shearer et al 2009 addiction

Upload: ahnaf-ahmed

Post on 06-Jul-2018

218 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/17/2019 Shearer Et Al 2009 Addiction

    1/10

    A double-blind, placebo-controlled trial of modafinil

    (200 mg/day) for methamphetamine dependence

     James Shearer 1, Shane Darke1, Craig Rodgers2, Tim Slade1, Ingrid van Beek2, John Lewis3,

    Donna Brady4, Rebecca McKetin1, Richard P. Mattick1 & Alex Wodak4

    National Drug and Alcohol Research Centre, University of New South Wales, Sydney, NSW, Australia,1 Kirketon Road Centre, Sydney Hospital, Sydney, NSW,

    Australia,2 Pacific Laboratory Medicine Ser vices, Sydney, NSW, Australia3 and St Vincent’s Hospital, Sydney, NSW, Australia4

    ABSTRACT

    Aim   To examine the safety and efficacy of modafinil (200 mg/day) compared to placebo in the treatment of meth-

    amphetamine dependence and to examine predictors of post-treatment outcome.  Participants and design   Eighty

    methamphetamine-dependent subjects in Sydney, Australia were allocated randomly to modafinil (200 mg/day)

    (n =  38) or placebo (n =  42) under double-blind conditions for 10 weeks with a further 12 weeks post- treatment

    follow-up.  Measures   Comprehensive drug use data (urine specimens and self-report) and other health and psycho-

    social data were collected weekly during treatment and research interviews at baseline, week 10 and week 22.

    Results   Treatment retention and medication adherence were equivalent between groups. There were no differences

    in methamphetamine abstinence, craving or severity of dependence. Medication-compliant subjects tended to provide

    more methamphetamine-negative urine samples over the 10-week treatment period (P =  0.07). Outcomes were better

    for methamphetamine-dependent subjects with no other substance dependence and those who accessed counselling.

    There were statistically significant reductions in systolic blood pressure (P =  0.03) and weight gain (P =  0.05) in

    modafinil-compliant subjects compared to placebo. There were no medication-related serious adverse events. Adverse

    events were generally mild and consistent with known pharmacological effects. Conclusions   Modafinil demonstrated

    promise in reducing methamphetamine use in selected methamphetamine-dependent patients. The study findings

    support definitive trials of modafinil in larger multi-site trials.

    Keywords   Abuse liability, counselling, methamphetamine dependence, modafinil, randomised controlled trial,safety, treatment.

    Correspondence to:   Shane Darke, National Drug and Alcohol Research Centre, University of New South Wales, Sydney, NSW 2052, Australia.

    E-mail: [email protected]

    Submitted 24 May 2008; initial review completed 22 August 2008; final version accepted 15 October 2008

    INTRODUCTION

    Methamphetamine dependence is a major public healthissue in many parts of the world [1–3] associated with a

    wide range of psychological, medical and social problems

    [4]. Long-term use of high-dose methamphetamine dys-

    regulates monoamine-based neurotransmission, which

    is hypothesized to form a neurobiological basis of meth-

    amphetamine dependence and withdrawal [5–8]. Dys-

    regulation of reward-related glutamate pathways, as

    observed in cocaine use, represents a further potential

    neurobiological substrate for methamphetamine depen-

    dence [7,9,10]. Despite increasing knowledge of the neu-

    robiological consequences of methamphetamine use, no

    medications to date have been any more successful than

    placebo in reducing methamphetamine use in dependent

    patients [11,12].Modafinil is a novel non-amphetamine-type stimulant

    approved in Australia for thetreatmentof narcolepsy, shift

    workers syndrome and sleep apnoea-related fatigue.

    Modafinil acts specifically to promote wakefulness and

    vigilance, although its precise pharmacological mecha-

    nism of action is yet to be delineated [13–16]. The

    dopamine- and glutamate-enhancing actions of modafi-

    nil have been postulated as beneficial in reducing

    withdrawal severity attributable to psychostimulant-

    induced neuroadaption [17,18].The stimulant properties

    of modafinil may also attenuate the disturbed sleep

    RESEARC H REPO RT   doi:10.1111/j.1360-0443.2008.02437.x

    © 2009 The Authors. Journal compilation © 2009 Society for the Study of Addiction   Addiction, 104, 224–233

    mailto:[email protected]:[email protected]

  • 8/17/2019 Shearer Et Al 2009 Addiction

    2/10

    patterns, poor concentration and low energy levels char-

    acteristic of methamphetamine withdrawal.

    Modafinil is a well-tolerated drug with a low overdose

    risk and few side effects [19]. Modafinil does not appear to

    be behaviourally reinforcing, as use does not produce

    euphoria and there is no discontinuation effect [20,21].

    The effects of modafinil cannot be intensified through

    high-dose intravenous or intrapulmonary use due to

    insolubility in water and instability at high temperatures

    [22]. The safety of modafinil in cocaine users has been

    investigated in double-blind placebo-controlled interac-

    tion studies. Combined cocaine and modafinil (0 mg,

    400 mg, 800 mg daily) produced no haemodynamic

    interactions and significantly reduced subjective drug

    effect ratings for cocaine [23,24].

    Cocaine-dependent patients who received modafinil

    (400 mg) as a single morning dose provided significantly

    more cocaine-negative urine samples compared to place-

    bos during an 8-week trial [25]. There were, however, nosignificant differences on self-reported outcomes, includ-

    ing frequency of cocaine use, spending on cocaine or the

    severity of cocaine withdrawal and craving. No serious

    adverse events or overuse of modafinil were reported. The

    safety and tolerability of modafinil (400 mg/day) during

    a 10-day in-patient methamphetamine withdrawal pro-

    gramme was examinedin an open-label comparison with

    mirtazapine (60 mg/day) and pericyazine (2.5–10 mg/

    day) [21]. Modafinil and mirtazapine were found to be

    safe and well tolerated, with no discernable reinforcing

    effects. Both were more effective than pericyazine in

    reducing withdrawal symptoms, with modafinil achiev-ing less severe withdrawal symptoms and less sleep

    disturbance than mirtazapine. Randomized controlled

    placebo trials of modafinil in methamphetamine users

    were recommended.

