, Song J, Peng WX. Pharmacokinetics of duloxetine hydrochloride enteric-coated tablets in healthy Chinese volunteers: a randomized, open-label, single- and multiple-dose study. Clin Ther. 2009;31:1022–1036.
    1. Hondebrink L, Meulenbelt J, Meijer M, van den Berg M, Westerink RH. High concentrations of MDMA (“ecstasy") and its metabolite MDA inhibit calcium influx and depolarization-evoked vesicular dopamine release in PC12 cells. Neuropharmacology. 2011;61:202–208.
    1. Gudelsky GA, Yamamoto BK, Nash JF. Potentiation of 3,4-methylenedioxymethamphetamine-induced dopamine release and serotonin neurotoxicity by 5-HT2 receptor agonists. Eur J Pharmacol. 1994;264:325–330.
    1. Gudelsky GA, Nash JF. Carrier-mediated release of serotonin by 3,4-methylenedioxymethamphetamine: implications for serotonin-dopamine interactions. J Neurochem. 1996;66:243–249.
    1. Gould GG, Javors MA, Frazer A. Effect of chronic administration of duloxetine on serotonin and norepinephrine transporter binding sites in rat brain. Biol Psychiatry. 2007;61:210–215.

Source: PubMed

3
Abonnieren