Download Opposite effects of MDMA

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

5-HT3 antagonist wikipedia , lookup

Environmental impact of pharmaceuticals and personal care products wikipedia , lookup

Prescription costs wikipedia , lookup

Neuropsychopharmacology wikipedia , lookup

Pharmaceutical industry wikipedia , lookup

Clinical trial wikipedia , lookup

Drug interaction wikipedia , lookup

Psychedelic therapy wikipedia , lookup

Pharmacogenomics wikipedia , lookup

Polysubstance dependence wikipedia , lookup

Urban legends about drugs wikipedia , lookup

Neuropharmacology wikipedia , lookup

Pharmacokinetics wikipedia , lookup

Pharmacognosy wikipedia , lookup

Theralizumab wikipedia , lookup

Psychopharmacology wikipedia , lookup

Stimulant wikipedia , lookup

MDMA wikipedia , lookup

Bilastine wikipedia , lookup

Transcript
Pharmacological interaction between duloxetine and
3,4-Methylenedioxymethamphetamine (MDMA, Ecstasy)
1
SYNOPSIS
Sponsor
Study title
Clinical Pharmacology & Toxicology, University Hospital Basel
Pharmacological interaction between duloxetine and
Methylenedioxymethamphetamine (MDMA, “Ecstasy”)
Study site
Divisions of Clinical Pharmacology & Toxicology, University Hospital
Basel
Primary study endpoints
1. Effect of duloxetine on the subjective response to MDMA
Secondary study endpoints
1. Effect of duloxetine on physiological responses to MDMA
2. Tolerability of MDMA and duloxetine
3. Effect of duloxetine on neuroendocrine effects of MDMA
4. Effects of duloxetine on pharmacokinetics of MDMA
5. Effect of MDMA on duloxetine pharmacokinetics
Randomized, placebo-controlled, cross-over study
Determination of plasma concentrations of MDMA, metabolites and
duloxetine by GC-MS/LC-MS/MS
16 healthy volunteers (men or women), age: 19-45 years
Duloxetine (Cymbalta®) and MDMA (3,4-methylenedioxymethamphetamine)
Start: 1.1.2010, End: 31.12.2010
Objectives
Design
Analytics
Subjects
Drugs
Study dates
3,4-
Duloxetine-MDMA Study Protocol
2
Matthias Liechti
INDEX
1
2
3
4
5
6
7
SYNOPSIS ...................................................................................................................................... 0
INDEX ............................................................................................................................................. 1
Abbreviations and Definitions.......................................................................................................... 3
SUMMARY ...................................................................................................................................... 4
INTRODUCTION ............................................................................................................................ 5
SIGNIFICANCE .............................................................................................................................. 5
BACKGROUND .............................................................................................................................. 5
7.1
Pharmacology of MDMA ........................................................................................................ 5
7.2
Expected interactive effects of duloxetine and MDMA ........................................................... 6
7.2.1
Pharmacodynamic interactions ..................................................................................... 6
7.2.2
Pharmacokinetic interactions......................................................................................... 7
8
STUDY OBJECTIVES .................................................................................................................... 7
8.1
Primary study endpoints ......................................................................................................... 7
8.2
Secondary study endpoints .................................................................................................... 7
9
HYPOTHESES................................................................................................................................ 7
9.1
Primary Hypotheses ............................................................................................................... 7
9.2
Secondary Hypotheses .......................................................................................................... 7
10 STUDY DESIGN AND METHODS ................................................................................................. 8
10.1
Study design ........................................................................................................................... 8
10.2
Study duration ........................................................................................................................ 8
10.3
Study site ................................................................................................................................ 8
10.4
Study population ..................................................................................................................... 8
10.4.1
Recruitment ................................................................................................................... 8
10.4.2
Inclusion criteria ............................................................................................................. 8
10.4.3
Exclusion criteria ........................................................................................................... 8
11 STUDY PROCEDURES ................................................................................................................. 9
11.1
Schedule of Events ................................................................................................................ 9
11.2
Screening procedure .............................................................................................................. 9
11.2.1
Informed consent ........................................................................................................... 9
11.2.2
Physical health ............................................................................................................... 9
11.2.3
Mental health ................................................................................................................. 9
11.2.4
History of Drug use ........................................................................................................ 9
11.2.5
Screening laboratory tests ........................................................................................... 10
11.2.6
CYP2D6 phenotyping .................................................................................................. 10
11.2.7
Genotyping .................................................................................................................. 10
11.2.8
Personality ................................................................................................................... 10
11.2.9
Menstrual cycle phase in women................................................................................. 10
11.3
Schedule of experimental session ....................................................................................... 10
11.4
Assessments and Measures ................................................................................................ 11
11.4.1
Psychometric assessments ......................................................................................... 11
11.4.1.1
Visual Analogue Scales (VAS) ............................................................................ 11
11.4.1.2
Adjective mood rating scale (AMRS) .................................................................. 11
11.4.1.3
State-trait anxiety inventory (STAI) ..................................................................... 11
11.4.1.4
Addiction Research Center Inventory (ARCI) ..................................................... 11
11.4.1.5
Altered states of consciousness (5D-ASC)......................................................... 12
11.4.2
Physiological assessments .......................................................................................... 12
11.4.2.1
Vital signs ............................................................................................................ 12
11.4.2.2
List of complaints (LC) and Adverse effects (AE) ............................................... 12
11.4.3
Cognitive and psychomotor function ........................................................................... 12
11.4.3.1
Reading the mind in the eyes test (RME) ........................................................... 12
11.4.3.2
Choice reaction time test (CRTT) ....................................................................... 12
11.4.3.3
End of Session Questionnarie ............................................................................ 13
11.4.4
Neuroendocrine function ............................................................................................. 13
11.4.5
Blood sample collection (pharmacokinetics) ............................................................... 13
11.4.5.1
Amount of blood samples ................................................................................... 13
11.4.6
End of Study (EOS) visit .............................................................................................. 14
11.4.6.1
EOS Examination................................................................................................ 14
11.4.6.2
EOS Questionnaire ............................................................................................. 14
Version 14.10.2009
1/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
11.5
Study Drugs .......................................................................................................................... 14
11.5.1
Duloxetine .................................................................................................................... 14
11.5.2
MDMA .......................................................................................................................... 15
11.5.2.1
Chemistry, manufacturing, control ...................................................................... 15
11.5.2.2
Metabolism and Pharmacokinetics ..................................................................... 15
11.5.2.3
Dose selection .................................................................................................... 15
11.5.3
Drug-Accountability, storage and return of study drugs............................................... 16
11.5.4
Concomitant medications ............................................................................................ 16
11.5.5
Randomization and Blinding ........................................................................................ 16
11.5.6
Compliance .................................................................................................................. 16
12 Analytics ........................................................................................................................................ 16
12.1
MDMA and metabolites ........................................................................................................ 16
12.2
CYP2D6 Phenotype ............................................................................................................. 16
12.3
Cortisol and prolactin............................................................................................................ 16
12.4
Catecholamines and metanephrines .................................................................................... 16
12.5
Copeptin ............................................................................................................................... 17
12.6
Duloxetine ............................................................................................................................ 17
13 Data analysis ................................................................................................................................. 17
13.1
Sample size estimation ........................................................................................................ 17
13.2
Analysis of outcomes ........................................................................................................... 17
14 Protection of subjects.................................................................................................................... 17
14.1
Potential risks ....................................................................................................................... 17
14.2
Monitoring of toxicity............................................................................................................. 18
14.2.1
Safety definitions ......................................................................................................... 18
14.2.2
Documentation ............................................................................................................ 19
14.2.3
Adverse events (AE) documentation ........................................................................... 19