    The aim of the present study was to examine the

    safety and efficacy of modafinil (200 mg/day) in

    reducing methamphetamine use and associated harms

    through a randomized, double-blind, placebo-controlled

    10-week trial of modafinil (200 mg/day) in 80

    methamphetamine-dependent subjects. Relapse and pre-

    dictors of methamphetamine use other than group (base-

    line characteristics, severity of dependence, treatmentexposure) were also examined 12 weeks post-treatment.

    METHODS

    Subjects

    Eighty methamphetamine-dependent subjects were

    recruited through a primary health care centre (n =  72)

    anda specialist drug andalcohol service(n =  8) located in

    the innercity area of Sydney. Subjects met Diagnosticand

    Statistical Manual (DSM-IV) criteria for methamphet-

    amine dependence operationalized using the Composite

    International Diagnostic Interview [26] and were

    current, regular (2–3 days of use perweek or more) meth-

    amphetamine users confirmed by a methamphetamine-

    positive urine spot test at baseline interview. Individuals

    who had an active, serious and uncontrolled medical or

    psychiatric illness were excluded; however, those with

    comorbiddrug dependence or mental illness were enrolled

    after consultation with their usual medical attendants.

    Women of child-bearing age were excluded if they were

    pregnant, nursing infants or unwilling to avoid preg-

    nancy through abstinence or barrier contraception. The

    latter requirement was necessary, as modafinil induces

    metabolism of steroidal contraceptives. Thirty-seven

    methamphetamine users who presented for treatment

    were excluded due to incomplete enrolment (n =  10),

    ineligibility (mainly infrequent use/long-term abstinence

    n = 13) or were eligible but declined either randomization

    or medication (n =  14). Seven sets of partners wererecruited into the trial and randomized together (modafi-

    nil n =  4, placebo n =  3);however, datawereincludedfrom

    onlythe presentingindexcase, as theirpartners would not

    constitute independent observations. Subjects provided

    written informed consent to participate in the trial

    approved by three institutional human research ethics

    committees, including that of the University of New

    South Wales. Subjects were reimbursed $10 for each

    research interview and urine specimen provided during

    the trial.

    Design

    Eighty eligible methamphetamine-dependent subjects

    seeking treatment were randomized equally to receive

    either modafinil (200 mg/day) (n =  38) or placebo

    (n =  42) under equivalent conditions for a period of 10

    weeks. The sample size of 40 per group was calculated

    based on an estimated between-group difference of 40%

    in the proportion of stimulant-positive urine samples at

    week 10, with missing samples deemed positive and

    allowing a 25% attrition rate (a =  0.05, 1-b =  0.9). A

    fixed-dose regimen was selected because modafinil has

    dose-independent pharmacokinetics between 200 and600 mg/day [19], and early research in cocaine users

    found no difference in cocaine suppression or reductions

    in subjective effects between 200-mg and 400-mg doses.

    Randomization was conducted independently through

    Sydney Hospital Pharmacy. Trial medication was dis-

    pensed on a weekly basis through Sydney Hospital Phar-

    macy using medication event monitoring system (MEMS)

    cap bottles to record unsupervised regimen adherence.

    All subjects were offered a brief four-session cognitive

    behavioural intervention developed specifically for meth-

    amphetamine users [27]. Data were collected on alldrug-

    Modafinil for methamphetamine dependence   225

    © 2009 The Authors. Journal compilation © 2009 Society for the Study of Addiction   Addiction, 104, 224–233

  • 8/17/2019 Shearer Et Al 2009 Addiction

    3/10

    related counselling received by subjects whether within

    or outside the trial during the 10-week treatment phase.

    Subjects attended weekly medical reviews for side effects,

    adverse events, craving, vital observations, renewal of 

    weekly scripts and to provide study urine samples. Sub-

     jects were re-interviewed 12 weeks after they completed

    treatment about their post-treatment progress and drug

    use. There were no between-group differences in study

    follow-up at week 10 (modafinil 84%; placebo 88%) and

    week 22 (modafinil 68%; placebo 62%) (see Fig. 1).

    Measures

    The Opiate Treatment Index (OTI) [28,29] was used tomeasure self-reported methamphetamine use, other drug

    use and other treatment outcomes. A ‘cost of habit’

    section was added to capture changes in average expen-

    diture on methamphetamine. Additional self-reported

    methamphetamine use was collected using 28-day drug

    use diaries. Prospective drug use diaries are acknowl-

    edged as an accurate and valid method of collecting drug

    use data [30]. Psychopathology was assessed using the

    Brief Symptom Inventory (BSI) [31]. A positive BSI case

    was defined as a score above the 90th percentile for US

    non-patient norms on the Global Severity Index or in two

    or more of the psychiatric domains. Changes in the sever-

    ity of dependence were measured using the Severity of 

    Dependence Scale (SDS) [32]. Subjects rated their stron-

    gest methamphetamine craving in the past week on a

    100-mm visual analogue scale (VAS) from ‘none’ to

    ‘strongest ever’. Urine specimens were collected weekly,

    whilesubjectsremainedin treatmentand also at research

    follow-up interviews. Urine was collected in jars with

    heat-sensitive strips to confirm that the samples were at

    body temperature and not substituted or otherwise con-

    taminated. The specimens were analysed by immunoas-

    say for amphetamine and cocaine metabolites using a

    cut-off of 300 mg/l. High-performance thin-layer chro-

    matography and mass spectrometry were used for

    unequivocal confirmation of methamphetamine [33]

    indicative of recent use (within the past 48–72 hours). In

    order to monitor further adherence with modafinil, all

    urine samples were tested for the presence of modafinilic

    acid (urinary metabolite of modafinil) using mass

    spectrometry.

    Data analysis

    Data were analysed using SPSS for Windows (version

    15.0) and SAS (version 9.1) PROC GENMOD. An

    Baseline

    MODAFINIL

    (n=38)

    All commenced 

    treatment

    Week 10

    Completed (n=11)

    Followed Up

    (n=32)

    Week 22

    Followed Up (n=26)

    Analysed (n=38)

    Week 10

    Completed (n=15)

    Followed Up

    (n=37)

    Week 22

    Followed Up (n=26)

    Analysed (n=42)

    Lost (n=16)

     No reason n=8

    Moved n=3

    Jail n=2

    Adverse event n=3

    Baseline

    PLACEBO

    (n=42)

    All commenced 

    treatment

    Lost (n=12)

     No reason n=7

    Moved n=3

    Jail n=1

    Adverse event n=1

    Assessed 

    (n=124)

    Randomised 

    (n=80)

    Excluded (n=44)

    Incomplete n=10

    Ineligible n=13

    Declined n=14

    Partnern

    =7

    Figure 1   Study flow chart

    226   James Shearer  et al.

    © 2009 The Authors. Journal compilation © 2009 Society for the Study of Addiction   Addiction, 104, 224–233

  • 8/17/2019 Shearer Et Al 2009 Addiction

    4/10

    intention-to-treat approach was followed. Data were col-

    lected at research interviews conducted at baseline,

    week 10 and week 22, regardless of treatment status.

    Data for subjects lost to study follow-up were imputed

    from baseline using a worst-case scenario assumption of 

    no change from baseline. Baseline scores and group

    variables were entered through the analysis of covari-

    ance (ANCOVA) approach as predictors in the relevant

    analyses [34]. All statistical tests were two-tailed using

    a 0.05 significance level and 95% confidence (CI)intervals;   t-tests were used for normally distributed or

    transformed continuous variables, the   c2 statistic for

    categorical variables and the Mann–Whitney  U -statistic

    for skewed data. Survival analysis based on the log-rank

    (Mantel–Cox) statistic was used to examine between-

    group differences in retention. Outcome predictors other

    than group (comorbidity, other drug dependence, coun-

    selling uptake) were examined by linear regression. Cat-

    egorical and continuous repeated measures, including

    vital signs, craving and urine drug screens collected at

    weekly intervals while subjects remained in treatment,

    were examined using generalized estimating equations[35,36]. After examination of the within-subject corre-

    lation structures, an autoregressive working matrix

    (which assumes that pairs of measures are correlated

    more highly than measures that are further apart) was

    adopted. Missing samples and observations due to varied

    scheduling, lost, de-identified or testing failures were

    deemed missing completely at random and results were

    imputed on a last observation carried forward basis.

    Missing data due to treatment dropout were not imputed

    in order not to obscure medication effects, whether posi-

    tive or negative [37].

    RESULTS

    Baseline characteristics

    Thetypical subject was male, aged mid-30s, andwelledu-

    cated (Table 1). Most subjects injected methamphet-

    amine on a daily basis, with a third who preferred to

    smoke crystalline methamphetamine. There tended to be

    more human immunodeficiency virus (HIV)-positive

    cases in the modafinil group (all in gay men) and more

    subjects in the placebo group were employed at baseline.

    Retention

    Eleven modafinil-group subjects (29%) and 15 placebo-

    group subjects (36%) completed the 10-week course

    of treatment. There was no difference in retention over

    the 10-week medication phase [log-rank (Mantel–Cox)

    c2 = 0.69,   P =  0.41] (Fig. 2). Subjects were asked their

    reasons for ceasing medication. Similar proportions

    between groups reported medication dropout due to no

    perceived effect/benefit (modafinil 33%, nine of 27;

    placebo 30%, eight of 27), unacceptable side effects

    (modafinil 11%, three of 27; placebo 7%, two of 27) or

    moving, entering drug rehabilitation or jail (modafinil

    11%, three of 27; placebo 33%, seven of 27). Modafinil

    subjects tended to report ceasing medication early due to

    achieving abstinence (22%, six of 27) compared to

    placebo (7%, two of 27) (c2 = 2.7, P =  0.1).

    Adherence

    Subjects who received modafinil remained on medication

    for a mean of 40 days (26 days) and were medication-

    adherent on a mean of 31 days (21 days) as confirmed

    Table 1  Baseline characteristics.

    Modafinil (n =  38) Placebo (n =  42)

    Mean age (years) 35.8 6.9

    range 22–50

    36.1 9.1

    range 20–58

    t = -1.85, P  =  0.85

    Gender (male) 63% 62%   c2 = 0.13,   P =  0.91

    Education (completed year 12) 68% 62%   c2 = 0.372,   P =  0.54

    Employed 43% 61%   c2 = 2.243,   P =  0.17

    Years of regular methamphetamine use 6.9 6.3 7.2 9.7   U  =  693.5,   P =  0.310

    Injecting drug use 68% 57%   c2 = 1.083,   P =  0.30

    Current opioid treatment 13% 12%   c2 = 0.029,   P =  0.87

    Alcohol  >6 standard drinks daily 3% 10%   c2 = 1.617,   P =  0.36

    HIV-positive (self-report) 38% 18%   c2 = 3.506,   P =  0.08

    HCV-positive (self-report) 38% 26%   c2 = 1.144,   P =  0.29

    Daily or almost daily use 55% 69%   c2 = 1.617,   P =  0.20

    Mean days used (past 28) 18.4 6.8 20.5 6.5   t =  1.371,   P =  0.17

    Daily cost of methamphetaminea $95 $79 $107 $90   t = -0.694,   P =  0.49

    HCV = hepatitis C virus; HIV = human immunodeficiency virus.  aAnalysis performed after log-transformation of skewed data.