14.2.4
SAE and SUSAR reporting .......................................................................................... 20
14.2.5
Medical follow-up of adverse events ........................................................................... 20
14.2.6
Monitoring for specific toxicity...................................................................................... 20
14.3
Risk-Benefit Assessment ..................................................................................................... 21
14.4
Ethical standards .................................................................................................................. 21
14.4.1
Institutional Review Board ........................................................................................... 21
14.4.2
Protocol amendments.................................................................................................. 21
14.4.3
Early study termination ................................................................................................ 22
14.4.4
Insurance ..................................................................................................................... 22
14.4.5
Compensation ............................................................................................................. 22
14.4.6
Premature withdrawal of subjects................................................................................ 22
14.4.7
Replacement policy ..................................................................................................... 22
14.5
Legal authorizations ............................................................................................................. 22
14.6
Trial registration.................................................................................................................... 22
14.7
Study documentation and record keeping ............................................................................ 22
14.8
Quality control and quality assurance................................................................................... 23
14.8.1
Training of personnel and SOPs ................................................................................. 23
14.8.2
Monitoring .................................................................................................................... 23
14.8.3
Direct access to source data ....................................................................................... 23
14.8.4
Inspection .................................................................................................................... 23
14.9
Confidentiality of trial documents and subject records ......................................................... 23
15 Time plan ...................................................................................................................................... 23
16 Publications ................................................................................................................................... 23
17 Budget and Funding ...................................................................................................................... 24
18 Certifications of principal investigator ........................................................................................... 24
19 Conflict of interest ......................................................................................................................... 24
20 Research Environment ................................................................................................................. 24
20.1
Study site .............................................................................................................................. 24
21 References .................................................................................................................................... 24
Version 14.10.2009
2/30
Duloxetine-MDMA Study Protocol
3
Matthias Liechti
Abbreviations and Definitions
ASAT
ALAT
AE
ANOVA
AMRS
ARCI
AUC
BAG
BP
CHF
CRF
CRTT
CRC
CRP
CYP
Cmax
Cl
Ca
5D-ASC
EWL
Ecstasy
EKBB
ECG
EOS
GCP
GC-MS
GMP
-GT
HPLC
5-HT
HR
HMA
HMMA
aspartate transferase
alanine aminotransferase
adverse event
analysis of variance
adjective mood rating scale
addiction research center inventory
area under the concentration time curve
Bundesamt für Gesundheitswesen = Swiss Federal Office of Public Health
blood pressure
Swiss francs
case report form
choice reaction time test
clinical research center
C-reactive proteine
cytochrome P450 enzyme
maximum concentration
chloride
calcium
5 dimensions of altered states of consciousness
Eigenschaftswörterliste = AMRS
may contain MDMA or other substances at various amounts
Ethische Kommission beider Basel
electrocardigram
end of study
good clinical practice
gas chromatography-mass spectrometry
good manufacturing practice
gamma-glutamyl transferase
high-pressure liquid chromatography
serotonin
heart rate
5-HIAA
4-hydroxy-3-methoxyamphetamine
4-hydroxy-3-mehoxymethamphetamine
5-hydroxy indole acetic acid
IRB
K
LSD
LC
LDH
LC-MS
MDMA
MDA
MD
NE
Na
P
PK
PI
Psys
PhD
THC
SAE
SSRI
STAI
SD
T1/2
Tmax
VAS
institutional ethics board = EKBB
potassium
lysergic acid diethylamine
list of complaints
laccate dehydrogenase
liquid chromatography-mass spectroscopy
3,4-Methylenedioxymethamphetamine
3,4-methylenedioxyamphetamine
medical doctor
norepinephrine = noradrenaline
sodium
phosphorus
pharmacokinetic
principal investigator
systolic blood pressure
Philosophical doctor (Dr. Phil. II)
tetrahydrocannabinol
serious adverse event
selective serotonin uptake inhibitor
state-trait anxiety inventory
standard deviation
Plasma eliminiation half-life
time to reach Cmax
visual analogue scale
Version 14.10.2009
3/30
Duloxetine-MDMA Study Protocol
4
Matthias Liechti
SUMMARY
3,4-methylenedioxymethamphetamine (MDMA, “ecstasy”) is widely used by young people for
its euphoric effects. MDMA releases serotonin (5-HT), norepinephrine (NE), and dopamine
through an interaction with the corresponding presynaptic monoamine uptake transporter. 5HT transport inhibitors block MDMA-induced 5-HT release in vitro or in animals and also
attenuate the subjective and cardiovascular response to MDMA in humans. NE transport
inhibitors similarly prevent the MDMA-induced release of NE in cell assays and attenuate
behavioral effects of MDMA in animals. Effects of the NE transporter inhibitor reboxetine on
the response to MDMA in humans are currently investigated. Here we suggest evaluating
effects of pretreatment with the combined 5-HT and NE transport blocker duloxetine on the
pharmacodynamics and pharmacokinetics of MDMA. The study will use a randomized
double-blind cross-over design with four experimental sessions. Duloxetine (120 mg) or
placebo will be administered 16 h and 4 h before the administration of MDMA (125 mg) or
placebo to 16 healthy volunteers. Subjective and cardiovascular responses and plasma
samples for pharmacokinetics will be repeatedly assessed throughout the experiments. We
hypothesize that duloxetine will attenuate the subjective and cardiovascular response to
MDMA.
Key words: MDMA, 3,4-Methylenedioxymethamphetamine, Ecstasy, norepinephrine,
reboxetine, pharmacokinetics
Version 14.10.2009
4/30
Duloxetine-MDMA Study Protocol
5
Matthias Liechti
INTRODUCTION
3,4-methylenedioxymethamphetamine (MDMA, “ecstasy”) is widely used by young people for
its euphoric effects. MDMA acts at the presynaptic monoamine transporter and releases
serotonin (5-HT), dopamine (DA), and norepinephrine (NE) by reversing the transport of
these monoamines. While the neurochemical effects of MDMA have been well studied in
preclinical models, it is less clear how the neurochemistry translates into to the psychotropic
and physiological effects in humans. Here, we suggest evaluating the functional role of 5-HT
and NE in the pharmacology of MDMA in humans. We plan to test the effects of
pretreatment with the selective and potent dual NE and 5-HT transport inhibitor duloxetine on
the pharmacodynamics and pharmacokinetics of MDMA in healthy human volunteers. We
hypothesize that duloxetine will attenuate subjective and cardiovascular responses to
MDMA. Such a result would indicate that 5-HT and NE are critically involved in the
pharmacology of MDMA and may provide helpful in the use and development of treatments
for Ecstasy intoxications. The study shall take place in the Clinical Research Centre (CRC)
of the Clinical Pharmacology Unit of the University Hospital of Basel.
6
SIGNIFICANCE
Use of stimulant drugs including Ecstasy is highly prevalent in our society. In Switzerland,
2.2% of 15-39 year olds reported having used Ecstasy at least once [1]. Besides from being
abused as a recreational drug, MDMA, the active substance usually found in Ecstasy pills, is
being evaluated as a treatment for post-traumatic stress disorder in several countries
including Switzerland. MDMA has also been suggested to be useful to treat and study mood
disorders due to its potential to increase empathy and rapidly elevate mood [2, 3]. Ecstasy
use can result in adverse and potentially fatal medical complications [4, 5]. Serious toxicity
due to Ecstasy use is relatively rare but may be seen in up to ten percent of patients
presenting to Emergency Departments with medical problems due to Ecstasy use [4].
Extensive Ecstasy use has also been associated with neurotoxic effects to serotonergic
brain neurons [6]. Because MDMA is widely used, a better understanding of its
pharmacology and toxicology is warranted.
7
BACKGROUND
7.1 Pharmacology of MDMA
In animals, MDMA releases presynaptic 5-HT, NE and to a lesser extent DA [7-9]. The
MDMA-induced release of these monoamines is thought to be due to reverse-transport of
monoamines through the corresponding uptake transporter. By blocking the interaction of
MDMA with the 5-HT uptake site, 5-HT uptake inhibitors (SSRIs) decrease 5-HT release [9,
10] and the behavioral response to MDMA in animals [11, 12]. In humans, SSRIs including
citalopram [13-15], fluoxetine [16], and paroxetine [17] similarly attenuated the subjective
effects of MDMA. These findings confirm that the interaction of MDMA with the 5-HT uptake
site is an important mode of action of this drug of abuse. However, blockade of MDMAinduced 5-HT release or postsynaptic antagonism of serotonin or dopamine receptors only
partly blocked MDMA’s effects and other neurotransmitter systems are likely to contribute to
the pharmacological response to MDMA [18]. In particular, recent evidence from in vitro and
animal studies indicates an important role for NE in the pharmacology of MDMA. MDMA
exhibits higher affinity for the NE transporter compared to the human 5-HT or DA uptake site
[8, 19]. In addition, MDMA was more potent in releasing NE than 5-HT or DA from
monoamine preloaded human embryonic kidney cells transfected with the corresponding
human monoamine transporters [19]. The NE uptake transporter inhibitor desipramine
Version 14.10.2009
5/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
reversed acute cognitive effects of MDMA in resus monkeys [20]. Further, doses of
stimulants, which produce amphetamine-type subjective effects in humans, correlated with
their potency to release NE and not DA release [8]. NE also plays an important role in
mediating peripheral effects of MDMA. MDMA dramatically increases circulating plasma
levels of NE [21]. The postsynaptic adrenergic 1 receptor antagonist prazosin reversed
MDMA-associated locomotor stimulation in rats [22]. In addition, blockade of the effects of
NE at postsynaptic 1 and  1-3 receptors reversed the hyperthermic response to MDMA in
rats [21]. Further, blockade of the NE transporter abolished contraction of the rat aorta
produced by 4-methylthioampethamine [23], a compound with a similar pharmacology to
MDMA. Clinically, MDMA stimulates the sympathetic nervous system and increases heart
rate, blood pressure and body temperature in controlled settings [15, 24]. Serious adverse
effects of uncontrolled Ecstasy use also include hypertensive and hyperthermic reactions
that are likely to be mediated by an activation of the adrenergic system by MDMA [5].
Together, the preclinical and clinical findings suggest that 5-HT and NE mediate most of the
psychotropic and cardiovascular effects of MDMA in humans.
7.2
Expected interactive effects of duloxetine and MDMA
7.2.1 Pharmacodynamic interactions
Duloxetine (Cymbalta) is a potent inhibitor of the 5-HT and NE transporter [25-27]. In
radioligand binding assays utilizing rat cortex synaptosomes, duloxetine possesses high
affinity for both the NE and 5-HT transporter with Ki values of 2.1 and 0.53 nM, respectively
[27]. Duloxetine is selective for the NE and 5-HT system and does not directly modulate the
DA system. The affinity of duloxetine for the 5-HT transporter is similar to that of citalopram
for this transporter (1.8 nM). We have previously shown that citalopram pretreatment
significantly reduced the psychological and cardiovascular response to MDMA in healthy
subjects [13, 15]. Because duloxetine is at least equally potent in blocking the 5-HT