    Modafinil for methamphetamine dependence   227

    © 2009 The Authors. Journal compilation © 2009 Society for the Study of Addiction   Addiction, 104, 224–233

  • 8/17/2019 Shearer Et Al 2009 Addiction

    5/10

    by MEMS caps and pharmacy records. This represented

    78% adherence to daily medication, almost identical to

    placebo adherenceof 77%(mean34 21 days adherent

    divided by mean 44 21 days retained). Thus, on an

    intention-to-treat basis, adherence was 44% in the

    modafinil group and 49% in the placebo group. The

    majority of unsupervised doses were taken before 3 p.m.

    in both groups (modafinil 79%; placebo 84%). Modafinil

    (as modafinilic acid) was detected in the urine of all

    modafinil-treated subjects and was not found in any of 

    the placebo group during the medication phase. Medica-tion overuse was measured through the dispensing of 9

    days of drug supply for each 7-day period in MEMS con-

    tainers and tablet returns for both groups. There was no

    difference in the proportion of subjects who over-used

    their takeaway medication [modafinil 47%; placebo 51%

    (P =  0.8)] or in the average number of additional tablets

    consumed [modafinil 13 10, range 2–32; placebo

    22 22, range 2–74 (P =  0.5)]. There was no difference

    between either the mean total number of internal

    counselling sessions attended [modafinil 0.6 1.6;

    placebo 0.5 0.9 (P =  0.7)], external counselling

    sessions attended [modafinil 1.1 2.4; placebo 1.4 3.3 (P =  0.6)] or the proportions who did not attend any

    counselling [modafinil 53%; placebo 47% (P =  0.6)].

    Stimulant use

    Urinalysis results

    The chart in Fig. 3 illustrates the proportion of urine

    samples positive for illicit psychostimulants [cocaine,

    methamphetamine or 3,4-methylenedioxymethamphe-

    tamine (MDMA)] over the 10-week medication period.

    There was a trend (P  

  • 8/17/2019 Shearer Et Al 2009 Addiction

    6/10

    most measures and follow-up points (Table 2). Modafinil

    subjects provided more stimulant negative weekly urine

    samples in the 4 weeks matching the 28-day self-reportat week 10 (modafinil 1.4 1.4; placebo 0.7 0.9,

    P =  0.16). When analyses were adjusted post hoc for HIV-

    positive subjects who were over-represented in the

    modafinil group, week 10 estimates improved in the

    direction of treatment [B =  3.59 (95% CI:  -0.45, 7.64),

    P =  0.08] but did not alter appreciably at week 22. When

    the analysis was limited   post hoc   to single diagnosis,

    methamphetamine-dependent subjects with no other

    drug dependence (i.e. opioid maintenance patients

    excluded) self-reported days of stimulant use at week 10

    were 8.3 8.2 (modafinil) and 13.5 10.2 (placebo)

    [B =  4.22 (95% CI: 0.08, 8.35), P <

     0.05] and at week 22were 11.6 9.2 (modafinil) and 16.4 10.4 (placebo)

    [B =  3.82 (95% CI:  -0.60, 8.25), P =  0.09].

    Study power

    A sample size of 58 per group was calculated retros-

    pectively as necessary to detect reliably the observed

    between-group difference of 20% using a repeated-

    measures design without imputation for missing data

    (a =  0.05, 1-b =  0.9, T =  10, r = 0.5) [38]. Alternatively,

    a sample size of 136 per group would be necessary to

    detect the between-group difference in frequency of 

    stimulantuse at week 10 with missing data imputed from

    baseline (a =  0.05, 1-b =  0.8, D = 3.15/9.2 =  0.34). The

    effect size estimate (D) was calculated using the adjusted

    between group difference in 28-day stimulantuse at week

    10 divided by the pooled standard deviation (see Table 2).

    The difference in samplesize estimates reflects the greater

    statistical power of repeated-measures designs.

    Tolerability

    There were no medication-related serious adverse events

    during the study. The most commonly reported adverse

    events are reported in Table 3. Insomnia was the most

    common complaint, which did not differ between groups.

    Headache was the second most common complaint, anddid not differ between groups except when severity was

    taken into account. The modafinil group reported more

    transient mild/moderate headache consistent with

    product information. Mood problems were unique to the

    modafinil group, although all cases were due to an exac-

    erbation of pre-existing symptoms. One subject ceased

    modafinil due to panicattacks and elevated anxiety, while

    another managed anxiety symptoms through dose

    reduction to 100 mg/day. There was a statistically signifi-

    cant reduction in systolic blood pressure over time in

    the modafinil group compared to placebo (c2 = 19.55,

    P =  0.03). Differences in weight gain/loss between thegroups were due mainly to acute weight loss (>4 kg)

    observed in four subjects in the placebo group.

    Other self-report outcomes

    There were no apparent between group differences on a

    range of secondary outcomes. There was no difference in

    weekly craving during the medication phase (F =  1.839,

    P =  0.19) or the SDS score at either treatment follow-up

    (modafinil 7.3 3.4; placebo 7.3 3.4) or post-treat-

    ment follow-up (modafinil 8.1 3.4; placebo 7.7

    4.3). Both groups experienced a substantial regression tothe mean in terms of the overall proportion of BSI cases

    (modafinil 95% at baseline to 66% at week 22; placebo

    90% at baseline to 60% at week 22). Sixty per cent of 

    modafinil subjects guessed correctly when asked at week

    22 if they had received active medication compared to

    55% of placebo subjects (c2 = 0.271, P =  0.6).