transporter compare to citalopram and also blocks the NE transporter with high potency we
expect that duloxetine will be at least equally effective in decreasing the response to MDMA
as citalopram. Actually, we hypothesize that duloxetine will be considerably more effective in
reducing the cardiovascular effects of MDMA due to its additional binding to the NE
transporter which is expected to prevent MDMA from interacting with the NE transporter to
release NE [19]. The 5-HT and NE transport inhibition by duloxetine results in slight
increases in brain 5-HT and NE [26]. In the present study, however, the ability of duloxetine
to bind to the NE and 5-HT transporter is used to block the effects of MDMA which releases
5-HT and NE through an interaction with the NE and 5-HT transporter [8]. In preclinical
studies, duloxetine prevented effects of different amphetamine derivatives with structural
similarity to MDMA [26]. For example, duloxetine was shown to antagonize depletion of brain
5-HT by p-chloroamphetamine or of brain or heart NE by 6-HO-dopamine [26, 28]. No
preclinical studies have studied the interaction of duloxetine and MDMA at the level of the
NE or 5-HT transporter. Also no previous clinical study has addressed whether dual
blockade of NE and 5-HT transport effectively blocks effects of stimulant drugs in humans.
However, several compounds that block either 5-HT or NE transport have consistently been
shown to block MDMA-induced release of NE or 5-HT in various assays [7, 8, 20, 29]. As
noted above, 5-HT blockade reduced responses to MDMA in humans. Further, NE transport
blockade with desipramine decreased acute cognitive effects of MDMA in resus monkeys
[20]. In addition, recent clinical studies found that the NE transport blocker atomoxetine
attenuated cocaine-induced systolic blood pressure increases [30] and cardiovascular and
subjective responses to d-amphetamine [31]. These clinical data indicate that pretreatment
with an NE transport antagonist decreases the cardiovascular stimulant effects of cocaine or
d-amphetamine that have a very similar pharmacological action to MDMA. Together the data
indicate that duloxetine will decrease the response to MDMA in healthy subjects.
Version 14.10.2009
6/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
7.2.2 Pharmacokinetic interactions
Duloxetine is a substrate and inhibitor of CYP 2D6 [32] similar to MDMA [33]. Duloxetine
may therefore increase MDMA plasma levels and vice versa. However, because MDMA itself
inhibits CYP 2D6 (autoinhibition) the additive effect of duloxetine is expected to be small.
Pretreatment with paroxetine which is considerably more potent in inhibiting CYP 2D6 than
duloxetine increased MDMA plasma levels by only 30% [17]. Thus, we do not expect a
relevant increase in MDMA Cmax or AUC when duloxetine is administered before MDMA
(<30% increase in Cmax or AUC). On the other hand, MDMA may moderately increase Cmax
and AUC of duloxetine thereby enhancing the pharmacodynamic interaction. This potential
effect is not unwanted on the test day. However, we want to eliminate any carry-over effect
from MDMA to the next test session to avoid elevated duloxetine levels due to MDMAinduced 2D6 inhibition. Therefore, test sessions will be separated by at least 10 days [33] to
eliminate potential carry-over effects between sessions. The effect of CYP 2D6 inhibition on
the PK of duloxetine has previously been studied and is moderate. Specifically, the
maximum plasma concentration of duloxetine and the AUC increased 1.6-fold after
pretreatment with the potent CYP 2D6 inhibitor paroxetine [32]. This moderate effect of 2D6
inhibition on the metabolism of duloxetine is explained by the fact that duloxetine is mainly
metabolised by CYP 1A2 and only to a minor degree by CYP 2D6 [34].
8
STUDY OBJECTIVES
We aim to test the effect of pretreatment with duloxetine on the subjective and
cardiovascular effects of MDMA and on the pharmacokinetics of MDMA in humans.
8.1 Primary study endpoints
1. Effect of duloxetine on the subjective response to MDMA
8.2
1.
2.
3.
4.
5.
9
Secondary study endpoints
Effect of duloxetine on cardiovascular effects of MDMA
Effect of duloxetine on pharmacokinetics of MDMA
Effect of MDMA on duloxetine pharmacokinetics
Tolerability of MDMA and duloxetine
Effect of duloxetine on neuroendocrine responses to MDMA
HYPOTHESES
9.1 Primary Hypotheses
1. Duloxetine will attenuate positive mood effects of MDMA. Specifically, coadministration of duloxetine with MDMA will result in a relative decrease in “good
drug effects” VAS scores by 20% compared to the MDMA alone condition.
9.2
1.
2.
3.
4.
5.
Secondary Hypotheses
Duloxetine will reduce blood pressure and heart-rate increases produced by MDMA.
Duloxetine will increase Cmax and AUC0-6h of MDMA by <30%
MDMA will increase Cmax and AUC0-6h of duloxetine by <30%
MDMA and duloxetine will produce no severe adverse effects
Duloxetine will decrease neuroendocrine effects of MDMA
Version 14.10.2009
7/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
10 STUDY DESIGN AND METHODS
10.1 Study design
We will use a double-blind cross-over design with four treatment conditions. Treatment
conditions are placebo-placebo, duloxetine-placebo, placebo-MDMA, and duloxetine-MDMA.
Order will be balanced and pseudo-random. Each subject will participate in four 1-day study
sessions with a washout period of two weeks (>10 days) between sessions. We previously
used similar designs [18].
10.2 Study duration
1.1.2010 to 31.12.2010
10.3 Study site
Clinical Research Center, Division of Clinical Pharmacology and Toxicology, University
Hospital of Basel, Switzerland,
10.4 Study population
10.4.1 Recruitment
Subjects will be recruited by word of mouth and by advertisements placed on advertising
boards of university institutes of the University of Basel and on the homepage of the
University. Similar to previous studies [18], we intend to include mainly healthy University
students. Six-teen male or female subjects will be enrolled. Drop-outs will be replaced.
10.4.2
1.
2.
3.
4.
5.
6.
7.
8.
9.
Inclusion criteria
Age between 18 and 45 years
Sufficient understanding of the German language
Subjects understand the procedures and the risks associated with the study
Participants must be willing to adhere to the protocol and sign the consent form
Participants must be willing to refrain from taking illicit psychoactive substances
during the study.
Participants must be willing to drink only alcohol-free liquids and no xanthinecontaining liquids (such as coffee, black or green tea, red bull, chocolate) after
midnight of the evening before the study session. Subjects must agree not to smoke
tobacco for 1 h before and 4 hours after MDMA administration.
Participants must be willing not to drive a traffic vehicle in the evening of the study
day.
Women of childbearing potential must have a negative pregnancy test at the
beginning of the study and must agree to use an effective form of birth control.
Pregnancy tests are repeated before each study session.
Body mass index: 18-25 kg/m2
10.4.3 Exclusion criteria
1. Chronic or acute medical condition including clinically relevant abnormality in physical
exam, laboratory values, or ECG. In particular: Hypertension (>140/90 mmHg).
Personal or first-grade history of seizures. Cardiac or neurological disorder.
2. Current or previous psychotic or affective disorder
3. Psychotic or affective disorder in first-degree relatives
4. Prior illicit drug use (except THC-containing products) more than 5 times or any time
within the previous 2 months.
5. Pregnant or nursing women.
6. Participation in another clinical trial (currently or within the last 30 days)
Version 14.10.2009
8/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
7. Use of medications that are contraindicated or otherwise interfere with the effects of
the study medications (monoamine oxidase inhibitors, antidepressants, sedatives
etc.)
11 STUDY PROCEDURES
11.1 Schedule of Events
The schedule of events for a subject is shown on table 1. Subjects take part in a 2 h
screening session, four 10 h test sessions, and a 1 h end of study (EOS) visit.
Table 1: Schedule of Events
Time
Event
Duration (h)
Informed consent
Interview
Medical exam
BP, HR
ECG
Drug screen
Pregnancy test
Blood tests
CYP2D6 phenotyp
PK blood samples
Adverse events
Week 1
Week 2
screening Session 1
2
10
x
x
x
x
x
x
x
(x)
(x)
(x)
x
x
x
x
Week 4
Session 2
10
Week 6
Session 3
10
Week 8
Session 4
10
Week 8
EOS
1
x
x
x
x
x
x
(x)
(x)
(x)
(x)
(x)
(x)
x
x
x
x
x
x
x
x
BP=blood pressure, HR=heart rate, ECG=electrocardiogram, CYP=cytochrome P450 enzyme, PK=pharmacokinetik,
EOS=end of study tests
11.2 Screening procedure
11.2.1 Informed consent
The subjects will be informed about the study both verbally and by the approved written
consent form regarding MDMA and duloxetine, the study procedures, and associated risks,
in particular including those with the use of MDMA [35]. The investigator and the subject will
both personally sign and date the consent form as confirmation of consent.
11.2.2 Physical health
Subjects will be examined by a research physician. Basic health will be ensured by general
medical examination including medical history, physical examination, ECG, and blood
chemistry and hematology. Body weight will also be measured.
11.2.3 Mental health
Subjects will be screened by an MD using a semi-structured clinical interview for DSM IV [36]
to exclude those with a personal or family (first-degree relative) axis I major psychiatric
disorder or a history of illicit drug dependence.
11.2.4 History of Drug use
Occasional recreational drug use in the past is not an exclusion criterion if no adverse
reactions occurred and if use was moderate and controlled. Drugs screens are primarily
intended to document and control for concomitant drug use and not as a means to exclude
subjects from the study. However, subjects will be asked to abstain from any illicit drug use
during the study and drug screens are performed during screening and before each session.
Positive screens for stimulants, opioid or hallucinogens will result in exclusion from the study
(next study session). Subjects will also be asked to abstain from excessive alcohol
consumption between test-sessions and in particular to limit their use to one glass on the
day before the test sessions. Smokers will be asked to maintain their usual smoking habit
Version 14.10.2009
9/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
but will not be allowed to smoke before and up to for four hours after MDMA administration.
Positive screens for THC (cannabis) will be recorded but do not result in study exclusion.
11.2.5 Screening laboratory tests
The following laboratory blood tests are performed at the screening examination: Na, K,
creatinine, ASAT, ALAT, -GT, hemoglobin, hematocrit, white blood cell count, red blood cell
count, platelet cell count. Urine drug screen (Triage-8, Biosite, Morges, CH). Urine
pregnancy test.
11.2.6 CYP2D6 phenotyping
Subjects will be asked to ingest a tablet of dextromethorphan (Bexin®) 25 mg in the evening
of the screening day or on a day prior to the test sessions (10-11PM) and to collect and
measure the amount of urine during the next 8 h (night urine plus first morning urine). Of this
urine two test vials need to be sent to our laboratory in a prepared envelope.
11.2.7 Genotyping
Polymorphisms in gens that code for monoamine transporters (SLC6A2A and others)
contribute to interindividual variations in the clinical response to amphetamines [37]. We will
collect a blood sample at the screening visit for genotyping of monoamine transporter genes.
In addition genotyping of CYP enzymes are planned (CYP2D6 genotype).
11.2.8 Personality
Personality traits are known to affect subjective responses to amphetamines [38]. The
Multidimensional Personality Questionnaire (MPQ) [39] and the Freiburger Personality
Inventory [40] will be used to assess personality traits and their potential modulatory effects
on the response to MDMA.
11.2.9 Menstrual cycle phase in women
Women exhibit differential subjective effects to amphetamines during the follicular as
compared to the luteal phase of the menstrual cycle [41]. Therefore, female subjects will
preferentially be tested during the follicular phase (day 2-14) of the cycle.
11.3 Schedule of experimental session
Table 2 shows the schedule of events for each experimental session. Each of the four test
sessions will last 10 h from 8:00 AM to 6:00 PM. Subjects will arrive at 8:00 AM at our CRC.
Subjects will take duloxetine (120 mg) or placebo the night before the study session at 8:00
PM and another dose of duloxetine (120 mg) or placebo in the morning of the study day
(8:00 AM). An indwelling intravenous catheter will be inserted into a subcutaneous vein of
the forearm of the nondominant arm. MDMA (125 mg) or placebo is administered 4 h after
duloxetine (12:00 AM). The acute effects of MDMA are expected to last for 4 h [42, 43].
Outcome measures (see below) will repeatedly be assessed during the study session. The
sessions end at 6 PM. Subjects will be under continuous medical control until the effects of