    Predictors of post-treatment outcome other than group

    The relationship between potential predictors of post-

    treatment outcome (other than group) and self-reported

    Table 2  Self-reported stimulant use.

    Measure Period Modafinil (n =  38) Placebo (n =  42) Coefficient B (95% CI) P-value

    Mean 28-day dr ug use Baseline 18.4 6.8 20.5 6.5

    Week 10a 8.8 8.3 13.2 10.1 3.15 (-0.71, 7.01) 0.11

    Week 22a 11.8 9.0 15.9 10.7 2.88 (-1.22, 6.99) 0.17

    Change score from baseline Week 10   -9.6 8.4   -7.3 9.4   U  =  650, 0.15

    Week 22   -6.7 8.8   -4.5 9.9   U  =  699, 0.20

    Mean quantity used (OTI) Baseline 2.3 2.0 2.7 2.3

    Week 10a,b 0.6 1.0 1.5 2.2 0.78 (0.03, 1.53) 0.05

    Week 22a,b 1.2 1.3 1.8 2.0 0.54 (-0.19, 1.27) 0.20

    Mean spending ($/day) Baseline $95 79 $107 90

    Week 10a,b $41 70 $64 86 17.5 (-17, 52) 0.24

    Week22a,b $57 82 $75 95 13 (-12, 39) 0.23

    CI = confidence interval; OTI = Opiate TreatmentIndex. aBaseline score included in analysisof covariance; banalysis performed after log-transformation.

    Modafinil for methamphetamine dependence   229

    © 2009 The Authors. Journal compilation © 2009 Society for the Study of Addiction   Addiction, 104, 224–233

  • 8/17/2019 Shearer Et Al 2009 Addiction

    7/10

    28-day stimulant use were examined by linear regression

    (Table 4). The most pronounced effect was from attend-

    ing counselling. Each session of counselling attended

    during the 10-week treatment period reduced 28-day

    stimulant self-report by 1 day (95% CI:  -1.7,  -0.3) at

    post-treatment follow-up. Simply attending any form of 

    counselling reduced 28-day self-report by6 days (95% CI:

    -10.8,   -1.8). HIV-positive subjects who were over-

    represented in the modafinil group had poorer metham-

    phetamine use outcomes. BSI cases also had poorer

    methamphetamine use outcomes; however, there werevery few non-cases and these were divided equally

    between the groups [modafinil two subjects (5%); placebo

    four subjects (10%)]. Opioid maintenance therapy

    (methadone or buprenorphine) had no significant direct

    effect on outcome and the small number of opioid main-

    tenance patients were distributed equally between the

    groups (five in each group). Overall, however, the lack of 

    difference at week 22 between opioid-dependent patients

    in the modafinil and placebo groups appeared to mask

    greater between-group effect size for single-diagnosis

    methamphetamine users (see Self-reported stimulant

    use). Other baseline and treatment characteristics did not

    appear to influence post-treatment outcome.

    DISCUSSION

    In this treatment population modafinil showed

    promise in reducing methamphetamine use in selected

    methamphetamine-dependent patients. However, there

    were no differences between modafinil and placebo in

    retention, methamphetamine abstinence, methamphet-

    amine craving or severity of dependence. Subjects whoremained on medication tended to provide more illicit

    psychostimulant negative urine samples than those who

    received placebo over the 10-week treatment period. This

    was consistent with greater declines in the modafinil

    group in self-reported 28-day stimulant use and the

    quantity of stimulants used. The between-group differ-

    ences in favour of modafinil achieved at week 10 per-

    sisted at week 22, consistent with a group-related

    treatment effect. Emergent treatment effects may have

    been masked by over-representation of HIV-positive sub-

     jects in the modafinil group, who had poorer metham-

    Table 3  Adverse events and vital signs in medication-compliant subjects.

    Event Severity/frequency Modafinil Placebo (P)

    Insomnia All reports 12/38 (32%) 17/42 (40%)   c2 = 0.683,   P =  0.41

    Headache Mild/moderate 7/38 (18%) 2/42 (5%)   c2 = 3.728,   P  <  0.05

    Severe 2/38 (5%) 4/42 (10%)   c2 = 0.522,   P =  0.47

    Nausea Mild/moderate 2/38 (5%) 5/42 (12%)   c2 = 1.102,   P =  0.29

    Severe 3/38 (11%) 1/42 (2%)   c2 = 1.277   P =  0.26

    Gastric pain/discomfort All reports 5/38 (13%) 3/42 (7%)   c2 = 0.802,   P =  0.37

    Thought disorder All reports 2/38 (5%) – Nc

    Anxiety Severe 3/38 (8%) – Nc

    Fatigue All reports – 4/42 (10%) Nc

    High blood pressurea Baseline 9/38 (24%) 10/42 (24%)   c2 = 0.234,   P =  0.63

    High blood pressurea Final observation 5/34 (15%) 5/39 (13%)   c2 = 0.054,   P =  0.82

    Weight loss Between baseline and 10 weeks 2/11 (18%) 8/15 (53%)   c2 = 3.331,   P =  0.07

    Weight change (mean kg) Between baseline and 10 weeks 2.7 2.8 0.2 3.4   U  =  66,   P  <  0.05

    Nc = Not calculated.  aHigh blood pressure was defined as either a systolic reading of 155 mmHg or higher, or a diastolic reading of 90 mmHg or higher.

    Table 4  Predictors of stimulant use 12 weeks post-treatment.