the drug have completely subsided (within 4-6 h after MDMA administration).
Version 14.10.2009
10/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
Table 2: Schedule of events for each session
Time
relative Time (min/h)
Duloxetine (120 mg)
MDMA (125 mg)
neuroendocrine samples
cortisol, prolactin
copeptin
sodium, urea, osmol
catecholamines, 5-HT
urine samples
sodium, osmol
PK blood sample
Psychometrics
VAS
AMRS, STAI
ARCI
LC
5D-ASC
AE
Physiological measures
blood pressure
heart rate1
body temperature
pupil diameter
Cognitive Tests
RME
CRTT
20:00
X
08:00 12:00 12:20 12:40 13:00 13:15 13:30 14:00 14:30 15:00 16:00 17:00 18:00
-4
0
20
40
60
75
90
120 150 180
4
5
6
X
X
0
0
0
0
0
-1
-1
0
0
0
1
1
1
2
1
0
1
2
3
0
1
2
3
12:00 blood
24 samples
4
4
1
1
5
5
2
2
2
2
3
6
7
8
6
7
8
1
9
9
3
2
11
10
1
2
1
2
1
-1
-1
-1
-1
0
0
0
0
-1
0
1
1
1
1
2
2
2
2
3
3
3
3
4
4
4
4
5
5
5
5
6
6
6
6
7
7
7
7
8
8
8
8
9
9
9
9
10
10
10
10
1
1
PK=pharmacokinetic, AMRS=adjectiv mood rating scale, STAI=state-trait anxiety inventory, 5D-ASC=altered states of
consciousness scale, VAS=visual analogue scales, ARCI=adiction reserach inventory, LC=list of complaints, AE=adverse
events, CRTT= Choice reaction time test, RME= Reading the mind in the eye, 1heart rate is also measured continuously
20
11.4 Assessments and Measures
11.4.1 Psychometric assessments
11.4.1.1 Visual Analogue Scales (VAS)
Previously tested and sensitive VAS will be repeatedly used to better characterize subjective
drugs effects over time [16, 44-46]. VAS will be presented as are 100 mm long horizontal
lines marked with “not at all” on the left and “extremely” on the right [46]. The following VAS
will be used: “drug effect”, “good effect”, “bad effect”, “liking”, “high”, “energy level”, “feelings
of closeness to others”, “mind racing”, “ability to concentrate”, “fear”, “hungry”, “sedated”,
“talkative”, “open”, and “confused”. Scales will be administered 4 h before and 0, 20, 60, 90,
120, 150, 180 min, and 4, 5, and 6 h after MDMA/placebo administration [45].
11.4.1.2 Adjective mood rating scale (AMRS)
An adjective mood rating scale (AMRS or EWL60S) is a 60-item Likert scale that allows
repeated assessment of mood in 6 dimensions: activation, inactivation, well-being,
anxiety/depressed mood, extro- and introversion, and emotional excitability. The scale is
sensitive to the effects of MDMA [13, 24, 42, 47]. It will be used before, 75, 120 min and 5 h
after MDMA administration. The German EWL60S version is used [48].
11.4.1.3 State-trait anxiety inventory (STAI)
The state-trait anxiety inventory (STAI) yields a score for state and trait anxiety levels [49].
The trait scale will be administered once. The state anxiety scale will be used before, 75, 120
min and 5 h after MDMA administration.
11.4.1.4 Addiction Research Center Inventory (ARCI)
The Addiction Research Center Inventory (ARCI) is a true-false questionnaire with five
empirically derived scales: pentobarbital-chlorpromazine-alcohol group, a measure of
Version 14.10.2009
11/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
sedation; morphine-benzedrine group, a measure of euphoria; lysergic acid diethylamine
group (LSD), a measure of dysphoria and somatic symptoms; benzedrine group, a stimulant
scale consisting mainly of items relating to intellectual efficiency and energy; and
amphetamine, an empirically derived scale sensitive to the effects of d-amphetamine [50]. It
has previously been shown to be sensitive to the effects of MDMA [16, 17] and is available in
a validated German version [51]. The ARCI will be administered 4 h before and 150 min after
MDMA administration.
11.4.1.5 Altered states of consciousness (5D-ASC)
The 5 dimensions of altered states of consciousness (5D-ASC) scale is a visual analogue
scale consisting of 94 items [52]. The instrument is constructed of five scales and allows
assessing mood, anxiety, derealization, depersonalization, changes in perception, auditory
alterations, and reduced vigilance. The scale has repeatedly been used to evaluate
subjective responses to MDMA [24, 42, 47] or other psychoactive substances [53]. The 5DASC scale will be administered once 4 h after MDMA administration to retrospectively
assess drug peak effects.
11.4.2 Physiological assessments
11.4.2.1 Vital signs
Blood pressure, heart rate, body temperature, and pupil diameter will be recorded at -4 h, 0,
20, 40, 60, 90, 120, 150, 180 min, and 4, 5, and 6 h after MDMA/placebo administration.
Blood pressure (systolic and diastolic) and heart rate will be measured on the leading arm
using an automatic oscillometric device. Heart rate will also be measured continuously with a
RS 800 Polar device (Polar Electro Oy, Kempele, FIN), set to sample the average heart rate
every 5 s, and analyzed using Polar Precision Performance software. Body temperature will
be measured using an ear thermometer. Ambient temperature is also recorded trice on the
experimental day. Pupil diameter and the reaction to a light stimulus will be recorded using a
handheld dynamic pupillometer (PRL-200, Neuroptics, Irvine, CA, USA).
11.4.2.2 List of complaints (LC) and Adverse effects (AE)
Adverse effects (AE) will be assessed systematically during the session using a modified list
of complaints (LC) [54]. The scale consists of 66 items, yielding a global score measuring
physical and general discomfort. The LC list is administered 4 h before and 180 min after
MDMA/placebo. An additional LC is completed 24 h after MDMA/placebo in the absence of
the experimenter. Additional AE during the session will be listed in the case report form
(CRF) 5 h after MDMA/placebo administration. Subjects will also be asked to report AE
between study sessions at the beginning of the next session.
11.4.3 Cognitive and psychomotor function
11.4.3.1 Reading the mind in the eyes test (RME)
A 10-min computerized “Reading the Mind in the Eyes Test” is performed 90 min after
MDMA/placebo administration (before the CRTT). The test assesses drug effects on
inferring mental states from the eye region. 36 pictures of the eye regions of different
persons will be presented to the participants on a computer screen with four alternative
labels describing what the person displayed might be thinking or feeling at the moment [55].
11.4.3.2 Choice reaction time test (CRTT)
This five choice reaction time test (CRTT) assesses a combination of sustained attention
and executive motor function [56]. The CRTT will last for 3 min and requires the subject to
Version 14.10.2009
12/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
quickly and accurately press a button of the same color as the one of five (red, blue, white,
yellow, or green) which flashed. The stimuli are presented in random sequence. Using a
computer-based algorithm, the interstimulus interval is adjusted by 50 ms according to
performance in a continuously moving window of the last seven stimulus–response pairs.
The interstimulus interval decreases when four or more stimulus–response pairs were
correct; otherwise, the interval increases. Thus, test difficulty is adjusted to achieve a subject
false response rate of 50%, which makes for a demanding task. The dependent variable is
the mean reaction time (for correct responses) for the last 2 min of the test. Reaction times
less than 300 ms will be considered artifactual and are therefore discarded. Subjects will go
through one training session at the beginning of each session. Tests will then be performed
0 min and 90 min after MDMA administration.
11.4.3.3 End of Session Questionnarie
At the end of each session, participants will be asked to indicate how much they liked the
drug overall and how strong the overall effect was on separate VAS [16].
11.4.4 Neuroendocrine function
MDMA increases cortisol, prolactin, catecholamine, and 5-HT concentrations in plasma in
humans [43, 57-59]. MDMA also increases vasopressin (AVP) potentially leading to
hyponatriemia [59, 60]. We will test the effect of duloxetine on MDMA-induced increases in
plasma cortisol, prolactin, catecholamines, serotonin, and metanephrines (norepinephrine
and epniephrine metabolites). Plasma cortisol and prolactin samples will be collected at 0
min and at 120 min (tmax) after MDMA/placebo administration (2x7.5 ml blood). Copeptin is
the C-terminal part of provasopressin. Copeptin is released stoichiometrically together with
AVP and is easier to measure than AVP. Copeptin samples will be collected at 0 min and
120 min after MDMA/placebo administration when peak AVP values are expected [59] (2x7.5
ml blood). Plasma and urine sodium, urea, and osmolality are simultaneously assessed
(2x7.5 ml blood, 2x10 ml urine). Catecholamine and metanephrine samples will be collected
at 0, 60, and 120 min after MDMA/placebo administration (3x7.5 ml blood).
11.4.5 Blood sample collection (pharmacokinetics)
Blood samples (11x7.5 ml) will be collected on the study day -4, 0, 0.33, 0.67, 1, 1.5, 2, 2.5,
3, 4, and 6 h after MDMA or placebo administration. This will allow us to assess the
pharmacokinetics of both MDMA and duloxetine and potential PK interactions of the two
compounds. The design is optimized to assess effects of duloxetine on peak plasma levels
of MDMA (time to Cmax = 2h) and AUC0-6h for MDMA [43]. Blood samples will be collected in
heparinized tubes, centrifuged at 4C at 2000 rpm for 10 min, and the plasma is then stored
at -20C (two aliquots) until analysis. Collection tubes labels will include the following
information: study number, study day, subject number, and sample number. The date and
actual blood sampling time will be recorded on the sample collection page in the CRF.
11.4.5.1 Amount of blood samples
On each study day a total of 20 blood samples (20x7.5 ml = 150 ml) will be collected on the
study day. During the whole study approximately 600 ml of blood are collected. This amount
to 20% more than a standard blood donation. However, blood is collected in amounts of 150
ml every 2 weeks spread out over 6 weeks and not at once which allows for regeneration. In
order to blind the study, plasma samples will be collected also on the placebo day.
Version 14.10.2009
13/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
11.4.6 End of Study (EOS) visit
11.4.6.1 EOS Examination
Clinical examination and vital signs. Blood tests: Na, K, creatinine, ASAT, ALAT, -GT,
hemoglobin, hematocrit, white blood cell count, red blood cell count, platelet cell count.
Adverse effects.
11.4.6.2 EOS Questionnaire
A debriefing interview will be performed including a retrospective comparative evaluation of
the subjective experience of all four study sessions. Subjects will be asked to rate the overall
drug effect for each of the four sessions. Subjects will also be asked to guess the
assignment of the study drugs to the four study sessions to assess whether subjects were
able to distinguish the study conditions (duloxetine vs. placebo). They will also be asked to
provide a feed-back on the study and their experience and to indicate whether they would
take the drug again in a controlled or recreational setting [15].
11.5 Study Drugs
11.5.1 Duloxetine
Duloxetine (Cymbalta) is a potent and selective 5-HT and NE reuptake inhibitor [27].
Duloxetine (60 mg gelatine capsules) will be obtained from Eli Lilly (Suisse) SA, Vernier. The
investigator pays for the product. Eli Lilly does not act as a sponsor for the study. The
original company product will be used. However, capsules will be encapsuled with opaque
dark-colored gelatine capsules and similar placebo capsules containing lactose will be
prepared by a pharmacist. Clinically used doses: Duloxetine is used clinically in the
treatment of depression, generalized anxiety disorder, diabetic and other neuropathy, and
fibromyalgia. The clinically used common starting dose is 60 mg/day. Maintenance doses
are 60-120 mg/day [25, 61, 62]. Several clinical trials have used 120 mg/day given once or in
two doses of 60 mg twice per day. The suggested maximum dose is 120 mg daily.
Tolerability: Duloxetine was well tolerated at doses of 60-120 mg/day [63]. See 14.1. for
expected adverse effects of duloxetine. Pharmakokinetics (dose finding and dosing
intervals): Elimination half-life of duloxetine is 12 h and Tmax is 6 h [61, 64]. On the test day,
duloxetine (120 mg) will be given at 8 AM 4 h before MDMA administration at 12 AM so that
the duloxetine Cmax will be reached shortly after the onset of the subjective MDMA effect (1h
after MDMA administration) and together with Cmax of MDMA. An additional dose of
duloxetine (120 mg) will be administered the day before MDMA at 8 PM, 16 h before MDMA
administration. At both time-points duloxetine will be administered in a dose of 120 mg
(maximum suggested therapeutic dose). Because the study uses only one dose of
duloxetine it is important to use a relatively high dose to ensure effective plasma levels.
Mean duloxetine plasma Cmax values were 22, 44, and 61 ng/mL after single oral doses of
30, 60, and 90 mg of duloxetine in healthy volunteers [64]. At steady state 40 mg once daily
resulted in a mean Cmax level of 35 ng/mL and a trough level of approximately 10 ng/mL at
24h [32]. Population PK studies showed mean Cmax levels of 30-60 ng/mL follwing daily
administration of 60 mg and depending on gender, smoking status, age and other variables
[65]. Mean trough levels at 24h were in the range of 25 ng/mL. Women typically had 64%
higher average steady-state concentrations than men (with considerable overlap). Similarly,
nonsmokers had 43% higher average concentrations than smokers. Doubling the duloxetine
dose from 30 mg to 60 mg and from 60 mg to 120 mg resulted in 2.3 fold and 2.6 fold higher
average concentration values, respectively [65]. Based on these data we expect mean Cmax
Version 14.10.2009
14/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
levels of 120-150 6 h after the administration of the second dose of 120 mg of duloxetine
with the first dose (120 mg) given 12 h before the second dose. This Cmax is similar to the
one seen in patients treated with daily doses of 120 mg under steady state conditions (peak
100 ng/mL, trough 50 ng/mL) [65]. PD and PK interactions of duloxetine and MDMA (see
7.2.1/2). Relevant clinical studies using duloxetine in healthy volunteers: Duloxetine (60
mg for 14 days) was not able to block the thyramine-induced increases in systolic blood
pressure, indicating that the dose might be too low to block the NE transporter to a relevant
extent in this test [66]. A dose of 80-120 mg was effective in suppressing the blood pressure
increase due to thyramine [25]. However, there is clinical evidence that lower doses of
duloxetine block NE transport. For example, urine catecholamine changes indicative of NE
reuptake inhibition were present already at 80 mg. We suggest that a dose of 120 mg should
be used for the present study. Of note, desipramine completely blocked thyramine-induced
SBP responses which could indicate that desipramine interacts with stimulants at a different
site on the NE transporter than duloxetine.
11.5.2 MDMA
11.5.2.1 Chemistry, manufacturing, control
Pharmaceutically pure racemic MDMA HCl (±3,4-methylenedioxymethamphetamine
hydrochloride) (Lipomed AG, Arlesheim, Switzerland) will be used similar to previous studies
[47, 67, 68]. Quality control of the drug confirmed purity (>98%) and content of the batch and
absence of decomposition products. MDMA from this lot has previously been used in human
studies [67]. MDMA from the same batch is also being used in clinical trials in several
countries including Switzerland [69]. MDMA will be prepared as capsules (100 mg and 25
mg) by a BAG authorized laboratory according to good manufacturing practice (GMP)
criteria. Identically looking placebo capsules will contain lactose and will be provided by the
same laboratory. An updated quality control certificate for the final study drug preparations
and all labeling information will be provided before study initiation.
11.5.2.2 Metabolism and Pharmacokinetics
After a single dose of MDMA tmax is attained at 120-144 min [44, 70]. T1/2 of MDMA (100 -125
mg dose) is about 8-9 h [71]. MDMA inhibits its own metabolism possibly by forming an
enzyme-metabolite complex with CYP2D6. This autoinhibition likely explains the suggested
non-linear pharmacokinetics of MDMA [33, 70-72]. There are two main metabolic pathways
for the metabolism of MDMA [71]. MDMA is N-demethylated to 3,4methlyenedioxyamphetamine (MDA). MDA is an active, but minor metabolite. CYP2D6mediated O-demethylenation followed by COMT-catalyzed methylation leads to 4-hydroxy-3mehoxymethamphetamine (HMMA) or 4-hydroxy-3-methoxyamphetamine (HMA). HMMA is
the main metabolite of MDMA [71]. HMMA and HMA are rapidly glucuronidated or sulphateconjugated.
11.5.2.3 Dose selection
An oral dose of 125 mg MDMA corresponding to about 1.5-1.7 mg/kg will be used similar to
previous studies [42 , 44, 46, 47, 58, 70]. This dose of pure MDMA produces moderat
subjective and cardiovascular effects [24, 42, 47, 67] and is considered safe in a controlled
clinical setting [2, 35, 70, 73]. The same dose of 125 mg followed 2.5 h later by 62.5 mg
MDMA is currently being used in a clinical study [69]. Higher doses (150 mg or 2 mg/kg)
have also been used without adverse events in MDMA studies [42, 69, 74, 75] and are used
in clinical studies as well (150 mg plus 75 mg 2.5h later). Ecstasy tablets usually contain 80160 mg of MDMA [76]. Use of more than one tablet at once is common.
Version 14.10.2009
15/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
11.5.3 Drug-Accountability, storage and return of study drugs
The study drugs will be stored in a locked drug cabinet in a lockable room with restricted
access at room temperature (15-25C) at the clinical research center. A drug dispensing and
accountability log for all capsules will be kept current. The log includes the date, origin, and
amount of incoming study drug. The study physician/investigator who dispenses the drug,
registers and signs out the drug quantity, dispensing date of dispensing, participant number
for all outgoing medications.
11.5.4 Concomitant medications
Concomitant use of medications will be recorded at the screening visit and at the beginning
of each study session. Subjects are asked to abstain from using drugs that may interfere
with the effects of the study medications including sleeping aids, cough medications,
betablocker or other substances with potentially relevant psychoactive effects. The PI may
exclude subjects from study participation or reschedule sessions in the case of relevant
pharmacokinetic of pharmacodynamic interactions with concomitant medications.
11.5.5 Randomization and Blinding
Each subject will receive all treatments. Subjects and study personnel will be blinded to
treatment order. Order is balanced and pseudo-random. A study subject number will be
assigned to each subject. A treatment order is assigned to each subject number (code).
Treatments are prepared and labeled according to the code by a pharmacist not involved in
the study. The PI will also receive the code enclosed in an envelope. In case of a medical
emergency the code can be accessed.
11.5.6 Compliance
Subjects are provided with the duloxetine medication box for each session and are reminded
to take the medication by a phone call the day before the study session. Compliance is
assessed by asking the subject to report the time of drug intake, by collection of the empty
medication box, and by determination of duloxetine in plasma (PK sample -0 and 1). During
the study day medications are administered under observation by the study personnel.
12 Analytics
12.1 MDMA and metabolites
MDMA, its active metabolite MDA and the main inactive metabolites HMMA and HMA will be
determined in plasma by gas chromatography-mass spectrometry (GC-MS) by established
techniques as previously described [77, 44, 78].
12.2 CYP2D6 Phenotype
Dextrorphan/dextromethorphan ratio will be used for CYP2D6 phenotyping [79, 80].
12.3 Cortisol and prolactin
Plasma cortisol and prolactin concentrations will be determined by standard immunoassay
techniques.
12.4 Catecholamines and metanephrines
Plasma metanephrines, catecholamines, and 5-HT will be measured by HPLC with
electrochemical detection at the University Hospital Lausanne.
Version 14.10.2009
16/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
12.5 Copeptin
Plasma copeptin levels will be measured with a sandwich immunoassay (BRAHMS AG,
Hennigsdorf, Berlin, Germany) [81].
12.6 Duloxetine
Analysis of duloxetine plasma levels will be performed using LC-MS-MS.
13 Data analysis
13.1 Sample size estimation
Power analysis was performed with PASS®, Hintze J. Kaysville, Utah, US. We consider a
relative reduction of 20% (MDMA vs. duloxetine+MDMA) in “good drug effects” to be
clinically meaningful [13, 16, 17, 68]. A sample size of 13 achieves 82% power to detect a
difference of 20 between the null hypothesis mean of 100 and the alternative hypothesis
mean of 80 with a known standard deviation of 25 [13, 68] and with a significance level
(alpha) of 0.05 using a two-sided one-sample t-test. For the most relevant secondary
outcome, which is Psys, we expect that duloxetine will decrease the MDMA-induced increase
in Psys by 10 mmHg from +30 mmHg (MDMA vs. placebo) to +20 mmHg MDMA+duloxetine
vs. duloxetine). This effect of duloxetine would be relevant and similar to the one of
citalopram on MDMA-induced increases in Psys [15]. A sample size of 8 achieves 81% power
to detect a difference of 10 between the null hypothesis mean of +30 and the alternative
hypothesis mean of +20 with a known standard deviation of the difference of 10 [15] and
with a significance level (alpha) of 0.05 using a two-sided one-sample t-test. Assuming a
drop-out rate before inclusion of 50% we will have to screen 32 subjects to include 16 in the
study. Lower (8-12) [16, 17] or similar sample sizes [13, 47, 68, 82] were used in previous
comparable studies. Subjects who prematurely discontinued the study for any reason will be
replaced if the number of discontinued subjects exceeds three. A samples size of 8 or less is
commonly used in PK studies. A larger sample is used here to produce meaningful results
for the pharmacodynamic parameters.
13.2 Analysis of outcomes
Repeated-measures analysis of variance (ANOVA) with drug (MDMA and duloxetine) as
within-subject factors followed by Tukey’s post-hoc tests will be use to analyze differences
between the different treatment conditions. Transformations will be performed before
analysis, if response variables are nonnormally distributed. Pharmacokinetic parameters
(Cmax, tmax, AUC from 0 to 6 h) will be obtained using PK Functions for Excel®. AUC values
will be calculated by the trapezoidal rule. Additional PK analyses may be performed using
WinNonLin® and GraphPad®. Statistical analyses will be performed using Statistica®
(StatSoft).
14 Protection of subjects
Trained study personnel will place venous accesses, administer the study drugs and collect
blood samples. Subjects will be supervised by trained study personnel during the entire test
sessions, backed-up by study physicians. After the test session subjects are allowed to leave
only if the subjective and cardiovascular effects of MDMA have completely ceased (see
14.2.5). In the case of unexpected prolonged or severe adverse reactions subjects will
remain at the CRC and are monitored as long as necessary. Subjects will be provided with
the phone number of the study physician on call in case of an emergency between/after
study sessions.
14.1 Potential risks
Physical risks:
Version 14.10.2009
17/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
MDMA: A large series of phase I like studies have been performed using MDMA [13-16, 24,
42, 44-47, 57, 58, 67, 68, 70, 71, 74, 75, 82-91]. This considerable body of evidence
indicates that the likelihood for significant toxicity from doses of MDMA such as the one to be
used in the present study and in controlled settings is very low. Thus, no lasting biological or
psychological injury is expected. See below for specific MDMA toxicity and monitoring.
Expected adverse effects of MDMA: MDMA is expected to produce moderate acute adverse
effects such as increased jaw muscle tension, dry mouth, dizziness, palpitations, tremor,
nausea, headache, insomnia, and transient anxiety in more than 10% of subjects, [15, 42].
MDMA is expected to produce sequelae (1-3 days post MDMA) including difficulty
concentrating, irritability, or slightly depressed mood in more than 10% of subjects [15]. No
severe or serious adverse effects are expected. Duloxetine: In placebo-controlled doubleblind trials in patients duloxetine (60-120 mg/day) produced nausea (20-29%), headache
(20%), dry mouth (15-18%), insomnia (11-15%), fatigue (14%), dizziness (9-11%),
constipation (11%), decreased appetite (8%), and sweating (6%) [63]. Adverse effects of
duloxetine in healthy volunteers similary include nausea, dry mouth, insomnia, dizziness,
sweating, and clinically non-significant increases in blood pressure or heart rate [25, 61, 92].
No severe or serious adverse effects are expected.
Venipunction: There is a risk for pain, bruising and thrombophlebitis.
Risk to privacy of subjects: Potential risks of data collection include breach of confidentiality.
Financial risks: There is no risk of expense to the subject. Insurance coverage is provided.
Travel expenses are expected to be minimal and are included in the compensation of 1000
CHF per subject.
14.2 Monitoring of toxicity
14.2.1 Safety definitions
Adverse Event (AE):
Any untoward medical occurrence in a clinical trial subject administered an
investigational medicinal product (IMP) and which does not necessarily have a causal
relationship with this treatment. An AE can therefore be any unfavorable and
unintended sign (including an abnormal laboratory finding), symptom, or disease
temporally associated with the use of an (IMP), whether or not considered related to
the IMP.
Adverse Reaction (AR):
All untoward and unintended responses to an IMP judged by investigator/sponsor as
having a reasonable causal relationship to the IMP. The expression reasonable
causal relationship means to convey in general that there is evidence or argument to
suggest a causal relationship.