    Variable Rationale Coefficient B (95% CI) P-value

    No. of counselling sessions Treatment exposure   -0.99 (-1.73, -0.26)  

  • 8/17/2019 Shearer Et Al 2009 Addiction

    8/10

    phetamine outcomes, and the enrolment of subjects in

    concurrent opioid maintenance treatment. Methamphe-

    tamine-dependent subjects with no other substance

    dependence who received modafinil reported the greatest

    reductions in methamphetamine use compared to

    placebo.

    Modafinil appeared to be safe and well tolerated. There

    were no medication-related serious adverse events. The

    modafinil group reported more transient mild/moderate

    headaches and also tended to report more nausea/gastric

    discomfort, all of which resolved after the first week of 

    treatment. Reports of thought disorder and anxiety were

    unique to the modafinil group and indicate caution in

    patients with pre-existing psychotic symptoms or anxiety.

    Modafinil did not appear to be associated with any eleva-

    tion of blood pressure or heart rate. The clinical signifi-

    cance of a reduction in systolic blood pressure over the

    10-week treatment period in medication-adherent sub-

     jects receiving modafinil should not be underestimatedgiven the cardiotoxicity of methamphetamine [4]. There

    was no evidence of excessive use of modafinil, consistent

    with previous reports of low abuse liability [20]. Signifi-

    cant weight gain in the modafinil group could be inter-

    preted as a positive treatment outcome, as modafinil

    usually decreases food intake [39]. The measurement of 

    body mass index (BMI) should be considered in future

    studies. Finally, reports of fatigue were unique to the

    placebo group and were consistent with the wake-

    promoting properties of modafinil.

    More than one-third of subjects who received modafi-

    nil experienced no discernable effects on sleep or atten-tion. The expectation of a rapid, strong stimulating effect

    may have contributed to disappointment and early treat-

    ment dropout for those who believed they were receiving

    placebo. Another potential factor has been identified in

    sleep studies, where some patients with previous amphet-

    amine treatment or illicit methamphetamine histories

    appeared to be less sensitive to the stimulating effects of 

    modafinil [40–42]. Motivational enhancement therapy

    may improve adherence and treatment retention in

    future studies, particularly in those where the effect of 

    modafinil was weak or diminishing. The relatively low

    dose used in this study may not have been optimal.Higher doses in the usual range (up to 200 mg b.i.d.)

    could improve adherence and treatment retention.

    Attendance at any form of counselling although

    self-selected, was significantly predictive of better post-

    treatment outcome. Although uptake of the specific

    cognitive–behavioural treatment (CBT) programme

    offered to subjects was minimal, many had pre-existing

    counselling arrangements and study participants

    accessed a diverse array of counselling services during

    the course of the trial. The psychosocial platforms on

    which medications targeted toward psychostimulant use

    are delivered are critical to their success, and these may

    vary according to the needs of target populations [43].

    Potential strategies to improve the uptake of counselling

    could include contingent incentives to attend counsel-

    ling, such as travel reimbursement. Narrative therapy

    has also shown recent promise in methamphetamine

    users [44].

    There was no group difference in methamphetamine

    craving. These data suggest that modafinil may not

    achieve its effects as an anti-craving agent. This finding

    wasconsistent with the findings of Dackis and colleagues,

    who found no reduction in craving in cocaine users mea-

    sured using a similar visual analogue scale [25]. Lack of 

    effect on craving may simply be evidence for lack of effect

    on methamphetamine use; however, the analyses that

    found downward trends in weekly methamphetamine-

    negative urine samples and self-reported frequency of 

    methamphetamine were not matched by any changes in

    self-reported craving. It may be more likely that modafinilworks as a drug effect agonist and improves cognitive

    functioning, rather than as an anti-craving agent. Emerg-

    ing research is drawing attention to the importance of 

    cognitive impairment, impulsivity and stress as critical

    features of problematic psychostimulant use [45,46].

    Chronic psychostimulant use reduces activity in areas

    of the brain associated with cognitive functioning,

    regions where modafinil appears to achieve its wake and

    vigilance-promoting effects. Through restoring cognitive

    functioning, modafinil may help patients to deal with the

    life stressors, cravings, cues and compulsive/obsessive

    drug-seeking behaviour which are common features of relapse among psychostimulant users.

    The main limitations affecting interpretation of the

    results of this study were the absence of an objective

    quantitative measure, reliance on self-reported outcomes

    and a sample size which was too small to detect reliably

    the small differences between modafinil and placebo.

    Between-group differences based on self-report were not

    supported by urinalysis of specimens collected at the

    week 10 and week 22 research follow-up interviews.

    While prospective self-report is considered valid and reli-

    able in drug and alcohol research, there may be a risk of 

    differential reporting between groups. The substantialplacebo effect of 45% and the high proportion of modafi-

    nilsubjects with no discernable medication effect of 40%,

    however, mitigated against potential systematic bias in

    self-report in favour of modafinil. The weekly urine drug

    screens captured changes in drug use over the 10-week

    treatment period time, albeit in compliant subjects.

    Increased dose and motivational enhancement therapy

    informed by the experience of this trial could improve

    medication adherence and avoid the treatment disap-

    pointment that may have contributed to selective treat-

    ment attrition. Alternative biological measures more

    Modafinil for methamphetamine dependence   231

    © 2009 The Authors. Journal compilation © 2009 Society for the Study of Addiction   Addiction, 104, 224–233

  • 8/17/2019 Shearer Et Al 2009 Addiction

    9/10

    sensitive or appropriate to longer-term quantitative

    changes could be considered, such as hair analysis. Given

    the predominance of behavioural and psychosocial

    factors in methamphetamine dependence, it may be

    unrealistic to expect medication alone to achieve large

    treatment effect sizes.

    Modafinil appeared to be safe, non-reinforcing and

    moderately beneficial in reducing methamphetamine use

    in this treatment population. These findings need to be

    confirmed by larger multi-centre trials. Benefits were

    selective for medication-adherent patients who engaged

    in any form of psychosocial therapy or support. Future

    studies could benefit from the experience in this trial by

    encouraging completion of the full course of medication

    and engagement in psychosocial therapies. Future trials

    could also examine thevalueof longer maintenance regi-

    mens or titration to the higher standard dose of 200 mg

    b.i.d.

    Clinical trial registration

    NCT00123370 Clinicaltrials.gov.

    Declarations of interest

    None.

    Acknowledgements

    We thank the staff and clients of the Kirketon Road

    Centre, Rankin Court and Sydney Hospital Pharmacy for

    their support. Funding was provided by the Australian

    Government Department of Health and Ageing. Match-

    ing active and placebo trial medication was supplied as a

    grant by Cephalon Inc.

    References

    1. McKetin R., Kozel N., Douglas J., Ali R., Vicknasingam B.,

    Lund J.  et al.  The rise of methamphetamine in Southeast

    and East Asia. Drug Alcohol Rev 2008; 27: 220.

    2. Maxwell J., Rutowski B. The prevalence of methamphet-

    amine and amphetamine abuse in North America: a review

    of the indicators. Drug Alcohol Rev 2008; 27: 229.

    3. Degenhardt L., Roxburgh A., Black E., Bruno R., Campbell

    G., Kinner S.  et al. The epidemiology of methamphetamineuse andharm in Australia. Drug Alcohol Rev 2008; 27: 243.

    4. Darke S., Kaye S., McKetin R., Duflou J. Major physical and

    psychological harms of methamphetamine use.   Drug

    Alcohol Rev 2008; 27: 253.

    5. Everitt B. J., Wolf M. E. Psychomotor stimulant addiction: a

    neural systems perspective. J Neurosci 2002; 22: 3312–20.

    6. Volkow N. D.,Chang L., Wang G.-J., Fowler J.S., Leonido-Yee

    M., Franceschi D. et al. Associationof dopamine transporter

    reduction with psychomotor impairment in methamphet-

    amine abusers. Am J Psychiatry 2001; 158: 377–82.

    7. Spanagel R., Weiss F. The dopamine hypothesis of reward:

    past and current status.  Trends Neurosci   1999;   22: 521– 

    7.

    8. White F., Kalivas P. Neuroadaptions involved in amphet-

    amine and cocaine addiction.  Drug Alcohol Depend   1998;

    51: 141–53.

    9. Trudeau L.-E. Glutamate co-transmission as an emerging

    concept in monoamine neuron function.   J Psychiatry

    Neurosci  2004; 29: 296–310.

    10. Kalivas P., McFarland K., Bowers S., Szumlinski K., Xi Z.,

    Baker D. Glutamate transmission and addiction to cocaine.

    Ann N Y Acad Sci 2003; 1003: 169–75.

    11. Shearer J., Gowing L. Pharmacotherapies for problematic

    psychostimulant use: a review of current research.   Drug

    Alcohol Rev 2004; 23: 203–11.

    12. Srisurapanont M., Jarusuraisin N., Kittirattanapaiboon P.

    Treatment for Amphetamine Dependence and Abuse (Cochrane

    Review). Oxford: The Cochrane Library, Update Software;

    2001.

    13. Murillo-Rodriguez E., Haro R., Palomero-Rivero M., Millan-

    Aldaco D., Drucker-Colin R. Modafinil enhances extracellu-

    lar levels of dopamine in the nucleus accumbens and

    increases wakefulness in rats.  Behav Brain Res 2007;  176:

    353–7.

    14. Madras B. K., Xie Z., Lin Z., Jassen A., Panas H., Lynch L.

    et al.   Modafinil occupies dopamine and norepinephrinetransporters   in vivo   and modulates the transporters and

    trace amine activity   in vitro.  J Pharmacol Exp Ther   2006;

    319: 561–9.

    15. Willie J., Renthal W., Chemelli R., Miller M., Scammell T.,

    Yanagisawa M.   et al.   Modafinil more effectively induces

    wakefulness in norexin-null mice than wild-type litter-

    mates.  Neuroscience 2005; 130: 983–95.

    16. Scammell T., Estabrooke I., McCarthy M., Chemelli R.,

    Yanagisawa M., Miller M.   et al.   Hypothalamic arousal

    regions are activated during modafinil-induced wakeful-

    ness. J Neurosci 2000; 20: 8620–8.

    17. Dackis C. New treatments for cocaine abuse.  Drug Discov

    Today 2005; 2: 79–86.

    18. Stoops W., Lile J., Fillmore M., Glaser P., Rush C. Reinforcingeffects of modafinil: influence of dose and behavioural

    demands following drug administration.   Psychopharmacol-

    ogy 2005; 182: 186–93.

    19. Robertson P., Hellriegel E. Clinical pharmacokinetic profile

    of modafinil. Clin Pharmacokinet 2003; 42: 123–37.

    20. O’Brien C., Dackis C., Kampman K. Does modafinil produce

    euphoria? Am J Psychiatry 2006; 163: 6.

    21. McGregor C., Srisurapanont M., Mitchell A., Wickes W.,

    White J. Symptoms and sleep patterns during inpatient

    treatment of methamphetamine withdrawal: a comparison

    of mirtazapine and modafinil with treatment as usual.   J 

    Subst Abuse Treat 2008; 35: 334–42.

    22. Jasinski D.,Kovacevic-Ristanovic R. Evaluation of the abuse

    liability of modafinil and other drugs for excessive daytimesleepiness associated with narcolepsy. Clin Neuropharmacol

    2000; 23: 149–56.