Unexpected Adverse Reaction (UAR):
An AR, the nature or severity of which is not consistent with the applicable product
information (e.g. investigator’s brochure for an unapproved investigational product or
summary of product characteristics (SmPC) for an authorized product). When the
outcome of the adverse reaction is not consistent with the applicable product
information this adverse reaction should be considered as unexpected. Side effects
documented in the IB or SmPC which occur in a more severe form than anticipated
are also considered as being unexpected.
Serious Adverse Event (SAE) or Serious Adverse Reaction:
Any untoward medical occurrence or effect that at any dose results in death, is lifethreatening, requires hospitalization, or prolongation of existing hospitalization,
results in persistent or significant disability or incapacity or is a congenital anomaly or
Version 14.10.2009
18/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
birth defect. In this context, the term life threatening refers to an event in which the
trial participant was at immediate risk of death at the time of the event; it does not
refer to an event, which might have caused death if it were more severe.
Suspected Unexpected Serious Adverse Reaction (SUSAR):
Any suspected adverse reaction related to an IMP that is both unexpected and
serious.
Causality:
Most adverse events and adverse reactions that occur in this study, whether they are
serious or not, will be expected treatment-related toxicities due to the medication
used in this study. The assignment of the causality will be made by the investigator
using the definitions in the table below.
Relationship
Description
Unrelated
There is no evidence of any causal relationship.
Unlikely
Possible
Probable
Definitely
Not assessable
There is little evidence to suggest there is a causal relationship
(e.g. the event did not occur within a reasonable time after
administration of the IMP). There is another reasonable
explanation for the event (e.g. the participant’s clinical condition,
other concomitant treatment).
There is some evidence to suggest a causal relationship (e.g.
because the event occurs within a reasonable time after
administration of the IMP). However, the influence of other
factors may have contributed to the event (e.g. the participant’s
clinical condition, other concomitant treatments).
There is evidence to suggest a causal relationship and the
influence of other factors is unlikely.
There is clear evidence to suggest a causal relationship and
other possible contributing factors can be ruled out.
There is insufficient or incomplete evidence to make a clinical
judgment of the causal relationship.
14.2.2 Documentation
All adverse events occuring after the subject has signed the informed consent will be fully
recorded in the subject’s CRF. Each event will be described in detail along with start and
stop dates, severity, relationship to investigational product, action taken and outcome.
14.2.3 Adverse events (AE) documentation
AE events will be described and recorded on the subject’s CRF, regardless of the severity or
relationship to the drug. Adverse effects are assessed by the standardized list of complaints
(LC) [54]. Additional AEs are entered in the CRF at the end of the study session. AEs are
also assessed 24 h after MDMA/placebo using the LC. Further, AEs are assessed before the
next session 2 weeks after MDMA/placebo. AEs are rated for severity and the potential
relationship to the study medications will be evaluated by the investigator according standard
criteria. Subjects with AEs will be treated appropriately. Abnormal laboratory values will be
repeated until normal or until the abnormality can be explained and the subject’s safety is not
at risk. In cases of a medical emergency, treatment is available in house and includes the
24h-availability of the reanimation team and intensive care facilities.
Version 14.10.2009
19/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
14.2.4 SAE and SUSAR reporting
Serious adverse events (SAE) are not expected. Should a SAE occur the PI will be informed
and relationship to the study drug will be assessed by the PI using the definitions shown
above (14.2.1.). Complete information concerning the SAE will be collected by the PI and
documented on a standard SAE form. Any SUSAR will be reported to the IRB and
Swissmedic by the PI according to guidelines and within 7 days. The PI acts as sponsor in
this investigator-initiated study. Therefore no reporting to a sponsor is needed. An annual
safety report will be sent to Swissmedic by the PI.
14.2.5 Medical follow-up of adverse events
The investigator will ensure the subject receives medical follow-up as necessary until the
condition has stabilized or returned to normal state, even if the period of the trial is over.
14.2.6 Monitoring for specific toxicity
Duration of monitoring: Subjective and cardiovascular effects of MDMA last for a mean of
3.5 h and completely subside within 5-6 h [13, 42, 43, 58, 67]. Accordingly subjects are
monitored for 6 h after MDMA administration. In our previous studies, that similarly evaluated
interactive effects of MDMA with various pretreatments, subjects were dismissed 4 h after
MDMA administration [13, 15, 68, 82]. Another interaction study used an observation time of
6 h after MDMA administration similar to the present study [43]. Only one to two subjects are
tested simultaneously on the same test day.
Cardiovascular effects: Blood pressure and heart rate are measured at repeated intervals
(Table 2). Closer monitoring will be implemented if blood pressure values exceed 180/120
mm Hg. Subjects will be monitored for signs of hypertensive crisis such as headache, angina
pectoris or neurological deficits. Treatment of a hypertensive reaction (Psys>220 mmHg)
would include administration of oxygen, lorazepam, nitroglycerin or nifedipin or/and
carvedilol. In the case of angina pectoris, oxygen is given and ECG monitoring is initiated
and cardiac enzymes will be measured. Subjects will remain at the CRC overnight for further
observation if blood pressure is >140/95 mmHg or pulse rate is >100/min at the end of the
test session at 6 PM.
Psychological effects: The effects of MDMA are mostly pleasurable. Dysphoric reactions to
MDMA and moderate anxiety may occur at the onset of the MDMA effect. Such effects have
always resolved within hours [42]. Unexpected severe anxiety would be treated with a
benzodiazepine. Up to one third of the subjects is expected to experience slightly depressed
mood including emotional irritability, lack of energy, brooding and bad dreams for up to 3
days following MDMA administration [42]. These effects will be specifically monitored using
an adverse effects check list. After the test session subjects are allowed to leave only if the
subjective effects of MDMA have completely ceased (VAS score “drug effect” back to
predose ± 10%).
Hyperthermia: Increases in body temperature of approximately 0.5C are expected [42]. If
body temperature rises by more than 1.5C appropriate treatment including hydration and
cooling will be initiated [5]. Subjects will remain at the CRC overnight for further observation
if body temperature has not returned to pretreatment levels ± 0.5 C at the end of the test
session at 6 PM.
Dehydration/Hyperhydration: Ecstasy use has been associated with both dehydration and
hyperhydration leading to hyponatremia [5]. Subjects will be encouraged to drink no less
than 1 but no more than 3 l during one session. If symptoms of hyponatremia such as
confusion or vomiting are present, subjects will be further examined and serum sodium
levels checked.
Hepatotoxicity: Ecstasy use has been associated with idiosyncratic liver toxicity [5]. Liver
enzymes will be measured at the beginning and end of the study and any pathological
results will be followed up.
Neurotoxicity: Recreational use of Ecstasy has been linked to reversible neurotoxicity [93,
94], especially when the drug is taken frequently and in high doses [6, 93]. In contrast,
Version 14.10.2009
20/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
several studies document no lasting effects of Ecstasy in moderate Ecstasy users [93, 9597]. There is no indication that administration of MDMA in a controlled setting has any
adverse effects on cognitive function [2, 89, 98]. The use of pure MDMA in controlled
settings is considered safe with regard to neurotoxic effects [35, 73]. No changes in
serotonin transporter density were seen using positron emission tomography and [11C]McN5652 four weeks after MDMA administration (1.5-1.7 mg/kg) in MDMA-naïve human
volunteers [99, 100]. We will administer only two single moderate doses of MDMA similar to
previous clinical studies [13, 16, 17, 47, 67, 68, 82].
Abuse liability: The likelihood for future drug abuse triggered by the administration of MDMA
in a controlled setting is assumed to be very low. MDMA has rewarding effects in animals
and humans [74] but the risk for developing dependence is considered low when compared
with other drugs of abuse [101]. In previous studies using MDMA in controlled settings, none
of the participants expressed any interest in taking MDMA as a recreational drug after
completing the study [15, 42]. Subjects with known risk factors for future MDMA use
including depression or anxiety disorders [102] and prior drug dependence or other relevant
illicit drug use [103] will be excluded from the study. Illicit drug use will also be monitored
during the study using repeated urine drug screens.
14.3 Risk-Benefit Assessment
Potential benefits: First, the present study will generate objective, scientific information about
the effects of MDMA that could not be obtained with observational studies. MDMA is used
non-medically by millions of people and scientific information on the pharmacological and
toxicological effects of MDMA is needed. Second, research into the mechanisms of action of
MDMA, which has strong mood-enhancing effects, will provide insight into the physiology of
mood and pathophysiological processes involved in mood disorders. Third, normative data
on the effects of MDMA in normals are useful to interpret results form studies in Ecstasy
users. Forth, pharmacodynamics and pharmacokinetics of MDMA should be better
characterized in the light of ongoing clinical phase II trials using similar doses of MDMA. The
importance of these issues is recognized by researchers and regulatory authorities as
demonstrated by the increase in number of approved controlled studies in humans.
Risk/benefit Ratio: Overall, the risk for severe adverse effects or lasting physical harm is
considered very low. With regard to subjective distress, the overall effect of MDMA is clearly
a pleasurable experience with comparatively minimal risks for dysphoric reactions and
tolerable adverse effects. The potential risk of further abuse of MDMA is expected to be very
low in healthy subjects with low risk of drug abuse liability such as previous significant drug
use or preexisting psychiatric disorder. Taken together, the risks are minimal. Because
healthy volunteers will participate in this phase I study, the benefits mainly consist in
increased information that will not directly benefit the study participants but will be of benefit
for the society and patients with Ecstasy intoxications. However, based on our experience
with similar studies, subject mostly experienced pleasure and considered the use of MDMA
in a controlled setting to be of some interesting and personal value.
14.4 Ethical standards
The study will be performed in accordance with the Declaration of Helsinki, International
Conference on Harmonization, Topic E6 Guidelines for Good Clinical Practice (GCP), the
“Verordnung über klinische Versuche mit Heilmitteln” (VKlin), the “Heilmittelgesetz” (HMG),
and standard operating procedures of the CRC/CTU of the University Hospital of Basel.
14.4.1 Institutional Review Board
The PI will submit the study protocol and informed consent form to an approved IRB
(institutional review board = Ethische Kommission beider Basel, EKBB).
14.4.2 Protocol amendments
Version 14.10.2009
21/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
Any protocol amendments will require formal approval of the ethics committee prior to their
implementation.
14.4.3 Early study termination
The study may be ended early in the case of unexpected adverse events or other reasons
based on the PIs decision and on criteria determined as outlined below.
 An interim analysis will be performed after 8 subjects have completed the study. If
this analysis indicates a significant increase in measures that indicate increased
monoamine release the study will be terminated early. Measures included in the
analysis are: heart rate, blood pressure, the total of adverse effects (list of
complaints), and adverse events.
 In addition, in the case of severe adverse events in a subject indicating excessive
monoamine release, the code will be opened and results analyzed by the principal
investigator. The following findings when due to MDMA and duloxetine