    23. Dackis C., Lynch K., Yu E., Samaha F., Kampman K.,

    Cornish J.   et al.   Modafinil and cocaine: a double-blind,

    placebo-controlled drug interaction study.   Drug Alcohol

    Depend  2003; 70: 29–37.

    24. Malcolm R., Swayngim K., Donovan J., Devane C., Elkashef 

    A., Chiang N. et al. Modafinil and cocaine interactions. Am J 

    Drug Alcohol Abuse 2006; 32: 577–87.

    25. Dackis C., Kampman K., Lynch K., Pettinati H., O’Brien C.

    A double-blind, placebo-controlled trial of modafinil for

    cocaine dependence.   Neuropsychopharmacology   2005;   30:

    205–11.

    232   James Shearer  et al.

    © 2009 The Authors. Journal compilation © 2009 Society for the Study of Addiction   Addiction, 104, 224–233

  • 8/17/2019 Shearer Et Al 2009 Addiction

    10/10

    26. WHO.  Composite International Diagnostic Interview. Sydney:

    WHO Collaborating Centre for Mental Health and Sub-

    stance Abuse; 1997.

    27. Baker A., Lee N., Claire M., Lewin T., Grant T., Pohlman S.

    et al.   Brief cognitive behavioural interventions for regular

    amphetamine users: a step in the right direction.   Addiction

    2005; 100: 367–78.

    28. Darke S., Hall W., Wodak A., Heather N., Ward J. Develop-

    ment and validation of a multi-dimensional instrument for

    assessing outcome of treatment among opiate users: the

    Opiate Treatment Index. Br J Addict 1992; 87: 733–42.

    29. Barrowcliff A., Champney-Smith J., McBride A. Use of a

    modified version of the Opiate Treatment Index with

    amphetamine users: validation and pilot evaluation of a

    prescribing service. J Subst Use 1999; 14: 98–103.

    30. Del Boca F., Darkes J. Enhacing the validity and utility of 

    randomized clinical trials in addictions treatment research:

    II. Participant samples and assessment.   Addiction   2007;

    102: 1194–203.

    31. Derogatis L. Brief Symptom Inventory (BSI): Administration,

    Scoring and Procedures Manual. Minneapolis: National Com-

    puter Systems; 1993.

    32. Gossop M., Darke S., Griffiths P., Hando J., Powis B., Hall W.et al. The Severity of Dependence Scale (SDS): psychometric

    properties of the SDS in English and Australian samples of 

    heroin, cocaine and amphetamine users.  Addiction   1995;

    90: 607–14.

    33. Lewis J. Working to recognised standards: a prerequisite for

    drug testing in Australia.  Ther Drug Monit 2002; 24: 182– 

    6.

    34. Vickers A., AltmanD. Analysing controlled trials with base-

    line and follow up measurements.  BMJ  2001; 323: 1123– 

    4.

    35. Zeger S., Liang K.-Y. Longitudinal data analysis for discrete

    and continuous outcomes.   Biometrics   1986;   42: 121– 

    30.

    36. Zeger S., Liang K.-Y., Albert P. Models for longitudinal data:

    a generalized estimating equation approach.   Biometrics

    1988; 44: 1049–60.

    37. Mazumdar S., Tang G., Houck P., Dew M., Begley A., Scott J.

    et al.  Statistical analysis of longitudinal psychiatric data

    with dropouts. J Psychiatr Res 2007; 41: 1032–41.

    38. Twisk J. Applied Longitudinal Data Analysis for Epidemiology.

    Cambridge: Cambridge University Press; 2003.

    39. Mackris A., Rush C., Frederich R., Kelly T. Wake-promoting

    agents with different mechanisms of action: comparison of 

    effects of modafinil and amphetamine on food intake and

    cardiovascular activity. Appetite 2004; 42: 185–95.

    40. Broughton R. J., Fleming J. A. E., George C. F. P., Hill J. D.,

    KrygerM. H.,Moldofsky H. et al. Randomized, double-blind,

    placebo-controlled crossover trial of modafinil in the treat-

    ment of excessive daytime sleepiness in narcolepsy. Neurol-

    ogy 1997; 49: 444–51.

    41. Guilleminault C., Aftab F., Karadeniz D., Philip P., Leger D.

    Problems associated with switch to modafinil—a novel

    alerting agent in narcolepsy.  Eur J Neurol 2000; 7: 381–4.

    42. Girish M., Shephard J., Silber M. Change from conventional

    stimulants to modafinil in patients with narcolepsy or idio-

    pathic hypersomnia. Sleep 2000; 23: A302.

    43. Schmitz J., Mooney M., Moeller F., Stotts A., Green C.,Grabowski J.  et al.  Levodopa pharmacotherapy for cocaine

    dependence: choosing the optimal behavioral platform.

    Drug Alcohol Depend  2008; 94: 142–50.

    44. Cruikshank C., Montebello M., Dyer K., Quigley A.,

    Blaszczyk J., Tomkins S.  et al.   A placebo-controlled trial of 

    mirtazapine for the management of methamphetamine

    withdrawal.  Drug Alcohol Rev 2008; 27: 326–33.

    45. Sinha R., Garcia M., Paliwal P., Kreek M. J., Rounsaville B. J.

    Stress-induced cocaine craving and hypothalamic– 

    pituitary–adrenal responses are predictive of cocaine

    relapse outcomes. Arch Gen Psychiatry  2006; 63: 324–31.

    46. Volkow N., Fowler J. Addiction, a disease of complusion and

    drive: involvement of the orbitofrontal cortex. Cereb Cortex

    2000; 10: 318–25.

    Modafinil for methamphetamine dependence   233

    © 2009 The Authors. Journal compilation © 2009 Society for the Study of Addiction   Addiction, 104, 224–233