administration will trigger early analysis of results and potential study termination:
o Hypertension >Psys 200 mm Hg or Pdia 140 mm Hg
o Tachykardia 120 beats/min
o Serotonin syndrome (cloni, hyperthermia, delir)
o Other unexpected severe adverse effects that could be linked to excessive
release of monoamines
The PI will inform the IRB and Swissmedic when the study ends within 90 days or 15 days if
the study is terminated early. A final report is sent to both within six months of study
termination by the PI.
14.4.4 Insurance
Subjects will be insured for this trial by Zurich Insurances via the University Hospital. The PI
will be responsible for the initiation of all necessary action in the case of harm to a subject.
14.4.5 Compensation
Subjects will receive a financial compensation of 1’000 CHF for the completed trial. Pro rata
payments will be done for early discontinuation. There are no other compensations.
14.4.6 Premature withdrawal of subjects
Subjects have the right to withdraw from the study at any time. The investigator may
withdraw subjects from the study in the event of adverse events, protocol violations, or other
reasons. An end of study assessment is made at the time of the subject's withdrawal. If the
reason for removal of a subject from the study is an adverse event or an abnormal laboratory
test result, the event or test will also be recorded on the CRF.
14.4.7 Replacement policy
Subjects who withdraw from the study will be replaced if the number exceeds three subjects,
to maintain at least 13 subjects. Replacement subject will follow the same treatment
sequence as the withdrawn subject.
14.5 Legal authorizations
MDMA is a scheduled illegal substance in Switzerland (Anhang d der BetmV-Swissmedic).
We will apply to the BAG for permission to use MDMA in the present study as soon as the
study has been assessed by the IRB and Swissmedic.
14.6 Trial registration
The study will be registered at www.clinicaltrials.gov.
14.7 Study documentation and record keeping
Version 14.10.2009
22/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
The investigator will maintain adequate records to enable the conduct of the study to be fully
documented. Copies of protocols, CRFs, originals of test result reports, drug dispensing
logs, correspondence, records of informed consent and other documents pertaining to the
conduct of the study will be kept on file for 10 years in the CRC archive. Identification codes
will be kept for at least two years after study completion. All forms should be typed or filled
out using a blue or black ball-point pen, and must be legible. Errors should be crossed out
but not obliterated, the correction inserted, and the change initialed and dated by the
investigator or an authorized person. For each subject enrolled a CRF will be completed and
signed by the investigator. This also applies to those subjects who fail to complete the study.
14.8 Quality control and quality assurance
14.8.1 Training of personnel and SOPs
The study is performed in accordance with ICH GCP E6 and the SOPs of the phase 1 unit
and the study coordination center of the clinical trial unit (CTU) Basel. The study personnel
has completed a clinical investigator course of the CTU Basel and additional on-site training.
14.8.2 Monitoring
The trial will be monitored by the CTU Basel to verify the accuracy and completeness of
CRFs, to ensure that all protocol requirements, applicable local authority regulations and
investigator’s obligations are being fulfilled, and to resolve any inconsistencies in the study
records.
14.8.3 Direct access to source data
The investigator will permit study related monitoring visits, audits, ethics committee reviews,
and regulatory inspections, and provide direct access to all source data. Source data are all
information, original records of clinical findings, observations, or other activities in a clinical
trial necessary for the reconstruction and evaluation of the trial.
14.8.4 Inspection
Inspections by regulatory authorities during study or after study closure might be performed
to ensure proper study conduct and data handling procedures according to ICH-GCP
guidelines and regulatory requirements. Inspections may include verification of all source
documents, check of CRFs and site files and a visual inspection of the study site.
14.9 Confidentiality of trial documents and subject records
The investigator will assure that subjects' anonymity will be maintained. On CRFs subjects
should not be identified by their names, but by an identification code. The investigator will
keep a separate log of subjects' codes, names and addresses.
15 Time plan
Start of the study:
Study duration per subject:
Study completion:
Final report:
1.1.2010
7 weeks
31.12.2010
31.3.2011
16 Publications
All results will be presented at a congress meeting and published in a peer-reviewed
scientific journal. The PI will be senior and corresponding author on all publications. PhD
candidates who will substantially contribute to the work will be first authors.
Version 14.10.2009
23/30
Duloxetine-MDMA Study Protocol
Matthias Liechti
17 Budget and Funding
The costs will be covered by a grant of the Swiss National Science Foundation (SNF). The
researchers do not receive any payments for the conduct of this research besides from their
salaries.
18 Certifications of principal investigator
The PI is a MD specialized in internal medicine and clinical pharmacology and toxicology. He
holds a MAS degree in Clinical Research (2005-2007, UCSD, CA, US). He has successfully
been conducting clinical research and similar studies using MDMA in healthy volunteers [1315, 18, 24, 42, 68, 82, 104] and works as senior physician in the Clinical Pharmacology and
Toxicology Unit.
19 Conflict of interest
None
20 Research Environment
20.1 Study site
The study will take place in the fully equipped eight-bed Clinical Research Center of the
University Hospital Basel, Switzerland. This controlled setting allows for close monitoring of
adverse effects and know-how and staff for PK sampling is available.
21 References
[1]
[2]
[3]
[4]
[5]
[6]
[7]
[8]
[9]
[10]
[11]
[12]
[13]
SFA. Schweizerische Fachstelle für Alkohol- und andere Drogenprobleme. 2002.
Sessa B, Nutt DJ. MDMA, politics and medical research: have we thrown the baby out with the
bathwater? J Psychopharmacol 2007;21:787-91.
Johansen P, Krebs T. How could MDMA (ecstasy) help anxiety disorders? A neurobiological
rationale. J Psychopharmacol 2009;23:389-91.
Liechti ME, Kunz I, Kupferschmidt H. Acute medical problems due to Ecstasy use. Case-series
of emergency department visits. Swiss Med Wkly 2005;135:652-7.
Liechti ME. ["Ecstasy" (MDMA): pharmacology, toxicology, and treatment of acute intoxication].
Dtsch Med Wochenschr 2003;128:1361-6.
McCann UD, Szabo Z, Scheffel U, Dannals RF, Ricaurte GA. Positron emission tomographic
evidence of toxic effect of MDMA ("Ecstasy") on brain serotonin neurons in human beings.
Lancet 1998;352:1433-7.
Rothman RB, Baumann MH. Therapeutic and adverse actions of serotonin transporter
substrates. Pharmacol Ther 2002;95:73-88.
Rothman RB, Baumann MH, Dersch CM, Romero DV, Rice KC, Carroll FI, et al.
Amphetamine-type central nervous system stimulants release norepinephrine more potently
than they release dopamine and serotonin. Synapse 2001;39:32-41.
Gudelsky
GA,
Nash
JF.
Carrier-mediated
release
of
serotonin
by 3,4methylenedioxymethamphetamine: implications for serotonin-dopamine interactions. J
Neurochem 1996;66:243-9.
Mechan AO, Esteban B, O'Shea E, Elliott JM, Colado MI, Green AR. The pharmacology of the
acute
hyperthermic
response
that
follows
administration
of
3,4methylenedioxymethamphetamine (MDMA, 'ecstasy') to rats. Br J Pharmacol 2002;135:17080.
Callaway CW, Wing LL, Geyer MA. Serotonin release contributes to the locomotor stimulant
effects of 3,4-methylenedioxymethamphetamine in rats. J Pharmacol Exp Ther 1990;254:45664.
Bengel D, Murphy DL, Andrews AM, Wichems CH, Feltner D, Heils A, et al. Altered brain
serotonin homeostasis and locomotor insensitivity to 3, 4-methylenedioxymethamphetamine
("Ecstasy") in serotonin transporter-deficient mice. Mol Pharmacol 1998;53:649-55.
Liechti ME, Baumann C, Gamma A, Vollenweider FX. Acute psychological effects of 3,4methylenedioxymethamphetamine (MDMA, "Ecstasy") are attenuated by the serotonin uptake
inhibitor citalopram. Neuropsychopharmacology 2000;22:513-21.
Version 14.10.2009
24/30
Duloxetine-MDMA Study Protocol
[14]
[15]
[16]
[17]
[18]
[19]
[20]
[21]
[22]
[23]
[24]
[25]
[26]
[27]
[28]
[29]
[30]
[31]
[32]
[33]
[34]
[35]
Matthias Liechti
Liechti ME, Geyer MA, Hell D, Vollenweider FX. Effects of MDMA (ecstasy) on prepulse
inhibition and habituation of startle in humans after pretreatment with citalopram, haloperidol,
or ketanserin. Neuropsychopharmacology 2001;24:240-52.
Liechti ME, Vollenweider FX. The serotonin uptake inhibitor citalopram reduces acute
cardiovascular and vegetative effects of 3,4-methylenedioxymethamphetamine ('Ecstasy') in
healthy volunteers. J Psychopharmacol 2000;14:269-74.
Tancer M, Johanson CE. The effects of fluoxetine on the subjective and physiological effects
of 3,4-methylenedioxymethamphetamine (MDMA) in humans. Psychopharmacology (Berl)
2007;189:565-73.
Farre M, Abanades S, Roset PN, Peiro AM, Torrens M, O'Mathuna B, et al. Pharmacological
interaction between 3,4-methylenedioxymethamphetamine (ecstasy) and paroxetine:
pharmacological effects and pharmacokinetics. J Pharmacol Exp Ther 2007;323:954-62.
Liechti ME, Vollenweider FX. Which neuroreceptors mediate the subjective effects of MDMA in
humans? A summary of mechanistic studies. Hum Psychopharmacol 2001;16:589-98.
Verrico CD, Miller GM, Madras BK. MDMA (Ecstasy) and human dopamine, norepinephrine,
and serotonin transporters: implications for MDMA-induced neurotoxicity and treatment.
Psychopharmacology (Berl) 2007;189:489-503.
Verrico CD, Lynch L, Fahey MA, Fryer AK, Miller GM, Madras BK. 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.
Sprague JE, Moze P, Caden D, Rusyniak DE, Holmes C, Goldstein DS, et al. Carvedilol
reverses
hyperthermia
and
attenuates
rhabdomyolysis
induced
by
3,4methylenedioxymethamphetamine (MDMA, Ecstasy) in an animal model. Crit Care Med
2005;33:1311-6.
Selken J, Nichols DE. Alpha1-adrenergic receptors mediate the locomotor response to
systemic administration of (+/-)-3,4-methylenedioxymethamphetamine (MDMA) in rats.
Pharmacol Biochem Behav 2007;86:622-30.
Quinn ST, Guiry PJ, Schwab T, Keenan AK, McBean GJ. Blockade of noradrenaline transport
abolishes 4-methylthioamphetamine-induced contraction of the rat aorta in vitro. Auton
Autacoid Pharmacol 2006;26:335-44.
Vollenweider FX, Gamma A, Liechti M, Huber T. Psychological and cardiovascular effects and
short-term sequelae of MDMA ("ecstasy") in MDMA-naive healthy volunteers.
Neuropsychopharmacology 1998;19:241-51.
Trivedi MH, Desaiah D, Ossanna MJ, Pritchett YL, Brannan SK, Detke MJ. Clinical evidence
for serotonin and norepinephrine reuptake inhibition of duloxetine. Int Clin Psychopharmacol
2008;23:161-9.
Karpa KD, Cavanaugh JE, Lakoski JM. Duloxetine pharmacology: profile of a dual monoamine
modulator. CNS Drug Rev 2002;8:361-76.
Wong DT. Duloxetine (LY 248686): an inhibitor of serotonin and noradrenaline uptake and an
antidepressant drug candidate. Expert Opin Investig Drugs 1998;7:1691-9.
Fuller RW, Hemrick-Luecke SK, Snoddy HD. Effects of duloxetine, an antidepressant drug
candidate, on concentrations of monoamines and their metabolites in rats and mice. J
Pharmacol Exp Ther 1994;269:132-6.
Han DD, Gu HH. Comparison of the monoamine transporters from human and mouse in their
sensitivities to psychostimulant drugs. BMC Pharmacol 2006;6:6.
Stoops WW, Blackburn JW, Hudson DA, Hays LR, Rush CR. Safety, tolerability and subjectrated effects of acute intranasal cocaine administration during atomoxetine maintenance. Drug
Alcohol Depend 2008;92:282-5.
Sofuoglu M, Sewell RA. Norepinephrine and stimulant addiction. Addict Biol 2008.
Skinner MH, Kuan HY, Pan A, Sathirakul K, Knadler MP, Gonzales CR, et al. Duloxetine is
both an inhibitor and a substrate of cytochrome P4502D6 in healthy volunteers. Clin
Pharmacol Ther 2003;73:170-7.
O'Mathuna B, Farre M, Rostami-Hodjegan A, Yang J, Cuyas E, Torrens M, et al. The
consequences of 3,4-methylenedioxymethamphetamine induced CYP2D6 inhibition in
humans. J Clin Psychopharmacol 2008;28:523-9.
Lantz RJ, Gillespie TA, Rash TJ, Kuo F, Skinner M, Kuan HY, et al. Metabolism, excretion, and
pharmacokinetics of duloxetine in healthy human subjects. Drug Metab Dispos 2003;31:114250.
Lieberman
JA,
Aghajanian
GK.
Caveat
emptor:
researcher
beware.
Neuropsychopharmacology 1999;21:471-3.
Version 14.10.2009
25/30
Duloxetine-MDMA Study Protocol
[36]
[37]
[38]
[39]
[40]
[41]
[42]
[43]
[44]
[45]
[46]
[47]
[48]
[49]
[50]
[51]
[52]
[53]
[54]
[55]
[56]
[57]
[58]
[59]
Matthias Liechti
Wittchen H-U, Wunderlich U, Gruschwitz S, Zaudig M. SKID-I: Strukturiertes Klinisches
Interview für DSM-IV. Göttingen, D.: Hogrefe-Verlag, 1997.
Dlugos A, Freitag C, Hohoff C, McDonald J, Cook EH, Deckert J, et al. Norepinephrine
transporter gene variation modulates acute response to D-amphetamine. Biol Psychiatry
2007;61:1296-305.
White TL, Lott DC, de Wit H. Personality and the subjective effects of acute amphetamine in
healthy volunteers. Neuropsychopharmacology 2006;31:1064-74.
Patrick CJ, Curtin JJ, Tellegen A. Development and validation of a brief form of the
Multidimensional Personality Questionnaire. Psychol Assess 2002;14:150-63.
Fahrenberg J, Hampel R, Selg H. Das Freiburger Persönlichkeitsinventar (FPI). Göttingen:
Hogrefe, 1984.
White TL, Justice AJ, de Wit H. Differential subjective effects of D-amphetamine by gender,
hormone levels and menstrual cycle phase. Pharmacol Biochem Behav 2002;73:729-41.
Liechti ME, Gamma A, Vollenweider FX. Gender differences in the subjective effects of
MDMA. Psychopharmacology (Berl) 2001;154:161-8.
Dumont G, Kramers C, Sweep F, Touw D, van Hasselt J, de Kam M, et al. Cannabis
Coadministration Potentiates the Effects of "Ecstasy" on Heart Rate and Temperature in
Humans. Clin Pharmacol Ther 2009.
Kolbrich EA, Goodwin RS, Gorelick DA, Hayes RJ, Stein EA, Huestis MA. Plasma
pharmacokinetics of 3,4-methylenedioxymethamphetamine after controlled oral administration
to young adults. Ther Drug Monit 2008;30:320-32.
Farre M, de la Torre R, Mathuna BO, Roset PN, Peiro AM, Torrens M, et al. Repeated doses
administration of MDMA in humans: pharmacological effects and pharmacokinetics.
Psychopharmacology (Berl) 2004;173:364-75.
Cami J, Farre M, Mas M, Roset PN, Poudevida S, Mas A, et al. Human pharmacology of 3,4methylenedioxymethamphetamine ("ecstasy"): psychomotor performance and subjective
effects. J Clin Psychopharmacol 2000;20:455-66.
Hasler F, Studerus E, Lindner K, Ludewig S, Vollenweider F. Investigation of serotonin-1A
receptor function in the human psychopharmacology of MDMA. J Psychopharmacol 2008.
Janke W, Debus G. Die Eigenschaftswörterliste. Göttingen, D.: Hogrefe, 1978.
Spielberger CD, Gorsuch RL, Lusheme RE. STAI, Manual for the Stat-Trait-Anxiety Inventory.
Palo Alto, CA, US: Cunsulting Psychologists Press., 1970.
Martin WR, Sloan JW, Sapira JD, Jasinski DR. Physiologic, subjective, and behavioral effects
of amphetamine, methamphetamine, ephedrine, phenmetrazine, and methylphenidate in man.
Clin Pharmacol Ther 1971;12:245-58.
Bopp G, Bender W, Schütz CG. Validierung der deutschen Version des Addiction Research
Center Inventory (ARCI). Suchtmedizin 2005;7:152-3.
Dittrich A. The standardized psychometric assessment of altered states of consciousness
(ASCs) in humans. Pharmacopsychiatry 1998;31 Suppl 2:80-4.
Gouzoulis-Mayfrank E, Heekeren K, Neukirch A, Stoll M, Stock C, Obradovic M, et al.
Psychological effects of (S)-ketamine and N,N-dimethyltryptamine (DMT): a double-blind,
cross-over study in healthy volunteers. Pharmacopsychiatry 2005;38:301-11.
Zerssen DV. Die Beschwerden-Liste. Münchener Informationssystem. . München, D: Psychis,
1976.
Baron-Cohen S, Wheelwright S, Hill J, Raste Y, Plumb I. The "Reading the Mind in the Eyes"
Test revised version: a study with normal adults, and adults with Asperger syndrome or highfunctioning autism. J Child Psychol Psychiatry 2001;42:241-51.
Schachinger H, Cox D, Linder L, Brody S, Keller U. Cognitive and psychomotor function in
hypoglycemia: response error patterns and retest reliability. Pharmacol Biochem Behav
2003;75:915-20.
Grob CS, Poland RE, Chang L, Ernst T. Psychobiologic effects of 3,4methylenedioxymethamphetamine in humans: methodological considerations and preliminary
observations. Behav Brain Res 1996;73:103-7.
Mas M, Farre M, de la Torre R, Roset PN, Ortuno J, Segura J, et al. Cardiovascular and
neuroendocrine effects and pharmacokinetics of 3, 4-methylenedioxymethamphetamine in
humans. J Pharmacol Exp Ther 1999;290:136-45.
Forsling M, Fallon JK, Kicman AT, Hutt AJ, Cowan DA, Henry JA. Arginine vasopressin
release in response to the administration of 3,4-methylenedioxymethamphetamine ("ecstasy"):
is metabolism a contributory factor? J Pharm Pharmacol 2001;53:1357-63.
Version 14.10.2009
26/30
Duloxetine-MDMA Study Protocol
[60]
[61]
[62]
[63]
[64]
[65]
[66]
[67]
[68]
[69]
[70]
[71]
[72]
[73]
[74]
[75]
[76]
[77]
[78]
[79]
[80]
[81]
Matthias Liechti
Wolff K, Tsapakis EM, Winstock AR, Hartley D, Holt D, Forsling ML, et al. Vasopressin and
oxytocin secretion in response to the consumption of ecstasy in a clubbing population. J
Psychopharmacol 2006;20:400-10.
AK. Arzneimittelkompendium der Schweiz. Basel: Documed AG, http://www.documed.ch,
2009.
Goldstein DJ, Mallinckrodt C, Lu Y, Demitrack MA. Duloxetine in the treatment of major
depressive disorder: a double-blind clinical trial. J Clin Psychiatry 2002;63:225-31.
Choy EH, Mease PJ, Kajdasz DK, Wohlreich MM, Crits-Christoph P, Walker DJ, et al. Safety
and tolerability of duloxetine in the treatment of patients with fibromyalgia: pooled analysis of
data from five clinical trials. Clin Rheumatol 2009.
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;31:1022-36.
Lobo ED, Quinlan T, O'Brien L, Knadler MP, Heathman M. Population pharmacokinetics of
orally administered duloxetine in patients: implications for dosing recommendation. Clin
Pharmacokinet 2009;48:189-97.
Turcotte JE, Debonnel G, de Montigny C, Hebert C, Blier P. Assessment of the serotonin and
norepinephrine reuptake blocking properties of duloxetine in healthy subjects.
Neuropsychopharmacology 2001;24:511-21.
Kolbrich EA, Goodwin RS, Gorelick DA, Hayes RJ, Stein EA, Huestis MA. Physiological and
subjective responses to controlled oral 3,4-methylenedioxymethamphetamine administration. J
Clin Psychopharmacol 2008;28:432-40.
Liechti ME, Saur MR, Gamma A, Hell D, Vollenweider FX. Psychological and physiological
effects of MDMA ("Ecstasy") after pretreatment with the 5-HT(2) antagonist ketanserin in
healthy humans. Neuropsychopharmacology 2000;23:396-404.
Oehen P. Phase II Pilot Randomized Double-Blind Placebo-Controlled Study of 3,4Methylenedioxymethamphetamine (MDMA)Assisted Psychotherapy in Posttraumatic Stress
Disorder
(PTSD)Switzerland.
www.clinicaltrials.gov.
ClinicalTrials.gov
Identifier:
NCT00353938. 2007.
de la Torre R, Farre M, Ortuno J, Mas M, Brenneisen R, Roset PN, et al. Non-linear
pharmacokinetics of MDMA ('ecstasy') in humans. Br J Clin Pharmacol 2000;49:104-9.
de la Torre R, Farre M, Roset PN, Pizarro N, Abanades S, Segura M, et al. Human
pharmacology of MDMA: pharmacokinetics, metabolism, and disposition. Ther Drug Monit
2004;26:137-44.
Yang J, Jamei M, Heydari A, Yeo KR, de la Torre R, Farre M, et al. Implications of
mechanism-based inhibition of CYP2D6 for the pharmacokinetics and toxicity of MDMA. J
Psychopharmacol 2006;20:842-9.
Vollenweider FX, Gamma A, Liechti M, Huber T. Is a single dose of MDMA harmless?
Neuropsychopharmacology 1999;21:598-600.
Tancer M, Johanson CE. Reinforcing, subjective, and physiological effects of MDMA in
humans: a comparison with d-amphetamine and mCPP. Drug Alcohol Depend 2003;72:33-44.
Tancer ME, Johanson CE. The subjective effects of MDMA and mCPP in moderate MDMA
users. Drug Alcohol Depend 2001;65:97-101.
Giroud C, Augsburger M, Sadeghipour F, Varesio E, Veuthey JL, Rivier L. [Ecstasy--the status
in French-speaking Switzerland. Composition of seized drugs, analysis of biological specimens
and short review of its pharmacological action and toxicity]. Praxis (Bern 1994) 1997;86:51023.
Helmlin HJ, Bracher K, Bourquin D, Vonlanthen D, Brenneisen R. Analysis of 3,4methylenedioxymethamphetamine (MDMA) and its metabolites in plasma and urine by HPLCDAD and GC-MS. J Anal Toxicol 1996;20:432-40.
Kolbrich EA, Lowe RH, Huestis MA. Two-dimensional gas chromatography/electron-impact
mass spectrometry with cryofocusing for simultaneous quantification of MDMA, MDA, HMMA,
HMA, and MDEA in human plasma. Clin Chem 2008;54:379-87.
Wenk M, Todesco L, Krahenbuhl S. Effect of St John's wort on the activities of CYP1A2,
CYP3A4, CYP2D6, N-acetyltransferase 2, and xanthine oxidase in healthy males and females.
Br J Clin Pharmacol 2004;57:495-9.
Wenk M, Todesco L, Keller B, Follath F. Determination of dextromethorphan and dextrorphan
in urine by high-performance liquid chromatography after solid-phase extraction. J Pharm
Biomed Anal 1991;9:341-4.
Morgenthaler NG, Struck J, Alonso C, Bergmann A. Assay for the measurement of copeptin, a
stable peptide derived from the precursor of vasopressin. Clin Chem 2006;52:112-9.
Version 14.10.2009
27/30
Duloxetine-MDMA Study Protocol
[82]
[83]
[84]
[85]
[86]
[87]
[88]
[89]
[90]
[91]
[92]
[93]
[94]
[95]
[96]
[97]
[98]
[99]
[100]
[101]
[102]
Matthias Liechti
Liechti ME, Vollenweider FX. Acute psychological and physiological effects of MDMA
("Ecstasy") after haloperidol pretreatment in healthy humans. Eur Neuropsychopharmacol
2000;10:289-95.
Camarasa J, Marimon JM, Rodrigo T, Escubedo E, Pubill D. Memantine prevents the cognitive
impairment induced by 3,4-methylenedioxymethamphetamine in rats. Eur J Pharmacol 2008.
Kuypers KP, Ramaekers JG. Acute dose of MDMA (75 mg) impairs spatial memory for
location but leaves contextual processing of visuospatial information unaffected.
Psychopharmacology (Berl) 2007;189:557-63.
de la Torre R, Farre M, Mathuna BO, Roset PN, Pizarro N, Segura M, et al. MDMA (ecstasy)
pharmacokinetics in a CYP2D6 poor metaboliser and in nine CYP2D6 extensive metabolisers.
Eur J Clin Pharmacol 2005;61:551-4.
Pacifici R, Pichini S, Zuccaro P, Farre M, Segura M, Ortuno J, et al. Paroxetine inhibits acute
effects of 3,4-methylenedioxymethamphetamine on the immune system in humans. J
Pharmacol Exp Ther 2004;309:285-92.
Vollenweider FX, Liechti ME, Gamma A, Greer G, Geyer M. Acute psychological and
neurophysiological effects of MDMA in humans. J Psychoactive Drugs 2002;34:171-84.
Hernandez-Lopez C, Farre M, Roset PN, Menoyo E, Pizarro N, Ortuno J, et al. 3,4Methylenedioxymethamphetamine (ecstasy) and alcohol interactions in humans: psychomotor
performance, subjective effects, and pharmacokinetics. J Pharmacol Exp Ther 2002;300:23644.
Liechti ME, Gamma A, Vollenweider FX. No lasting psycho-physiological effects of a single
dose of MDMA ("Ecstasy") in controlled conditions in healthy human volunteers. Eur
Neuropsychopharmacol 2001;11.
Gamma
A,
Buck
A,
Berthold
T,
Liechti
ME,
Vollenweider
FX.
3,4Methylenedioxymethamphetamine (MDMA) modulates cortical and limbic brain activity as
measured by [H(2)(15)O]-PET in healthy humans. Neuropsychopharmacology 2000;23:38895.
Chang L, Grob CS, Ernst T, Itti L, Mishkin FS, Jose-Melchor R, et al. Effect of ecstasy [3,4methylenedioxymethamphetamine (MDMA)] on cerebral blood flow: a co-registered SPECT
and MRI study. Psychiatry Res 2000;98:15-28.
Sharma A, Goldberg MJ, Cerimele BJ. Pharmacokinetics and safety of duloxetine, a dualserotonin and norepinephrine reuptake inhibitor. J Clin Pharmacol 2000;40:161-7.
Reneman L, Booij J, de Bruin K, Reitsma JB, de Wolff FA, Gunning WB, et al. Effects of dose,
sex, and long-term abstention from use on toxic effects of MDMA (ecstasy) on brain serotonin
neurons. Lancet 2001;358:1864-9.
Buchert R, Thiele F, Thomasius R, Wilke F, Petersen K, Brenner W, et al. Ecstasy-induced
reduction of the availability of the brain serotonin transporter as revealed by [11C](+)McN5652PET and the multi-linear reference tissue model: loss of transporters or artifact of tracer kinetic
modelling? J Psychopharmacol 2007;21:628-34.
de Win MM, Reneman L, Jager G, Vlieger EJ, Olabarriaga SD, Lavini C, et al. A prospective
cohort study on sustained effects of low-dose ecstasy use on the brain in new ecstasy users.
Neuropsychopharmacology 2007;32:458-70.
de Win MM, Schilt T, Reneman L, Vervaeke H, Jager G, Dijkink S, et al. Ecstasy use and selfreported depression, impulsivity, and sensation seeking: a prospective cohort study. J
Psychopharmacol 2006;20:226-35.
Halpern JH, Pope HG, Jr., Sherwood AR, Barry S, Hudson JI, Yurgelun-Todd D. Residual
neuropsychological effects of illicit 3,4-methylenedioxymethamphetamine (MDMA) in
individuals with minimal exposure to other drugs. Drug Alcohol Depend 2004;75:135-47.
Ludewig S, Ludewig K, Hasler F, Vollenweider FX. No lasting effects of moderate doses of
MDMA (Ecstasy) on memory performance and mood states in healthy humans. Biol Psychiatry
2003;53.
Vollenweider FX, Gucker P, Schönbächler R, Kamber E, Vollenweider-Scherpenhuyzen MFI,
Schubiger G, et al. Effects of MDMA on 5-HT uptake sites using PET and [11C]-McN5652 in
humans. . Nervenarzt 2000;71.
Vollenweider FX, Jones RT, Baggott MJ. Caveat emptor: editors beware.
Neuropsychopharmacology 2001;24:461-3.
Nutt D, King LA, Saulsbury W, Blakemore C. Development of a rational scale to assess the
harm of drugs of potential misuse. Lancet 2007;369:1047-53.
Huizink AC, Ferdinand RF, van der Ende J, Verhulst FC. Symptoms of anxiety and depression
in childhood and use of MDMA: prospective, population based study. Bmj 2006;332:825-8.
Version 14.10.2009
28/30
Duloxetine-MDMA Study Protocol
[103]
[104]
Matthias Liechti
Pedersen W, Skrondal A. Ecstasy and new patterns of drug use: a normal population study.
Addiction 1999;94:1695-706.
Vollenweider FX, Liechti ME, Paulus MP. MDMA affects both error-rate dependent and
independent aspects of decision-making in a two-choice prediction task. J Psychopharmacol
2005;19:366-74.
Version 14.10.2009
29/30